Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

JBIC Journal of Biological Inorganic Chemistry (2019) 24:929–941

https://doi.org/10.1007/s00775-019-01717-7

MINI REVIEW

Antimicrobial activities of biologically synthesized metal


nanoparticles: an insight into the mechanism of action
Parveen Nisar1 · Nasir Ali1 · Lubna Rahman1 · Muhammad Ali1 · Zabta Khan Shinwari1,2,3

Received: 22 February 2019 / Accepted: 28 August 2019 / Published online: 12 September 2019
© Society for Biological Inorganic Chemistry (SBIC) 2019

Abstract
Increasing antimicrobial resistance is a clinical crisis worldwide. Recent progress in the field of green synthesis has fascinated
scientists and researchers to explore its potentials against pathogenic microbes. Bioinspired-metal-based nanoparticles (silver,
copper, gold, zinc, etc.) have been reported to be tested against both Gram-positive and Gram-negative bacteria such as B.
subtilis, E. coli, Staphylococcus aureus, etc., as well as some pathogenic fungi including A. niger, F. oxysporum, A. fumigatus,
etc., and are testified to exhibit inhibitory effects against pathogenic microbes. The possible modes of action of these metal
nanoparticles include: (a) excess production of reactive oxygen species inside microbes; (b) disruption of vital enzymes in
respiratory chain via damaging microbial plasma membranes; (c) accumulation of metal ions in microbial membranes; (d)
electrostatic attraction between metal nanoparticles and microbial cells which disrupt metabolic activities; and (e) inhibition
of microbial proteins/enzymes by increased production of H ­ 2O2. Although these pathways are interconnected, information
on potential mechanism of most of these biogenic nanoparticles is still limited. Further exploration of these mechanisms
could help in tackling the burning issue of antibiotics resistance.

Keywords  Green synthesis · Metal nanoparticles · Antimicrobial activity · Antimicrobial mode of action

Introduction The burden of microbial infectious diseases is a serious


health issue and economic burden around the globe. Due
Nanoparticles are the particles with one dimension (at least) to antimicrobial-resistant microorganisms, the number and
in the size range of 10–100 nm [1]. They can be synthesized severity of infections have increased; and in USA alone;
by physical and chemical methods [2]. However, these meth- annually, a cost of extra $125 billion increase has been
ods have potential hazards due to manipulation of certain resulted in hospital charges [5, 6].
stabilizing agents, high radiation, and high concentrated There are tons of nanoparticle-based products marketed
reductants [3]. An alternative way to develop ecofriendly, for bacterial diagnosis, medical devices, and antibiotic
non-toxic, and reliable methods is based on biological syn- delivery. Various organic and inorganic nanoparticles with
thesis of nanoparticles. The safest of these methods is the their unique physicochemical characteristics are playing
one that uses medicinal plants [4]. Another class of biologi- important role in rapid, sensitive, and selective detection of
cal synthesis is hybrid methods that combine organic and infections. In addition, some of these biosynthesized metal
inorganic compounds [2]. nanoparticles have the potential to heal microbial infections
involving multidrug-resistant pathogens. Thus, popular to
combat microbial infections [7].
There are various metal nanoparticles that can be used
* Muhammad Ali as antimicrobial agents, e.g., ZnO and ­TiO2 nanocrystals.
alibiotech01@gmail.com These nanoparticles may alter microbial cell integrity and
1 metabolism processes [8] resulting in microbial death.
Molecular Systematics and Applied Ethnobotany
Laboratory, Department of Biotechnology, Quaid-i-Azam Along with antimicrobial properties of ZnO nanoparticles,
University, Islamabad 45320, Pakistan it can also exhibit a photocatalytic activity under the sun-
2
National Council for Tibb (NCT), Islamabad, Pakistan light towards the degradation of rhodamine-B (RhB). RhB
3 is one of the common water pollutants released by paper and
Pakistan Academy of Sciences, Islamabad, Pakistan

13
Vol.:(0123456789)

930 JBIC Journal of Biological Inorganic Chemistry (2019) 24:929–941

textile industries [9]. Similarly, CdSe nanoparticles exhibit search results in different figures. Silver and gold nanopar-
antibacterial potential against a vast variety of bacterial spe- ticles have remained in the focus of most researchers due
cies; E. coli, Staphylococcus aureus, E. hermannii, and P. to their numerous biomedical applications. These biogenic-
vulgaris. Among these P. vulgaris are more susceptible to metal nanoparticles have been extensively manipulated for
these nanoparticles [10]. in vitro and in vivo biological screening with multifarious
Among other methods, “green synthesis” of metallic nan- antimicrobial applications. However, very limited informa-
oparticles is popular because of their harmless procedure. tion is available on the mechanism of bioinspired-metal NPs
Several studies have shown green biosynthesis of different action. In this study, we have discussed possible mechanism
metallic nanoparticles (such as copper, silver, gold, iron of antimicrobial activities of different biosynthesized metal
oxide, zinc oxide, etc.) from various plants. We have sum- nanoparticles.
marized the studies of plant-inspired synthesis of metallic
nanoparticles in Tables 1 and 2. The plants extracts used by
these methods exhibit various antimicrobial properties [11]. Metal nanoparticles and their antimicrobial
Table 1 shows green synthesis of various metallic nanoparti- potency
cles from different plants, their sizes, and nature of activity.
Table 2 shows the summary of studies conducted on green Metals have been used as antimicrobial agent for centuries
synthesis of metallic particles, the plants extracts used, NPs in several countries. In 1500 BP, the Egyptians have used
sizes, and their possible mechanisms. the copper salt as a constringent. The Greek, Egyptians,
Although the précised mechanism of nanoparticles action Persians, Romans, and Indians have used silver and copper
against microbes is still under investigation; some of the to disinfect water and for food preservation [40, 41]. Small
important proposed possibilities exist which include free size and larger surface area of metal nanoparticles have
metal ion toxicity generating from dissolution from the sur- shown improved antimicrobial activities. By the advent
face of synthesized nano-metals, and oxidative stress arising of nanotechnology, several studies have been conducted
from the reactive oxygen species (ROS) on the surface of to synthesize and investigate metal nanoparticles with
nanoparticles [39]. better antimicrobial properties [42]. ZnO are reported
Several articles are available dealing with antimicrobial to be significantly efficient against pathogenic microbes
activities of biosynthesized nanoparticles. Most of these and viruses. However, ZnONPs are much more efficient
reports are limited to specific type of metals or biological against pathogens as compared to zinc oxide due to their
source or pathogen. Comprehensive data (based on the dec- small size. Tabernaemontana divaricata-based zinc oxide
ade 2009–2018) from the peer reviewed journals indexed nanoparticles have a promising photocatalytic activity for
by web of knowledge regarding plant-mediated synthesis methylene blue dye degradation under sunlight [43]. Due
and green synthesis of different metallic nanoparticles to the selective nature of rare-earth metal oxides (REMO),
are shown in Figs. 1 and 2, respectively. It is worth noting it can widely be used in certain fields: energy, ecological,
that in the literature, both key terms “synthesis and green and biomedical. Cerium oxide ­(CeO2) is one of the most
synthesis” are used alternatively for biological synthesis accessible earth metal oxides meant for the excitation via
methods. Therefore, we have shown that both key terms light-harvesting performance. ­C eO 2 nanostructure has

