Characterization and potential applications of silver nanoparticles: an insight on different mechanisms published by Ural Federal University eISSN 2411-1414; chimicatechnoacta.ru REVIEW 2022, vol. 9(4), No. 20229402 DOI: 10.15826/chimtech.2022.9.4.02 1 of 20 Characterization and potential applications of silver nanoparticles: an insight on different mechanisms Anton N. Kodintcev * Institute of High Temperature Electrochemistry of the Ural Branch of the Russian Academy of Sciences, Ekaterinburg 620066, Russia * Corresponding author: antonkodintsev@mail.ru This invited review belongs to a Regular Issue. © 2022, the Authors. This article is published in open access under the terms and conditions of the Creative Commons Attribution (CC BY) license (http:creativecommons:licenses/by/4.0/). Abstract In the 21st century, a great interest is devoted to biomedical applica- tion of various nanoparticles, particularly, as a means of improving the effectiveness of therapy for different diseases. Silver nanoparti- cles (AgNPs) are among the most studied types of nanoparticles. Due to the wide spectrum of their action, silver nanoparticles may be used both to influence pathogenic microorganisms and to improve the treatment of cancer. The basic physicochemical characteristics and stabilizing agents play an important modifying role in the phar- macokinetics and pharmacodynamics of nanoparticles, determining the severity of the caused effect and their potential toxicity. This re- view summarizes the main physicochemical properties of AgNPs and their impact on the biological effects. Additionally, biochemical and pathophysiological mechanisms of silver nanoparticles activity against various microorganisms and tumor cells are considered. Fi- nally, we address the problems associated with determining the op- timal characteristics of nanoparticles in order to increase their effi- ciency and reduce their toxicity for the macroorganism. Keywords silver nanoparticles physicochemical properties biological effects toxicity Received: 23.06.22 Revised: 25.06.22 Accepted: 25.06.22 Available online: 11.07.22 1. Introduction Currently, the research on biomedical properties of vari- ous nanoparticles, synthesized from gold or silver, attracts a considerable interest. Despite the lack of data on the mechanisms of silver toxicity in vivo, the nanoparticles obtained from it are widely used as antibacterial sub- stances in medical, cosmetic, food and textile industries. Silver nanoparticles (AgNPs) have unique binding proper- ties, including internal antimicrobial activity and poten- tially low toxicity. Silver, and, in particular, its ions have the strongest antibacterial effect among metals; AgNPs may be used in wound dressings, local drug delivery sys- tems, orthopedic and orthodontic materials, antiseptic solutions, catheters, bandages, tissue scaffolds and protec- tive clothing [1–3]. Thus, AgNPs can be an effective alter- native to local antibacterial drugs because of special phys- icochemical properties and wide spectrum of action against various Gram-negative (Escherichia coli, Neisseria gonorrhea, P. aeruginosa) and Gram-positive (S. aureus, including MRSA) bacteria, as well as intracellular micro- organisms (Chlamydia trachomatis) and different viruses [3–5]. AgNPs may be considered as a means to modernize the treatment regimens for Mycobacteria, such as tubercu- losis [6]. The systematic review of Fakhruddin et al. demonstrates the existence of the evidence of antimicrobi- al properties of silver nanoparticles against cariogenic flora in vitro and prevention of the dentin destruction [7]. Moreover, there is evidence of synergistic action of nanoparticles and various antibiotics. Combining AgNPs with these class of drugs could increase the antibacterial activity of imipenem, tetracycline, aminopenicillins, met- ronidazole, gentamicin, kanamycin, streptomycin, and vancomycin [8–14]. Silver nanoparticles may interact with bacterial membranes and penetrate into the cell with the subse- quent initiation of vital malfunction and structural changes leading to destruction and death of pathogenic microorganisms [15]. The key factor is the activation of oxidative stress processes leading to dysfunction of cellu- lar structures such as DNA (genotoxic effect). Disturb- ance of protein structure occurs due to the formation of Ag–S complexes, which leads to malfunction of mem- brane pumps and respiratory chain. Activation of lipid peroxidation processes is another important factor of cellular dysfunction [16]. http://chimicatechnoacta.ru/ https://doi.org/10.15826/chimtech.2022.9.4.02 mailto:antonkodintsev@mail.ru http://creativecommons.org/licenses/by/4.0/ https://orcid.org/0000-0002-3978-8902 https://crossmark.crossref.org/dialog/?doi=https://doi.org/10.15826/chimtech.2022.9.4.02&domain=pdf&date_stamp=2022-7-11 Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 2 of 20 A comparative analysis of the antibacterial effective- ness of silver ions and silver nanoparticles elucidated that they boost the development of many pores in the cell membrane and, consequently, cause the outside leakage of the cytoplasm and macromolecules. Besides, there is a significant damage of the flagella, which impairs the mo- bility of bacteria [19, 20, 21]. AgNPs have an antifungal effect (Candida albicans, As- pergillus niger) when adding material to silicone- containing linings and resins. In addition, there are data on their effectiveness against Penicillium citrinum and Trichophyton mentagrophytes [20–22]. Beyond antibacte- rial and antifungal activity, AgNPs demonstrate anti- inflammatory and antitumor properties [2, 3, 21, 22]. It is assumed that AgNPs are able to enhance the immunogen- icity of vaccines, have an antidiabetic effect and also con- tribute to bone regeneration and wound healing [3]. Despite the large number of studies indicating antibacte- rial properties of silver, there is evidence of the possible phe- notypic resistance of bacteria to AgNPs. During repeated ap- plying of nanoparticles, the increasing aggregation under the influence of flagellin protein was detected. This process leads to subsequent loss of beneficial antibacterial effect. Inhibition of flagellin, for example, with pomegranate peel extract, helps to overcome this resistance mechanism [23, 24]. It should be noted that the bactericidal and bacterio- static functions of AgNPs could be changed in the bacte- rial environment. For example, oxidation processes can induce the changes of nanoparticles in oxygen-containing liquids, which distinguishes AgNPs from the potentially stable gold nanoparticles. However, the synthesis of AgNPs is much cheaper and more affordable, so it is very important to find new ways to prevent any loss of basic silver characteristics in solutions. For example, the po- tential instability of AgNPs could be improved by stabili- zation with organic, inorganic, synthetic, natural, biotic, and abiotic substrates [15]. Silver nanoparticles are zero-dimensional materials with particle sizes ranging from 1 to 100 nm. Currently, special synthetic methods have been developed to obtain particles with certain shapes and sizes. For example, chemical (reduction of ions to atoms), physical (mechani- cal and pear-shaped) and biological (extracts of microor- ganisms and plants) methods of production are actively used (Figure 1). The most common approach is chemical synthesis; however, this method is characterized by high toxicity and pollution. Biological methods are cheaper and more ecologically friendly. Moreover, the substrates ob- tained during biological synthesis can be used as stabi- lizers for the nanoparticles [3]. 2. Materials and methods 2.1. Data sources A literature review on the properties of silver nanoparti- cles was performed using the following databases: MEDLINE (PubMed interface), SCOPUS, Cochrane Library, Google Scholar. The published data on the in vitro and in vivo studies were accessed between January 1990 and De- cember 2021. Backward and forward reference searching was applied to find the most relevant articles. 2.2. Inclusion and exclusion criteria The current review includes predominantly full-text arti- cles presented in English language and focused on silver nanoparticles chemical/physical/biological properties. The selected articles were original studies (observational and experimental), systematic reviews, narrative reviews and meta-analyses. The studies were excluded if they were not consistent with the research objectives and the actual purposes of the current review. All studies were carried out in vitro (cell lines) or on mice/rats due to ethic issues. 2.3. Search strategy Search terms included the following: silver nanoparticles, AgNP, physical, chemical, biological, biocidal, properties, effects, toxicity, microorganism, virus, bacteria, fungi, protozoa, parasites, cancer and tumor. The main search structures consisted of keywords according to each objec- tive of the review: ((silver nanoparticle*) OR AgNP*) AND synth*; ((silver nanoparticle*) OR AgNP*) AND (chemic* OR physic* OR bio*); ((silver nanoparticle*) OR AgNP*) AND microorg*; ((silver nanoparticle*) OR AgNP*) AND propert*; ((silver nanoparticle*) OR AgNP*) AND (anti- bacter* OR bacter*); ((silver nanoparticle*) OR AgNP*) AND antivirus*; ((silver nanoparticle*) OR AgNP*) AND (protoz* OR parasite*); ((silver nanoparticle*) OR AgNP*) AND (anticancer* OR tumor*); ((silver nanoparticle*) OR AgNP*) AND toxic*. Grey literature and unpublished in- formation were neither considered nor used. After refer- ence screening the duplicates were excluded. The key research objectives were: 1. Studying different chemical and physical properties of AgNPs, which can change biological effects. 2. Uncovering some important biological and bio- chemical mechanisms of AgNPs effects and toxicity against bacteria, viruses, fungi, protozoa, and cancer. Figure 1 Characteristics of silver nanoparticles. Different ways of AgNPs synthesis. Adapted from refs [17, 18]. Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 3 of 20 3. Elucidating potential harm and toxicity of AgNPs for a macroorganism and a host. The current review was prepared according to a scale for the quality assessment of narrative review articles – SANRA [25]. 3. Results and discussion 3.1. Physical and chemical properties of silver nano- particles 3.1.1. Size The antibacterial activity of silver nanoparticles is closely related to their size. Agnihotri et al. have investigated an- tibacterial activity of silver nanoparticles of different size (5, 7, 10, 15, 20, 30, 50, 63, 85, and 100 nm) against Esch- erichia coli MTCC 443 (E. сoli) and Staphylococcus aureus NCIM 5201. Antibacterial activity of AgNPs was enhanced with decreasing size. The smallest nanoparticles of 5 nm demonstrated the best results and the highest bactericidal activity against all tested strains [26]. Baker et al. studied the antibacterial activity of nanoparticles with different sizes ranging from 5 to 70 nm (average size 15 nm) and from 50 nm to more than 100 nm (average size 75 nm), respectively. The results showed that smaller AgNPs (15 nm) were more effective against E. coli [27]. Conse- quently, the smaller size results in the higher antibacterial activity. This rule is explained by the fact that small AgNPs have larger specific surface area and more surface-acting centers, resulting in the increase in Ag+ release rate and, therefore, toxicity [28, 29]. Dobias et al. demonstrated that in natural ponds, the dissolution of 5 nm AgNPS oc- curred faster than that of 10 nm silver nanoparticles. Smaller nanoparticles released more silver ions than 50 nm AgNPs [30]. Morones et al. showed that only nanopar- ticles with a diameter of 10 nm or less could penetrate through the bacterial cell membrane [31]. In addition, ap- plying the nanoparticles with the size of 8.3±1.9 nm in- creases the damage of cellular DNA by influencing the nu- cleotide excision repair [32]. Conversely, Bélteky et al. noticed that in spite of the fact that nanoparticles with the smallest diameter have the greatest toxicity, an increase in the size of the nanoparticles promotes colloidal stability and also provides greater resistance to environmental conditions and aggregation [33]. 3.1.2. Shape Another property that determines the antibacterial activi- ty of silver nanoparticles is their shape. There are quasi- spheres, nanotubes, rods, disks, cubes, prisms, octahedral, and triangular nanoplates [34–37] (Figure 2). Pal et al. investigated the antibacterial activity of spherical, rod- shaped, and triangular silver nanoparticles against E. coli. Truncated triangular nanoplates demonstrated the highest bactericidal activity compared to nanospheres and nano- rods. The result could be explained by the presence of spe- cial active faces that determine their high reactivity. This triangular structure of nanoplates promotes more effec- tive interaction with the bacterial cell, leading to its lysis [38]. On the contrary, in one of the latest studies, triangu- lar nanoplates showed a smaller antibacterial effect com- pared to nanospheres against E. coli, S. aureus, P. aeru- ginosa. This can be put down to the fact that the surface area of the nanospheres (1.307±5 cm2) exceeded the sur- face area of triangular nanoplates (1.028±35 cm2) [39]. In the other studies, the greatest bactericidal activity was also demonstrated by nanoparticles with the largest sur- face area, which caused the accelerated formation of ions on the nanoparticle surface during their dissolution, there- fore, increasing the antibacterial effect [40, 41]. Figure 2 Different shapes of silver nanoparticles. Adapted from ref [42]. 3.1.3. Concentration of nanoparticles The dissolution of silver nanoparticles depends on their initial concentration. If this concentration is lower than the solubility of AgNPs (which depends on the size of the nanoparticles, the presence of ligands forming a complex with silver (I) and the physicochemical properties of the solution), all nanoparticles will eventually dissolve. Ag- gregation also explains why the initial concentration of AgNPs affects the release kinetics of silver ions. For exam- ple, if other parameters are fixed, the higher initial con- centration of nanoparticles results in the slower initial release of silver ions. [43]. This is explained by the fact that at the high initial concentration AgNPs tend to aggre- gate more rapidly, which decreases the soluble AgNPs sur- face area [44]. The initial concentration of AgNPs ranging from 300 to 600 μg/L increases the aggregation rate for all three sizes of AgNPs (20, 40 and 80 nm) [45]. The an- tibacterial activity of AgNPs is also determined by the con- centration of nanoparticles. Panáček et al. found that AgNPs are able to exhibit antibacterial activity at a very low concentration of 1.69 μg/ml [46]. Shu et al. demon- strated that the antibacterial activity of AgNPs of 13.8 nm against E. coli depended on their concentration. The growth inhibition analysis illustrated a complete suppres- sion of the E. coli growth at AgNPs concentrations above Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 4 of 20 20.0 μg/ml [47]. Ugwoke et al. found that bactericidal activity enhanced as the dose of silver nanoparticles in- creased from 2 to 8 μg/ml. The smallest AgNPs (3.4±1.2 nm) demonstrated high sensitivity to bacterial strains (Coliform) at lower concentrations (8 μg/ml), compared with 30 μg of gentamicin [48]. In addition, the minimal concentration of AgNPs for antibacterial proper- ties is mainly determined by their shape. Thus, truncated triangular silver nanoparticles showed the inhibition of bacterial growth at the 1 μg concentration, while nano- spheres and nanorods induce an inhibiting effect at con- centrations of 12.5 μg and 50-100 μg, respectively [31, 38]. But, the shape cannot be considered as a single factor that affects antibacterial activity, because the particle size var- ies with the form, which influences the overall rate of par- ticles dissolving [34]. 3.1.4. Stabilizing agents As a rule, stabilizing agents are used during AgNPs syn- thesis to provide an electrostatic repulsion between indi- vidual particles and to prevent their aggregation [43]. As opposed to other silver forms, nanoparticles exhibit high surface energy values due to their small size and, there- fore, they are more likely to clump and to form agglomer- ates. Choosing a suitable stabilizing agent is an important requirement for the nanoparticles stabilization, because coating agent affects the structural properties of the na- noparticle, including its size, shape, surface charge, and interaction with environment [49]. It is worth noting that coatings probably detach from the surface after interact- ing with environment [50]. Among stabilizing mechanisms of coating agents, there are electrostatic stabilization, ste- ric stabilization, and stabilization by hydration forces, depletion stabilization and stabilization by the Van der Waals forces [51]. Organic coating agents are widely used as stabilizers for silver nanoparticles. In some cases, the stabilizing agent acts simultaneously as a reducing agent of Ag+ ion to Ag0 [52]. Also, organic molecules contribute to the complexation of silver ions, thereby accelerating their dissolution [53]. The most common coating agents for silver nanoparti- cles are citrate, polyvinyl alcohol (PVA), sodium dodecyl sulfate (SDS), polyvinylpyrrolidone (PVP), Tween 80 [54– 58]. Citrate is one of the most commonly used stabilizers and reducing agents used for the AgNPs synthesis. Citrate- coated particles are electrostatically stabilized by nega- tively charged anions. However, as the pH value decreas- es, the citrate-anion protonates, which causes the loss of stabilization [59]. In vivo AgNPs capped by citrate or PVP demonstrate the greater antibacterial activity against Salmonella, compared to uncapped AgNPs, which could be explained by minimal interaction with serum proteins. Uncapped AgNPs, by contrast, lose their antibacterial ac- tivity due to interaction with bovine serum albumin (BSA) [58]. Kvítek et al. proved that any of three mentioned sta- bilizing agents (Twin-80, sodium dodecyl sulfate (SDS) or polyvinylpyrrolidone (PVP)) increased the antibacterial activity and excellent stabilization of silver nanoparticles dispersion against aggregation. Among all AgNPs ligands, SDS-modified AgNPs proved to be the most stable due to the electrostatic repulsion and steric effect. SDS-modified AgNPs demonstrated the highest antibacterial activity as- sociated with good silver nanoparticles dispersibility and effective interaction with the cell membrane [60]. Ajitha et al. found that PVA-coated AgNPs had the smallest size and demonstrated high stability and antibacterial activity, compared to nanoparticles stabilized by other coatings [49]. Currently, the biosynthesis of metals and metal oxide nanoparticles, using biological agents such as bacteria, fungi, yeast, plant and algae extracts, has gained popu- larity in the field of nanotechnology [61]. Thus, AgNPs, which are synthesized by various microorganisms, pro- vide high stability due to the fact that microbes produce large amounts of protein [62]. AgNPs, which are synthe- sized using cyanobacterial extract of Oscillatoria lim- netic, have high antibacterial activity against multidrug- resistant bacteria (Escherichia coli and Bacillus cereus) as well as cytotoxicity against breast cancer and colon cancer cells at low concentrations of 6.147 μg/ ml and 5.369 μg/ml, respectively [63]. Plants contain carbohy- drates, fats, proteins, nucleic acids, pigments and several types of secondary metabolites that act as stabilizers and reducing agents in the biosynthesis of silver nanoparti- cles [64]. Caffeine and theophylline are widely used as stabilizing agents and could be found in water-alcohol extracts of Coffea arabica and Camellia sinensis as well as in extracts of black tea [65, 66]. Utilization of fungi as reducing and stabilizing agents in the biogenic synthesis of silver nanoparticles is also attractive because they produce large amounts of protein. During biological syn- thesis, nanoparticles are coated with biomolecules de- rived from the fungus, which leads to improved stability and increased biological activity [17, 67]. Konappa et al. capped silver nanoparticles with secondary metabolites secreted by the T. Harzianum fungus. The obtained AgNPs had high stability as well as a wide range of anti- bacterial activity against two gram-positive bacteria (S. aureus and B. subtilis) and two gram-negative bacteria (E. coli and R. solanacearum) [68]. Silver nanoparticles also could be stabilized with pol- ymeric carbohydrates such as starch, sodium alginate and chitosan [69–71]. Muhammad et al. used sericin (a protein that is a part of silk) as a stabilizing agent. Sericin-coated nanoparticles proved to be highly effective against bacte- ria and maintained stability over a wide range of tempera- tures and pH concentrations. The authors suggested wide use of sericin in the future because of low cost and high stability [72]. Hydroxyl groups of sericin form complexes with silver ions, thus preventing their aggregation and deposition [71]. Azócar et al. used diclofenac (d) and ke- torolac (k) as stabilizing agents, which are widely used as anti-inflammatory drugs in medicine. The results demon- Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 5 of 20 strated that AgNPs-k were more stable than the uncoated nanoparticles. Under the influence of UV light (wavelength of 365 nm), capped nanoparticles generated anion radi- cals. This effect is probably associated with capping agents, because bare nanoparticles do not promote the formation of superoxide anion [73]. 3.1.5. Surface charge of the nanoparticles Gao et al. found that dispersion and stability of AgNPs are related to their surface charge. The negative surface charge contributes to the electrostatic stabilization of the nanoparticles against aggregation. However, the disper- sion charge of particle can change depending on the pH value. The surface charge of AgNPs becomes more nega- tive at higher pH, as confirmed by the high zeta potential value equal to –32.5 mV, which promotes the stability of the suspension. Conversely, lower pH values of 5 and 3 are characterized by a decrease in zeta potential to –22.5 and –18.2 mV, which, therefore, reduces the repulsive forces and stability [59]. Moreover, small AgNPs have lower zeta potentials than large AgNPs; thus, small particles have less electrostatic repulsion and more rapid aggregation. Surface charge is one of the most important factors of AgNPs toxicity. Recently, the antibacterial activity of posi- tively and negatively charged AgNPs has been studied. Some studies demonstrated that positively charged AgNPs have a higher bactericidal activity against all microorgan- isms compared with negatively or neutral charged AgNPs [74, 75]. El Badawy et al. found out that positively charged BPEI-AgNP were more toxic against bacteria, compared with negatively charged citrate-AgNP. It could be ex- plained by negative membrane charge of both gram- positive and gram-negative bacteria. Consequently, there is an electrostatic barrier between negatively charged cit- rate-AgNP and bacterial membranes that limits interaction between the cell and nanoparticles, thus reducing toxicity [76, 77]. Qiao et al. synthesized zwitterion-modified AgNPs, which could change the charge depending on the environmental pH. These AgNPs demonstrated a pH- dependent transformation of negative charge into positive charge. Therefore AgNPs were innocuous for healthy tis- sue cells (pH=7.4), while interacted effectively with nega- tively charged bacterial surfaces in foci of infection (pH=5.5) [78]. 3.2. Influence of biological conditions on the nanopar- ticles properties Such factors as pH, presence of dissolved oxygen, electro- lytes and organic substances, in particular, proteins affect significantly the physicochemical properties and antibac- terial activity of silver nanoparticles [79, 80]. 3.2.1. Oxygen availability The dissolution of the nanoparticles occurs in the presence of dissolved oxygen. The surface of AgNPs is easily oxi- dized by O2 and other molecules in ecological and biologi- cal systems, resulting in the release of Ag+ and defining their toxicity [81]. However, silver nanoparticles do not dissolve completely in the presence of molecular oxygen. A complete dissolution requires a stronger oxidizer such as H2O2. The AgNPs aggregation in oxygenated water is 3–8 times faster than under anaerobic conditions, indicat- ing that dissolved molecular oxygen can also significantly affect this process [45]. 3.2.2. Interaction with proteins It is well known that nanomaterials can interact with various biomolecules of living organisms, primarily with proteins that are able to adsorb on the nanoparticles sur- face, forming the biomolecular corona [82]. About 300– 500 human plasma proteins could be bound to different nanoparticles. Nanomaterials are rapidly coated by pro- teins in physiological fluids. The formation of the protein corona leads to the change in the physicochemical prop- erties of nanoparticles, including hydrodynamic size, surface charge, and aggregation [84]. The structure of protein corona depends primarily on the nanoparticle material, size and surface properties, as well as on the composition of protein environment and experi- mental/physiological conditions [84–86]. Tai et al. and Alarcon et al. demonstrated that the protein corona serves as a kind of biological identity of the nanoparticle. In addition, it contributes to the colloidal stability of the particle, that presents the aggregation of the nanoparti- cles and protective from aggressive environmental condi- tions [87, 88]. Functionalization of silver nanoparticles surface significantly affects the formation dynamics of protein corona. The presence of a protein coating on the surface of the nanoparticles strongly reduces the binding degree to the protein. On the contrary, the formation of the corona on the uncoated nanoparticles improved sig- nificantly their stability in biological environment. The in vitro experiments showed that the physiological sta- bility of AgNps caused by the corona formation may be directly associated with their binding to the cell, capture and toxicity [89]. The interaction between nanoparticles and different cells is determined by the composition of protein corona [90]. 3.2.3. pH The solution pH affects significantly the surface charge and oxidative dissolution of AgNPs. The behavior of nano- particles differs under acidic and alkaline conditions. AgNPs are found to destabilize in acid and neutral pH, which results in higher aggregation rate. Under alkaline conditions, negatively charged hydroxyl ions promote the stabilization of nanoparticles [91]. Bélteky et al. found that AgNPs are more stable at alkaline and neutral values of pH than under acid conditions. An increase in the pH level leads to higher deprotonation degree of free organic functional groups and increasing negative charge on the nanoparticles surface. It facilitates the increasing electro- static repulsion between particles and reduces the degree of aggregation [82]. Sivera et al. showed that gelatin- Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 6 of 20 stabilized AgNPs have robust resistance to aggregation in a wide range of pH (2 to 13). Moreover, these AgNPs demonstrated long-term stability against aggregation and maintained high antibacterial activity under environmen- tal conditions for several months [92]. 3.2.4. Electrolyte concentration An increase in the electrolyte concentration leads to an increase in the AgNPs aggregation rate [43]. Stebounova et al. concluded that AgNPs aggregate in solutions with high ionic strength regardless of stabilization [79]. Bé- lteky et al. found that in solutions containing 50 mm of NaCl, the aggregation is slow and the size of AgNPs ag- glomerates does not change significantly. However, the addition of 150 mm of NaCl induces the rapid aggrega- tion of AgNPs up to micrometer size range. Sudden changes in aggregation occur due to an increase in the concentration of Na+, because in large quantities these ions can shield negatively charged surface groups, which provide electrostatic stabilization. With reduced repul- sion, the particles form larger aggregates during colli- sions [82]. The presence of chloride ions in solution causes silver chloride deposition [53]. However, sulfide ligands significantly reduce the toxicity of AgNPs caused by generation of insoluble silver compounds [93]. The nanoparticle aggregation is very prominent in solutions with high concentration of divalent cations, such as Mg 2+ and Ca2+, due to the stronger neutralization of the sur- face charge. On the other hand, monovalent cations (K+ and Na+) can also enhance the aggregation of AgNPs, but they are much less effective in the shielding of surface charge than divalent ones [43]. Finally, the effect of the ionic strength on the AgNPs aggregation is more signifi- cant for smaller particles [93]. 3.3. Mechanisms of AgNPs biological activity 3.3.1. The main mechanisms of antibacterial activity The main antibacterial properties of silver nanoparticles are provided directly by silver ions; in other words, nano- particles are a kind of transporters of the main active sub- stance [97]. Moreover, AgNPs obtained by the "green" syn- thesis are more toxic than AgNPs obtained by non- biological approaches [64]. AgNPs release silver ions that interact with sulfa groups of membrane proteins, which disturb the mem- brane integrity and thereby increase its permeability. AgNPs interact with the cytoskeletal protein MREB, which plays an important role in the survival and formation of the bacterial cell [98]. Active adhesion of silver ions to the cell membrane leads to the change in its charge (depolari- zation and desensitization), lysis of cellular components and rupture of organelles. Moreover, silver ions partici- pate in transduction processes by dephosphorylation of tyrosine residues, initiating the launch of the bacterial cell apoptosis program [13, 99]. Once in the cell, free silver ions interact with respira- tory chain enzymes (dehydrogenases) and bind to func- tional electron donor groups (thiols, phosphates, imidaz- ole, indoles, and hydroxyl groups), disrupting the ATP syn- thesis and K+ transport [14, 98, 100]. In addition, the pathological effect of silver nanopar- ticles is induced by the formation of reactive oxygen spe- cies (ROS): superoxide anion radical (O2•−), peroxide (O2•−2), hydroxyl radical (•OH), hydrogen peroxide (H2O2), hydroxyl ions (OH−). Different ROS have various pathological effects, for instance: OH−, H2O2 and O2•− have the greatest antibacterial activity. OH− interacts with positively charged cell membranes, while H2O2 has the greatest penetrating effect. High concentration of ROS leads to a decrease in the concentration of glutathi- one, an increase in lactate dehydrogenase, dysregulation of calcium channels, matrix metalloproteases and intra- cellular redox homeostasis [101]. Actually, ROS interacts with the thioredoxin system of S. aureus, which is one of the most important disulfide reductase systems that counteract the processes of oxidative stress. Oligomeri- zation and dysregulation of thiol-redox homeostasis due to the depletion of intracellular thiol leads to disruption of the protective components and to activation of the oxidative stress [102]. Moreover, ROS interact with DNA, thereby triggering the processes of its modification. In addition to the indi- rect effect through oxidative stress, silver ions interact with sulfur and phosphorus groups of DNAs, which leads to the rupture of hydrogen bonds between the chains; the processes of replication and reproduction are disrupted (i.e., genome splitting is observed) [103]. Unipolar charge of DNA and AgNPs leads to additional destabilization of the chains. Damage to DNA molecules occurs due to oxida- tion and alkylation of its bases, which leads to the for- mation of various compounds: 8-oxoguanine, 7,8-dihydro- 8-oxoguanine, 8-oxoadenine, unsubstituted and substitut- ed with an imidazole ring of purines. These compounds are integrated into DNA under the influence of hydroxyl radicals [103, 104]. In addition to DNA, silver ions also interact with RNA and block the subunits of ribosomes (30S), which are nec- essary for binding tRNA [105, 106]. Being in cytoplasm AgNPs induce the processes of ribosome denaturation, inhibition of translation, protein synthesis and carbohy- drate metabolism [102, 107]. The disturbance of the signal transduction processes is equally important because suppressing the phosphoryla- tion of tyrosine residues results in blocking the cell cycle, the synthesis of exopolysaccharides and capsular polysac- charides, which ultimately leads to the interruption of the bacterial cell division [103]. Thus, the antibacterial activity of silver has a complex effect on the microorganism, disrupting various aspects of its vital activity. Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 7 of 20 Gram-negative bacteria are more sensitive to the ef- fects of silver ions than Gram-positive bacteria, which is due to a thinner layer of peptidoglycans in the cell wall [108]. For example, S. aureus, a Gram-positive coccus with a cell wall width of 30 nm, can effectively prevent the in- ward penetration of nanoparticles due to the high affinity of the peptidoglycan layer [109, 110]. The antibacterial effect may depend not only on the size of the nanoparticles (nanoparticles smaller than 10 nm have high penetrating power), but also on the physico- chemical properties, in particular, surface characteristics. So, positively charged nanoparticles have a greater bind- ing ability with a negatively charged cell membrane due to the electrostatic interaction, in contrast to the negatively charged AgNPs. The higher positive charge of the ions re- sults in the lower severity of the electrostatic barrier [97, 108, 109, 111]. Despite the fact that silver ions play a crucial role in disrupting the basic processes of microorganisms’ activity, nanoparticles also have an antibacterial effect due to the mechanism of the "contact destruction". The antibacterial properties of silver nanoparticles also depend on their shape. For example, spherical and trian- gular AgNPs have greater activity against E. coli and S. aureus is than AgNPs of irregular shape, which may be due to a larger surface and increased release of silver ions [5, 13]. However, Pal et al. elucidated that the triangular shape’s AgNPs have a greater antibacterial effectiveness against E. coli, compared to spherical and rod-shaped par- ticles [38]. Moreover E.coli is more sensitive to AgNP ex- posure than S. aureus regardless of AgNP size and surface properties [112]. There are published data indicating the ability of silver nanoparticles to suppress the formation of bacterial films on medical instruments without significant accumulation of silver ions in surrounding organs and tissues. The most promising direction is the approbation of this coating for catheters, drains and medical masks [5]. Moreover, numerous studies pinpoint the anti- inflammatory properties of AgNPs, which are associated with a decrease in the pro-inflammatory cytokines synthe- sis (TNF, IL-12, IL-1, NF-kb) and the induction of apoptosis. In addition, the modulation role of silver in the process of wound and periodontal healing has been discovered [104]. Thus, the main antibacterial mechanisms of AgNPs (Figure 3) are as follows: 1. Interaction with the membrane, impairment of its permeability and change in charge. 2. Disturbance of intracellular processes and organelles dysfunction. 3. Inhibition of mitochondrial processes (respiratory chain malfunction), activation of oxidative stress, synthe- sis of reactive oxygen species and lipid peroxidation. 4. Interaction with DNA and RNA, blocking the replica- tion, transcription, and translation. 5. Inhibition of transduction of signals due to dephosphorylation of tyrosine residues. Figure 3 Antibacterial properties of AgNPs. General mechanisms of antimicrobial mode of action of silver nanoparticles (a); Bacte- ricidal effect of green synthesized AgNPs on different bacterial strains. Dose-dependent activity of AgNPs synthesized using Allo- phylus cobbe leaves. The bacterial strains were incubated at vari- ous AgNPs concentrations ranging from 0.1 to 1.0 mg/ml and bac- terial rate survival was estimated by colony forming unit (CFU) assay at 4 h (b); Zone of inhibition of silver nanoparticles against various pathogenic microorganisms compared to banana peel extract (BPE) (c). Presented from refs [61, 113]. (a) (b) (c) Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 8 of 20 3.3.2. The main mechanisms of antiviral activity In vitro AgNPs could adhere to the virus surface, bind the viral ligand and therefore prevent its spreading in cell cultures (Figure 4a, 4b). Lv et al. indicate the property of silver nanoparticles to inhibit TGEV (transmissible gastro- enteritis virus in pigs) induced apoptosis by binding to S glycoprotein. The suppression of the proapoptotic pathway is one of the possible mechanisms because TGEV causes an increase in concentration of protein BAX [114]. Based on the published data, there is assumption that the nanopar- ticles can also be effective against the novel coronavirus infection (COVID-19) because they have a possibility to interact with the spike glycoprotein and to decrease pH of respiratory epithelium, which would effectively prevent intracellular invasion of the virus (Figure 4c) [98, 115]. There is a description of AgNPs possibility to bind the G- protein of the respiratory syncytial virus in the hep-2 cell culture. AgNPs coated with polyvinylpyrrolidone inhibited the reproduction of RSC virus by 44%, while silver nano- particles coated with biomolecules and RF-112 did not have a visible effect on the pathogen's life cycle [104, 116]. In addition to the binding to surface proteins, AgNPs interact with the viruses’ nucleic acids (for instance, of the hepatitis B virus) and disrupt their replication in the host cells [103]. The inhibition of HBV RNA/DNA synthesis and creation of extracellular virions are observed in the hu- man hepatoma HepAD38 cell line [117]. Biologically synthesized AgNPs of small size (<20 nm) have greater effectiveness against herpes simplex virus type 1/2 and human parainfluenza virus type 3 due to a more pronounced blocking effect on the interaction be- tween the virus and a cell of the Veroline cell line. Nano- particles are able to interact non-covalently with the thy- midine kinase ligand, thereby suppressing the activity of herpesviruses. In addition to inhibition of HPV 1 and type 2, there is an inhibition of oncogenic herpesviruses, for example, Epstein-Barr virus [13, 120]. Moreover, AgNPs block the interaction of herpesviruses with heparan sul- fate proteoglycans of cell membranes, preventing invasion processes. These properties could be enhanced by combin- ing AgNPs with tannic acid [121, 122]. Particles smaller than 10 nm can prevent the spreading of influenza virus in cell culture MDCK [123]. In 2017, Lin et al. published the data about the activity of combination treatment with Zanamivir and AgNPs against the H1A1 influenza virus in MDCK cell line. This combination not only demonstrated high thermodynamic and kinetic stabil- ity, but also suppressed effectively the replication of the influenza virus by regulating neuraminidase activity [124]. There are published data about the ability of silver na- noparticles to interact with HIV and inhibit significantly its reproduction by binding to the regions of disulfide bond (sulfur-containing residues of CD4-binding domain) of gp120 [125, 126]. Moreover, silver nanoparticles could prevent infection of cervical tissue with HIV type 1 with- out cytotoxic effect in vitro [127]. Figure 4 Antiviral properties of AgNPs. Schematic model of a virus infecting an eukaryotic cell and antiviral mechanism of metal nanoparticles. (a); The activity of silver nanoparticles against the prevalent viruses (b); Mechanisms of antiviral activity against SARS-CoV2 (c). Adapted from refs [118, 119]. Silver nanoparticles have a wide antiviral spectrum of action against different viruses that spread through mos- quito bites: dengue fever, West Nile fever, Zika virus and Chikungunya virus. Both uncoated AgNPs and polysaccha- Influenza inhibition of new virions formation and their spreading Hepatitis B virus inhibition of new virions formation and their spreading Herpes virus Inhibition of replication and invasion processes in a healthy cell HIV Binding to gp120 and preventing virus from entering the cell. Inhibition of virus reproduction Respiratory syncytial virus Binding to G-protein, inhibition of virus reproduction (a) ))k k)) (b) (c) Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 9 of 20 rides- coated AgNPs have a suppressing effect on the repli- cation of Tacaribe virus and Monkeypox virus [13, 128, 129]. 3.3.3. The main mechanisms of antifungal activity The mechanisms of AgNPs antifungal activity are not fully understood. There is a suggestion that the interaction of nanoparticles with the membrane leads to disruption of its function (the current of transmembrane ions, including protons) and division processes (especially in yeast). Also, AgNPs induce an inhibition of germ tube formation, growth of biofilm and secretion of hydrolytic enzymes [130, 131]. Beyond the membranotoxic effect, silver nano- particles could initiate a cascade of intracellular patholog- ical processes leading to the fungus death: oxidative stress, interaction with thiol groups and phosphorus- containing molecules, blocking protein synthesis. Thus, the fundamental antifungal mechanisms are similar to antibacterial ones (Figure 5a) [132]. The majority of the published data was acquired by studying the activity of AgNPs against plant pathogens and mold fungi [133]. However, AgNPs have antifungal proper- ties against pathogenic fungi that cause dermatophytosis: Candida sp. (including Candida albicans) and Tri- chophyton mentagrophytes [134, 135]. Even amphotericin- B resistant strains of Candida glabrate were found to be sensitive to nanoparticles [136]. Silver nanoparticles also have a biocidal effect on biofilms that are formed by Can- dida spp. [137]. There are data on the inhibition effect of particles on fungal keratitis pathogens (Fusarium spp., Aspergillus spp., Alternaria alternate) in vitro. At the same time, AgNPs had a potentially greater antifungal effect than an- timycotic drug – natamycin [140]. AgNPs can enhance the effect of antifungal drugs. There are data about synergy of silver nanoparticles and ketoconazole against the main cause of seborrheic derma- titis - Malassezia furfur. Combination therapy leads to a decrease in the frequency of drug use and the frequency of relapses not only of seborrheic dermatitis, but also of oth- er malasesiosis [141]. Also, AgNPs suppress the growth of different pathogenic Aspergillus species (Aspergillus ni- ger, Aspergillus flavus, Aspergillus fumigatus), which can cause a wide range of diseases. Due to this effect, silver nanoparticles can play an important role in the treatment of aspergillosis, especially in patients with drug-resistant strains [142]. It is worth mentioning that Aspergillus niger can be used to synthesize AgNPs (one of the directions of biological synthesis). The synthesized nanoparticles have the ability to inhibit the growth and development of Allo- vahlkampfia spelaea, which causes resistant keratitis [143]. Thus, silver nanoparticles can play an important role in the treatment of fungal infections, especially due to the scarcity of the antifungal drugs and increasing number of drug-resistant species (Figure 5b). The combination of nanoparticles and drugs can boost the effect of the latter and have an independent bactericidal action in the case of multiple resistance of pathogenic fungi. AgNPs can also be used to prevent the spreading of mold on different surfac- es [133]. However, the inhibition of fungal growth by sil- ver nanoparticles is less prominent than that of bacteria, which may be due to the presence of chitin in the wall of the fungus, rather than peptidoglycans [142]. Figure 5 Antifungal activity of silver nanoparticles. Schematic repre- sentation for the proposed model for mechanism of action of AgNPs against Candida cells, depicting possible cellular targets and existence of ROS-dependent and ROS-independent pathways for fungicidal action of AgNPs (a); Antifungal activity of silver nanoparticles against: A.niger (1) and P.chrysogenum (2) at the concentrations of 200, 300, 400 and 500 µg/ml(b); Colonies of Trichophyton tonsurans (1), Microsporum gypseum (2) and Trichophyton mentagrophytes (3). On PDA and PDA supplemented with different concentrations of AgNP. The bar charts represent the diameter of the colonies in the function of AgNP concentration (b). Reproduced from [138, 139]. 3.3.4. Mechanisms of antiparasitic activity There are reliable data confirming the antiparasitic prop- erties of silver nanoparticles, in particular, against the causative agent of cutaneous leishmaniasis (Leishmania tropica) (Figure 6a). AgNPs bind to the sulfo- and phos- phorus-containing membrane and DNA proteins, blocking DNA synthesis and activation of oxidative stress. AgNPs have an anti-promastigote effect because of blocking the proliferation of promastigotes. In addition, AgNPs sup- (a) (c) (b) Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 10 of 20 press the vital activity of amastigotes and reduce their survival in infected host cells. Anti-amastigote properties are boosted by additional exposure of ultraviolet light. Probably, when exposed to ultraviolet light, there is an increase in the concentration of monosulfide radicals, which are formed from complexes. These complexes are formed by interaction of silver ions and cysteine groups of parasitic proteins [144, 145]. Figure 6 Antiparasitic properties of AgNP. AgNPs leishmanicidal activity. L. amazonensis promastigotes forms were subjected to different concentrations of AgNPs-bio (0.125, 0.25 and 0.50 μg/mL) and the parasite viability was assessed at 0, 24, 48 and 72 h (a); Effect of AgNPs in Toxoplasma gondii morphology as as- sessed by fluorescence microscopy. In the control sample, intact oocyst, in the presence AgNPs at 45 min and 60 min (b); Scanning electron microscopy (SEM) micrograph of protoscolices of E. granulosus. Protoscolices without nanoparticles and 1 mg/mL concentrations of Ag-NPs that covered protoscolex (c). Repro- duced from [146–148]. Apart from Leishmania, the antibacterial activity of silver nanoparticlesis was observed against oocysts Enta- moeba histolytica, Cryptosporidium parvum and several other protozoa [131, 149]. For example, in the study of Costa et al. biogenic AgNPs inhibited the replication of Toxoplasma gondi (which causes toxoplasmosis) in cell cultures, such as BeWo, HTR-8/svneo, Hela and in villous explants. Moreover, the nanoparticles induced secretion of inflammatory cytokines in cells, for example, in the BeWo line: IL-4 and IL-10; in the HTR-8/SVneo line: IL-4 and the macrophage migration inhibitory factor (MIF). Toxoplas- ma gondi increased the MIF concentration in the BeWo cell culture and IL-6 in HTR-8/SVneo line. In villous explants the synthesis of IL-4, IL-6 and IL-8 decreased after infest- ing. In Hela cell line an increase in the NO concentration, oxidative stress and reduction of pro-inflammatory cyto- kines, in particular, IL-8 were observed. Thus, silver na- noparticles can significantly inhibit the spread of T. Gondi without developing dysfunction of the host cells (Figure 6b) [150, 151]. In addition to the cell cultures and chorion- ic villi, AgNPs suppresses toxoplasma’s replication in liver and spleen tissues [152]. One of the possible antiparasitic mechanisms includes the suppression of mitochondrial function, disturbing mitochondrial membrane potential, redox signaling and destruction of leucine aminopeptidase (LAP) [153]. The current data plays an important role in the devel- opment of alternative approaches to the treatment of tox- oplasmosis, particularly in pregnancy, because standard drugs used for the treatment of this pathology have tera- togenic and myelosuppressive properties. Moreover, toxo- plasmosis is part of the TORCH complex, which includes a group of intrauterine infections that lead to impaired fetal development and even death. The results of Younis et al. demonstrated the effective- ness of AgNPs against Blastocystis hominis, which is the causative agent of blastomycosis. The most prominent an- tiparasitic effect in vitro was observed with a combination of particles and metronidazole. The concentration of B. Hominis decreased by 71.69% in the metronidazole group, by 79.67% in the AgNPs group and by 62.65% in the com- bination therapy group (AgNPs + metronidazole) after 3 hours (p<0.05). The nanoparticles are likely to interact with and modify glycoprotein and lipophosphoglycan mole- cules on the parasite surface they may induce oxidative stress, and inhibit ROS synthesis and DNA replication [154]. There are data on the possible sporicidal action of na- noparticles against Echinococcus (Figure 6c). Moreover, AgNPs have a synergistic effect with albendazole and are able to prevent the development of adverse reactions in the liver associated with this drug. For example, they de- crease the severity of necrosis, steatosis, and reduce the level of transaminase and IFN-γ. Combination therapy is associated with a greater degree of structural changes in echinococcal cysts (reduction of cyst size and cyst mass) [155, 156]. Additionally, silver nanoparticles are used for the devel- opment of new directions in the treatment of giardiasis (Gi- ardia lamblia). The combination with chitosan and curcu- min leads to the complete eradication of giardia in the in- testine and feces of rodents without the development of adverse reactions [157]. The study of tropical malaria (Plasmodium falciparum) using AgNPs for the treatment is ongoing [158]. Moreover, silver nanoparticles could be used in ophthalmology for prevention of Acanthamoeba adhesion on contact lenses (amoebic keratitis prophylaxis) [159]. 3.3.5. Main mechanisms of anticancer activity Silver nanoparticles are promising for developing and modifying approaches to antitumor therapy. AgNPs can have a cytotoxic effect on tumor cells with subsequent suppression of the pathological process. The decreased lymphatic outflow in malignant tumors allows nanoparti- cles to accumulate and act longer [160]. Moreover, tumor cells absorb AgNPs (by endocytosis) to greater than nor- mal cells [2]. Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 11 of 20 There are main mechanisms of anticancer activity: in- duction of oxidative stress, changing the structural- cellular morphology and activation of pro-apoptotic pro- cesses (caspase 3 and 9, regulation of p53, p38 MAPK, HIF-1α, increase in BAX concentration and decrease in Bcl- 2 concentration) [161]. Apart from apoptosis, there are also necrosis and autophagy in cancer cells are activated by stimulation of autophagosomes formation through the PtdIns3K signaling pathway. Except direct pro-apoptotic effect, there is also an indirect activation of apoptosis through oxidative stress and synthesis of pro- inflammatory cytokines (IL-6). An increase in the concen- tration of TNF-alpha and "NF-kB" nuclear factor contrib- utes to the activation of pro-inflammatory processes in the tumor cell [162]. The increased concentration of oxygen radi- cals and significant depletion of glutathione leads to the dys- function of mitochondria and the NADP/NAD system, im- paired permeability of the outer mitochondrial membrane, destruction of the respiratory chain, blocking the ATP synthe- sis and release of cytochrome C into cytosol, which are im- portant activating factors of caspase 3 (through apaf-1) and caspase 9 (Figure 7) [3, 101, 163–165]. Besides inhibiting the mitochondrial activity, AgNPs af- fect structural and functional characteristics of DNA. AgNPs provoke the DNA methylation, increasing the num- ber of chromosomal aberrations and malfunction of the repair system. For example, they cause the downregula- tion of proliferating cell nuclear antigen, i.e. the clamp of DNA polymerase, which plays an important role in the synthesis and reparation of DNA. As in bacterial cells, the released silver ions are able to disrupt the hydrogen bonds between the DNA bases, which leads to disorganization [3]. The activation of c-Jun NH2 terminal kinase (JNK) is an additional factor in DNA fragmentation and the atopic bodies formation [162]. Moreover, the action of AgNPs is characterized by the disturbance of metabolic processes and sensitization of the tumor cell, increasing its sensitivity to antitumor drugs, in particular, to 5-fluorouracil due to the modulating effect on expression of uracil phosphoribosyl transferase, which ultimately leads to active induction of apoptosis [166]. In addition, there are data on the pharmacological synergism of AgNPs and doxorubicin [167]. In lung fibroblasts and glioblastoma cells AgNPs in- duce different processes, such as metallothionein up- regulation, downregulation of the actin-binding protein and filamine, cell cycle arrest in phase G (2)/M [2, 168]. Biosynthesized AgNPs can block the cell cycle in G1 phase. One possible mechanism of the cell cycle arrest is the downregulation of cyclin B and cyclin E, whose nor- mal functioning is of the paramount importance for divi- sion processes [162]. AgNPs have antiangiogenic properties. In particular, AgNPs inhibit growth of blood vessels in tumor, limiting its progression. Probably, this effect is associated with VEGF (vascular endothelial growth factor) blocking and angiogenic FGF-2 (fibroblast growth factor 2) synthesis, as well as the inhibition of the transduction processes of the signaling pathways through the phosphorylation of KDR tyrosinekinase (VEGFR-2) and PI3K/Akt [169–171]. Another mechanism of inhibiting cancer cell prolifera- tion, vascular growth, and tumor progression implies a disruption of signaling transduction by suppressing the effects of hypoxia-induced factor-1a (HIF-1α) and matrix metalloproteases. Active growth and progression of the tumor are accompanied by insufficiently active formation of the vascular network, which leads to the cell hypoxia and, consequently, an increase in the concentration of HIF-1α. HIF-1α regulates the expression of genes responsi- ble for cellular activity: division, growth, and angiogenesis [172]. Matrix metalloproteases have similar functions. Resistance to the therapy is often accompanied with high activity of these signaling pathways, so their blocking plays a potentially important role in the modification of contemporary treatment approaches [3]. The antitumor effect of nanoparticles can be enhanced by coating with nano transfers, for example, with chi- tosan. Chitosan-coated nanoparticles have a greater inhib- itory effect and cause apoptosis at a lower concentration than uncoated AgNPs [2, 173]. Smaller nanoparticles (10, 20 nm) have the strongest antineoplastic effect compared to the large AgNPs (100 nm), which may be due to the greater penetrating capacity. Figure 7 Anticancer properties of AgNPs. AgNPs have a cytotoxic effect, inhibit the mitochondrial functions and boost the processes of oxidative stress and damage of tumor cell membrane. AgNPs also contribute to the suppression of the vascular growth factor (VEGF), which leads to the inhibition of the new vessels for- mation. Moreover, processes of tumor cells division are disturbed and the phenomenon of autophagy is observed. Reproduced from refs [180, 181]. Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 12 of 20 Moreover, charge and electrostatic interaction also af- fect internalization. For example, positively charged parti- cles penetrate more quickly and have greater cytotoxicity than particles with a neutral or negative charge. A positive charge allows the interaction both with a negatively charged membrane and albumin, which leads to formation of a protein shell. The "protein corona" allows AgNPs to enter cells through receptor-mediated endocyto- sis with further implementation of cytotoxic and genotoxic effects [75, 174, 175]. There are published data on the antitumor activity of AgNPs against a wide spectrum of oncological diseases in vitro: blood cancer (acute myeloid leukemia), breast cancer, hepatocellular carcinoma, osteosarcoma, lung cancer, mela- noma of the skin and mucous membranes, squamous cell carcinoma of the skin, colon cancer, osteosarcoma, cervical cancer, prostate cancer, adenocarcinoma of the stomach, bladder cancer and pancreas cancer [1, 3, 13, 176–179]. 3.3.6. Potential toxicity of silver nanoparticles Despite the data indicating the broad therapeutic potential of AgNPs, it is important to assess their toxicity, since the main biochemical effects of nanoparticles do not have bio- logical selectivity and can interact with the macroorgan- ism cells. Studying of toxic effects allows us to determine the therapeutic properties of drugs contain AgNPs and to minimize adverse side effects. The toxic properties of AgNPs depend on their size, shape, surface feature (nega- tively charged particles are less toxic), stabilizing agent and coating. Moreover, local environmental factors have an equally important role in toxicity: the strength of the ion interaction, presence of ligands, macromolecules and bivalent cations, as well as pH parameters [74, 182]. Toxic effects of nanoparticles have been studied either in vitro on cell cultures or in vivo on rodents. Different methodol- ogies and approaches make it difficult to determine com- mon parameters and characteristics of AgNPs toxicity. Pharmacokinetic features of AgNPs have wide distribu- tion in organs and tissues (lungs, CNS, kidneys, heart, liv- er, spleen, etc.), independent of the route of administra- tion. The clearance of nanoparticles can differ from 17 days to 4 months. In tissues containing natural physio- logical barriers, for example, brain, excretion of silver proceeds more slowly (up to 260 days), which creates ad- ditional conditions for its accumulation [183, 184]. Exces- sive accumulation of AgNPs leads to disruption of cells activity in different organs and systems: skin (argyria, contact dermatitis), respiratory system (bronchitis, alveo- litis, fibrosis, provocation of bronchial asthma exacerba- tions) visual system (conjunctivitis, argyrias), gastrointes- tinal tract (hepatobiliary and intestinal dysfunction), im- mune system (dysregulation of cytokine synthesis and function of cells), CNS (cognitive impairment, Alzheimer's disease, epileptic seizures), urinary system (acute tubular necrosis, glomerular dysfunction), cardiovascular system (bradycardia, AV block, ventricular arrhythmias) (Fig- ure 8) [3, 161, 185]. The possible nanoparticles accumula- tion in the reproductive system and damage of the germ cells structures were discovered by studying the patholog- ical effects of AgNPs in mice. Apart from the metabolic disturbances in germ cells and reduction of female oocyte fertility, the dysfunction of Leydig and Sertoli cells in males, which leads to infertility and a decrease in testos- terone synthesis, was observed [3]. An embryotoxic effect was detected in mice and zebrafish. This effect depended on the size and coating of AgNPs nanoparticles. Smaller AgNPs of 20 nm have greater toxicity than large particles of 110 nm, and polypyrimidine-coated particles are more toxic than citrate-coated AgNPs [186]. AgNPs also have a genotoxic effect due to chromosome damage, oxidative stress, and interaction with DNA [185, 187]. Figure 8 Main spectrum of AgNPs toxicity. AgNPs have a wide spectrum of toxicity, which has been studied in mice. The collect- ed data made it possible to predict the different consequences of nanoparticles accumulation in various organs and systems. Re- produced rom ref. [180]. The process of the silver ions permutation and their in- teraction with oxygen or sulfur, which induce a pathologi- cal biochemical cascade in cell, are considered the main mechanism of potential AgNPs cytotoxicity. It is believed that formation of Ag+ plays a key role in activation of lyso- somal acid phosphatases, dysfunction of the actin cyto- skelet, inhibition of Na+/K+-ATPase, stimulation of apopto- sis (through the protein p53, ACT, TP38), induction of oxi- dative stress, depletion of glutathione and diffusion of cellular components [101, 185, 188]. However, according to another hypothesis, independent mechanisms of metal- lic ions toxicity, which can be found for various nanoparti- cles, cause the pathological effects [189]. Except the direct entering into cell through the membrane (diffusion, endo- cytosis), it is supposed that AgNPs may penetrate through ion channels ("flip-flop" mechanism) and by the "Trojan horse" approach. The latter method consists of phagocyto- sis and further induction of petrochemical changes in ac- tive cells by silver ionization in the cytosol [101]. It is Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 13 of 20 worth mentioning that the most prominent morphological changes are observed in the liver, lungs, and kidneys, which may be due to the greatest participation of these organs in the clearance of nanoparticles [190]. AgNPs were found to induce oxidative stress in rat liv- er cells due to disturbance of metabolism and mitochon- drialal malfunction. These changes resulted in the focal liver necrosis, spleen edema and apoptosis in thymus cor- tex [185, 191]. Exposure to the most effective small nano- particles (10 nm or less) caused much more prominent pathological shifts [192]. The excretion of silver from the body is mostly carried out by the hepatobiliary system (more than 50%), which may be associated with more severe hepatotoxicity because of the greatest accumulation in hepatocytes, Kupffer cells and sinusoidal endothelio- cytes [193]. Kidneys are the second important excretion system. Silver nanoparticles accumulate in all structural components of cortex and medulla in spite of a low uri- nary excretion fraction (less than 0.01%) [161, 193]. Beyond hepato- and nephrotoxicity, silver particles may provokepathological changes in the intestinal wall despite a rather low level of absorption in the intestine ranging from 0.12% to 0.88%, which can be caused by the binding of nanoparticles to undigested food [193]. Howev- er, oral administration of silver nanoparticles in mice causes destruction of epithelial villi and glands. The impli- cation of bowel dysfunction causes weight loss [194]. In addition, nanoparticles have toxic effect on hearing and retina due to the activation of oxidative stress in the mitochondria, which leads to the loss of hearing and vi- sion [3, 190]. The accumulation of AgNPs in the central nervous sys- tem leads to disorganization of the cytoskeleton, activa- tion of neuroinflammation and increase in the insoluble beta-amyloid concentration. These processes are im- portant pathophysiological entity of Alzheimer's disease. Thus, it is possible to conclude that AgNPs can induce the development of neurodegenerative disorders, particularly due to low clearance and pathological accumulation [195]. Positively charged silver nanoparticles are known to have toxic effects on myocardial INA and IK1 channels, which leads to significantly increased risk of severe brad- ycardia [196]. Prolonged inhalation of AgNPs causes re- duction of tidal volume and enhancing of inflammatory processes in bronchopulmonary system [131, 197]. Thus, it is necessary to study and compare different AgNPs in order to determine the possibilities to synthesize less toxic nanoparticles with the strongest therapeutic effect. The study of pharmacodynamics and pharmacoki- netics properties of AgNPs to prevent pathological accu- mulation is equally important. Moreover, it is necessary to conduct further research, devoted to direct comparison of coating agents and to selecting the optimal coating ap- proach, because the coating and stabilization have been- proved to have a huge modifying effect. 4. Conclusion Given the wide range of biological, physical, and chemical properties of silver nanoparticles, a potential role of this compound in clinical medicine may be suggested. Appro- bation and usage of AgNPs are highly promising, especially in the era of growing antimicrobial resistance. In addition to activity against pathogenic bacteria, viruses, protozoa and fungi, silver nanoparticles are able to inhibit the activ- ity of tumor cells or play the role of drug carriers in the structures of malignant neoplasms in order to increase the effectiveness of chemotherapy [198]. Despite the promis- ing results of the studies, most of them are conducted mainly in cell cultures or mice. Therefore, pharmacody- namics and pharmacokinetics of silver nanoparticles in human have not been fully studied due to the reliable data on possible multisystem toxicity, which could restrict per- forming these kinds of studies. Moreover, silver nanopar- ticles are among the most toxic nanocompounds. There- fore, it is of tremendous importance to develop and evalu- ate potential AgNPs antidotes, such as sulfides [199]. A careful selection of the minimum toxic and at the same time the most effective dose of AgNPs is an important as- pect of planning the clinical and paraclinical trials. The nanoparticles synthesized by the "green synthesis" meth- ods are likely to have less toxicity and a wider biological spectrum against microorganisms, which makes this ap- proach more preferable than chemical or physical synthe- sis [200]. Further studies of different properties of nano- particles are needed to determine the most optimal con- centration, shape, structure and enveloping substance in vivo. These may be achieved by evaluating the dose- response parameters via comparison of the equivalent val- ues with the results from animal studies and extrapolated potential effects on humans [188]. Thus, it can be concluded that the integration of nano- technologies, AgNPs in particular, for various medical purposes is highly promising due to the clear biocidal ef- fects. However, the lack of unified methodological ap- proaches and the inconsistencies in the data leave a wide field for further research and development of unified algo- rithms to prevent biases, trial heterogeneity, inaccurate data processing and compilation. Limitations The type of article is a narrative review. It means a more relaxed literature search strategy, and it is affected by subjective approach (in contrast with PRSIMA and multi- ple search strategy by two or more authors), which limits the comprehensive data extraction and strict study selec- tion. There is a high probability of heterogeneity among studies due to the lack of strict prespecified criteria (selec- tion bias), which leads to the impossibility of generalizing results and performing the statistical summary effect as- sessment. All studies are carried out in vitro (cell lines) or Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 14 of 20 on mice/rats which makes it difficult to extrapolate the obtained results to humans and assess a potential minimal clinical important difference. There is a high demand for systematic reviews and meta-analyses according to the PRISMA guidelines for every particular topic of the current review, including extensive analysis of the pub- lished/unpublished literature and following the evidence- based practice. Such approach could improve the accuracy and understanding of the research objective for the medi- cal application of silver nanoparticles. Supplementary materials No supplementary materials are available. Funding This research had no external funding. Acknowledgments The author is grateful to Anastasia Vlasova for assitance in the preparation of the current review. Author contributions There is only one author. Conflict of interest The author declares no conflict of interest. Additional information Kodintcev Anton Nikolaevich – adult general neurologist at polyclinic of the Institute of High Temperature Electro- chemistry of the Ural Branch of the Russian Academy of Sciences and a young scientist at the same institute. Education: (MD) at Ural State Medical University (USMU), Faculty of General Medicine. Postgraduate courses, 2019–2022 at Ural State Medical University (USMU), Department of Neurology, Neurosur- gery and Medical Genetics Neurology residency: 2018–2020 at Yekaterinburg Medical Research Center for Prophylaxis and Health Pro- tection in Industrial Workers is a Research Institute of the Russian Agency for Consumers Rights Protection (Rospo- trebnadzor), Department of Neurology. Scientific interest: researching of new and cheap diag- nostic methods of neurodegenerative disorders, in particu- lar, Alzheimer’s disease based on different biomarkers of biofluids. Website: Institute of High Temperature Electrochemistry, http://www.ihte.uran.ru/?page_id=3106. References 1. Wei L, Lu J, Xu H, Patel A, Chen Z-S, Chen G. Silver nanopar- ticles: synthesis, properties, and therapeutic applications. Drug Discov Today. 2015;20(5):595–601. doi:10.1016/j.drudis.2014.11.014 2. Zhang X-F, Liu Z-G, Shen W, Gurunathan S. Silver nanoparti- cles: synthesis, characterization, properties, applications, and therapeutic approaches. Int J Mol Sci. 2016;17(9):E1534. doi:10.3390/ijms17091534 3. Xu L, Wang Y-Y, Huang J, Chen C-Y, Wang Z-X, Xie H. Silver nanoparticles: synthesis, medical applications and biosafety. Theranostics. 2020;10(20):8996–9031. doi:10.7150/thno.45413 4. Marin S, Vlascean GM, Tiplea RE, Bucur IR, Lemnaru M, Mar- in MM, Grumezescu AM. Applications and toxicity of silver nanoparticles: a recent review. Curr Top Med Chem. 2015;15(16):1596–1604. doi:10.2174/1568026615666150414142209 5. Markowska K, Grudniak AM, Wolska KI. Silver nanoparticles as an alternative strategy against bacterial biofilms. Acta Bi- ochim Pol. 2013;60(4):523–530. doi:10.18388/abp.2013_2016 6. Tăbăran A-F, Matea CT, Mocan T, Tăbăran A, Mihaiu M, Iancu C, Mocan L. Silver nanoparticles for the therapy of tuberculo- sis. Int J Nanomed. 2020;15:2231–2258. doi:10.2147/IJN.S241183 7. Fakhruddin KS, Egusa H, Ngo HC, Panduwawala C, Pesee S, Samaranayake LP. Clinical efficacy and the antimicrobial po- tential of silver formulations in arresting dental caries: a sys- tematic review. BMC Oral Health. 2020;20(1):160. doi:10.1186/s12903-020-01133-3 8. Qaralleh H, Khleifat KM, Al-Limoun MO, Alzedaneen FY, Al- Tawarah N. Antibacterial and synergistic effect of biosynthe- sized silver nanoparticles using the fungi tritirachium oryzae W5H with essential oil of centaurea damascena to enhance conventional antibiotics activity. Adv Nat Sci Nanosci Nano- technol. 2019;10:025016. doi:10.1088/2043-6254/ab28671 9. Kaur A, Preet S, Kumar V, Kumar R, Kumar R. Synergetic effect of vancomycin loaded silver nanoparticles for en- hanced antibacterial activity. Colloids Surf B Biointerfaces. 2019;176:62–69. doi:10.1016/j.colsurfb.2018.12.043 10. Hussein EAM, Mohammad AA-H, Harraz FA, Ahsan MF. Bio- logically synthesized silver nanoparticles for enhancing tet- racycline activity against staphylococcus aureus and klebsiel- la pneumoniae. Braz Arch Biol Technol. 2019;62. doi:10.1590/1678-4324-2019180266 11. Saha S, Malik MM, Qureshi MS. Study of synergistic effects of antibiotics and triangular shaped silver nanoparticles, syn- thesized using uv-light irradiation, on s. aureus and p. aeru- ginosa. Mater Today Proceed. 2019;18:920–927. doi:10.1016/j.matpr.2019.06.525 12. Farjadian F, Akbarizadeh AR, Tayebi L. Synthesis of novel reducing agent for formation of metronidazole-capped silver nanoparticle and evaluating antibacterial efficiency in gram- positive and gram-negative bacteria. Heliyon. 2020;6(8):e04747. doi:10.1016/j.heliyon.2020.e04747 13. Gherasim O, Puiu RA, Bîrcă AC, Burdușel A-C, Grumezescu AM. An updated review on silver nanoparticles in biomedi- cine. Nanomater. 2020;10(11):2318. doi:10.3390/nano10112318 14. Franci G, Falanga A, Galdiero S, Palomba L, Rai M, Morelli G, Galdiero M. Silver nanoparticles as potential antibacterial agents. Mol. 2015:20(5):8856–8874. doi:10.3390/molecules20058856 15. Burdușel A-C, Gherasim O, Grumezescu AM, Mogoantă L, Ficai A, Andronescu E. Biomedical applications of silver na- noparticles: an up-to-date overview. Nanomater. 2018:8(9):E681. doi:10.3390/nano8090681 16. Lubojanski A, Dobrzynski M, Nowak N, Rewak-Soroczynska J, Sztyler K, Zakrzewski W, Dobrzynski W, Szymonowicz M, Rybak Z, Wiglusz K, Wiglusz RJ. Application of selected na- http://www.ihte.uran.ru/?page_id=3106 https://doi.org/10.1016/j.drudis.2014.11.014 https://doi.org/10.3390/ijms17091534 https://doi.org/10.7150/thno.45413 https://doi.org/10.2174/1568026615666150414142209 https://doi.org/10.18388/abp.2013_2016 https://doi.org/10.2147/IJN.S241183 https://doi.org/10.1186/s12903-020-01133-3 https://doi.org/10.1088/2043-6254/ab28671 https://doi.org/10.1016/j.colsurfb.2018.12.043 https://doi.org/10.1590/1678-4324-2019180266 https://doi.org/10.1016/j.matpr.2019.06.525 https://doi.org/10.1016/j.heliyon.2020.e04747 https://doi.org/10.3390/nano10112318 https://doi.org/10.3390/molecules20058856 https://doi.org/10.3390/nano8090681 Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 15 of 20 nomaterials and ozone in modern clinical dentistry. Nano- mater. 2021;11(2):259. doi:10.3390/nano11020259 17. Guilger-Casagrande M, de Lima R. Synthesis of silver nano- particles mediated by fungi: a review. Front Bioeng Biotech- nol. 2019;7:287. doi:10.3389/fbioe.2019.00287 18. Dikshit PK, Kumar J, Das AK, Sadhu S, Sharma S, Singh S, Gupta PK, Kim BS. Green Synthesis of Metallic Nanoparticles: Applications and Limitations. Catalysts. 2021;11:902. doi:10.3390/catal11080902 19. Li W-R, Sun T-L, Zhou S-L, Ma Y-K, Shi Q-S, Xie X, Huang X- M. A comparative analysis of antibacterial activity, dynamics, and effects of silver ions and silver nanoparticles against four bacterial strains. Int Biodeterior Biodegradation. 2017;123. doi:10.1016/j.ibiod.2017.07.015 20. Galdiero S, Falanga A, Vitiello M, Cantisani M, Marra V, Galdiero M. Silver nanoparticles as potential antiviral agents. Mol. 2011;16(10):8894–8918. doi:10.3390/molecules16108894 21. Wei Q-Y, He K-M, Chen J-L, Xu Y-M, Lau ATY. Phytofabrica- tion of nanoparticles as novel drugs for anticancer applica- tions. Mol. 2019;24(23):4246. doi:10.3390/molecules24234246 22. Acosta-Torres LS, Mendieta I, Nuñez-Anita RE, Cajero-Juárez M, Castaño VM. Cytocompatible antifungal acrylic resin con- taining silver nanoparticles for dentures. Int J Nanomed. 2012;7:4777–4786. doi:10.2147/IJN.S32391 23. Panáček A, Kvítek L, Smékalová M, Večeřová R, Kolář M, Röderová M, Dyčka F, Šebela M, Prucek R, Tomanec O, Zbořil R. Bacterial resistance to silver nanoparticles and how to overcome it. Nat Nanotechnol. 2018;13(1):65–71. doi:10.1038/s41565-017-0013-y 24. Asadishad B, Hidalgo G, Tufenkji N. Pomegranate materials inhibit flagellin gene expression and flagellar-propelled mo- tility of uropathogenic escherichia coli strain CFT073. FEMS Microbiol Lett. 2012;334(2):87–94. doi:10.1111/j.1574-6968.2012.02622.x 25. Baethge C, Goldbeck-Wood S, Mertens S. SANRA-a scale for the quality assessment of narrative review articles. