You are on page 1of 21

J Nanopart Res (2010) 12:1531–1551

DOI 10.1007/s11051-010-9900-y

REVIEW PAPER

A review of the antibacterial effects of silver nanomaterials


and potential implications for human health
and the environment
Catalina Marambio-Jones • Eric M. V. Hoek

Received: 29 July 2009 / Accepted: 6 March 2010 / Published online: 23 March 2010
Ó Springer Science+Business Media B.V. 2010

Abstract Here, we present a review of the antibac- well as solution pH, ionic strength, specific ions and
terial effects of silver nanomaterials, including pro- ligands, and organic macromolecules—all of which
posed antibacterial mechanisms and possible toxicity influence silver nanoparticle stability and bioavailabil-
to higher organisms. For purpose of this review, silver ity. Although none of the studies reviewed definitively
nanomaterials include silver nanoparticles, stabilized proved any immediate impacts to human health or the
silver salts, silver–dendrimer, polymer and metal oxide environment by a silver nanomaterial containing
composites, and silver-impregnated zeolite and acti- product, the entirety of the science reviewed suggests
vated carbon materials. While there is some evidence some caution and further research are warranted given
that silver nanoparticles can directly damage bacteria the already widespread and rapidly growing use of
cell membranes, silver nanomaterials appear to exert silver nanomaterials.
bacteriocidal activity predominantly through release of
silver ions followed (individually or in combination) Keywords Silver  Nanoparticle 
by increased membrane permeability, loss of the Antimicrobial  Antibacterial  Nanotechnology 
proton motive force, inducing de-energization of the Nanotoxicology  Safety  EHS
cells and efflux of phosphate, leakage of cellular
content, and disruption DNA replication. Eukaryotic
cells could be similarly impacted by most of these
mechanisms and, indeed, a small but growing body of Introduction
literature supports this concern. Most antimicrobial
studies are performed in simple aquatic media or cell The broad-spectrum antimicrobial properties of silver
culture media without proper characterization of silver encourage its use in biomedical applications, water
nanomaterial stability (aggregation, dissolution, and and air purification, food production, cosmetics,
re-precipitation). Silver nanoparticle stability is gov- clothing, and numerous household products. With
erned by particle size, shape, and capping agents as the rapid development of nanotechnology, applica-
tions have been extended further and now silver is the
engineered nanomaterial most commonly used in
C. Marambio-Jones  E. M. V. Hoek (&) consumer products (Rejeski 2009). Clothing, respi-
Department of Civil and Environmental Engineering, rators, household water filters, contraconceptives,
California NanoSystems Institute, University antibacterial sprays, cosmetics, detergent, dietary
of California, Los Angeles, 5732G Boelter Hall,
PO Box 951593, Los Angeles, CA 90095-1593, USA supplements, cutting boards, sox, shoes, cell phones,
e-mail: emvhoek@ucla.edu laptop keyboards, and children’s toys are among the

123
1532 J Nanopart Res (2010) 12:1531–1551

retail products that purportedly exploit the antimi- shown effective for S. aureus, Pseudomonas aeru-
crobial properties of silver nanomaterials. ginosa, E. coli, B. subtilis, and K. mobilis (Balogh
Different forms of silver nanomaterials already in et al. 2001; Zhang et al. 2008). Furthermore, silver
such products include: metallic silver nanoparticles loaded in nanoporous materials such as silver-
(Arora et al. 2008; Chi et al. 2009; Choi et al. 2008; exchanged zeolites exhibit antibacterial effects for
Hwang et al. 2008; Kim et al. 2007, 2008a, b; Kvitek Pseudomonas putida, E. coli, B. subtilis, S. aureus,
et al. 2008; Lok et al. 2006; Raffi et al. 2008; Schrand and P. aeruginosa (Cowan et al. 2003; Inoue et al.
et al. 2008; Sondi and Salopek-Sondi 2004; Vertelov 2002; Lind et al. 2009; McDonnell et al. 2005).
et al. 2008), silver chloride particles (Choi et al. Despite the vast number of papers touting the
2008), silver-impregnated zeolite powders and acti- beneficial antimicrobial effects of silver nanomateri-
vated carbon materials (Cowan et al. 2003; Inoue als, a relatively modest number of studies have
et al. 2002; Yoon et al. 2008a, b), dendrimer–silver attempted to elucidate the mechanisms by which
complexes and composites (Balogh et al. 2001; silver nanomaterials exert this antimicrobial activity.
Lesniak et al. 2005; Zhang et al. 2008), polymer- As a result, the mechanisms are not widely under-
silver nanoparticle composites (Bajpai et al. 2007; stood or agreed upon. For bacteria, commonly
Damm and Munstedt 2008; Damm et al. 2008; Hlidek proposed mechanisms in the literature begin with
et al. 2008; Jin et al. 2007; Kim et al. 2009a, b; the release of silver ions (Hwang et al. 2008; Smetana
Kvitek et al. 2008; Naidu et al. 2008; Nita 2008; et al. 2008) followed by generation of reactive
Sambhy and Sen 2008; Sanpui et al. 2008; Xu et al. oxygen species (ROS) (Hwang et al. 2008; Kim
2006), silver-titanium dioxide composite nanopow- et al. 2007) and cell membrane damage (Choi et al.
ders (Yeo and Kang 2008), and silver nanoparticles 2008; Raffi et al. 2008; Smetana et al. 2008; Sondi
coated onto polymers like polyurethane (Jain and and Salopek-Sondi 2004), but there are many
Pradeep 2005). While all of these forms of silver contradictory findings reported.
exert antimicrobial activity to some extent through The more widespread our use of silver nanoma-
release of silver ions, silver nanoparticles might terials becomes the more widespread will become the
exhibit additional antimicrobial capabilities not potential for human and ecosystem exposure. Silver
exerted by bulk or ionic silver (Chen and Schluesener nanoparticles may be released to the environment
2008). from discharges at the point of production, from
Already, silver nanoparticles have been shown to erosion of engineered materials in household prod-
be effective biocides against: (a) bacteria such as ucts (e.g., antibacterial coatings and silver-impreg-
Escherichia coli, Staphylococcus aureus, Staphylo- nated water filters), and from washing or disposal of
coccus epidermis, Leuconostoc mesenteroides, Bacil- silver-containing products (Benn and Westerhoff
lus subtilis, Klebsiella mobilis, and Klebsiella 2008). Silver released to both natural and engineered
pneumonia among others (Benn and Westerhoff systems will likely impact the lowest trophic levels
2008; Chen and Chiang 2008; Falletta et al. 2008; first, i.e., bacteria. However, little is known about
Hernandez-Sierra et al. 2008; Ingle et al. 2008; Jung trophic transfer of silver and impacts to higher
et al. 2009; Kim 2007; Kim et al. 2007, 2009a, b; organisms. Indeed, silver nanoparticles have already
Kvitek et al. 2008; Raffi et al. 2008; Ruparelia et al. been proven toxic to both aerobic and anaerobic
2008; Smetana et al. 2008; Sondi and Salopek-Sondi bacteria isolated from wastewater treatment plants
2004; Vertelov et al. 2008; Yang et al. 2009; Yoon (Choi and Hu 2008), which we speculate could lead
et al. 2008a, b); (b) fungi such as Aspergillus niger, to severe disruption of this critical environmental
Candida albicans, Saccharomyces cerevisia, Tricho- infrastructure if the load of silver into wastewater
phyton mentagrophytes, and Penicillium citrinum treatment plants increases significantly.
(Kim et al. 2007, 2008a, b, 2009a, b; Roe et al. Given the vast number of products leveraging the
2008; Vertelov et al. 2008; Zhang et al. 2008); and (c) benefits of silver, it seems prudent to assess the
virii such as Hepatitis B, HIV-1, syncytial virus potential human and ecosystem hazards associated
(Elechiguerra et al. 2005; Lu et al. 2008; Sun et al. with its increased utilization. The main routes of
2008; Zodrow et al. 2009). Hybrid silver nanocom- human exposure would be the respiratory system,
posites with dendrimers and polymers have been gastrointestinal system, and skin, which are interfaces

123
J Nanopart Res (2010) 12:1531–1551 1533

between the internal systems of the human body and of the state-of-knowledge about silver nanomaterial
the external environment (Chen and Schluesener antibacterial effects with insights toward better
2008). For example, silver nanomaterials may enter understanding potential implications for human
through the respiratory tract due to inhalation of dust health and the environment.
or fumes containing silver nanomaterials at the point
of manufacture, it may be ingested from water,
children’s toys, or food containers treated with silver, Typical forms of silver nanomaterials
or it may penetrate the skin via silver-containing
textiles and cosmetics. Additionally, other potential Herein, the term ‘‘silver nanomaterials’’ refers to any
entryways could include the female genital tract (due silver-containing materials with enhanced activity
to incorporation of silver nanoparticles into numerous due to their nanoscale features. In some cases,
female hygienic products), via systemic administra- commercial products containing metallic silver nano-
tion as it is used for some imaging and therapeutic particles in the range of 5–50 nm or ionic silver are
purposes (Chen and Schluesener 2008; Schrand et al. given the name ‘nanosilver’ (Panyala et al. 2008).
2008; West and Halas 2003), or by incorporation into Silver nanoparticles are nanoscale clusters of metallic
medical implants, catheters, and wound dressings silver atoms, Ag0, engineered for some practical
(Furno et al. 2004; Galiano et al. 2008; Maneerung purpose—most typically antimicrobial and sterile
et al. 2008; Roe et al. 2008). applications.
In addition to broad-spectrum antimicrobial The most common method of producing silver
effects, silver nanoparticles have produced toxic nanoparticles is chemical reduction of a silver salt
effects in higher cell lines like zebra fish, clams, dissolved in water with a reducing compound such as
rats, and humans (Arora et al. 2008, 2009; Asharani NaBH4, citrate, glucose, hydrazine, and ascorbate
et al. 2008; Braydich-Stolle et al. 2005; Hsin et al. (Gulrajani et al. 2008; Martinez-Castanon et al. 2009;
2008; Hussain et al. 2005; Kim et al. 2008a, b; Sung Panacek et al. 2006; Pillai and Kamat 2004). Strong
et al. 2008; Yeo and Kang 2008; Yeo and Yoon reductants lead to small monodisperse particles,
2009). Evidence in rodents shows that after entering while generating larger sizes can be difficult to
into the body silver nanoparticles can accumulate control. Weaker reductants produce slower reduction
and, in some cases, damage tissues such as the liver, reactions, but the nanoparticles obtained tend to be
lungs, and olfactory bulbs, or penetrate the blood– more polydisperse in size. In order to generate silver
brain barrier (Arora et al. 2009; Braydich-Stolle et al. nanoparticles with controlled sizes, a two-step
2005; Hussain et al. 2005; Sung et al. 2008). A study method is usually utilized. In this method, nuclei
in human cells concluded that silver can be genotoxic particles are prepared using a strong reducing agent
(Asharani et al. 2009). Additionally, a release of ionic and they are enlarged by a weak reducing agent
silver led to the sterility of Macoma balthica clams in (Schneider et al. 1994; Shirtcliffe et al. 1999). Since
the San Francisco Bay during the 1980s (Brown et al. reducing agents for silver nanoparticle synthesis are
2003). If silver nanomaterials exhibit similar or often considered toxic or hazardous, the use of green
stronger reactivity, the impacts of this isolated event synthesis methods is becoming a priority (Panacek
in San Francisco may foreshadow potential ecosys- et al. 2006). A recent review of green synthesis
tem impacts of silver nanomaterials. methods for silver nanoparticles discussed the use of
The various forms of silver nanomaterials are polysaccharides, polyphenols, Tollens agent, irradia-
among the most promising antimicrobial agents being tion, biological reduction, and polyoxometalate
developed from nanotechnology, but the preliminary (Sharma et al. 2009).
evidence of effects on higher organisms alerts us to Polysaccharides and polyphenols are typically
remain cautious of its widespread utilization. This used as capping agents during silver nanoparticle
cautiousness demands additional research to deter- synthesis, but they may also contribute to reduction
mine how to safely design, use, and dispose products of silver ions through as yet poorly understood
containing silver nanomaterials without creating new mechanisms. For polysaccharides, the reduction of
risks to humans or the environment. Consequently, silver may be linked to the oxidation of aldehyde
the goal of this article is to provide a critical review groups to carboxylic acid groups (Manzi and van