Table 1  Antimicrobial potential NPs type Plant Size Activity References


of green-synthesized metallic
nanoparticles from various AgNPs Abutilon indicum 50–100 nm Antimicrobial [12]
plants’ extracts
Cleome viscosa L. 20–50 nm Antimicrobial [13]
Shikakai and Reetha 20–40 nm Detection of Mycobacte- [14]
rium tuberculosis
Talinum triangulare (Jacq.) Not given Antimicrobial [15]
Datura stramonium 15–20 nm Antibacterial [16]
Banana (Musa paradisiaca) 23.7 nm Antimicrobial [17]
Azadirachtaindica 34 nm Antimicrobial [17]
AuNPs Panax ginseng 10–40 nm Antibacterial [18]
ZnONPs Aloe vera 8–18 nm Biofilm inhibition [19]
L. aculeate 12 nm Antifungal [20]
Aloe vera 8–18 nm Biofilm inhibition [21]
CuONPs Citrus medica 20 nm Antimicrobial [22]

13
JBIC Journal of Biological Inorganic Chemistry (2019) 24:929–941 931

Table 2  Selected studies reporting potential mechanism of antimicrobial activity of metal nanoparticles


NPs type Plant Size (nm) Activity Antimicrobial mechanisms References

AgNPs Ocimum gratissimum 31 nm Antibacterial


Excess production of ROS by damaging plasma [22]
membrane that prevent electron transport and disrupt
respiratory chain enzymes; eventually cause cell
death
Acacia rigidula 15 and 25 nm Antibacterial Release of ­Ag+ ions, production of ROS, and free [23]
radicals may interact with cell wall components
which produce toxicity; thus inhibit bacterial growth
by damaging bacterial cell wall
Syngonium podophyllum 10.41 nm Antifungal Silver ions and nucleic acid complexes, interact with [24]
proteins/nucleosides; or A ­ g+ that may accumulate
in membrane, penetrate into microbial cell, interact
with nitrogen bases and as a result denature the DNA
Acalyphaindica 20–30 nm Antibacterial Proteins and other elements in a cell that contain [25]
sulphur or phosphorus; such as sulphur-containing
proteins in membranes or within the cells; and other
biomolecules like DNA are the favorite reactive
sites for binding silver nanoparticles. Silver ions
disrupt the function of membrane bound enzymes
(vital enzymes in respiratory chain) and respiratory
enzymes that cause the complete destruction of
bacterial cell
AuNPs Galaxaura elongate 3.85–77.13 nm Antibacterial Inhibit bacterial growth by reaction of Au ions with [26]
SH groups of protein by uncoupling respiratory
electron transport from oxidative phosphorylation;
as a result disruption of respiratory chain enzymes or
interruption of membrane permeability to phosphate
and proton
Similarly, impairment of DNA replicated strands by
interacting with nucleic acids
Ananascomosus 16 nm Antibacterial Antibacterial mechanism of AuNPs depends upon the [27]
morphology and physiology of cell. As electrostatic
attraction occurs between AuNPs and bacterial cell;
Au ions are released which penetrate in bacterial cell
wall. These ions further disrupt the metabolic activi-
ties eventually cause cell death
CuO NPs Aloe vera 20 nm Antibacterial CuNPs rupture the bacterial cell wall by releasing Cu [28]
ions that bind negatively charged cell wall, attach-
ment of Cu ions with DNA molecule through forma-
tion of cross-linking strands with nucleic acids; thus
disorganization of DNA helical strand (denaturation
of protein) leads to cell death.It also interrupt bio-
chemical process of bacterial cell
Gloriosa superba L. 5–10 nm Antibacterial Due to electrostatic attraction; released Cu ions cause [29]
disruption of biochemical activities of bacterial cell
and transformation of helical structure of DNA by
interacting between and within nucleic acids
Citrus medica Linn. (Idilimbu) 20 nm Antimicrobial Metal nanoparticles release ions; and accumulation of [30]
these ions and nanoparticles on cell surface form pits
in the membrane; cause leakage of cellular compo-
nents. Furthermore, CuNPs penetrate in microbial
cell and inhibit their enzymes/proteins by increasing
production of H ­ 2O2 that finally cause cell death
It may also show antibacterial activity by interacting
of Cu ions with SH group; by producing ROS; and
by disorganizing DNA helical structure
Pam-ZnO Plectranthus amboinicus 20–50 nm Antibacterial Diffusion and penetration into the exopolysaccharide [31]
and deep inside the layers. Thus, removes the MRSA
ATCC 33591 biofilms and bacterial cells are reduced

13

932 JBIC Journal of Biological Inorganic Chemistry (2019) 24:929–941

Table 2  (continued)
NPs type Plant Size (nm) Activity Antimicrobial mechanisms References

Cu NPs Acalyphaindica 26–30 nm Antimicrobial Copper nanoparticles can inhibit cellular growth by [32]
destroying cell wall and as a consequently interrupt
the activity of cellular enzyme
Gloriosa superba L. 5–10 nm Antibacterial
Cu has high affinity towards carboxyl and amine [33]
groups present on cell surface. The released Cu ions
may bind with DNA and disrupt the helical structure
via cross-linking between nucleic acid strands. It
also disrupt the biochemical processes of bacterial
cell
ZnONPs Azadirachtaindica (L.) 18 nm Antimicrobial The antimicrobial activities of ZnO NPs are due to [34]
the increase production of ­H2O2; able to penetrate
the cell membrane that eventually causes microbial
death
Mostly metal oxide nanoparticles are associated with
larger band gap and this cause unfavourable condi-
tions for recombination of excitons. As a result,
higher concentration of reactive oxygen species,
enhance antimicrobial activity
Aloe 40–25 nm Antimicrobial ROS react with H ions to generate hydrogen perox- [35]
ide, which can enter bacterial cell wall and cause
bacterial death. Furthermore, nanoparticles attach to
SH groups of bacterial protein present on cell wall,
which decreases cell permeability and leads to cell
lysis
This damage also leads to leakage of proteins, genetic
material and minerals which cause cell death
Nyctanthes arbor-tristis 12–32 nm Antifungal ZnONPs change the permeability of outer surface of [36]
plasma membrane; help nanoparticles enter to the
cytoplasm and inhibit cell growth by interfering with
biochemical process within cell
Production of ­H2O2 and ROS (singlet oxygen and
hydroxyl radicals) also lead to the fungal cell death
Ongamia pinnata 100 nm Antibacterial ZnONPs generate reactive oxygen species, which enter [37]
into cell through small pores present in cell. These
ROS when enter to the cell; kill bacterial cell due
to leakage of minerals, proteins and some cellular
components from the cell
Trifolium pratense < 100–190 nm Antibacterial Mechanism of antibacterial activity of ZnO involves [38]
the disruption of bacterial cell membrane and leak-
age of cytoplasmic components due to the produc-
tion of ROS which eventually cause bacterial cell
lysis
Azadirachtaindica (L.) Not given Antimicrobial Generation of free radical such as ­H2O2 molecules that [34]
enter the cell membrane which leads to the bacterial
cell death

