You are on page 1of 22

Microbial Pathogenesis 123 (2018) 505–526

Contents lists available at ScienceDirect

Microbial Pathogenesis
journal homepage: www.elsevier.com/locate/micpath

Nanoparticles and their antimicrobial properties against pathogens T


including bacteria, fungi, parasites and viruses
Arezou Khezerloua, Mahmood Alizadeh-Sanib, Maryam Azizi-Lalabadib, Ali Ehsanic,∗
a
Talented Students Center, Students Research Committee, Department of Food Sciences and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of
Medical Sciences, Tabriz, Iran
b
Students Research Committee, Department of Food Sciences and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
c
Nutrition Research Center, Department of Food Sciences and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran

A R T I C LE I N FO A B S T R A C T

Keywords: In recent year, propagation and resistance of pathogenic microorganisms (bacteria, fungi and virals) to common
Nanoparticles antimicrobial agents has led to serious health and food problems. Today, nanotechnology science and nano-
Reactive oxygen species particles (NPs) have been identified as a new approach to deal with this problem because of their inherent
Antimicrobial activity antimicrobial activity. Several studies have reported that, NPs (metal and metal oxide) are considered as a group
Pathogenic microorganisms
of materials that can be studied due to their antimicrobial properties. In this review, we investigated recent
Cell toxicity
studies regarding the antimicrobial activity of NPs with their mechanism of action. Many research has proved
that particle size is a significant factor which indicates the antimicrobial effectiveness of NPs. The use of NPs as
antimicrobial component especially in the food additives and medical application can be one of the new and
considerable strategies for overcoming pathogenic microorganisms. Nevertheless, more studies must be con-
ducted to minimize the possible toxicity of NPs in order to use as suitable alternatives for disinfectants and
antibacterial agents in food applications.

1. Introduction mass transfer, dissolution rate and catalytic activity [4,6].


The exact mechanisms mentioned for the antimicrobial effects of
Emergence of pathogenic and spoilage bacteria resistant to anti- NPs are still being studied. Up to now two popular possibilities have
microbial agents has become a serious health issue; thus, many studies been proposed: (a), free metal ion toxicity arising from dissolution of
have been accomplished with the aim of improving the current anti- metals from the surface of NPs (b), oxidative stress via generation of
microbial methods. It is implicated that over 70% of bacterial causing reactive oxygen species (ROS) on the surface of NPs [7]. Moreover,
poisoning and infection are resistant to one or more of the antimicrobial morphological and physicochemical characteristics of NPs have been
agents that are generally used for eradicating infection treatment of demonstrated to affect the antimicrobial activities of metals [2,8]. It has
poisoning. Development of new and effective antimicrobial agents been proven that small NPs have stronger bactericidal effects
seems to be of paramount importance. Metal nanoparticles (NPs) such [4,7,9,10]. Positive surface charge of metal NPs facilitates their binding
as copper (Cu), titanium (Ti), silver (Ag), gold (Au), and zinc (Zn), each to bacteria with negative surface charge which may result in en-
have various antimicrobial activity properties, with potencies and hancement of bactericidal effects [2]. The shape of NPs also influences
spectra of activity, which have been known and applied for decades [1]. its antimicrobial activities [11,12]. NPs have been proposed as antiviral
The type of materials used for preparing NPs as well as particle size agents using the core material and/or the ligands shell [13]. In this
are two important factors affecting resultant antimicrobial efficiency article, we focused on the latest findings regarding antimicrobial effects
and effectiveness [2,3]. Generally, NPs have different properties com- of most commonly employed NPs and their mechanism of action. Due
pared to the same material with larger particles. In fact, surface/volume to the promising development and wide application of NPs, under-
ratio of NPs increases considerably with decrease in particle size [4,5]. standing their non-toxicity and properties is necessary. For this reason,
Indeed, in nanometer dimensions, fraction of surface molecule notice- nanotechnology and pharmaceutical sciences have been used NPs for
ably increases which in turn improves factors such as heat treatment, reducing the toxicity and side effects of drugs and other material;


Corresponding author. Department of Food Sciences and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Golgasht
Ave, Attar-Neishaboori St., Tabriz, Iran.
E-mail address: ehsani@tbzmed.ac.ir (A. Ehsani).

https://doi.org/10.1016/j.micpath.2018.08.008
Received 26 February 2018; Received in revised form 15 July 2018; Accepted 6 August 2018
Available online 07 August 2018
0882-4010/ © 2018 Elsevier Ltd. All rights reserved.
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

Fig. 1. Mechanisms of antimicrobial activity of NPs against pathogens (B). NPs and their ions (e.g., titanium, silver and zinc) generate free radicals, and lead to
induction of oxidative stress (i.e.,ROS) (A). The generated ROS can damage and destroy the cellular components of the pathogens irreversibly, (e.g., membrane, DNA,
protein and mitochondria), resulting in cell death.

nevertheless, there are few safety concerns regarding NPs. According to 2. Antimicrobial properties of selected types of NPs
reports, respiratory and neurological damage, circulatory problems and
toxic effects of NPs are the main concerns in using NPs [14–16]. Indeed, 2.1. Titanium dioxide NPs (TiO2)
several types of NPs are considered non-toxic and some of them are
provided non-toxic with beneficial health effects [17]. Using NPs due to Antimicrobial activity of TiO2 NPs is attributed to its crystal struc-
their antimicrobial activities for overcoming spoilage and pathogenic ture, size and shape (Fig. 3) [18]. Oxidative stress caused by ROS is
microorganisms can be considered as one of these valuable health ap- particularly the mechanism proposed for TiO2 NPs. As a result, ROS
proaches. cause site specific DNA damage Figs. 1 and 2, [19,20]. Resting stages,
particularly bacterial endospores, fungal spores and protozoan cysts,

506
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

Maneerat et al. (2006) were investigated the antifungal activity of


TiO2 photocatalytic reaction in the form of TiO2 powder and TiO2
coated on a plastic film against P. expansum in vitro and in fruit tests.
Their findings suggested that “TiO2 photocatalytic reaction” indicates
antifungal effects against P. expansum which may have potential for
postharvest disease control [25]. Bodaghi et al. (2013) evaluated the
photocatalytic antimicrobial effects of TiO2 nanocomposite LDPE film
on P. spp. and Rhodotorula mucilaginosa. The most effects were reported
by combining UVA illumination and composite LDPE film against tested
bacteria [26]. Roy et al. (2010) investigated the effect of TiO2 NPs
along with different antibiotics against methicillin-resistant S. aureus
(MRSA). They recorded that, TiO2 NPs improve the antimicrobial effect
of different antibiotics against MRSA. In another study, antimicrobial
resistance of MRSA against various antibiotics reduces in the presence
of TiO2 NPs [19]. Gumiero et al. (2013) investigated effect of TiO2
photocatalytic activity in a HDPE-based food active packaging on the
microbiological stability of a short-ripened cheese. According to their
Fig. 2. Mechanism of photocatalytic activity of Cu/Ag doped TiO2 or ZnO and results, the growth of lactic acid bacteria and coliforms is inhibited by
producing ROS under irradiation.
using this type of packaging [27]. In another study, Chorianopoulos
et al. (2011) showed the significant photocatalytic antibacterial activity
are generally more resistant than the vegetative forms, possibly due to of TiO2 NPs against L. monocytogenes bacterial biofilm disinfection in
the increased cell wall thickness. The killing mechanism involves de- food processing [28].
gradation of the cell wall and cytoplasmic membrane due to the pro- Haghighi et al. (2013) evaluated the antifungal activity of TiO2 NPs
duction of reactive oxygen species such as hydroxyl radicals and hy- on fungal biofilms standard strains of C. albicans. According to results,
drogen peroxide. This initially leads to leakage of cellular contents then the synthesized TiO2 NPs improved the antifungal effect of C. albicans
cell lysis and may be followed by complete mineralization of the or- biofilms on fluconazole resistant strain. The authors suggested that
ganism. The general scheme for the photocatalytic damage of micro- TiO2 NPs can effectively inhibit the fungal biofilms particularly those
organism cells by TiO2 photocatalytic properties involves several steps formed on the surface of instruments [18]. Alizadeh Sani et al. (2017)
[21]: (1) the photoexcited TiO2 catalyst produces electron-hole pairs investigated the antibacterial effects of bio-nanocomposite films in-
that migrate to the TiO2 surface; (2) photogenerated holes in TiO2 can corporated with TiO2 NPs on Gram positive (L. monocytogenes, S.
react with adsorbed H2O or OH- at the catalyst/water interface to aureus) and Gram negative (E. coli O157:H7, S. enteritidis and P. fluor-
produce the highly reactive hydroxyl radicals and the electrons can escens) bacteria in packaged lamb meat samples. They demonstrated
react with oxygen vacancies to form superoxide ions; (3) finally, the that TiO2 NPs significantly inhibited the growth of test bacteria, espe-
various highly active oxygen species generated can oxidize organic cially on the Gram positive bacteria [29]. Photocatalytic properties of
compounds/cells adsorbed on the TiO2 surface, resulting in the death of TiO2 NPs help them eradicate bacteria efficiently. In fact, it must be
the microorganisms. The efficiency of TiO2 photocatalytic properties indicated that TiO2 NPs produce ROS under UV light. Carre et al.
and antibacterial activities should depend on our abilities (i) to make (2014) mentioned that the antibacterial photocatalytic activity is ac-
stable nanostructured TiO2 particles or composites, (ii) to generate companied by lipid peroxidation which increase the membrane fluidity
electron-hole pairs by extending the excitation wavelength to the and disrupt cell integrity [30]. Nevertheless, using TiO2 NPs under UV
visible light region, and (iii) to achieve a reduced recombination rate on light is restricted due to genetic damage in human cells and tissues
the newly created electron-hole carriers. Several methods may be used [31]. It has been demonstrated that, doping TiO2 NPs with metal ions is
to improve the photo-catalytic efficiency, including increase of the a significant strategy for overcoming this problem. Moreover, anti-
surface area of TiO2 through tailoring particle size and pore-size dis- bacterial and photocatalytic properties of TiO2 NPs are significantly
tributions, generation of defect structures to induce space-charge se- increased by doping them with metal ions Fig. 2 [31,32]. In other
paration via metal dopants, and surface modification of the TiO2 with a words, doping with metal ions shifts TiO2 NPs light absorption range to
metal or another semiconductor [22–24]. visible light and therefore, there is no need to irradiate them with UV

Fig. 3. Physicochemical properties of the NPs involved in biological activity: size, shape, surface area, chemical composition, surface chemistry, and interactions
between NPs and proteins or receptors that lead to the agglomeration and aggregation in the cell membrane.

507
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

light (Abamor et al., 2016). Incorporation of TiO2 NPs with nontoxic They evaluated the antibacterial activity of ZnO NPs according to
polymers is a new another approach to overcome the toxicity problems concentration and particle size reduction. They indicated that the an-
of TiO2 NPs. tibacterial activity of ZnO NPs increases with reducing particle size and
increment concentration; however, ZnO bulk showed no significant
2.2. Zinc oxide NPs (ZnO) antibacterial activity [47]. Few studies have proven that preparation of
metal ion doped NPs could improve the antimicrobial properties of NPs
The safety of ZnO and its compatibility within human skin make it a [32,48,49]. Akbar et al. (2014) evaluated the antibacterial effects of
proper additive for food application and surfaces that come in contact ZnO NPs that were prepared using hydrothermal synthesis against S.
with the human body and food products (Ravindranadh et al., 2013; typhimurium and S. aureus. They demonstrated that the ZnO NPs loaded
Hajipour et al., 2012). Indeed, ZnO NPs show antibacterial effects on film effectively inhibited the growth of foodborne pathogens bacteria
Gram-positive and Gram-negative bacteria as well as spores which are [50]. Sun et al. (2014) synthesized titanium-doped ZnO powders from
resistant to high temperature and high pressure (Li et al., 2013). The different zinc salts. Results displayed that titanium doped ZnO powders
efficient antibacterial activity of ZnO NPs compared to micro-particles had antibacterial activity against E. coli and S. aureus. The authors
is related to nanoparticle's surface area enhancement [2,33,34]. Ya- claimed that the antibacterial properties of titanium-doped ZnO pow-
mamoto et al. (2001) investigated influence of concentration and par- ders are related to crystallinity and particle size reduction Fig. 2 [48].
ticle size on the antibacterial activity of ZnO NPs against S. aureus and Furthermore, ZnO NPs exhibit high photocatalytic characteristics which
E. coli. Based on his results, it was observed that the antibacterial ac- improve their antimicrobial properties and produce ROS under UV light
tivity of ZnO increased with reducing particle size and increasing as well [20,51]. An increasing antimicrobial effects of ZnO NPs was
powder concentration [35]. Padmavathy et al. (2008) evaluated the shown by Azam et al. (2012) against the following microorganisms, in
antibacterial effects of ZnO NPs with various particle sizes. The results the order of sensitivity: A. niger < C. albicans < P. aeruginosa
indicated that the bactericidal efficacy of ZnO NPs increases by decline (G−) < E. coli (G−) < S. aureus (G+) < B. subtilis (G+) [36]. The
in particle size [34]. antimicrobial activity of ZnO NPs have been proven against C. albicans.
Azam et al. (2012) illustrated the comparative antimicrobial activity ZnO was added into edible food linings which were used in packaging
of ZnO, CuO, and Fe2O3 NPs against Gram-negative (E. coli and P. to avoid spoilage and to maintain the color, its mechanical capability,
aeruginosa) and Gram-positive (S. aureus and B. subtilis) bacteria. constancy and food safety [52]. Moreover, ZnO represented antifungal
According to results, the highest Antibacterial activity was reported for activity against Pythium spp [53]. Antifungal activities of ZnO NPs and
ZnO NPs while Fe2O3 NPs exhibited the least bactericidal effect [36]. their mode of action against two postharvest pathogenic fungi (Botrytis
In particular, ZnO decreases bacteria viability; however, the main cinerea and P. expansum) were investigated. These results suggest that
and accurate mechanism of its antibacterial activity has not been well ZnO NPs could be used as an effective fungicide in agricultural and food
understood. One proposed possibility is the generation of hydrogen safety applications [44]. Another study, promising particle size de-
peroxide (H2O2) as the main factor of antibacterial activity. According pendent antibacterial and antifungal activities of the ZnO NPs have
to results, another mechanism mentioned for the antibacterial effect of been observed [54]. Thessicar et al. (2012) provided new insights into
ZnO NPs is the accumulation of particles on the bacteria surface due to the anti-HSV-2 effects of ZnO and rationalize their development as a
electrostatic forces [37]. In addition, ROS generation on the surface of HSV-2 trapping agent for the prevention and/or treatment of infection.
particles, zinc ion release, membrane dysfunction, and NPs inter- The observed results also demonstrate that blocking HSV-2 attachment
nalization could also be considered as possible reasons for cell damage can have prophylactic as well as therapeutic applications [55].
[38]. Moreover, in the case of few metal NPs such as Ag and Zn, in-
terruption of transmembrane electron transportation has been stated 2.3. Silver NPs (Ag/Ag2O)
[39–41]. Espitia et al. (2013) studied antimicrobial activity ZnO NPs
against foodborne pathogens and spoilage microorganisms. Based on According to evidences, Ag NPs are considered as the most common
their results, ZnO NPs had no considerable antimicrobial activity inorganic NPs used as antimicrobial agents [56]. The antimicrobial
against P. aeruginosa, L plantarum, and L. monocytogenes. However, it application of Ag additives is widely benefitted in various plastic and
showed significant antimicrobial activity against E. coli, S. choleraesuis, biopolymer products, textiles and coating-based materials [57]. Ag NPs
S. aureus, Saccharomyces cerevisiae, and A. niger [42]. Xie et al. (2011) also possess a wide range of biomedical applications [1]. It has been
investigated the antibacterial activity of ZnO NPs against C. jejuni. They revealed that, Ag NPs show high antimicrobial activities compared to
suggested that the antibacterial mechanism of ZnO NPs can be attrib- their ionic form [58]. It has also been indicated that Ag NPs are po-
uted to the disruption of cell membrane and oxidative stress in C. jejuni. tential antimicrobial agents against wide range bacteria [31]. Ac-
Results showed that ZnO NPs lead to morphological changes and cording to researches, the antibacterial action of Ag NPs is resulted
membrane leakage, and increase oxidative stress gene expression in C. from damage of the outer membrane bacteria [59]. Many literature
jejuni [33]. Interestingly, Ag NPs show antibacterial activities even in revealed that Ag NPs can induce pits and gaps in the bacterial mem-
ultra-low concentrations [20]; nevertheless, the antibacterial activity of brane and eventually fragment the cell [60,61]. Additionally, Ag ions
ZnO NPs is depended on concentration and surface area. Thus ZnO NPs interact with disulfide or sulfhydryl groups of enzymes that lead to the
display better antibacterial activity in higher concentrations and larger disruption of metabolic processes and finally, cell death [57]. Shahverdi
surface area [4]. In another study, Liu et al. (2009) and He et al. (2011) et al. (2007) studied the effect of size reduction on the antimicrobial
indicated the effects of changing the concentration of ZnO NPs on the properties of Ag NPs. They used Ag NPs to control S. aureus and E. coli
inhibition of E. coli O157:H7 (bacteria) and Botrytis cinerea and P. ex- [62]. Jo et al. (2009) tested the efficacy of Ag NPs on different types of
pansum (fungi). The antibacterial activity increase as the concentration pathogens such as foodborne fungi which rarely produce spore. Ac-
of ZnO NP increased [43,44]. Similarly, Jin et al. (2008) illustrated that cording to their results, Ag NPs (20–30 nm) penetrate and colonize
increasing the concentration of ZnO NPs inhibited the growth of better within the plant tissue. They proposed that, Ag NPs were effec-
foodborne pathogenic bacteria including; L. monocytogenes, S. En- tive in controlling spore-producing fungal plant pathogens. They also
teritidis, and E. coli O157:H7 [45]. Hossein-khani et al. (2011) evaluated offered that Ag NPs may be less toxic than synthetic fungicides [58]. In
the antibacterial effects of ZnO nanoparticle against Shigella dysenteriae. another study, Mie and et al. (2014) evaluated the antibacterial activity
According to their results, a significant reduce in bacteria count was of synthesized Ag NPs against eight micro-organisms using the disk
observed as a result of particles size reduction [46]. Emami-Karvani diffusion method. The results showed that Ag NPs had potential anti-
et al. (2011) studied the antimicrobial characteristics of ZnO NPs bacterial activity against gram-negative bacteria. Thus, the authors
against Gram-positive (S. aureus) and Gram-negative (E. coli) bacteria. suggested that synthesized Ag NPs could be applied in difference

508
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

industries including food, pharmaceutical and biomedical [63]. Choi Considering all above-mentioned arguments, further research should be
et al. (2008) illustrated the inhibitory effects of silver NPs against E. coli conducted in order to develop Ag compounds and composites with at
PHL628-gfp [64]. In another study, the comparative was done between least toxicity and maximum antimicrobial activity.
bactericidal activity of Ag, ZnO, and Au NPs on S. mutans. The results
demonstrated that Ag NPs exhibit several effects in controlling S. mu- 2.4. Copper NPs (Cu/CuO)
tans. Therefore, the authors suggested that Ag NPs can be used in dental
caries since they are commonly caused by S. mutans [65]. Emamifar Cu NPs due to their unique biological, chemical and physical
et al. (2011) indicated effect of nanocomposite packaging containing Ag properties, antimicrobial activities as well as inexpensiveness are of
and ZnO on inhibition of L. plantarum in orange juice [66]. Birla et al. great interest to scientists [80–82]. Yoon et al. (2007) examined sus-
(2009) evaluated the antibacterial effect of Ag NPs against foodborne ceptibility constants of B. subtilis and E. coli to Ag and Cu NPs. They
bacteria namely E. coli, P. aeruginosa and S. aureus by disc diffusion proved that reaction of Cu NPs with B. subtilis illustrated the highest
method [67]. Zarei et al. (2014) investigated the antibacterial effect of susceptibility whereas the reaction of Ag NPs with E. coli displayed the
Ag NPs against four foodborne spoilage and pathogenic bacteria such as lowest one [83]. Usman et al. (2013) evaluated the antimicrobial ac-
S. typhimurium, L. monocytogenes, V. parahaemolyticus and E. coli. Ac- tivities of Cu-chitosan NPs. In their studies they assessed the anti-
cording to the results, Ag NPs had significant antibacterial effects on the bacterial and antifungal activities of NPs on several microorganisms,
mentioned bacteria. They concluded that Ag NPs can be a suitable al- including B. subtilis, P. aeruginosa, methicillin-resistant S. aureus, S.
ternative for cleaning and disinfecting equipment and surfaces in food- choleraesuis, and C. albicans. Additionally, they introduced these NPs as
related environments [68]. antimicrobial agents [81]. Nevertheless, Cu NPs act at high con-
Researchers have also been studied the shape-dependent properties centrations as a prooxidant, and rapid oxidation of Cu NPs limit their
of NPs. Pal et al. (2007) assessed the shape dependent antibacterial application [81,84]. Ren et al. (2009) investigated the antibacterial
activities of Ag NPs (spherical, rod-shaped and truncated triangular). properties of CuO NPs against S. aureus EMRSA-16 (epidemic methi-
The findings showed that, truncated triangular NPs had higher anti- cillin-resistant S. aureus), EMRSA-15, methicillin-resistant S. aureus
microbial activity because of their high-atom-density surfaces [11]. In (MRSA) 252, S. aureus ‘Golden’ and S. aureus Oxford (NCTC 6571); S.
another study, Bera et al. (2014) evaluated the size and shape-depen- epidermidis SE-51 and SE-4; E. coli NCTC 9001; P. aeruginosa PAOI, and
dent antimicrobial activity of Ag NPs against P. aeruginosa (Gram ne- Proteus spp. Results revealed that CuO NPs incorporated into polymers
gative bacteria) and S. epidermidis and B. megaterium (Gram positive suggest release of ions might be required for optimum bactericidal [85].
bacteria). They confirmed that the shape and size of these particles are Mahapatra et al. (2008) examined the antibacterial activity of CuO NPs
controlled by their activity. These results displayed that, the smaller against S. paratyphi, P. aeruginosa, Klebsiell apneumoniae, and Shigella
particles were able to easily penetrate the cell wall and enhance anti- strains. According to their results, these NPs showed efficient anti-
microbial activity. The authors offered that Ag NPs can be used for bacterial activity against these bacteria. The authors suggested that
different purposes such as bio adhesives, clinical therapy, biofilms, crossing NPs through the bacteria cell membrane and then damaging
coating materials and food packaging [12]. Rajeshkumar et al. (2014) the vital enzymes of bacteria were the critical factors that triggered cell
showed the antibacterial effect of Ag NPs against B. subtilis, K. planti- death. They also demonstrated that these NPs were not cytotoxic on
cola, K. pneumoniae, S. nematodiphila, and E. coli by disc diffusion HeLa cell line [84]. Bogdanovic et al. (2014) investigated the anti-
method [69]. Bahrami studied on Ag-Au alloy NPs for evaluating their microbial activity of Cu small NPs (∼5 nm) with narrow size distribu-
antimicrobial properties against S. aureus. The antibacterial activity of tion against microorganisms of public concern (E. coli, S. aureus and C.
Ag-Au alloy NPs intensified when combined with penicillin G and pi- albicans). Based on their results, Cu NPs exhibited significant anti-
peracillin [70]. Ag2O NPs have also been mentioned for their significant microbial activities toward different kinds of microorganisms (E. coli, S.
antimicrobial activity [31]. It is believed that, metal oxide NPs may be aureus, and C. albicans) [86]. Azam et al. (2012) showed the size-de-
considered as a new alternative for most antibacterial agents [31,71]. pendent antibacterial activity of CuO NPs. They evaluated the anti-
Sondi and Salopek (2004) illustrated the antimicrobial efficacy of Ag2O bacterial activities of CuO NPs against Gram-positive bacteria (S. aureus
NPs against E. coli. They indicated that when E. coli is exposed to these and B. subtilis) and Gram-negative bacteria (P. aeruginosa and E. coli).
NPs, DNA loses its replication ability and the cell cycle is halted at the Based on their results, CuO NPs exhibited inhibitory effects against both
G2/M phase resulting in DNA damage. Eventually, the cells are affected groups of bacteria. The authors concluded that the bactericidal activity
by apoptosis and oxidative stress, occurs [72]. Besides, Ag has been of CuO NPs depends on their size, stability, and concentration along
reported to be less toxic than most disinfectants. Jones and Hoek (2010) with growth medium. The authors also proved that, NPs restrict bac-
reviewed the antibacterial mechanisms of the Ag NPs and potential terial growth via passing through nanometeric pores present on the
implications in human health and environment [73]. cellular membranes of most bacteria [36,87]. Ahamed et al. (2014)
Ag NPs have been used to determine antiviral activity against HIV-1 showed that CuO NPs had significant antimicrobial activity against
at non-cytotoxic concentrations [74]. Based on their results, Ag NPs various bacterial (E. coli, Shigella flexneri, P. aeruginosa, Klebsiella
revealed clear anti-HIV activity at an early stage of viral replication, pneumonia, S. typhimurium Enterococcus faecalis, Proteus vulgaris, and S.
and was considered as a virucidal or an inhibitor of viral entry agent. aureus). Among these pathogens, K. pneumonia exhibited almost re-
The author suggested that, it is possible that, Ag NPs prevent fusion, sistant to these NPs while E. coli and E. faecalis showed highest sensi-
infectivity and CD4-dependent virion binding, and act as an effective tivity to CuO NPs [82]. In another study, Ramyadevi et al. (2012) tested
virucidal agent against both cell-associated virus and cell-free virus. the antimicrobial activity of Cu NPs against S. aureus, Micrococcus lu-
Furthermore, Ag NPs prevent post-entry stages of the HIV-1 life cycle. It teus, Klebsiella pneumoniae, E. coli, and P. aeruginosa, fungus like A.
can be concluded that these characteristics made them a wide-spectrum flavus, A. niger and C. albicans. The Cu NPs revealed more inhibitory
antiviral agent that could be used preventively against variety of viral activity in bacteria compared with the fungus and it also exhibited more
strains. Pinto et al. (2009) investigated inhibition of Herpes Simplex zone of inhibition in E. coli than C. albicans [88].
Virus Type 1 infection by Ag NPs capped with mercaptoethane sulfo-
nate. Ag NPs compete with virus for binding to cellular heparan sulfate 2.5. Gold NPs (Au)
through their sulfonate end groups. This results led to inhibition of viral
entry into the cell and prevent of subsequent infection [75]. These NPs Au NPs are considered to be valuable in the development of anti-
have no toxic effects on the host cells. Several other studies have also bacterial agents because of their nontoxicity, polyvalent effects, high
reported the antiviral activity of Ag NPs against the hepatitis B virus, ability to functionalization, ease of detection and photo thermal activ-
HIV 1, respiratory syncytial virus, and monkey-pox virus [76–79]. ities [89–92]. Although generation of ROS is the main cause of cellular

509
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

death for most antibacterial agents and NPs; however, antimicrobial bacteria. Based on their results, these NPs decreased about 90–95% of S.
activity of Au NPs is not induced by ROS-related processes [93]. typhi and E. coli colonies. The authors stated that, the main factors that
Giancivincenzo et al. (2010) evaluated Au NPs capped with amphiphilic influenced the bactericidal properties were roughness and dispersion of
sulfate-ended ligands as anti-HIV agents. Their results showed that, the Au NPs on the medium [89]. It seems that Au NPs are safer on mam-
Au NPs coated with multiple copies of an amphiphilic sulfate-ended malian cells than other NPs because of ROS-independent mechanisms.
ligand inhibit the HIV infection at nanomolar concentrations in vitro. Furthermore, the high ability of these NPs for functionalizing make
Nanoparticle with multiple ligands creates a many local binding mo- them ideal nanomaterials for applying as targeted antimicrobial agents.
lecules which can help the targeted biological interaction. The authors
suggested that the multivalent Au NPs can be considered as a novel anti 2.6. Silica NPs (SiO2)
HIV agent for targeting the fusion/adsorption process of the virus in-
fection [13]. Sametband et al. (2011) investigated effective inhibition The antimicrobial activity of SiO2 is more significant at nano-scale
of influenza virus by anionic Au NPs. Their results showed that, the due to surface area enhancement [100]. Cousins et al. (2007) revealed
anionic Au NPs have effective inhibition properties against several in- that Si NPs inhibit bacteria adherence to oral biofilms [101]. The
fluenza strains. The initial mechanism of inhibition Au NPs usually combination usage of Si NPs with other biocidal NPs such as Ag have
attributed to the blocking of viral attachment to cell surface. The var- been extensively investigated in the recent years. Kim et al. (2006) were
iation in the degree of inhibition with the anionic groups demonstrated examined the antibacterial properties of the Cu/SiO2 nanocomposite by
that the charge density and the functional groups play an important role disk diffusion methods. For antibacterial test, S. aureus, E. coli, E.
in the antiviral activity of the NPs [94]. The small size of the Au NPs cloacae, C. albicans, and P. citrinum were selected as indicators. Results
enables them to penetrate into the cell through the endosome vesicle demonstrated that the antibacterial activities of Cu-SiO2 nanocomposite
and possibly interfere with the fusion step as well [95]. Many studies were clearly detected against Gram-negative, Gram positive bacteria,
indicated low or no Au NPs cytotoxicity, and their toxicity if present is and fungi due to Cu NPs of Cu-SiO2 nanocomposite were well formed on
related to the size, shape and nanoparticle's surface charge [96]. The the surface of SiO2 nanoparticle without aggregation of the Cu NPs and
NPs contain Au core and mercaptoethanesulfonate molecules have had large surface area [102]. Similarly, Kim et al. (2007) were eval-
bonded to its surface through the thiol group showed no cytotoxicity uated the antibacterial properties of the Ag/SiO2 nanocomposite by
and inhibited various influenza virus strains. Cui et al. (2012) in- disk diffusion methods against S. aureus, P. aeruginosa, E. coli, E. cloacae,
vestigated that the antibacterial activity of Au NPs is attributable to 1) C. albicans, A. niger and P. citrinum And achieved similar results [103].
attachment of these NPs to the bacteria membrane followed by mem- Egger et al. (2009) assessed the antimicrobial activity of novel Ag-Si
brane potential modification and ATP level decrease 2) inhibition of nanocomposite. Their results revealed better antimicrobial function of
binding tRNA to ribosome [93]. Tiwari et al. (2011) tested the anti- nanocomposite against a wide range of microorganisms compared to
bacterial and antifungal activities of Au NPs functionalized with 5- conventional materials, such as silver nitrate and silver zeolite [57]. In
fluorouracil against M. luteus, S. aureus, P. aeruginosa, E. coli, A. fumi- another study, Mukha et al. investigated synthesized Ag/SiO2 and Au/
gatus, and A. niger. The authors reported that these NPs are efficient on SiO2 nanostructures and their antimicrobial activity. Results indicated
Gram negative bacteria than Gram positive ones because of their easier that Ag/SiO2 nanocomposites improved antimicrobial properties
internalization into Gram negative bacteria. Additionally, they proved against E. coli, S. aurous, and C. albicans while Au/SiO2 nanocomposites
that these NPs indicate antifungal activity against A. fumigatus and A. did not show any antibacterial activity against the tested microorgan-
niger [90]. Zawrah et al. (2011) illustrated antimicrobial activities of isms. The authors claimed that these nanocomposites could be used for
drugs (fluconazole and ciprofloxacin) coated with Au NPs. Results water disinfection and food and pharmaceutical application [104]. In
showed that, antimicrobial activities of Au NPs had been increased with another study, Xu et al. (2009) were tested the antibacterial effects of
increasing volume. According to results, Best antifungal activity ob- Ag-SiO2 core-shell particles against E. coli and S. aureus by the usual
served on using fluconazole coated with Au NPs against A. niger, C. serial dilution method for determine minimal inhibitory concentration
albicans and A. flavus. Minimum inhibitory concentration test (MIC) (MIC) and minimal bactericidal concentration (MBC). According to
revealed synergistic effect of Au NPs with ciprofloxacin. Best results results, the NPs showed considerable antibacterial effects [105]. Sev-
were demonstrated against both Gram negative (S. Typhimurium, E. coli eral studies showed that Si nanowires could interface with living cells
O157:H7 and P. aeruginosa) and Gram positive (L. monocytogenes, B. and bacteria interrupting cell functions such as cell differentiation,
cereus and finally S. aureus) [97]. Zhou et al. (2012) evaluated the adhesion and spreading. Li et al. (2004) revealed the antibacterial ef-
antibacterial activities of Au and Ag NPs against E. coli and B. Calmette- fects of Ag NPs Si nanowires. Their results indicated high antibacterial
Guerin (BCG). They asserted that, Au and Ag NPs exhibit significant activity for these nanostructures. They also showed that, these nanos-
antibacterial activity against both Gram negative (E. coli) and Gram tructures are biocompatible with human cell namely lung adenocarci-
positive bacteria (BCG). They also functionalized Au NPs with strong noma epithelial cell [9,106]. Fellahi et al. (2013) assessed the anti-
bound capping (poly-allylamine hydrochloride) and weak bound cap- bacterial activity of Si nanowire substrates decorated with Ag or Cu
ping agents (citrate). Poly-allylamine hydrochloride is able to directly NPs. According to author's results, the NPs revealed strong antibacterial
contact with bacteria cell membrane because of its positively charged activity against E. coli. Ag decorated with Si nanowires was found to be
nature [91]. In addition, Au NPs functionalized by strong bound cap- biocompatible with human cell while Cu decorated with Si nanowires
ping agents can self-assemble into 4–5 μm long chains [98]. Zhou et al. show high cytotoxicity [10]. All of these studies signify that the de-
(2012) explained that the two mentioned processes facilitate the de- velopment of Si compounds and composites especially their nano-
livery of a large number of Au NPs on the bacteria cell wall. In contrast, composites together with NPs such as Ag show good potential on the
extra-accumulation of weak bound capping agents such as citrate de- development of antimicrobial agents. In addition, the non-toxicity
crease surface area and reduce interactions with NPs. Accordingly, Au properties of Si NPs introduce them as antimicrobial agents in food and
nanoparticle with the same shape and size but different capping agent biomedical applications.
display different antimicrobial activities [73]. Jayaseelan et al. (2013)
proved antifungal activity of green synthesized of Au NPs using seed 2.7. Magnesium oxide NPs (MgO) and calcium oxide NPs (CaO)
aqueous extract of Abelmoschus esculentus against Puccinia graminis tritci,
A. flavus, A. niger and C. albicans using standard well diffusion method. Due to the alkalinity and active oxygen species of CaO and MgO,
The authors suggested that the synthesized Au NPs can be act as an they provide strong antibacterial activity. It has been proved that the
effective antifungal agent [99]. In another study, Lima et al. in- antibacterial mechanism of CaO and MgO NPs attributed to the gen-
vestigated the antimicrobial effect of Au NPs against S. typhi and E. coli eration of superoxide on the surface of these particles, and also increase

510
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

Fig. 4. Schematic comparison of NPs-induced general toxicity via active internalization mechanisms, and endocytosis-free NPs.
“NPs enter cells by energy-dependent processes, and rapidly confine in vesicular structures, endosomes, and lysosomes. Acidic lysosomal pH, triggers a lysosome-
enhanced Trojan horse effect (LETH effect) that combines the abundant cellular internalization of the NPs via active processes which consequently enhances the
release of relatively toxic ions (e.g., Ag, Au ions). Significant amounts of intracellularly leaked ions may then exert ion-specific toxicity (e.g., enzyme depletion/
inactivation, protein denaturation, etc.) against particular cellular targets (e.g., mitochondria, RER) and/or lysosomal damage/dysfunction. Finally, it results in ROS
level increase, apoptosis, DNA and membrane damage”.

in pH value via the hydration of CaO and MgO [107]. According to negative (E. coli) and Gram-positive (S. aureus) bacteria. Results ex-
results, MgO NPs damage the cell membrane and then cause the leakage hibited more antibacterial action against S. aureus compared to E. coli.
of intracellular contents which in turn lead to bacterial cell death [108]. Based on the authors results, the antibacterial effect of CaO and MgO
Hewitt et al. (2001) indicated that MgO initiates the sensitivity changes was because of the generation of superoxide on their surface and also
of E. coli induced by active oxygen [109]. Nevertheless, Leung et al. increase in pH value due to CaO and MgO hydration [107].
(2014) described that strong antibacterial activity of MgO NPs can be Vidic et al. (2013) investigated the antimicrobial activity of mixed
observed in the absence of ROS production. They declared that the nanostructures of ZnO-MgO. They also compared the antimicrobial
mechanism of antimicrobial activity may be due to cell membrane activity of ZnO-MgO NPs with pure ZnO and MgO NPs. Based on their
damage [110]. In fact, MgO NPs initiate bactericidal activity against findings, ZnO nanocrystals illustrated high antimicrobial activity
both Gram-positive and Gram negative bacteria [111]. Sawai et al. against both Gram-negative (E. coli) and Gram-positive (B. subtilis)
(2000) evaluated the antibacterial activity of MgO against E. coli and S. bacteria. MgO NPs exhibited moderate activity and ZnO-MgO NPs re-
aureus. They stated that the presence of active oxygen, such as super- vealed high antibacterial activity against Gram-positive bacteria.
oxide, on the surface of MgO NPs is one of the main factors that affects Microscopic analysis displayed that B. subtilis cells were damaged after
antibacterial activity [112]. Jin et al. (2011) also investigated the an- contact with ZnO-MgO NPs. They suggested that nanostructured ZnO-
tibacterial activities of MgO NPs alone or in combination with other MgO can be used as a safe and new therapeutic agent for overcome
antimicrobials (nisin and ZnO NPs) against E. coli and S. Stanley. MgO bacterial [111]. Results indicate that, MgO and CaO NPs alone or in
NPs demonstrated strong bactericidal activity against these pathogens combination with other antibacterial agents show great antibacterial
According to their results, the antibacterial activity of MgO NPs im- activities. These NPs also are inexpensive, available materials and
proved as MgO concentrations increased. The authors showed that MgO biocompatible. Thus they are considered as promising antibacterial
NPs alone or in combination with niacin could be used as an effective agents [110]. Researchers suggest that, these materials can be utilized
antibacterial agent for enhancing food safety [108]. Yamamoto et al. in environmental preservation as well as in medical treatments and
(2010) examined the antimicrobial efficiency of CaCO3 NPs. According food processing [113].
to results, CaCO3 was converted to CaO due to heat treatment. CaO NPs
displayed bactericidal activity against E. coli, S. typhimurium, S. aureus
and B. subtilis [107]. Yamamoto et al. (2010) evaluated the anti- 2.8. Aluminum oxide NPs (Al2O3)
bacterial activity of CaCO3/MgO nanocomposites against Gram-
Aluminum oxide NPs is used for different applications in industrial

511
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

Fig. 5. Molecular mechanisms of ROS-mediated cell death pathway.