123
1534 J Nanopart Res (2010) 12:1531–1551

Halbeek 1999). Typical polysaccharides used are the metal atoms and solvent warms causing the
glucose, starch, and heparin (Batabyal et al. 2007; aggregation of metal atoms. SMAD can be performed
Huang and Yang 2004; Manno et al. 2008; Singh in conjunction with digestive ripening, in this way the
et al. 2009; Venediktov and Padokhin 2008). In the nanoparticles resulting from SMAD method are
Tollens method, a silver ammoniacal solution is further refined by heating them in inert atmosphere
reduced by an aldehyde forming silver nanoparticles. in the presence of selected ligands that encourage the
This method can be altered (i.e., ‘‘modified Tollens particles to reach a narrow size range. As a result,
method’’) by reducing Ag? using saccharides in the monodisperse spherical particles are obtained (Sme-
presence of ammonia resulting in films with nano- tana et al. 2005, 2008).
particles sizes ranging from 50 to 200 nm and silver The use of silver ions as antimicrobial agents is
hydrosols ranging from 20 to 50 nm (Panacek et al. limited by the solubility of silver ions in biological
2006; Saito et al. 2003; Yu and Yam 2004). Silver and environmental media containing Cl-, because
nanoparticles can also be synthesized by irradiating AgCl has a very low solubility and rapidly precip-
silver salts solutions containing reducing and capping itates out of solution. In some cases, silver salts are
agents. Different sources of irradiation have been stabilized with hyperbranched polymers or dendri-
used such as laser, microwave, ionization radiation, mers that act as nanoreactors, wherein silver ions
and radiolysis (Abid et al. 2002; Ha et al. 2006; Li are initially complexed with a specific moiety in the
et al. 2006; Long et al. 2007; Mahapatra et al. 2007; polymeric structure and then reduced to form silver
Pillai and Kamat 2004; Sharma et al. 2007, 2008; nanoparticles within the polymeric matrix (Fig. 1)
Yanagihara et al. 2001; Yin et al. 2004; Zeng et al. (Lesniak et al. 2005; Zhang et al. 2008). Dendri-
2007). mer–silver complexes prevent silver ions from
Biological methods involve the production of precipitating and keep silver dispersed in the media
silver nanoparticles utilizing extracts from bio-organ- long enough to be delivered where it is desired
isms as reductant, capping agents or both (Li et al. (Balogh et al. 2001; Lesniak et al. 2005; Zhang
2007; Sanghi and Verma 2009). Such extracts can et al. 2008). Silver ions can also be stabilized in
include proteins, amino acids, polysaccharides, and zeolite channels (Fig. 2) or deposited in activated
vitamins (Eby et al. 2009; Sharma et al. 2009). Plant carbon fibers (Inoue et al. 2002; Ogden et al. 1999;
extracts such as apiin (a glucoside compound) and Pal et al. 2009).
leaf extract from magnolia, Persimmon, geranium, Composites of silver coatings over titanium diox-
and Pine leaf have also been used as reducing agents ide nanoparticles are used in products such as baby
of Ag? to produce silver nanoparticles (Kasthuri bottles and blood-clotting agents to produce antibac-
et al. 2009; Shankar et al. 2003; Song and Kim 2009). terial activity (Yeo and Kang 2008). Other hybrid
Additionally, silver nanoparticles can be synthesized silver nanomaterials may include silver nanoparticles
by several microorganisms such as the bacterial coated onto polyurethane and silver–magnetite com-
strains Bacillus licheniformis, K. pneumonia, and posite nanoparticles (Fe3O4@Ag); both of these
fungi strains such as Verticillium and Fusarium hybrids are utilized for water disinfection (Gong
oxysporum, Aspergillus flavus (Ahmad et al. 2003; et al. 2007; Jain and Pradeep 2005). One of the
Kalishwaralal et al. 2008; Mokhtari et al. 2009; challenges of using silver (or any) nanoparticles for
Mukherjee et al. 2001; Senapati et al. 2004; Vig- water treatment is recovering the particles after the
neshwaran et al. 2007). It is not clear if these treatment process. Silver–magnetite nanoparticles
microbes are impacted (favorably or unfavorably) by offer the potential advantage of being removed by a
exposure to engineered forms of nano-scaled silver, magnet, avoiding release to the environment, and
but their seemingly favorable interactions with silver making possible direct reuse without additional
suggest resistance may be fairly widespread. separation processes. For example, in related
Another procedure utilized to synthesize silver research, magnetite particles proved effective for
nanoparticles is the solvated metal atom dispersion removal of arsenic from water (Mayo et al. 2007;
(SMAD) method (Stoeva et al. 2002). In this method, Yavuz et al. 2006). However, for silver materials, an
a metal is co-vaporized with a solvent onto a liquid additional concern is controlling the release of metal
nitrogen cooled surface, as liquid nitrogen is removed ions into the final produce water.

123
J Nanopart Res (2010) 12:1531–1551 1535

Fig. 1 General formation


scheme of PAMAM
dendrimer complexes and
nanocomposites. PAMAM
structural subunits:
core = ethylenediamine,
branching site = –N\,
chains connecting the
branching sites =
–CH2CH2CONHCH2CH2–.
Terminal groups on the
surface are marked as
–CH2CH2–CO–Z. Silver
ions (represented by M?)
can be pre-organize and
subsequently contained in
the form of solubilized and
stabilize, high-surface area
silver domains. Redrawn
based on (Balogh et al.
2001)

Fig. 2 Silver ions


stabilized in zeolite
channels and ionic
exchange of silver ions with
other cations in the media.
Left picture shows zeolite
type A and right picture
shows zeolite type X.
Adapted from (Auerbach
2003)

Evidence of silver toxicity in microbes Evidence of toxicity to bacteria


and higher organisms
Table 1 presents a concise summary of silver nano-
Here, toxicity refers to any deleterious effects on an material antibacterial studies. Kim et al. reported that
organism upon exposure to silver. Obviously, if the 13.4-nm silver nanoparticles prepared by reduction of
practical intent is to disinfect or sterilize a specific silver nitrate with sodium borohydride show mini-
type of organism, then toxicity may be interpreted mum inhibitory concentration (MIC) against E. coli
as a positive outcome (e.g., antibacterial, antiviral, below 6.6 nM and above 33 nM for S. aureus (Kim
etc.). However, if the same material exerts unin- et al. 2007). In another study, 16-nm silver nanopar-
tended or undesired impacts to other organisms, then ticles generated by gas condensation were able to
such toxicity may be interpreted as a potential completely inhibit colony forming units (CFU)
hazard. ability of E. coli at 60 lg/mL (Raffi et al. 2008).

123
1536 J Nanopart Res (2010) 12:1531–1551

Table 1 Bactericidal activity of nano-scaled silver and silver loaded in zeolite


Silver form Size data Bacterial strain Key aspects References

Silver nanoparticles/ 13.4 nmb E. coli, S. aureus Minimal inhibition concentration against (Kim et al. 2007)
nano-sized silver E. coli was lower than 6.6 nM and
powders higher than 33 nM for S. aureus
16 nm E. coli Complete inhibition of CFU ability at (Raffi et al. 2008)
60 lg/mL
1 lm E. coli, S. aureus CFU reduced by 4 to 5 log units (Smetana et al.
2008)
26 nm Standard strains and Minimal inhibition concentration from (Kvitek et al. 2008)
strains isolated 1.69 to 13.5 lg/mL
from clinical
material
10 nma E. coli, S. aureus Growth inhibition achieved at 5 lg/mL (Vertelov et al.
L. mesenteroides Growth inhibition achieved at 2.5 lg/mL 2008)
Silver nanoparticles 14.1–710 nmc B. subtilis 76% CFU reduction by applying silver (Yoon et al. 2008a,
applied as aerosol nanoparticles aerosol on B. subtilis b)
aerosol
Silver nanoparticles 1.4–7.1 nmb E. coli, S. aureus, Microbial activity increases as silver (Zhang et al. 2008)
stabilized in B. subtilis, content in polymer decreases since
hyperbranched K. mobilis decrease in silver nanoparticle size
polymers
Silver–dendrimer S. aureus, Antimicrobial activity was comparable or (Balogh et al. 2001)
complexes and P. aeruginosa, higher to those of silver nitrate
nanocomposites E. coli solutions. The activity and solubility did
not decrease even in presence of sulfate
or chloride ions
Silver zeolite E. coli CFU reduced by 7 log units in 5 min (Inoue et al. 2002)
Zeolite containing E. coli Minimal bactericidal concentration of (Cowan et al. 2003)
silver and zinc 78 lg/mL
(as Ag?) for bacteria grown is Luria-
Bertani broth
E. coli, S. aureus, Minimal bactericidal concentration of
P. aeruginosa 39 lg/mL
(as Ag?) for bacteria grown is Tryptic
Soy broth
a b c
Note: Size measured by dynamic light scattering, TEM images, and scanning mobility particle sizer

Commercially available silver nanopowders at a S. aureus, vancomycin-resistant Enterococcus


concentration of 300 lg/mL and SMAD-produced faecium, and K. pneumonia when exposed to
silver nanoparticles at a concentration of 30 lg/mL 26-nm silver nanoparticles prepared by the reduc-
were able to reduce (after 10 min contact time) tion of [Ag(NH3)2]? with D-maltose (Kvitek et al.
colony forming units of E. coli and S. aureus from 2008).
2 9 104 CFU/mL to 0 and \20, respectively (Smetana Silver nanoparticles also produced 76% reduction
et al. 2008). of B. subtilis CFU after applying silver nanoparticles
Additionally, MICs ranging from 13.5 to 1.69 lg/mL with a size distribution from 14 to 710 nm in an
were reported for bacterial strains such as S. aureus aerosol form (Yoon et al. 2008a, b). Likewise, silver
CCM 3953, Enterococcus faecalis CCM 4224, E. coli nanoparticles are effective against E. coli, S. aureus,
CCM 3954, and P. aeruginosa CCM 3955, and for and L. mesenteroides. Also, 10-nm MyramistinÒ
strains isolated from human clinical material like stabilized silver nanoparticles inhibit growth of
P. aeruginosa, methicillin-susceptible S. epidermidis, E. coli and S. aureus at 2.5 lg/mL and L. mesen-
methicillin-resistant S. epidermidis, methicillin-resistant teroides at 5 lg/mL (Vertelov et al. 2008).

123
J Nanopart Res (2010) 12:1531–1551 1537

Dendrimer–silver nanocomposites have also been zeolite (Inoue et al. 2002), and MICs of 78 lgAg?/mL
proven effective antibacterials. For example, for E. coli and 39 lgAg?/mL for S. aureus and
poly(amidoamine) dendrimer–silver composites have P. aeruginosa, plus some bactericidal activity against
been used against S. aureus, P. aeruginosa, E. coli, Listeria monocytogenes (Cowan et al. 2003).
B. subtilis, and K. mobilis (Balogh et al. 2001; Zhang
et al. 2008). Additionally, silver ions loaded in zeolites Evidence of toxicity to other microorganisms
elicit antibacterial properties. Two recent studies
demonstrated 7 log reduction in CFUs for E. coli, Silver nanoparticles also inactivate fungi, virii, and
from an initial concentration of 107 CFU/cm3, after algae (Table 2). For example, silver nanoparticles of
5 min of contact time with 333.3 lg/mL of Ag-loaded sizes ranging from 1.4 to 7.1 nm and stabilized in