become an attractive material, since there is less energy semiconductor, shows bio-sensing, electronic gas sens-
difference among higher ­(Ce4+) and lower ­(Ce3+) ions in ing, and electro-catalysis properties. Both cobalt oxide
ceria using their storage capacity and superior oxygen and their synthesized nano-composites have potential
mobility. Nowadays, erbium-doped ceria nanoparticles are applications in electrical, dielectric, and biosensors fields.
found to be beneficial for the decomposition of RhB dye Furthermore, these nano-composites have been reported
in aquatic industrial pollution [44]. Nanostructured metal with antibacterial activity when tested for Gram-positive
oxides (MnO, ZnO, ­MoO3, ­Co3O4, etc.) are increasingly and Gram-negative bacteria [45]. Biosynthesis of differ-
in-demand for their application as low-cost sensors due to ent metal nanoparticles (AgNPs, AuNPs, ZnONPs, and
their properties of semi conductivity and good compat- CuNPs) from several plants and their potential antimicro-
ibility. Mainly, cobalt oxide is a p-type antiferromagnetic bial activities are discussed below. In addition, chemical

13
JBIC Journal of Biological Inorganic Chemistry (2019) 24:929–941 933

200
AgNPs AuNPs ZnONPs CuNPs Silver nanoparticles (AgNPs)
180
160 Among nanoparticles, silver nanoparticles are mostly
Number of publicaons

140 exploited against various microbes due to its fast produc-


120 tion and toxic nature to bacterial cell [46]. Biosynthesis of
100 silver nanoparticles from biological sources (mostly from
80 plants) is a single step technique which involves the reduc-
60 tion and stabilization of silver ions through combination
40 of biomolecules (enzymes, polysaccharides, amino acids,
20 tannins, alkaloids, terpeniods, phenolic, and vitamins)
0 derived from medicinal plants [47].
2009 2010 2011 2012 2013 2014 2015 2016 2017 2018 Different plant extracts have been reported for the syn-
Year thesis of silver nanoparticles used against several micro-
bial activities. Kumar et al. [47] reported spherical-shaped
Fig. 1  Number of publications dealing with synthesis of metallic nan- silver nanoparticles biosynthesized using an aqueous mix-
oparticles from different plants. The literature search is based on the ture of Alternanthera dentate extract. These bioinspired
key term “Synthesis of AgNps, AuNps, and CuNps from plants”. The
x-axis represents years of publication (from 2009 to 2018) and y-axis
silver nanoparticles had potential antimicrobial activity
represents the number of articles published for each nanoparticle syn- against E. coli, Enterococcus facial, Pseudomonas aer-
thesis method uginosa, and Klebsiella pneumonia [48]. Reduction of
silver ions to nanoparticles using extract of Disodium
trifolium has been attributed to the presence of ascorbic
acids, hydrogen ions, and N ­ AD+ in the extract [49]. Study
reported by Anbazhagan et  al. [49] demonstrated that
AgNPs also exhibit broad spectrum antimicrobial activi-
300
ties against both Gram-negative and Gram-positive bacte-
250 AgNPs AuNPs ZnONPs CuNPs ria such as B. subtilis, E. coli, and S. aureus [50].
Number of publicaons

AgNPs have also been known to have inhibitory prop-


200
erties biosynthesized from S. aureus through a bioreduc-
150 tion of aqueous ­Ag+ ion with the culture supernatants of
S. aureus. These AgNPs were tested for their antimicro-
100
bial activities against certain infectious microbes such
50 as methicillin-resistant S. aureus, methicillin-resistant
Staphylococcus epidermidis, Streptococcus pyogenes, K.
0 pneumoniae, and S. typhi. Of these bacteria, methicillin-
2009 2010 2011 2012 2013 2014 2015 2016 2017 2018
resistant S. aureus, S. pyogenes, and methicillin-resistant
Year
S. epidermidis showed more sensitivity to AgNPs treat-
ment [50]. Maiti et al. [51] reported the antibacterial activ-
ity of AgNPs (biosynthesized from an aqueous extract of
red tomato; Lycopersicon esculentum) against a model
bacterium E. coli. For antibacterial evaluation, the E. coli
was inoculated on Luria broth agar plate in the presence
Fig. 2  Number of publications dealing with green synthesis of metal- of various amounts of AgNPs. Zones of inhibition showed
lic nanoparticles from different plants. The literature search is based bactericidal activity at high concentrations while bacterio-
on the key term “Green synthesis of AgNps, AuNps, and CuNps from
plants”. The x-axis represents years of publication (from 2009 to
static at low concentrations [51].
2018) and y-axis represents the number of articles published for each Variations in concentrations of biosynthesized AgNPs
nanoparticle synthesis method lead to the investigation for assessing the antifungal prop-
erties of silver nanoparticles against pathogenic fungi Tri-
chosporon asahii. For this purpose, T. asahii was grown
on potato dextrose agar medium having various concen-
trations of silver nanoparticles and the antifungal effect
structure of metal nanoparticles stabilized with various was determined using minimum inhibitory concentration
plant’s phytochemicals is schematically presented below (MIC) [52]. Xia et al. [52] reported that although MIC
(Fig. 3).

13

934 JBIC Journal of Biological Inorganic Chemistry (2019) 24:929–941

Fig. 3  Schematic diagram of bioactive compounds within Nicotiana tabacum and Aleo vera with the proposed mechanism of reaction. During
this process, NPs’ structures are stabilized with plant’s phytochemicals

for silver nanoparticles was lower than other antifungal radicals by the silver nanoparticles is believed to be another
drugs (used commonly in clinical practices); however, it mechanism which results in cell death. The electron spin
can inhibit the fungal growth more efficiently. They also resonance spectroscopy studies suggested that the formation
mentioned the mechanism that inhibits the growth of T. of free radicals occurs in the contact of silver nanoparticles
asahii which involves invading the fungal cell, damaging with bacterial cell. Hence, these free radicals can damage
its cell wall, and other basic cellular components. the cell membrane and make it porous that eventually lead
to cell death [55].
Reactive oxygen species are metabolic products gener-
Antimicrobial mechanism of silver ating from numerous cells; two cellular organelles, endo-
nanoparticles (AgNPs) plasmic reticulum and mitochondria, involved in their gen-
eration and metabolism. ROS are highly reactive molecules
Though the exact antimicrobial mechanism of silver nano- with the capability to oxidize cellular structures and bio-
particles remains unclear, still, there are certain theories molecules, and as a result cause DNA denaturation, pro-
regarding antimicrobial mechanism of silver nanoparticles tein modifications, and lipid peroxidation that leads to cell
[24, 53]. AgNPs have the ability to attach on the surface of death. Nanoparticles can produce ROS in the cell and con-
microbial cell wall, penetrate and disturb the structure of cell sequently cause oxidative stress to cellular structures. Das
membrane and ultimately cause cell death. The formation et al. [56] have demonstrated that the production of ROS
of ‘pits’ on treated bacterial cell surface shows the altera- may contribute to silver nanoparticles triggered cytotoxic-
tion and cell membrane damage [54]. The production of free ity in microbes. To monitor intracellular ROS production,