“TiO2 NPs and ZnO NPs induce ROS-mediated cell apoptosis, autophagy or necrosis. The increase of ROS level in cytoplasm results in thioredoxin (Trx) oxidation and
apoptosis signal-regulating kinase 1 (ASK1) activation. ASK1 phosphorylate mitogen-activated protein kinases (ERKs, JNKs, and p38 MAPKs) that control the AP-1-
mediated synthesis of pro- (Bak, Bax) and anti-apoptotic (Bcl-2, Bcl-xL) proteins, as well as death ligands (FasL), and eventually promote cell apoptosis. Likewise,
ERKs promote cell apoptosis trough attenuation of AKT kinase activity that controls NF-κB-mediated synthesis of Bcl-2 and Bcl-xL proteins. The oxidative damages of
the mitochondria results in the dissipation of mitochondrial membrane potential (ΔΨm), decrease of ATP level and release of cytochrome c (Cyt. c) to the cytosol. Cyt.
c, bind to apoptotic protease activating factor 1 (Apaf-1) and induce caspase-dependent apoptosis. The increase of ROS level in nucleus results in the activation of
ataxia telangiectasia mutated (ATM), as well as ataxia telangiectasia and Rad 3-related (ATR) kinases which induces p53-mediated apoptosis and increase DNA
double-strand breaks marker (γH2AX) level. ROS-induced endoplasmic reticulum stress results in JNK1-mediated increase of pro-autophagic protein (Beclin-1) level
and AMPK-mediated suppression of autophagic inhibitors and the rapamycin (mTOR) kinase. Both processes lead to cell autophagy. Oxidative injury of the plasma
membrane results in the downregulation of the plasma membrane calcium ATPase 1 (PMCA1), and decrease ATP level and increase Ca2+ level” [333].

and personal care products. The antimicrobial effects of aluminum NPs phenomenon of bacterial cell adhesion onto the NPs usually attributed
have been evaluated on E. coli [114]. Aluminum NPs exhibited a to the electrostatic interaction between bacteria cell and nanoparticle
moderate growth-inhibitory effects against tested bacteria, only at very surface, along with hydrophobic interactions and polymer cross-link.
high concentrations. The antimicrobial effects of aluminum NPs are The antimicrobial properties of NPs are related to the generation of
usually attributed to surface charge interactions between the NPs and reactive oxygen species (ROS) which leads to disruption of cell wall and
cells membrane and wall and also penetration inside the bacterial cells. membrane and subsequently cell death [117]. Versus, Al NPs may act as
However, it is believed that the free-radical scavenging properties of the free radical scavengers. NPs can rescue cells from oxidative stress-in-
NPs might have prevented intense antimicrobial action and cell wall duced cell death so that appears to be dependent on the structure of the
disruption [114]. nanoparticle and independent of its size [118]. Ansari et al. (2013)
Aluminum NPs has a corundum-like structure. It is thermo- investigated the antibacterial activity of Al NPs against methicillin-re-
dynamically stable over a wide range of temperature [115]. The Al NPs sistant coagulase negative S. aureus by serial dilution method and de-
at close-neutral pH carry a positive charge on its surface. The electro- termination of MIC. They presented that a novel Al NPs are effective
static interaction between the negatively charged microorganisms bactericidal agents regardless of the drug resistance mechanisms that is
namely E. coli cells and the NPs leads to the adhesion of Al NPs on the important to these bacteria as a pathogen [119]. Recently, the anti-
bacterial surfaces [116]. Increasing the concentration of NPs in the microbial activity of Al NPs to microalgae has been examined by Sadiq
suspension increased the reaction and adhesion levels. The et al. (2011) [120]. They evaluated micron and nano sized Al NPs

512
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

Table 1
NPs as antimicrobial agent, properties and their mechanisms of action.
NPs (NPs) The main factors for The proposed antimicrobial mechanism for NPs Important properties as antimicrobial agent Reference
antimicrobial activity

TiO2 NPs Crystal structure, shape No toxicity in dark condition; Oxidative stress via the Suitable photocatalytic properties; high stability; effective [170]
and size. generation of ROS; lipid peroxidation that lead to enhance antifungal for fluconazole resistant strains. [171]
membrane fluidity and disrupt the cell; Toxic only under UV [172]
illumination and killed approximately all bacteria. [173]
Photoactivation of TiO2 promotes bactericidal effect [174]
peroxidation of the polyunsaturated phospholipid of [175]
membrane; loss of respiratory activity [176]

ZnO NPs Particle size and ROS generation on the surface of the particles; zinc ion Photocatalytic activity; high stability; bactericidal effects on [177]
concentration. release; membrane dysfunction and NPs internalization into both Gram-positive and Gram-negative bacteria; antibacterial [178]
cell; Electrostatic interaction, morphological changes in the activity against spores which are resistant to high [179]
presence of ZnO NPs, increase in membrane permeability temperature and high pressure. [180]
and NPs accumulation in the cytoplasm
Ag NPs Particle size, Induction of pits and cavity in the bacterial cell membrane; High antimicrobial activity against both bacteria and drug- [181]
concentration and shape interact with disulfide or sulfhydryl groups of enzymes and resistant bacteria, antifungal activity on spore- producing [182]
of particles. disruption of metabolic processes. Ion release; DNA loses its fungal plant pathogens, high stability, nontoxicity [65]
replication ability and the cell cycle halts at the G2/M phase
owing to the DNA damage (in the case of Ag2O). Disturbs
permeability, respiration, and cell division, interacts with
cell membrane and sulfur- and phosphorus- containing
compounds, Ag NPs-ampicillin leads to cell wall lysis,
penetration of Ag NPs, and prevents DNA unwinding
CuO NPs Particle size and Crossing of NPs from the bacteria cell membrane and then Effective against gram positive and gram-negative bacteria; [85]
concentration. damaging the vital enzymes of bacteria. high stability; antifungal activity. killing bacteria [183]
Au NPs Roughness and particle Attachment of these NPs to membrane which change the Nontoxicity, not inducing any ROS-related process; high [184]
size. membrane potential and then cause the decrease the ATP ability to functionalization, polyvalent effects; ease of [185]
level; and inhibition of tRNA binding to the ribosome detection; photothermal activity.
SiO2 NPs Particle size and shape. Influencing the cell functions such as cell differentiation, Non-toxicity; stability. [102]
adhesion and spreading. [186]
MgO/CaO Particle size, pH and Damaging the cell membrane and then causing the leakage Effective against both grampositive and gram-negative [187]
NPs concentration. of intracellular contents and death of the bacterial cells. bacteria; high stability; low cost; availability. [111]
[188]
Al2O3 NPs Particle size and bacterial attachment (electrostatic interaction); Damage to Toxicity of NPs is from their higher tendency to attach to the [189]
concentration. the bacterial cell wall and increase the permeability cell walls [190]
[191]
[192]
Clay NPs Shape and concentration Electrostatic interaction, adsorption, and penetration of NPs The most effective against gram positive [136]
of particles. and toxicity [193]
[194]

against Scenedesmus sp. and Chlorella sp. According to their results, Al According to their results, nano-hybrids of Ag/clay NPs significantly
NPs exhibited a growth inhibitory effect against both the species and a inhibited the growth of pathogenic bacteria tested including methi-
considerable reduce in the chlorophyll content was also observed in the cillin-resistant S. aureus and E. coli, and Ag/clay NPs showed a stronger
cells treated with NPs. The authors were suggested that an interaction biocidal effect than Ag NPs. Antimicrobial effects are usually attributed
of the NPs with the cell surface can be as the possible mechanism for the to encapsulated bacteria and triggers death signals from the cell
toxicity Al NPs against microalgae's. membrane. These observations suggest that the high electrostatic affi-
nity of clay NPs onto the bacterial surface and facilitate Ag/clay NPs-
cell interaction, causing local membrane damage, finally, cell death
2.9. Clay NPs
[132]. Paulraj Kanmani et al. (2014) evaluated the antimicrobial
properties active nanocomposite films containing gelatin, Ag NPs and
Clay NPs known as layered mineral silicates. These NPs are orga-
nanoclay against E. coli and L. monocytogenes pathogenic bacteria using
nized into several classes such as montmorillonite, halloysite, kaolinite,
agar well diffusion and colony count methods. Based on their results,
bentonite and hectorite, depending on chemical composition and na-
the Ag NPs showed high antimicrobial activity against both bacteria,
noparticle morphology [121–124]. A new group of clay NPs called
but the clay NPs illustrated antimicrobial activity only against to Gram
Organically-modified nanoclays. Organoclays are an attractive class of
positive (L. monocytogenes) bacteria [133]. This property is usually re-
hybrid organic-inorganic NPs that are used for various purposes in
lated to the strong antimicrobial activity of alkyl quaternary ammo-
polymer nanocomposites such as rheological modifiers, gas absorbents,
nium salt group in the organoclay NPs [134]. Gram-positive bacteria
antimicrobial agent and drug delivery carriers [124–128]. Plate-like
have complex structure which is composed of a three-dimensional thick
montmorillonite (MMT) is the most common nanoclay used in nano-
peptidoglycan layer consisting of linear polysaccharide chains cross
materials applications. Montmorillonite depending on surface mod-
linked by peptides, which makes it difficult for Ag NPs to penetrate onto
ification of the clay layers, could be dispersed in a polymer matrix to
Gram-positive bacteria cell [135]. In contrast, Gram-negative bacteria
form nanocomposite [129,130].
are coated by thin peptidoglycan layer and negatively charged outer
Costa et al. (2011) investigated the antimicrobial activity of Ag/
membrane which facilitates the easy penetration of the nanoparticle
MMT NPs to increasing the shelf life of fresh fruit salad. Results showed
into the surface and inside the cell [135]. Bagchi et al. (2013) examined
that Ag/MMT NPs inhibited the growth of spoilage microorganisms
the antibacterial activity of Cu NPs loaded natural MMT clay based on
(mesophilic and psychrotrophic bacteria, coliforms, lactic acid bacteria,
contact inhibition and ion release. The authors reported that, significant
yeasts and molds) [131]. Su et al. (2011) synthesized a new nano-hy-
antimicrobial activity was observed on E. coli, S. aureus, P. aeruginosa
brids of Ag NPs on Clay platelets for inhibiting Ag-resistant bacteria.

513
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

Table 2
Classification antimicrobial activity of NPs.
Properties of NPs Mechanism of action NPs Target microorganism Reference

Antibacterial Interaction with cell membrane and phosphorus moieties in DNA, resulting in Ag NPs E. coli, [195]
inactivation of DNA replication. Generation of ROS, Reacts with sulfur-containing B. subtilis, [196]
proteins, leading to the inhibition of enzyme functions S. aureus, [197]
Methicillin-resistant coagulase-negative [198]
Staphylococci, [199]
Vancomycin-resistant Enterococcus faecium, [72]
ESBL-positive K. pneumonia, [200]
S. typhi, [201]
Vibrio cholera
Au NPs Methicillin-resistant S. aureus, [202]
Vancomycin-resistant Enterococcus faecium, [203]
E. coli, [204]
P. aeruginosa [205]
[184]
TiO2 NPs E. coli 0157:H7, [206]
S. aureus, [207]
L. monocytogenes [174]
S. enteritidis [172]
P. fluorescens, [208]
[209]
ZnO NPs E. coli 0157:H7, [189]
B. subtilis, [43]
P. fluorescens, [45]
L. monocytogenes, [210]
S. enteritidis, [211]
S. aureus,
S. typhimurium
CuO NPs B. subtilis [85]
L. monocytogenes [212]
S. aureus [117]
E. coli
MgO NPs B. subtilis, [213]
E. coli [214]
S. aureus [188]
B. megaterium [110]
CaO NPs S. aureus [215]
S. epidermidis [216]
E. coli [217]
S. mutans
Al2O3 NPs E. coli [116]
P. aeruginosa, [218]
S. aureus [219]
B. subtilis
K. aerogenes
P. desmolyticum
SiO2 NPs E. coli [220]
S. mutans [221]
B. subtilis
Clay NPs E. coli [136]
Enterococcus faecalis [222]
S. aureus,
P. aeruginosa
(continued on next page)

514
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

Table 2 (continued)

Properties of NPs Mechanism of action NPs Target microorganism Reference

Antifungal Disruption of cell membrane Ag NPs C. spp. [223]


Trichophyton. mentagrophytes [224]
Bipolaris sorokiniana [58]
Magnaporthe grisea
ZnO NPs Botrytis cinerea [44]
P. expansum [225]
A. flavus [226]
A. niger [227]
Saccharomyces cerevisiae [228]
C. albicans [227]
Rhizopus stolonifera
Fusarium oxysporum
TiO2 NPs C. spp. [229]
P. expansum [230]
A. niger spp. [231]
P. oxalicum [232]
CuO NPs A. niger, [233]
Rhizopus oryzae, [234]
A. flavus,
Cladosporium carrionii,
Mucor,
S. cerevisiae
P. notatum
Alternaria alternata
MgO NPs Saccharomyces cerevisiae [227]
C. albicans [235]
A. niger
Rhizopus stolonifer
CaO NPs Saccharomyces cerevisiae [227]
C. albicans [235]
A. niger
Rhizopus stolonifer
Au NPs Puccinia graminis tritci, [99]
A. flavus,
A. niger
C. albicans
SiO2 NPs C. spp. [236]
A. spp. [237]
Dermatophytes spp.
Al2O3 NPs C. spp. [238]
Scenedesmus quadricauda [239]
A. niger
Clay NPs Pycnoporus cinnabarinus [240]
Pleurotus ostreatus [241]
A. niger
C. albicans
Antiviral Blocking of viral attachment to cell surface Au NPs HIV virus, [13]
Influenza virus [74]
[242]
Ag NPs HIV-1, [75]
Influenza virus, [74]
Herpes Simplex virus, [76]
Respiratory syncytial virus, [77]
Monkey pox virus [78]
[79]
ZnO NPs Herpes simplex virus type-1 & 2, [55]
Transmissible gastroenteritis virus (TGEV) [243]
[244]
[245]
[246]
TiO2 NPs Inactivates bacteriophages, [247]
Viruses HSV-1 (Herpes simplex virus), [248]
Influenza virus, [249]
Inactivation of Qβ and T4 bacteriophages, [250]
Zika Virus Mosquito Vectors [251]
[252]
[253]
[254]
[255]
(continued on next page)

515
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

Table 2 (continued)

Properties of NPs Mechanism of action NPs Target microorganism Reference

Antiparasite Inhibition of promastigotes proliferation and metabolic activity, Ionic NPs released, TiO2 NPs Leishmania tropica [256]
Inactivation oocysts, Interaction of NPs with the surface of parasites, NPs impair the Leishmania infantum [31]
structure of lipophosphoglycan and glycoprotein molecules that are found on the Leishmania major [257]
surface of parasites and which are responsible for the infection and disturb DNA. Rhipicephalus (Boophilus) microplus [258]
Generated ROS form NPs and inhibition of parasite infection with oxidative stress, Haemaphysalis bispinosa [259]
coordination effects, and non-homeostasis effects. NPs can diffuse into the cell Anopheles stephensi, [260]
directly through the pores present in cell membrane due to their small size, or they Aedes aegypti [261]
get entry through ion channels and transporter proteins present on the plasma Culex quinquefasciatus [262]
membrane. NPs may enter into cells via endocytosis. Leishmania donovani [263]
Ag NPs Leishmania tropica [264]
Leishmania infantum [265]
Entamoeba histolytica [266]
Cryptosporidium parvum [267]
Giardia lamblia, [268]
Fasciola [269]
Plasmodium, [270]
Toxoplasma gondii [271]
Leishmania major [272]
Plasmodium falciparum (malaria parasite) [273]
larvae of malaria vector, [274]
Anopheles subpictus Grassi, filariasis vector Culex [275]
quinquefasciatus Say (Diptera: Culicidae), [276]
Rhipicephalus (Boophilus) microplus Canestrini [277]
(Acari: Ixodidae) [278]
Haemaphysalis bispinosa [279]
Hippobosca maculata [280]
CuO NPs Entamoeba histolytica [268]
Cryptosporidium parvum [281]
Plasmodium [282]
Hematophagous
Au NPs Giardia lamblia, [283]
Plasmodium, [284]
Toxoplasma, [285]
Leishmania donovani [286]
Leishmania major [287]
Cryptosporidium parvum [267]
Hydatid cyst protoscolices of Echinococcus [288]
granulosus [289]
ZnO NPs Leishmania major, [282]
Plasmodium [290]
[266]
MgO NPs Leishmania major [291]
[292]
Chitosan Giardia lamblia [270]
Curcumin Leishmania [293]
Selenium Leishmania donovani [294]
Toxoplasma gondii [295]
Plasmodium [296]
Echinococcus multilocularis albendazole [297]
Trichinella spiralis [298]
[299]

and Enterococcus faecalis. Cellular membrane damage by direct attach- chelate to metal cations such as Cu-penicillin [143], Zn-b-lactam
ment of the NPs and indirect damage caused by released Cu ion are the [144,145], Cu-aminoglycosides [146,147] and Zn-tetracycline [148]
main reasons of antibacterial action. NPs did not show any adverse and increase or decrease antibiotic activity [149–151]. The interaction
effects and cytotoxicity on the two human cell lines beyond the M.B.C. between antibiotic and metal cations reduce the activity of antibiotics
value for the microorganisms tested [136]. Das et al. (2014) reported and/or limits the bioavailability of metal cations against micro-
that Cu nanoparticle-decorated organically modified montmorillonite/ organism, this procedure may also change membrane penetration and
epoxy nanocomposites significantly inhibited the growth of Gram ne- displays antimicrobial activities [152,153]. Metal cations have synergic
gative (Klebsiella pneumonia) and Gram positive (S. aureus) bacteria effect against few microorganisms. The synergistic effects of NPs cation
[137]. is related to carbapenems, fluoroquinolones, ceftazidime and to-
bramycin against P. aeruginosa, Acinetobacter baumannii and Gram-ne-
3. The role of NPs versus ions release on antimicrobial activities gative pathogens [154,155]. However, regarding NPs, the impact is
related to antimicrobial peptides and Gram-positive and Gram-negative
Stress and environmental factors affect organisms' susceptibility to bacteria particularly P. aeruginosa [156,157]. Recently scientist have
antibiotics by antibiotic-resistant organisms which increase in the ab- discovered Cu-dependent antibiotics, which have Cu-dependent activity
sence of antibiotic reactions [138],environmental adaptation and pro- and are deactivate in Cu deficiency [158–160]. Recently antibiotic re-
tective cellular responses [139,140]. One of the most important causes sistance microorganisms have become of serious concern in public
of environmental stress is metal cations (namely Cu and Zn) of bacterial health [161,162]. Metal oxide NPs systems have shown considerable
cell activity at low concentrations [141,142], since high concentrations antimicrobial activities based on molecular surface. Metal oxide NPs
lead to selective pressure and resistance to antibiotics. Antibiotics with porous structure and active center act as novel solutions against

516
A. Khezerlou et al.

Table 3
Effects of NPs against various parasites.
NPs Parasites Findings Type of study Reference

Ag NPs Fasciola The percent of non-hatching eggs treated with the Triclabendazole drug was ∼70%, while this percent increased to ∼90% In vivo/In [271]
in combination with drug and Ag NPs. vitro
Ag/chitosan/curcumin NPs Giardia lamblia The most fighter effects obtained by combining the three nanomaterial. The parasite found to be eradicated from intestine In vivo [270]
and stool.
Chitosan NPs Trichinella spiralis Despite stimulation lymphocyte response by chitosan, but the effects of treatment was not protective In vitro [300]
Ag NPs Leishmania tropica Ag NPs indicated significant anti-leishmanial activities by inhibiting the proliferation and metabolic activity of In vitro [265]
promastigotes.
Albendazole/chitosan NPs Echinococcus multilocularis Metacestode grown was greatly suppressed during treatment with nanomaterials. In vivo [301]
Chitosan NPs Leishmania infantum Chitosan NPs did not show significant antileishmanial activity against Leishmania infantum. In vitro [302]
CuO nano-hybrid solids Plasmodium falsiparum The NPs showed significant antimalarial activities against the parasites tested. In vitro [303]
Nano-Nitazoxanide Cryptosporidium parvum Nano nitazoxanide was efficiency on parasites during the study (6th). In vivo [304]

517
Au NPs Leishmania major Simultaneous use of Au NPs and MW irradiation was more deadly for promastigotes and amastigotes compared with MW In vitro [288]
alone.
Ag NPs Leishmania major The combined using of direct current electricity and Ag NPs had a cosiderable synergistic effect on promastigote mortality. In vitro [305]
CuO/Ag NPs E. histolytica, C. parvum The combined using CuO NPs and Ag NPs illustrated a remarkable effect in overcoming cryptosporidiosis and amoebiasis. In vitro [268]
Ag/Chitosan NPs Toxoplasma gondii Using Ag NPs singly or combined with chitosan demonstrated promising anti-toxoplasma potentials. In vivo [306]
Ag/Selenium NPs Leishmania major Ag NPs indicated anti-Leishmanial effects but selenium did not have any significant effect In vivo [307]
Chitosan/tripolyphosphate conjugated chloroquine Plasmodium berghei The highest effects of nanoconjugated chloroquine (Nch) observed at 250 mg/kg Bw concentration during the course of In vivo [308]
NPs treatment (∼15th)
Ag NPs Leishmania tropica The IC50 obtained for Ag NPs solutions in high values (∼15 pg/ml). In vitro [309]
Au NPs Giardia lamblia Au NPs at a concentration of ∼0.3 mg/ml was effective for killing Giardia. In vitro [285]
TiO2/Ag2O NPs Leishmania TiO2 and Ag2O NPs displayed significant antiparasite effects. In vitro [31]
Ag NPs Leishmania major Ag NPs alone had no important lethal effects on Leishmania major Promastigotes. In vitro [305]
Curcumin-loaded lipid NPs Plasmodium berghei Antimalarial activity of curcumin increased with entrapping in lipid NPs against malaria. In vivo [310]
Ag NPs Plasmodium falciparum The Ag NPs indicated considerable antiplasmodial activity against P. falciparum. In vitro [311]
Microbial Pathogenesis 123 (2018) 505–526
A. Khezerlou et al.

Table 4
A summary of the in vitro and in vivo toxicity studies NPs.
NPs Assays Doses Cell/tissue or Animal Methods Findings References

TiO2 In vitro 20–100 μg/ml Human lymphocytes Cell viability tested by MTT and trypan blue exclusion assays; ROS levels measured Apoptosis induced by p38/JNK and caspase-8- [327]
using H2 DCFDA; TiO2 measured using AAS; Apoptosis tested by Caspase-8 activity dependent bid pathways; Size- and shape dependent
assay; expression levels of phosphorylated stress-responsive p38 and JNK/SAPK oxidative stress
analyzed by Western blotting
TiO2 In vivo 500 μg per mice Mice Intranasal instillation; TiO2 contents determined in the sub-brain regions by ICP-MS; Rutile had lower adverse effects on the CNS than did [334]
pathological changes observed by optical microscopy and TEM anatase
CuO In vitro 0.01–100 μM Human neuroglioma (H4) Cell viability assay using Hoechst and ethidium homodimer-1 dye Dose-dependent toxicity induced [335]
cells
CuO In vivo 50,000–200,000 μg/kg bw Rats Analyzed serum with automatic analyzer; used optical light microscopy for evaluating Hepato- and nephrotoxicity [336]
histopathology; urine, serum and organs analyzed by NMR spectroscopy
ZnO In vitro 0.001–5 μg/ml Human epidermal cell line Cell viability assayed by MTT and neutral red uptake method; DNA damage assessed Genotoxicity and oxidative stress induced [337]
(A431) using comet assay; oxidative stress assayed using markers (GSH level, catalase
activity, SOD activity)
ZnO In vivo 5 μg/kg bw Mice Gastrointestinal administration; serum assayed by automatic biochemical analyzer; Toxic; severe renal damage occurs in the nanoscale [338]
blood element and blood coagulation tested using automatic hematology analyzer ZnO-treated mice

518
and ELISA; histopathology utilized light microscopy
Ag In vitro 0.7 μg/ml Human hepatoma (HepG2) Evaluated toxicity by MTT, Alamar blue, and LDH assays; monitored oxidative stress Oxidative stress mediated toxicity [313]
cells using DCFH-DA; evaluated expression of metal responsive metallothionein by RT-PCR
Ag In vivo 49–515 μg/m3 Rat Exposed whole body in inhalation chamber; observed mortality, body weight, food Higher dose induces inflammatory response; lungs and [339]
consumption; tested pulmonary function; organs histopathologically examined liver major target tissues
Au In vitro 0.2–2 nM Human cell line, HeLa MTT assay performed; TEM analysis performed for internalization study; gene Nontoxic [340]
expression in stress-related genes analyzed
Au In vivo 0.750 μg/ml Mytilus edulis Determined toxicity by measuring protein ubiquitination and carbonylation; Oxidative stress induced [341]
measured catalase activity, lysosomal membrane stability; gill proteins analyzed by
2D gel electrophoresis
Al In vitro 10–100 μg/ml Type II lung epithelium cell LDH, WST-1, and XTT assayed cytotoxicity; intracellular NP accumulation and Cytotoxic [342]
line (A549) distribution studied using TEM
Al In vivo 0–250 μg/ml Daphnia magna Toxicity evaluated by the Microtox test Toxic [343]
Co In vitro 0.1–0.2 μM Human peripheral blood Assayed Co uptake; cytokinesis-block micronucleus assay; DNA damage detected by Internalized by human leukocytes; Genotoxic [344]
leukocytes (PBLs) Comet assay
Co In vitro 0–5 μg/ml Human U937 monocytes Evaluated cell viability using Celltiter 96® aqueous nonradioactive cell proliferation Toxic; act by increasing the transcription and activities [345]
assay; determined ROS using H2 DCF-DA; analyzed expression and secretion of MMP- of MMP-2 and MMP-9
2 and MMP-9 and their specific tissue inhibitors by RT-PCR
SiO2 In vitro 4–400 μg/cm2 Laryngeal epithelial cells Tested cell viability using alamar blue assay; measured cellular ROS by H 2 DCFDA; Nontoxic, induced reversible ROS generation [346]
Human (hep-2 cells) analyzed SOD, catalase, GR and GPx enzymes activity; detected LPO by competitive
ELISA
Microbial Pathogenesis 123 (2018) 505–526
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

antibiotic resistant microorganisms. The most important factors in an- DNA damage and oxidative stress in the human cell line [320]. The
timicrobial activity are: chemical properties, particle size, shape and reason of toxicity induction is unclear. Despite release of ions from the
zeta potential energetic in ROS [163,164]. According to mentioned NPs, other mechanisms have also been suggested to be responsible for
studies results, the authors suggested that the main mechanism of an- toxicity. The NPs also cause oxidative lesions by significantly increasing
timicrobial activity of clay NPs is disruption of bacterial membrane intracellular ROS levels [321].
integrity by ROS generation, similar to others NPs, and clay shows good Oesterling et al. (2008) reported that Al NPs induces the risk of
effects for use in therapeutic, pharmaceutical and food applications. inflammatory diseases. Exposure to Al NPs increases level of in-
Metal oxide in nano scale demonstrate antibacterial activity greater flammatory markers such as VCAM-1, ICAM-1, and ELAM-1, mRNA
than that of metal oxide in micro scale [165,166]. These NPs must be level and protein expression in human umbilical vein endothelial cells.
stabilized in order to inhibit their interaction with the outside en- Moreover, these NPs increase the adhesion of activated monocytes in
vironment. Nano-sized metal ions are semiconductors, affecting anti- human endothelial cells [322]. Al NPs also induce dose-dependent
microbial activity based on the formation of reactive oxygen species toxicity in cell lines. Exposure to lower doses of Al NPs (5–50 μg/ml),
(ROS) [167–169]. Except ultraviolet (UV) ray, NPs are not energetic in induce little or no toxicity, whereas higher doses (100–250 μg/ml)
ROS [163,164]. produce irregular cell shapes and cell shrinkage. The toxicity produced
As a final statement, several mechanisms have been mentioned in by these NPs is independent of size and results from presence of in-
respect to the antimicrobial activities of NPs. Figs. 1 and 2 shows dif- organic particles [323]. The toxicity produced by exposure to foregoing
ferent antimicrobial mechanisms mentioned for NPs. Fig. 3 display NPs is generally resulted from chemical nature or dissolution products
physicochemical properties of the NPs involved in biological activity: and/or agglomerates.
size, shape, surface area, chemical composition, surface chemistry, and Regarding, NPs have tendency to cause toxicity by different routes,
interactions between NPs and proteins or receptors. Also, Figs. 4 and 5 depending on concentration and size. It has been reported that NPs
illustrate comparison of NPs-induced general toxicity via active inter- cause abnormal sedimentation, hem-agglutination, and hemolysis in
nalization mechanisms and molecular mechanisms of ROS-mediated erythrocytes [324]. Studies have shown that NPs have the tendency to
cell death pathway. The common NPs used as antimicrobial agents increase level of cellular nitric oxide, hydrogen peroxide, induction of
together with their mechanisms of action are summarized in Table 1. oxidative stress and inflammation-related genes and ROS in human cell
Table 2 classifies antimicrobial NPs based on target microorganism and line, and are size- and shape dependent [325]. NPs also interfere with
antibacterial, antiparasite, antifungal and antiviral characteristics. In chromosome segregation, centrosome duplication, cytokinesis, and
addition, Table 3 presents a summary of NP's antiparasite activities. functional regulation of protein PLK1. Short-term exposure to NPs is
Finally, Table 4 demonstrate a summary of the in vitro and in vivo reported to enhance cell survival, cell proliferation, ERK signaling ac-
toxicity the studied NPs. tivation, and ROS production. While, long-term exposure disrupt cell
cycle, duplicated genomes segregation, chromosomal instability, and
4. Cell toxicity of NPs cell transformation in cultured human fibroblast cells [326]. NPs have
also been observed to induce apoptosis via the mitochondrial pathway
NPs are increasingly being used as industrial catalysts. and necrosis in cultured human lymphocytes and in human mono-
Unfortunately, only limited data are available regarding the environ- blastoid cells [327]. The Easy and rapid entry of NPs, has also been
mental or organismal effects of NPs. The large-scale production of NPs suggested as a possible reason for their toxicity [328].
inevitably risks human health, and the environment. It has been sug- Both particle types have been found to agglomerate into micro-
gested that NP's chemical stability significantly effects their cytotoxi- meter-sized particles in cell culture media, and induce toxicity through
city. NPs with oxidizing/reducing or dissolving abilities have the ca- apoptotic pathways [329].
pacity to be toxic in cellular organisms [312]. Therefore, prudence Comparative toxicity studies have also been conducted in order to
suggests that toxicity testing should be performed before releasing and evaluate the relative toxicity of various NPs. NPs such as ZnO have a
using such NP forms. A literature review summary of in vitro and in tendency to induce apoptosis- and necrosis-like cell death in human
vivo toxicity studies regarding NPs is presented in Table 4. astrocytoma cells and in human fibroblasts. Solubility has been found to
Studies reported that NPs induced oxidative stress mediates cyto- play a key role in the cytotoxicity of metal oxide NPs on human fi-
toxicity in human cells. These NPs tend to agglomerate in the cytoplasm broblast cell lines. High soluble NPs exhibit higher toxicity [330].
and nucleus, and cause intracellular oxidative stress. For example, Both However, it has been reported that NPs induce toxicity in cell mem-
Ag NPs, and Ag + ions induce cytotoxicity, but by different mechan- branes [331]. The chemical properties of NPs also influence their cy-
isms [313]. Because these NPs are smaller in size, they interact with totoxicity. At higher doses (100–250 m g ml−1), all NPs exhibit cyto-
cellular genetic material. Unfortunately, little is known about the toxicity which result in irregular cell shape, and cell shrinkage [332].
genotoxicity of NPs. Higher concentrations (50–100 μg/ml) stimulate
cell proliferation, while lower concentrations (1–10 μg/ml) inhibit 5. Conclusion
proliferation, and impair endothelial nitric oxide (NO) synthase
[314–316]. The entry of NPs into cells and their cytotoxicity depend Several studies reported that NPs because of their biological and
both on the type of material absorbed and their relative orientation on physiochemical properties are promising as antimicrobials and ther-
nanoscale surfaces. Surface charge also affects the toxicity of NPs. This apeutic agents. But it must be remembered that they can also possibly
cytotoxic response is concentration dependent. It has been suggested led to adverse biological effects at the cellular levels. Therefore, after
that concentration-dependent electrostatic binding of these NPs on cells the determination non-cytotoxicity and clinical studies the NPs can find
results in cell lysis [317]. NPs penetrate into the human cells by un- vast application as antimicrobials in the consumer and industrial pro-
known mechanisms, different from phagocytosis and endocytosis. Wi- ducts. Application of NPs could be considered as a proper alternative
wanitkit et al. (2009) documented that Au NPs are easily accumulated for many antimicrobial methods. These studies suggest that NPs with
inside white blood cells (WBCs), either via phagocytosis or direct pe- functionalized groups have enhanced antimicrobial activity and may
netration [318]. NPs are used in industrial and healthcare applications. possibly help in eradicating microbial infections. Antimicrobial NPs
NPs are quite small and easily penetrate through the skin by inhalation could be beneficial in the food, pharmaceutical and biomedical appli-
and/or ingestion. They can be transported from nerve endings to the cations. NPs (particularly metal oxide NPs) exhibit significant anti-
skin and reach the somatosensory neurons. Indeed, toxicity is size and microbial effects. Factors such as effectiveness of NPs on resistant
concentration-dependent. High concentrations and smaller sized NPs strains of microbial pathogens as well as heat resistance consider them
provide maximum toxic effects [319]. NPs induced cytotoxicity causes as potent antimicrobial agents. However, application of few NPs is

519
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

limited due to their toxicity at higher concentrations. It has also been with different antibiotics against methicillin-resistant Staphylococcus aureus, J.
proposed that, ion doping and polymer incorporation of these NPs Biomaterials Nanobiotechnol. 1 (2010) 37.
[20] M. Rai, A. Yadav, N. Cioffi, Silver nanoparticles as nano-antimicrobials: bioac-
could be useful for decreasing associated toxicity. Finally, it may be tivity, benefits and bottlenecks, Nano. Antimicrob.: Spring (2012) 211–224.
concluded that, NPs with minimized toxicity could possibly be used in [21] A.L. Linsebigler, G. Lu, J.T. Yates Jr., Photocatalysis on TiO2 surfaces: principles,
the near future for eradicating several microorganism pathogenic and mechanisms, and selected results, Chem. Rev. 95 (1995) 735–758.
[22] A. Dawson, P.V. Kamat, Semiconductor− metal nanocomposites. Photoinduced
infectious conditions. We believe that the development of simple and fusion and photocatalysis of gold-capped TiO2 (TiO2/gold) nanoparticles, J. Phys.
low cost inorganic antimicrobial agents such as NPs as alternatives for Chem. B 105 (2001) 960–966.
traditional antimicrobial agents might be promising for the future of [23] J.C. Yu, L. Zhang, Z. Zheng, J. Zhao, Synthesis and characterization of phosphated
mesoporous titanium dioxide with high photocatalytic activity, Chem. Mater. 15
pharmaceutic, food and medical industry. (2003) 2280–2286.
[24] S.T. Martin, C.L. Morrison, M.R. Hoffmann, Photochemical mechanism of size-
Conflicts of interest quantized vanadium-doped TiO2 particles, J. Phys. Chem. 98 (1994)
13695–13704.
[25] C. Maneerat, Y. Hayata, Antifungal activity of TiO 2 photocatalysis against
The authors declare no conflict of interests. Penicillium expansum in vitro and in fruit tests, Int. J. Food Microbiol. 107 (2006)
99–103.
Acknowledgments [26] H. Bodaghi, Y. Mostofi, A. Oromiehie, Z. Zamani, B. Ghanbarzadeh, C. Costa, et al.,
Evaluation of the photocatalytic antimicrobial effects of a TiO 2 nanocomposite
food packaging film by in vitro and in vivo tests, LWT - Food Sci. Technol. 50
This review was not supported by organizational and conducted at (2013) 702–706.
Tabriz University of Medical Sciences, Tabriz, Iran. [27] M. Gumiero, D. Peressini, A. Pizzariello, A. Sensidoni, L. Iacumin, G. Comi, et al.,
Effect of TiO 2 photocatalytic activity in a HDPE-based food packaging on the
structural and microbiological stability of a short-ripened cheese, Food Chemistry
Appendix A. Supplementary data 138 (2013) 1633–1640.
[28] N. Chorianopoulos, D. Tsoukleris, E. Panagou, P. Falaras, G.-J. Nychas, Use of
titanium dioxide (TiO 2) photocatalysts as alternative means for Listeria mono-
Supplementary data related to this article can be found at https:// cytogenes biofilm disinfection in food processing, Food Microbiol. 28 (2011)
doi.org/10.1016/j.micpath.2018.08.008. 164–170.
[29] M.A. Sani, A. Ehsani, M. Hashemi, Whey protein isolate/cellulose nanofibre/TiO 2
nanoparticle/rosemary essential oil nanocomposite film: its effect on microbial
References and sensory quality of lamb meat and growth of common foodborne pathogenic
bacteria during refrigeration, Int. J. Food Microbiol. 251 (2017) 8–14.
[1] C. Malarkodi, S. Rajeshkumar, K. Paulkumar, M. Vanaja, G. Gnanajobitha, [30] G. Carré, E. Hamon, S. Ennahar, M. Estner, M.-C. Lett, P. Horvatovich, et al., TiO2
G. Annadurai, Biosynthesis and antimicrobial activity of semiconductor nano- photocatalysis damages lipids and proteins in Escherichia coli, Appl. Environ.
particles against oral pathogens, Bioinorgan. Chem. Appl. (2014) 2014. Microbiol. 80 (2014) 2573–2581.
[2] J.T. Seil, T.J. Webster, Antimicrobial applications of nanotechnology: methods [31] A.M. Allahverdiyev, E.S. Abamor, M. Bagirova, M. Rafailovich, Antimicrobial ef-
and literature, Int. J. Nanomed. 7 (2012) 2767. fects of TiO2 and Ag2O nanoparticles against drug-resistant bacteria and leish-
[3] K. Adibkia, M. Alaei-Beirami, M. Barzegar-Jalali, G. Mohammadi, M.S. Ardestani, mania parasites, Future Microbiology 6 (2011) 933–940.
Evaluation and optimization of factors affecting novel diclofenac sodium-eudragit [32] A. Zaleska, Doped-TiO2: a review, Recent Pat. Eng. 2 (2008) 157–164.
RS100 nanoparticles, Afr. J. Pharm. Pharmacol. 6 (2012) 941–947. [33] Y. Xie, Y. He, P.L. Irwin, T. Jin, X. Shi, Antibacterial activity and mechanism of
[4] C. Buzea, Pacheco II, K. Robbie, Nanomaterials and nanoparticles: sources and action of zinc oxide nanoparticles against Campylobacter jejuni, Appl. Environ.
toxicity, Biointerphases 2 (2007) MR17–MR71. Microbiol. 77 (2011) 2325–2331.
[5] K. Adibkia, M. Barzegar-Jalali, A. Nokhodchi, M. Siahi Shadbad, Y. Omidi, [34] N. Padmavathy, R. Vijayaraghavan, Enhanced bioactivity of ZnO nanoparti-
Y. Javadzadeh, et al., A review on the methods of preparation of pharmaceutical cles—an antimicrobial study, Sci. Technol. Adv. Mater. (2008) 9 035004.
nanoparticles, Pharmaceut. Sci. 15 (2009) 303–314. [35] O. Yamamoto, Influence of particle size on the antibacterial activity of zinc oxide,
[6] V. Ravishankar Rai, A. Jamuna Bai, A. Méndez-Vilas A (Ed.), Nanoparticles and Int. J. Inorg. Mater. 3 (2001) 643–646.
Their Potential Application as Antimicrobials, Formatex, Mysore, 2011. [36] A. Azam, A.S. Ahmed, M. Oves, M.S. Khan, S.S. Habib, A. Memic, Antimicrobial
[7] A. Besinis, T. De Peralta, R.D. Handy, The antibacterial effects of silver, titanium activity of metal oxide nanoparticles against Gram-positive and Gram-negative
dioxide and silica dioxide nanoparticles compared to the dental disinfectant bacteria: a comparative study, Int. J. Nanomed. 7 (2012) 6003.
chlorhexidine on Streptococcus mutans using a suite of bioassays, Nanotoxicology [37] L. Zhang, Y. Ding, M. Povey, D. York, ZnO nanofluids–A potential antibacterial
8 (2014) 1–16. agent, Prog. Nat. Sci. 18 (2008) 939–944.
[8] G. Mohammadi, H. Valizadeh, M. Barzegar-Jalali, F. Lotfipour, K. Adibkia, [38] M.R.K. Ravindranadh, T.R. Mary, Development of ZnO nanoparticles for clinical
M. Milani, et al., Development of azithromycin–PLGA nanoparticles: physico- applications. Journal of chemical, J. Chem. Biol. Phys. Sci. 4 (2013) 469.
chemical characterization and antibacterial effect against Salmonella typhi, [39] M.J. Hajipour, K.M. Fromm, A.A. Ashkarran, D.J. de Aberasturi, I.R. de
Colloids Surfaces B Biointerfaces 80 (2010) 34–39. Larramendi, T. Rojo, et al., Antibacterial properties of nanoparticles, Trends
[9] O. Fellahi, R.K. Sarma, M.R. Das, R. Saikia, L. Marcon, Y. Coffinier, et al., The Biotechnol. 30 (2012) 499–511.
antimicrobial effect of silicon nanowires decorated with silver and copper nano- [40] L.-H. Li, M.-Y. Yen, C.-C. Ho, P. Wu, C.-C. Wang, P.K. Maurya, et al., Non-cytotoxic
particles, Nanotechnology 24 (2013) 495101. nanomaterials enhance antimicrobial activities of cefmetazole against multidrug-
[10] G. Mohammadi, A. Nokhodchi, M. Barzegar-Jalali, F. Lotfipour, K. Adibkia, resistant Neisseria gonorrhoeae, PLoS One 8 (2013) e64794.
N. Ehyaei, et al., Physicochemical and anti-bacterial performance characterization [41] A. Atkinson, D.R. Winge, Metal acquisition and availability in the mitochondria,
of clarithromycin nanoparticles as colloidal drug delivery system, Colloids Chem. Rev. 109 (2009) 4708–4721.
Surfaces B Biointerfaces 88 (2011) 39–44. [42] P.J.P. Espitia, NdFF. Soares, R.F. Teófilo, D.M. Vitor, J.S. dos Reis Coimbra, N.J. de
[11] S. Pal, Y.K. Tak, J.M. Song, Does the antibacterial activity of silver nanoparticles Andrade, et al., Optimized dispersion of ZnO nanoparticles and antimicrobial
depend on the shape of the nanoparticle? A study of the gram-negative bacterium activity against foodborne pathogens and spoilage microorganisms, J.
Escherichia coli, Appl. Environ. Microbiol. 73 (2007) 1712–1720. Nanoparticle Res. 15 (2013) 1324.
[12] R. Bera, S. Mandal, C.R. Raj, Antimicrobial activity of fluorescent Ag nano- [43] Y. Liu, L. He, A. Mustapha, H. Li, Z. Hu, M. Lin, Antibacterial activities of zinc
particles, Lett. Appl. Microbiol. 58 (2014) 520–526. oxide nanoparticles against Escherichia coli O157: H7, J. Appl. Microbiol. 107
[13] P. Di Gianvincenzo, M. Marradi, O.M. Martínez-Ávila, L.M. Bedoya, J. Alcamí, (2009) 1193–1201.
S. Penadés, Gold nanoparticles capped with sulfate-ended ligands as anti-HIV [44] L. He, Y. Liu, A. Mustapha, M. Lin, Antifungal activity of zinc oxide nanoparticles
agents, Bioorg. Med. Chem. Lett 20 (2010) 2718–2721. against Botrytis cinerea and Penicillium expansum, Microbiol. Res. 166 (2011)
[14] J.S. Tsuji, A.D. Maynard, P.C. Howard, J.T. James, C-w Lam, D.B. Warheit, et al., 207–215.
Research strategies for safety evaluation of nanomaterials, part IV: risk assessment [45] T. Jin, D. Sun, J. Su, H. Zhang, H.J. Sue, Antimicrobial efficacy of zinc oxide
of nanoparticles, Toxicol. Sci. 89 (2005) 42–50. quantum dots against Listeria monocytogenes, Salmonella enteritidis, and
[15] W.H. De Jong, P.J. Borm, Drug delivery and nanoparticles: applications and ha- Escherichia coli O157: H7, J. Food Sci. (2009) 74.
zards, Int. J. Nanomed. 3 (2008) 133. [46] P. Hosseinkhani, A. Zand, S. Imani, M. Rezayi, S. Rezaei Zarchi, Determining the
[16] U. Zimper, J. Aaltonen, K. Krauel-Goellner, K.C. Gordon, C.J. Strachan, T. Rades, antibacterial effect of ZnO nanoparticle against the pathogenic bacterium, Shigella
The influence of milling on the dissolution performance of simvastatin, dysenteriae (type 1), Int. J. Nano Dimens. (IJND) 1 (2011) 279–285.
Pharmaceutics 2 (2010) 419–431. [47] Z. Emami-Karvani, P. Chehrazi, Antibacterial activity of ZnO nanoparticle on
[17] A. Elsaesser, C.V. Howard, Toxicology of nanoparticles, Adv. Drug Deliv. Rev. 64 gram-positive and gram-negative bacteria, Afr. J. Microbiol. Res. 5 (2011)
(2012) 129–137. 1368–1373.
[18] F. Haghighi, S. Roudbar Mohammadi, P. Mohammadi, S. Hosseinkhani, [48] T. Sun, H. Hao, W-t Hao, S-m Yi, X-p Li, J-r Li, Preparation and antibacterial
R. Shipour, Antifungal activity of TiO2 nanoparticles and EDTA on Candida al- properties of titanium-doped ZnO from different zinc salts, Nanoscale research
bicans biofilms, Infect. Epidemiol. Immunobiol. 1 (2013) 33–38. letters 9 (2014) 98.
[19] A.S. Roy, A. Parveen, A.R. Koppalkar, M.A. Prasad, Effect of nano-titanium dioxide [49] K. Gupta, R. Singh, A. Pandey, A. Pandey, Photocatalytic antibacterial

520
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

performance of TiO2 and Ag-doped TiO2 against S. aureus. P. aeruginosa and E. et al., Interaction of silver nanoparticles with HIV-1, J. Nanobiotechnol. 3
coli, Beilstein Journal of Nanotechnology 4 (2013) 345. (2005) 6.
[50] A. Akbar, A.K. Anal, Zinc oxide nanoparticles loaded active packaging, a challenge [79] J.V. Rogers, C.V. Parkinson, Y.W. Choi, J.L. Speshock, S.M. Hussain, A preliminary
study against Salmonella typhimurium and Staphylococcus aureus in ready-to-eat assessment of silver nanoparticle inhibition of monkeypox virus plaque formation,
poultry meat, Food Contr. 38 (2014) 88–95. Nanoscale Res. Lett. 3 (2008) 129.
[51] J.W. Rasmussen, E. Martinez, P. Louka, D.G. Wingett, Zinc oxide nanoparticles for [80] H.-Q. Wu, X.-W. Wei, M.-W. Shao, J.-S. Gu, M.-Z. Qu, Synthesis of copper oxide
selective destruction of tumor cells and potential for drug delivery applications, nanoparticles using carbon nanotubes as templates, Chem. Phys. Lett. 364 (2002)
Expet Opin. Drug Deliv. 7 (2010) 1063–1077. 152–156.
[52] P.J.P. Espitia, NdFF. Soares, J.S. dos Reis Coimbra, N.J. de Andrade, R.S. Cruz, [81] M.S. Usman, M.E. El Zowalaty, K. Shameli, N. Zainuddin, M. Salama,
E.A.A. Medeiros, Zinc oxide nanoparticles: synthesis, antimicrobial activity and N.A. Ibrahim, Synthesis, characterization, and antimicrobial properties of copper
food packaging applications, Food Bioprocess Technol. 5 (2012) 1447–1464. nanoparticles, Int. J. Nanomed. 8 (2013) 4467.
[53] Z. Zabrieski, E. Morrell, J. Hortin, C. Dimkpa, J. McLean, D. Britt, et al., Pesticidal [82] M. Ahamed, H.A. Alhadlaq, M. Khan, P. Karuppiah, N.A. Al-Dhabi, Synthesis,
activity of metal oxide nanoparticles on plant pathogenic isolates of Pythium, characterization, and antimicrobial activity of copper oxide nanoparticles, J.
Ecotoxicology 24 (2015) 1305–1314. Nanomater. 2014 (2014) 17.
[54] D. Sharma, J. Rajput, B. Kaith, M. Kaur, S. Sharma, Synthesis of ZnO nanoparticles [83] K.-Y. Yoon, J.H. Byeon, J.-H. Park, J. Hwang, Susceptibility constants of
and study of their antibacterial and antifungal properties, Thin Solid Films 519 Escherichia coli and Bacillus subtilis to silver and copper nanoparticles, Sci. Total
(2010) 1224–1229. Environ. 373 (2007) 572–575.
[55] T.E. Antoine, Y.K. Mishra, J. Trigilio, V. Tiwari, R. Adelung, D. Shukla, [84] O. Mahapatra, M. Bhagat, C. Gopalakrishnan, K.D. Arunachalam, Ultrafine dis-
Prophylactic, therapeutic and neutralizing effects of zinc oxide tetrapod structures persed CuO nanoparticles and their antibacterial activity, J. Exp. Nanosci. 3
against herpes simplex virus type-2 infection, Antivir. Res. 96 (2012) 363–375. (2008) 185–193.
[56] S. Zinjarde, Bio-inspired nanomaterials and their applications as antimicrobial [85] G. Ren, D. Hu, E.W. Cheng, M.A. Vargas-Reus, P. Reip, R.P. Allaker,
agents, Chronicles Young Sci. 3 (2012) 74. Characterisation of copper oxide nanoparticles for antimicrobial applications, Int.
[57] S. Egger, R.P. Lehmann, M.J. Height, M.J. Loessner, M. Schuppler, Antimicrobial J. Antimicrob. Agents 33 (2009) 587–590.
properties of a novel silver-silica nanocomposite material, Appl. Environ. [86] U. Bogdanović, V. Lazić, V. Vodnik, M. Budimir, Z. Marković, S. Dimitrijević,
Microbiol. 75 (2009) 2973–2976. Copper nanoparticles with high antimicrobial activity, Mater. Lett. 128 (2014)
[58] Y.-K. Jo, B.H. Kim, G. Jung, Antifungal activity of silver ions and nanoparticles on 75–78.
phytopathogenic fungi, Plant Dis. 93 (2009) 1037–1043. [87] A. Azam, A.S. Ahmed, M. Oves, M. Khan, A. Memic, Size-dependent antimicrobial
[59] C.-N. Lok, C.-M. Ho, R. Chen, Q.-Y. He, W.-Y. Yu, H. Sun, et al., Silver nano- properties of CuO nanoparticles against Gram-positive and-negative bacterial
particles: partial oxidation and antibacterial activities, J. Biol. Inorg. Chem. 12 strains, Int. J. Nanomed. 7 (2012) 3527.
(2007) 527–534. [88] J. Ramyadevi, K. Jeyasubramanian, A. Marikani, G. Rajakumar, A.A. Rahuman,
[60] H. Yun, J.D. Kim, H.C. Choi, C.W. Lee, Antibacterial activity of CNT-Ag and GO-Ag Synthesis and antimicrobial activity of copper nanoparticles, Mater. Lett. 71
nanocomposites against gram-negative and gram-positive bacteria, Bull. Kor. (2012) 114–116.
Chem. Soc. 34 (2013) 3261–3264. [89] E. Lima, R. Guerra, V. Lara, A. Guzmán, Gold nanoparticles as efficient anti-
[61] I. Iavicoli, L. Fontana, V. Leso, A. Bergamaschi, The effects of nanomaterials as microbial agents for Escherichia coli and Salmonella typhi, Chem. Cent. J. 7
endocrine disruptors, Int. J. Mol. Sci. 14 (2013) 16732–16801. (2013) 11.
[62] A.R. Shahverdi, A. Fakhimi, H.R. Shahverdi, S. Minaian, Synthesis and effect of [90] P.M. Tiwari, K. Vig, V.A. Dennis, S.R. Singh, Functionalized gold nanoparticles and
silver nanoparticles on the antibacterial activity of different antibiotics against their biomedical applications, Nanomaterials 1 (2011) 31–63.
Staphylococcus aureus and Escherichia coli, Nanomed. Nanotechnol. Biol. Med. 3 [91] Y. Zhou, Y. Kong, S. Kundu, J.D. Cirillo, H. Liang, Antibacterial activities of gold
(2007) 168–171. and silver nanoparticles against Escherichia coli and bacillus Calmette-Guérin, J.
[63] R. Mie, M.W. Samsudin, L.B. Din, A. Ahmad, N. Ibrahim, S.N.A. Adnan, Synthesis Nanobiotechnol. 10 (2012) 19.
of silver nanoparticles with antibacterial activity using the lichen Parmotrema [92] S. Lokina, V. Narayanan, Antimicrobial and anticancer activity of gold nano-
praesorediosum, Int. J. Nanomed. 9 (2014) 121. particles synthesized from grapes fruit extract, Chem. Sci. Tran. 2 (2013)
[64] O. Choi, K.K. Deng, N.-J. Kim, L. Ross, R.Y. Surampalli, Z. Hu, The inhibitory S105–S110.
effects of silver nanoparticles, silver ions, and silver chloride colloids on microbial [93] Y. Cui, Y. Zhao, Y. Tian, W. Zhang, X. Lü, X. Jiang, The molecular mechanism of
growth, Water Research 42 (2008) 3066–3074. action of bactericidal gold nanoparticles on Escherichia coli, Biomaterials 33
[65] J.F. Hernández-Sierra, F. Ruiz, D.C.C. Pena, F. Martínez-Gutiérrez, A.E. Martínez, (2012) 2327–2333.
AdJP. Guillén, et al., The antimicrobial sensitivity of Streptococcus mutans to [94] M. Sametband, S. Shukla, T. Meningher, S. Hirsh, E. Mendelson, R. Sarid, et al.,
nanoparticles of silver, zinc oxide, and gold. Nanomedicine: Nanotechnology, Biol. Effective multi-strain inhibition of influenza virus by anionic gold nanoparticles,
Med. 4 (2008) 237–240. Med. Chem. Comm. 2 (2011) 421–423.
[66] A. Emamifar, M. Kadivar, M. Shahedi, S. Soleimanian-Zad, Effect of nanocompo- [95] A. Verma, F. Stellacci, Effect of surface properties on nanoparticle–cell interac-
site packaging containing Ag and ZnO on inactivation of Lactobacillus plantarum tions, Small 6 (2010) 12–21.
in orange juice, Food Contr. 22 (2011) 408–413. [96] C.A. Simpson, B.J. Huffman, A.E. Gerdon, D.E. Cliffel, Unexpected toxicity of
[67] S. Birla, V. Tiwari, A. Gade, A. Ingle, A. Yadav, M. Rai, Fabrication of silver na- monolayer protected gold clusters eliminated by PEG-thiol place exchange reac-
noparticles by Phoma glomerata and its combined effect against Escherichia coli, tions, Chem. Res. Toxicol. 23 (2010) 1608–1616.
Pseudomonas aeruginosa and Staphylococcus aureus, Lett. Appl. Microbiol. 48 [97] M. Zawrah, S.A. El-Moez, D. Center, Antimicrobial activities of gold nanoparticles
(2009) 173–179. against major foodborne pathogens, Life Sci. J. 8 (2011) 37–44.
[68] M. Zarei, A. Jamnejad, E. Khajehali, Antibacterial effect of silver nanoparticles [98] S. Kundu, H. Liang, Polyelectrolyte-mediated non-micellar synthesis of mono-
against four foodborne pathogens, Jundishapur J. Microbiol. (2014) 7. dispersed ‘aggregates’ of gold nanoparticles using a microwave approach, Colloid.
[69] S. Rajeshkumar, C. Malarkodi, In vitro antibacterial activity and mechanism of Surface. Physicochem. Eng. Aspect. 330 (2008) 143–150.
silver nanoparticles against foodborne pathogens, Bioinorgan. Chem. Appl. (2014) [99] C. Jayaseelan, R. Ramkumar, A.A. Rahuman, P. Perumal, Green synthesis of gold
2014. nanoparticles using seed aqueous extract of Abelmoschus esculentus and its an-
[70] K. Bahrami, P. Nazari, M. Nabavi, M. Golkar, A. Almasirad, A.R. Shahverdi, tifungal activity, Industrial Crops and Products 45 (2013) 423–429.
Hydroxyl capped silver-gold alloy nanoparticles: characterization and their com- [100] V. Dhapte, S. Kadam, V. Pokharkar, P.K. Khanna, V. Dhapte, Versatile SiO 2 na-
bination effect with different antibiotics against Staphylococcus aureus, J. noparticles@ polymer composites with pragmatic properties, ISRN Inorg. Chem.
Nanomed. 1 (2014) 155–161. (2014) 2014.
[71] M.B. Sathyanarayanan, R. Balachandranath, Y. Genji Srinivasulu, S.K. Kannaiyan, [101] B. Cousins, H. Allison, P. Doherty, C. Edwards, M. Garvey, D. Martin, et al., Effects
G. Subbiahdoss, The effect of gold and iron-oxide nanoparticles on biofilm-forming of a nanoparticulate silica substrate on cell attachment of Candida albicans, J.
pathogens, ISRN Microbiol. (2013) 2013. Appl. Microbiol. 102 (2007) 757–765.
[72] I. Sondi, B. Salopek-Sondi, Silver nanoparticles as antimicrobial agent: a case study [102] Y.H. Kim, D.K. Lee, H.G. Cha, C.W. Kim, Y.C. Kang, Y.S. Kang, Preparation and
on E. coli as a model for Gram-negative bacteria, J. Colloid Interface Sci. 275 characterization of the antibacterial Cu nanoparticle formed on the surface of SiO2
(2004) 177–182. nanoparticles, The Journal of Physical Chemistry B 110 (2006) 24923–24928.
[73] C. Marambio-Jones, E.M. Hoek, A review of the antibacterial effects of silver na- [103] Y.H. Kim, D.K. Lee, H.G. Cha, C.W. Kim, Y.S. Kang, Synthesis and characterization
nomaterials and potential implications for human health and the environment, J. of antibacterial Ag− SiO2 nanocomposite, J. Phys. Chem. C 111 (2007)
Nanoparticle Res. 12 (2010) 1531–1551. 3629–3635.
[74] H.H. Lara, N.V. Ayala-Nuñez, L. Ixtepan-Turrent, C. Rodriguez-Padilla, Mode of [104] I. Mukha, А. Eremenko, G. Korchak, А. Michienkova, Antibacterial action and
antiviral action of silver nanoparticles against HIV-1, J. Nanobiotechnol. 8 physicochemical properties of stabilized silver and gold nanostructures on the
(2010) 1. surface of disperse silica, J. Water Resour. Protect. 2 (2010) 131.
[75] D. Baram-Pinto, S. Shukla, N. Perkas, A. Gedanken, R. Sarid, Inhibition of herpes [105] K. Xu, J.-X. Wang, X.-L. Kang, J.-F. Chen, Fabrication of antibacterial mono-
simplex virus type 1 infection by silver nanoparticles capped with mercaptoethane dispersed Ag–SiO 2 core–shell nanoparticles with high concentration, Mater. Lett.
sulfonate, Bioconjugate Chemistry 20 (2009) 1497–1502. 63 (2009) 31–33.
[76] L. Lu, R. Sun, R. Chen, C.-K. Hui, C.-M. Ho, J.M. Luk, et al., Silver nanoparticles [106] M. Lv, S. Su, Y. He, Q. Huang, W. Hu, D. Li, et al., Long-term antimicrobial effect of
inhibit hepatitis B virus replication, Antivir. Ther. 13 (2008) 253. silicon nanowires decorated with silver nanoparticles, Adv. Mater. 22 (2010)
[77] L. Sun, A.K. Singh, K. Vig, S.R. Pillai, S.R. Singh, Silver nanoparticles inhibit re- 5463–5467.
plication of respiratory syncytial virus, J. Biomed. Nanotechnol. 4 (2008) [107] O. Yamamoto, T. Ohira, K. Alvarez, M. Fukuda, Antibacterial characteristics of
149–158. CaCO 3–MgO composites, Mater. Sci. Eng. B 173 (2010) 208–212.
[78] J.L. Elechiguerra, J.L. Burt, J.R. Morones, A. Camacho-Bragado, X. Gao, H.H. Lara, [108] T. Jin, Y. He, Antibacterial activities of magnesium oxide (MgO) nanoparticles

521
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

against foodborne pathogens, J. Nanoparticle Res. 13 (2011) 6877–6885. aeruginosa, Antimicrob. Agents Chemother. 55 (2011) 1068–1074.
[109] C.J. Hewitt, S.R. Bellara, A. Andreani, G. Nebe-von-Caron, C.M. McFarlane, An [139] K. Poole, Bacterial stress responses as determinants of antimicrobial resistance, J.
evaluation of the anti-bacterial action of ceramic powder slurries using multi- Antimicrob. Chemother. 67 (2012) 2069–2089.
parameter flow cytometry, Biotechnol. Lett. 23 (2001) 667–675. [140] M. Fruci, K. Poole, Bacterial Stress Responses as Determinants of Antimicrobial
[110] Y.H. Leung, A. Ng, X. Xu, Z. Shen, L.A. Gethings, M.T. Wong, et al., Mechanisms of Resistance. Stress and Environmental Regulation of Gene Expression and
antibacterial activity of MgO: non-ROS mediated toxicity of MgO nanoparticles Adaptation in Bacteria, 2 Volume Set, (2016), p. 2.
towards Escherichia coli, Small 10 (2014) 1171–1183. [141] A.W. Foster, D. Osman, N.J. Robinson, Metal preferences and metallation, J. Biol.
[111] J. Vidic, S. Stankic, F. Haque, D. Ciric, R. Le Goffic, A. Vidy, et al., Selective an- Chem. 289 (2014) 28095–28103.
tibacterial effects of mixed ZnMgO nanoparticles, J. Nanoparticle Res. 15 (2013) [142] J.A. Lemire, J.J. Harrison, R.J. Turner, Antimicrobial activity of metals: me-
1595. chanisms, molecular targets and applications, Nat. Rev. Microbiol. 11 (2013)
[112] J. Sawai, H. Kojima, H. Igarashi, A. Hashimoto, S. Shoji, T. Sawaki, et al., 371–384.
Antibacterial characteristics of magnesium oxide powder, World J. Microbiol. [143] W. Cressman, E. Sugita, J. Doluisio, P. Niebergall, Complexation of penicillins and
Biotechnol. 16 (2000) 187–194. penicilloic acids by cupric ion, J. Pharm. Pharmacol. 18 (1966) 801–808.
[113] A.A. Tayel, N.M. Sorour, A.F. El-Baz, F. Wael, Nanometals appraisal in food pre- [144] Y. Guo, D.C. Tsang, X. Zhang, X. Yang, Cu (II)-catalyzed degradation of ampicillin:
servation and food-related activities, Food Preserv.: Elsevier (2017) 487–526. effect of pH and dissolved oxygen, Environ. Sci. Pollut. Control Ser. (2017) 1–10.
[114] I.M. Sadiq, B. Chowdhury, N. Chandrasekaran, A. Mukherjee, Antimicrobial sen- [145] H. Si, J. Hu, Z. Liu, Zl Zeng, Antibacterial effect of oregano essential oil alone and
sitivity of Escherichia coli to alumina nanoparticles, Nanomed. Nanotechnol. Biol. in combination with antibiotics against extended-spectrum β-lactamase-producing
Med. 5 (2009) 282–286. Escherichia coli, Pathogens and Disease 53 (2008) 190–194.
[115] E. Martı́, J. Negrete, G.T. Villaseñor, Structure and properties of Zn–Al–Cu alloy [146] H. Kozłowski, T. Kowalik-Jankowska, M. Jeżowska-Bojczuk, Chemical and biolo-
reinforced with alumina particles, Mater. Des. 24 (2003) 281–286. gical aspects of Cu 2+ interactions with peptides and aminoglycosides, Coord.
[116] B. Li, B.E. Logan, Bacterial adhesion to glass and metal-oxide surfaces, Colloids Chem. Rev. 249 (2005) 2323–2334.
Surfaces B Biointerfaces 36 (2004) 81–90. [147] K. Poole, At the nexus of antibiotics and metals: the impact of Cu and Zn on an-
[117] J.P. Ruparelia, A.K. Chatterjee, S.P. Duttagupta, S. Mukherji, Strain specificity in tibiotic activity and resistance, Trends Microbiol. 25 (10) (2017) 820–832.
antimicrobial activity of silver and copper nanoparticles, Acta Biomaterialia 4 [148] R. Pulicharla, K. Hegde, S.K. Brar, R.Y. Surampalli, Tetracyclines metal com-
(2008) 707–716. plexation: significance and fate of mutual existence in the environment, Environ.
[118] G. Mohammad, V.K. Mishra, H. Pandey, Antioxidant properties of some nano- Pollut. 221 (2016) 1–14.
particle may enhance wound healing in T2DM patient, Digest J. Digest J [149] B. Zawisza, A. Baranik, E. Malicka, E. Talik, R. Sitko, Preconcentration of Fe (III),
Nanomater Digest J Nanomater Biostruct 3 (2008) 159–162. Co (II), Ni (II), Cu (II), Zn (II) and Pb (II) with ethylenediamine-modified graphene
[119] M.A. Ansari, H.M. Khan, A.A. Khan, R. Pal, S.S. Cameotra, Antibacterial potential oxide, Microchimica Acta 183 (2016) 231–240.
of Al2O3 nanoparticles against multidrug resistance strains of [150] A. Zarkan, H.-R. Macklyne, A.W. Truman, A.R. Hesketh, H.-J. Hong, The frontline
Staphylococcusaureus isolated from skin exudates, J. Nanoparticle Res. 15 (2013) antibiotic vancomycin induces a zinc starvation response in bacteria by binding to
1970. Zn (II), Sci. Rep. (2016) 6.
[120] I.M. Sadiq, S. Pakrashi, N. Chandrasekaran, A. Mukherjee, Studies on toxicity of [151] S.K. Mohammed, M.H. Al-Amery, Metal complexes of mixed ligands (quinolone
aluminum oxide (Al2O3) nanoparticles to microalgae species: Scenedesmus sp. antibiotics and α-aminonitrile derivatives) their applications: an update with Mn
and Chlorella sp, J. Nanoparticle Res. 13 (2011) 3287–3299. (II), Cu (II) and Cr (III) ions and study the biological activity, Org. Chem.: Ind. J.
[121] J.C. Mierzwa, V. Arieta, M. Verlage, J. Carvalho, C.D. Vecitis, Effect of clay na- 12 (2016) 29–45.
noparticles on the structure and performance of polyethersulfone ultrafiltration [152] D. Wu, Z. Huang, K. Yang, D. Graham, B. Xie, Relationships between antibiotics
membranes, Desalination 314 (2013) 147–158. and antibiotic resistance gene levels in municipal solid waste leachates in
[122] A. Karkamkar, C. Aardahl, T. Autrey, Advanced applications of engineered na- Shanghai, China, Environ. Sci. Technol. 49 (2015) 4122–4128.
nomaterials, Materials Matters 2 (2007) 10–15. [153] Y. Tan, Y. Guo, X. Gu, C. Gu, Effects of metal cations and fulvic acid on the ad-
[123] J.-W. Rhim, S.-I. Hong, C.-S. Ha, Tensile, water vapor barrier and antimicrobial sorption of ciprofloxacin onto goethite, Environ. Sci. Pollut. Control Ser. 22 (2015)
properties of PLA/nanoclay composite films, LWT - Food Sci. Technol. 42 (2009) 609–617.
612–617. [154] E. Bayroodi, R. Jalal, Modulation of antibiotic resistance in Pseudomonas aeru-
[124] S.G. Jahromi, A. Khodaii, Effects of nanoclay on rheological properties of bitumen ginosa by ZnO nanoparticles, Iran. J. Microbiol. 8 (2016) 85.
binder, Construct. Build. Mater. 23 (2009) 2894–2904. [155] W. Elkhatib, A. Noreddin, In vitro antibiofilm efficacies of different antibiotic
[125] W.H. Awad, G. Beyer, D. Benderly, W.L. Ijdo, P. Songtipya, M. del Mar Jimenez- combinations with zinc sulfate against Pseudomonas aeruginosa recovered from
Gasco, et al., Material properties of nanoclay PVC composites, Polymer 50 (2009) hospitalized patients with urinary tract infection, Antibiotics 3 (2014) 64–84.
1857–1867. [156] W.F. Walkenhorst, Using adjuvants and environmental factors to modulate the
[126] R. Suresh, S. Borkar, V. Sawant, V. Shende, S. Dimble, Nanoclay drug delivery activity of antimicrobial peptides, Biochim. Biophys. Acta Biomembr. 1858 (2016)
system, Int. J. Phar. Sci. Nanotechnol. 3 (2010) 901–905. 926–935.
[127] D.P. de Abreu, P.P. Losada, I. Angulo, J. Cruz, Development of new polyolefin [157] K. Lohner, Membrane-active antimicrobial peptides as template structures for
films with nanoclays for application in food packaging, Eur. Polym. J. 43 (2007) novel antibiotic agents, Curr. Top. Med. Chem. 17 (2017) 508–519.
2229–2243. [158] K.Y. Djoko, M.M. Goytia, P.S. Donnelly, M.A. Schembri, W.M. Shafer,
[128] M.A. Busolo, P. Fernandez, M.J. Ocio, J.M. Lagaron, Novel silver-based nanoclay A.G. McEwan, Copper (II)-bis (thiosemicarbazonato) complexes as antibacterial
as an antimicrobial in polylactic acid food packaging coatings, Food Addit. agents: insights into their mode of action and potential as therapeutics,
Contam. 27 (2010) 1617–1626. Antimicrob. Agents Chemother. 59 (2015) 6444–6453.
[129] K. Shameli, M.B. Ahmad, M. Zargar, W.M.Z.W. Yunus, A. Rustaiyan, N.A. Ibrahim, [159] S. Shah, A.G. Dalecki, A.P. Malalasekera, C.L. Crawford, S.M. Michalek, O. Kutsch,
Synthesis of silver nanoparticles in montmorillonite and their antibacterial beha- et al., 8-Hydroxyquinolines are boosting agents of copper-related toxicity in my-
vior, Int. J. Nanomed. 6 (2011) 581. cobacterium tuberculosis, Antimicrob. Agents Chemother. 60 (2016) 5765–5776.
[130] K. Shameli, M.B. Ahmad, M. Zargar, W.M.Z.W. Yunus, N.A. Ibrahim, [160] A.G. Dalecki, M. Haeili, S. Shah, A. Speer, M. Niederweis, O. Kutsch, et al.,
P. Shabanzadeh, et al., Synthesis and characterization of silver/montmorillonite/ Disulfiram and copper ions kill Mycobacterium tuberculosis in a synergistic
chitosan bionanocomposites by chemical reduction method and their antibacterial manner, Antimicrob. Agents Chemother. 59 (2015) 4835–4844.
activity, Int. J. Nanomed. 6 (2011) 271. [161] S.M. Dizaj, F. Lotfipour, M. Barzegar-Jalali, M.H. Zarrintan, K. Adibkia,
[131] C. Costa, A. Conte, G. Buonocore, M. Del Nobile, Antimicrobial silver-montmor- Antimicrobial activity of the metals and metal oxide nanoparticles, Mater. Sci.
illonite nanoparticles to prolong the shelf life of fresh fruit salad, Int. J. Food Eng. C 44 (2014) 278–284.
Microbiol. 148 (2011) 164–167. [162] P. Mukherjee, Stenotrophomonas and microbacterium: mediated biogenesis of
[132] H.-L. Su, S.-H. Lin, J.-C. Wei, I.-C. Pao, S.-H. Chiao, C.-C. Huang, et al., Novel copper, silver and iron nanoparticles—proteomic insights and antibacterial
nanohybrids of silver particles on clay platelets for inhibiting silver-resistant properties versus biofilm formation, J. Cluster Sci. 28 (2017) 331–358.
bacteria, PLoS One 6 (2011) e21125. [163] P. Bleckenwegner, C.C. Mardare, C. Cobet, J.P. Kollender, A.W. Hassel,
[133] P. Kanmani, J.-W. Rhim, Physical, mechanical and antimicrobial properties of A.I. Mardare, Compositionally dependent nonlinear optical bandgap behavior of
gelatin based active nanocomposite films containing AgNPs and nanoclay, Food mixed anodic oxides in niobium–titanium system, ACS Comb. Sci. 19 (2017)
Hydrocolloids 35 (2014) 644–652. 121–129.
[134] H.M.C. de Azeredo, Antimicrobial activity of nanomaterials for food packaging [164] F. Di Quarto, A. Zaffora, F. Di Franco, M. Santamaria, Critical re-
applications, Nano. Antimicrob.: Spring (2012) 375–394. view—photocurrent spectroscopy in corrosion and passivity studies: a critical
[135] S. Priyadarshini, V. Gopinath, N.M. Priyadharsshini, D. MubarakAli, P. Velusamy, assessment of the use of band gap value to estimate the oxide film composition, J.
Synthesis of anisotropic silver nanoparticles using novel strain, Bacillus flexus and Electrochem. Soc. 164 (2017) C671–C681.
its biomedical application, Colloids Surfaces B Biointerfaces 102 (2013) 232–237. [165] N.A. Rongione, S.A. Floerke, E. Celik, Developments in antibacterial disinfection
[136] B. Bagchi, S. Kar, S.K. Dey, S. Bhandary, D. Roy, T.K. Mukhopadhyay, et al., In situ techniques: applications of nanotechnology, Applying Nanotechnology for
synthesis and antibacterial activity of copper nanoparticle loaded natural mon- Environmental Sustainability: IGI Global (2017) 185–203.
tmorillonite clay based on contact inhibition and ion release, Colloids Surfaces B [166] N. Dasgupta, S. Ranjan, C. Ramalingam, Applications of nanotechnology in agri-
Biointerfaces 108 (2013) 358–365. culture and water quality management, Environ. Chem. Lett. (2017) 1–15.
[137] G. Das, R.D. Kalita, P. Gogoi, A.K. Buragohain, N. Karak, Antibacterial activities of [167] C. Zhang, Z. Hu, B. Deng, Silver nanoparticles in aquatic environments: physio-
copper nanoparticle-decorated organically modified montmorillonite/epoxy na- chemical behavior and antimicrobial mechanisms, Water Research 88 (2016)
nocomposites, Appl. Clay Sci. 90 (2014) 18–26. 403–427.
[138] S. Fraud, K. Poole, Oxidative stress induction of the MexXY multidrug efflux genes [168] E. McGillicuddy, I. Murray, S. Kavanagh, L. Morrison, A. Fogarty, M. Cormican,
and promotion of aminoglycoside resistance development in Pseudomonas et al., Silver nanoparticles in the environment: sources, detection and

522
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

ecotoxicology, Sci. Total Environ. 575 (2017) 231–246. nanocrystalline silver powders and particles, Langmuir 24 (2008) 7457–7464.
[169] F. Vatansever, W.C. de Melo, P. Avci, D. Vecchio, M. Sadasivam, A. Gupta, et al., [198] A. Panáček, L. Kvitek, R. Prucek, M. Kolář, R. Večeřová, N. Pizúrová, et al., Silver
Antimicrobial strategies centered around reactive oxygen species–bactericidal colloid nanoparticles: synthesis, characterization, and their antibacterial activity,
antibiotics, photodynamic therapy, and beyond, FEMS Microbiol. Rev. 37 (2013) J. Phys. Chem. B 110 (2006) 16248–16253.
955–989. [199] A. Nanda, M. Saravanan, Biosynthesis of silver nanoparticles from Staphylococcus
[170] M.C. Sportelli, R.A. Picca, N. Cioffi, Recent advances in the synthesis and char- aureus and its antimicrobial activity against MRSA and MRSE, Nanomed.
acterization of nano-antimicrobials, Trac. Trends Anal. Chem. 84 (2016) 131–138. Nanotechnol. Biol. Med. 5 (2009) 452–456.
[171] Y. Liu, X. Wang, F. Yang, X. Yang, Excellent antimicrobial properties of meso- [200] J.S. Kim, E. Kuk, K.N. Yu, J.-H. Kim, S.J. Park, H.J. Lee, et al., Antimicrobial effects
porous anatase TiO2 and Ag/TiO2 composite films, Microporous Mesoporous of silver nanoparticles, Nanomed. Nanotechnol. Biol. Med. 3 (2007) 95–101.
Mater. 114 (2008) 431–439. [201] V.K. Sharma, R.A. Yngard, Y. Lin, Silver nanoparticles: green synthesis and their
[172] S.H. Othman, A. Salam, N. Raudhah, N. Zainal, R. Kadir Basha, R.A. Talib, antimicrobial activities, Adv. Colloid Interface Sci. 145 (2009) 83–96.
Antimicrobial activity of TiO2 nanoparticle-coated film for potential food packa- [202] J. Gil-Tomás, S. Tubby, I.P. Parkin, N. Narband, L. Dekker, S.P. Nair, et al., Lethal
ging applications, Int. J. Photoenergy (2014) 2014. photosensitisation of Staphylococcus aureus using a toluidine blue
[173] H. Zhang, G. Chen, Potent antibacterial activities of Ag/TiO2 nanocomposite O–tiopronin–gold nanoparticle conjugate, J. Mater. Chem. 17 (2007) 3739–3746.
powders synthesized by a one-pot sol− gel method, Environ. Sci. Technol. 43 [203] S. Perni, C. Piccirillo, J. Pratten, P. Prokopovich, W. Chrzanowski, I.P. Parkin,
(2009) 2905–2910. et al., The antimicrobial properties of light-activated polymers containing me-
[174] C. Chawengkijwanich, Y. Hayata, Development of TiO2 powder-coated food thylene blue and gold nanoparticles, Biomaterials 30 (2009) 89–93.
packaging film and its ability to inactivate Escherichia coli in vitro and in actual [204] H. Gu, P. Ho, E. Tong, L. Wang, B. Xu, Presenting vancomycin on nanoparticles to
tests, Int. J. Food Microbiol. 123 (2008) 288–292. enhance antimicrobial activities, Nano Letters 3 (2003) 1261–1263.
[175] V. Subramanian, E. Wolf, P.V. Kamat, Semiconductor− metal composite nanos- [205] D. MubarakAli, N. Thajuddin, K. Jeganathan, M. Gunasekaran, Plant extract
tructures. To what extent do metal nanoparticles improve the photocatalytic ac- mediated synthesis of silver and gold nanoparticles and its antibacterial activity
tivity of TiO2 films? J. Phys. Chem. B 105 (2001) 11439–11446. against clinically isolated pathogens, Colloids Surfaces B Biointerfaces 85 (2011)
[176] J.C. Yu, W. Ho, J. Lin, H. Yip, P.K. Wong, Photocatalytic activity, antibacterial 360–365.
effect, and photoinduced hydrophilicity of TiO2 films coated on a stainless steel [206] T. Matsunaga, R. Tomoda, T. Nakajima, N. Nakamura, T. Komine, Continuous-
substrate, Environ. Sci. Technol. 37 (2003) 2296–2301. sterilization system that uses photosemiconductor powders, Appl. Environ.
[177] L.S. Reddy, M.M. Nisha, M. Joice, P. Shilpa, Antimicrobial activity of zinc oxide Microbiol. 54 (1988) 1330–1333.
(ZnO) nanoparticle against Klebsiella pneumoniae, Pharmaceut. Biol. 52 (2014) [207] B. Kim, D. Kim, D. Cho, S. Cho, Bactericidal effect of TiO2 photocatalyst on se-
1388–1397. lected food-borne pathogenic bacteria, Chemosphere 52 (2003) 277–281.
[178] R. Saraf, Cost effective and monodispersed zinc oxide nanoparticles synthesis and [208] K. Sunada, Y. Kikuchi, K. Hashimoto, A. Fujishima, Bactericidal and detoxification
their characterization, Int. J. Adv. Appl. Sci. 2 (2013) 85–88. effects of TiO2 thin film photocatalysts, Environ. Sci. Technol. 32 (1998) 726–728.
[179] R. Sinha, R. Karan, A. Sinha, S. Khare, Interaction and nanotoxic effect of ZnO and [209] C. Hu, Y. Lan, J. Qu, X. Hu, A. Wang, Ag/AgBr/TiO2 visible light photocatalyst for
Ag nanoparticles on mesophilic and halophilic bacterial cells, Bioresour. Technol. destruction of azodyes and bacteria, J. Phys. Chem. B 110 (2006) 4066–4072.
102 (2011) 1516–1520. [210] N. Jones, B. Ray, K.T. Ranjit, A.C. Manna, Antibacterial activity of ZnO nano-
[180] K. Chitra, G. Annadurai, Antimicrobial activity of wet chemically engineered particle suspensions on a broad spectrum of microorganisms, FEMS Microbiol.
spherical shaped ZnO nanoparticles on food borne pathogen, Int. Food. Res. J. Lett. 279 (2008) 71–76.
(2013) 20. [211] K. Ghule, A.V. Ghule, B.-J. Chen, Y.-C. Ling, Preparation and characterization of
[181] J.R. Morones, J.L. Elechiguerra, A. Camacho, K. Holt, J.B. Kouri, J.T. Ramírez, ZnO nanoparticles coated paper and its antibacterial activity study, Green Chem. 8
et al., The bactericidal effect of silver nanoparticles, Nanotechnology 16 (2005) (2006) 1034–1041.
2346. [212] N. Cioffi, L. Torsi, N. Ditaranto, G. Tantillo, L. Ghibelli, L. Sabbatini, et al., Copper
[182] A.M. Fayaz, K. Balaji, M. Girilal, R. Yadav, P.T. Kalaichelvan, R. Venketesan, nanoparticle/polymer composites with antifungal and bacteriostatic properties,
Biogenic synthesis of silver nanoparticles and their synergistic effect with anti- Chem. Mater. 17 (2005) 5255–5262.
biotics: a study against gram-positive and gram-negative bacteria. Nanomedicine: [213] R. Richards, W. Li, S. Decker, C. Davidson, O. Koper, V. Zaikovski, et al.,
Nanotechnology, Biol. Med. 6 (2010) 103–109. Consolidation of metal oxide nanocrystals. Reactive pellets with controllable pore
[183] O. Akhavan, E. Ghaderi, Cu and CuO nanoparticles immobilized by silica thin films structure that represent a new family of porous, inorganic materials, J. Am. Chem.
as antibacterial materials and photocatalysts, Surf. Coating. Technol. 205 (2010) Soc. 122 (2000) 4921–4925.
219–223. [214] O.B. Koper, J.S. Klabunde, G.L. Marchin, K.J. Klabunde, P. Stoimenov, L. Bohra,
[184] X. Li, S.M. Robinson, A. Gupta, K. Saha, Z. Jiang, D.F. Moyano, et al., Functional Nanoscale powders and formulations with biocidal activity toward spores and
gold nanoparticles as potent antimicrobial agents against multi-drug-resistant vegetative cells of bacillus species, viruses, and toxins, Curr. Microbiol. 44 (2002)
bacteria, ACS Nano 8 (2014) 10682–10686. 49–55.
[185] D. Pissuwan, C.H. Cortie, S.M. Valenzuela, M.B. Cortie, Functionalised gold na- [215] A. Roy, S.S. Gauri, M. Bhattacharya, J. Bhattacharya, Antimicrobial activity of
noparticles for controlling pathogenic bacteria, Trends Biotechnol. 28 (2010) CaO nanoparticles, J. Biomed. Nanotechnol. 9 (2013) 1570–1578.
207–213. [216] J. Sawai, Quantitative evaluation of antibacterial activities of metallic oxide
[186] K. Xu, J.-X. Wang, X.-L. Kang, J.-F. Chen, Fabrication of antibacterial mono- powders (ZnO, MgO and CaO) by conductimetric assay, J. Microbiol. Meth. 54
dispersed Ag–SiO2 core–shell nanoparticles with high concentration, Mater. Lett. (2003) 177–182.
63 (2009) 31–33. [217] M. Catauro, M. Raucci, F. De Gaetano, A. Marotta, Antibacterial and bioactive
[187] O. Yamamoto, T. Ohira, K. Alvarez, M. Fukuda, Antibacterial characteristics of silver-containing Na 2 O· CaO· 2SiO 2 glass prepared by sol–gel method, J. Mater.
CaCO3–MgO composites, Mater. Sci. Eng. B 173 (2010) 208–212. Sci. Mater. Med. 15 (2004) 831–837.
[188] K. Krishnamoorthy, G. Manivannan, S.J. Kim, K. Jeyasubramanian, [218] P. Prashanth, R. Raveendra, R.H. Krishna, S. Ananda, N. Bhagya,
M. Premanathan, Antibacterial activity of MgO nanoparticles based on lipid per- B. Nagabhushana, et al., Synthesis, characterizations, antibacterial and photo-
oxidation by oxygen vacancy, J. Nanoparticle Res. 14 (2012) 1063. luminescence studies of solution combustion-derived α-Al2O3 nanoparticles, J.
[189] W. Jiang, H. Mashayekhi, B. Xing, Bacterial toxicity comparison between nano- Asian. Cera. Soc. 3 (2015) 345–351.
and micro-scaled oxide particles, Environ. Pollut. 157 (2009) 1619–1625. [219] A. Jastrzębska, E. Radziun, M. Rosłon, A. Kunicki, A. Olszyna, J. Dudkiewicz-
[190] A. Simon-Deckers, S. Loo, M. Mayne-L’hermite, N. Herlin-Boime, N. Menguy, Wilczyńska, et al., In vitro assessment of antibacterial properties and cytotoxicity
C. Reynaud, et al., Size-, composition-and shape-dependent toxicological impact of of Al2O3–Ag nanopowders, Adv. Appl. Cera. 110 (2011) 353–359.
metal oxide nanoparticles and carbon nanotubes toward bacteria, Environ. Sci. [220] N. Silvestry-Rodriguez, E.E. Sicairos-Ruelas, C.P. Gerba, K.R. Bright, Silver as a
Technol. 43 (2009) 8423–8429. Disinfectant. Reviews of Environmental Contamination and Toxicology: Springer,
[191] M. Ansari, H. Khan, A. Khan, S.S. Cameotra, Q. Saquib, J. Musarrat, Interaction of (2007), pp. 23–45.
Al2O3 nanoparticles with Escherichia coli and their cell envelope biomolecules, J. [221] L.K. Adams, D.Y. Lyon, P.J. Alvarez, Comparative eco-toxicity of nanoscale TiO2,
Appl. Microbiol. 116 (2014) 772–783. SiO2, and ZnO water suspensions, Water Research 40 (2006) 3527–3532.
[192] M.A. Ansari, H.M. Khan, M.A. Alzohairy, M. Jalal, S.G. Ali, R. Pal, et al., Green [222] S.E. Haydel, C.M. Remenih, L.B. Williams, Broad-spectrum in vitro antibacterial
synthesis of Al2O3 nanoparticles and their bactericidal potential against clinical activities of clay minerals against antibiotic-susceptible and antibiotic-resistant
isolates of multi-drug resistant Pseudomonas aeruginosa, World J. Microbiol. bacterial pathogens, J. Antimicrob. Chemother. 61 (2007) 353–361.
Biotechnol. 31 (2015) 153–164. [223] A. Panáček, M. Kolář, R. Večeřová, R. Prucek, J. Soukupová, V. Kryštof, et al.,
[193] S.C. Motshekga, S.S. Ray, M.S. Onyango, M.N. Momba, Microwave-assisted Antifungal activity of silver nanoparticles against Candida spp, Biomaterials 30
synthesis, characterization and antibacterial activity of Ag/ZnO nanoparticles (2009) 6333–6340.
supported bentonite clay, J. Hazard Mater. 262 (2013) 439–446. [224] K.-J. Kim, W.S. Sung, S.-K. Moon, J.-S. Choi, J.G. Kim, D.G. Lee, Antifungal effect
[194] S. Magana, P. Quintana, D. Aguilar, J. Toledo, C. Angeles-Chavez, M. Cortes, et al., of silver nanoparticles on dermatophytes, J. Microbiol. Biotechnol. 18 (2008)
Antibacterial activity of montmorillonites modified with silver, J. Mol. Catal. 1482–1484.
Chem. 281 (2008) 192–199. [225] R.S. Yehia, O.F. Ahmed, In vitro study of the antifungal efficacy of zinc oxide
[195] Q.L. Feng, J. Wu, G. Chen, F. Cui, T. Kim, J. Kim, A mechanistic study of the nanoparticles against Fusarium oxysporum and Penicilium expansum, Afr. J.
antibacterial effect of silver ions on Escherichia coli and Staphylococcus aureus, J. Microbiol. Res. 7 (2013) 1917–1923.
Biomed. Mater. Res. 52 (2000) 662–668. [226] P. Rajiv, S. Rajeshwari, R. Venckatesh, Bio-Fabrication of zinc oxide nanoparticles
[196] W.K. Jung, H.C. Koo, K.W. Kim, S. Shin, S.H. Kim, Y.H. Park, Antibacterial activity using leaf extract of Parthenium hysterophorus L. and its size-dependent anti-
and mechanism of action of the silver ion in Staphylococcus aureus and fungal activity against plant fungal pathogens, Spectrochim. Acta Mol. Biomol.
Escherichia coli, Appl. Environ. Microbiol. 74 (2008) 2171–2178. Spectrosc. 112 (2013) 384–387.
[197] A.B. Smetana, K.J. Klabunde, G.R. Marchin, C.M. Sorensen, Biocidal activity of [227] J. Sawai, T. Yoshikawa, Quantitative evaluation of antifungal activity of metallic