Table 2 Summary of silver nanoparticles toxicity to other microorganisms


Strain Silver nanoparticles Size (nm) Key aspects Reference

Fungi
A. niger MyramistinÒ stabilized 10a MIC were found to be 5 mg/L (Vertelov et al. 2008)
silver nanoparticles
Silver nanoparticles 1.4–7.1b Formation of inhibition zones (Zhang et al. 2008)
stabilized in hyper around silver nanoparticles
branched polymers inoculated spots in agar plates
S. cerevisiae MyramistinÒ stabilized 10a MIC were found to be 5 mg/L (Vertelov et al. 2008)
silver nanoparticles
T. mentagrophytes Silver nanoparticles 3b IC80 between 1 and 4 mg/L (Kim et al. 2008a, b)
C. Albanicas Silver nanoparticles 3b Silver nanoparticles inhibited (Kim et al. 2008a, b)
micelial formation, which is
responsible for pathogenicity
Silver nanoparticles 3b Antifungal activity may be exerted (Kim et al. 2009a, b)
by cell membrane structure
disruption and inhibition of
normal budding process
Silver nanoparticles 3–18b Catheter coated with silver (Roe et al. 2008)
coated on plastic nanoparticles inhibited growth
catheters and biofilm formation.
Yeast (isolated Silver nanoparticles 13.4b MIC estimated between 6.6 nM (Kim et al. 2007)
from bovine and 13.2 nM
mastitis)
P. citrinum Silver nanoparticles 1.4–7.1b Formation of inhibition zones (Zhang et al. 2008)
stabilized in hyper around silver nanoparticles
branched polymers inoculated spots in agar plates
Viruses
Hepatitis B virus Silver nanoparticles 10b Inhibition of virus replication (Lu et al. 2008)
HIV-1 Silver nanoparticles 16.19 ± 8.68b Only 1–10 nm nanoparticles (Elechiguerra et al. 2005)
attached to virus restraining virus
from attaching to host cells.
Syncitial virus Silver nanoparticles 44% inhibition of Syncitial virus (Sun et al. 2008)
infection
Algae
C. reinhardtii Silver nanoparticles 10–200a EC50 for the photosynthetic yield (Navarro et al. 2008)
was found in 0.35 mg/L of total
silver content after 1 h of
exposure
a b
Note: Size measured by dynamic light scattering and TEM images

123
1538 J Nanopart Res (2010) 12:1531–1551

hyperbranched polymers, and silver nanoparticles are able to inhibit hepatitis B virus replication (Lu
stabilized with MyramistinÒ (size 10 nm) inhibited et al. 2008). Additionally, PVP-coated silver nano-
the growth of A. niger (Tomsic et al. 2009; Vertelov particles in the range 1–10 nm attach to HIV-1 virus,
et al. 2008; Zhang et al. 2008); the same MyramistinÒ inhibiting the virus from attaching to host cells
stabilized particles were found toxic toward S. cere- (Elechiguerra et al. 2005). In other study, PVP-coated
visiae showing a MIC of 5 mg/L. Further, 3-nm silver silver nanoparticles reduced respiratory syncytial
nanoparticles showed IC80 values from 1 to 4 lg/mL virus infection by 44% (Sun et al. 2008). Polysulfone
against T. mentagrophytes and 2 to 4 lg/mL for ultrafiltration membranes impregnated with silver
C. albanicans (Kim et al. 2008a, b); while in other study, nanoparticles of sizes ranging from 1 to 70 nm
it was reported that the antifungal activity of silver showed enhanced virus removal, thus improving
nanoparticles against C. albanicans could be exerted water disinfection via low pressure membrane filtra-
by cell membrane structure disruption leading to tion (Zodrow et al. 2009).
reproduction inhibition (Kim et al. 2009a, b). Addi-
tionally, silver nanoparticles (sizes ranging from 3 to
18 nm) coated in catheters were able to inhibit growth Evidence of toxicity for mammalian cells
of C. albicans. Although, in this case, no analysis of
the antifungal molecular mechanism was done, the Significant evidence has been reported in relation to
authors speculate that silver ions (Ag?) released from the toxicity of silver nanoparticles to higher organ-
the matrix were the antifungal agents (Kim et al. isms. It has been shown toxic to fish such as zebrafish
2008a, b, 2009a, b; Roe et al. 2008). Furthermore, (Asharani et al. 2008; Yeo and Kang 2008; Yeo and
silver nanoparticles suppress yeast growth and show Yoon 2009), Diptera species such as Drosophila
MIC between 6.6 and 13.2 nM (Kim et al. 2007). melanogaster, known asfruit fly (Ahamed et al. 2010)
Silver nanoparticles of sizes from 1.4 to 7.1 nm and and different mammalian cell lines of mice (Brayd-
stabilized in hyperbranched polymers (HPAMAM- ich-Stolle et al. 2005; Hussain et al. 2005), rats (Kim
N(CH3)2/AgNPs composite) inhibit P. citrinum et al. 2008a, b; Sung et al. 2008), and also humans
growth (Zhang et al. 2008). Although information (Asharani et al. 2009; Braydich-Stolle et al. 2005;
about toxicity for algae is limited, silver nanoparticles Hsin et al. 2008; Hussain et al. 2005). This review
reduced the photosynthetic yield of Chlamydomonas presents only a few, brief examples of silver nano-
reinhardtii; in this case, the observed toxicity was material toxicity for mammalian cells (Table 3).
attributed to Ag? ions (Navarro et al. 2008). More detailed, focused reviews on this topic are
Evidence of virus inactivation is also reported in available elsewhere (Chen and Schluesener 2008;
literature. For example, silver nanoparticles of 10 nm Panyala et al. 2008).

Table 3 Evidence of nano-scaled silver toxicity for mammalian cells


Target cell/organism Key aspects Reference

Rat lung cells Reduction in lung function and inflammatory lesions (Sung et al. 2008)
Sprague-Dawley rats Silver nanoparticles accumulation in olfactory bulb (Kim et al. 2008a, b)
and subsequent translocation to the brain
Mouse stem cells Cell leakage and reduction of mitochondrial function (Braydich-Stolle et al. 2005)
Rat liver cells Cell leakage and reduction of mitochondrial function (Hussain et al. 2005)
Human fibrosarcoma Oxidative stress. Low doses produced apoptosis (Arora et al. 2008)
and human skin/carcinoma and higher dose necrosis
Mouse fibroblast 50 lg/mL induced apoptosis to 43.4% of cells (Arora et al. 2009)
Human colon cancer 100 lg/mL produced necrosis to 40.2% of cells
Human glioblastoma Silver nanoparticles were found cytotoxic, (Asharani et al. 2009)
genotoxic and antiproliferative
Human fibroblast Silver nanoparticles were found cytotoxic, (Asharani et al. 2009)
genotoxic and antiproliferative

123
J Nanopart Res (2010) 12:1531–1551 1539

Evidence of silver nanoparticle toxicity for mam- Asharani, a possible mechanism of toxicity to human
malian cells was presented in the in vivo studies cells was proposed (Asharani et al. 2009). Silver
performed by Sung et al. (2008) and Kim et al. nanoparticles would affect the mitochondrial respira-
(2008a, b). In the former, a 90-day inhalation study in tory chain, causing ROS generation and affecting the
rats showed that silver nanoparticles reduce lung production of ATP, which subsequently leads to DNA
function and produce inflammatory lesions in the damage. In this study, the authors also concluded that
lungs. In the later, silver nanoparticles accumulated ‘‘even and small dose of Ag-NP (silver nanoparticles)
in the olfactory bulbs of Sprague-Dawley rats and has the potential to cause toxicity’’ and that silver
also accumulated in the brain. nanoparticles are cytotoxic, genotoxic, and antipro-
Evidence from in vitro studies is also available in the liferative, being as toxic for human glioblastoma as
literature. For example, silver nanoparticles have been for normal human fibroblasts cells.
shown to reduce mitochondrial function and to
increase membrane leakage of mouse spermatogonial
stem cell and rat liver cells (Braydich-Stolle et al. 2005; Mechanisms of silver’s antibacterial properties
Hussain et al. 2005). Studies performed on human
fibrosarcoma and human skin/carcinoma cells with Although the mechanisms behind the activity of
silver nanoparticles used in a topical antimicrobial nano-scaled silver on bacteria are not yet fully
agent concluded that in the presence of the nanopar- elucidated, the three most common mechanisms of
ticles the cellular levels of glutathione are reduced, toxicity proposed to date are: (1) uptake of free silver
indicating oxidative stress, that results in cellular ions followed by disruption of ATP production and
damage and lipid peroxidation (Arora et al. 2008). DNA replication, (2) silver nanoparticle and silver
However, in the study performed by Arora et al. the ion generation of ROS, and (3) silver nanoparticle
dose required to induce apoptosis (0.78–1.56 lg/mL) direct damage to cell membranes. The various
was much smaller than that required to produce observed and hypothesized interactions between
necrosis (12.5 lg/mL) in both cell types. Therefore, silver nanomaterials and bacteria cells are conceptu-
the authors concluded that, after the required in vivo ally illustrated in Fig. 3.
studies, it would be possible to define a safe range for
the application of silver nanoparticles as a topical
antimicrobial agent. Similar differences of the required
concentration to cause apoptosis or necrosis were
found in a second study by the same authors in mouse
fibroblasts and liver cells (Arora et al. 2009). In this
second article, it was suggested that although silver
nanoparticles may enter into the cells, the cellular
antioxidant mechanisms would limit oxidative stress.
Mechanistic studies of silver nanoparticle toxicity
in mammalian cells have considered mouse fibroblast
and human colon cancer cells (Hsin et al. 2008). In
this study, silver nanoparticle doses of 50 lg/mL
induced apoptosis to 43.4% of fibroblast cells, while
100 lg/mL produced necrosis to 40.2% of the cancer
cells. The authors concluded that the apoptotic Fig. 3 Diagram summarizing nano-scaled silver interaction
with bacterial cells. Nano-scaled silver may (1) release silver
mechanisms in fibrosblast cells are a mitochondrial ions and generate ROS; (2) interact with membrane proteins
mediated pathway including the generation of ROS in affecting their correct function; (3) accumulate in the cell
the cell, which activate the apoptosis regulators JNK membrane affecting membrane permeability; and (4) enter into
and p53 proteins inducing protein Bax to migrate to the cell where it can generate ROS, release silver ions, and
affect DNA. Generated ROS may also affect DNA, cell
the surface of the mitochondria. That subsequently membrane, and membrane proteins, and silver ion release will
induces cytochrome C release from mitochondria and likely affect DNA and membrane proteins. Similar pictures
cleavage of PARP. Additionally, in a study done by have been published in (Damm et al. 2008; Neal 2008)

123
1540 J Nanopart Res (2010) 12:1531–1551

Free silver ion uptake proteins (Holt and Bard 2005; Liau et al. 1997;
Petering 1976). Additionally, it has been reported that
Silver nanoparticles have been reported to dissolve Ag? increases DNA mutation frequencies during
generating silver ions and it is thought that in vivo this polymerase chain reactions (Yang et al. 2009).
release would be product of reactions of silver nano- Bacterial cells exposed to milli-molar Ag? doses
particles with H2O2 (Asharani et al. 2009). Asharani has suffer morphological changes such as cytoplasm shrink-
proposed the following reaction as a possible mecha- age and detachment of cell wall membrane, DNA
nism for silver nanoparticle oxidative dissolution. condensation and localization in an electron-light region
in the center of the cell, and cell membrane degradation
2Ag þ H2 O2 þ 2Hþ ! 2Agþ þ 2H2 O E0 ¼ 0:17 V:
allowing leakage of intracellular contents (Feng et al.
2000; Jung et al. 2008). Physiological changes occur
Asharani suggests that in eukaryotic cells this
together with the morphological changes. Bacterial cells
reaction could occur in the mitochondria, where
enter an active, but non-culturable state in which
exists an important concentration of H?. Similarly,
physiological levels can be measured but bacteria are
we hypothesized that a similar mechanism could
not able to growth and replicate.
occur in the bacterial cell membrane where proton
Several studies have linked the toxicity of silver
motive force takes place.
nanoparticles to the release of silver ions. For example,
Another possible mechanism for the oxidative
Smetana et al. observed that silver ions eroded from
dissolution of silver nanoparticles has been reported
high-surface area silver powders prepared by SMAD
by Choi et al., in this case silver is oxidized in the
method interacted and destroyed bacterial cells (Sme-
presence of oxygen. Choi et al. speculated that the
tana et al. 2008). In the same study, a second
observed changed of color of their silver nanoparti-
preparation of silver nanoparticles using water-soluble
cles suspensions, over a week period, would be
ligands was used to obtain silver nanoparticles with
attributed to this mechanism.
higher surface area to improve their antibacterial
4Ag þ O2 þ 2H2 O $ 4Agþ þ 4OH : efficacy. However, the second preparation of silver
nanoparticles showed lower toxicity toward bacterial
The amount of free silver ion measured in this case cells than uncoated powders, suggesting the ligands
was approximately 2.2% of the total silver content in prevented silver ion erosion; thus, diminishing the
the silver nanoparticle suspension (Choi et al. 2008). resulting toxicity. Another possible reason for this
In other article, a 0.1% content of the total silver in result is that the surface coatings prevented adhesion
partially oxidized silver nanoparticles suspensions of silver nanoparticles to the bacterial cell surface, but
was attributed to silver ions (Lok et al. 2007). the authors did not explore this option.
Ionic silver has known antibacterial properties; Hwang et al. observed that Ag? induced the same
thus, it is expected that eluted ions from silver effect in bioluminescence bacteria sensitive to mem-
nanoparticles are responsible for at least a part of brane protein damage and slightly less effect in a strain
their antibacterial properties. At sub-micromolar sensitive to superoxides compared to silver nanopar-
concentrations, Ag? interacts with enzymes of the ticles (Hwang et al. 2008). The authors suggested that
respiratory chain reaction such as NADH dehydro- silver nanoparticles produce silver ions that move
genase resulting in the uncoupling of respiration from inside the cell producing ROS through redox reactions
ATP synthesis. Silver ions also bind with transport with oxygen. In other research, bacterial activity of
proteins leading to proton leakage, inducing collapse activated carbon fiber supported silver was attributed
of the proton motive force (Dibrov et al. 2002; Holt to the synergistic action of silver ions, superoxides,
and Bard 2005; Lok et al. 2006). Silver inhibits the and hydrogen peroxide (Le Pape et al. 2004).
uptake of phosphate and causes the efflux of intra-
cellular phosphate (Schreurs and Rosenberg 1982). Generation of reactive oxygen species
The interaction with respiratory and transport pro-
teins is due to the high affinity of Ag? with thiol Reactive oxygen species (ROS) are natural byprod-
groups present in the cysteine residues of those ucts of the metabolism of respiring organisms. While