13
JBIC Journal of Biological Inorganic Chemistry (2019) 24:929–941 935

2,7-dichlorofluorescin-diacetate ­(DCFH2-DA) was used for Gold nanoparticles (AuNPs)


the silver nanoparticles treated bacterial cells. After treat-
ment with silver nanoparticles, cells were stained with Gold nanoparticles have unique and tunable surface plas-
­DCFH2-DA. It was observed that nanoparticles treated bac- mon resonance (SPR) with various applications in differ-
teria became DCF+ which indicates ROS production. These ent fields of science especially in biomedical sciences and
nanoparticles attached to the cell membrane and penetrate research [60]. Several studies have reported antimicrobial
inside the cell and produce ROS. The ROS productions activities of gold nanoparticles. MubarakAli et al. [61]
destabilize plasma membrane integrity and decrease level biosynthesized gold nanoparticles from plant extracts
of intracellular ATP, damaging cellular respiratory chain (leaves) of Mentha peperita (Lamiaceae). Nanoparti-
(cellular enzymes) and DNA damage consequently cause cles’ synthesis was studied under UV–Vis spectroscopy
cell lysis and death [56]. and characterized by SEM equipped with EDS and FTIR.
Prabhu and Poulose [53] described the antibacterial The diameter of these biosynthesized nanoparticles was
mechanism of silver nanoparticles that involves the release reported to be 150 nm in size that exhibited strong bacte-
of nanoparticles’ silver ions which can inactivate many vital ricidal effects against pathogenic bacteria, E. coli and S.
enzymes by the interaction of their thiol groups. When silver aureus [61].
ions contact with bacterial cell, they inhibit various basic Green-based gold nanoparticles were also synthesized
functions in the cell. Such alteration of cellular function can using benign (saponin with no capping or any other special
damage the cells, and as a result, generating ROS due to reducing agents) solvent isolated from Trianthema decandra.
the inhibition of cellular respiratory enzymes, can trigger Stable gold nanoparticles were rapidly produced by treat-
microbial cell self-destruction [54]. ment of aqueous solutions containing ­AgNO3 or chloroauric
Another fact is that phosphorous and sulphur are the vital acid with a saponin solution. Ultraviolet–visible spectros-
components of DNA. As these sulphur and phosphorous are copy was used to analyze the reduction of gold nanoparticles
soft bases, while silver is soft acid, they have natural ten- formed in various shapes (hexagonal, spherical, and cubi-
dency to react with each other. Nanoparticles can destroy cal). The Kirby–Bauer method was employed to examine
DNA by interrupting these soft bases which can cause tar- the antimicrobial activity of AuNPs. These nanoparticles
geted cell death. The interaction between silver nanopar- exhibited excellent bactericidal effects against S. aureus,
ticles with sulphur and phosphorous of the DNA can also E. coli, P. vulgaris, S. faecalis, and Y. enterocolitica [34].
cause DNA replication complications in the pathogens. It Vijayan et al. [62] conducted study on the synthesis of gold
has also been noticed that nanoparticles can influence signal nanoparticles from medicinal plant Indigofera tinctoria.
transduction in the bacterial cell by the phosphorylation of Different techniques including UV–Vis spectroscopy, EDX,
protein substrates and can alter the phosphotyrosine profile XRD, TEM, FTIR spectroscopy, and AFM were used for
of bacterial peptides. The NPs dephosphorylate the peptide the confirmation and characterization of biosynthesized
substrates on tyrosine residues of Gram-negative bacteria. It gold nanoparticles. Biosynthesized AuNPs showed high
stops the bacterial growth by signal transduction inhibition antimicrobial activity against certain pathogenic strains
(Fig. 4) [57]. including S. aureus, B. pumilis, E. coli and Pseudomonas
Another antibacterial mechanism of biosynthesized sil- species, A. niger, and A. fumigatus [62]. Jayaseelan et al.
ver nanoparticle involves the denaturation of bacterial pro- [63] also reported biosynthesis of AuNPs using the extract
tein. Several studies have stated that the electrostatic forces of Abelmoschus esculentus. The crystalline nature of the bio-
between the nanoparticles and cell membrane of microbes synthesized AuNPs (size; 62 nm) was confirmed through
are vital for antimicrobial effects. Microorganisms such as XRD. The results confirmed that these nanoparticles pos-
bacteria, fungi, and viruses have negatively charged surface sessed high antifungal efficacy against Candida albicans and
membranes, while nanoparticles often have positive charge P. graminis with significant potential in the preparation of
on their surface. AgNPs (due to large surface areas) have the certain antifungal drugs as well [63].
ability to bind with functional group (–SH) of proteins and
consequently cause protein denaturation [58].
According to earlier studies, AgNPs might inhibit fungal
cell growth by proton pump damaging and cause fungal sur- Antimicrobial mechanism of gold
face protein denaturation. It influences protein lipid bilayer nanoparticles
or membrane permeability and consequently disrupts cell
membrane. Silver nanoparticles (Fig. 5) also initiate the Cui et al. [64] illustrated the antibacterial mechanism of
accumulation of silver ions, efflux of intracellular ions, AuNPs by elucidating the proteomic and transcriptomic
blocking respiration, and metabolism process by damaging approaches. AuNPs use their antimicrobial capabilities
the electron transport system [59].

13

936 JBIC Journal of Biological Inorganic Chemistry (2019) 24:929–941

Fig. 4  Illustration of possible antibacterial mechanism of action of biosynthesized metal nanoparticles

against multidrug-resistant (Gram-negative) bacteria 4,6-diaminopyrimidine thiol is an analogue of bacterial


through two approaches: degrading membrane potential tRNA base. Hence, it can potentially inhibit bacterial
to decrease the ATP level by inhibiting the activity of tRNA function [65] and eventually lead to cessation of
ATPase, and inhibiting the binding of ribosome subunit microbial growth.
with tRNA. AuNPs induce the ATP level (oxidative phos- The ATP synthesis in the terminal step of oxidative phos-
phorylation pathway), down-regulation of F-type ATP phorylation pathway is catalyzed by F-type ATP synthase.
synthase and ribosome pathways, resulting in transient F-type ATP synthase also works in a reverse manner as
upregulation of chemotaxis. The results also showed that an ATPase to produce the transmembrane proton electro-
4,6-diaminopyrimidine thiol-modified AuNPs have the chemical gradient which are essential for molecular trans-
ability to affect protein synthesis through inhibiting the location [66]. AuNPs down-regulate the F-type synthase
function of bacterial tRNA [64]. The modifier of AuNPs, activity which leads to the decreased ATP levels as reported

