Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Materials Science for Energy Technologies 3 (2020) 756–769

Contents lists available at ScienceDirect

Materials Science for Energy Technologies


CHINESE ROOTS
GLOBAL IMPACT
journal homepage: www.keaipublishing.com/en/journals/materials-science-for-energy-technologies

Bactericidal activity of silver nanoparticles: A mechanistic review


Syed Anees Ahmad a, Sabya Sachi Das b, Ayesha Khatoon c, Mohammed Tahir Ansari d, Mohd. Afzal e,
Md Saquib Hasnain f,⇑, Amit Kumar Nayak g,⇑
a
Department of Pharmacology, Hygia Institute of Pharmaceutical Education and Research, Lucknow, U.P., India
b
Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835 215, Jharkhand, India
c
Molecular Cell Biology Laboratory, Integral Information and Research Centre-4 (IIRC-4), Department of Biosciences, Faculty of Science, Integral University, Kursi Road,
Lucknow 226026, India
d
School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, Semenyih, Selangor 43500, Malaysia
e
Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
f
Department of Pharmacy, Shri Venkateshwara University, NH-24, Rajabpur, Gajraula, Amroha 244236, U.P., India
g
Department of Pharmaceutics, Seemanta Institute of Pharmaceutical Sciences, Mayurbhanj 757086, Odisha, India

a r t i c l e i n f o a b s t r a c t

Article history: Metallic nanoparticles such as silver nanoparticles (AgNPs) have gained lots of attention due to the con-
Received 19 July 2020 tinuous upsurge in microbial infections and diseases, and also inefficient treatment. Also, due to rapid
Revised 9 September 2020 intensification in the antibiotic resistance in this period has revived the consideration of the researchers
Accepted 9 September 2020
and scientists to explore the therapeutic abilities of silver and its nanoparticulate systems as potential
Available online 29 September 2020
antimicrobial agents. Indeed, numerous studies suggested that AgNPs have exhibited significant antimi-
crobial actions, specifically against the bacterial infections. Ample of studies have been performed to
Keywords:
evaluate and estimate the antibacterial potentials of silver and its associated products, and it was noticed
Silver
Silver nanoparticles
that the they these Ag-particles triggers the oxidative stress, protein dysfunction, membrane and DNA
Bacterial resistance damage, leading to microbial cell damage. Although it has exhibited antimicrobial activities, but its
Antibacterial activity adverse effects over human health has been a major concern. Further, in this review we have summarized
Mechanism of action the significance of silver and AgNPs, various approaches for the preparation of AgNPs, antibacterial mech-
Toxicity profile anism and probable toxicity in the cell environment. Furthermore, this review has been focused to con-
sider the major mechanisms behind the bactericidal actions of the silver and silver nanoparticles-based
systems, followed by comprehensive ideas related to their toxic potentials. This review would open new
robust possibilities to develop silver nanoparticle-based antimicrobial systems with desired properties
for effective treatment of bacterial infections in humans.
Ó 2020 The Authors. Production and hosting by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-
nd/4.0/).

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 757
2. Silver: an effective antibacterial agent . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 757
2.1. Synthesis of silver nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 758
2.1.1. Physical method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 758
2.1.2. Chemical method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 759
2.1.3. Biological method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 759

⇑ Corresponding authors.
E-mail addresses: msaquibhasnain@gmail.com (M. Saquib Hasnain), amitkrnayak@yahoo.co.in (A. Kumar Nayak).
Peer review under responsibility of KeAi Communications Co., Ltd.

Production and hosting by Elsevier

https://doi.org/10.1016/j.mset.2020.09.002
2589-2991/Ó 2020 The Authors. Production and hosting by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
S. Anees Ahmad, S. Sachi Das, A. Khatoon et al. Materials Science for Energy Technologies 3 (2020) 756–769

3. Antibacterial mechanism of AgNPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 760


3.1. Antibacterial applications of AgNPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 763
4. Toxicity of AgNPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 764
5. Conclusion and future perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 764
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 765
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 765

1. Introduction the thiol (-SH) links of crucial compounds and disables them [15].
Trials have showed that the replication capacity of the DNA gets
The metallic nanoparticles (NPs) are the most encouraging as lost, when the microbes are treated with Ag-based particles. Differ-
they show great antibacterial properties because of their extensive ent reviews have demonstrated confirmation of basic alterations in
surface range to volume proportion, which is coming up as the ebb the cell layer and the arrangement of little electron-thick granules
and flow enthusiasm for research because of the development of shaped by silver and sulphur [16].
microbial resistance against metallic particles, anti-toxins and Ag-based particles have exhibited helpful and compelling
the advancement of safe strains [1]. Metallic particles in the antibacterial applications, yet because of the remarkable activities
nanometre range exhibits physical properties which do not resem- of nanotechnology based materials have given immense support to
ble with both the particle and the mass material. This makes them improve the activities of Ag. Metallic particles with a size in
show astounding properties, for example, expanded reactant nanometre range have exhibited physical characteristics with
movement because of morphologies with very dynamic aspects unique relation to both the particle and the mass material. This
[2]. Microorganisms, for example, microbes and organisms, now makes them show astounding properties, for example, expanded
assume a critical part in the remediation of poisonous metals synergist action because of morphologies with profoundly
through the decrease of the metal particles [3]. Reaction surface dynamic aspects [17–19]. A few electron microscopy strategies
system (RSM) is an accumulation of measurable and scientific pro- could be applied to concentrate the system by which AgNPs inter-
cedures helpful for creating, enhancing and streamlining forms [4]. face with these microbes. Also, a high point annular dull field
RSM is an outstanding strategy connected in the streamlining of (HAADF) filtering transmission electron microscopy (STEM) could
medium constituents and other basic factors in charge of the cre- be used, followed by the formation of a novel specimen planning
ation of biomolecules [5,6]. Notwithstanding dissecting the that maintains a strategic distance from the utilization of substan-
impacts of the autonomous factors, this test approach creates a sci- tial metal-based mixtures [2].
entific model that precisely depicts the general procedure. It has The advancement of new benign microbial strains to the pre-
been effectively connected to advance the conditions in suste- sent anti-infective agents [20] has been a major issue in common
nance, compound and natural procedures [4,7]. health systems; subsequently, there is a perquisite to develop
The routine of single component streamlining by keeping up novel bactericidal agents [21]. The microbial strains have been
different variables at an unspecified steady level does not portray reported to exist with varied film assemblies which allows to rec-
the joined impact of the considerable number of elements ognize them as Gram-negative (G ve) or Gram positive (G+ve).
included. The one component at once technique requires countless The main mechanism lies behind the conjugation with the main
to decide ideal levels, which is dreary and tedious. Streamlining all sites of the layers of peptidoglycan (PG). G ve microorganisms
the affecting parameters can dispense with these restrictions of a have a thin PG layer of 1–5 nm that exists between the external
solitary element enhancement prepare all in all by measurable test layer and the cytoplasmic film, however the G+ve microscopic
configuration utilizing RSM [5,8]. Gamma-light amalgamation of organisms do not have the external film yet have a thick PG layer
metallic NPs [9] has been additionally utilized as a standout of ~30 nm [2].
amongst the most encouraging techniques to deliver AgNPs The complexes of Ag have likewise been utilized as a part of the
because of some imperative focal points. When contrasted with therapeutics to treat blazes and variety of diseases [16]. Also Ag
ordinary synthetic/photochemical systems, the radiochemical pro- and its salt forms have been significantly utilized as a potential tool
cedure can be performed to diminish Ag+ particles at the encom- as bactericidal agents for the treatment of various microbial infec-
passing temperature without utilizing over the top decreasing tions [22]. Excellent endeavours have been performed to investi-
operators or creating undesirable by-results of the reductant. In gate the bactericidal features of Ag-based materials through
addition, diminishing operator can be consistently disseminated electron microscopy and observed the possible interfaces of AgNPs
in the arrangement and AgNPs are delivered in exceptionally with microorganisms [23]. AgNPs have also been used as a material
immaculate and stable shape [10]. The advancement of novel medium for transportation of antibiotics [24], as a blending agent
benign strains of microbes to develop anti-microbial agents have to form composites used in purifications processes [25] and also as
showed significant potentials and thus thusly, there a strong moti- casing material [24]. Although in any phase, the antibacterial prop-
vation has developed to establish new bactericidal agents [11,12]. erties of these AgNPs depends upon their steadiness in the system,
as this exhibits greater reaction time for the interaction between
2. Silver: an effective antibacterial agent the AgNPs and bacterial membranes. Thus, the stability of the
AgNPs in reaction medium plays a crucial role for hindering the
Silver (Ag) has been known to have a toxicity effect over an growth and progression of the microbes [2].
extensive variety of small organisms [13], hence silver-based com- For a long time, Ag has been utilized by the people as an
binations have been widely exploited for its antibacterial applica- antibacterial agent due to immense antibacterial properties against
tions [14,15]. A few salt types of Ag and their subtypes are a wide range of bacterial species. NPs, for the most time are pre-
economically used as bactericidal agents. The bactericidal impact ferred over the basic Ag-based materials due to the diverse quali-
of silver particles on microorganism’s is extremely notable; in ties of the NPs. The smaller size and shape of the AgNPs makes it
any case, the bactericidal system is just incompletely caught on. a potential bactericidal agent with enriched antibacterial impact.
Reports have demonstrated that the Ag+ ions firmly connects with Moreover, due to considerable surface area the AgNPs can loan

757
S. Anees Ahmad, S. Sachi Das, A. Khatoon et al. Materials Science for Energy Technologies 3 (2020) 756–769

antimicrobial impacts to many square meters of its host material study, mono-dispersed AgNPs were developed through oleylamine
[26]. AgNPshave as of now been tried in different field of organic and paraffin framework [47]. It was demonstrated that the
science, medicate conveyance, water treatment and an antibacte- arrangement process of AgNPs could be parted into three phases:
rial compound against both G+ve and G ve microorganisms by dif- development, brooding and Ostwald aging phases. In this tech-
ferent scientists. The greater part of the microscopic organisms has nique, just three chemicals, including AgNO3, oleylamine and
yet created imperviousness to anti-microbial and in this view in paraffin, are utilized all through the entire procedure. The liquid
future it is expected to build up a substitute for anti-infection paraffin has higher melting point and this makes it usage more fea-
agents. AgNPs are alluring as these are non-lethal to human body sible even at higher temperatures and thus are potentially used in
at low fixation and having wide range antibacterial nature. AgNPs preparation of AgNPs at varying temperatures. Something else, the
repress the bacterial development at low fixation than anti- extent of the colloidal AgNPs can be directed not just by altering
infection agents and starting at now no reactions are accounted the warming temperature, or the growth time, additionally by
for [26]. modifying the proportion of oleylamine to the Ag supernatants.
The acting system of Ag has been known in some cases. For Most often the synthesizing procedure of AgNPs preparations does
instance, Ag+ ions repress the phosphate uptake and growth of not uses convoying three prime parts: (i) metal precursors, (ii)
Escherichia coli (E. coli) leading to efflux of aggregated phosphates, diminishing specialists and (iii) settling/topping operators. The
glutamine, mannitol, proline, succinate. Besides, Ag particles generation of colloidal preparations from the decreased of Ag salts
restrain oxidation of glucose, glycerol, fumarate, and succinate in comprises of two stages, nucleation and subsequent development.
E. coli. The AgNPs demonstrate proficient bactericidal properties Furthermore, the size and shape of the colloidal AgNPs mostly
contrasted with different salts due to greater degree of substantial depends upon the conditions of the two phase mentioned above.
surface zone, which gives better contact microorganisms. It has Moreover, for the combination of monodispersed AgNPs with iden-
been observed that when the AgNPs enter into cell membrane of tical size distribution, all cores are prerequisite to form in the
bacteria, they frame a low sub-atomic weight within the cell. Con- interim. For this phase, each of the core have the similar or equiv-
sequently, the microbes aggregate to shield the DNA from the alent size, followed by the development of similar sized NPs. The
AgNPs. Thus, the AgNPs ideally assault the respiratory system, cel- underlying nucleation and subsequent development of initial cores
lular division for longer duration prompting to cell passing [27]. could be significantly regulated by monitoring the response fac-
tors, including temperature, pH, precursors, degeneration opera-
2.1. Synthesis of silver nanoparticles tors (glucose,ethylene glycol, etc.) and stabilizing agents
(polyvinyl alcohol, PVP etc.) [48–50].
AgNPs could be efficiently prepared through physical, chemical,
and biological approaches [28]. At present, top-down and bottom- 2.1.1. Physical method
up are the two methods that have been mostly utilized for the syn- For a physical approach, the metallic NPs are mostly formed by
thesis of AgNPs [29]. The top-down method is used to reduce the a robust method of evaporation followed by rapid condensation,
size of the bigger structures into nanoscale range while preserves possibly performed by using a tube heater at climatic load. How-
their original characteristics without alterations ion the atomic or ever, this approach has been reported with certain limitations,
sub-atomic levels [30]. The bottom-up method, also described as including a substantial space of tube heater, incredible utilization
molecular nanotechnology, includes the concept of assembling of vitality for enhancing the ecological temperature across the base
materials at atomic or molecular level by a procedure of assem- material. Hence, different strategies for amalgamation of AgNPs
bling or self-assembling [30]. Presently, the most of the technolo- in view of the physical approaches have been produced. A thermal
gies depends upon the top-down method, however, the bottom- disintegration technique was created to orchestrate AgNPs in pow-
up method are frequently used for improvements in material prop- dered shape [51]. The AgNPs were framed by decay of an Ag+-
erties [31–33], electronic engineering [34], energy and dynamics oleate composite, formed by response with AgNO3 and sodium ole-
[35–37], biotechnology-based material development [38–40] ate in aqueous media, at higher temperature (~290 °C). Normal
medicines and healthcare systems [41–44]. In recent years, various molecular size of the AgNPs was acquired of around 10 nm and
approaches have been reported for the blend of AgNPs by employ- thus it showed that the AgNPs have an extremely limit measure
ing constituents, photochemical, physical and organic progresses. appropriation.
Each approach has crucial facts and flaws with some common The physical methods have been reported with various advan-
problems, including expenditures, compliance, particle sizes and tages for the development and establishment of high-quality
distribution. Among the current techniques, the synthetic strate- AgNPs. However, it is a low yield process, which often requires
gies are mostly utilized for generation of AgNPs. Concoction high temperature and power consumption, thereby limiting their
approaches provides a meek tactic to incorporate AgNPs in application in the industrial sector. In contrast, AgNPs can be pro-
arrangements. Mono-disperse tests of Ag-nanocubes were orches- duced in large quantities through chemical and biological routes
trated in huge quantities by treating silver nitrate (AgNO3) with due to their cost-effectiveness. In this respect, we focus mainly
ethylene glycol surrounded by the polyvinylpyrrolidone (PVP) on nanoparticle fabrication usingchemical and biological routes
[45]. It demonstrated that PVP amount and its molar ratio with in this section. The physical synthesis process generally employs
AgNO3, both exhibited significant impacts over the geometric thermal, laser, or arc discharge power to form AgNPs with nearly
shape and size of material. Thus, it was possible regulate the mea- narrow size distribution. In this context, evaporation condensation
sure of Ag-nanocubes by monitoring the various experimental and laser ablation strategies are commonly adopted to form AgNPs.
conditions. The former approach is carried out by simply placing a source
Circular AgNPs with a manageable size and greater mono- metal, i.e., silver target inside the tube furnace, and then vaporized
dispersity were incorporated by utilizing the polyol procedure silver into a carrier gas under atmospheric pressure at high tem-
and an altered forerunner infusion method [46]. In the forerunner peratures. The vapor is then condensed to form nanoparticles.
infusion technique, the infusion rate and response temperature The drawbacks of this approach include high power consumption
were imperative components for delivering identical-sized AgNPs and long heating time to reach thermal stability [28]. Jung et al.
with a lessened size. The infusion of the forerunner arrangement modified this process by using a small ceramic heater with a local
within a warm arrangement was a crucial factor for quicker nucle- heating area for vaporizing silver metal [52]. The temperature gra-
ation within less time, guaranteeing the manufacture of AgNPs. In a dient in the vicinity of the heater surface is very steep, such that
758
S. Anees Ahmad, S. Sachi Das, A. Khatoon et al. Materials Science for Energy Technologies 3 (2020) 756–769