523
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

oxide powders (MgO, CaO and ZnO) by an indirect conductimetric assay, J. Appl. Mosq. Contr. Assoc. 28 (2012) 123–126.
Microbiol. 96 (2004) 803–809. [255] K. Murugan, A. Jaganathan, R. Rajaganesh, U. Suresh, J. Madhavan, S. Senthil-
[228] A. Lipovsky, Y. Nitzan, A. Gedanken, R. Lubart, Antifungal activity of ZnO na- Nathan, et al., Poly (styrene sulfonate)/poly (allylamine hydrochloride) en-
noparticles—the role of ROS mediated cell injury, Nanotechnology 22 (2011) capsulation of TiO2 nanoparticles boosts their toxic and repellent activity against
105101. zika virus mosquito vectors, J. Cluster Sci. 29 (2018) 27–39.
[229] G. Rajakumar, A.A. Rahuman, S.M. Roopan, V.G. Khanna, G. Elango, C. Kamaraj, [256] A.M. Allahverdiyev, E.S. Abamor, M. Bagirova, S.Y. Baydar, S.C. Ates, F. Kaya,
et al., Fungus-mediated biosynthesis and characterization of TiO2 nanoparticles et al., Investigation of antileishmanial activities of Tio2@ Ag nanoparticles on
and their activity against pathogenic bacteria, Spectrochim. Acta Mol. Biomol. biological properties of L. tropica and L. infantum parasites, in vitro, Exp.
Spectrosc. 91 (2012) 23–29. Parasitol. 135 (2013) 55–63.
[230] N. Gómez-Ortíz, S. De la Rosa-García, W. González-Gómez, M. Soria-Castro, [257] E.S. Abamor, A.M. Allahverdiyev, A nanotechnology based new approach for
P. Quintana, G. Oskam, et al., Antifungal coatings based on Ca (OH) 2 mixed with chemotherapy of cutaneous leishmaniasis: TIO2@ AG nanoparticles–Nigella sativa
ZnO/TiO2 nanomaterials for protection of limestone monuments, ACS Appl. oil combinations, Exp. Parasitol. 166 (2016) 150–163.
Mater. Interfaces 5 (2013) 1556–1565. [258] E.S. Abamor, A.M. Allahverdiyev, M. Bagirova, M. Rafailovich, Meglumine
[231] A. Herman, A.P. Herman, Nanoparticles as antimicrobial agents: their toxicity and antımoniate-TiO2@ Ag nanoparticle combinations reduce toxicity of the drug
mechanisms of action, J. Nanosci. Nanotechnol. 14 (2014) 946–957. while enhancing its antileishmanial effect, Acta Tropica 169 (2017) 30–42.
[232] P.A. Zapata, H. Palza, K. Delgado, F.M. Rabagliati, Novel antimicrobial poly- [259] S. Marimuthu, A.A. Rahuman, C. Jayaseelan, A.V. Kirthi, T. Santhoshkumar,
ethylene composites prepared by metallocenic in situ polymerization with TiO2- K. Velayutham, et al., Acaricidal activity of synthesized titanium dioxide nano-
based nanoparticles, J. Polym. Sci. Polym. Chem. 50 (2012) 4055–4062. particles using Calotropis gigantea against Rhipicephalus microplus and
[233] K. Mageshwari, R. Sathyamoorthy, Flower-shaped CuO nanostructures: synthesis, Haemaphysalis bispinosa, Asian Pac. J. Trop. Med. 6 (2013) 682–688.
characterization and antimicrobial activity, J. Mater. Sci. Technol. 29 (2013) [260] P.R. Gandhi, C. Jayaseelan, E. Vimalkumar, R.R. Mary, Larvicidal and pediculi-
909–914. cidal activity of synthesized TiO2 nanoparticles using Vitex negundo leaf extract
[234] K. Giannousi, G. Sarafidis, S. Mourdikoudis, A. Pantazaki, C. Dendrinou-Samara, against blood feeding parasites, J. Asia Pac. Entomol. 19 (2016) 1089–1094.
Selective synthesis of Cu2O and Cu/Cu2O NPs: antifungal activity to yeast sac- [261] T.Y. Suman, R.R.S. Ravindranath, D. Elumalai, P.K. Kaleena, R. Ramkumar,
charomyces cerevisiae and DNA interaction, Inorg. Chem. 53 (2014) 9657–9666. P. Perumal, et al., Larvicidal activity of titanium dioxide nanoparticles synthesized
[235] J. Sawai, T. Yoshikawa, Measurement of fungi by an indirect conductimetric assay, using Morinda citrifolia root extract against Anopheles stephensi, Aedes aegypti
Lett. Appl. Microbiol. 37 (2003) 40–44. and Culex quinquefasciatus and its other effect on non-target fish, Asia pac J. Trop.
[236] J.A. Gutierrez, S.J. Caballero, L.A. Díaz, M.A. Guerrero, J. Ruiz, C.C. Ortiz, High Med. 5 (2015) 224–230.
antifungal activity against Candida species of monometallic and bimetallic nano- [262] G. Rajakumar, A.A. Rahuman, C. Jayaseelan, T. Santhoshkumar, S. Marimuthu,
particles synthesized in nanoreactors, ACS Biomater. Sci. Eng. 4 (2) (2017) C. Kamaraj, et al., Solanum trilobatum extract-mediated synthesis of titanium
647–653. dioxide nanoparticles to control Pediculus humanus capitis, Hyalomma anato-
[237] L. Zamani, B.B.F. Mirjalili, K. Zomorodian, M. Namazian, S. Khabnadideh, licum anatolicum and Anopheles subpictus, Parasitol. Res. 113 (2014) 469–479.
E.F. Mirzaei, Synthesis of benzimidazoles in the presence of nano-TiCl4. SiO2 as [263] A.A. Zahir, I.S. Chauhan, A. Bagavan, C. Kamaraj, G. Elango, J. Shankar, et al.,
antifungal agents and tautomerism theoretical study of some products, Synthesis Green synthesis of silver and titanium dioxide nanoparticles using Euphorbia
62 (2014) 3. prostrata extract shows shift from apoptosis to G0/G1 arrest followed by necrotic
[238] M. Jalal, M.A. Ansari, A.K. Shukla, S.G. Ali, H.M. Khan, R. Pal, et al., Green cell death in Leishmania donovani, Antimicrob. Agents Chemother. 59 (2015)
synthesis and antifungal activity of Al 2 O 3 NPs against fluconazole-resistant 4782–4799.
Candida spp isolated from a tertiary care hospital, RSC Adv. 6 (2016) [264] P. Baiocco, A. Ilari, P. Ceci, S. Orsini, M. Gramiccia, T. Di Muccio, et al., Inhibitory
107577–107590. effect of silver nanoparticles on trypanothione reductase activity and Leishmania
[239] N. Doskocz, K. Affek, M. Załęska-Radziwiłł, Effects of Aluminium Oxide infantum proliferation, ACS Med. Chem. Lett. 2 (2010) 230–233.
Nanoparticles on Bacterial Growth. E3S Web of Conferences, EDP Sciences, 2017, [265] A.M. Allahverdiyev, E.S. Abamor, M. Bagirova, C.B. Ustundag, C. Kaya, F. Kaya,
p. 00019. et al., Antileishmanial effect of silver nanoparticles and their enhanced anti-
[240] Y. Ling, Y. Luo, J. Luo, X. Wang, R. Sun, Novel antibacterial paper based on parasitic activity under ultraviolet light, Int. J. Nanomed. 6 (2011) 2705.
quaternized carboxymethyl chitosan/organic montmorillonite/Ag NP nano- [266] A. Nadhman, S. Nazir, M.I. Khan, S. Arooj, M. Bakhtiar, G. Shahnaz, et al.,
composites, Ind. Crop. Prod. 51 (2013) 470–479. PEGylated silver doped zinc oxide nanoparticles as novel photosensitizers for
[241] K. Malachová, P. Praus, Z. Rybková, O. Kozák, Antibacterial and antifungal ac- photodynamic therapy against Leishmania, Free Radic. Biol. Med. 77 (2014)
tivities of silver, copper and zinc montmorillonites, Appl. Clay Sci. 53 (2011) 230–238.
642–645. [267] A. Ahmad, F. Syed, A. Shah, Z. Khan, K. Tahir, A.U. Khan, et al., Silver and gold
[242] M.-C. Bowman, T.E. Ballard, C.J. Ackerson, D.L. Feldheim, D.M. Margolis, nanoparticles from Sargentodoxa cuneata: synthesis, characterization and antil-
C. Melander, Inhibition of HIV fusion with multivalent gold nanoparticles, J. Am. eishmanial activity, RSC Adv. 5 (2015) 73793–73806.
Chem. Soc. 130 (2008) 6896–6897. [268] H. Saad, M.I. Soliman, A.M. Azzam, B. Mostafa, Antiparasitic activity of silver and
[243] W. Chai, S.S. Zakrzewski, D. Günzel, R. Pieper, Z. Wang, S. Twardziok, et al., High- copper oxide nanoparticles against Entamoeba histolytica and Cryptosporidium
dose dietary zinc oxide mitigates infection with transmissible gastroenteritis virus parvum cysts, J. Egypt. Soc. Parasitol. 45 (2015) 593–602.
in piglets, BMC Vet. Res. 10 (2014) 75. [269] M. Rai, K. Kon, A. Ingle, N. Duran, S. Galdiero, M. Galdiero, Broad-spectrum
[244] Z. Wei, M. Burwinkel, C. Palissa, E. Ephraim, M.F. Schmidt, Antiviral activity of bioactivities of silver nanoparticles: the emerging trends and future prospects,
zinc salts against transmissible gastroenteritis virus in vitro, Vet. Microbiol. 160 Appl. Microbiol. Biotechnol. 98 (2014) 1951–1961.
(2012) 468–472. [270] D. Said, L. Elsamad, Y. Gohar, Validity of silver, chitosan, and curcumin nano-
[245] Y.K. Mishra, R. Adelung, C. Röhl, D. Shukla, F. Spors, V. Tiwari, Virostatic po- particles as anti-Giardia agents, Parasitol. Res. 111 (2012) 545–554.
tential of micro–nano filopodia-like ZnO structures against herpes simplex virus-1, [271] Y.A. Gherbawy, I.M. Shalaby, M.S.A. El-sadek, H.M. Elhariry, A.A. Banaja, The
Antivir. Res. 92 (2011) 305–312. anti-fasciolasis properties of silver nanoparticles produced by Trichoderma har-
[246] J.-H. Han, D. Lee, C.H.C. Chew, T. Kim, J.J. Pak, A multi-virus detectable mi- zianum and their improvement of the anti-fasciolasis drug triclabendazole, Int. J.
crofluidic electrochemical immunosensor for simultaneous detection of H1N1, Mol. Sci. 14 (2013) 21887–21898.
H5N1, and H7N9 virus using ZnO nanorods for sensitivity enhancement, Sensor. [272] K. Murugan, C.M. Samidoss, C. Panneerselvam, A. Higuchi, M. Roni, U. Suresh,
Actuator. B Chem. 228 (2016) 36–42. et al., Seaweed-synthesized silver nanoparticles: an eco-friendly tool in the fight
[247] V.I. Syngouna, C.V. Chrysikopoulos, Inactivation of MS2 bacteriophage by tita- against Plasmodium falciparum and its vector Anopheles stephensi? Parasitol. Res.
nium dioxide nanoparticles in the presence of quartz sand with and without am- 114 (2015) 4087–4097.
bient light, J. Colloid Interface Sci. 497 (2017) 117–125. [273] I. Dimier-Poisson, R. Carpentier, T.T.L. N'Guyen, F. Dahmani, C. Ducournau,
[248] P. Hajkova, P. Spatenka, J. Horsky, I. Horska, A. Kolouch, Photocatalytic effect of D. Betbeder, Porous nanoparticles as delivery system of complex antigens for an
TiO2 films on viruses and bacteria, Plasma Process. Polym. (2007) 4. effective vaccine against acute and chronic Toxoplasma gondii infection,
[249] R. Nakano, H. Ishiguro, Y. Yao, J. Kajioka, A. Fujishima, K. Sunada, et al., Biomaterials 50 (2015) 164–175.
Photocatalytic inactivation of influenza virus by titanium dioxide thin film, [274] S. Jiang, E. Hua, M. Liang, B. Liu, G. Xie, A novel immunosensor for detecting
Photochem. Photobiol. Sci. 11 (2012) 1293–1298. toxoplasma gondii-specific IgM based on goldmag nanoparticles and graphene
[250] H. Cui, J. Jiang, W. Gu, C. Sun, D. Wu, T. Yang, et al., Photocatalytic inactivation sheets, Colloids Surfaces B Biointerfaces 101 (2013) 481–486.
efficiency of anatase nano-TiO2 sol on the H9N2 avian influenza virus, [275] A. Jaganathan, K. Murugan, C. Panneerselvam, P. Madhiyazhagan, D. Dinesh,
Photochem. Photobiol. 86 (2010) 1135–1139. C. Vadivalagan, et al., Earthworm-mediated synthesis of silver nanoparticles: a
[251] H. Ishiguro, Y. Yao, R. Nakano, M. Hara, K. Sunada, K. Hashimoto, et al., potent tool against hepatocellular carcinoma, Plasmodium falciparum parasites
Photocatalytic activity of Cu2+/TiO2-coated cordierite foam inactivates bacter- and malaria mosquitoes, Parasitol. Int. 65 (2016) 276–284.
iophages and Legionella pneumophila, Appl. Catal. B Environ. 129 (2013) 56–61. [276] U. Muthukumaran, M. Govindarajan, M. Rajeswary, Mosquito larvicidal potential
[252] H. Ishiguro, R. Nakano, Y. Yao, J. Kajioka, A. Fujishima, K. Sunada, et al., of silver nanoparticles synthesized using Chomelia asiatica (Rubiaceae) against
Photocatalytic inactivation of bacteriophages by TiO 2-coated glass plates under Anopheles stephensi, Aedes aegypti, and Culex quinquefasciatus (Diptera:
low-intensity, long-wavelength UV irradiation, Photochem. Photobiol. Sci. 10 Culicidae), Parasitol. Res. 114 (2015) 989–999.
(2011) 1825–1829. [277] K. Veerakumar, M. Govindarajan, M. Rajeswary, U. Muthukumaran, Mosquito
[253] I.B. Ditta, A. Steele, C. Liptrot, J. Tobin, H. Tyler, H.M. Yates, et al., Photocatalytic larvicidal properties of silver nanoparticles synthesized using Heliotropium in-
antimicrobial activity of thin surface films of TiO 2, CuO and TiO 2/CuO dual dicum (Boraginaceae) against Aedes aegypti, Anopheles stephensi, and Culex
layers on Escherichia coli and bacteriophage T4, Appl. Microbiol. Biotechnol. 79 quinquefasciatus (Diptera: Culicidae), Parasitol. Res. 113 (2014) 2363–2373.
(2008) 127. [278] S. Subarani, S. Sabhanayakam, C. Kamaraj, Studies on the impact of biosynthe-
[254] M.J. Turell, Members of the Culex pipiens complex as vectors of viruses, J. Am. sized silver nanoparticles (AgNPs) in relation to malaria and filariasis vector

