toxicity of silver nanoparticles in fish: a critical revie · which changes as the particles are...

17
J. Bio. & Env. Sci. 2015 211 | Khan et al. REVIEW PAPER OPEN ACCESS Toxicity of silver nanoparticles in fish: a critical review Muhammad Saleem Khan 1 , Farhat Jabeen 1* , Naureen Aziz Qureshi 2 , Muhammad Saleem Asghar 1 , Muhammad Shakeel 1 and Aasma Noureen 1 Department of Zoology, GC University Faisalabad, Pakistan 2 GC Women University Faisalabad, Pakistan Article published on May 18, 2015 Key words: Silver, Nanoparticles, Uses, Toxicity, Fish model. Abstract The variable spectrum of applications largely depends upon silver physicochemical and biological properties which changes as the particles are decreased to nano-scale. This unique behavior is responsible for the larger use of silver in consumer product and industry. Since little information is available about toxicity to the organisms practically in the aquatic environment, the predication of possible environmental hazards and remedy are the hot topics of current research studies. Researchers are drawing more and more data from appropriate model organisms. Fish being aquatic organism is badly affected by Ag-NPs, so concern of potential risk to aquatic organism increases. The toxicity endpoints include growth and reproduction impairment, mortality and biochemical changes in both adult fish and embryos. Being a healthy food for human, the researchers try to know how Ag-NPs can affect the fish and its body when sizes decrease to nano-scale. Therefore, fish is extensively studied model in the toxicological studies. It examined some of these studies which address the adverse effects of Ag-NPs on biological systems of different fish group predicting the dose dependent toxicity. The organisms more acutely sensitive have lower LC50 values. All the studies also indicated that silver ions released from Ag-NPs surface contributes to toxicity. Therefore, it is suggested that emphasis should be placed on upcoming investigations for the evaluation of environmental impact of nano-silver in both in vitro and in vivo studies. The effect of long term exposure and bioaccumulation of silver through food web should be unmasked and discussed in detail. * Corresponding Author: Dr Farhat Jabeen [email protected] Journal of Biodiversity and Environmental Sciences (JBES) ISSN: 2220-6663 (Print) 2222-3045 (Online) Vol. 6, No. 5, p. 211-227, 2015 http://www.innspub.net

Upload: hathien

Post on 30-Apr-2019

213 views

Category:

Documents


0 download

TRANSCRIPT

J. Bio. & Env. Sci. 2015

211 | Khan et al.

REVIEW PAPER OPEN ACCESS

Toxicity of silver nanoparticles in fish: a critical review

Muhammad Saleem Khan1, Farhat Jabeen1*, Naureen Aziz Qureshi2, Muhammad

Saleem Asghar1, Muhammad Shakeel1 and Aasma Noureen

1Department of Zoology, GC University Faisalabad, Pakistan

2GC Women University Faisalabad, Pakistan

Article published on May 18, 2015

Key words: Silver, Nanoparticles, Uses, Toxicity, Fish model.

Abstract

The variable spectrum of applications largely depends upon silver physicochemical and biological properties

which changes as the particles are decreased to nano-scale. This unique behavior is responsible for the larger use

of silver in consumer product and industry. Since little information is available about toxicity to the organisms

practically in the aquatic environment, the predication of possible environmental hazards and remedy are the hot

topics of current research studies. Researchers are drawing more and more data from appropriate model

organisms. Fish being aquatic organism is badly affected by Ag-NPs, so concern of potential risk to aquatic

organism increases. The toxicity endpoints include growth and reproduction impairment, mortality and

biochemical changes in both adult fish and embryos. Being a healthy food for human, the researchers try to know

how Ag-NPs can affect the fish and its body when sizes decrease to nano-scale. Therefore, fish is extensively

studied model in the toxicological studies. It examined some of these studies which address the adverse effects of

Ag-NPs on biological systems of different fish group predicting the dose dependent toxicity. The organisms more

acutely sensitive have lower LC50 values. All the studies also indicated that silver ions released from Ag-NPs

surface contributes to toxicity. Therefore, it is suggested that emphasis should be placed on upcoming

investigations for the evaluation of environmental impact of nano-silver in both in vitro and in vivo studies. The

effect of long term exposure and bioaccumulation of silver through food web should be unmasked and discussed

in detail.

*Corresponding Author: Dr Farhat Jabeen [email protected]

Journal of Biodiversity and Environmental Sciences (JBES) ISSN: 2220-6663 (Print) 2222-3045 (Online)

Vol. 6, No. 5, p. 211-227, 2015

http://www.innspub.net

J. Bio. & Env. Sci. 2015

212 | Khan et al.

Introduction

The commercial applications of the nanomaterial

research are maximum in the present era. It is

estimated that approximately 60,000 tons of

nanomaterial is produced annually (Jovanovic et al.,

2011) with 1628 nano based products in 30 countries

(Woodrow Wilson Database, 2015). Among the nano

industry, silver nanoparticles is the most rapidly

growing class with 320 tons of production per year

(Nowack et al., 2011). Its importance is due to unique

properties and consumers demands. About 30% of all

the currently registered nanoproducts are claiming to

contain the nano-silver (Project on emerging

nanotechnologies, 2013). It is estimated that 383

nanoproducts are available in the market (Woodrow

Wilson Database, 2015).

The extensive use of Ag nanoproducts might increase

the discharge of these particles into aquatic

environment (Benn and Westerhoff, 2008; Taju et al.,

2014). It may be released into water and air through

different sources including weathering of rocks,

processing of ores, cement manufacturing and

burning of fossil fuel. Rain is responsible to release

the silver in the ground and water reservoirs

(Wijnhoven et al., 2009). About 68% of nano silver

load will be increased in the waste water due to

biocidal products from 2010 to 2015 (Blaser et al,

2008). In the aquatic environment, it exits in four

(Ag, Ag+, Ag+2 and Ag+3) oxidation states. But Ag and

Ag+ exist more commonly (Smith and Carson, 1997).

Metallic silver is insoluble whereas salts (AgCl,

AgNO3) are soluble in water (WHO, 2002). Silver

nanomaterial is found in the form of colloidal

particles in the aquatic medium.

In spite of existence in the aquatic environment,

limited information regarding the toxicity of Ag-NPs

is available (Wijnhoven et al., 2009). Depending

upon the existing literature, it can be hypothesized

that Ag-NPs are more toxic than other forms because

of it’s more readily absorbance than metallic silver

(Drake and Hazelwood, 2005). The fate and behavior

of silver nanomaterial is influenced by many factors

including; size, surface area, surface chemistry and

chemical composition (coatings and purity). The

other factors like water and lipid solubility, vapor

pressure and aggregation or coagulation state

(Wijnhoven et al., 2009).

In the aquatic environment, Ag-NPs most likely enter

the ecosystems produce a physiological response in

many animals, altering their fitness and population

densities (Luoma and Rainbow, 2008). On the other

hand, the detailed studies on the effect of these

particles on the target organisms and their

environment have just begun. Numbers of

toxicological studies have been performed but it

showed huge variations due to lack of proper particles

characterization (Gliga et al., 2014). The researchers

focus on toxicity of Ag-NPs in aquatic environment

including fish in the recent studies (Asharani et al.,

2008; Yeo and Kang 2008; Bar-Ilan et al., 2009;

Chae et al., 2009; Choi et al., 2009; Griffitt et al.,

2009; Bilberg et al., 2010; Powers et al., 2010; Wu et

al., 2010). Toxicity induced by Ag-NPs to vertebrate

cell line include generation of the reactive oxygen

species (Hussain et al., 2005; Schrand et al., 2008),

apoptosis (Braydich-Stolle et al., 2005; Park et al.,

2007), reduced mitochondrial function (Braydich-

Stolle et al., 2005; Hussain et al., 2005; Schrand et

al., 2008), increase lipid peroxidation (Arora et al.,

2008) and depletion of the oxidative stress markers

(Hussain et al., 2005; Arora et al., 2008). Further a

study by Larese et al. (2009) demonstrated that Ag-

NPs can pass through stratum corneum and the outer

layer of epidermis and even blood-brain barriers

causing damage to human skin, liver, lungs and

olfactory blabs (Braydich-Stolle et al., 2005; Hussain

et al., 2005; Arora et al. 2008; Sung et al., 2008). So

the current review will present the toxicity caused by

Ag-NPs on some groups of fish.

Silver uses

Silver is among basic elements that formed our

planet. Its size ranges 5-50 nm in many commercial

products so refers as nanosilver (Panyala et al.,

2008). It has a long history of use. Ancient Egypt,

J. Bio. & Env. Sci. 2015

213 | Khan et al.