the vapor can cool at a rapid rate, thereby condensing into spher- microwave irradiation is more energy efficient than thermal heat-
ical AgNPs with diameters of 6.2–21.5 nm without agglomeration. ing in the polyol process. Microwave irradiation offers fast and
Alternatively, laser ablation can be used to synthesize AgNPs from homogeneous heating of the reaction medium, typically in a time
a silver target placed in a solution under laser beam irradiation. period of few seconds. As such, it provides uniform nucleation
The nanoparticle size of colloids depends on the laser wavelength, and growth conditions for AgNPs [66].
ablation time, and duration of laser pulses [53–55]. The limitation
of this process is the high cost of laser facility. 2.1.3. Biological method
The limitations associated with both the physical and synthetic
2.1.2. Chemical method strategies for the development of AgNPs includes expensive tech-
The wet chemical route is by far the most economical and com- nique and usage of harmful, perilous chemicals, which might cause
monly used process for preparing nanosilver colloidal dispersions impending ecological and biological hazards. Therefore, it becomes
in water or organic solvents. This route includes chemical reduc- necessary to establish methods which could possibly overcome
tion, microwave-assisted synthesis, microemulsion, photoreduc- these limitations and produce stable and therapeutically active
tion, electrochemical approach, etc. [56,57]. AgNPs. This has led to the evolve of biological strategies, which
Chemical method of synthesis can be subdivided into chemical has encouraged the essential needs and potential usage of AgNPs
reduction, electrochemical, irradiationassisted chemical and pyrol- [67].
ysis methods [58]. Ag NPs synthesis in solution requires metal pre- Integration of green science systems and philosophies into nan-
cursor, reducing agents and stabilizing or capping agent. otechnology is of awesome intrigue which has increased much
Commonly used reducing agents are ascorbic acid, alcohol, borohy- consideration over the previous decade [68]. Green science which
dride, sodium citrate and hydrazine compounds. Sotiriou and Prat- is the utilization of science standards to decrease or take out utiliz-
sinis [59] have shown that the Ag NPs supported on ing of lethal reagents, has come about to noteworthy lessening in
nanostructured SiO2 were obtained by flame aerosol technology, the measure of unsafe deposits to social wellbeing and atmo-
which allows close control of silver content and size. Also, silver/ sphere. Green science is characterized as science supported proce-
silica nanoparticles with relatively narrow size distribution were dures for contamination anticipation which can be reached out to
obtained by flame spray pyrolysis [59]. particular zones including green diagnostic science, naturally well-
Among these, chemical reduction is a relatively simple, high disposed explanatory science and clean expository techniques [69].
yield and cost-effective process through the chemical reduction Green union of AgNPs has pulled in extensive consideration as
of silver salt in water or an organic solvent to form a colloidal sus- of late. In such manner, plants concentrates and normal assets,
pension. This strategy requires silver salt, reducing agent and sta- for example, microbes and proteins are observed to be of optional
bilizing/capping agent. Silver nitrate is the most widely used components in AgNPs amalgamation. Using green constituents has
silver salt precursor. Sodium borohydride (NaBH4), ascorbic acid, a few favourable circumstances including low vitality utilization
glucose, hydrazine, sodium citrate, and ethylene glycol (EG) are and direct process conditions (e.g. weight and temperature)
typical reductants for reducing silver ions [60,61]. deprived of utilizing any lethal chemical [70]. Thus, green blend
Sodium borohydride is a strong reductant for forming fined and strategies utilizing different natural living beings, for example,
monodispersed AgNPs. Weak reducing agents such as sodium yeast, form, green growth and microscopic organisms, and plant
citrate, ascorbic acid, and glucose lead to the formation of rela- extricates have been produced for NPs union [71]. When con-
tively large AgNPs, having a wider size distribution; sodiumcitrate trasted with the metallic components in mass state, metallic NPs
plays the role of a reducing and a stabilizing agent [61]. As recog- display irregular compound, optical, physical, and warm features
nized, colloidal AgNPs tend to contact or link with each other to because of their higher surface area to volume proportion [72].
form aggregates as a result of the attractive Van derWaals forces. Consequently, these novel features make AgNPs (>100 nm) a
The agglomeration of colloidal AgNPs can be prevented by the potential agent for a wide range of utilizations [73].
use of stabilizing agents. The stabilizing effect of polymer-based It is realized that Ag and its associated composites are very
capping agents or non-ionic surfactants of AgNPs suspensions is effective against the real types of microbes and to the infections
based on the charge repulsion or steric hindrance to counteract spread by them [74–76]. Although the main mechanism behind
the van derWaals attraction between colloidal nanoparticles. As the antibacterial and antifungal activities of Ag and Ag-based
such, electrostatic or steric stabilization of colloidal AgNPs is materials is still not clear, but studies have showed that the Ag
achieved through adsorption of macromolecules or organic com- could restrain cell transduction and furthermore causes cell death
pounds to the surfaces of the nanoparticles. Citrate-capped AgNPs [67]. AgNPs, because of their surface design and little size which
exhibit negative surface charge due to the carboxylic moiety of thus, increment their surface to volume proportion, display stun-
citrate. This leads to electrostatic repulsion between AgNPs, ning bactericidal properties [77]. Antimicrobial exercises of silver
thereby preventing agglomeration of nanoparticles [62,63]. Typical settles on it much intriguing decision for usages in various fields
polymer-based capping agents are polyvinyl alcohol (PVA), including, industrial sectors, biomedical applications, tissue engi-
polyvinylpyrrolidone (PVP), polyethylene glycol (PEG) and neering, food industries, safety concerns, environmental applica-
polysaccharides, while non-ionic surfactants including Brij, Tween, tions etc. [67,78].
and Triton X-100 are employed to stabilize AgNPs during the for- A wide range of strategies have been created for union of AgNPs
mation process [63]. These stabilizers also play important roles including physical, synthetic and green (natural) methods (Fig. 1).
in controlling the size and shape of AgNPs. The properties and bac- In all strategies, settled NPs are shaped by decreasing of the silver
tericidal activities of AgNPs are greatly influenced by their size and particles to silver components utilizing lessening specialists took
shape. Thus, researchers have spent much effort on the size- and after by nucleation and developing procedures [79–81]. In various
shape-controlled synthesis of AgNPs by using different capping physical and synthetic strategies associated with the formation of
agents. Organic solvents have some advantages over aqueous solu- NPs, it is conceivable to get fabricated materials having desired
tions such as high yield and narrow particle size distribution. In potentials. However, the utilization of costly and dangerous mate-
certain cases, solvent itself can also serve as a reducing agent rials for their production trails them as unconvinced strategies
[64]. As an example, the polyol process utilizing EG as both solvent [82,83]. Green union of AgNPs by microorganisms and plant
and reductant in the presence of PVP at 160 °C is known to be removes as an option practical blend system which has increased
effective to reduce silver nitrate to yield AgNPs [65]. In addition, much consideration and application nowadays. Different plant-
759
S. Anees Ahmad, S. Sachi Das, A. Khatoon et al. Materials Science for Energy Technologies 3 (2020) 756–769

in the formation of NPs blend [68]. Utilizing plant concentrates can


likewise be beneficial over microorganisms for NPs blend, by end
of the distinguishing proof, confinement, and keeping up cell soci-
eties forms for microorganisms [87]. Some of the significant
approaches have been summarized in Tables 1 and 2.

3. Antibacterial mechanism of AgNPs

The antibacterial properties of AgNPs mostly depends upon the


size, pH and ionic strength of the medium, and also on the type of
capping agent. However, the specific mechanism of antibacterial or
toxicity activities by the AgNPs is still indefinite and has not been
Fig. 1. Various approaches for the synthesis and development of AgNPs. completely explained. Perhaps, the AgNPs could frequently release
the silver ions (Ag+), which might be considered as one of the
mechanism behind the bactericidal activity of AgNPs [125]. The
based metabolites such as carbohydrates, alkaloids, terpenoids, positively charged Ag+ play a vital role to exhibit the antibacterial
tannins, phenolic acids, flavonoids, proteins, and others, plays sig- or toxicity activities of the silver (Ag) and to maintain its antibac-
nificant role in the fabrication of AgNPs from the Ag materials [84]. terial or toxicity activities, the Ag essentially should be in its ion-
The polyol parts, polysaccharides and aqueous-solvent hetero- ized state [126]. It has been noticed that the Ag+ ions forms
cyclic blends are the most important plant constituents that are complexes with the nucleic acids and as compared to the phos-
primarily responsible for the reduction of Ag particles, followed phate groups, it specifically interacts with the nucleosides of
by the formation of AgNPs. In green combination of AgNPs utilizing nucleic acids. Therefore, all types of silver or silver comprising
plant separates, a few components including plant sources, sorts of composites having antibacterial activities are ultimately the
natural mixes in the rough leaf remove, convergence of beginning sources of Ag+ ions [127,128]. In some of the works, the electro-
silver particles, temperature and the sort and centralization of leaf static attraction amongst the negatively charged microbial cells
concentrate shades are the vital variables on the effectiveness of and positively charged NP have been showed [129] and these
AgNPs creation prepare [85]. In correlation with physical and con- nanoparticulate systems have been proposed as the most apt bac-
coction techniques, amalgamation of NPs by plant concentrates is tericidal agents [130,131]. Due to the electrostatic attractions and
financially savvy and eco-accommodating strategy which allows affinity towards the sulfur proteins, the Ag+ ions adheres to the
the objective of formation of NPs which are apt for human uses cytoplasm and cell wall, and significantly enhances the permeabil-
[86]. This technique can be utilized as a monetary and significant ity; leading to the disruptions of bacterial casings [132]. As soon as
option for the huge scale creation of AgNPs. Moreover, separates the free Ag+ ions are uptaken by the cells, the respiratory enzymes
from plants might perform as reducing as well as stabilizing agents gets deactivated, leading to the production of reactive oxygen spe-

Table 1
Antimicrobial activities of plant extracted silver nanoparticles.

Plant (extracts) Particle size (nm) Remark References


Brassica oleracea extract 42–83 Antibacterial action against Escherichia coli and Staphylococcus aureus [88]
Carica papaya extract 15 Antibacterial action against Escherichia coli [89]
Trianthemadecandra extract 10 Antibacterial action against Escherichia coli [90]
Garcinia mangostana extract 1–100 Antibacterial and antioxidant activity action against E. coli and S. aureus [91]
Withania somnifera extract 5–30 Antimicrobial activity against S. aureus, E. coli, Candida albicans, Aspergillus niger, [92]
and Aspergillus flavus
Acorus calamus extract 31.83 Antibacterial activity against Bacillus subtilis, Bacillus cereus, and S. aureus [93]
Ocimum tenuiflorum extract 25–40 Antibacterial activity against E. coli and B. subtilis [94]
Sensitivum portulacastrum 5–20 Antibacterial and antifungal activity against S. aureus, Listeria monocytogenes, and [95]
extract Penicilliumitalicum
Tribuluster restris extract 16–28 Antibacterial activity against B. subtilis, E. coli, and S. aureus [96]
Cocos nuciferain florescence 22 Antimicrobial activity against Salmonella paratyphi and B. subtilis [97]
extract
Lantana camara extract 12.55–12.99 Antibacterial activity against B. subtilis [98]
Cymbopogon citrates extract 32 Antibacterial effect against E. coli, Shigella flexneri, and S. Sonnei [99]
Boerhaavia diffusa extract 25 Antibacterial activity against Flavobacterium [100]
Cassia roxburghii extract 10–30 Antibacterial activity against B. subtilis [101]
Musa paradisiaca peel extract 23.7 antibacterial activity against B. subtilis, E. coli, and S. aureus [102]
Afzelia quanzensis bark extract 10–80 Antimicrobial activity of against E. coli and S. aureus [103]
Eucalyptus globules extract 1.9 to 4.3 and 5–25 Antimicrobial activity against E. coli and S. aureus [104]
Origanum vulgare extract 63–85 Antibacterial activity against E. coli, S. dysenteriae, S. typhi, and Shigellasonnei [105]
Pulicaria glutinosa extract 40–60 Antibacterial activity against E. coli and S. aureus [106]
Petroselinum crispum leaf extract 30–32 Antibacterial activity against E. coli and S. aureus [107]
Dioscorea bulbifera extract 8–20 Antibacterial activity against E. coli, B. subtilis, S. typhi, and S. aureus [108]
Mangosteen leaf extract 35 Antimicrobial activity against E. coli and S. aureus [109]
Prunus japonica leaves extract 26 Medicinal and pharmaceutical applications [110]
Rheum palmatum root extract 121 Antimicrobial activity against S. aureus and P. aeruginosa [111]
Phoenix dactylifera extract 15–40 Antibacterial activity against E. coli [112]
Phyllanthus emblica extract 40 Antibacterial activity against Acidovoraxoryzae strain RS-2 [113]
Rosa indica leaves 50–100 Antibacterial activity against P. aeruginosa and B.subtilis [114]
Taraxacum officinale leaf extract 15 Antibacterial activity against Xanthomona saxonopodis and P. syringae. [115]

760
S. Anees Ahmad, S. Sachi Das, A. Khatoon et al. Materials Science for Energy Technologies 3 (2020) 756–769

Table 2
Antimicrobial activities of some microbes-mediated silver nanoparticles.

Bacterial Species Particle Major Outcomes References


size (nm)
Glucono bacterroseus 10 The AgNPs exhibited dose-dependent antiplatelet pattern against the ADP-induced activated blood platelets. [116]
The AgNPs conferred no toxicity.
Bacillus flexus 12–65 The synthesized AgNPs improved the efficiency of the antibacterial activity against multi-drug resistant [117]
(MDR) microbes.
Staphylococcus epidermidis 10–80 Metallic NPs synthesized from the supernatants of the bacterial cultures within 24 hours and the production [118]
rate was comparatively high at 50 °C.
CorynebacteriumSH09 10–15 The bio-sorption of the diamine Ag-complex was superior than that of Ag+ ions. [119]
Morganella species 10–40 As per study, the molecular mechanism of Ag resistance and its gene products (silE, silP and silS) showed a [120]
vital role in the entire synthesis procedure of AgNPs by Morganella sp.
Ureibacillus 10–100 The biosynthesis of AgNPs from the supernatants of bacterial strains were found to be maximum at the [121]
thermosphaericus raised temperatures and higherAg+ ion concentrations.
ShewanellaoneidensisMR-1 2–20 Results showed that the presence of bacterial cells was necessary for the development of AgNPs. Also, the [122]
reactions conditions effected the yield of AgNPs significantly, however did not effected the size very
effectively.
Geobacillus 5–35 Increased stability of the formed AgNPs was accredited to the release of certain capping proteins from the [123]
stearothermophilus bacterial cells present in the reactions mixture.
Bacillus safensis LAU 13 5–30 The AgNPs exhibited significant inhibitory activities against the clinical substrates of the bacterial strains of [124]
Escherichia coli. Also, the keratinase of the bacterial strain was found to be apt for the preparation of AgNPs.

cies (ROS) and interruption in adenosine triphosphate (ATP) potentially dephosphorylate the tyrosine residuals over the pep-
release [133]. tide substrates. Furthermore, the disruption of microbial signal
ROS could be a vital agent in the aggravation of the disruption of transduction could significantly cause to cell apoptosis and inhibi-
the cellular membrane and alteration in the deoxyribonucleic acid tion of cell propagation (Fig. 3). One of the major limitation of the
(DNA). DNA majorly comprises of phosphorus and sulfur and when currently available antibacterial or antimicrobial agents is resistant
its gets interacted with the Ag+ ions, could cause different issues of bacteria against these agents [136]. This is mainly due to anti-
related to DNA, including DNA replication, cell propagation and genic alterations in the bacteria which are incompetently accom-
others (Fig. 2). Additionally, the Ag+ ions could efficiently hinder plished with the current treatments. The resistance to
the protein synthesis by denaturing cytoplasmic ribosomal compo- antibacterial or antimicrobial agents by the bacteria or microbes
nents [134]. Apart from the ability to release the Ag+ ions, the has turn out to be a severe issue and therefore, is has become very
AgNPs could themselves eradicate the microbes or bacteria. The important to develop novel therapeutic approaches. The research-
amassed AgNPs trigger denaturation of the cell membranes and ers and formulators have explored the therapeutic potentials of Ag-
due to nanoscale size they have the capability to permeate through based nanoparticulate systems to overcome these issues. The main
the cell wall of bacteria and consequently modify the cell mem- mechanism behind the includes the interaction of Ag+ ions with
brane arrangements [135]. The organelles might get ruptured the disulphide bonds of the protein clusters or glycoproteins of
due to denaturation of cell membrane, and also might cause cell pathogens, leading to alterations in the three dimensional struc-
lysis. Also, the AgNPs could be included in microbial signal trans- tures of the protein [137,138]. The antibacterial efficacy and mech-
duction. The microbial signal transduction is impacted by phos- anism of the AgNPs are affected by their dissolution profile in the
phorylation of protein substrates, and the AgNPs could reaction media. The dissolution efficacy is highly dependent upon

Fig. 2. Probable mechanism of action of silver nanoparticles as antimicrobial agent [118] (Reproduced from Pareek et al., 2018).

761
S. Anees Ahmad, S. Sachi Das, A. Khatoon et al. Materials Science for Energy Technologies 3 (2020) 756–769

Fig. 3. Cell apoptosis in bacteria in presence of AgNPs [120] (Reproduced from Riaz Ahmed et al., 2017).

the synthesis and processing parameters, including intrinsic char- more rapidly in the acidic media [142]. Studies have showed that
acteristics of AgNPs and the dissolution media [139]. the gram-negative (G ve) bacteria are more liable to AgNPs than
Apart from these, the size and shape of the AgNPs influences the the gram-positive (G+ve) bacteria. The cell wall of G-ve bacteria
production of Ag+ ions and this phenomenon is described theoret- is more tapered as compared to G+ve strains (Fig. 4). The thick cell
ically by the Ostwald–Freundlich equation. According to it, the wall decrease the diffusion of the AgNPs into the cellular environ-
AgNPs having small size and sphere-shaped or quasi-spherical ment [143]. The diverse antibacterial activities of AgNPs over the
arrangement are more susceptible to the release, of Ag+ ions owing G ve and G+ve bacteria suggested that the uptake of AgNPs is
to their greater surface area [140]. Thus, the aggregation of AgNPs essential for effective antibacterial or antimicrobial action [139].
reduces the the release of Ag+ ions and this issue could be resolved As above-mentioned, G+ve pathogens are composed of very thick
with the usage of capping agents, which effectively modifies the cell wall comprised pf various peptidoglycan coats, thus serves as
surfaces of AgNPs, and thus could improve the dissolution activi- a barrier for the permeation of the Ag+ ions through cytoplasmic
ties of the AgNPs [141]. Addition to these inherent properties of membranes. However, G ve pathogens are mostly comprised of
AgNPs, the surrounding media also influences the release of Ag+ only a single peptidoglycan coat, thus Ag+ ions easily penetrate
ions. The existence of organic or inorganic constituents in the sur- into the cytoplasm and causes cell lysis.
rounding media affects the dissolution of AgNPs by accumulating The antibacterial effects of AgNPs is also influenced by the pan-
with AgNPs or forming complexes with the Ag+ ions. Moreover, icles properties such as shape, size, surface charge, dosing and dif-
as compared to the neutral media, the AgNPs releases the Ag+ ions fusion state [56,144–147]. It is usually recognized that AgNPs

Fig. 4. Probable bactericidal mechanism of AgNPs in G+ve and G ve bacteria [127] (Reproduced from Duval et al., 2019).

762
S. Anees Ahmad, S. Sachi Das, A. Khatoon et al. Materials Science for Energy Technologies 3 (2020) 756–769

smaller than 10 nm could directly modify cell penetrability, go into spect the surface modification of AgNPs through chemical func-
the bacterial cells and initiate cell lysis. Generally, the formation of tionalization has gained much considerations. Functionalized NPs
biofilm in the oral environment hinders the transport of Ag+ ions or could be useful in numerous areas such as medical, engineering,
AgNPs and ultimately the bacteria gets protected from the effects and biological uses [172–180]. It could be categorized into two dis-
of Ag+ ions and AgNPs [148]. Apart from these, some other mech- tinct groups, covalent and non-covalent type of binding physisorp-
anisms are also reported, including interaction of Ag+ ions with tion. Although in the physisorption process, the conjugations are
the biological macromolecules (enzymes and DNA), based on the merely weak where the ligands and the nanomaterials are conju-
mechanism of electron-discharge or free radical generation [149]. gated through weak electrostatic bonds, hydrogen bonds and
As per the reported studies, AgNPs triggers the inhibition of both hydrophobic interfaces. This property of the surface functionalisa-
protein synthesis and cell wall synthesis, evidenced by the amass- tion is helpful for the formation of surface layers for stabilizing the
ing of envelope protein precursor or disruption of the outer cellular discrete NPs in the solution medium. However, the chemisorption
membrane, leading to ATP leakage [150]. has showed greater potentials than the non-covalent bonding
developed by physisorption. Furthermore, the natural polysaccha-
3.1. Antibacterial applications of AgNPs rides could significantly improve the surface functionalization of
AgNPs [181–186]. Most of the surface functionalization approaches
AgNPs have been showed wide applications in the treatment are centred upon the formation of covalent bonds. This method
and management of injuries, burns and microbial infections offers stronger bonds, allowing an increase in the stability of the
[151,152]. Moreover, different salts of Ag and their by-products ligands attached with the surface of the NPs, thus makes the asso-
are significantly used as bactericidal agents [153]. Abundant study ciation stronger. Furthermore, the functional groups existing on
has been performed which reports the bactericidal activities of the the ligands, reacts with the NPs and through chemisorption
AgNPs. AgNPs have also been explored as a tool for the delivery of approach attach to the surface of the NPs and yields self-
antibiotics, production of complexes used as sterilizing filters and assembled arrangements [187,188]. Surface modifications of the
encasing materials [138,154–156]. Most importantly, due to smal- AgNPs might be helpful for the development of novel and fascinat-
ler size and greater surface to volume proportions, the AgNPs holds ing bactericidal agents. Also, some of the studies have been
an extensive contact area with the microbes. These properties of reported in which people have developed distinctive protein-
AgNPs significantly enhances their biological and chemical proper- based natural antibacterial systems such as lysozyme-based and
ties, and makes them a robust bactericidal material. The AgNPs also peptide-based materials with improved efficacy to inhibit bacterial
have the capability to interrupt the cellular functions, including the growth [189,190]. These systems usually caused cell lysis in bacte-
cell permeation and respiration. Additionally, the AgNPs when ria and hence the bacteria find it perplexing to achieve resistance
reach within the cell membranes, potentially disrupts the cellular against these developed and modified bactericidal agents.
components by reacting with the sulphur-mediated proteins and AgNPs, due to their killing activities against bacteria, are recog-
phosphorus-mediated complexes like deoxyribonucleic acid nized for their remarkable antibacterial activities. Moreover, the
(DNA) [157–160]. bactericidal activities of the AgNPs are mostly related with the
Even though the AgNPs have exhibited potential bactericidal direct interaction of the NPs to the cell wall of bacteria, trailed
activities, yet they have been also reported with certain limita- by permeating into the cytoplasmic fluid and this could damage
tions, including toxicity against cells/tissues, particularly the mam- the membrane which causes leaking of the cellular component
malian cells [161–163]. Though, current studies have revealed that and ultimately cellular death [191,192]. In one of the study, it
the conjugation of AgNPs with bactericidal agents might decrease was represented the AgNPs (~10 nm) attached to the microbial cell
the toxic effect towards the mammalian cells and simultaneously wall, thus interrupted the penetrability and cellular metabolic
improve their bactericidal activities synergistically [164–166]. Fur- pathways and caused cell death [191]. Moreover, the generation
thermore, the amalgation of bactericidal agents with AgNPs in a of ROS has been found to be the major cause for bacterial death
suitable fraction would reduce the loading amount of both the con- as it promotes lipid peroxidation, however hinders the ATP gener-
stituents and thus with reduced dosage also they would be able to ation and DNA replication. Also, the increased level of ROS in the
show greater antibacterial efficacy. Also, the conjugation leads to bacterial cells could lead to oxidative stress in these cells [193].
an increase in the amount of bactericidal agents or antibiotics at In general, the AgNPs that are finely dispersed in the physiological
the specific bacterial sites and thus enhances the therapeutic media, exhibited more anti-bacterial efficiency than that of the
potential of the antibiotics against the bacterial infections [167]. agglomerated AgNPs. Furthermore, it has been observed that the
Apart from this, an alternative approach to overcome the toxic- killing effects of the AgNPs against G ve and G+ve bacteria
ity issues of the AgNPs is the potential usages of peptide/peptide- enhanced with a decrease in the size of the AgNPs [194,195]. In
based linkages with AgNPs. The interactions between the metallic another study, the anti-bacterial activity of the sphere-shaped
ions and the biomolecules such as proteins, peptides, amino acids and rod-shaped AgNPs against G+ve and G ve bacterial strains
and others, have showed significant effects over the therapeutic were investigated. Results showed that the antibacterial activity
potential of the metallic NPs. These biomolecules also exhibit vital of both the sphere-shaped and rod-shaped AgNPs followed a
role for the effective delivery of the conjugated metallic NPs dose- dependent and time-dependent pattern. Also, the sphere-
through various biological paths such as electron transmission shaped AgNPs were found to be more efficient as compared to
responses, metal transportation and storing. Furthermore, these rod-shaped AgNPs against the Klebsiella bacterial strain [196].
conjugates metallic systems significantly inhibit the enzymes The antibacterial efficacy of the AgNPs is highly effected by the sur-
which play crucial role in bacterial metabolism. This process is car- ficial charge over the NPs and in this context various studies have
ried over by ligand interchange responses where the ligand exist- been reported for the use of the capping agents as well as stabiliz-
ing with the loaded drug moiety is replaced by the specific ers which prevents accumulation of the AgNPs, and thus certainly
enzyme that acts as a primary source for various metal-mediated influences the surface charge over these NPs [197,198]. In one of
systems. Thus, it has been reported as one of the most significantly the study, the significant effect of surface charge of AgNPs coated
applied method in bio-inorganic chemistry [168–171]. with polyvinyl pyrolidone (PVP), citrate (CT) and branched-
Moreover, the modifications in AgNPs surface could also assist polyethyleneimine (BPEI) over the antibacterial efficacy against
in modifying their features to ensemble numerous applications the bacillus strains. Results revealed that the positively charged
and govern the interactions of the constituents, and in this pro- AgNPs exhibited the maximum antibacterial activity against all
763
S. Anees Ahmad, S. Sachi Das, A. Khatoon et al. Materials Science for Energy Technologies 3 (2020) 756–769