123
J Nanopart Res (2010) 12:1531–1551 1541

small levels can be controlled by the antioxidant scavengers, it was determined that superoxide anions,
defenses of the cells such glutathione/glutathione hydrogen peroxide, hydroxyl radicals, and singlet
disulfide (GSH/GSSG) ratio, excess ROS production oxygen contributed to the antibacterial activity
may produce oxidative stress (Nel et al. 2006). The against E. coli (Inoue et al. 2002). In another article,
additional generation of free radicals can attack the bactericidal activity of activated carbon fiber
membrane lipids and lead to a breakdown of mem- supported silver occurred only in the presence of
brane and mitochondrial function or cause DNA oxygen and could not be explained by the silver ions
damage (Mendis et al. 2005). eluted from the fiber (Yoon et al. 2008a, b). These
Metals can act as catalysts and generate ROS in observations together with a study of gene expression
the presence of dissolved oxygen (Stohs and Bagchi related to oxidative stress lead the authors to suggest
1995). In this context, silver nanoparticles may that the strong bactericidal activity of this materials
catalyze reactions with oxygen leading to excess free resulted from the combined effects of silver ions,
radical production. Studies done in eukaryotic cells superoxides ions, and hydrogen peroxide (Le Pape
suggest that silver nanoparticles inhibit the antioxi- et al. 2004).
dant defense by interacting directly with GSH, A study of the toxicity effects of different silver
binding GSH reductase or other GSH maintenance forms on nitrifying bacteria showed that not only
enzymes (Carlson et al. 2008). This could decrease Ag? and AgCl, but also silver nanoparticles are able
the GSH/GSSG ratio and, subsequently, increase to induce intracellular ROS generation. Moreover, the
ROS in the cell. Silver ions eluted from nano-scaled bacterial inhibition of each of these silver species
silver or chemisorbed on its surface may also be correlates with their individual generation of intra-
responsible for the generation of ROS by serving as cellular ROS concentration. However, at the same
electron acceptor. In bacterial cells, silver ions would level of intracellular ROS concentration, silver
likely induce the generation of ROS by impairing the nanoparticles appeared more toxic than the other
respiratory chain enzymes through direct interactions species indicating that other factors besides the
with thiol groups in these enzymes or the superoxide- generation of intracellular ROS played a role in the
radical scavenging enzymes such as superoxide overall toxicity (Choi et al. 2008). In other research, a
dismutases (Park et al. 2009). ROS generation from panel of recombinant bioluminescent bacteria was
silver nanoparticles and silver ions may also be used to test different pathways of silver nanoparticles
induced photocatalytically; however, no correlations toxicity such as oxidative stress, DNA damage and
have been reported between bacterial toxicity and protein or membrane damage in bacterial strains. The
photocatalytic ROS concentration (Choi and Hu addition of silver nanoparticles to growing bacterial
2008). cultures leaded to the production of superoxide
Evidence of toxicity related to ROS generated radicals, but not of hydroxyl radicals. The authors
from silver nanoparticles and silver ions, released conclude that toxicity occurs via membrane protein
from or chemisorbed on its surface, has been shown damage and oxidative stress, but not DNA damage
previously. Kim et al. determined the existence of (Hwang et al. 2008).
free radicals from silver nanoparticles by means of
spin resonance measurements (Kim et al. 2007). They Direct cell membrane damage
observed that silver nanoparticles and silver nitrate
toxicity was abolished in the presence of an antiox- Silver nanoparticles interact with the bacterial mem-
idant, these results leaded them to suggest that the brane and are able to penetrate inside the cell.
antimicrobial mechanisms of silver nanoparticles Transmission electron microscopy data show that
against S. aureus and E. coli was related to the silver nanoparticles adhere to and penetrate into
formation of free radicals from the surface of silver E. coli cells and also are able to induce the formation
nanoparticles and subsequent free radical induced of pits in the cell membrane (Choi et al. 2008; Raffi
membrane damage. et al. 2008; Sondi and Salopek-Sondi 2004). Silver
Bactericidal activity of silver ions loaded in nanoparticles have been observed within E. coli cells
nanoporous materials such as zeolites has also been albeit at sizes much smaller than the original particles;
related to the generation of ROS. Using ROS moreover, silver nanoparticles with oxidized surfaces

123
1542 J Nanopart Res (2010) 12:1531–1551

induce the formation of ‘‘huge holes’’ in E. coli depletion, i.e., the formation of irregular-shaped pits
surfaces after the interaction and large portions of the in the outer membrane and change in membrane
cellular content seemed to be ‘‘eaten away’’ (Smetana permeability by the progressive release of LPS
et al. 2008). molecules and membrane proteins (Amro et al.
Silver nanoparticle accumulation on the cell mem- 2000; Sondi and Salopek-Sondi 2004). This may be
brane and uptake within the cell has also been reported fairly general for gram-negative bacteria. It has been
for other bacteria such as V. cholera, P. aeruginosa, reported that extrusion pump systems such as Mex-
and S. typhus. In these cases, only nanoparticles AB-OprM system of P. aeruginosa could also play an
smaller than 10 nm attached to bacteria cell mem- important role in controlling the accumulation of
branes or where observed inside the bacteria (Morones silver nanoparticles in living cells (Xu et al. 2004).
et al. 2005). However, in other research silver The same pump systems are responsible for the silver
nanoparticles with sizes up to 80 nm were transported resistance of several bacteria (see ‘‘Mechanisms of
through the inner and outer membrane of P. aerugin- silver resistance’’ section).
osa (Xu et al. 2004). Silver nanoparticle composites Despite the mechanism of interaction involved, it is
and silver nanoparticles stabilized with surfactants are evident that silver nanoparticles attached to bacterial
also thought to interact with cell membranes. Myram- cell membranes increase permeability and disturb
istinÒ capped silver nanoparticles showed notable respiration. Proteomic data show the accumulation of
antibacterial activity against gram-positive and envelope protein precursors in E. coli cells after
-negative bacteria, it was speculated that MyramistinÒ exposure to silver nanoparticles (Lok et al. 2006).
anchors to the cell wall and weakens it allowing Energy from ATP and proton motive force is required
penetration of silver nanoparticles inside the cells in order to newly synthesize envelope proteins to be
(Vertelov et al. 2008). Composites of HPAMAM- translocated to the membrane, therefore cytoplasmic
N(CH3)2 silver nanoparticle are thought to catch accumulation of protein precursors suggests dissipa-
bacteria by ionic interactions between the cationic tion of proton motive force and depletion of intracel-
polymer and the negative cell membrane, followed by lular levels of ATP. In a similar way, silver ions have
release the silver nanoparticles (Zhang et al. 2008). also been linked to the collapse of proton motive force
The detailed mechanism by which silver nanopar- and destabilization of the cell membrane, but the
ticles interact with cytoplasmic membranes and are concentrations at which this occur are much higher
able to penetrate inside cells is not fully determined. than the ones of silver nanoparticles (micromolar vs.
One hypothesis is that the interaction between nanomolar) (Dibrov et al. 2002; Lok et al. 2006).
nanoparticles and bacterial cells are due to electro-
static attraction between negatively charged cell Mechanisms of silver resistance
membranes and positively charged nanoparticles
(Raffi et al. 2008). However, this mechanism does Bacterial strains resistant to specific toxicants are
not likely explain the adhesion and uptake of naturally selected in environments where these agents
negatively charged silver nanoparticles. It has been are present (Gupta and Silver 1998). In this way, the
also proposed that the preferential sites of interaction widespread use of silver, whether in nano or bulk
for silver nanoparticles and membrane cells might be form, could lead to selection of bacterial communities
sulfur containing proteins—in a similar way as silver exhibiting silver resistance. Since most commercial
interacts with thiol groups of respiratory chain uses of silver and silver nanoparticles relate to
proteins and transport proteins, interfering with their fighting infection, widespread silver resistance is a
proper function (Morones et al. 2005). This seems growing concern. In fact, it has been suggested that it
more likely than electrostatic attraction because already occurs extensively, but it not recognized due
evidence of protein membrane damage on bacteria to a lack of testing for silver resistance (Silver et al.
after silver nanoparticle exposure has been demon- 2006). For example, 10% of the enteric bacteria
strated with bioluminescence bacteria (Hwang et al. tested randomly in a hospital in Chicago, IL, showed
2008). genes for Ag? resistance (Silver 2003).
Another proposed mechanism of E. coli membrane Silver resistance in bacteria (and other heavy
damage by silver nanoparticles relates to metal metals) is often encoded by plasmids genes, as for

123
J Nanopart Res (2010) 12:1531–1551 1543

example the Salmonella plasmid pMG101, within the This increase in bioavailability increases sensitivity
IncHI incompatibility group, as well as in other five of the silver-resistant and silver-sensitive bacteria.
plasmids in the same group (Gupta et al. 2001). Similar, behavior is also observed for silver in the
Although resistance is mainly encoded by plasmids, presence of other halides (Gupta et al. 1998).
bacterial chromosomes have also been reported to
encode Ag? resistance genes (sil) in strains such as Further insights about potential toxicity to higher
E. coli K-12 and O157:H7 (Gupta et al. 2001; Silver organisms
et al. 2006). The plasmid pMG101 has been studied in
detail (Gupta et al. 1999; Silver 2003; Silver et al. Silver nanomaterials exhibit outstanding antibacterial
2006), this plasmid has nine genes, whose products properties that give rise to many potentially beneficial
have been identified to be proteins responsible for applications. However, misuses of silver nanoparti-
heavy metal resistance. Here, the resistance is cles as a bactericide may also result in unintended
achieved by two efflux systems SilCBA and SilP exposure to higher organisms, including humans.
acting in combination with two periplasmic binding This is of concern because silver nanoparticles appear
proteins SilE and SilF. The complex SilCBA is to have toxic effects in higher organisms (‘‘Evidence
constituted by three proteins SilC in the outer mem- of silver toxicity in microbes and higher organisms’’
brane, SilA in the inner membrane and SilB that links section). While eukaryotic and prokaryotic cells are
SilA and SilC. This complex acts as an antiporter that different in many ways, understanding the funda-
transports Ag? out of the cell by pumping a proton mental mechanisms governing silver nanoparticle
inside. In turn, SilC is a P-type ATPase that transports toxicity in bacterial cells may shed light on the
Ag? from the cytoplasm to the perisplasm. These two potential effects of silver nanoparticles on important
efflux pumps work jointly with proteins SilE and SilF, organelles such as mitochondria.
which bind to Ag?. SilF is thought to act as a Mitochondria are believed to have evolved from
‘‘chaperone’’ by taking one Ag? ion from its release bacteria (i.e., the ‘‘serial primary endosymbiosis
site in SilP to its uptake site in SilCBA while SilE theory’’) and they share several characteristics
binds 5 Ag? ions to 10 histidines preventing silver ions (Kutschera 2009). For example, the inner membrane
entrance to the cytoplasm (Gupta et al. 1999). of a mitochondrion and the prokaryotic cell mem-
Limited evidence has been reported of the resis- brane are structurally very similar. Respiration occurs
tance shown by silver-resistant strains to silver in bacterial cells similarly as in the mitochondria of
nanoparticles. However, in one study, it was reported eukaryotes, including electron transport, ATP syn-
that albumin-stabilized oxidized silver nanoparticles thesis, and proton motive force. Likewise mitochon-
were unable to inhibit growth of 116 AgNO3R and drial DNA, the bacterial DNA are analogous, and the
J53 (pMG101) silver-resistant strains even when division of the mitochondria is similar to fission of
apply at a concentration of up to 80 nM (Lok et al. prokaryotes (Slonczewski and Foster 2009). Based on
2007). Therefore, it seems that silver resistance may these similarities, it is hypothesized that silver
also be of concern for the efficacy of silver nanopar- nanoparticles may affect mitochondria in higher
ticles use as antibacterial agent. organisms via similar mechanisms as in bacterial
Bacterial resistance and sensitivity to silver is cells—assuming they can be internalized by the cell.
affected by environmental conditions such as the
presence of halides in the media due to mainly the
variation of silver bioavailability experienced at Factors affecting the toxicity of silver
different concentrations of Cl- and other halides nanomaterials
such as Br- and I- (Gupta et al. 1998). For moderate
Cl- concentration, silver is precipitated as AgCl, Several factors have been reported to influence silver
which decreases silver bioavailability and increases nanoparticle toxicity like particle size, shape, crys-
bacterial silver resistance, as observed for E. coli J53 tallinity, surface chemistry, capping agents, as well,
(pMG101). Nevertheless, silver bioavailability rises as for environmental factors such as pH, ionic
when ionic complexes such as AgCl2-, AgCl32- are strength, and the presence of ligands, divalent
formed as product of a higher halide concentration. cations, and macromolecules (Carlson et al. 2008;