Rome, Italy and Greece were aware about the use of

silver (Reidy et al., 2013). They use silver for the

preparation of storage vessels that keep the water

fresh (Russell and Hugo 1994). It was used in the

form of colloidal silver more than 150 years ago and

first time registered as biocidal material in 1954

(Nowack et al., 2011). In the present time, it is mostly

used in the fields of chemistry, material science and

physics (Syrvatka et al., 2014). It is used worldwide in

photography, batteries as coatings of solar energy

absorption, water treatment filters (Li et al., 2008),

washing machines (Jung et al., 2007), fabrics

(Perelshtein et al., 2008), Heat sink, sensors

(Schrand et al., 2008), catalysts (Kumar et al., 2008),

superconductors, cloud seeding, shampoo, food

packing, Electroplating, kitchen utensils, and odor

resistant textiles (Sondi and Sondi, 2004; Cohen et

al., 2007; Yon and Lead, 2008). Furthermore silver is

also combined with other substance to develop

combined functions.

More attention is devoted towards silver due to its

medical importance. It has been used in the fields of

biotechnology, medicine, environmental technology

and as a broad spectrum antimicrobial agent (Kim et

al., 2007; Kim et al., 2013). Table 1 provides the

comprehensive medical uses of naonosilver in the

current time. The silver containing biocidal products

has reached to 110 to 230 tons in the European

market and significant portion is consists of Ag-NPs

(Blaser et al., 2008).

Fig. 1. Uses of silver in different sectors (SOURCE:

World Silver Survey 2014, The Silver institute, 2014).

The demand of silver use and production increases

every year. It is because of silver used in industrial,

medical and photography.

The humans, animals and microorganisms are

exposed to nanosilver through three major products.

These products are food, medical and consumer’s

products. Some common medical uses of Ag-NPs are

provided in the table-1.

Table 1. Some common uses of Ag-NPs in the medical fields.

Medical applications of silver

A. Treatment and repair

1. Treatment of ulcerative colitis and acne (Bhol and Schechter, 2007)

2. Treatment of dermatitis (Bhol et al., 2004)

3. Allergy prophylaxis (Gulbranson et al., 2000; Silver, 2003)

4. Inhibition of HIV-1 replication (Elechiguerra et al., 2005; Sun et al., 2005)

5. Bone cement additive (Alt et al., 2004)

6. Orthopedic stockings (Pohle et al., 2007)

7. Rheumatoid arthritis-associated leg ulcers (Coelho et al., 2004)

8. Coating of implant for joint replacement (Chen et al., 2006)

9. Coating of catheter for cerebrospinal fluid drainage (Bayston et al., 2007)

10. Coating of surgical mesh for pelvic reconstruction (Cohen et al., 2007)

11. Coating of intramedullary nail for long bone fractures (Alt et al., 2006)

12. In the form of silver proteinate for treatment of conjunctivitis in newborn babies (NCCAM, 2012)

13. In the form of lunar caustic for treatment of corns and warts (NCCAM, 2012)

J. Bio. & Env. Sci. 2015

214 | Khan et al.

Medical applications of silver

14. Anti-inflammatory medicine (Kirsner et al., 2001)

15. Modulate cytokines in wound healing (Tian et al., 2007)

16. Treatment of burns (Tredget et al.,1998)

17. Silver diamine fluoride to reduce tooth decay (Rosenblatt et al., 2009; Deery, 2009)

18. Silver acetate antismoking agent (Lancaster Stead, 2012)

B. Laboratory diagnosis

1. Detection of viral strain (SERS and silver nanorods) (Zhao et al., 2006)

2. Dendrimer nanocomposite for cell labeling (Lesniak et al., 2005)

3. Ag pyramids enhance bio-detection (Walt, 2005)

4. Sensitive diagnosis of myocardial infarction (Aslan and Geddes, 2006)

5. Fluorescence-based RNA sensing (Aslan et al., 2006)

6. Molecular imaging of cancer cells (Tai et al., 2007)

7. Protein biosensor any protein or any antibody (Ananth et al., 2011)

8. Clinical diagnosis of myocardial infraction (Aslan and Geddes, 2006)

9. Genosensors silver (I) and hydroquinone (He et al., 2009)

C. Antiseptic uses

1. Antimicrobial agent against infectious organisms (Yves and Philippe, 2012)

2. Hydrogel for wound dressing (silver-containing hydrocolloid) (Yu et al., 2007)

3. Antifungal uses (Wright et al., 1999)

4. Effective against yeast isolated from bovine mastitis (Kim et al., 2007)

D. Medical utensils

1. Coating of driveline for ventricular assist devices (Drake and Hazelwood, 2005)

2. Coating of endotracheal tube ventilatory support (Bouadma et al., 2012)

3. Coating of hospital textile (e.g., surgical gowns and face mask) (Lee et al., 2003)

4. Remote laser light-induced opening of microcapsules Skirtach et al., (2006)

5. Coating of breathing mask patent (Drake and Hazelwood, 2005)

6. Needles, catheters, dental amalgams (Drake and Hazelwood, 2005)

7. Surgical instruments production (Chen and Schluesener, 2008)

8. Coating of contact lens (Weisbarth et al., 2007)

9. Drug carrier in the medical products (Chen et al., 2015)

Causes of toxicity

According to the Thomson Reuters, the number of

papers on the toxicological aspects of Ag-NPs has

been increased since 1990. Currently more than 3500

research articles are published on this theme

annually. These research articles suggest that the

toxicity of Ag-NPs depends upon many factors such as

shape, size, surface area, chemical composition and

surface charges (Hedayati et al., 2012). In most of the

studies, it is observed that physical and chemical

properties change when we decrease the particle size

to nonoscale (Hedayati et al., 2012). This concluded

the nano-sized particles show variations of optical,

electrical and magnetic properties from large particles

of the same compounds (Dowling et al., 2009).

Further the toxicity may also affects due to particles

size (Nowack and Bucheli, 2007; Carlson et al., 2008;

Inoue et al., 2010). However, the relationship

between the biological effects and particle size of Ag-

NPs is still unclear (Ivask et al., 2014). One proposed

argument is that the smaller size of particles might

allow the Ag-NPs to enter an organism more readily

than larger particles (Hedayati et al., 2012; Ivask et

al., 2014). To prove this, Lvask et al. (2014) used four

J. Bio. & Env. Sci. 2015

215 | Khan et al.

different sizes of Ag-NPs (10, 20, 60, and 80 nm) on

different organisms. The analysis showed that 10nm

particles were more toxic than all the types. Ag-NPs

are coated with different organic compounds in the

synthesis process which can change fate, toxicity and

stability of the particles in the aqueous and biological

mediums. Several organic coated particles may

damage cell membrane directly, interfare in DNA

replication and ATP synthesis, cause alteration in the

expression of genes and produce reactive oxygen

species (Sherma et al., 2014).

Ag-NPs enter organism’s body through oral

absorption, inhalation, through damage skin (ATSDR,

1990; Drake and Hazelwood, 2005) even through

barrier of retina (Soderstjerna et al., 2014) in adult

and diffusion or endocytosis through the skin of

embryos (Asharani et al., 2008). Colloidal silver

nanoparticles and silver compounds enter in

organism s body through ingestion from food

containing silver preservatives, water and children s

toys. Inhalation of the dust or fumes having silver

occurs in the industrial and jewelry processing (Drake

and Hazelwood, 2005). It also enters in the body

through damage skin from the application of silver

containing burn creams (Wan et al., 1991) or through

cosmetics and textiles (Jones et al., 2010). It can also

enter through female genital tract as most of the

female use various hygienic products containing Ag-

NPs (West and Halas, 2003; Chen and Schluesener,

2008; Schrand et al., 2008). Other routes may

include through acupuncture needles, dental

amalgams (Drake and Hazelwood, 2005) and contact

of jewelry with body (Catsakis and Sulica, 1978).

Absorption of soluble silver compound is greater than

insoluble or metallic silver in this way causing adverse

health effects (Drake and, Hazelwood 2005). Is has

been reported that after the administration Ag-NPs

accumulate in the some organs and cause

hepatotoxicity or renal toxicity after administration

(Sung et al., 2009; Kim et al., 2010).

Fish being aquatic organisms is more venerable to

xenobiotic exposure containing the silver waste. Fish

gills are the primary site for Ag-NPs entrance in the

fish body and histological alterations occur very soon

(Hawkins et al., 2015). The possible effects of Ag-NPs

in different groups of fish are discussed in details.