the bacterial species, while negatively charged AgNPs exhibited the icity of AgNPs caused modifications in the organs of the mammals
least and neutrally-charged AgNPs showed transitional bacterici- that were related with their propagative and growth processes
dal activity [199]. Apart from these, certain modification with car- [226]. They also reported that the oxidative stress and inflamma-
bon nanotubes and quantum dots have also improved the tion due to the formation of ROS were the intervening factors of
therapeutic efficacy of these NPs [200–208]. The Ag-based disinfec- cytotoxicity. Moreover, the probable interaction of AgNPs with dif-
tants have been widely used and has exhibited utmost potentials ferent environmental constituents might considerably alter the
in treating the resistance caused by the microbes against effective toxicity profile of the AgNPs. Apart from this, toxicity of AgNPs
antibacterial treatments. The Ag-based antiseptics have greater was found to be widely assessed amongst various microorganisms
reactivity and binding efficacy with the cellular proteins, so allows of medical concern [136,227,228].
for alterations inside the assembly of cellular membrane and Studies have demonstrated that the cytotoxicity of AgNPs
nuclear membrane, and finally causes disruptions and cell death mostly depends upon its physical and chemical properties, and
[208]. also the microorganism type against which the toxicity is assessed
The AgNPs are also used in the food industries during the pro- [229]. AgNPs produced from the green methods are usually less
cess of packing, as they prevent destruction of the nutrients or toxic than those attained from the non-green methods. Application
foodstuffs by the pathogens [209]. One of the most important of green methods has abridged the usage of perilous chemical reac-
properties of the AgNPs or AgNPs-based biomaterials is that their gents and enhanced the synthesis efficiency of AgNPs through
essential antibacterial activity is exhibited against both the plank- chemical processes [230–232]. Moreover, the toxicity of AgNPs is
tonic and biofilm prearranged microbes. Apart from this the pro- generally observed due to the stimulation of oxidative stress or
gressive strategies to improve the bactericidal efficacy of the production of reactive oxygen species (ROS) in the system [193].
AGNPs could be established by the method of bio- AgNPs produced from the green methods are usually more toxic
functionalization by using some bactericidal peptides (like poly- than those attained from the non-green methods. Although, the
myxin B), bovine serum albumin (BSA), and others [210]. Bio- development and application of AgNPs has ensued in public cog-
functionalization improves the bactericidal activity, bioactivity nizance related to their toxicity and environmental impacts
and protects the surface of the AgNPs from microbial attack [211]. [233,234]. Interestingly, a contrasting fact has been observed
The Ag-based nanocomposites have also showed significant related to the dose of AgNPs indorsing toxicity. In some studies,
antibnacterial actions as that of AgNPs [212–214]. Chen et al. the AgNPs of particle size >50 nm reduced the [235] sustainability
developed a graphene oxide-silver nanocomposites (GO-Ag NCs) in the tested human mesenchymal cells at a lower dose of 10 lg/
and evaluated their antibacterial efficacy against multidrug- mL, however in an another study no toxicity was reported even
resistant E. coli strains. The GO-Ag NCs exhibited synergistic acticv- at a higher dose of 100 lg/mL. Also, the stability of the samples
ity with improved antibacterial efficacy. Additionally, the pho- has showed significant role in an upsurge in toxicity by the stored
tothermal effect for efficient sterilization was attained through AgNPs (water for 6 months) which is correlated with the release of
these nanocomposites [215]. The AgNPs adsorbed over graphene Ag+ ions [236].
or carbon nanotubes could efficiently enhance the antibacterial
activity of AgNPs [216–218]. In a study, a simplistic glucose-
mediated one-step approach was established for the production 5. Conclusion and future perspectives
AgNPs incorporated within carbon nanotubes (AgNPs-CNTs). The
nanocomposites were developed in the presence of CNTs (nano- The potential advantages of nanotechnology in modern
reactor), silver nitrite (precursor) and glucose (reductant). The biomedical applications are broadly acknowledged and have found
AgNPs were within the particle range of 6–10 nm and were uni- to be most encouraging segment for era in developing new bacte-
formly assembled within the CNT channels. Furthermore, the ricidal agents for treating bacterial infections. It is presently evi-
AgNPs-CNTs exhibited an improved antibacterial activity dent that AgNPs have a solid bactericidal and antiviral activities,
against E. coli strains [219]. emphasized by a few reviews. AgNPs can collaborate with different
microorganisms, (for example, microscopic organisms) and fur-
thermore affect both the development of and develop bacterial bio-
4. Toxicity of AgNPs films and, in this way, could be utilized as expansive range
bactericidal agents. Despite the fact that lipopolysaccharides are
AgNPs are amongst the most extensively used metal NPs and made out of lipids covalently conjugated with the polysaccharides,
has played crucial role in different field of biomedical and pharma- an absence of inflexibility of the general basic envelope is noticed.
ceutical applications. The smaller size of the AgNPs allows them for Also, the negative charge over the lipopolysaccharides are pulled in
easy passage through the biological membranes and reach within to the frail positive charges of AgNPs. Then again, the cell mass of
the cells, producing toxicity at various levels based upon the exag- Gram+ve microbes is essentially made out of a thick peptidoglycan
gerated microorganisms. The toxicity of the AgNPs is found to be layer comprised of chains of polysaccharides interlinked with short
associated with its features like size, shape, amount, accumulation, peptides to frame three-dimensional inflexible structures. The
surface charge, and the methods applied for synthesis [220,221]. solidness and the broad cross-connecting not only decrease the
Also, the toxicity of AgNPs depends upon the category of targeted bacterial cell divider tying down destinations for AgNPs addition-
microbe, which is correlated with the resistance mechanism of the ally render the divider itself more troublesome to enter. A multi-
microbe used eradicate the unwanted complexes. Furthermore, the faceted system against microorganisms appears to be because of
bacterial response in the toxicity assessments is highly effected by NPs communications with the bacterial surfaces, and in addition
the culture media, in which the bacterial species are exposed [222– to their specific structure. These days, there is significant need
224]. Primarily, both the AgNPs and Ag+ ions released from the for establishment of specific instruments characterization of AgNPs
AgNPs are presumed to employ toxicity by stimulating membrane and their activities. Also, apt research is essentially required on the
impairment, ROS generation, oxidation and denaturation of pro- biocompatibility and bioactivity of the AgNPs. This review on the
teins, dysfunction of mitochondria, damage of DNA and obstruc- consolidated utilization of AgNPs with other antimicrobial opera-
tion of cell propagation [136,225]. tors could definitely help lessen the issue of lethality and to keep
Ema et al. reviewed toxicity mechanism of AgNPs in the mam- away from the potential for improvement of resistance and, most
mals, particularly the rabbits and rats, and described that the tox- importantly, unequivocally upgrade the bactericidal impacts. The
764
S. Anees Ahmad, S. Sachi Das, A. Khatoon et al. Materials Science for Energy Technologies 3 (2020) 756–769

expansive range of bioactivity of AgNPs makes them promising [24] Y. Li, P. Leung, L. Yao, Q. Song, E. Newton, Antimicrobial effect of surgical
masks coated with nanoparticles, J. Hosp. Infect. 62 (2006) 58–63.
operators to battle contaminations, as well as in numerous other
[25] P. Jain, T. Pradeep, Potential of silver nanoparticle-coated polyurethane foam
biomedical regions. as an antibacterial water filter, Biotechnol. Bioeng. 90 (2005) 59–63.
[26] M.T. Ansari, N.B. Ibrahim, F. Sami, S. Majeed, M.S. Hasnain, V.B. Badgujar,
Design and evaluation of topical herbal antifungal stick containing extracts of
Declaration of Competing Interest Rhinacanthus nasutus, J. Herb. Med. 100290 (2019).
[27] W.R. Li, X.B. Xie, Q.S. Shi, H.Y. Zeng, O.Y. You-Sheng, Y.B. Chen, Antibacterial
activity and mechanism of silver nanoparticles on Escherichia coli, Appl.
The authors declare that they have no known competing finan-
Microbiol. Biotechnol. 85 (2010) 1115–1122.
cial interests or personal relationships that could have appeared [28] S. Iravani, H. Korbekandi, S.V. Mirmohammadi, B. Zolfaghari, Synthesis of
to influence the work reported in this paper. silver nanoparticles: chemical, physical and biological methods, Res. Pharm.
Sci. 9 (2014) 385–406.
[29] P.A. Sabatier, Top-down and bottom-up approaches to implementation
References research: a critical analysis and suggested synthesis, J. Public Policy 6
(1986) 21–48.
[30] E. Bharathi, G. Sivakumari, J. Kamalakkannan, B. Karthikeyan, S. Senthilvelan,
[1] A.M. Fayaz, K. Balaji, M. Girilal, R. Yadav, P.T. Kalaichelvan, R. Venketesan,
Synergetic execute pressure, temperature on mixed Ac/Ag@CuO and its multi
Biogenic synthesis of silver nanoparticles and their synergistic effect with
properties of solar light elucidation and antibacterial activity by
antibiotics: a study against gram-positive and gram-negative bacteria,
hydrothermal technique, Mater. Sci. Energy Technol. 3 (2020) 407–419.
Nanomed. Nanotechnol. 6 (2010) 103–109.
[31] L. Zhi, K. Müllen, A bottom-up approach from molecular nanographenes to
[2] M. Singh, S. Singh, S. Prasad, I. Gambhir, Nanotechnology in medicine and
unconventional carbon materials, J. Mater. Chem. 18 (2008) 1472–1484.
antibacterial effect of silver nanoparticles, Dig. J. Nanomater. Bio. 3 (2008)
[32] S. Stankovich, D.A. Dikin, G.H. Dommett, K.M. Kohlhaas, E.J. Zimney, E.A.
115–122.
Stach, R.D. Piner, S.T. Nguyen, R.S. Ruoff, Graphene-based composite
[3] K. Kalishwaralal, V. Deepak, S. Ramkumarpandian, H. Nellaiah, G.
materials, Nature 442 (2006) 282–286.
Sangiliyandi, Extracellular biosynthesis of silver nanoparticles by the
[33] S.H. Huang, M. Uppal, J. Shi, A product driven approach to manufacturing
culture supernatant of Bacillus licheniformis, Mater. Lett. 62 (2008) 4411–
supply chain selection, Supply Chain Manage. 7 (2002) 189–199.
4413.
_ [34] W. Lu, C.M. Lieber, Nanoelectronics from the bottom up, Nature Mater. 6
[4] I.H. Boyacı, A new approach for determination of enzyme kinetic constants
(2007) 841–850.
using response surface methodology, Biochem. Eng. J. 25 (2005) 55–62.
[35] A. Budi, A. Basile, G. Opletal, A.F. Hollenkamp, A.S. Best, R.J. Rees, A.I. Bhatt, A.
[5] Y.H. Xiong, J.Z. Liu, H.Y. Song, L.N. Ji, Enhanced production of extracellular
P. O’mullane, S.P. Russo, Study of the initial stage of solid electrolyte
ribonuclease from Aspergillus niger by optimization of culture conditions
interphase formation upon chemical reaction of lithium metal and N-methyl-
using response surface methodology, Biochem. Eng. J. 21 (2004) 27–32.
N-propyl-pyrrolidinium-Bis (Fluorosulfonyl) imide, J. Phys. Chem. C 116
[6] H.S. Shin, H.C. Choi, Y. Jung, S.B. Kim, H.J. Song, H.J. Shin, Chemical and size
(2012) 19789–19797.
effects of nanocomposites of silver and polyvinyl pyrrolidone determined by
[36] V. Balzani, A. Credi, M. Venturi, The Bottom-up approach to molecular level
X-ray photoemission spectroscopy, Chem. Phy. Lett. 383 (2004) 418–422.
devices and machines, Chem.–A Eur. J. 8 (2002) 5524–5532.
[7] S.N. Senanayake, F. Shahidi, Lipase-catalyzed incorporation of
[37] E.M. Smeets, A.P. Faaij, I.M. Lewandowski, W.C. Turkenburg, A bottom-up
docosahexaenoic acid (DHA) into borage oil: optimization using response
assessment and review of global bio-energy potentials to 2050, Prog. Energy
surface methodology, Food Chem. 77 (2002) 115–123.
Combust. 33 (2007) 56–106.
[8] G. Dey, A. Mitra, R. Banerjee, B. Maiti, Enhanced production of amylase by
[38] A. Bausch, K. Kroy, A bottom-up approach to cell mechanics, Nature Phys. 2
optimization of nutritional constituents using response surface methodology,
(2006) 231–238.
Biochem. Eng. J. 7 (2001) 227–231.
[39] N.J. Guido, X. Wang, D. Adalsteinsson, D. Mcmillen, J. Hasty, C.R. Cantor, T.C.
[9] A.I. El-Batal, A.A.M. Hashem, N.M. Abdelbaky, Gamma radiation mediated
Elston, J. Collins, A bottom-up approach to gene regulation, Nature 439 (2006)
green synthesis of gold nanoparticles using fermented soybean-garlic
856–860.
aqueous extract and their antimicrobial activity, SpringerPlus 2 (2013) 129.
[40] O.V. Salata, Applications of nanoparticles in biology and medicine, J.
[10] A.N. Krklješ, M.T. Marinović-Cincović, Z.M. Kacarevic-Popovic, J.M.
Nanobiotechnol. 2 (2004) 3.
Nedeljković, Radiolytic synthesis and characterization of Ag-PVA
[41] S.S. Das, P. Bharadwaj, M. Bilal, M. Barani, A. Rahdar, P. Taboada, S. Bungau, G.
nanocomposites, Eur. Polym. J. 43 (2007) 2171–2176.
Z. Kyzas, Stimuli-responsive polymeric nanocarriers for drug delivery,
[11] M. Guzman, J. Dille, S. Godet, Synthesis and antibacterial activity of silver
imaging, and theragnosis, Polymers 12 (2020) 1397.
nanoparticles against gram-positive and gram-negative bacteria, Nanomed.
[42] J. Hanrieder, A. Nyakas, T. Naessén, J. Bergquist, Proteomic analysis of human
Nanotechnol. 8 (2012) 37–45.
follicular fluid using an alternative bottom-up approach, J. Proteome Res. 7
[12] A. Sadeghi, B. Rahimi, M. Shojapour, Molecular detection of metallo–
(2007) 443–449.
lactamase genes blaVIM-1, blaVIM-2, blaIMP-1, blaIMP-2 and blaSPM-1 in
[43] X. Chen, H. Schluesener, Nanosilver: a nanoproduct in medical application,
Pseudomonas aeruginosa isolated from hospitalized patients in Markazi
Toxicol. Lett. 176 (2008) 1–12.
province by Duplex-PCR, Afr. J. Microbiol. Res. 6 (2012) 2965–2969.
[44] A. Solanki, J.D. Kim, K.B. Lee, Nanotechnology for regenerative medicine:
[13] S. Liau, D. Read, W. Pugh, J. Furr, A. Russell, Interaction of silver nitrate with
nanomaterials for stem cell imaging, Nanomedicine 3 (2008) 567–578.
readily identifiable groups: relationship to the antibacterialaction of silver
[45] Y. Sun, Y. Xia, Shape-controlled synthesis of gold and silver nanoparticles,
ions, Lett. Appl. Microbiol. 25 (1997) 279–283.
Science 298 (2002) 2176–2179.
[14] K. Nomiya, A. Yoshizawa, K. Tsukagoshi, N.C. Kasuga, S. Hirakawa, J.
[46] D. Kim, S. Jeong, J. Moon, Synthesis of silver nanoparticles using the polyol
Watanabe, Synthesis and structural characterization of silver (I), aluminium
process and the influence of precursor injection, Nanotechnology 17 (2006)
(III) and cobalt (II) complexes with 4-isopropyltropolone (hinokitiol) showing
4019.
noteworthy biological activities. Action of silver (I)-oxygen bonding
[47] M. Chen, Y.G. Feng, X. Wang, T.C. Li, J.Y. Zhang, D.J. Qian, Silver nanoparticles
complexes on the antimicrobial activities, J. Inorg. Biochem. 98 (2004) 46–60.
capped by oleylamine: formation, growth, and self-organization, Langmuir 23
[15] A. Gupta, Silver as a biocide: will resistance become a problem?, Nature
(2007) 5296–5304.
Biotechnol. 16 (1998) 888–898.
[48] S.F. Chen, H. Zhang, Aggregation kinetics of nanosilver in different water
[16] Q. Feng, J. Wu, G. Chen, F. Cui, T. Kim, J. Kim, A mechanistic study of the
conditions, Adv. Nat. Sci. Nanosci. Nanotechnol. 3 (2012) 035006.
antibacterial effect of silver ions on Escherichia coli and Staphylococcus aureus,
[49] T.M.D. Dang, T.T.T. Le, E. Fribourg-Blanc, M.C. Dang, Influence of surfactant on
J. Biomed. Mater. Res. 52 (2000) 662–668.
the preparation of silver nanoparticles by polyol method, Adv. Nat. Sci.
[17] M.J. Yacamán, J. Ascencio, H. Liu, J. Gardea-Torresdey, Structure shape and
Nanosci. Nanotechnol. 3 (2012) 035004.
stability of nanometric sized particles, J. Vac. Sci. Technol. 19 (2001) 1091–
[50] R.S. Patil, M.R. Kokate, C.L. Jambhale, S.M. Pawar, S.H. Han, S.S. Kolekar, One-
1103.
pot synthesis of PVA-capped silver nanoparticles their characterization and
[18] G.A. Somorjai, On the move, Nature 430 (2004) 730.
biomedical application, Adv. Nat. Sci. Nanosci. Nanotechnol. 3 (2012) 015013.
[19] M. Haruta, Size-and support-dependency in the catalysis of gold, Catal. Today
[51] D.K. Le, Y.S. Kang, Synthesis of silver nanocrystallites by a new thermal
36 (1997) 153–166.
decomposition method and their characterization, Etri J. 26 (2004) 252–256.
[20] S.V. Kyriacou, W.J. Brownlow, X.H.N. Xu, Using nanoparticle optics assay for
[52] J. Jung, H. Oh, H. Noh, J. Ji, S. Kim, Metal nanoparticle generation using a small
direct observation of the function of antimicrobial agents in single live
ceramic heater with a local heating area, J. Aerosol. Sci. 37 (2006) 1662–1670.
bacterial cells, Biochemistry 43 (2004) 140–147.
[53] T. Tsuji, K. Iryo, N. Watanabe, M. Tsuji, Preparation of silver nanoparticles by
[21] A. PanáčEk, L. Kvítek, R. Prucek, M. Kolář, R. VečEřová, N. Pizúrová, V.K.
laser ablation in solution: influence of laser wavelength on particle size, Appl.
Sharma, T.J. NevěčNá, R. Zbořil, Silver colloid nanoparticles: synthesis,
Surf. Sci. 202 (2002) 80–85.
characterization, and their antibacterial activity, J. Phys. Chem. B 110
[54] B. Perito, E. Giorgetti, P. Marsili, M. Muniz-Miranda, Antibacterial activity of
(2006) 16248–16253.
silver nanoparticles obtained by pulsed laser ablation in pure water and in
[22] R.J. Holladay, W. Moeller, D. Mehta, R. Roy, J.H. Brooks, M.G. Mortenson,
chloride solution, Beilstein J. Nanotechnol. 7 (2016) 465–473.
Silver/water, silver gels and silver-based compositions; and methods for
[55] M.C. Sportelli, M. Izzi, A. Volpe, M. Clemente, R.A. Picca, A. Ancona, P.M.
making and using the same. Google Patents, 2005.
Lugara, G. Palazzo, N. Cioffi, The pros and cons of the use of laser ablation
[23] J.R. Morones, J.L. Elechiguerra, A. Camacho, K. Holt, J.B. Kouri, J.T. Ramírez, M.J.
synthesis for the production of silver nano-antimicrobials, Antibiotics 7
Yacaman, The bactericidal effect of silver nanoparticles, Nanotechnology 16
(2018) 67.
(2005) 2346.