13
JBIC Journal of Biological Inorganic Chemistry (2019) 24:929–941 937

differences of protons across diverse biological membranes


by primary active transport of ­H+. Sequentially, these can
be used to drive various secondary active transport systems
through ­H+-dependent antiporters, symporters, and channel-
mediated transport systems. Interactions involving AuNPs
and microbial enzyme (ATPases) might change their normal
conformations, resulting in loss of their function and eventu-
ally leading to cell death [68].
Antifungal activities of nanoparticles have been reported
associated with particle size, smaller the particle size higher
its antifungal potency. This property of nanoparticle can be
Fig. 5  Effect of AgNPs on the activity of proton pump in fungal cell attributed to larger surface area of these nanoparticles result-
can lead to cell death
ing in higher/increased interaction with the target sites on
plasma membranes. In addition, nanoparticles with smaller
particle size can easily penetrate through cell membrane.
Gold (soft acid) has greater tendency to react with sulphur
and phosphorus containing soft bases. Therefore, AuNPs
possibly interact with the phosphorus containing bases in
the DNA or the sulphur-containing proteins in the mem-
brane. These interactions could cause interruption in normal
functioning of DNA such as repair mechanism, replication,
and synthesis of new strands and ultimately cause cell death
[69]. However, the process needs further elaboration that
could lead to understanding of specific pathway associated
with antifungal activity of nanoparticles.

Zinc oxide nanoparticles (ZnONPs)


Fig. 6  Possible antifungal mechanism of AuNPs. Interaction of fun- There are various reported studies about the synthesis of sev-
gal cell and AuNPs, (1) can result in bursting of fungal cell wall (2)
eral types of inorganic metal oxides such as ZnO, ­TiO2, and
CuO. Among these, ZnO has gained much interest because
in E. coli; and consequently cause metabolism failure. The of low expenses on production, ease preparation, and safe
transcriptome analysis confirmed that gold nanoparticles handling [70].
can up-regulate genes involved in the flagellar motility and ZnO is an n-type semiconductor metal oxide with broad
chemotaxis pathway [67]. range of nanostructures and significant antimicrobial prop-
Figure 6 shows the schematic diagram of the possible erties [71].
mechanism of interaction between AuNPs and fungi cell Zinc oxide in nanoscale has also indicated various antimi-
components. The fungicidal effect of gold nanoparticles is crobial properties and potential applications in food preser-
spontaneous and irreversible. AgNPs affect hydrogen ion vations. ZnONPs have been integrated in polymeric matrices
extrusion through plasma membrane H ­ +-ATPase. AuNPs- to improve packaging and provide antimicrobial activity to
treated candida cells showed the inhibition of proton the packaging materials [72]. For food industry and custom-
pumping (transmembrane ­H+-ATPase). Glucose-induced ers, antimicrobial packaging is very important, since these
acidification of the external medium using yeast cells is a packaging can retard microbial growth, maintain food safety,
suitable measure of H­ +-ATPase-mediated proton pumping. and extend product shelf life [73].
The enzymes might be in various conformational states in Several nanoparticles have been synthesized success-
both situations. It was also observed that vanadate could fully employed for food safety applications including zinc
completely inhibits ­H+-efflux; however, fluconazole had no oxide, silver oxide, and magnesium oxide nanoparticles [74].
substantial effect on ­H+-ATPase. Therefore, AuNPs might ZnONPs have shown effective activities against food-borne
interact directly with enzymes present in regulation of the pathogens such as L. plantarum, E. coli, B. subtilis, and S.
proton gradients through plasma membrane. H ­ +-ATPase is aureus [75]. ZnONPs have also many promising bacteri-
one of the ATP-driven enzyme and its function is to convert cidal potentials against a wide variety of both Gram-positive
the energy of ATP hydrolysis to electro-chemical potential and Gram-negative bacteria including other multifarious

13

938 JBIC Journal of Biological Inorganic Chemistry (2019) 24:929–941

properties such as stability, mechanical strength, and bar- species, P. expansum and F. oxysporum. The zinc oxide
rier properties [76, 77]. nanoparticles with size of ~ 70 nm showed notable inhibitory
Researchers have also tested the in vitro antibacterial effects against pathogenic fungi P. expansum and B. cinerea
action of zinc oxide; utilizing nanoparticles suspension in a concentration-dependent manner. These nanoparticles
called nanofluids or pure nanoparticles. ZnO shows signifi- may cause inhibition of fungal hyphae. Hence, it might be
cant obstruction in bacterial growth on a wide range of bac- used as effective fungicide in agriculture and food preserva-
teria, particularly by the catalysis of ROS formation from tion applications [81]. Phytosynthesis-based ZnONPs (size
water and oxygen. ZnO nanofluids are ideal formulation for ranging from 12 to 32 nm) were synthesized from an aque-
antibacterial agents in liquid phases, but due to the hydro- ous extract of Nyctanthes arbor-tristis and were reported
phobic nature of ZnONPs, it tends to aggregate into large with high fungicidal activities against fungal pathogens
flocculates in aqueous media and, therefore, cannot interact including P. expansum, B. cinerea, A. alternata, F. oxyspo-
with microorganisms effectively. Gordon and co-workers rum, and A. niger, indicating that these ZnONPs could be
combined FeO with ZnO to generate magnetic composite efficient fungicidal agents in field of agriculture [36].
nanoparticles with enhanced colloidal suspension ability and
antibacterial activity. Results of the study showed that bio-
genic nano-composites had comparatively higher antibacte- Antimicrobial mechanism of ZnONPs
rial activity for S. aureus than E. coli. However, antibacterial
action of composite nanoparticles depends on the ratio of The bactericidal activity of ZnONPs may be contributed to
Zn and Fe [78]. the size of NPs, their shapes, and surface-capping agents.
Parthenium hysterophorus is an annual herbaceous nox- Previous literature has reported that the smaller size of
ious weed, native to North and South America, West Indies ZnONPs is associated with greater antibacterial efficacy
America, Asia, Africa, and Australia. The weed is known [82]. Xie et al. [82] elucidated that the bactericidal mecha-
for its vigorous growth toxicity to humans and livestock. nism of ZnONPs is attributed to increased levels of oxidative
However, despite all the problems related with this noxious stress exerted in bacterial cells. As the majority of bacte-
weed, recent studies suggested their pharmacological prop- rial virulence genes were observed to be down regulated by
erties. P. hysterophorus confers several health benefits such applying NPs; proposing that bacterial virulence is weaken
as anticancer, antioxidant, antimicrobial, anti-inflammatory, by ZnONPs’ treatment [83].
antitrypanosomal, antimalarial, skin diseases, urinary tract ZnONPs showed remarkable bactericidal and toxic effects
infection, and much more. Furthermore, the weed can be against Campylobacter jejuni even at low concentrations.
used as pesticides, herbicides, insecticides, and phyto-reme- ZnONPs showed considerable increase (up to 52 fold) in
dial agent for metal and dye removal from industrial waste. the expression of oxidative stress gene, measurable mem-
It is a rich source of minerals such as P, N, K, Mn, Fe, Zn, brane leakage, and morphological differences in C. jejuni.
and Cu. It is also used as spice in several parts of the world, In view of these phenomena and cell responses, a conceiv-
useful as low-cost substrate for oxalic acid production, xyla- able mode of ZnO-induced microbial inactivation includes
nase production, and edible protein. In addition, its use in directly interaction between ZnONPs and cell surfaces.
nano-medicine is being carried out with some initially suc- These interactions influence membrane permeability and
cess [79]. allow nanoparticles to enter and incite oxidative stress in
Zinc oxide nanoparticles also possess significant antifun- bacterial cells which inhibit bacterial cell growth and ulti-
gal activities. Gunalan et al. [34] biosynthesized ZnONPs mately cause microbial cell death.
from P. hysterophorus L., and the study was based on the The antifungal mechanism of ZnONPs has not been yet
comparison between chemically synthesized ZnO nanoparti- fully understood. However, an investigation of ZnONPs’
cles and plant-based synthesis of ZnO nanoparticles. Results effects against the fungi B. cinerea and P. expansum dem-
of the study showed that green ZnO nanoparticles enhanced onstrated inhibition of conidial development by distortion
fungicidal activity against certain fungal pathogens includ- of conidiophores of P. expansum. Conidia of P. expansum
ing: R. stolonifera, A. nidulans, T. harzianum, and A. flavus was completely inhibited and conidial development was sup-
as compared to chemically synthesized ZnONPs [35]. More- pressed by ZnONPs. While the inhibition of B. cinerea is
over, P. hysterophorus-based ZnO nanoparticles showed due to the deformation of fungal hyphae [84], this deforma-
antifungal activity against pathogenic fungi: F. oxysporium, tion may be due to higher accumulation of carbohydrates
A. flavus, A. fumigatus, and A. niger. Thus, green-synthe- and nucleic acids. ZnONPs can affect cell function by
sized ZnONPs can inhibit the growth and exert biocidal the production of excessive amount of nucleic acids. The
effect for plant pathogenic fungi and bacteria [80]. increased level of nucleic acid in fungal hyphae may be due
In another study, antifungal efficacy of zinc oxide nan- to the NP-mediated stress response. Similarly, the increase
oparticles was examined against two pathogenic fungal in carbohydrates level can be considered as self-protection