Zebra fish (Danio rerio)

Zebra fish is extensively studied model in the Ag-NPs

toxicological studies (Asharani et al., 2008; Kanan et

al., 2011). The toxicity indicators may include, drop in

heart rate, hatching delay and higher mortality rate

(Asharani et al., 2008). The LC50 value is 250 mg L-1

in case of embryo (Choi et al., 2010). Bar-llan et al.

(2009) calculated the LC50 values of 3nm to 100nm of

Ag-NPs. The calculated values were 93.31 µM for 3nm

particle size and 137.26 µM for 100 nm. The higher

value of LC50 for lager particles suggest that the

toxicity increases as the particle size decreases. In

some studies free Ag+ also demonstrated the almost

same cytotoxicity as Ag-NPs with almost same LC50

values in zebra fish model (Kim et al., 2009). The Ag-

NPs also accumulated in the different organs like

intestine, gills, blood, liver and brain upon exposing

fish to particles (Handy et al., 2008). The

concentration of accumulated Ag in the liver tissues

was found 0.29 and 2.4ng/mg liver when treated with

30 and 120mgL-1 (Choi et al., 2010). The accumulated

Ag-NPs cause number of cellular alterations in the

liver. These alterations are haptic cell cords, apoptotic

changes, condensation of chromatin and pyknosis in

adult (Gonzalez et al., 2006) and circulatory and

morphological abnormalities in embryo (Asharani et

al., 2008; Bar-Ilan et al., 2009). 2 to 4 mgL-1

exposure for 14 days causes decrease the

Na(+)/K(+)ATPase activity in the gills and

erythrocytes acetylcholinestrase activity (Katuli et al.,

2014). The Ag-NPs treatment also causes oxidative

damage in the hepatic cells. The DNA damage

includes double strand breaks cause lesions in cells

(Rothkamm and Lobrich, 2003).

Toxicity to silver carp (Hypophthalmicthys molitrix)

Hedayati et al. (2012a) suggested Ag-NPs are very

toxic to the silver carp than the metallic silver. The

recorded LC50 value was 0.34 ppm in case of nanocid

J. Bio. & Env. Sci. 2015

216 | Khan et al.

(Hedayati et al., 2012a) and 66.4 ppm in case of

nanosil (Jahanbakhsi et al., 2012). The mortality also

increases as the time of exposure and concentration

increases. There was 100% mortality seen in case of

1ppm and 96 hours of exposure (Hedayati et al.,

2012). In different studies difference in toxicity was

also seen due to change in the size time, age and

condition of test organisms (Rathore and Khangarot,

2002). Shalui et al. (2013) found 0.810 mg L-1 LC50

value for 24h explore and 0.64, 0.383, 0.202 mg L-1

for 48, 72 and 96h respectively. The Ag-NPs also

decrease the RBC, hemoglobin and hematocrit level

in the silver carp (Shalui et al., 2013).

Common Carp (Cyprinus carpio)

The results of the comparative toxicities of Ag-NPs

and Ag ions suggested that Ag-NPs are slightly more

toxic than Ag ions (Hedayati et al., 2012b). Ag-NPs

alter the metabolic enzymes in the organs like gills,

kidney, brain and liver (Reddy et al., 2013). The liver

was found most susceptible to change in Ag-NPs

concentration among all the examined tissues (Lee et

al., 2012). Jung et al. (2014) found mean concentra-

tion of 5.61 mg kg-1 in the liver when exposed to

0.06±0.12 mgL-1 for 7 days. The other organs were

found to have concentration of 3.32 mg kg-1 in gills,

2.93 mg kg-1 in gastrointestinal tract, 0.48 mg kg-1 in

the skeletal muscle 0.48 mg kg-1 in skeletal muscle,

0.14 mg kg-1 in brain and 0.02 mg kg-1 in blood. The

localized Ag-NPs badly reduce the activities of the

metabolic enzymes (SOD, CAT and GST) in brain and

other tissues (Lee et al., 2012). Silver salts (AgNO3),

Nanocid and Nanosil are mostly used in the

toxicological studies of Ag-NPs in the case of the

juvenile common carp (Hedayati et al., 2012b). The

recorded values of LC50 for 96 hours exposure are

0.49±0.90 ppm (Nanocid), 73.8±0.38 (Nanosil) and

0.33± 0.3 ppm (AgNO3) (Hedayati et al., 2012b).

Thala (Catla catla)

Little work has been done for Ag-NPs toxicity in the

case of catla catla. Reddy et al. (2013) found a

significant change in the lipid peroxidation level in

the gills when fish was exposed to 1/5th concentration

of 100 µgL-1 of Ag-NPs for a period of 48 h. But after

the 96 h the lipid peroxidation levels in the gills were

declined at the same concentration. This is due to the

gills endogenous antioxidant system mitigating the

free radical generation (Diehl, 2000). Taju et al.

(2014) also found lipid peroxidation level, decrease in

level of antioxidant enzymatic level due to Ag-NPs

explore.

Rohu (Labeo rohita)

Chemically synthesized Ag-NPs show dose dependent

toxicity in the Labeo rohita. 500 mg kg-1 causes 100%

mortality and 50% mortality was observed at 100 mg

kg-1 in the studies of Rajkumar et al. (2015). The Ag-

NPs creates stressful condition which causes

alteration in the WBC, RBC and total protein level in

the serum. Acid phosphate (ACP) and alkaline

phosphate (ALP) level increases in the Ag-NPs treated

tissues. Orally administrated Ag-NPs also cause the

reduction of GST (glutathione-S-transferase), SOD

(superoxide dismutase) and catalase activities

(Rajkumar et al., 2015).

Crucian carp (Perca fluviatilis)

Exposure of nanosilver causes impairment of

tolerance to hypoxia in crucian carp. It affects gills

and causes reduction in diffusion of oxygen through

gills epithelium leading to hypoxia (Bilberg et al.,

2010). The exposure of 45mgL-1 also suppresses

olfactory responses. It hyperpolarized the olfactory

epithelium membrane interfere the odor detection

mechanism. The free Ag ions release from the surface

of Ag-NPs form complex with receptors and prevent

the odor to combine with olfactory receptors (Klaprat

et al., 1992).

Rainbow trout (Oncorhynchus mykiss)

In different studies, the sized effect of Ag-NPs on

rainbow trout has been studied. Scown et al. (2010)

for example, treated the rainbow trout with 10 nm, 35

nm and 600 to 1600 nm through water medium for

ten days. The uptake level was found very low. 10nm

Ag-NPs were found highest among all the other size

and concentrated more in the gills than liver and

J. Bio. & Env. Sci. 2015

217 | Khan et al.

kidney. Fish liver also showed significant decrease in

weight (p< 0.05). In hepatic parenchyma, local

congestion was decrease in size when exposed to Ag-

NPs (Monfared et al., 2013). The Ag-NPs coated with

PVP (Polyvinylpyrrolidone) and citrate also

accumulate in the gills transport through gill

epithelium and cause cytotoxicity (Farkas et al.,

2010).

The calculated LC50 values were 0.25 and 28.25 mgL-1

for colloidal and suspended powder respectively in

alevins for 96 hours exposure and 2.16 mgL-1 for

colloidal in juveniles. No mortality was seen in case of

powder Ag-NPs (Kalbassi et al., 2013). Johari et al.

(2013) also calculated the LC50 values for the colloidal

particles. Their calculated values were 0.25 mgL-1,

0.71 and 2.16 mgL-1 for eleutheroembryo, larvae and

juveniles, respectively. According to the European

Union Directive (EC, 1999) number 67/548/EEC

dated 27 June 1967 and European legislation (EC,

2008), any substance that has less than 1 mgL-1 LC50

value for 96 hours must be classified as very toxic. It

must have long term adverse effect on the aquatic

organisms. So according to the findings of Johari et

al. (2013), the colloidal Ag-NPs should be classified

very toxic for eleutheroembryo and larvae and toxic to

juveniles in rainbow trout.

The serum level of total protein decreases by

elevation of Ag-NPs concentration (Monfared et al.,

2013). Reduction of potassium and chloride ions and

increase of cortisol and cholinsterse present in blood

plasma were seen when exposed to Ag-NPs (Johari et

al., 2013). Glucose level was also increases (Webb and

Wood, 1998). These changes were dose-dependent

(Johari et al., 2013). Ag-NPs increase the lipid

peroxidation where Ag+ increase the DNA damage

(Massarsky et al., 2014) and inhibits the Na(+),K(+)-

ATPase activity (Schultz et al., 2013).