765
S. Anees Ahmad, S. Sachi Das, A. Khatoon et al. Materials Science for Energy Technologies 3 (2020) 756–769

[56] M.A. Raza, Z. Kanwal, A. Rauf, A.N. Sabri, S. Riaz, S. Naseem, Size- and shape- [83] G. Geoprincy, B.V. Srri, U. Poonguzhali, N.N. Gandhi, S. Renganathan, A review
dependent antibacterial studies of silver nanoparticles synthesized by wet on green synthesis of silver nanoparticles, Asian J. Pharm. Clin. Res. 6 (2013)
chemical routes, Nanomaterials 6 (2016) 74. 8–12.
[57] H.D. Beyene, A.A. Werkneh, H.K. Bezabha, T.G. Ambaye, Synthesis paradigm [84] M. Rahman, A.K. Nayak, S. Beg, M.S. Hasnain, Metallic nanoparticles for drug
and applications of silver nanoparticles (AgNPs), a review, Sustain. Mater. delivery and biomedical applications: patent perspectives, Curr. Nanomed. 8
Technol. 13 (2017) 18–23. (2018) 176.
[58] W. Zhang, X. Qiao, J. Chen, Synthesis of silver nanoparticles-effects of [85] A. Leela, M. Vivekanandan, Tapping the unexploited plant resources for the
concerned parameters in water/oil microemulsion, Mater. Sci. Eng. B 142 synthesis of silver nanoparticles, Afr. J. Biotechnol. 7 (2008) 3162.
(2007) 1–15. [86] K. Velayutham, A.A. Rahuman, G. Rajakumar, S.M. Roopan, G. Elango, C.
[59] G.A. Sotiriou, A. Teleki, A. Camenzind, F. Krumeich, A. Meyer, S. Panke, S.E. Kamaraj, S. Marimuthu, T. Santhoshkumar, M. Iyappan, C. Siva, Larvicidal
Pratsinis, Nanosilver on nanostructured silica: antibacterial activity and Ag activity of green synthesized silver nanoparticles using bark aqueous extract
surface area, Chem. Eng. J. 170 (2011) 547–554. of Ficus racemosa against Culex quinquefasciatus and Culex gelidus, Asian Pac. J.
[60] J. Helmlinger, C. Sengstock, C. Groß-Heitfeld, C. Mayer, T.A. Schildhaue, M. Trop. Med. 6 (2013) 95–101.
Koller, M. Epple, Silver nanoparticles with different size and shape: equal [87] J.Y. Song, H.K. Jang, B.S. Kim, Biological synthesis of gold nanoparticles using
cytotoxicity, but different antibacterial effects, RSC Adv. 6 (2016) 18490– Magnolia kobus and Diopyros kaki leaf extracts, Process Biochem. 44 (2009)
18501. 1133–1138.
[61] K. Ranoszek-Soliwoda, E. Tomaszewska, E. Socha, P. Krzyczmonik, A. [88] V. Sridhara, K. Pratima, G. Krishnamurthy, B. Sreekanth, Vegetable assisted
Ignaczak, P. Orlowski, M. Krzyzowska, G. Celichowski, J. Grobeln, The role synthesis of silver nanoparticles and its antibacterial activity against two
of tannic acid and sodium citrate in the synthesis of silver nanoparticles, J. human pathogens, Asian J. Pharm. Clin. Res. 6 (2013) 53–57.
Nanopart. Res. 19 (2017) 273. [89] D. Jain, H.K. Daima, S. Kachhwaha, S. Kothari, Synthesis of plant-mediated
[62] L. Malassis, R. Dreyfus, R.J. Murphy, L.A. Hough, B. Donnio, C.B. Murray, One- silver nanoparticles using papaya fruit extract and evaluation of their anti
step green synthesis of gold and silver nanoparticles with ascorbic acid and microbial activities, Dig. J. Nanomater. Biostruct. 4 (2009) 557–563.
their versatile surface post-functionalization, RSC Adv. 6 (2016) 33092– [90] R. Geethalakshmi, D. Sarada, Synthesis of plant-mediated silver nanoparticles
33100. using Trianthema decandra extract and evaluation of their anti microbial
[63] L. Kvitek, A. Panacek, J. Soukupova, M. Kolar, R. Vecerova, R. Prucek, M. activities, Int. J. Eng. Sci. Technol. 2 (2010) 970–975.
Holecova, R. Zboril, Effect of surfactants and polymers on stability and [91] S. Rajakannu, S. Shankar, S. Perumal, S. Subramanian, G.P. Dhakshinamoorthy,
antibacterial activity of silver nanoparticles (NPs), J. Phys. Chem. C 112 (2008) Biosynthesis of silver nanoparticles using Garcinia mangostana fruit extract
5825–5834. and their antibacterial, antioxidant activity, Int. J. Curr. Microbiol. Appl. Sci. 4
[64] Y. Sun, Controlled synthesis of colloidal silver nanoparticles in organic (2015) 944–952.
solutions: empirical rules for nucleation engineering, Chem. Soc. Rev. 42 [92] R.W. Raut, V.D. Mendhulkar, S.B. Kashid, Photosensitized synthesis of silver
(2013) 2497–2511. nanoparticles using Withania somnifera leaf powder and silver nitrate, J.
[65] T.M. Dang, T.T. Le, E. Fribourg-Blanc, M.C. Dang, Influence of surfactant on the Photochem. Photobiol. B 132 (2014) 45–55.
preparation of silver nanoparticles by polyol method, Adv. Nat. Sci. Nanosci. [93] J.R. Nakkala, R. Mata, A.K. Gupta, S.R. Sadras, Biological activities of green
Nanotechnol. 3 (2012) 035004. silver nanoparticles synthesized with Acorous calamus rhizome extract, Eur. J.
[66] N.T. Nguyen, B.H. Nguyen, D.T. Ba, D.G. Pham, T.V. Kahi, L.T. Nguyen, L.D. Tran, Med. Chem. 85 (2014) 784–794.
Microwave-assisted synthesis of silver nanoparticles using chitosan: a novel [94] R.S. Patil, M.R. Kokate, S.S. Kolekar, Bioinspired synthesis of highly stabilized
approach, Mater. Manuf. Process. 29 (2014) 418–421. silver nanoparticles using Ocimum tenuiflorum leaf extract and their
[67] S. Prabhu, E.K. Poulose, Silver nanoparticles: mechanism of antimicrobial antibacterial activity, Spectrochim. Acta A 91 (2012) 234–238.
action, synthesis, medical applications, and toxicity effects, Int. Nano Lett. 2 [95] A. Nabikhan, K. Kandasamy, A. Raj, N.M. Alikunhi, Synthesis of antimicrobial
(2012) 32. silver nanoparticles by callus and leaf extracts from saltmarsh plant,
[68] B. Hu, S.B. Wang, K. Wang, M. Zhang, S.H. Yu, Microwave-assisted rapid facile Sesuvium portulacastrum L, Colloids Surf. B 79 (2010) 488–493.
‘‘green” synthesis of uniform silver nanoparticles: self-assembly into [96] V. Gopinath, D. Mubarakali, S. Priyadarshini, N.M. Priyadharsshini, N.
multilayered films and their optical properties, J. Phys. Chem. C 112 (2008) Thajuddin, P. Velusamy, Biosynthesis of silver nanoparticles from Tribulus
11169–11174. terrestris and its antimicrobial activity: a novel biological approach, Colloids
[69] W.R. Melchert, B.F. Reis, F.R. Rocha, Green chemistry and the evolution of flow Surf. B 96 (2012) 69–74.
analysis. A review, Anal. Chim. Acta 714 (2012) 8–19. [97] R. Mariselvam, A. Ranjitsingh, A.U.R. Nanthini, K. Kalirajan, C. Padmalatha, P.
[70] R. Mie, M.W. Samsudin, L.B. Din, A. Ahmad, N. Ibrahim, S.N.A. Adnan, M. Selvakumar, Green synthesis of silver nanoparticles from the extract of the
Synthesis of silver nanoparticles with antibacterial activity using the lichen inflorescence of Cocos nucifera (Family: Arecaceae) for enhanced antibacterial
Parmotrema praesorediosum, Int. J. Nanomed. 9 (2014) 121. activity, Spectrochim. Acta A 129 (2014) 537–541.
[71] S. Kaviya, J. Santhanalakshmi, B. Viswanathan, J. Muthumary, K. Srinivasan, [98] P. Sivakumar, C. Nethradevi, S. Renganathan, Synthesis of silver nanoparticles
Biosynthesis of silver nanoparticles using Citrus sinensis peel extract and its using Lantana camara fruit extract and its effect on pathogens, Asian J. Pharm.
antibacterial activity, Spectrochim. Acta A 79 (2011) 594–598. Clin. Res. 5 (2012) 97–101.
[72] K. Caswell, C.M. Bende, C.J. Murphy, Seedless, surfactantless wet chemical [99] N. Geetha, T. Geetha, P. Manonmani, M. Thiyagarajan, Green synthesis of
synthesis of silver nanowires, Nano Lett. 3 (2003) 667–669. silver nanoparticles using Cymbopogan citratus (Dc) Stapf. extract and its
[73] K.D. Bhatte, K.M. Deshmukh, Y.P. Patil, D.N. Sawant, S.I. Fujita, M. Arai, B.M. antibacterial activity, J. Basic Appl. 8 (2014) 324–331.
Bhanage, Synthesis of powdered silver nanoparticles using hydrogen in [100] P.V. Kumar, S. Pammi, P. Kollu, K. Satyanarayana, U. Shameem, Green
aqueous medium, Particuology 10 (2012) 140–143. synthesis and characterization of silver nanoparticles using Boerhaavia diffusa
[74] R. Sukirtha, K.M. Priyanka, J.J. Anton, S. Kamalakkannan, R. Thangam, P. plant extract and their anti bacterial activity, Ind. Crops Prod. 52 (2014) 562–
Gunasekaran, M. Krishnan, S. Achiraman, Cytotoxic effect of green 566.
synthesized silver nanoparticles using Melia azedarach against in vitro [101] P. Balashanmugam, P.T. Kalaichelvan, Biosynthesis characterization of silver
HeLa cell lines and lymphoma mice model, Process Biochem. 47 (2012) nanoparticles using Cassia roxburghii DC. aqueous extract, and coated on
273–279. cotton cloth for effective antibacterial activity, Int. J. Nanomed. 10 (2015) 87.
[75] T. Suma, S.R. Rajasree, A. Kanchana, S.B. Elizabeth, Biosynthesis, [102] H.M. Ibrahim, Green synthesis and characterization of silver nanoparticles
characterization and cytotoxic effect of plant mediated silver using banana peel extract and their antimicrobial activity against
nanoparticles using Morinda citrifolia root extract, Colloids Surf. B 106 representative microorganisms, J Radiat. Res. Appl. Sc. 8 (2015) 265–275.
(2013) 74–78. [103] K. Singh, M. Panghal, S. Kadyan, U. Chaudhary, J.P. Yadav, Green silver
[76] V. Vadlapudi, D. Kaladhar, Review: green synthesis of silver and gold nanoparticles of Phyllanthus amarus: as an antibacterial agent against multi
nanoparticles, Middle East J. Sci. Res. 19 (2014) 834–842. drug resistant clinical isolates of Pseudomonas aeruginosa, J. Nanobiotechnol.
[77] V. Thirumalai Arasu, D. Prabhu, M. Soniya, Stable silver nanoparticle 12 (2014) 40.
synthesizing methods and its applications, J. Biol. Sci. Res. 1 (2010) 259– [104] K. Ali, B. Ahmed, S. Dwivedi, Q. Saquib, A.A. Al-Khedhairy, J. Musarrat,
270. Microwave accelerated green synthesis of stable silver nanoparticles with
[78] T.J.I. Edison, M. Sethuraman, Biogenic robust synthesis of silver nanoparticles Eucalyptus globulus leaf extract and their antibacterial and antibiofilm
using Punica granatum peel and its application as a green catalyst for the activity on clinical isolates, PLoS One 10 (2015) e0131178.
reduction of an anthropogenic pollutant 4-nitrophenol, Spectrochim. Acta A [105] M.S. Hasnain, M.N. Javed, M.S. Alam, P. Rishishwar, S. Rishishwar, S. Ali, A.K.
104 (2013) 262–264. Nayak, S. Beg, Purple heart plant leaves extract-mediated silver nanoparticle
[79] Y.H. Chen, C.S. Yeh, Laser ablation method: use of surfactants to form the synthesis: optimization by Box-Behnken design, Mater. Sci. Eng. C, Mater.
dispersed Ag nanoparticles, Colloids Surf. A 197 (2002) 133–139. Biol. Appl. 99 (2019) 1105–1114.
[80] P. Sen, J. Ghosh, A. Abdullah, P. Kumar, Preparation of Cu, Ag, Fe and Al [106] M. Khan, S.T. Khan, M. Khan, S.F. Adil, J. Musarrat, A.A. Al-Khedhairy, A. Al-
nanoparticles by the exploding wire technique, J. Chem. Sci. 115 (2003) 499– Warthan, M. Siddiqui, H.Z. Alkhathlan, Antibacterial properties of silver
508. nanoparticles synthesized using Pulicaria glutinosa plant extract as a green
[81] S.A. Agnihotri, N.N. Mallikarjuna, T.M. Aminabhavi, Recent advances on bioreductant, Int. J. Nanomed. 9 (2014) 3551–3565.
chitosan-based micro and nanoparticles in drug delivery, J. Control. Release [107] K. Roy, C. Sarkar, C. Ghosh, Plant-mediated synthesis of silver nanoparticles
100 (2004) 5–28. using parsley (Petroselinum crispum) leaf extract: spectral analysis of the
[82] S. Majeed, E.L. Joel, M.S. Hasnain, Novel green approach for synthesis of particles and antibacterial study, Appl. Nanosci. 5 (2015) 945–951.
metallic nanoparticles and its biomedical application, Curr. Nanomed. 8 [108] B. Chopade, S. Ghosh, S. Patil, M. Ahire, R. Kitture, A. Jabgunde, S. Kale, K.
(2018) 177. Pardesi, S.S. Cameotra, J. Bellare, Synthesis of silver nanoparticles using