524
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

control against Anopheles stephensi Liston and Culex quinquefasciatus Say [304] F. Sedighi, R.A. Pourkabir, A. Maghsood, M. Fallah, Comparison of therapeutic
(Diptera: Culicidae), Parasitol. Res. 112 (2013) 487–499. effect of anti-cryptosporidium nano-nitazoxanide (NTZ) with free form of this drug
[279] G. Rajakumar, A.A. Rahuman, Acaricidal activity of aqueous extract and synthe- in neonatal rat, Sci. J. Hamad. Univ. Med. Sci. 23 (2016) 134–140.
sized silver nanoparticles from Manilkara zapota against Rhipicephalus [305] M. Karimi, A. Dalimi, F. Jamei, F. Ghaffarifar, A. Dalimi, The killing effect of silver
(Boophilus) microplus, Res. Vet. Sci. 93 (2012) 303–309. nanoparticles and direct electric current induction on Leishmania major promas-
[280] A.A. Zahir, A.A. Rahuman, Evaluation of different extracts and synthesised silver tigotes in vitro, Pathobiol. Res. 18 (3) (2015) 87–96.
nanoparticles from leaves of Euphorbia prostrata against Haemaphysalis bispinosa [306] M. Gaafar, R. Mady, R. Diab, T.I. Shalaby, Chitosan and silver nanoparticles:
and Hippobosca maculata, Vet. Parasitol. 187 (2012) 511–520. promising anti-toxoplasma agents, Exp. Parasitol. 143 (2014) 30–38.
[281] J. Ramyadevi, K. Jeyasubramanian, A. Marikani, G. Rajakumar, A.A. Rahuman, [307] F. Jameii, A.D. Asl, M. Karimi, F. Ghaffarifar, Healing effect comparison of sele-
T. Santhoshkumar, et al., Copper nanoparticles synthesized by polyol process used nium and silver nanoparticles on skin leishmanial lesions in mice, Sci. J. Hamad.
to control hematophagous parasites, Parasitol. Res. 109 (2011) 1403–1415. Univ. Med. Sci. 22 (2015) 217–223.
[282] K.B. Paul, S. Kumar, S. Tripathy, S.R.K. Vanjari, V. Singh, S.G. Singh, A highly [308] S. Tripathy, S. Das, S.P. Chakraborty, S.K. Sahu, P. Pramanik, S. Roy, Synthesis,
sensitive self assembled monolayer modified copper doped zinc oxide nanofiber characterization of chitosan–tripolyphosphate conjugated chloroquine nano-
interface for detection of Plasmodium falciparum histidine-rich protein-2: targeted particle and its in vivo anti-malarial efficacy against rodent parasite: a dose and
towards rapid, early diagnosis of malaria, Biosens. Bioelectron. 80 (2016) 39–46. duration dependent approach, Int. J. Pharm. 434 (2012) 292–305.
[283] K.L. Rule, P.J. Vikesland, Surface-enhanced resonance Raman spectroscopy for the [309] A. Khosravi, I. Sharifi, M. Barati, M. Zarean, M. Hakimi-Parizi, Anti-leishmanial
rapid detection of Cryptosporidium parvum and Giardia lamblia, Environ. Sci. effect of nanosilver solutions on Leishmania tropica promastigotes by in-vitro
Technol. 43 (2009) 1147–1152. assay, Zehed. J. Res. Med. Sci. 13 (2011) 8–12.
[284] D.J. Javier, A. Castellanos-Gonzalez, S.E. Weigum, A.C. White, R. Richards- [310] A.P. Nayak, W. Tiyaboonchai, S. Patankar, B. Madhusudhan, E.B. Souto,
Kortum, Oligonucleotide-gold nanoparticle networks for detection of Curcuminoids-loaded lipid nanoparticles: novel approach towards malaria treat-
Cryptosporidium parvum heat shock protein 70 mRNA, J. Clin. Microbiol. 47 ment, Colloids Surfaces B Biointerfaces 81 (2010) 263–273.
(2009) 4060–4066. [311] S. Ponarulselvam, C. Panneerselvam, K. Murugan, N. Aarthi, K. Kalimuthu,
[285] Z. Bavand, S. Gholami, S. Honary, E.B. RAHIMI, N. Torabi, H. Barabadi, In Vitro S. Thangamani, Synthesis of silver nanoparticles using leaves of Catharanthus
Evaluation of the Effect of Gold Nanoparticles on Giardia Lamblia Cyst, (2014). roseus Linn. G. Don and their antiplasmodial activities, Asia. Pac. J. Trop. Biomed.
[286] J. Subramaniam, K. Murugan, C. Panneerselvam, K. Kovendan, P. Madhiyazhagan, 2 (2012) 574–580.
D. Dinesh, et al., Multipurpose effectiveness of Couroupita guianensis-synthesized [312] M. Auffan, J. Rose, M.R. Wiesner, J.-Y. Bottero, Chemical stability of metallic
gold nanoparticles: high antiplasmodial potential, field efficacy against malaria nanoparticles: a parameter controlling their potential cellular toxicity in vitro,
vectors and synergy with Aplocheilus lineatus predators, Environ. Sci. Pollut. Environ. Pollut. 157 (2009) 1127–1133.
Control Ser. 23 (2016) 7543–7558. [313] S. Kim, J.E. Choi, J. Choi, K.-H. Chung, K. Park, J. Yi, et al., Oxidative stress-
[287] H. Wang, C. Lei, J. Li, Z. Wu, G. Shen, R. Yu, A piezoelectric immunoagglutination dependent toxicity of silver nanoparticles in human hepatoma cells, Toxicol. Vitro
assay for Toxoplasma gondii antibodies using gold nanoparticles, Biosens. 23 (2009) 1076–1084.
Bioelectron. 19 (2004) 701–709. [314] M. Safaepour, A.R. Shahverdi, H.R. Shahverdi, M.R. Khorramizadeh, A.R. Gohari,
[288] A. Sazgarnia, A.R. Taheri, S. Soudmand, A.J. Parizi, O. Rajabi, M.S. Darbandi, Green synthesis of small silver nanoparticles using geraniol and its cytotoxicity
Antiparasitic effects of gold nanoparticles with microwave radiation on promas- against fibrosarcoma-wehi 164, Avicenna Journal of Medical Biotechnology 1
tigotes and amastigotes of Leishmania major, Int. J. Hyperther. 29 (2013) 79–86. (2009) 111.
[289] H. Barabadi, S. Honary, M.A. Mohammadi, E. Ahmadpour, M.T. Rahimi, [315] R.Y. Parikh, S. Singh, B. Prasad, M.S. Patole, M. Sastry, Y.S. Shouche, Extracellular
A. Alizadeh, et al., Green chemical synthesis of gold nanoparticles by using synthesis of crystalline silver nanoparticles and molecular evidence of silver re-
Penicillium aculeatum and their scolicidal activity against hydatid cyst proto- sistance from Morganella sp.: towards understanding biochemical synthesis me-
scolices of Echinococcus granulosus, Environ. Sci. Pollut. Control Ser. 24 (2017) chanism, Chembiochem 9 (2008) 1415–1422.
5800–5810. [316] M. Srivastava, S. Singh, W.T. Self, Exposure to silver nanoparticles inhibits sele-
[290] M. Delavari, A. Dalimi, F. Ghaffarifar, J. Sadraei, In vitro study on cytotoxic effects noprotein synthesis and the activity of thioredoxin reductase, Environ. Health
of ZnO nanoparticles on promastigote and amastigote forms of Leishmania major Perspect. 120 (2012) 56.
(MRHO/IR/75/ER), Iran. J. Parasitol. 9 (2014) 6. [317] C.M. Goodman, C.D. McCusker, T. Yilmaz, V.M. Rotello, Toxicity of gold nano-
[291] A. Jebali, S. Hekmatimoghaddam, B. Kazemi, A. Allaveisie, A. Masoudi, K. Daliri, particles functionalized with cationic and anionic side chains, Bioconjugate
et al., Lectin coated MgO nanoparticle: its toxicity, antileishmanial activity, and Chemistry 15 (2004) 897–900.
macrophage activation, Drug Chem. Toxicol. 37 (2014) 400–409. [318] V. Wiwanitkit, A. Sereemaspun, R. Rojanathanes, Effect of gold nanoparticle on
[292] A.F. Bafghi, M. Daghighi, K. Daliri, A. Jebali, Magnesium oxide nanoparticles the microscopic morphology of white blood cell, Cytopathology 20 (2009)
coated with glucose can silence important genes of Leishmania major at sub-toxic 109–110.
concentrations, Colloids Surfaces B Biointerfaces 136 (2015) 300–304. [319] B.M. Prabhu, S.F. Ali, R.C. Murdock, S.M. Hussain, M. Srivatsan, Copper nano-
[293] M. Yarahmadi, M. Fakhar, M.A. Ebrahimzadeh, A. Chabra, B. Rahimi-esboei, The particles exert size and concentration dependent toxicity on somatosensory neu-
anti-giardial effectiveness of fungal and commercial chitosan against Giardia in- rons of rat, Nanotoxicology 4 (2010) 150–160.
testinalis cysts in vitro, J. Parasit. Dis. 40 (2016) 75–80. [320] R.H. Rice, E.A. Vidrio, B.M. Kumfer, Q. Qin, N.H. Willits, I.M. Kennedy, et al.,
[294] P. Chaubey, R.R. Patel, B. Mishra, Development and optimization of curcumin- Generation of oxidant response to copper and iron nanoparticles and salts: sti-
loaded mannosylated chitosan nanoparticles using response surface methodology mulation by ascorbate, Chem. Biol. Interact. 181 (2009) 359–365.
in the treatment of visceral leishmaniasis, Expet Opin. Drug Deliv. 11 (2014) [321] H.L. Karlsson, P. Cronholm, J. Gustafsson, L. Moller, Copper oxide nanoparticles
1163–1181. are highly toxic: a comparison between metal oxide nanoparticles and carbon
[295] P. Chaubey, B. Mishra, S.L. Mudavath, R.R. Patel, S. Chaurasia, S. Sundar, et al., nanotubes, Chem. Res. Toxicol. 21 (2008) 1726–1732.
Mannose-conjugated curcumin-chitosan nanoparticles: efficacy and toxicity as- [322] E. Oesterling, N. Chopra, V. Gavalas, X. Arzuaga, E.J. Lim, R. Sultana, et al.,
sessments against Leishmania donovani, Int. J. Biol. Macromol. 111 (2018) Alumina nanoparticles induce expression of endothelial cell adhesion molecules,
109–120. Toxicol. Lett. 178 (2008) 160–166.
[296] A. Jebali, B. Kazemi, Nano-based antileishmanial agents: a toxicological study on [323] K.-E. Cha, H. Myung, Cytotoxic effects of nanoparticles assessed in vitro and in
nanoparticles for future treatment of cutaneous leishmaniasis, Toxicol. Vitro 27 vivo, J. Microbiol. Biotechnol. 17 (2007) 1573.
(2013) 1896–1904. [324] S.-Q. Li, R.-R. Zhu, H. Zhu, M. Xue, X.-Y. Sun, S.-D. Yao, et al., Nanotoxicity of
[297] F. Akhtar, M.M.A. Rizvi, S.K. Kar, Oral delivery of curcumin bound to chitosan TiO2 nanoparticles to erythrocyte in vitro, Food Chem. Toxicol. 46 (2008)
nanoparticles cured Plasmodium yoelii infected mice, Biotechnol. Adv. 30 (2012) 3626–3631.
310–320. [325] J.-R. Gurr, A.S. Wang, C.-H. Chen, K.-Y. Jan, Ultrafine titanium dioxide particles in
[298] B. Rahimi-Esboei, M. Fakhar, A. Chabra, M. Hosseini, In vitro treatments of the absence of photoactivation can induce oxidative damage to human bronchial
Echinococcus granulosus with fungal chitosan, as a novel biomolecule, Asia. Pac. epithelial cells, Toxicology 213 (2005) 66–73.
J. Trop. Biomed. 3 (2013) 811–815. [326] S. Huang, P.J. Chueh, Y.-W. Lin, T.-S. Shih, S.-M. Chuang, Disturbed mitotic
[299] J. Priotti, A.V. Codina, D. Leonardi, M.D. Vasconi, L.I. Hinrichsen, M.C. Lamas, progression and genome segregation are involved in cell transformation mediated
Albendazole microcrystal formulations based on chitosan and cellulose deriva- by nano-TiO2 long-term exposure, Toxicol. Appl. Pharmacol. 241 (2009) 182–194.
tives: physicochemical characterization and in vitro parasiticidal activity in [327] S.J. Kang, B.M. Kim, Y.J. Lee, S.H. Hong, H.W. Chung, Titanium dioxide nano-
Trichinella spiralis adult worms, AAPS PharmSciTech 18 (2017) 947–956. particles induce apoptosis through the JNK/p38-caspase-8-Bid pathway in phy-
[300] Brodaczewska K, Wolaniuk N, Donskow-Lysoniewska K, Doligalska M. Chitosan tohemagglutinin-stimulated human lymphocytes, Biochem. Biophys. Res.
Stimulates Lymphocyte Proliferation during the Muscle Phase of Trichinella Commun. 386 (2009) 682–687.
spiralis Infection in Mice. Front Immunol Conference Abstract: 15th International [328] E. Papis, F. Rossi, M. Raspanti, I. Dalle-Donne, G. Colombo, A. Milzani, et al.,
Congress of Immunology (ICI) doi: 103389/conf fimmu2013. Engineered cobalt oxide nanoparticles readily enter cells, Toxicol. Lett. 189 (2009)
[301] M. Abulaihaiti, X.-W. Wu, L. Qiao, H.-L. Lv, H.-W. Zhang, N. Aduwayi, et al., 253–259.
Efficacy of albendazole-chitosan microsphere-based treatment for alveolar echi- [329] P.J. Moos, K. Chung, D. Woessner, M. Honeggar, N.S. Cutler, J.M. Veranth, ZnO
nococcosis in mice, PLoS Neglected Tropical Diseases 9 (2015) e0003950. particulate matter requires cell contact for toxicity in human colon cancer cells,
[302] Salah-Tazdaït R, Tazdaït D, Harrat Z, Eddaikra N, Abdi N, Mameri N. Antiparasite Chem. Res. Toxicol. 23 (2010) 733–739.
Activity of Chitosan. [330] T.J. Brunner, P. Wick, P. Manser, P. Spohn, R.N. Grass, L.K. Limbach, et al., In
[303] S.C. Mohapatra, H.K. Tiwari, M. Singla, B. Rathi, A. Sharma, K. Mahiya, et al., vitro cytotoxicity of oxide nanoparticles: comparison to asbestos, silica, and the
Antimalarial evaluation of copper (II) nanohybrid solids: inhibition of plasmepsin effect of particle solubility, Environ. Sci. Technol. 40 (2006) 4374–4381.
II, a hemoglobin-degrading malarial aspartic protease from Plasmodium falci- [331] L. Bregoli, F. Chiarini, A. Gambarelli, G. Sighinolfi, A.M. Gatti, P. Santi, et al.,
parum, J. Biol. Inorg. Chem. 15 (2010) 373–385. Toxicity of antimony trioxide nanoparticles on human hematopoietic progenitor

525
A. Khezerlou et al. Microbial Pathogenesis 123 (2018) 505–526

cells and comparison to cell lines, Toxicology 262 (2009) 121–129. [339] J.H. Sung, J.H. Ji, J.D. Park, J.U. Yoon, D.S. Kim, K.S. Jeon, et al., Subchronic
[332] S. Hussain, K. Hess, J. Gearhart, K. Geiss, J. Schlager, In vitro toxicity of nano- inhalation toxicity of silver nanoparticles, Toxicol. Sci. 108 (2008) 452–461.
particles in BRL 3A rat liver cells, Toxicol. Vitro 19 (2005) 975–983. [340] J.A. Khan, B. Pillai, T.K. Das, Y. Singh, S. Maiti, Molecular effects of uptake of gold
[333] J. Bogdan, J. Pławińska-Czarnak, J. Zarzyńska, Nanoparticles of titanium and zinc nanoparticles in HeLa cells, Chembiochem 8 (2007) 1237–1240.
oxides as novel agents in tumor treatment: a review, Nanoscale research letters 12 [341] D. Sheehan, B. McDonagh, Oxidative stress and bivalves: a proteomic approach,
(2017) 225. Invertebr. Surviv. J. 5 (2008) 110–123.
[334] J. Wang, Y. Liu, F. Jiao, F. Lao, W. Li, Y. Gu, et al., Time-dependent translocation [342] A. Simon-Deckers, B. Gouget, M. Mayne-L’Hermite, N. Herlin-Boime, C. Reynaud,
and potential impairment on central nervous system by intranasally instilled TiO2 M. Carriere, In vitro investigation of oxide nanoparticle and carbon nanotube
nanoparticles, Toxicology 254 (2008) 82–90. toxicity and intracellular accumulation in A549 human pneumocytes, Toxicology
[335] F. Li, X. Zhou, J. Zhu, J. Ma, X. Huang, S.T. Wong, High content image analysis for 253 (2008) 137–146.
human H4 neuroglioma cells exposed to CuO nanoparticles, BMC Biotechnol. 7 [343] N. Strigul, L. Vaccari, C. Galdun, M. Wazne, X. Liu, C. Christodoulatos, et al., Acute
(2007) 66. toxicity of boron, titanium dioxide, and aluminum nanoparticles to Daphnia
[336] R. Lei, C. Wu, B. Yang, H. Ma, C. Shi, Q. Wang, et al., Integrated metabolomic magna and Vibrio fischeri, Desalination 248 (2009) 771–782.
analysis of the nano-sized copper particle-induced hepatotoxicity and ne- [344] R. Colognato, A. Bonelli, J. Ponti, M. Farina, E. Bergamaschi, E. Sabbioni, et al.,
phrotoxicity in rats: a rapid in vivo screening method for nanotoxicity, Toxicol. Comparative genotoxicity of cobalt nanoparticles and ions on human peripheral
Appl. Pharmacol. 232 (2008) 292–301. leukocytes in vitro, Mutagenesis 23 (2008) 377–382.
[337] V. Sharma, R.K. Shukla, N. Saxena, D. Parmar, M. Das, A. Dhawan, DNA damaging [345] R. Wan, Y. Mo, X. Zhang, S. Chien, D.J. Tollerud, Q. Zhang, Matrix metallopro-
potential of zinc oxide nanoparticles in human epidermal cells, Toxicol. Lett. 185 teinase-2 and-9 are induced differently by metal nanoparticles in human mono-
(2009) 211–218. cytes: the role of oxidative stress and protein tyrosine kinase activation, Toxicol.
[338] B. Wang, W. Feng, M. Wang, T. Wang, Y. Gu, M. Zhu, et al., Acute toxicological Appl. Pharmacol. 233 (2008) 276–285.
impact of nano-and submicro-scaled zinc oxide powder on healthy adult mice, J. [346] B. Fahmy, S.A. Cormier, Copper oxide nanoparticles induce oxidative stress and
Nanoparticle Res. 10 (2008) 263–276. cytotoxicity in airway epithelial cells, Toxicol. Vitro 23 (2009) 1365–1371.

526

You might also like