Medaka (Oryzias latipes)

Kim et al. (2013) demonstrated aged (old) Ag-NPs are

more toxic than fresh one as aged particles release

more Ag ions. They performed toxicity assay in

medaka (Japanese rice fish) and found lower LC50

value (1.44mgL-1) in case of aged Ag-NPs than (3.53

mgL-1) fresh Ag-NPs. Wu et al. (2010) found 100%

mortality at 2.0 mgL-1 in 48h toxicity test and no

mortality at 0.5 mgL-1.

In embryo development retardation, reduce

pigmentation and reduction of width of optic tectum

were seen at 400 µgL-1 concentration (Wu et al.,

2010). Other malfunctions include spinal card

abnormalities, heart deformation, edema and defects

in eyes development at different concentrations (Wu

et al., 2010). 0.8 mgL-1 concentration causes heart

beat retardation, where lower concentration also

causes dose dependent decrease in hatching rate and

body length posing same toxicity to embryo and adult

(Cho et al., 2013). Histological analysis found that Ag-

NPs can also penetrate through chorion of egg

(embryo) and skin membrane and then distributed to

the other tissues (Lee et al., 2014). The principal sites

of uptake are gills in Japanese medaka (Kwok et al.,

2012).

Table 3. Values of 96h LC50 of different forms of Nano-silver in different fish groups.

Sr# Form of Ag-NPs Fish Group LC50 values Reference

1 Nanocid Silver carp 0.34ppm Hedayati et al., 2012a

2 Nanocil Silver carp 66.4ppm Jahanbakhsi et al., 2012

3 Nanpcid (61nm) Silver carp 0.202 mgL-1 Shaluei et al., 2013

4 Nanocid Common carp (juvenile) 0.49±0.90ppm Hedayati et al., 2012b

5 Nanosil Common carp (juvenile) 73.8±0.38ppm Hedayati et al., 2012b

6 AgNO3 Common carp (juvenile) 0.33±0.3ppm Hedayati et al., 2012b

7 Ag-NPs (50-100) Rohu 100 mg kg-1 Rajkumar et al., 2015

8 Ag-NPs (powder) Zebra fish (embryo) 250 mgL-1 Choi et al., 2010

J. Bio. & Env. Sci. 2015

218 | Khan et al.

Sr# Form of Ag-NPs Fish Group LC50 values Reference

9 Ag-NPs (3nm) Zebra fish (juvenile) 93.11µM Bar-llan et al., 2009

10 Ag-NPs (100nm) Zebra fish (juvenile) 137.26 µM Bar-llan et al., 2009

11 Colloidal Zebra fish (adult) 7.07 mg AgL-1 Griffitt et al., 2008

12 Colloidal Rainbow trout (eleutheroembryo) 0.25 mgL-1 Johari et al., 2013

13 Colloidal Rainbow trout (larvae) 2.16 mgL-1 Johari et al., 2013

14 Colloidal Rainbow trout (alevins) 0.25 mgL-1 Kalbassi et al., 2013

15 Colloidal Rainbow trout (juvenile) 0.25 mgL-1 Kalbassi et al., 2013

16 Colloidal Rainbow trout (juvenile) 2.16 mgL-1 Johari et al., 2013

17 Suspended powder Rainbow trout (alevins) 28.25 mgL-1 Kalbassi et al., 2013

18 Ag-NPs (aged) Medaka (adult) 1.44 mgL-1 Kim et al., 2013

19 Ag-NPs (fresh) Medaka (adult) 3.53 mgL-1 Kim et al., 2013

20 Ag-NPs (stirred) Fathead minnow(embryo 10.6 mgL-1 Laban et al., 2010

21 Ag (sonicated) Fathead minnow 1.36 mgL-1 Laban et al., 2010

Fathead minnow (Pimephales promelas)

The treatment of silver nanoproducts and

nanoparticles cause pericardial, yolk sac edema and

hemorrhages to head region in the Fathead minnow’ s

embryo (Laban et al., 2010) and gills alterations.

Disturbance in the blood circulation is the prevalent

alteration in the gills (Hawkins et al., 2015). 1.3 µg L-1

of AgNO3 increases the total goblet cells in the

mucous. Both Ag-NO3 and Ag-NPs decreases the

Na+/K+-ATPase immune-reactivity in the gills

(Hawkins et al., 2015). The calculated LC50 values

were 9.4mgL-1, 10.6mgL-1 for nanoAmor and Ag-NPs

in case of stirred particles and 1.25mgL-1, 1.36mgL-1

for sonicated particles. Ag-NPs were less toxic than

silver nitrate in fathead minnow (Laban et al., 2010).

Conclusion

Silver nanoparticles attract much attention of

researchers not because of its wide applications in the

fields of medicine, catalysts, biotechnology, nano

biotechnology, electronics, optics, textile engineering

and water treatment but due to toxicity. It can cause

damage to brain cells, liver cells and even to stem

cells in the human body. So instead of using human in

toxicological studies, it is preferable to use animal

models. Among all the models, fish is most

dominantly use model in the toxicological studies.

From the published literature, it is concluded that Ag-

NPs poses toxicity to all the life stages of fish model.

Variation in the toxicity due to size, form and

condition of target model, the researchers are

encouraged to further investigate the different aspects

of Ag-NPs toxicity.

References

Alt V, Bechert T, Steinrucke P, Wagener M,

Seidel P, Dingeldein E, Domann E, Schnettler

R. 2004. An in vitro assessment of the antibacterial

properties and cytotoxicity of nanoparticulate silver

bone cement. Biomaterials 25(18), 4383-4391.

doi:10.1016/j.biomaterials.2003.10.078

Alt V, Wagener M, Salz D, Bechert T,

Steinrucke P, Schnettler R. 2006. Plasma

polymer high-porosity silver composite coating for

infection prophylaxis in intramedullary nailing.

Practice of Intramedullary Locked Nails pp. 297-303.

DOI: 10.1007/3-540-32345-7_30

Ananth AN, Daniel SCG, Sironmani TA,

Umapathi. 2011. PVA and BSA stabilized silver

nanoparticles based surface–enhanced plasmon

resonance probes for protein detection Colloids and

Surfaces B: Biointerfaces 85(2), 138-144.

doi:10.1016/ j.colsurfb.2011.02.012

Arora S, Jain J, Rajwade JM, Paknikar KM.

2009. Interactions of silver nanoparticles with

J. Bio. & Env. Sci. 2015

219 | Khan et al.

primary mouse fibroblasts and liver cells. Toxicology

and Applied Pharmacology 236(3), 310–318.

doi:10.1016/j.taap.2009.02.020.

Asharani PV, Wu YL, Gong Z, Valiyaveetti S.

2008. Toxicity of silver nanoparticles in zebrafish

models. Nanotechnology 19(25), 55-102.

-doi:10.1088/0957-4484/19/25/ 255102

Aslan K, Geddes CD. 2006. Microwave-accelerated

and metalenhanced fluorescence myoglobin detection

on silvered surfaces: Potential application to myo-

cardial infarction diagnosis. Plasmonics 1(1), 53-59.

DOI: 10.1007/s11468-006-9006-7

Aslan K, Huang J, Wilson GM, Geddes CD.

2006. Metalenhanced fluorescence-based RNA

sensing. Journal of American Chemical Society 128,

4206-4207.

ATSDR (Agency for Toxic Substances and

Disease Registry). 1990. Toxicological profile for

Silver. Prepared by Clement international

corporation, under Contract 205-88-0608). U.S.

public Health Service. ATSDR/TP-90-24.

Bar-Ilan O, Albrecht RM, Fako VE, Furgeson

DY. 2009. Toxicity assessments of multisized gold

and silver nanoparticles in Zebrafish embryos. Small

5(16), 1897-910. DOI: 10.1002/smll.200801716.

Bayston R, Ashraf W, Fisher L. 2007. Prevention

of infection in neurosurgery: Role of ‘antimicrobial’

catheters. Journal of Hospital Infection 65(2), 39-42.

DOI: http://dx.doi.org/10.1016/S0195-6701(07)60013-9.

Benn TM, Westerhoff P. 2008. Nanoparticle silver

released into water from commercially available sock

fabrics. Environmental Science and Technology

42(11), 4133–4139. DOI: 10.1021/es7032718.

Bhol KC, Alroy J, Schechter PJ. 2004. Anti-

inflammatory effect of topical nanocrystalline silver

cream on allergic contact dermatitis in a guinea pig

model. Clinical Experimental Dermatology 29(3),

282-287. DOI: 10.1111/j.1365-2230.2004.01515.x.