766
S. Anees Ahmad, S. Sachi Das, A. Khatoon et al. Materials Science for Energy Technologies 3 (2020) 756–769

Dioscorea bulbifera tuber extract and evaluation of its synergistic potential in [133] S.S. Das, S. Alkahtani, P. Bharadwaj, M.T. Ansari, M.D. ALKahtani, Z. Pang, M.S.
combination with antimicrobial agents, Int. J. Nanomed. 483 (2012). Hasnain, A.K. Nayak, T. Aminabhavi, Molecular insights and novel approaches
[109] R. Veerasamy, T.Z. Xin, S. Gunasagaran, T.F.W. Xiang, E.F.C. Yang, N. for targeting tumor metastasis, Int. J. Pharm. 585 (2020) 119556.
Jeyakumar, S.A. Dhanaraj, Biosynthesis of silver nanoparticles using [134] V. Pareek, R. Gupta, J. Panwar, Do physico-chemical properties of silver
mangosteen leaf extract and evaluation of their antimicrobial activities, J. nanoparticles decide their interaction with biological media and bactericidal
Saudi Chem. Society. 15 (2011) 113–120. action? A review, Mater. Sci. Eng. C Mater. Biol. Appl. 90 (2018) 739–749.
[110] A. Saravanakumar, M.M. Peng, M. Ganesh, J. Jayaprakash, M. [135] C. Liao, Y. Li, S.C. Tjong, Bactericidal and cytotoxic properties of silver
Mohankumar, H.T. Jang, Low-cost and eco-friendly green synthesis of nanoparticles, Int. J. Mol. Sci. 2019 (20) (2019) 449.
silver nanoparticles using Prunus japonica (Rosaceae) leaf extract and [136] K.B. Riaz Ahmed, A.M. Nagy, R.P. Brown, Q. Zhang, S.G. Malghan, P.L. Goering,
their antibacterial, antioxidant properties, Artif. Cells Nanomed. Silver nanoparticles: Significance of physicochemical properties and assay
Biotechnol. 45 (2017) 1–7. interference on the interpretation of in vitro cytotoxicity studies, Toxicol. In
[111] S. Arokiyaraj, S. Vincent, M. Saravanan, Y. Lee, Y. KOh, K.H. Kim, Green Vitro 38 (2017) 179–192.
synthesis of silver nanoparticles using Rheum palmatum root extract and [137] S. Beg, A. Jain, S. Saini, T. Sharma, M.S. Hasnain, S.S. Imam, I. Kazmi, M.
their antibacterial activity against Staphylococcus aureus and Pseudomonas Rahman, S. Akhter, B. Singh, Metal–organic frameworks as expanding hybrid
aeruginosa, Artif. Cells Nanomed. Biotechnol. 45 (2017) 372–379. carriers with diverse therapeutic applications, in: A.M. Grumezescu (Ed.),
[112] M. Oves, M. Aslam, M.A. Rauf, S. Qayyum, H.A. Qari, M.S. Khan, M.Z. Alam, S. Organic Materials as Smart Nanocarriers for Drug Delivery, William Andrew
Tabrez, A. Pugazhendhi, I. Ismail, Antimicrobial and anticancer activities of Publishing, 2018, pp. 1–34.
silver nanoparticles synthesized from the root hair extract of Phoenix [138] M. Rai, A. Yadav, A. Gade, Silver nanoparticles as a new generation of
dactylifera, Mater. Sci. Eng. C Mater. Biol. Appl. 89 (2018) 429–443. antimicrobials, Biotechnol. Adv. 27 (2009) 76–83.
[113] M.M.I. Masum, M.M. Siddiqa, K.A. Ali, Y. Zhang, Y. Abdallah, E. Ibrahim, W. [139] V.T. Noronha, A.J. Paula, G. Durán, et al., Silver nanoparticles in dentistry,
Qiu, C. Yan, B. Li, Biogenic synthesis of silver nanoparticles using Phyllanthus Dent Mater. 33 (2017) 1110–1126.
emblica fruit extract and its inhibitory action against the pathogen Acidovorax [140] R. Shanmuganathan, D. MubarakAli, D. Prabakar, et al., An enhancement of
oryzae strain RS-2 of rice bacterial brown stripe, Front. Microbiol. 26 (2019) antimicrobial efficacy of biogenic and ceftriaxone-conjugated silver
820. nanoparticles: green approach, Environ. Sci. Pollut. Res. Int. 25 (2018)
[114] R. Amrita, R. Lawerence, Green synthesis and charcterization of ZnO 10362–10370.
nanoparticles from leafs extracts of Rosa indica and its antibacterial [141] S. Khorrami, F. Jafari, A. Zarrabi, A. Zarepour, Is Astragalus gossypinus honey a
activity, Rasayan J. Chem. 11 (2018) 1339–1348. natural antibacterial and cytotoxic agent? An investigation on A. gossypinus
[115] R.G. Saratale, G. Benelli, G. Kumar, D.S. Kim, G.D. Saratale, Bio-fabrication of honey biological activity and its green synthesized silver nanoparticles,
silver nanoparticles using the leaf extract of an ancient herbal medicine, Bionanoscience 9 (2018) 603–610.
dandelion (Taraxacum officinale), evaluation of their antioxidant, anticancer [142] J.M. Jacob, M.S. John, A. Jacob, P. Abitha, S.S. Kumar, R. Rajan, S. Natarajan, A.
potential, and antimicrobial activity against phytopathogens, Environ. Sci. Pugazhendhi, Bactericidal coating of paper towels via sustainable
Pollut. Res. Int. 25 (2018) 10392–10406. biosynthesis of silver nanoparticles using Ocimum sanctum leaf extract.
[116] R.N. Krishnaraj, S. Berchmans, In vitro antiplatelet activity of silver Bactericidal Coat Pap Towels Sustainable Biosynth Silver Nanopart Ocimum
nanoparticles synthesized using the microorganism Gluconobacter roseus: Sanctum Leaf Extr, Mater. Res. Express 6 (2019) 045401.
an AFM-based study, RSC Adv. 3 (2013) 8953–8959. [143] R.E. DuvaL, J. Gouyau, E. Lamouroux, Limitations of recent studies dealing
[117] S. Priyadarshini, V. Gopinath, N.M. Priyadharsshini, A.D. Mubarak, P. with the antibacterial properties of silver nanoparticles: fact and opinion,
Velusamy, Synthesis of anisotropic silver nanoparticles using novel strain, Nanomaterials 9 (2019) 1–22.
Bacillus flexus and its biomedical application, Coll. Surf. B. Biointerface 102 [144] S. Agnihotri, S. Mukherji, S. Mukherji, Size-controlled silver nanoparticles
(2013) 232–237. synthesized over the range 5–100 nm using the same protocol and their
[118] P. Pourali, M. Baserisalehi, S. Afsharnezhad, J. Behravan, H. Alavi, A. Hosseini, antibacterial efficacy, RSC Adv. 4 (2014) 3974–3983.
Biological synthesis of silver and gold nanoparticles by bacteria in [145] X. Hong, J. Wen, X. Xiong, Y. Hu, Shape effect on the antibacterial activity of
different temperatures (37 °C and 50 °C), J. Pure. Appl. Microbiol. 6 (2012) silver nanoparticles synthesized via a microwave-assisted method, Environ.
757–763. Sci. Pollut. Res. 23 (2016) 4489–4497.
[119] H. Zhang, Q. Li, Y. Lu, D. Sun, X. Lin, X. Deng, N. He, S. Zheng, Biosorption and [146] Z. Lu, K. Rong, J. Li, H. Yang, R. Chen, Size-dependent antibacterial activities of
bioreduction of diamine silver complex by Corynebacterium, J. Chem. silver nanoparticles against oral anaerobic pathogenic bacteria, J. Mater. Sci.
Technol. Biotechnol. 80 (2005) 285–290. Mater. Med. 24 (2013) 1465–1471.
[120] R.Y. Parikh, S. Singh, B.L.V. Prasad, M.S. Patole, M. Sastry, Y.S. Shouche, [147] D.H. Kim, J.C. Park, G.E. Jeon, C.S. Kim, J.H. Seo, Effect of the size and shape of
Extracellular synthesis of crystalline silver nanoparticles and molecular silver nanoparticles on bacterial growth and metabolism by monitoring
evidence of silver resistance from Morganella sp.: towards understanding optical density and fluorescence intensity, Biotechnol. Bioprocess Eng. 22
biochemical synthesis mechanism, Chem. Bio. Chem. 9 (2008) 1415–1422. (2017) 210–217.
[121] M.M. Juibari, S. Abbasalizadeh, G.S. Jouzani, M. Noruzi, Intensified [148] M. Saravanan, S. Arokiyaraj, T. Lakshmi, A. Pugazhendhi, Synthesis of silver
biosynthesis of silver nanoparticles using a native extremophilic nanoparticles from Phenerochaete chrysosporium (MTCC-787) and their
Ureibacillus thermosphaericus strain, Mater. Lett. 65 (2011) 1014–1017. antibacterial activity against human pathogenic bacteria, Microb. Pathog.
[122] V.G. Debabov, T.A. Voeikova, A.S. Shebanova, K.V. Shaitan, L.K. Emel’yanova, L. 117 (2018) 68–72.
M. Novikova, M.P. Kirpichnikov, Bacterial synthesis of silver sulfide [149] V.K. Sharma, R.A. Yngard, Y. Lin, Silver nanoparticles: green synthesis and
nanoparticles, Nanotechnol Russ. 8 (2013) 269–276. their antimicrobial activities, Adv. Colloid Interface Sci. 145 (2009) 83–96.
[123] A.M. Fayaz, M. Girilal, M. Rahman, R. Venkatesan, P.T. Kalaichelvan, [150] J. Park, D.H. Lim, H.J. Lim, T. Kwon, J.S. Choi, S. Jeong, Size dependent
Biosynthesis of silver and gold nanoparticles using thermophilic bacterium macrophage responses and toxicological effects of Ag nanoparticles, Chem.
Geobacillus stearothermophilus, Process Biochem. 46 (2011) 1958–1962. Commun. 47 (2011) 4382–4384.
[124] A. Lateef, I.A. Adelere, E.B. Gueguim-Kana, T.B. Asafa, L.S. Beukes, Green [151] B.S. Atiyeh, M. Costagliola, S.N. Hayek, S.A. Dibo, Effect of silver on burn
synthesis of silver nanoparticles using keratinase obtained from a strain of wound infection control and healing: review of the literature, Burns 33
Bacillus safensis LAU 13, Int. Nano Lett. 5 (2015) 29–35. (2007) 139–148.
[125] R.A. Bapat, T.V. Chaubal, C.P. Joshi, et al., An overview of application of silver [152] J. Tian, K.K. Wong, C.M. Ho, C.N. Lok, W.Y. Yu, C.M. Che, J.F. Chiu, P.K. Tam,
nanoparticles for biomaterials in dentistry, Mater. Sci. Eng. C 91 (2018) 881– Topical delivery of silver nanoparticles promotes wound healing,
898. ChemMedChem 2 (2007) 129–136.
[126] U. Klueh, V. Wagner, S. Kelly, A. Johnson, J.D. Bryers, Efficacy of silver- [153] A. Melaiye, W.J. Youngs, Silver and its Application as an Antimicrobial Agent,
coated fabric to prevent bacterial colonization and subsequent device- Taylor & Francis, 2005.
based biofilm formation, J. Biomed. Mater. Res. Part B: Appl. Biomater. 53 [154] J.S. Kim, E. Kuk, K.N. Yu, J.H. Kim, S.J. Park, H.J. Lee, S.H. Kim, Y.K. Park, Y.H.
(2000) 621–631. Park, C.Y. Hwang, Antimicrobial effects of silver nanoparticles, Nanomed.
[127] Y. Yakabe, T. Sano, H. Ushio, T. Yasunaga, Kinetic studies of the interaction Nanotechnol. 3 (2007) 95–101.
between silver ion and deoxyribonucleic acid, Chem. Lett. 4 (1980) 373–376. [155] P. Li, J. Li, C. Wu, Q. Wu, J. Li, Synergistic antibacterial effects of b-lactam
[128] S. Chouhan, S. Guleria, Green synthesis of AgNPs using Cannabis sativa leaf antibiotic combined with silver nanoparticles, Nanotechnology 16 (2005)
extract: characterization, antibacterial, anti-yeast and a-amylase inhibitory 1912.
activity, Mater. Sci. Energy Technol. 3 (2020) 536–544. [156] N.L. Rosi, N.L., C.A. Mirkin, Nanostructures in biodiagnostics. Chem. Rev. 105
[129] Y.W. Cao, R. Jin, C.A. Mirkin, DNA-modified core–shell Ag/Au nanoparticles, J. (2005), 1547-1562.
Am. Chem. Soc. 123 (2001) 7961–7962. [157] S. Pal, Y.K. Tak, J.M. Song, Does the antibacterial activity of silver
[130] J.B. Wright, K. Lam, D. Hanson, R.E. Burrell, Efficacy of topical silver against nanoparticles depend on the shape of the nanoparticle? A study of the
fungal burn wound pathogens, Am. J. Infect. Control. 27 (1999) 344–350. gram-negative bacterium Escherichia coli, Appl. Environ. Microbiol. 73
[131] E.D. Matthew, N.M. Schaeublin, K.E. Farrington, S.M. Hussain, G.R. Johnson, (2007) 1712–1720.
Lysozyme catalyzes the formation of antimicrobial silver nanoparticles, ACS [158] M. Raffi, F. Hussain, T. Bhatti, J. Akhter, A. Hameed, M. Hasan, Antibacterial
Nano 3 (2009) 984–994. characterization of silver nanoparticles against E. coli ATCC-15224, J. Mater.
[132] S. Khorrami, A. Zarrabi, M. Khaleghi, M. Danaei, M. Mozafari, Selective Sci. Technol. 24 (2008) 192–196.
cytotoxicity of green synthesized silver nanoparticles against the MCF-7 [159] O. Choi, K.K. Deng, N.J. Kim, L. Ross, R.Y. Surampalli, Z. Hu, The inhibitory
tumor cell line and their enhanced antioxidant and antimicrobial properties, effects of silver nanoparticles, silver ions, and silver chloride colloids on
Int. J. Nanomed. 13 (2018) 8013–8024. microbial growth, Water Res. 42 (2008) 3066–3074.

767
S. Anees Ahmad, S. Sachi Das, A. Khatoon et al. Materials Science for Energy Technologies 3 (2020) 756–769

[160] M. Ahamed, M.S. Alsalhi, M. Siddiqui, Silver nanoparticle applications and Polysaccharides-Based Multiple-Unit Systems for Oral Drug Delivery,
human health, Clin. Chim. Acta 41 (2010) 1841–1848. Springer Briefs in Applied Sciences and Technology, Springer, Singapore,
[161] H.A. Jeng, J. Swanson, Toxicity of metal oxide nanoparticles in mammalian 2019, pp. 117–121.
cells, J. Environ. Sci. Health. A 41 (2006) 2699–2711. [187] X. Wang, L.H. Liu, O. Ramström, M. Yan, Engineering nanomaterial surfaces
[162] S. Hussain, K. Hess, J. Gearhart, K. Geiss, J. Schlager, In vitro toxicity of for biomedical applications, Exp. Biol. Med. 23 (2009) 1128–1139.
nanoparticles in BRL 3A rat liver cells, Toxicol. In Vitro 19 (2005) 975–983. [188] L. Prescott, Bacteria: The high G+ C gram positives, in: L.M. Prescott, J.P.
[163] N. Lewinski, V. Colvin, R. Drezek, Cytotoxicity of nanoparticles, Small 4 (2008) Harley, D.A. Klein (Eds.), Microbiology, McGraw-Hill, 1999.
26–49. [189] M.U. Niemann, S.S. Srinivasan, A.R. Phani, A. Kumar, D.Y. Goswami, E.K.
[164] A.R. Shahverdi, A. Fakhimi, H.R. Shahverdi, S. Minaian, Synthesis and effect of Stefanakos, Nanomaterials for hydrogen storage applications: a review, J.
silver nanoparticles on the antibacterial activity of different antibiotics Nanomaterial. 2008 (2008) 1–9.
against Staphylococcus aureus and Escherichia coli, Nanomed. Nanotechnol. 3 [190] K.A. Brogden, Antimicrobial peptides: pore formers or metabolic inhibitors in
(2007) 168–171. bacteria?, Nat Rev. Microbiol. 3 (2005) 238–250.
[165] M.A. Barkat, S.S Das Harshita, S. Beg, F.J. Ahmad, Nanotechnology-based [191] G. Gahlawat, S. Shikha, B.S. Chaddha, S.R. Chaudhuri, S. Mayilraj, A.R.
phytotherapeutics: current status and challenges, in: S. Beg, M. Barkat, F. Choudhury, Microbial glycolipoprotein-capped silver nanoparticles as
Ahmad (Eds.), Nanophytomedicine, Springer, Singapore, 2020, pp. 1–17. emerging antibacterial agents against cholera, Microb. Cell Fact. 15 (2016)
[166] B.S. Sekhon, Metalloantibiotics and antibiotic mimics-an overview, J. Pharm. 25.
Edu. Res. 1 (2010) 1. [192] C.H. Barros, S. Fulaz, D. Stanisic, L. Tasic, Biogenic nanosilver against
[167] A.M. Allahverdiyev, K.V. Kon, E.S. Abamor, M. Bagirova, M. Rafailovich, Coping multidrug-resistant bacteria (MRDB), Antibiotics 7 (2018) 69.
with antibiotic resistance: combining nanoparticles with antibiotics and [193] M.A. Quinteros, V.C. Aristizabal, P.R. Dalmasso, M.G. Paraje, P.L. Paez,
other antimicrobial agents, Expert Rev. Anti Infect. Ther. 9 (2011) 1035–1052. Oxidative stress generation of silver nanoparticles in three bacterial genera
[168] C.M. Che, F.M. Siu, Metal complexes in medicine with a focus on enzyme and its relationship with the antimicrobial activity, Toxicol. In Vitro 36
inhibition, Curr. Opin. Chem. Biol. 14 (2010) 255–261. (2016) 216–223.
[169] J. Yu, S.A. Patel, R.M. Dickson, In vitro and intracellular production of peptide- [194] X. Xia, J. Zeng, C.H. Moran, Y. Xia, Recent developments in shape-controlled
encapsulated fluorescent silver nanoclusters, Angew. Chem. 119 (2007) synthesis of silver nanocrystals, J. Phys. Chem. C 116 (2012) 21647–21656.
2074–2076. [195] Z. Hosseinidoust, M. Basenet, T.G. van de Ven, N. Tufenkji, One-pot green
[170] N. Durán, P.D. Marcato, M. Durán, A. Yadav, A. Gade, M. Rai, Mechanistic synthesis of anisotropic silver nanoparticles, Environ. Sci. Nano 3 (2016)
aspects in the biogenic synthesis of extracellular metal nanoparticles by 1259–1264.
peptides, bacteria, fungi, and plants, Appl. Microbiol. Biot. 90 (2011) 1609– [196] D. Acharya, K.M. Singha, P. Pandey, B. Mohanta, J. Rajkumari, L.P. Singha,
1624. Shape dependent physical mutilation and lethal effects of silver
[171] K.J. Waldron, N.J. Robinson, How do bacterial cells ensure that nanoparticles on bacteria, Sci. Rep. 8 (2018) 201.
metalloproteins get the correct metal?, Nat Rev. Microbiol. 7 (2009) 25–35. [197] A. Abbaszadegan, Y. Ghahramani, A. Gholami, B. Hemmateenejad, S.
[172] N.P. Shetti, D.S. Nayak, S.J. Malode, K.R. Reddy, S.S. Shukla, T.M. Aminabhavi, Dorostkar, M. Nabavizadeh, H. Shargh, The effect of charge at the surface of
Electrochemical behavior of flufenamic acid at amberlite XAD-4 resin and silver nanoparticles on antimicrobial activity against Gram-positive and
silver-doped titanium dioxide/ amberlite XAD-4 resin modified carbon Gram-negative bacteria: a preliminary study, J. Nanomater. 201 (2015)
electrodes, Colloids Surf. B: Biointerfaces 177 (2019) 407–415. 720654.
[173] N.P. Shetti, S.J. Malode, D.S. Nayak, T.M. Aminabhavi, K.R. Reddy, [198] A.M. Badawy, R. Silva, B. Morris, K.G. Scheckel, M.T. Suidan, T.M. Tolaymat,
Nanostructured silver doped TiO2/CNTs hybrid as an efficient Surface charge-dependent toxicity of silver nanoparticles, Environ. Sci.
electrochemical sensor for detection of anti-inflammatory drug, cetirizine, Technol. 45 (2011) 283–287.
Microchem. J. 150 (2019) 104124. [199] C.M.J. Silvero, D.M. Rocca, E.A. de la Villarmois, K. Fournier, A.E. Lanterna, M.F.
[174] S.S. Das, K. Neelam, S. Hussain, A. Singh, A. Hussain, M. Faruk, Tebyetekerwa, Pérez, M.C. Becerra, J.C. Scaiano, Selective photoinduced antibacterial activity
laponite-based nanomaterials for biomedical applications: a review, Curr. of amoxicillin-coated gold nanoparticles: from one-step synthesis to in vivo
Pharm. Des. 25 (2019) 424–443. cytocompatibility, ACS Omega 3 (2018) 1220–1230.
[175] N.P. Shetti, S.J. Malode, D.S. Nayak, G.B. Bagihalli, S.S. Kalanur, R.S. Malladi, C. [200] A.K. Nayak, M.S. Hasnain, Synthesis of carbon nanotubes, in: A.K. Nayak, M.S.
V. Reddy, T.M. Aminabhavi, K.R. Reddy, Fabrication of ZnO nanoparticles Hasnain (Eds.), Carbon Nanotubes for Targeted Drug Delivery, Springer Briefs
modified sensor for electrochemical oxidation of methdilazine, Appl. Surf. Sci. in Applied Sciences and Technology. Springer Nature, Singapore, 2019, pp.
496 (2019) 143656. 17–20.
[176] N.P. Shetti, D.S. Nayak, S.J. Malode, R.R. Kakarla, S.S. Shukla, T.M. Aminabhavi, [201] A.K. Nayak, M.S. Hasnain, Functionalization of carbon nanotubes, in: A.K.
Sensors based on ruthenium-doped TiO2 nanoparticles loaded into multi- Nayak, M.S. Hasnain (Eds.), Carbon Nanotubes for Targeted Drug Delivery,
walled carbon nanotubes for the detection of flufenamic acid and mefenamic Springer Briefs in Applied Sciences and Technology. Springer Nature,
acid, Anal. Chim. Acta 1051 (2019) 58–72. Singapore, 2019, pp. 21–28.
[177] N.P. Shetti, S.J. Malode, D. Ilager, K.R. Reddy, S.S. Shukla, T.M. Aminabhavi, A [202] A.K. Nayak, M.S. Hasnain, Characterization of carbon nanotubes, in: A.K.
novel electrochemical sensor for detection of molinate using ZnO Nayak, M.S. Hasnain (Eds.), Carbon Nanotubes for Targeted Drug Delivery,
nanoparticles loaded carbon electrode, Electroanalysis 31 (2019) 1040–1049. Springer Briefs in Applied Sciences and Technology. Springer Nature,
[178] P.V. Kulkarni, C.A. Roney, P.P. Antich, F.J. Bonte, A.V. Raghu, T.M. Aminabhavi, Singapore, 2019, pp. 29–31.
Quinoline-n-butylcyanoacrylate-based nanoparticles for brain targeting for [203] A.K. Nayak, M.S. Hasnain, Applications of carbon nanotubes, in: A.K. Nayak,
the diagnosis of Alzheimers Disease, in: WIREs Nanomedicine and M.S. Hasnain (Eds.), Carbon Nanotubes for Targeted Drug Delivery, Springer
Nanotechnology, John Wiley & Sons Inc, New York, 2010, pp. 35–47. Briefs in Applied Sciences and Technology. Springer Nature, Singapore, 2019,
[179] C. Roney, P.V. Kulkarni, V. Arora, P. Antich, F. Bonte, A. Wu, N.N. Mallikarjuna, pp. 33–36.
S. Manohar, H.F. Liang, A.R. Kulkarni, H.W. Sung, M. Sairam, T.M. Aminabhavi, [204] A.K. Nayak, M.S. Hasnain, CNTs in solubility enhancement, in: A.K. Nayak, M.
Targetted nanoparticles for drug delivery through blood brain barrier, J. S. Hasnain (Eds.), Carbon Nanotubes for Targeted Drug Delivery, Springer
Control. Release 108 (2005) 193–214. Briefs in Applied Sciences and Technology. Springer Nature, Singapore, 2019,
[180] N.N. Mallikarjuna, S.K. Manohar, P.V. Kulkarni, A. Venkataramana, T.M. pp. 55–57.
Aminabhavi, Novel high dielectric constant nanocomposites of polyaniline [205] A.K. Nayak, M.S. Hasnain, Carbon nanotubes in vaccine delivery, in: A.K.
dispersed with c-Fe2O3 nanoparticles, J. Appl. Polym. Sci. 97 (2005) 1868– Nayak, M.S. Hasnain (Eds.), Carbon Nanotubes for Targeted Drug Delivery,
1874. Springer Briefs in Applied Sciences and Technology. Springer Nature,
[181] A.K. Nayak, M.S. Hasnain, Plant polysaccharides in drug delivery applications, Singapore, 2019, pp. 69–73.
in: A.K. Nayak, M.S. Hasnain (Eds.), Plant Polysaccharides-Based Multiple- [206] A.K. Nayak, M.S. Hasnain, Classification of carbon nanotubes, in: A.K. Nayak,
Unit Systems for Oral Drug Delivery, SpringerBriefs in Applied Sciences and M.S. Hasnain (Eds.), Carbon Nanotubes for Targeted Drug Delivery, Springer
Technology, Springer, Singapore, 2019, pp. 19–23. Briefs in Applied Sciences and Technology. Springer Nature, Singapore, 2019,
[182] A.K. Nayak, M.S. Hasnain, Okra gum based multiple units for oral drug pp. 11–15.
delivery, in: A.K. Nayak, M.S. Hasnain (Eds.), Plant Polysaccharides-Based [207] A.K. Nayak, M.S. Hasnain, Carbon nanotubes as quantum dots for therapeutic
Multiple-Unit Systems for Oral Drug Delivery, SpringerBriefs in Applied purpose, in: A.K. Nayak, M.S. Hasnain (Eds.), Carbon Nanotubes for Targeted
Sciences and Technology, Springer, Singapore, 2019, pp. 83–92. Drug Delivery, SpringerBriefs in Applied Sciences and Technology. Springer
[183] K.S. Soppimath, T.M. Aminabhavi, A.R. Kulkarni, W.E. Rudzinski, Nature, Singapore, 2019, pp. 59–64.
Biodegradable polymeric nanoparticles as drug delivery devices, J. Control. [208] K.S. Siddiqi, A. Husen, R.A.K. Rao, A review on biosynthesis of silver
Release 70 (2001) 1–20. nanoparticles and their biocidal properties, J. Nanobiotechnol. 16
[184] A.K. Nayak, M.S. Hasnain, Sterculia gum based multiple units for oral drug (2018).
delivery, in: A.K. Nayak, M.S. Hasnain (Eds.), Plant Polysaccharides-Based [209] K. Gupta, C. Sanjay, Biofunctionalization of silver nanoparticles with
Multiple-Unit Systems for Oral Drug Delivery, Springer Briefs in Applied lactonase leads to altered antimicrobial and cytotoxic properties, Front.
Sciences and Technology, Springer, Singapore, 2019, pp. 67–82. Mol. Biosci. 6 (2019) 1–14.
[185] A.K. Nayak, M.S. Hasnain, Potato starch based multiple units for oral drug [210] H.A. Barkat, S.S. Das, M.A. Barkat, S. Beg, H.A. Hadi, Selective targeting of
delivery, in: A.K. Nayak, M.S. Hasnain (Eds.), Plant Polysaccharides-Based cancer signaling pathways with nanomedicines: challenges and progress,
Multiple-Unit Systems for Oral Drug Delivery, Springer Briefs in Applied Future Oncol. (2020), https://doi.org/10.2217/fon-2020-0198.
Sciences and Technology, Springer, Singapore, 2019, pp. 113–116. [211] N. Duran, M. Duran, C.E. de Souza, Silver and silver chloride nanoparticles
[186] A.K. Nayak, M.S. Hasnain, Linseed polysaccharide based multiple units for and their anti-tick activity: a mini review, J. Braz. Chem. Soc. 28 (2017) 927–
oral drug delivery, in: A.K. Nayak, M.S. Hasnain (Eds.), Plant 932.