13
JBIC Journal of Biological Inorganic Chemistry (2019) 24:929–941 939

mechanism from ZnONPs, which could lead to deformed to disturb bacterial cell membrane and localize in the cell to
hyphal cells structures and eventually cause cell death. Thus, disrupt cellular enzyme functions and cause inactivation or
these investigations suggest that ZnO nanoparticles have dif- death of bacterial cells [89].
ferent mechanisms of action than that of antibacterial activ- Figure 7 shows fungicidal activity of metal nanoparticles
ity [72]. It was also found that various fungi are known for against A. niger. Electrospun cellulose acetate (CA) com-
their mycotoxin production, i.e., patulin and fusaric acid. It plexes having copper and silver nanoparticles supported
was determined that inhibition rate of these mycotoxin is in mesoporous silica and sepiolite were prepared and used
gradually decreased with an increase in the concentration as fungistatic membranes. The size of nanoparticles for
of ZnONPs. These inhibitions of mycotoxin production may mesoporous silica is range in 5–8 nm and the size of sepio-
be due to the inactivation of specific enzymes involved in lite (3–50 nm) and both are well dispersed inside the fiber.
biosynthesis pathway of these toxins by ZnONPs [85]. Large aggregates in µm range are used and allowed a con-
trolled release of metals to make a fungistatic environment.
The finding showed that when spores were incubated either
Copper nanoparticles (CuNPs) in direct contacts with nanoparticles or included CA com-
plex membrane; the silver and copper nanoparticles resulted
Metal nanoparticles have potential applications in various in impaired growth of fungi. The fungistatic effect against A.
disciplines including nano-devices, nano-sensors, catalysis, niger occurred in germinating spores before the formation
nano-electronics, and information storage. Among different of hyphae growth conidiophore. Sepiolite containing copper
nanoparticles, copper nanoparticles have also gained special has been reported to effect the fungal growth by a decreas-
attention due to their potential applications in antimicrobial ing its metabolic activity. These metal-loaded nanoparticles
and other activities [86]. Cioffi et al. [86] have explained work as reservoirs for the controlled release of metals ions
the effective antibacterial and antifungal activities of cop- that eventually effect fungal growth [90]. Shende et al. [29]
per nanoparticles. Stable copper nanoparticles (size ranging showed likely mechanism of copper nanoparticles in fungi
from 40 to 100 nm) were successfully synthesized using which is based on changes that occur in the function and
Mangolia virginiana leaf extract treated with the aqueous structure of fungal cell. In addition, it can cause cell death
solution of ­CuSO4.5H2O as reducing agent with higher anti- by affecting its DNA and disrupt its replication and tran-
bacterial potentials [87]. The antibacterial test carried out scription mechanisms. Moreover, CuNPs could inactivate
against E. coli and showed remarkable antibacterial activity proteins by interacting with its –SH (sulfhydryl) groups and
[88]. Another plant, citron juice (Citrus medica Linn.) was may cause growth cessation. Another concept suggests that
used to synthesize biogenic nanoparticles (size; 10–60 nm). CuNP-mediated oxidative stress inside microbes could result
These copper nanoparticles showed antimicrobial impact in their cell death [30]. However, further studies are needed
against certain human and plant pathogens. The results
exhibited a substantial inhibitory activity against various
bacteria including K. pneumoniae, E. coli, S. aureus, P. vul-
garis, Shigella flexneri, Propionibacterium acnes, S. typhi, P.
aeruginosa, and E. faecalis. Among these pathogens, E. coli
and E. faecalis were reported with high sensitivity towards
CuO nanoparticles [89].
Sivaraj et al. [31] conducted a study elucidating both anti-
fungal and antibacterial activities of biosynthesized, highly
stable CuONPs using aqueous extract of Acalyphaindica
leaf. The size of the biosynthesized nanoparticles ranged
from 26 to 30 nm. These biosynthesized nanoparticles were
reported with significant antifungal effects against C. albi-
cans as well as noteworthy antibacterial activity against P.
fluorescens and E. coli [32].