123
1544 J Nanopart Res (2010) 12:1531–1551

Choi and Hu 2008; Kvitek et al. 2008; Lok et al. stabilize silver nanoparticle dispersions and enhance
2007; Morones et al. 2005; Pal et al. 2007; Smetana biocidal activity (Dorjnamjin et al. 2008; Kvitek et al.
et al. 2008). Many publications have shown size- 2008; Yu 2007). However, some ligand-capped silver
dependent toxicities of silver nanoparticles (Carlson nanoparticles—although highly stable and monodis-
et al. 2008; Choi and Hu 2008; Martinez-Castanon perse in suspension—were less bioactive because the
et al. 2008; Morones et al. 2005). As particle size capping agent hindered release of silver ions (Sme-
decreases, the specific surface area increases leaving tana et al. 2008).
a higher number of atoms exposed on the surface Environmental conditions such as pH, ionic
available for redox, photochemical, and biochemical strength, presence of complexing agents, and natural
reactions in addition to physicochemical interactions organic matter, also affect the toxicity of silver
with cells. nanoparticles. High salt concentrations and pH values
As discussed previously, one of the key mecha- close to the isoelectric point promote nanoparticle
nisms for silver nanomaterials to exert biocidal aggregation by screening electrical double layer
activity is through the release of silver ions. As the repulsion (Nel et al. 2009). However, water chemistry
rate of ion release, in general, is proportional to also governs silver dissolution and/or re-precipitation
particle surface area, nanoparticles can release ions through various possible redox and precipitation
more rapidly than larger particles and macroscopic reactions (Lok et al. 2007). Dissolved Ag? ions are
materials. In effect, chemisorbed silver ions would be sparingly soluble in the presence of various ligands
the cause of the antibacterial activity observed by such as chloride (pK = 9.75), sulfate (pK = 4.9),
Lok et al., who reported that only partially oxidized sulfide (pK = 49), hydroxide (pK = 7.8) and dis-
silver nanoparticles exhibit antibacterial activities, solved organic carbon (pK [ 7.5) (Choi et al. 2008;
while zero valent silver nanoparticles do not (Lok Gao et al. 2009). The released Ag? can also form Ag0
et al. 2007). This surface area effect also influences -containing clusters through light or chemical reduc-
RO generation. For example, at the same silver tion (Morones et al. 2005). Therefore, nanomaterials in
concentration, silver nanoparticles of 15 nm gener- aqueous suspensions must be considered in a contin-
ated higher levels of ROS in macrophages than 30 uous state of flux where the apparent speciation is
and 50 nm particles (Carlson et al. 2008). controlled by the aquatic media pH, redox potential,
High atom densities at h111i facets increased the ionic composition, and exposure to light.
toxicity of silver nanoparticles to several bacterial Previously, cysteine ligands and chloride were
strains; the increased toxicity was the result of the used to scavenge or precipitate eluted silver ions from
higher reactivity presented by the h111i facets silver nanoparticles; thus, diminishing their toxicity
(Morones et al. 2005). Silver nanoparticle shape (Navarro et al. 2008). Sulfide ligands have also been
may also be a factor. Truncated triangular nanoplates used to reduce the inhibition of silver nanoparticles to
exert stronger antibacterial activity than spherical- nitrifying bacteria (Choi et al. 2009). More recently,
and rod-shaped silver nanoparticles because they it was reported that halide ions act as precipitating
contain more h111i facets; thus, they would be more agents and profoundly affect the ‘‘bioavailability’’ of
reactive (Pal et al. 2007). Hence, the antibacterial Ag? in unexpected ways (Silver 2003). For example,
properties of silver nanoparticles are related to both at low chloride concentrations, soluble Ag? binds to
size (i.e., reactive surface area) and crystallinity (i.e., cell membranes impacting respiration and producing
surface reactivity). other toxics effects (Gupta et al. 1998). As chloride
Stability of silver nanoparticles also influences concentration increases, silver becomes less bioavail-
toxicity since the formation of aggregates tends to able because the solubility of AgCl is very low.
decrease biocidal activity (Kvitek et al. 2008; Shriv- However, further increase in halide concentration
astava et al. 2007; Teeguarden et al. 2007). Different results in the formation of water-soluble anionic
surfactants and polymers (e.g., sodium dodecyl complexes in the form of AgCl2- and AgCl32-.
sulfate, polyoxyethylene-sorbitan monooleate, poly- These anionic complexes are more bioavailable and
vinylpyrrolidone, Na?-carrying poly(glutamic acid), increase silver toxicity to both silver-sensitive and
hydroxyl functionalized ionic liquids and hydroxyl silver-resistant bacteria. Other halides such as Br-
functionalized cationic surfactants) have been used to and I- have similar effects, but the concentrations at

123
J Nanopart Res (2010) 12:1531–1551 1545

which ionic complexes form depends on their However, if the amount of nano-scaled silver entering
respective solubility limits. sewers becomes higher than the tolerable levels for
Finally, aerobic versus anaerobic conditions are microbial communities in wastewater treatment
important when assessing the antibacterial properties plants, critical environmental infrastructure might
of silver loaded in zeolite or activated carbon fibers be impacted. Further, there is mounting evidence that
(Inoue et al. 2002; Le Pape et al. 2002; 2004). silver nanoparticles exhibit an array of cytotoxic and
Dissolved oxygen is important in these cases because genotoxic effects in higher organisms. This raises
ROS contributes significantly to bactericidal activity. concern about possible impacts to higher organisms
Dissolved organic carbon (DOC) seems also to including humans.
influence the toxicity of silver nanoparticles, specif- The antibacterial mechanisms of silver nanomate-
ically, toxicity may decrease for aquatic invertebrates rials are not fully elucidated, but the prevailing
as the concentration DOC increases (Gao et al. 2009). paradigm suggests various combinations of: (1) silver
A concise summary of the factors discussed above is ion release followed by cellular uptake and a cascade
presented in Table 4. Since water chemistry dramat- of intracellular reactions, (2) extracellular and intra-
ically impacts the toxicity for virtually all forms of cellular generations of ROS, and (3) direct interac-
silver nanomaterials, it may be difficult to predict tions between nano-scaled silver and cell membranes.
environmental impacts using laboratory data produced At sub-micromolar concentrations, silver ions are
in relatively simple aquatic media or media that is not internalized and react with thiol groups of cellular
very representative of natural aquatic chemistries. This proteins, which lead to uncoupling of ATP synthesis
presents a major hurdle to science-based policy from respiration, loss of proton motive force, and
development for environmental protection. interference with the phosphate efflux system. At
milli-molar levels, silver nanoparticles induce detach-
ment of the cell wall membrane from the cytoplasm,
Summary and future research possible release of intracellular content, DNA con-
densation and loss of replication ability. ROS
In summary, silver nanomaterials exhibit broad- produce oxidative stress resulting in membrane lipid
spectrum biocidal activity toward bacteria, fungi, and DNA damage. Finally, silver nanoparticles
viruses, and algae. This motivates its use in a large increase cell membrane permeability and, subse-
number of biomedical and environmental applica- quently, penetrate inside cells to induce any one or
tions as well as a growing list of consumer products. the entire cascade of effects just described.

Table 4 Factors affecting nano-scaled silver toxicity


Factor Tendency Possible explanation

Particle size Smaller particles sizes tend to enhance As size decrease, there is lager number of atoms on
antibacterial properties the surface available to interact with bacteria or to
release a higher amount of silver ions
Particle stability Higher stability produces a higher Non-stable nanoparticles will tend to form
antibacterial properties aggregates thus surface area will be reduced and
the density of atoms available on the surface will
be lower (Kvitek et al. 2008; Shrivastava et al.
2007; Teeguarden et al. 2007)
Particle shape Particles with shapes containing more\111[ \111[ facets would contain larger atom densities
facets like triangular particles tend to have thus more atoms available for interaction
strongest antibacterial properties (Morones et al. 2005)
Water chemistry Depending in a case to case base Since water chemistry affects particle suspension/
solubility, particle size distribution, as well as,
bacterial ability to face environmental stresses,
water chemistry will affect the interaction between
nano-scaled silver and bacteria thus influencing the
resulting toxicity