768
S. Anees Ahmad, S. Sachi Das, A. Khatoon et al. Materials Science for Energy Technologies 3 (2020) 756–769

[212] E.I. Hassanen, E. Ragab, In vivo and in vitro assessments of the antibacterial contaminated sea water solutions and synthetic, natural based dye-
potential of chitosan-silver nanocomposite against methicillin-resistant sensitized solar cells, Mater. Sci. Energy Technol. 3 (2020) 183–192.
Staphylococcus aureus-induced infection in rats, Biol. Trace Elem. Res. [224] M.A. Harshita, S.S. Barkat, F.H. Das, S. Pottoo, Z. Beg, Rahman, lipid-based
(2020), https://doi.org/10.1007/s12011-020-02143-6 [published online nanosystem as intelligent carriers for versatile drug delivery applications,
ahead of print]. Curr. Phamr. Des. 26 (2020) 1167–1180.
[213] W. Shao, H. Liu, X. Liu, H. Sun, S. Wang, R. Zhang, pH-responsive release [225] M. Ema, H. Okuda, M. Gamo, K. Honda, A review of reproductive and
behavior and anti-bacterial activity of bacterial cellulose-silver developmental toxicity of silver nanoparticles in laboratory animals, Reprod.
nanocomposites, Int. J. Biol. Macromol. 76 (2015) 209–217. Toxicol. 67 (2017) 149–164.
[214] J.J. Martínez-Sanmiguel, D. Zarate-Triviño, R. Hernandez-Delgadillo, A.L. [226] W. Rolim, J. Pieretti, D. Reno, B. Lima, M. Nascimento, F. Ambrosio, C.
Giraldo-Betancur, N. Pineda-Aguilar, S.A. Galindo-Rodríguez, M.A. Franco- Lombello, M. Brocchi, A.C.S. de Souza, A.B. Seabra, Antimicrobial activity and
Molina, S.P. Hernández-Martínez, C. Rodríguez-Padilla, Anti-inflammatory cytotoxicity to tumor cells of nitric oxide donor and silver nanoparticles
and antimicrobial activity of bioactive hydroxyapatite/silver containing PVA/ PEG films for topical applications, ACS Appl. Mater.
nanocomposites, J. Biomater. Appl. 33 (2019) 1314–1326. Interfaces 11 (2019) 6589–6604.
[215] Y. Chen, W. Wu, Z. Xu, C. Jiang, S. Han, J. Ruan, Y. Wang, Photothermal- [227] W.R. Rolim, M.T. Pelegrino, B.A. Lima, L.S. Ferraz, F.N. Costa, J.S. Bernardes, T.
assisted antibacterial application of graphene oxide-Ag nanocomposites Rodigues, M. Brocchi, A.B. Seabra, Green tea extract mediated biogenic
against clinically isolated multi-drug resistant Escherichia coli, Royal Soc. synthesis of silver nanoparticles: characterization, cytotoxicity evaluation
Open Sci. 7 (2020) 192019. and antibacterial activity, Appl. Surf. Sci. 463 (2019) 66–74.
[216] L. Yang, F. Meng, X. Qu, L. Xia, F. Huang, S. Qin, M. Zhang, F. Xu, L. Sun, H. Liu, [228] R. Brayner, The toxicological impact of nanoparticles, Nanotoday 3 (2008)
Multiple-twinned silver nanoparticles supported on mesoporous graphene 48–55.
with enhanced antibacterial activity, Carbon 155 (2019) 397–402. [229] A. Hussain, S. Singh, S.S. Das, K. Anjireddy, S. Karpagam, F. Shakeel,
[217] M. Cobos, I. De-La-Pinta, G. Quindós, M.J. Fernández, M.D. Fernández, Nanomedicines as drug delivery carriers of anti-tubercular drugs: from
Synthesis, physical, mechanical and antibacterial properties of pathogenesis to infection control, Curr. Drug Deliv. 16 (2019) 400–429.
nanocomposites based on poly(vinyl alcohol)/graphene oxide-silver [230] I. Hussain, N.B. Singh, A. Singh, H. Singh, S.C. Singh, Green synthesis of
nanoparticles, Polymers 12 (2020) 723. nanoparticles and its potential application, Biotechnol. Lett. 38 (2016) 545–
[218] A. Usman, Z. Hussain, A. Riaz, A.N. Khan, Enhanced mechanical, thermal and 560.
antimicrobial properties of poly(vinyl alcohol)/graphene oxide/starch/silver [231] S. Iravani, Green synthesis of metal nanoparticles using plants, Green Chem.
nanocomposites films, Carbohydr. Polym. 153 (2016) 592–599. 13 (2011) 2638–2650.
[219] L. Xia, M. Xu, G. Cheng, L. Yang, Y. Guo, D. Li, D. Fang, Q. Zhang, H. Liu, Facile [232] V.K. Sharma, R.A. Yngard, Y. Lin, Silver nanoparticles: green synthesis and
construction of Ag nanoparticles encapsulated into carbon nanotubes with their antimicrobial activities, Adv. Colloid Interface Sci. 145 (2009) 83–96.
robust antibacterial activity, Carbon 130 (2018) 775–781. [233] K.K. Panda, V.M.M. Achary, R. Krishnaveni, B.K. Padhi, S.N. Sarangi, S.N. Sahu,
[220] M. Akter, M.D.T. Sikder, M.D.M. Rahman, A.K.M.A. Ullah, K.F.B. Hossain, S. B.B. Panda, In vitro biosynthesis and genotoxicity bioassay of silver
Banik, T. Hosokawa, T. Saito, M. Kurasaki, A systematic review on silver nanoparticles using plants, Toxicol. Vitro 25 (2011) 1097–1105.
nanoparticles- induced cytotoxicity: physicochemical properties and [234] S. Hackenberg, A. Scherzed, M. Kessler, S. Hummel, A. Technau, K. Froelich, C.
perspectives, Adv. Res. 9 (2018) 1–16. Ginzkey, C. Koehler, R. Hagen, N. Kleinsasser, Silver nanoparticles: evaluation
[221] S. Lekamge, A. Miranda, A. Abraham, V. Li, R. Shukla, V. Bansal, D. Nugegoda, of DNA damage, toxicity and functional impairment in human mesenchymal
The toxicity of silver nanoparticles (AgNPs) to three freshwater invertebrates stem cells, Toxicol. Lett. 201 (2011) 27–33.
with different life strategies: Hydra vulgaris, Daphnia carinata, and Paratya [235] M.E. Samberg, E.G. Loboa, S.J. Oldenburg, N.A. Monteiro-Riviere, Silver
australiensis, Front. Environ. Sci. 6 (2018) 1–13. nanoparticles do not influence stem cell differentiation but cause minimal
[222] T. Zhang, L. Wang, Q. Chen, C. Chen, Cytotoxic potential of silver toxicity, Nanomedicine 7 (2012) 1197–1209.
nanoparticles, Yonsei Med. J. 55 (2014) 283–291. [236] S. Kittler, C. Greulich, J. Diendorf, M. Koller, M. Epple, Toxicity of silver
[223] A.M. Ibraheem, J. Kamalakkannan, Sustainable scientific advancements nanoparticles increases during storage because of slow dissolution under
modified Ag2O-ZnO thin films characterization and application of release of silver ions, Chem. Mater. 22 (2010) 4548–4554.
photocatalytic purification of carcinogenic dye in deionizer water and

769

You might also like