Bhol KC, Schechter PJ. 2007. Effects of

nanocrystalline silver (NPI 32101) in a rat model of

ulcerative colitis. Digestive Diseases and Sciences 52,

2732-2742. DOI: 10.1007/s10620-006-9738-4.

Bilberg K, Doving KB, Beedholm K, Baatrup

E. 2011. Silver nanoparticles disrupt olfaction in

Crucian carp (Carassius carassius) and Eurasian

perch (Perca fluviatilis). Aquatic Toxicology 104,

145–152. doi: 10.1016/j.aquatox.2011. 04.010.

Blaser SA, Scheringer M, MacLeod M,

Hungerbühler K. 2008. Estimation of cumulative

aquatic exposure and risk due to silver: Contribution

of nano-functionalized plastics and textiles. Science of

Total Environment 390 (2-3), 396–409.

DOI: 10.1016/ j.scitotenv. 2007.10.010.

Bouadma L, Wolff M, Lucet JC. 2012. Ventilator-

associated pneumonia and its prevention. Current

opinion in infectious diseases 25 (4), 395–404. doi:

10.1097/ QCO.0b013e328355a835.

Braydich-Stolle L, Hussain S, Schlager JJ,

Hofmann MC. 2005. In vitro cytotoxicity of

nanoparticles in mammalian germline stem cells.

Toxicological Science 88(2), 412–419.

doi: 10.1093/toxsci/kfi340.

Carlson C, Hussain S, Schrand A, Braydich-

Stolle L, Hess K, Jones R, Schlager J. 2008.

Unique cellular interaction of silver nanoparticles:

Size-dependent generation of reactive oxygen species.

The Journal of Physical Chemistry 112(43), 13608-

13619. DOI: 10.1021/jp712087m.

Catsakis LH, Sulica VI. 1978. Allergy to silver

amalgams. Oral Surgery Medicine Oral Pathology

Oral Radiology 46(3), 371-375.

DOI: http://dx.doi.org/ 10.1016/0030-4220(78)90402-4.

J. Bio. & Env. Sci. 2015

220 | Khan et al.

Chae YJ, Pham CH, Lee J, Bae E, Yi J, Gu MB.

2009. Evaluation of the toxic impact of silver

nanoparticles on Japanese medaka (Oryzias latipes).

Aquatic Toxicology 94(4), 320–327.

doi:10.1016/j.aquatox.2009.07.019

Chen LQ, Fang L, Ling J, Ding CZ, Kang B, Huang

CZ. 2015. Nanotoxicity of silver nanoparticles to red

blood cells: size dependent adsorption, uptake, and

hemolytic activity. Chemical Research in Toxicology

28(3), 501-9. doi: 10.1021/tx 500479m.

Chen W, Liu Y, Courtney HS, Bettenga M,

Agrawal CM, Bumgardner JD, Ong JL. 2006. In

vitro anti-bacterial and biological properties of

magnetron co-sputtered silver-containing hydroxyapatite

coating. Biomaterials 27(32), 5512-5517.

doi:10.1016/j.biomaterials.2006.07.003.

Chen X, Schluesener HJ. 2008. Nanosilver: A

nanoproduct in medical application. Toxicological

Letters 176(1), 1–12.

doi:10.1016/j.toxlet.2007.10.004.

Cho JG, Kim KT, Ryu TK, Lee JW, Kim JE,

Kim J, Lee BC, Jo EH, Yoon J, Eom IC, Choi

K, Kim P. 2013. Stepwise Embryonic Toxicity of

Silver Nanoparticles on Oryzias latipes. BioMed

Research International, Article ID 494671, 7 pages.

http:// dx. doi.org/10.1155/2013/494671.

Choi JE, Kim S, Ahn JH, Youn P, Kang JS,

Park K, Yi J, Ryu D. 2009. Induction of oxidative

stress and apoptosis by silver nanoparticles in the

liver of adult Zebrafish. Aquatic Toxicology 100(2),

151-159. doi: 10.1016/j.aquatox. 2009.12.012.

Coelho S, Amarelo M, Ryan S, Reddy M,

Sibbald RG. 2004. Rheumatoid arthritis-associated

inflammatory leg ulcers: A new treatment for

recalcitrant wounds. International Wound Journal

1(1), 81-84. DOI: 10.1111/j.1742-481x.2004. 0002.x.

Cohen MS, Stern JM, Vanni AJ, Kelley RS,

Baumgart E, Field D, Libertino JA, Summer-

hayes IC. 2007. In vitro analysis of a nanocrystalline

silver-coated surgical mesh. Surgical Infections

(Larchmt.) 8(3), 397-403. DOI: 10.1089/sur.

2006.032

Deery C. 2009. Silver lining for caries cloud?

Evidence-Based Dentistry 10(3), 68. doi:10.1038/ sj.

ebd.6400661

Diehl AM. 2000. Cytokine regulation of liver injury

and repair. Immunological Reviews 174(1), 160-171.

DOI: 10.1034/j.1600-0528.2002.017411.x

Dowling A, Clift R, Grobert N, Hutton D,

Oliver R, Neill O, Pethica J, Inoue KI, Takano

H, Yanagisawa R, Koike E, Shimada A. 2009.

Size effects of latex nanomaterials on lung

inflammation in mice. Toxicology and Applied

Pharmacology 234(1), 68-76.

doi:10.1016/j.taap.2008.09.012

Drake PL, Hazelwood KJ. 2005. Exposure-related

health effects of silver and silver compounds: A

review. The Annals of Occupational Hygiene 49(7),

575-585. doi: 10.1093/annhyg/mei019

EC. 1999. Annex VI of Directive 1999/45/EC to

consolidated version of directive 67/548/EEC.

General classification and labeling requirements for

dangerous substances and preparations.

ec.europa.eu/environment/archives/dansub/pdfs/an

nex6_ en.pdf

EC. 2008. Regulation (EC) No 1272/2008 of the

European Parliament and Council of 16 December

2008 on classification, labeling and packaging of

substances and mixtures, Official Journal of the

European Union, 31.12.2008.

http://eur-lex.europa.eu/legal-content/en/TXT/?uri

=CELEX:32008R1272.

J. Bio. & Env. Sci. 2015

221 | Khan et al.

Elechiguerra JL, Morones JR, Camacho A,

Holt K, Kouri JB, Ramirez JT, Yacaman MJ.

2005. Interaction of silver nanoparticles with HIV-1.

Journal of Nanotechnology 16, 23-46.

DOI: 10.1186/1477-3155-3-6

Farkas J, Christian P, Gallego JA, Urrea N,

Roos, Hassellöv M, Tollefsen KE, Thomas KV.

2010. Effects of silver and gold nanoparticles on

rainbow trout (Oncorhynchus mykiss) hepatocytes.

Aquatic Toxicology 96(1), 44-52. doi:10.1016/j.

aquatox.2009.09. 016

Gliga AR, Skoglund S, Wallinder IO, Fadeel B,

Karlsson HL. 2014. Size-dependent cytotoxicity of

silver nanoparticles in human lung cells: the role of

cellular uptake, agglomeration and Ag release.

Particle and Fibre Toxicology 11(11), 1-17 doi:

10.1186/1743-8977-11-11

Gonzalez P, Baudrimont M, Boudou A,

Bourdineaud JP. 2006. Comparative effects of

direct cadmium contamination on gene expression in

gills, liver, skeletal muscles and brain of the zebrafish

(Danio rerio). Biometals 19(3), 225–235. DOI:

10.1007/s10534-005-5670-x.

Griffitt RJ, Hyndman K, Denslow ND, Barber

DS. 2009. Comparison of molecular and histological

changes in zebrafish gills exposed to metallic

nanoparticles. Toxicological Science 107(2), 404-415.

doi: 10.1093/toxsci/kfn256

Griffitt RJ, Luo J, Gao J, Bonzongo JC, Barber

DS. 2008. Effects of particle composition and species

on toxicity of metallic nanomaterials in aquatic

organisms. Environmental Toxicology and Chemistry

27(9), 1972–1978. DOI: 10.1897/08-002.1

Gulbranson SH, Hud JA, Hansen RC. 2000.

Argyria following the use of dietary supplements

containing colloidal silver protein. Cutis 66, 373-376.

Handy RH, Owen R, Valsami-Jones E. 2008.