123
1546 J Nanopart Res (2010) 12:1531–1551

Silver nanoparticle toxicity is influenced by intrin- Ahmad A, Mukherjee P, Senapati S, Mandal D, Khan M,
sic nanoparticle features like size, shape, chemistry, Kumar R, Sastry M (2003) Extracellular biosynthesis of
silver nanoparticles using the fungus Fusarium oxyspo-
crystallinity, and capping agents, but also by the rum. Colloids Surf B 28:313–318. doi:10.1002/cbic.2007
aquatic chemistry through such factors as solution 00592
pH, redox state, ionic strength, and ionic composi- Amro N, Kotra L, Wadu-Mesthrige K, Bulychev A, Mobashery S,
tion. Most of the toxicity data presented in the Liu G (2000) High-resolution atomic force microscopy studies
of the Escherichia coli outer membrane: structural basis for
literature are obtained in relatively simple media like permeability. Langmuir 16:2789–2796. doi:10.1021/la9910
distilled water or cell culture media, which do not 13x
reflect the aquatic environment inside living organ- Arora S, Jain J, Rajwade J, Paknikar K (2008) Cellular responses
isms or the natural environment. Hence, the surface induced by silver nanoparticles: in vitro studies. Toxicol
Lett 179:93–100. doi:10.1016/j.toxlet.2008.04.009
chemistry, reactivity, and state of dispersion achieved Arora S, Jain J, Rajwade JM, Paknikar KM (2009) Interactions of
in the laboratory may not be relevant for assessing silver nanoparticles with primary mouse fibroblasts and liver
behavior in real systems. In natural waters, the wide cells. Toxicol Appl Pharmacol 236:310–318. doi:10.1016/
variation of pH, ionic strength, ionic composition, j.taap.2009.02.020
Asharani P, Wu Y, Gong Z, Valiyaveettil S (2008) Toxicity of
and natural organic matter will induce widely varying silver nanoparticles in zebrafish models. Nanotechnology
aggregation states of silver nanoparticles; thus, 19:1–8. doi:10.1088/0957-4484/19/25/255102
resulting in widely varying antimicrobial activities Asharani PV, Mun GLK, Hande MP, Valiyaveettil S (2009)
and toxicities. In addition, most toxicity data are Cytotoxicity and genotoxicity of silver nanoparticles in
human cells. ACS Nano 3:279–290. doi:10.1021/nn800
obtained by dispersing silver nanomaterials in or on 596w
nutrient rich growth media; however, oligotrophic Auerbach SM (2003) Zeolite science and technology. Marcel
conditions prevail in most natural environmental Dekker, New York
systems. Microorganisms in these conditions may be Bajpai S, Mohan Y, Bajpai M, Tankhiwale R, Thomas V
(2007) Synthesis of polymer stabilized silver and gold
more or less vulnerable to silver than predicted by nanostructures. J Nanosci Nanotechnol 7:2994–3010. doi:
laboratory studies. 10.1166/jnn.2007.911
Although significant progress has been made to Balogh L, Swanson D, Tomalia D, Hagnauer G, McManus A
elucidate the mechanisms of silver nanomaterial (2001) Dendrimer–silver complexes and nanocomposites
as antimicrobial agents. Nano Lett 1:18–21. doi:10.1021/
toxicity, further research is required to fully under- nl005502p
stand the processes involved and to safely exploit the Batabyal S, Basu C, Das A, Sanyal G (2007) Green chemical
tremendous antimicrobial properties of silver without synthesis of silver nanowires and microfibers using starch. J
jeopardizing human health, critical infrastructure, and Biobased Mater Bioenergy 1:143–147. doi:10.1166/jbmb.
2007.016
the environment. Future in vivo, in vitro, and Benn T, Westerhoff P (2008) Nanoparticle silver released into
environmental studies should consider more system- water from commercially available sock fabrics. Environ
atically the various effects of aquatic chemistry on Sci Technol 42:4133–4139. doi:10.1021/es7032718
nano-scaled silver fate, transport, and toxicity. Braydich-Stolle L, Hussain S, Schlager J, Hofmann M (2005) In
vitro cytotoxicity of nanoparticles in mammalian germline
stem cells. Toxicol Sci 88:412–419. doi:10.1093/toxsci/
Acknowledgments Financial support for this research was kfi256
provided by the University of California Toxic Substances Brown C, Parchaso F, Thompson J, Luoma S (2003) Assessing
Research and Training Program: Lead Campus Program in toxicant effects in a complex estuary: a case study of
Nanotoxicology. effects of silver on reproduction in the bivalve, Potamo-
corbula amurensis, in San Francisco Bay. Hum Ecol Risk
Assess 9:95–119. doi:10.1080/713609854
References Carlson C, Hussain SM, Schrand AM, Braydich-Stolle LK,
Hess KL, Jones RL, Schlager JJ (2008) Unique cellular
Abid J, Wark A, Brevet P, Girault H (2002) Preparation of interaction of silver nanoparticles: size-dependent gener-
silver nanoparticles in solution from a silver salt by laser ation of reactive oxygen species. J Phys Chem B 112:
irradiation. Chem Commun 792–793. doi: 10.1039/ 13608–13619. doi:10.1021/jp712087m
b200272h Chen C, Chiang C (2008) Preparation of cotton fibers with anti-
Ahamed M, Posgai R, Gorey TJ, Nielsen M, Hussain SM, bacterial silver nanoparticles. Mater Lett 62:3607–3609. doi:
Rowe JJ (2010) Silver nanoparticles induced heat shock 10.1016/j.matlet.2008.04.008
protein 70, oxidative stress and apoptosis in Drosophila Chen X, Schluesener H (2008) Nanosilver: a nanoproduct in
melanogaster. Toxicol Appl Pharmacol 242:263–269. doi: medical application. Toxicol Lett 176:1–12. doi:10.1016/
10.1016/j.taap.2009.10.016 j.toxlet.2007.10.004

123
J Nanopart Res (2010) 12:1531–1551 1547

Chi Z, Liu R, Zhao L, Qin P, Pan X, Sun F, Hao X (2009) A fluid drainages. Neurol Res 30:285–287. doi:10.1179/
new strategy to probe the genotoxicity of silver nano- 016164107x229902
particles combined with cetylpyridine bromide. Spectro- Gao J, Youn S, Hovsepyan A, Llaneza VL, Wang Y, Bitton G,
chim Acta A 72:577–581. doi:10.1016/j.saa.2008.10.044 Bonzongo JCJ (2009) Dispersion and toxicity of selected
Choi O, Hu Z (2008) Size dependent and reactive oxygen species manufactured nanomaterials in Natural River water sam-
related nanosilver toxicity to nitrifying bacteria. Environ ples: effects of water chemical composition. Environ Sci
Sci Technol 42:4583–4588. doi:10.1021/es703238h Technol 43:3322–3328. doi:10.1021/es803315v
Choi O, Deng K, Kim N, Ross L, Surampalli R, Hu Z (2008) Gong P, Li HM, He XX, Wang KM, Hu JB, Tan WH, Zhang
The inhibitory effects of silver nanoparticles, silver ions, SC, Yang XH (2007) Preparation and antibacterial activ-
and silver chloride colloids on microbial growth. Water ity of Fe3O4@Ag nanoparticles. Nanotechnology 18. doi:
Res 42:3066–3074. doi:10.1016/j.watres.2008.02.021 10.1088/0957-4484/18/28/285604
Choi O, Cleuenger T, Deng B, Surampalli R, Ross L, Hu Z Gulrajani M, Gupta D, Periyasamy S, Muthu S (2008) Prepa-
(2009) Role of sulfide and ligand strength in controlling ration and application of silver nanoparticles on silk for
nanosilver toxicity. Water Res 43:1879–1886. doi:10.1016/ imparting antimicrobial properties. J Appl Polym Sci 108:
j.watres.2009.01.029 614–623. doi:10.1002/app.27584
Cowan M, Abshire K, Houk S, Evans S (2003) Antimicrobial Gupta A, Silver S (1998) Molecular genetics—silver as a bio-
efficacy of a silver-zeolite matrix coating on stainless cide: Will resistance become a problem? Nat Biotechnol 16:
steel. J Ind Microbiol Biotechnol 30:102–106. doi:10.1007/ 888. doi: 10.1038/nbt1098-888
s10295-002-0022-0 Gupta A, Maynes M, Silver S (1998) Effects of halides on
Damm C, Munstedt H (2008) Kinetic aspects of the silver ion plasmid-mediated silver resistance in Escherichia coli.
release from antimicrobial polyamide/silver nanocom- Appl Environ Microbiol 64:5042–5045
posites. Appl Phys A 91:479–486. doi:10.1007/s00339- Gupta A, Matsui K, Lo JF, Silver S (1999) Molecular basis for
008-4434-1 resistance to silver cations in Salmonella. Nat Med 5:183–
Damm C, Munstedt H, Rosch A (2008) The antimicrobial 188. doi:10.1038/5545
efficacy of polyamide 6/silver-nano- and microcomposites. Gupta A, Phung LT, Taylor DE, Silver S (2001) Diversity of
Mater Chem Phys 108:61–66. doi:10.1016/j.matchemphys. silver resistance genes in IncH incompatibility group
2007.09.002 plasmids. Microbiology 147:3393–3402
Dibrov P, Dzioba J, Gosink K, Hase C (2002) Chemiosmotic Ha H, Xu W, An J, Li D, Zhao B (2006) A simple method to
mechanism of antimicrobial activity of Ag? in Vibrio synthesize triangular silver nanoparticles by light irradi-
cholerae. Antimicrob Agents Chemother 46:2668–2670. ation. Spectrochim Acta A 64:956–960. doi:10.1016/
doi:10.1128/AAC.46.8.2668-2670.2002 j.saa.2005.09.004
Dorjnamjin D, Ariunaa M, Shim Y (2008) Synthesis of silver Hernandez-Sierra J, Ruiz F, Pena D, Martinez-Gutierrez F,
nanoparticles using hydroxyl functionalized ionic liquids Martinez A, Guillen A, Tapia-Perez H, Castanon G
and their antimicrobial activity. Int J Mol Sci 9:807–819. (2008) The antimicrobial sensitivity of Streptococcus
doi:10.3390/ijms9050807 mutants to nanoparticles of silver, zinc oxide, and gold.
Eby D, Schaeublin N, Farrington K, Hussain S, Johnson G Nanomed Nanotechnol 4:237–240. doi:10.1016/j.nano.
(2009) Lysozyme catalyzes the formation of antimicrobial 2008.04.005
silver nanoparticles. ACS Nano 3:984–994. doi:10.1021/ Hlidek P, Biederman H, Choukourov A, Slavinska D (2008)
nn900079e Behavior of polymeric matrices containing silver inclu-
Elechiguerra J, Burt J, Morones J, Camacho-Bragado A, Gao X, sions. 1—Review of adsorption and oxidation of hydro-
Lara H, Yacaman M (2005) Interaction of silver nanopar- carbons on silver surfaces/interfaces as witnessed by FT-
ticles with HIV-1. J Nanobiotechnol 3:6. doi:10.1186/ IR spectroscopy. Plasma Process Polym 5:807–824. doi:
1477-3155-3-6 10.1002/ppap.200800083
Falletta E, Bonini M, Fratini E, Lo Nostro A, Pesavento G, Holt K, Bard A (2005) Interaction of silver(I) ions with the
Becheri A, Lo Nostro P, Canton P, Baglioni P (2008) respiratory chain of Escherichia coli: an electrochemical
Clusters of poly(acrylates) and silver nanoparticles: struc- and scanning electrochemical microscopy study of the
ture and applications for antimicrobial fabrics. J Phys Chem antimicrobial mechanism of micromolar Ag. Biochemis-
C 112:11758–11766. doi:10.1021/jp8035814 try 44:13214–13223. doi:10.1021/bi0508542
Feng Q, Wu J, Chen G, Cui F, Kim T, Kim J (2000) A Hsin Y, Chena C, Huang S, Shih T, Lai P, Chueh P (2008) The
mechanistic study of the antibacterial effect of silver ions apoptotic effect of nanosilver is mediated by a ROS- and
on Escherichia coli and Staphylococcus aureus. J Biomed JNK-dependent mechanism involving the mitochondrial
Mater Res 52:662–668 pathway in NIH3T3 cells. Toxicol Lett 179:130–139. doi:
Furno F, Morley KS, Wong B, Sharp BL, Arnold PL, Howdle 10.1016/j.toxlet.2008.04.015
SM, Bayston R, Brown PD, Winship PD, Reid HJ (2004) Huang H, Yang X (2004) Synthesis of polysaccharide-stabilized
Silver nanoparticles and polymeric medical devices: a gold and silver nanoparticles: a green method. Carbohydr
new approach to prevention of infection? J Antimicrob Res 339:2627–2631. doi:10.1016/j.carres.2004.08.005
Chemother 54:1019–1024. doi:10.1093/Jac/Dkh478 Hussain S, Hess K, Gearhart J, Geiss K, Schlager J (2005) In vitro
Galiano K, Pleifer C, Engelhardt K, Brossner G, Lackner P, toxicity of nanoparticles in BRL 3A rat liver cells. Toxicol
Huck C, Lass-Florl C, Obwegeser A (2008) Silver seg- In Vitro 19:975–983. doi:10.1016/j.tiv.2005.06.034
regation and bacterial growth of intraventricular catheters Hwang E, Lee J, Chae Y, Kim Y, Kim B, Sang B, Gu M (2008)
impregnated with silver nanoparticles in cerebrospinal Analysis of the toxic mode of action of silver nanoparticles