Antimicrobial mechanism of CuNPs Fig. 7  Possible antifungal mechanism of metal nanoparticles. The


illustration shows copper and silver nanoparticles supported in
Early research on the antimicrobial capabilities of CuNP- mesoporous silica and sepiolite. Metal ions and nanoparticles are
slowly released from these complexes and effect growth of microbes.
­ u2+ ions can change local conductivity
described release of C (1) metal loaded particles (represented by black dots), (2) damaged
and pH. This salvation of C­ u2+ ions into the solution is able spores, (3) Aspergillus niger, (4) spores, (5) metal releasing mat

13

940 JBIC Journal of Biological Inorganic Chemistry (2019) 24:929–941

to understand molecular mechanisms for CuNP-mediated 18. Gomathi M, Rajkumar P, Prakasam A, Ravichandran K (2017)
inhibition of microbial growth. Resour Effic Technol 3:280–284
19. Narendhran S, Sivaraj R (2016) Bull Mater Sci 39:1–5
20. Ahmed S, Ahmad M, Swami BL, Ikram S (2016) J Radiat Res
Appl Sci 9:1–7
21. Das B, Dash SK, Mandal D, Ghosh T, Chattopadhyay S, Tripathy
Conclusion and future prospects S, Das S, Dey SK, Das D, Roy S (2017) Arab J Chem 10:862–876
22. Escárcega-González CE, Garza-Cervantes J, Vázquez-Rodríguez
Green synthesis of metallic nanoparticles has rapidly cap- A, Montelongo-Peralta LZ, Treviño-González M, Castro EDB,
Saucedo-Salazar E, Morales RC, Soto DR, González FT (2018)
tured scientists’ attention thanks to its involvement in the Int J Nanomed 13:2349
utilization of various biological entities for a variety of effi- 23. Yasir M, Singh J, Tripathi MK, Singh P, Shrivastava R (2018)
cient antimicrobial properties. Several green-based metal Pharmacogn Mag 13:S840
and metal oxide (gold, silver, copper, and zinc oxide) nano- 24. Krishnaraj C, Jagan E, Rajasekar S, Selvakumar P, Kalaichelvan
P, Mohan N (2010) Colloids Surf B 76:50–56
particles possessing substantial antimicrobial features have 25. Abdel-Raouf N, Al-Enazi NM, Ibraheem IB (2017) Arab J Chem
been successfully generated from various biological sources 10:S3029–S3039
of which plants are the most prominent. These bio-nano- 26. Bindhu M, Umadevi M (2014) Mater Lett 120:122–125
materials have been proved to be most effective against dif- 27. Kumar PV, Shameem U, Kollu P, Kalyani R, Pammi S (2015)
BioNanoScience 5:135–139
ferent strains of (plant and human) pathogenic bacteria and 28. Naika HR, Lingaraju K, Manjunath K, Kumar D, Nagaraju G,
fungi. However, the precise mechanism of antimicrobial Suresh D, Nagabhushana H (2015) J Taibah Univ Sci 9:7–12
characteristics for biogenic-metal nanoparticles needs fur- 29. Shende S, Ingle AP, Gade A, Rai M (2015) World J Microbiol
ther experimental support and systematic and clinical trials. Biotechnol 31:865–873
30. Dobrucka R, Długaszewska J (2016) Saudi J Biol Sci 23:517–523
31. Sivaraj R, Rahman PK, Rajiv P, Narendhran S, Venckatesh
R (2014) Spectrochim Acta Part A Mol Biomol Spectrosc
129:255–258
References 32. Elumalai K, Velmurugan S (2015) Appl Surf Sci 345:329–336
33. Sundrarajan M, Ambika S, Bharathi K (2015) Adv Powder Tech-
1. Thakkar KN, Mhatre SS, Parikh RY (2010) Nanomed Nanotech- nol 26:1294–1299
nol Biol Med 6:257–262 34. Gunalan S, Sivaraj R, Rajendran V (2012) Prog Nat Sci Mater Int
2. Liu J, Qiao SZ, Hu QH, Lu GQ (2011) Small 7:425–443 22:693–700
3. Parveen K, Banse V, Ledwani L (2016) AIP conference proceed- 35. Jamdagni P, Khatri P, Rana J (2018) J King Saud Univ Sci
ings. AIP Publishing, New York, p 020048 30:168–175
4. Li X, Xu H, Chen Z-S, Chen G (2011) J Nanomater 36. Vijayakumar S, Vinoj G, Malaikozhundan B, Shanthi S, Vasee-
2011(270974):1–16 haran B (2015) Spectrochim Acta Part A Mol Biomol Spectrosc
5. Gao W, Thamphiwatana S, Angsantikul P, Zhang L (2014) Wiley 137:886–891
Interdiscip Rev Nanomed Nanobiotechnol 6:532–547 37. Chandirika JU, Annadurai G (2018) Glob J Biotechnol Biochem
6. Grass G, Rensing C, Solioz M (2011) Appl Environ Microbiol 13:07–11
77:1541–1547 38. Besinis A, De Peralta T, Handy RD (2014) Nanotoxicology
7. Wilson BA, Salyers AA, Whitt DD, Winkler ME (2011) Bacte- 8:1–16
rial pathogenesis: a molecular approach. American Society for 39. Lemire JA, Harrison JJ, Turner RJ (2013) Nat Rev Microbiol
Microbiology (ASM), Washington, D.C. 11:371
8. Kaviyarasu K, Geetha N, Kanimozhi K, Magdalane CM, Sivaran- 40. Gold K, Slay B, Knackstedt M, Gaharwar AK (2018) Adv Ther
jani S, Ayeshamariam A, Maaza M (2017) Mater Sci Eng C 1:1700033
74:325–333 41. Jeon H-J, Yi S-C, Oh S-G (2003) Biomaterials 24:4921–4928
9. Kaviyarasu K, Magdalane CM, Kanimozhi K, Kennedy J, Sid- 42. Klueh U, Wagner V, Kelly S, Johnson A, Bryers J (2000) J Biomed
dhardha B, Reddy ES, Mola GT (2017) J Photochem Photobiol B Mater Res 53:621–63143
173:466–475 43. Raja A, Ashokkumar S, Marthandam RP, Jayachandiran J, Khati-
10. Kaviyarasu K, Kanimozhi K, Matinise N, Magdalane CM, Mola wada CP, Kaviyarasu K, Swaminathan M (2018) J Photochem
GT, Kennedy J, Maaza M (2017) Mater Sci Eng C 76:1012–1025 Photobiol B Biol 181:53–58
11. Govindaraju K, Basha SK, Kumar VG, Singaravelu G (2008) J 44. Magdalane CM, Kaviyarasu K, Raja A, Arularasu MV, Mola
Mater Sci 43:5115–5122 GT, Isaev AB, Maaza M (2018) J Photochem Photobiol B
12. Lakshmanan G, Sathiyaseelan A, Kalaichelvan P, Murugesan K 185:275–282
(2018) Karbala Int J Mod Sci 4:61–68 45. Amanulla AM, Shahina SJ, Sundaram R, Magdalane CM, Kavi-
13. Sur UK, Ankamwar B, Karmakar S, Halder A, Das P (2018) Mater yarasu K, Letsholathebe D, Maaza M (2018) J Photochem Photo-
Today Proc 5:2321–2329 biol B 183:233–241
14. Ojo OA, Oyinloye BE, Ojo AB, Afolabi OB, Peters OA, Olaiya 46. Roy S, Das T (2015) J Appl Spectrosc 82:598–606
O, Fadaka A, Jonathan J, Osunlana O (2017) J Bionanosci 47. Kumar DA, Palanichamy V, Roopan SM (2014) Spectrochim
11:292–296 Acta Part A Mol Biomol Spectrosc 127:168–171. https​://doi.
15. Singh P, Kim YJ, Yang DC (2016) Artif Cells Nanomed Biotech- org/10.1016/j.saa.2014.02.058
nol 44:1949–1957 48. Ahmad N, Sharma S, Singh V, Shamsi S, Fatma A, Mehta B
16. Ali K, Dwivedi S, Azam A, Saquib Q, Al-Said MS, Alkhedhairy (2011) Biotechnol Res Int 2011(454090):1–8
AA, Musarrat J (2016) J Colloid Interface Sci 472:145–156 49. Anbazhagan S, Azeez S, Morukattu G, Rajan R, Venkatesan K,
17. Ibrahim HM (2015) J Radiat Res Appl Sci 8:265–275 Thangavelu KP (2017) 3 Biotech 7:333

13
JBIC Journal of Biological Inorganic Chemistry (2019) 24:929–941 941

50. Nanda A, Saravanan M (2009) Nanomed Nanotechnol Biol Med 73. Neethirajan S, Jayas DS (2011) Food Bioprocess Technol 4:39–47
5:452–456. https​://doi.org/10.1016/j.nano.2009.01.012 7 4. Jones N, Ray B, Ranjit KT, Manna AC (2008) FEMS Microbiol
51. Maiti S, Krishnan D, Barman G, Ghosh SK, Laha JK (2014) J Lett 279:71–76
Anal Sci Technol 5:40 75. Jin T, Gurtler J (2011) J Appl Microbiol 110:704–712
52. Xia Z-K, Ma Q-H, Li S-Y, Zhang D-Q, Cong L, Tian Y-L, Yang 76. Seil JT, Webster TJ (2011) Acta Biomater 7:2579–2584
R-Y (2016) J Microbiol Immunol Infect 49:182–188 77. Vicentini DS, Smania A Jr, Laranjeira MC (2010) Mater Sci Eng
53. Prabhu S, Poulose EK (2012) Int Nano Lett 2:32 C 30:503–508
54. Sondi I, Salopek-Sondi B (2004) J Colloid Interface Sci 78. Gordon T, Perlstein B, Houbara O, Felner I, Banin E, Margel S
275:177–182 (2011) Colloids Surf A 374:1–8
55. Danilczuk M, Lund A, Sadlo J, Yamada H, Michalik J (2006) 79. Rajiv P, Rajeshwari S, Venckatesh R (2013) Spectrochim Acta
Spectrochim Acta Part A Mol Biomol Spectrosc 63:189–19157 Part A Mol Biomol Spectrosc 112:384–387
56. Das B, Dash SK, Mandal D, Ghosh T, Chattopadhyay S, Tripathy 80. Patel S (2011) 3 Biotech 1(1):1–9
S, Roy S (2017) Arab J Chem 10(6):862–876 81. He L, Liu Y, Mustapha A, Lin M (2011) Microbiol Res
57. Shrivastava S, Bera T, Roy A, Singh G, Ramachandrarao P, Dash 166:207–215
D (2007) Nanotechnology 18:225103 82. Xie Y, He Y, Irwin PL, Jin T, Shi X (2011) Appl Environ Micro-
58. Kim J, Lee J, Kwon S, Jeong S (2009) J Nanosci Nanotechnol biol 77:2325–2331
9:1098–1102 83. Abbasi BH, Anjum S, Hano C (2017) RSC Adv

59. Du H, Lo T-M, Sitompul J, Chang MW (2012) Biochem Biophys 7(26):15931–15943
Res Commun 424:657–662 84. Huang Z, Cui F, Kang H, Chen J, Zhang X, Xia C (2008) Chem
60. Huang S-H (2006) Clin Chim Acta 373:139–143 Mater 20:5090–5099
61. MubarakAli D, Thajuddin N, Jeganathan K, Gunasekaran M 85. Yehia RS, Ahmed OF (2013) Afr J Microbiol Res 7:1917–1923
(2011) Colloids Surf B Biointerfaces 85:360–365. https​://doi. 86. Cioffi N, Torsi L, Ditaranto N, Tantillo G, Ghibelli L, Sabbatini L,
org/10.1016/j.colsu​rfb.2011.03.009 Bleve-Zacheo T, D’Alessio M, Zambonin PG, Traversa E (2005)
62. Vijayan R, Joseph S, Mathew B (2018) Artif Cells Nanomed Bio- Chem Mater 17:5255–5262
technol 46:861–871 87. Lee H-J, Lee G, Jang NR, Yun JH, Song JY, Kim BS (2011)
63. Jayaseelan C, Ramkumar R, Rahuman AA, Perumal P (2013) Ind Nanotechnology 1:371–374
Crops Prod 45:423–429 88. Ahamed M, Alhadlaq HA, Khan M, Karuppiah P, Al-Dhabi NA
64. Cui Y, Zhao Y, Tian Y, Zhang W, Lü X, Jiang X (2012) Biomateri- (2014) J Nanomater 2014:17
als 33:2327–2333 89. Ren G, Hu D, Cheng EW, Vargas-Reus MA, Reip P, Allaker RP
65. Carbon J, David H, Studier MH (1968) Science 161:1146–1147 (2009) Int J Antimicrob Agents 33:587–590
66. Zharova TV, Vinogradov AD (2004) J Biol Chem
90. Quirós J, Gonzalo S, Jalvo B, Boltes K, Perdigón-Melón JA, Rosal
279:12319–12324 R (2016) Sci Total Environ 563:912–920
67. Wani IA, Ahmad T (2013) Colloids Surf B 101:162–170
68. Ahmad T, Wani IA, Lone IH, Ganguly A, Manzoor N, Ahmad A, Publisher’s Note Springer Nature remains neutral with regard to
Al-Shihri AS (2013) Mater Res Bull 48(1):12–20 jurisdictional claims in published maps and institutional affiliations.
69. Tan YN, Lee KH, Su X (2011) Anal Chem 83:4251–4257
70. Jayaseelan C, Rahuman AA, Kirthi AV, Marimuthu S, Santho-
shkumar T, Bagavan A, Gaurav K, Karthik L, Rao KB (2012)
Spectrochim Acta Part A Mol Biomol Spectrosc 90:78–84
71. Szabó T, Németh J, Dékány I (2003) Colloids Surf A 230:23–35
72. Espitia PJP, Soares NDFF, dos Reis Coimbra JS, de Andrade
NJ, Cruz RS, Medeiros EAA (2012) Food Bioprocess Technol
5:1447–1464

13

You might also like