The ecotoxicology of nanoparticles and nanomaterials:

current status, knowledge gaps, challenges, and

future needs. Ecotoxicology 17(5), 315-325. doi:

10.1007/s10646-008-0206-0.

Hawkins AD, Thornton C, Kennedy AJ, Bu

K, Cizdziel J, Jones BW, Steevens JA, Willett

KL. 2015. Gill histopathologies following exposure to

nanosilver or silver nitrate. Journal of Toxicology and

Environmental Health A 78(5), 301-15.

doi: 10.1080/15287394.2014.971386.

He J, Lin L, Liu H, Zhang P, Lee M, Sankey OF,

Lindsay SM. 2009. A hydrogen-bounded electron-

tunneling circuit reads the base composition of

unmodified DNA. Nanotechnology 20(7), 075102.

doi: 10.1088/0957-4484/20/7/075102

Hedayati A, Kolangi H, Jahanbakhshi A,

Shaluei F. 2012a. Evaluation of Silver nanoparticles

Ecotoxicity in Silver carp (Hypophthalmicthys molitrix)

and Goldfish (Carassius auratus). Bulgarian Journal

of Veterinary Medicine 15(3), 172−177. Article id:

80158939

Hedayati A, Shaluei F, Jahanbakhshi A. 2012b.

Comparison of Toxicity Responses by Water Exposure to

Silver Nanoparticles and Silver Salt in Common Carp

(Cyprinus carpio). Global Veterinaria 8(2), 179-184.

Hussain SM, Hess KL, Gearhart JM, Geiss KT,

Schlager JJ. 2005. In vitro toxicity of nanoparticles

in BRL 3A rat liver cells. Toxicology in Vitro 19(7),

975–983. doi:10.1016/j.tiv.2005.06.034.

Inoue Y, Uota M, Torikai T, Watari T, Noda I,

Hotokebuchi T. 2010. Antibacterial properties of

nanostructured silver titanate thin films formed on a

titanium plate. Journal of Biomedical Materials

Research Part A 92A(3), 1171-1180.

doi: 10.1002/jbm.a.32456.

J. Bio. & Env. Sci. 2015

222 | Khan et al.

Ivask A, Kurvet I, Kasemets K, Blinova I,

Aruoja V. 2014. Size-Dependent Toxicity of Silver

Nanoparticles to Bacteria, Yeast, Algae, Crustaceans

and Mammalian Cells In Vitro. PLoS ONE 9(7),

e102108. doi:10.1371/journal.pone.0102108

Jahanbakhshi A, Shaluei F, Hedayati A. 2012b.

Detection of Silver Nanoparticles (Nanosil®) LC50 in

Silver Carp (Hypophthalmichthys molitrix) and

Goldfish (Carassius auratus). World Journal of

Zoology 7(2), 126-130. DOI: 10.5829/idosi.

wjz.2012.7.2.62129.

Jang MH, Kim WK, Lee SK, Henry TB, Park

JW. 2014. Uptake, tissue distribution, and

depuration of total silver in common carp (Cyprinus

carpio) after aqueous exposure to silver nanoparticles.

Environmental Science and Technology 48(19),

11568-74. doi: 10.1021/es5022813.

Johari SA, Kalbassi MR, Soltani M, Yu IJ.

2013. Toxicity comparison of colloidal silver

nanoparticles in various life stages of rainbow trout

(Oncorhynchus mykiss). Iranian Journal of Fisheries

Science 12(1), 76 -95.

Jones CM, Hoek EM. 2010. A review of the

antibacterial effects of silver nanomaterials and

potential implications for human health and the

environment. Journal of Nanoparticle Research 12,

1531–1551.

Jovanovic B, Anastasova L, Rowe EW, Zhang Y,

Clapp AR, Palic D. 2011. Effects of nanosized titanium

dioxide on innate immune system of fathead minnow

(Pimephales promelas Rafinesque, 1820). Ecotoxicology

and Environmental Safety 74(7), 675-683.

DOI: 10.1016/j.ecoenv.2010.10.017

Jung WK, Kim SH, Koo HC, Shin S, Kim JM,

Park YK, Hwang SY, Yang H, Park YH. 2007.

Antifungal activity of the silver ion against

contaminated fabric. Mycoses 50(4), 265–269.

DOI: 10.1111/j.1439-0507.2007.01372.x

Kalbassi MR, Johari SA, Soltani M, Yu LJ.

2013. Particle Size and Agglomeration Affect the

Toxicity Levels of Silver Nanoparticle Types in

Aquatic Environment. ECOPERSIA 1(3), 273-290.

Kannan R, Jerley A, Ranjani M, Prakash V.

2011. Antimicrobial silver nanoparticle induces organ

deformities in the developing Zebra fish (Danio rerio)

embryos. Journal of Biomedical Science and

Engineering 4, 248-254.

doi: 10.4236/ jbise.2011.44034.

Katuli KK, Massarsky A Hadadi A, Pourmehran

Z. 2014. Silver nanoparticles inhibit the gill Na⁺/K⁺-

ATPase and erythrocyte AChE activities and induce

the stress response in adult zebrafish (Danio rerio).

Ecotoxicology and Environmental Safety 106, 173-80

doi: 10.1016/j.ecoenv.2014.04.001.

Kim J, Kuk E, Yu K, Park S, Lee H, Kim S,

Park Y, Hwang C, Kim Y, Lee Y, Jeong D, Cho

M. 2007. Antimicrobial effects of silver nanoparticles.

Nanomedicine: Nanotechnology, Biology and Medicine

3(1), 95-101. doi:10.1016/j.nano. 2006.12.001

Kim J, Lee J, Kwon S, Jeong S. 2009.

Preparation of biodegradable polymer/silver nano-

particles composite and its antibacterial efficacy.

Journal of Nanoscience and Nanotechnology 9(2),

1098–1102. doi:10.1166/jnn.2009.C096

Kim JY, Kim KT, Lee BG, Lim BJ, Kim SD.

2013. Developmental toxicity of Japanese medaka

embryos by silver nanoparticles and released ions in

the presence of humic acid. Ecotoxicology and

Environmental Safety 92(1), 57-63. doi: 10.1016/j.

ecoenv.2013.02.004.

Kirsner RS, Orstead H, Wright JB. 2001. Matrix

metalloproteinases in normal and impaired wound

healing: a potential role for nanocrystalline silver.

Wounds 13(3), 5-12.

J. Bio. & Env. Sci. 2015

223 | Khan et al.

Klaprat DA, Evans RE, Hara TJ. 1992.

Environmental contaminants and chemoreception in

fishes. In Fish Chemoreception Fish and Fisheries

Series 6, 321-341. DOI: 10.1007/978-94-011-2332-

7_15.

Kumar PS, Sivakumar R, Anandan S,

Madhavan J, Maruthamuthu P, Ashokkumar

M. 2008. Photocatalytic degradation of Acid Red 88

using Au TiO2 nanoparticles in aqueous solutions.

Water Research 42(19), 4878–4884. doi:10.1016/j.

watres.2008.09.027

Kwok KW, Auffan M, Badireddy AR, Nelson

CM, Wiesner MR, Chilkoti A, Liu J, Marinakos

SM, Hinton DE. 2012. Uptake of silver nanoparticles

and toxicity to early life stages of Japanese medaka

(Oryzias latipes): effect of coating materials. Aquatic

Toxicology 120(121), 59-66.

doi: 10.1016/j.aquatox.2012.04.012.

Laban G, Nies LF, Turco RF, Bickham JW,

Sepulveda MS. 2010. The effects of silver

nanoparticles on fathead minnow (Pimephales

promelas) embryos. Ecotoxicology 19(1), 185-195.

DOI: 10.1007/s10646-009-0404-4

Lancaster T, Stead LF. 2012. Silver acetate for

smoking cessation. The Cochrane Collaboration.

(Online) 9, CD000191.

DOI: 10.1002/14651858.CD000191.

Lansdown A. 2006. Silver in health care:

antimicrobial effects and safety in use. Current

Problems in Dermatology 33, 17-34.

DOI: 10.1159/000093928.

Larese FF, Dagostin F, Crosera M, Adami G,

Renzi N, Bovenzi M, Maina G. 2009. Human

skin penetration of silver nanoparticles through intact

and damaged skin. Toxicology 255(1-2), 33–37.

doi:10.1016/j.tox.2008.09.025

Lee B, Duong C, Cho J, Lee J, Kim K, Seo Y,

Kim P, Choi K, Yoon J. 2012. Toxicity of Citrate-

Capped Silver Nanoparticles in Common Carp

(Cyprinus carpio). Journal of Biomedicine and

Biotechnology 2012, 262670.

doi: 10.1155/2012/262670.

Lee BC, Kim J, Cho JG, Lee JW, Duong CN, Bae

E, Yi J, Eom IC, Choi K, Kim P, Yoon J. 2014.

Effects of ionization on the toxicity of silver

nanoparticles to Japanese medaka (Oryzias latipes)

embryos. Toxic/Hazardous Substances and Environ-

mental Engineering 49(3), 287-93.

doi: 10.1080/10934529.2014. 846614.

Lee HJ, Yeo SY, Jeong SH. 2003. Antibacterial

effect of nanosized silver colloidal solution on textile

fabrics. Journal of Materials Science 38(10), 2199-

2204. DOI: 10.1023/A: 1023736416361.

Lesniak W, Bielinska AU, Sun K, Janczak KW,

Shi X, Baker JR, Balogh LP, 2005. Silver

/dendrimer nanocomposites as biomarkers:

Fabrication, characterization, in vitro toxicity, and

intracellular detection. Nano Letters 5(11), 2123-

2130. DOI: 10.1021/nl051077u.

Li Q, Mahendra S, Lyon, DY, Brunet L, Liga

MV, Li D, Alvarez PJJ. 2008. Antimicrobial

nanomaterials for water disinfection and microbial

control: potential applications and implications.

Water Research 42 (18), 4591–4602.

doi: 10.1016/j.watres.2008.08.015.

Luoma SN, Rainbow PS. 2008. Metal

contamination in aquatic environments: science and

lateral management. Journal of Fish Biology 75,

1911–1912.

DOI: 10.1111/j.1095-8649.2009.02440_4.x.

Massarsky A, Abraham R, Nguyen KC,

Rippstein P, Tayabali AF, Trudeau VL, Moon

TW. 2014. Nanosilver cytotoxicity in rainbow trout

(Oncorhynchus mykiss) erythrocytes and hepatocytes.

J. Bio. & Env. Sci. 2015

224 | Khan et al.

Comparative Biochemistry and Physiology Part C:

Pharmacology, Toxicology and Endocrinology; 159,

0-21. doi: 10.1016/j. cbpc.2013.09.008.

Moaddab S, Ahari H, Shahbazzadeh D,

Motallebi A, Anvar A, Rahman-Nya J,

Shokrgozar MA. 2011. Toxicity study of nanosilver

(Nanocid®) on osteoblast cancer cell Line.

International Nano Letters 1(1), 11-16.

Monfared AL, Soltani S. 2013. Effects of silver

nanoparticles administration on the liver of rainbow

trout (Oncorhynchus mykiss): histological and

biochemical studies. European Journal of

Exponential Biology 3(2), 285-289.

NCCAM. 2012. Colloidal Silver Products. National

Center for Complementary and Alternative Medicine.

February 2012.

https://nccih.nih.gov/health/providers/digest/topsu

pplements

Nowack B, Bucheli TD. 2007. Occurrence,

behavior and effects of nanoparticles in the

environment. Environmental Pollution 150(1), 5-22.

doi:10.1016/j. envpol.2007.06.006

Nowack B, Krug HF, Height M. 2011. 120 years of

nanosilver history: implications for policy makers.

Environmental Science and Technology 45(4), 1177–

1183. DOI: 10.1021/es103316q.

Pal S, Tak YK, Song JM. 2007. Does the

Antibacterial Activity of Silver Nanoparticles Depend

on the Shape of the Nanoparticle? A Study of the

Gram-Negative Bacterium Escherichia coli. Applied

and Environmental Microbiology 73(6), 1712-1720.

doi:10.1128/AEM.02218-06.

Panyala NR, Pena-Mendez EM, Havel J. 2008.

Silver or silver nanoparticles: a hazardous threat to

the environment and human health? Journal of

Applied Biomedicine 6, 117–129.

Perelshtein I, Applerot G, Perkas N, Guibert

G, Mikhailov S, Gedanken A. 2008. Sonochemical

coating of silver nanoparticles on textile fabrics

(nylon, polyester and cotton) and their antibacterial

activity. Nanotechnology 19, 245705.

doi:10.1088/0957-4484/19/24/245705

Pohle D, Damm C, Neuhof J, Rosch A,

Munstedt H. 2007. Antimicrobial properties of

orthopaedic textiles after in-situ deposition of silver

nanoparticles. Polymers & Polymer Composites

15(5), 357-363. Accession# 28655926.

Powers CM, Yen J, Linney EA, Seidler FJ,

Slotkin TA. 2010. Silver exposure in developing

Zebrafish (Danio rerio): Persistent effects on larval

behavior and survival. Neurotoxicology and

Teratology 32(3), 391-397.

doi:10.1016/j.ntt.2010.01.009

Project on emerging nanotechnologies. 2013.

Available online: http://www. nanotechproject. org/

inventories/consumer/ (accessed on 3 June 2013).

Rajkumar KS, Kanipandian N, Thirumurugan

R. 2015. Toxicity assessment on haemotology,

biochemical and histopathological alterations of silver

nanoparticles-exposed freshwater fish Labeo rohita.

Applied Nanoscience DOI 10.1007/s13204-015-0417-7.

Rathore RS, Khangarot BS. 2002. Effect of

temperature on the sensitivity of sludge worm Tubifex

tubifex (Muller) to selected heavy metals. Ecotoxi-

cology and Environmental Safety 53(1), 27–36.

doi:10.1006/eesa.2001.2100

Reddy TK, Reddy SJ, Prasad TNVKV. 2013.

Effect of Silver Nanoparticles on Energy Metabolism

in Selected Tissues of Aeromonas Hydrophila

Infected Indian Major Carp, Catla Catla. IOSR

Journal of Pharmacy 3(1), 49-55.

Reidy B, Haase A, Luch A, Dawson KA, Lynch

A. 2013. Mechanisms of Silver Nanoparticle Release,

J. Bio. & Env. Sci. 2015

225 | Khan et al.

Transformation and Toxicity: A Critical Review of

Current Knowledge and Recommendations for Future

Studies and Applications. Materials 6, 2295-2350.

doi: 10.3390/ma6062295.

Rivero P, Urrutia A, Goicoechea J, Zamarreno

C, Arregui F, Matias I. 2011. An antibacterial

coating based on a polymer/solgel hybrid matrix

loaded with silver nanoparticles. Nanoscale Research

Letters 6(305). doi:10.1186/1556-276X-6-305.

Rosenblatt A, Stamford TCM, Niederman R.

2009. Silver Diamine Fluoride: A Caries Silver-

Fluoride Bullet. Journal of Dental Research 88 (2),

116–125. DOI: 10.1177/0022034508329406

Rothkamm K, Lobrich M. 2003. Evidence for a

lack of DNA double-strand break repair in human

cells exposed to very low X-ray doses. Proceding of

National and Academy of Sciences U.S.A. 100, 5057-

5062. doi: 10.1073/pnas.0830918100

Russell AD, Hugo WB. 1994. Antimicrobial

activity and action of silver. Progress in Medicinal

Chemistry 31, 351-370.

Safaepour M, Shahverdi A, Shahverdi H,

Khorramizadeh M, Gohari A. 2009. Green

synthesis of small silver nanoparticles using geraniol and

its cytotoxicity against Fibro sarcoma-Wehi 164.

Avicenna Journal of Medical Biotechnology 1(2), 111-

115.

Samuel U, Guggenbichler J. 2004. Prevention of

catheter-related infections: the potential of a new

nano-silver impregnated catheter. International

Journal of Antimicrob Agents 23, 75-78.

DOI: 10.1016/j.ijantimicag.2003.12.004

Schrand AM, Braydich-Stolle LK, Schlager JJ,

Dai L, Hussain SM. 2008. Can silver nanoparticles

be useful as potential biological labels? Nanotech-

nology 19(2), 235104.

doi: 10.1088/0957-4484/19/23/235104

Schultz AG, Ong KJ, MacCormack T, Ma G,

Veinot JG, Goss GG. 2012. Silver nanoparticles

inhibit sodium uptake in juvenile rainbow trout

(Oncorhynchus mykiss). Environmental Science and

Technology 46(18), 10295-301.

doi: 10.1021/es3017717.

Scown TM, Santos EM, Johnston BD, Gaiser

B, Baalousha M, Mitov S, Lead JR, Stone V,

Fernandes TF, Jepson M, Van Aerle R, Tyler

CR. 2010. Effects of aqueous exposure to silver

nanoparticles of different sizes in rainbow trout.

Toxicological Science 115(2), 521–534. doi:

10.1093/toxsci/ kfq076.

Shaluei F, Hedayati A, Jahanbakhshi A, Kolangi

H, Fotovat M. 2013. Effect of subacute exposure

to silver nanoparticle on some hematological and

plasma biochemical indices insilver carp (Hypoph-

thalmichthys molitrix). Human& Experimental

Toxicology 32(12), 1270-7.

doi: 10.1177/0960327113485258.

Sharma VK, Siskova KM, Zboril R, Gardea-

Torresdey JL. 2014. Organic-coated silver

nanoparticles in biological and environmental

conditions: fate, stability and toxicity. Advances in

Colloid and Interface Science 204, 15-34. doi:

10.1016/j. cis.2013.12.002.

Silver Institute, 2014. World Silver Survey 2014.

https://www.silverinstitute.org/ site/supply- demand/

Silver S. 2003. Bacterial silver resistance: Molecular

biology and uses and misuses of silver compounds.

FEMS Microbiology Reviews 27, 341-353.

DOI: http://dx. doi.org/10.1016/S0168-6445(03) 000

47-0

Skirtach AG, Antipov AA, Shchukin DG,

Sukhorukov GB. 2004. Remote activation of

capsules containing Ag nanoparticles and IR dye by

laser light. Langmuir 20(17), 6988-6992.

DOI: 10.1021/la048873k

J. Bio. & Env. Sci. 2015

226 | Khan et al.

Smith I, Carson B. 1977. Trace metals in the

environment. Trace Metals in the Environment 469

pp. ISBN: 978-0-444-50352-7

Soderstjerna E, Bauer P, Cedervall T, Abdshill

H, Johansson F. 2014. Silver and Gold

Nanoparticles Exposure to In Vitro Cultured Retina

Studies on Nanoparticle Internalization, Apoptosis,

Oxidative Stress, Glial- and Microglial Activity. PLoS

ONE 9(8), e105359.

doi:10.1371/journal.pone.0105359.

Sondi I, Sondi BS. 2004. Silver nanoparticles as

antimicrobial agent: a case study on E. coli as a model

for Gram-negative bacteria. Journal of Colloid and

Interface Science 275(1), 177–182.

doi:10.1016/j.jcis.2004.02.012

Sung J, Ji J, Yoon J, Kim D, Song M, Jeong J,

Han B, Han J, Chung Y, Kim J, Kim T, Chang

H, Lee E, Lee J, Yu I. 2008. Lung function changes

in Sprague-Dawley rats after prolonged inhalation

exposure to silver nanoparticles. Inhalation

Toxicology 20(6), 567–574. doi:10.1080/089583707

01874671.

Syrvatka V, Rozgoni I, Slyvchuk Y, Milovanova

G, Hevkan I, Matyukha I. 2014. Effects of Silver

nanoparticles in Solution and liposomal form on

some blood Parameters in female rabbits during

fertilization and early embryonic development.

Journal of microbiology, biotechnology and food

sciences 3 (4), 274-278. ICID: 1092144

Tai SP, Wu Y, Shieh BD, Chen LJ, Lin KJ, Yu

CH, Chu SW, Chang CH, Shi XY, Wen YC, Lin

KH, Liu TM, Sun CK. 2007. Molecular imaging of

cancer cells using plasmonresonant- enhanced third-

harmonic-generation in silver nanoparticles. Advance

Materials 19, 4520-4523.

DOI: 10.1002/adma.200602213.

Taju G, Majeed AS, Nambi KS, Sahul Hameed

AS. 2014. In vitro assay for the toxicity

of silver nanoparticles using heart and gill cell lines of

Catla catla and gill cell line of Labeo rohita.

Comparative Biochemistry and Physiology Part C:

Pharmacology, Toxicology and Endocrinology 161,

41-52. doi: 10.1016/j.cbpc.2014.01.007.

Tian J, Wong KK, Ho CM, Lok CN, Yu WY, Che

CM, Chiu JF, Tam PK. 2007. Topical delivery of

silver nanoparticles promotes wound healing. Chem

Med Chem 2(1), 129-136.

DOI: 10.1002/cmdc.200600171

Tredget EE, Shankowsky HA, Groeneveld A,

Burrell R. 1998. A matched-pair randomized study

evaluating the efficacy and safety of Acticoat silver-

coated dressing for the treatment of burn wounds.

Journal of Burn Care & Rehabilitation 19, 531-537.

DOI: 10.1097/00004630-199811000-00013.

Walt DR. 2005. Miniature analytical methods for

medical diagnostics. Science 308(5719), 217-219.

DOI: 10.1126/science.1108161

Wan AT, Conyers RA, Coombs CJ, Masterton

JP. 1991. Determination of silver in blood, urine and

tissues of volunteers and burn patients. Clinical

Chemistry 37(10), 1683-1687.

Webb NA, Wood CM. 1998. Physiological analysis

of the stress response associated with acute silver

nitrate exposure in freshwater rainbow trout

(Oncorhynchus mykiss). Environmental Toxicology

and Chemistry 17(4), 579–588.

DOI: 10. 1002/etc.5620170408

Weisbarth RE, Gabriel MM, George M,

Rappon J, Miller M, Chalmers R, Winterton L.

2007. Creating antimicrobial surfaces and materials

for contact lenses and lens cases. Eye and Contact

Lens 33, 426-429.

West JL, Halas NJ. 2003. Engineered nanomaterials

for biophotonics applications: Improving sensing,

imaging, and therapeutics. Annual Review of

J. Bio. & Env. Sci. 2015

227 | Khan et al.

Biomedical Engineering 5, 285–292. doi: 10.1146/

annurev.bioeng.5.011303.120723.

Wijnhoven SWP, Peijnenburg WJGM,

Herberts CA, Hagens WI, Oomen AG,

Heugens EHW, Roszek B, Bisschops J, Gösens

I, Van de Meent D, Dekkers S, De Jong WH,

Van Zijverden M, Sips AJAM, Geertsma RE.

2009. Nano-silver - A review of available data and

knowledge gaps in human and environmental risk

assessment. Nanotoxicology 3(2), 109-138.

doi:10.1080/17435390902725914.

Woodrow Wilson Database. 2015. Nanotechnology

consumer product inventory

http://www.nanotechproject.org/cpi/about/analysis/.

World Health Organization. 2002. Silver and

silver compounds: Environmental aspects. (Concise

international chemical assessment document; 44). 1.

Silver _ adverse effects 2. Water pollutants, Chemical

3. Risk assessment 4. Environmental exposure I.

International Programme on Chemical Safety II.

Series ISBN 92 4 153044 8 (NLM Classification: QV

297). ISSN 1020 6167.

http://www.who.int/ipcs/publications/cicad /en/cicad

44.Pdf

Wright JB, Lam K, Hansen D, Burrell RE.

1999. Efficacy of topical silver against fungal burn

wound pathogens. American Journal of Infection

Control 27(4), 344-350. doi:10.1016/S0196-

6553(99)70055-6

Wu Y, Zhoua Q, Li H, Liua W, Wanga T,

Jianga G. 2010. Effects of silver nanoparticles on the

development and histopathology biomarkers of

Japanese medaka (Oryzias latipes) using the partial-

life test. Aquatic Toxicology 100(2), 160-167. doi:

10.1016/j.aquatox.2009.11.014

Yeo M, Kang M. 2008. Effects of nanometer sized

silver materials on biological toxicity during Zebra

fish embryogenesis. Bulletin of the Korean Chemical

Society 29(6), 1179-1184.

Yon JN, Lead JR. 2008. Manufactured

nanoparticles: An overview of their chemistry,

interactions and potential environmental

implications. Science of Total Environment 400(1-

3), 396–414. doi:10.1016/j.scitotenv.2008.06.042.

Yu H, Xu X, Chen X, Lu T, Zhang P, Jing X.

2007. Preparation and antibacterial effects of PVA-

PVP hydrogels containing silver nanoparticles.

Journal of Applied Polymer Science 103, 125-133.

DOI: 10.1002/app.24835

Yves MJ, Philippe H. 2012. Silver as an

antimicrobial: Facts and gaps in knowledge. Critical

Reviews in Microbiology 39(4), 373-83. doi:

10.3109/1040841X.2012. 713323.

Zhao Y, Shanmukh S, Liu Y, Jones L, Dluhy

RA, Tripp RA. 2006. Silver nanorod arrays can

distinguish virus strains. Nanotech SPIE Newsroom

DOI: 10.1117/2. 1200610.0438.