123
1548 J Nanopart Res (2010) 12:1531–1551

using stress-specific bioluminescent bacteria. Small 4:746– Kutschera U (2009) Symbiogenesis, natural selection, and the
750. doi:10.1002/smll.200700954 dynamic Earth. Theory Biosci 128:191–203. doi:10.1007/
Ingle A, Gade A, Pierrat S, Sonnichsen C, Rai M (2008) s12064-009-0065-0
Mycosynthesis of silver nanoparticles using the fungus Kvitek L, Panacek A, Soukupova J, Kolar M, Vecerova R,
Fusarium acuminatum and its activity against some Prucek R, Holecova M, Zboril R (2008) Effect of sur-
human pathogenic bacteria. Curr Nanosci 4:141–144. doi: factants and polymers on stability and antibacterial
10.2174/157341308784340804 activity of silver nanoparticles (NPs). J Phys Chem C 112:
Inoue Y, Hoshino M, Takahashi H, Noguchi T, Murata T, 5825–5834. doi:10.1021/jp711616v
Kanzaki Y, Hamashima H, Sasatsu M (2002) Bactericidal Le Pape H, Solano-Serena F, Contini P, Devillers C, Maftah A,
activity of Ag-zeolite mediated by reactive oxygen spe- Leprat P (2002) Evaluation of the anti-microbial properties
cies under aerated conditions. J Inorg Biochem 92:37–42. of an activated carbon fibre supporting silver using a dynamic
doi:10.1016/S0162-0134(02)00489-0 method. Carbon 40:2954. doi:10.1016/S0008-6223(02)
Jain P, Pradeep T (2005) Potential of silver nanoparticle-coated 00246-4
polyurethane foam as an antibacterial water filter. Bio- Le Pape H, Solano-Serena F, Contini P, Devillers C, Maftah A,
technol Bioeng 90:59–63. doi:10.1002/bit.20368 Leprat P (2004) Involvement of reactive oxygen species in
Jin W, Jeon H, Kim J, Youk J (2007) A study on the preparation of the bactericidal activity of activated carbon fibre supporting
poly(vinyl alcohol) nanofibers containing silver nanoparti- silver bactericidal activity of ACF(Ag) mediated by ROS.
cles. Synthetic Met 157:454–459. doi:10.1016/j.synthmet. J Inorg Biochem 98:1054–1060. doi:10.1016/j.jinorgbio.
2007.05.011 2004.02.025
Jung W, Koo H, Kim K, Shin S, Kim S, Park Y (2008) Antibac- Lesniak W, Bielinska A, Sun K, Janczak K, Shi X, Baker J,
terial activity and mechanism of action of the silver ion in Balogh L (2005) Silver/dendrimer nanocomposites as
Staphylococcus aureus and Escherichia coli. Appl Environ biomarkers: fabrication, characterization, in vitro toxicity,
Microbiol 74:2171–2178. doi:10.1128/AEM.02001-07 and intracellular detection. Nano Lett 5:2123–2130. doi:
Jung R, Kim Y, Kim H, Jin H (2009) Antimicrobial properties 10.1021/nl051077u
of hydrated cellulose membranes with silver nanoparti- Li Y, Kim Y, Lee E, Cai W, Cho S (2006) Synthesis of silver
cles. J Biomater Sci Polym Ed 20:311–324. doi:10.1163/ nanoparticles by electron irradiation of silver acetate.
156856209X412182 Nucl Instrum Methods B 251:425–428. doi:10.1016/
Kalishwaralal K, Deepak V, Ramkumarpandian S, Nellaiah H, j.nimb.2006.06.019
Sangiliyandi G (2008) Extracellular biosynthesis of silver Li S, Shen Y, Xie A, Yu X, Qiu L, Zhang L, Zhang Q (2007)
nanoparticles by the culture supernatant of Bacillus licheni- Green synthesis of silver nanoparticles using Capsicum
formis. Mater Lett 62:4411–4413. doi:10.1016/j.matlet. annuum L. extract. Green Chem 9:852–858. doi:10.1039/
2008.06.051 b615357g
Kasthuri J, Veerapandian S, Rajendiran N (2009) Biological Liau S, Read D, Pugh W, Furr J, Russell A (1997) Interaction
synthesis of silver and gold nanoparticles using apiin as of silver nitrate with readily identifiable groups: rela-
reducing agent. Colloids Surf B 68:55–60. doi:10.1016/ tionship to the antibacterial action of silver ions. Lett Appl
j.colsurfb.2008.09.021 Microbiol 25:279–283. doi:10.1046/j.1472-765X.1997.
Kim J (2007) Antibacterial activity of Ag? ion-containing 00219.x
silver nanoparticles prepared using the alcohol reduction Lind ML, Jeong BH, Subramani A, Huang XF, Hoek EMV
method. J Ind Eng Chem 13:718–722 (2009) Effect of mobile cation on zeolite-polyamide thin
Kim J, Kuk E, Yu K, Kim J, Park S, Lee H, Kim S, Park Y, film nanocomposite membranes. J Mater Res 24:1624–
Park Y, Hwang C, Kim Y, Lee Y, Jeong D, Cho M (2007) 1631. doi:10.1557/Jmr.2009.0189
Antimicrobial effects of silver nanoparticles. Nanomed Lok C, Ho C, Chen R, He Q, Yu W, Sun H, Tam P, Chiu J, Che
Nanotechnol 3:95–101. doi:10.1016/j.nano.2006.12.001 C (2006) Proteomic analysis of the mode of antibacterial
Kim K, Sung W, Moon S, Choi J, Kim J, Lee D (2008a) action of silver nanoparticles. J Proteome Res 5:916–924.
Antifungal effect of silver nanoparticles on dermato- doi:10.1021/pr0504079
phytes. J Microbiol Biotechnol 18:1482–1484 Lok C, Ho C, Chen R, He Q, Yu W, Sun H, Tam P, Chiu J, Che
Kim Y, Kim J, Cho H, Rha D, Kim J, Park J, Choi B, Lim R, C (2007) Silver nanoparticles: partial oxidation and anti-
Chang H, Chung Y, Kwon I, Jeong J, Han B, Yu I (2008b) bacterial activities. J Biol Inorg Chem 12:527–534. doi:
Twenty-eight-day oral toxicity, genotoxicity, and gender- 10.1007/s00775-007-0208-z
related tissue distribution of silver nanoparticles in Spra- Long D, Wu G, Chen S (2007) Preparation of oligochitosan
gue-Dawley rats. Inhal Toxicol 20:575–583. doi:10.1080/ stabilized silver nanoparticles by gamma irradiation. Radiat
08958370701874663 Phys chem 76:1126–1131. doi:10.1016/j.radphyschem.
Kim J, Lee J, Kwon S, Jeong S (2009a) Preparation of biode- 2006.11.001
gradable polymer/silver nanoparticles composite and its Lu L, Sun R, Chen R, Hui C, Ho C, Luk J, Lau G, Che C
antibacterial efficacy. J Nanosci Nanotechnol 9:1098–1102. (2008) Silver nanoparticles inhibit hepatitis B virus rep-
doi:10.1166/jnn.2009.C096 lication. Antivir Ther 13:253–262
Kim K, Sung W, Suh B, Moon S, Choi J, Kim J, Lee D (2009b) Mahapatra S, Bogle K, Dhole S, Bhoraskar V (2007) Synthesis
Antifungal activity and mode of action of silver nano- of gold and silver nanoparticles by electron irradiation at 5–
particles on Candida albicans. Biometals 22:235–242. 15 keV energy. Nanotechnology 18. doi: 10.1088/0957-
doi:10.1007/s10534-008-9159-2 4484/18/13/135602

123
J Nanopart Res (2010) 12:1531–1551 1549

Maneerung T, Tokura S, Rujiravanit R (2008) Impregnation of Neal AL (2008) What can be inferred from bacterium-nano-
silver nanoparticles into bacterial cellulose for antimi- particle interactions about the potential consequences of
crobial wound dressing. Carbohydr Polym 72:43–51. doi: environmental exposure to nanoparticles? Ecotoxicology
10.1016/j.carbpol.2007.07.025 17:362–371. doi:10.1007/s10646-008-0217-x
Manno D, Filippo E, Di Giulio M, Serra A (2008) Synthesis Nel A, Xia T, Madler L, Li N (2006) Toxic potential of
and characterization of starch-stabilized Ag nanostruc- materials at the nanolevel. Science 311:622–627. doi:
tures for sensors applications. J Non-Cryst Solids 354: 10.1126/science.1114397
5515–5520. doi:10.1016/j.jnoncrysol.2008.04.059 Nel AE, Madler L, Velegol D, Xia T, Hoek EMV, Somas-
Manzi AE, van Halbeek H (1999) Saccharide structure and undaran P, Klaessig F, Castranova V, Thompson M
nomenclature. In: Varki A, Cummings R, Esko J, Freeze (2009) Understanding biophysicochemical interactions at
H, Hart G, Marth J (eds) Essentials of glycobiology, 1st the nano–bio interface. Nat Mater 8:543–557. doi:10.1038/
edn. Cold Spring Harbor Laboratory Press, New York nmat2442
Martinez-Castanon GA, Nino-Martinez N, Martinez-Gutierrez Nita T (2008) Synthesis of antimicrobial polymer composition
F, Martinez-Mendoza JR, Ruiz F (2008) Synthesis and and in vitro drugs release study. e-Polymers
antibacterial activity of silver nanoparticles with different Ogden J, Bogdanchikova N, Corker J, Petranovskii V (1999)
sizes. J Nanopart Res 10:1343–1348. doi:10.1007/s11051- Structure of silver clusters embedded in erionite channels.
008-9428-6 Eur Phys J D 9:605–608. doi:10.1007/s100530050509
Martinez-Castanon G, Nino-Martinez N, Loyola-Rodriguez J, Pal S, Tak Y, Song J (2007) Does the antibacterial activity of
Patino-Marin N, Martinez-Mendoza J, Ruiz F (2009) silver nanoparticles depend on the shape of the nanopar-
Synthesis of silver particles with different sizes and mor- ticle? A study of the gram-negative bacterium Escherichia
phologies. Mater Lett 63:1266–1268. doi:10.1016/j.matlet. coli. Appl Environ Microbiol 73:1712–1720. doi:10.1128/
2009.02.061 AEM.02218-06
Mayo JT, Yavuz C, Yean S, Cong L, Shipley H, Yu W, Falkner Pal S, Tak YK, Joardar J, Kim W, Lee JE, Han MS, Song JM
J, Kan A, Tomson M, Colvin VL (2007) The effect of (2009) Nanocrystalline silver supported on activated car-
nanocrystalline magnetite size on arsenic removal. Sci bon matrix from hydrosol: antibacterial mechanism under
Technol Adv Mater 8:71–75. doi:10.1016/j.stam.2006.10. prolonged incubation conditions. J Nanosci Nanotechnol
005 9:2092–2103. doi:10.1166/jnn.2009.427
McDonnell AMP, Beving D, Wang AJ, Chen W, Yan YS Panacek A, Kvitek L, Prucek R, Kolar M, Vecerova R,
(2005) Hydrophilic and antimicrobial zeolite coatings for Pizurova N, Sharma V, Nevecna T, Zboril R (2006) Silver
gravity-independent water separation. Adv Funct Mater colloid nanoparticles: synthesis, characterization, and
15:336–340. doi:10.1002/adfm.200400183 their antibacterial activity. J Phys Chem B 110:16248–
Mendis E, Rajapakse N, Byun H, Kim S (2005) Investigation 16253. doi:10.1021/jp063826h
of jumbo squid (Dosidicus gigas) skin gelatin peptides for Panyala NR, Peña-Méndez EM, Havel J (2008) Silver or silver
their in vitro antioxidant effects. Life Sci 77:2166–2178. nanoparticles: a hazardous threat to the environment and
doi:10.1016/j.lfs.2005.03.016 human health? J Appl Biomed 6:117–129
Mokhtari N, Daneshpajouh S, Seyedbagheri S, Atashdehghan Park H, Kim J, Kim J, Lee J, Hahn J, Gu M, Yoon J (2009)
R, Abdi K, Sarkar S, Minaian S, Shahverdi H, Shahverdi Silver-ion-mediated reactive oxygen species generation
A (2009) Biological synthesis of very small silver nano- affecting bactericidal activity. Water Res 43:1027–1032.
particles by culture supernatant of Klebsiella pneumonia: doi:10.1016/j.watres.2008.12.002
the effects of visible-light irradiation and the liquid mix- Petering H (1976) Pharmacology and toxicology of heavy-
ing process. Mater Res Bull 44:1415–1421. doi:10.1016/ metals-silver. Pharmacol Ther A 1:127–130. doi:10.1016/
j.materresbull.2008.11.021 0362-5478(76)90002-4
Morones J, Elechiguerra J, Camacho A, Holt K, Kouri J, Pillai Z, Kamat P (2004) What factors control the size and shape
Ramirez J, Yacaman M (2005) The bactericidal effect of of silver nanoparticles in the citrate ion reduction method?
silver nanoparticles. Nanotechnology 16:2346–2353. doi: J Phys Chem B 108:945–951. doi:10.1021/jp037018r
10.1088/0957-4484/16/10/059 Raffi M, Hussain F, Bhatti T, Akhter J, Hameed A, Hasan M
Mukherjee P, Ahmad A, Mandal D, Senapati S, Sainkar S, Khan (2008) Antibacterial characterization of silver nanoparti-
M, Parishcha R, Ajaykumar P, Alam M, Kumar R, Sastry M cles against E. coli ATCC-15224. J Mater Sci Technol
(2001) Fungus-mediated synthesis of silver nanoparticles 24:192–196
and their immobilization in the mycelial matrix: a novel Rejeski D (2009) Nanotechnology and consumer products.
biological approach to nanoparticle synthesis. Nano Lett http://www.nanotechproject.org/publications/archive/
1:515–519. doi:10.1021/nl0155274 nanotechnology_consumer_products/. Accessed 22
Naidu B, Park J, Kim S, Park S, Lee E, Yoon K, Lee S, Lee J, February 2010
Gal Y, Jin S (2008) Novel hybrid polymer photovoltaics Roe D, Karandikar B, Bonn-Savage N, Gibbins B, Roullet J
made by generating silver nanoparticles in polymer: ful- (2008) Antimicrobial surface functionalization of plastic
lerene bulk-heterojunction structures. Sol Energy Mater catheters by silver nanoparticles. J Antimicrob Chemother
Sol Cells 92:397–401. doi:10.1016/j.solmat.2007.09.017 61:869–876. doi:10.1093/jac/dkn034
Navarro E, Piccapietra F, Wagner B, Marconi F, Kaegi R, Ruparelia J, Chatteriee A, Duttagupta S, Mukherji S (2008)
Odzak N, Sigg L, Behra R (2008) Toxicity of Silver Strain specificity in antimicrobial activity of silver and
Nanoparticles to Chlamydomonas reinhardtii. Environ Sci copper nanoparticles. Acta Biomater 4:707–716. doi:
Technol 42:8959–8964. doi:10.1021/es801785m 10.1016/j.actbio.2007.11.006

123
1550 J Nanopart Res (2010) 12:1531–1551

Saito Y, Wang J, Batchelder D, Smith D (2003) Simple Singh M, Sinha I, Mandal R (2009) Role of pH in the green
chemical method for forming silver surfaces with con- synthesis of silver nanoparticles. Mater Lett 63:425–427.
trolled grain sizes for surface plasmon experiments. doi:10.1016/j.matlet.2008.10.067
Langmuir 19:6857–6861. doi:10.1021/la0301240 Slonczewski J, Foster J (2009) Microbiology: an evolving
Sambhy V, Sen A (2008) Novel process of incorporating silver science. W. W. Norton & Co, New York
biocides into polymers. Chim Oggi 26:16–18 Smetana A, Klabunde K, Sorensen C (2005) Synthesis of
Sanghi R, Verma P (2009) Biomimetic synthesis and charac- spherical silver nanoparticles by digestive ripening, sta-
terisation of protein capped silver nanoparticles. Biore- bilization with various agents, and their 3-D and 2-D su-
sour Technol 100:501–504. doi:10.1016/j.biortech.2008. perlattice formation. J Colloid Interface Sci 284:521–526.
05.048 doi:10.1016/j.jcis.2004.10.038
Sanpui P, Murugadoss A, Prasad P, Ghosh S, Chattopadhyay A Smetana A, Klabunde K, Marchin G, Sorensen C (2008) Biocidal
(2008) The antibacterial properties of a novel chitosan- activity of nanocrystalline silver powders and particles.
Ag-nanoparticle composite. Int J Food Microbiol 124: Langmuir 24:7457–7464. doi:10.1021/la800091y
142–146. doi:10.1016/j.ijfoodmicro.2008.03.004 Sondi I, Salopek-Sondi B (2004) Silver nanoparticles as anti-
Schneider S, Halbig P, Grau H, Nickel U (1994) Reproducible microbial agent: a case study on E. coli as a model for
preparation of silver sols with uniform particle-size for Gram-negative bacteria. J Colloid Interface Sci 275:177–
application in surface-enhanced raman-spectroscopy. 182. doi:10.1016/j.jcis.2004.02.012
Photochem Photobiol 60:605–610. doi:10.1111/j.1751- Song J, Kim B (2009) Rapid biological synthesis of silver
1097.1994.tb05156.x nanoparticles using plant leaf extracts. Bioproc Biosyst
Schrand A, Braydich-Stolle L, Schlager J, Dai L, Hussain S Eng 32:79–84. doi:10.1007/s00449-008-0224-6
(2008) Can silver nanoparticles be useful as potential Stoeva S, Klabunde K, Sorensen C, Dragieva I (2002) Gram-scale
biological labels? Nanotechnology 19. doi: 10.1088/0957- synthesis of monodisperse gold colloids by the solvated
4484/19/23/235104 metal atom dispersion method and digestive ripening and
Schreurs W, Rosenberg H (1982) Effect of silver ions on their organization into two- and three-dimensional struc-
transport and retention of phosphate by Escherichia coli. tures. J Am Chem Soc 124:2305–2311. doi:10.1021/
J Bacteriol 152:7–13 ja012076g
Senapati S, Mandal D, Ahmad A, Khan M, Sastry M, Kumar R Stohs SJ, Bagchi D (1995) Oxidative mechanisms in the tox-
(2004) Fungus mediated synthesis of silver nanoparticles: a icity of metal-ions. Free Radic Biol Med 18:321–336. doi:
novel biological approach. Indian J Phys A 78A:101–105 10.1016/0891-5849(94)00159-H
Shankar SS, Ahmad A, Sastry M (2003) Geranium leaf assisted Sun L, Singh A, Vig K, Pillai S, Singh S (2008) Silver nano-
biosynthesis of silver nanoparticles. Biotechnol Progr 19: particles inhibit replication of respiratory syncytial virus.
1627–1631. doi:10.1021/Bp034070w J Biomed Nanotechnol 4:149–158. doi:10.1166/jbn.2008.
Sharma S, Thakur M, Deb M (2007) Synthesis of silver 012
nanoparticles using N-1, N-2-diphenylbenzamidine by Sung J, Ji J, Yoon J, Kim D, Song M, Jeong J, Han B, Han J,
microwave irradiation method. J Exp Nanosci 2:251–256. Chung Y, Kim J, Kim T, Chang H, Lee E, Lee J, Yu I
doi:10.1080/17458080701753744 (2008) Lung function changes in Sprague-Dawley rats
Sharma S, Thakur M, Deb MK (2008) Preparation of silver after prolonged inhalation exposure to silver nanoparti-
nanoparticles by microwave irradiation. Curr Nanosci 4: cles. Inhal Toxicol 20:567–574. doi:10.1080/089583707
138–140 01874671
Sharma VK, Yngard RA, Lin Y (2009) Silver nanoparticles: Teeguarden J, Hinderliter P, Orr G, Thrall B, Pounds J (2007)
green synthesis and their antimicrobial activities. Adv Particokinetics in vitro: dosimetry considerations for in
Colloid Interface Sci 145:83–96. doi:10.1016/j.cis.2008. vitro nanoparticle toxicity assessments. Toxicol Sci 95:
09.002 300–312. doi:10.1093/toxsci/kfl165
Shirtcliffe N, Nickel U, Schneider S (1999) Reproducible Tomsic B, Simoncic B, Orel B, Zerjav M, Schroers H, Si-
preparation of silver sols with small particle size using moncic A, Samardzija Z (2009) Antimicrobial activity of
borohydride reduction: for use as nuclei for preparation of AgCl embedded in a silica matrix on cotton fabric. Car-
larger particles. J Colloid Interface Sci 211:122–129. doi: bohydr Polym 75:618–626. doi:10.1016/j.carbpol.2008.09.
10.1006/jcis.1998.5980 013
Shrivastava S, Bera T, Roy A, Singh G, Ramachandrarao P, Venediktov E, Padokhin V (2008) Synthesis of silver nanocl-
Dash D (2007) Characterization of enhanced antibacterial usters in starch aqueous solutions. Russ J Appl Chem
effects of novel silver nanoparticles. Nanotechnology 18. 81:2040–2042. doi:10.1134/S1070427208110323
doi: 10.1088/0957-4484/18/22/225103 Vertelov G, Krutyakov Y, Efremenkova O, Olenin A, Lisich-
Silver S (2003) Bacterial silver resistance: molecular biology kin G (2008) A versatile synthesis of highly bactericidal
and uses and misuses of silver compounds. FEMS MyramistinÒ stabilized silver nanoparticles. Nanotech-
Microbiol Rev 27:341–353. doi:10.1016/S0168-6445(03) nology 19. doi: 10.1088/0957-4484/19/35/355707
00047-0 Vigneshwaran N, Ashtaputre N, Varadarajan P, Nachane R,
Silver S, Phung LT, Silver G (2006) Silver as biocides in burn Paralikar K, Balasubramanya R (2007) Biological syn-
and wound dressings and bacterial resistance to silver thesis of silver nanoparticles using the fungus Aspergillus
compounds. J Ind Microbiol Biotechnol 33:627–634. doi: flavus. Mater Lett 61:1413–1418. doi:10.1016/j.matlet.2006.
10.1007/s10295-006-0139-7 07.042

123
J Nanopart Res (2010) 12:1531–1551 1551

West JL, Halas NJ (2003) Engineered nanomaterials for bio- microwave irradiation. Mater Chem Phys 83:66–70. doi:
photonics applications: improving sensing, imaging, and 10.1016/j.matchemphys.2003.09.006
therapeutics. Annu Rev Biomed Eng 5:285–292. doi: Yoon K, Byeon J, Park C, Hwang J (2008a) Antimicrobial effect
10.1146/annurev.bioeng.5.011303.120723 of silver particles on bacterial contamination of activated
Xu X, Brownlow W, Kyriacou S, Wan Q, Viola J (2004) Real- carbon fibers. Environ Sci Technol 42:1251–1255. doi:
time probing of membrane transport in living microbial cells 10.1021/es0720199
using single nanoparticle optics and living cell imaging. Yoon K, Byeon J, Park J, Ji J, Bae G, Hwang J (2008b)
Biochemistry 43:10400–10413. doi:10.1021/bi036231a Antimicrobial characteristics of silver aerosol nanoparti-
Xu X, Yang Q, Wang Y, Yu H, Chen X, Jing X (2006) Bio- cles against Bacillus subtilis bioaerosols. Environ Eng Sci
degradable electrospun poly(L-lactide) fibers containing 25:289–293. doi:10.1089/ees.2007.0003
antibacterial silver nanoparticles. Eur Phys J D 42:2081– Yu D (2007) Formation of colloidal silver nanoparticles sta-
2087. doi:10.1016/j.eurpolymj.2006.03.032 bilized by Na?-poly(gamma-glutamic acid)-silver nitrate
Yanagihara N, Tanaka Y, Okamoto H (2001) Formation of complex via chemical reduction process. Colloids Surf B
silver nanoparticles in poly(methyl methacrylate) by UV 59:171–178. doi:10.1016/j.colsurfb.2007.05.007
irradiation. Chem Lett 30:796–797 Yu D, Yam V (2004) Controlled synthesis of monodisperse
Yang W, Shen C, Ji Q, An H, Wang J, Liu Q, Zhang Z (2009) silver nanocubes in water. J Am Chem Soc 126:13200–
Food storage material silver nanoparticles interfere with 13201. doi:10.1021/ja046037r
DNA replication fidelity and bind with DNA. Nanotech- Zeng H, Zhao C, Qiu J, Yang Y, Chen G (2007) Preparation
nology 20. doi: 10.1088/0957-4484/20/8/085102 and optical properties of silver nanoparticles induced by a
Yavuz CT, Mayo JT, Yu WW, Prakash A, Falkner JC, Yean S, femtosecond laser irradiation. J Cryst Growth 300:519–
Cong L, Shipley HJ, Kan A, Tomson M, Natelson D, 522. doi:10.1016/j.jcrysgro.2006.11.308
Colvin VL (2006) Low-field magnetic separation of mono- Zhang Y, Peng H, Huang W, Zhou Y, Yan D (2008) Facile
disperse Fe3O4. Nanocrystals 314:964–967. doi:10.1126/ preparation and characterization of highly antimicrobial
science.1131475 colloid Ag or Au nanoparticles. J Colloid Interface Sci 325:
Yeo M, Kang M (2008) Effects of nanometer sized silver 371–376. doi:10.1016/j.jcis.2008.05.063
materials on biological toxicity during zebrafish embryo- Zodrow K, Brunet L, Mahendra S, Li D, Zhang A, Li QL,
genesis. Bull Korean Chem Soc 29:1179–1184 Alvarez PJJ (2009) Polysulfone ultrafiltration membranes
Yeo M, Yoon J (2009) Comparison of the effects of nano-silver impregnated with silver nanoparticles show improved
antibacterial coatings and silver ions on zebrafish embryo- biofouling resistance and virus removal. Water Res 43:
genesis. Mol Cell Toxicol 5:23–31 715–723. doi:10.1016/j.watres.2008.11.014
Yin H, Yamamoto T, Wada Y, Yanagida S (2004) Large-scale
and size-controlled synthesis of silver nanoparticles under

123

You might also like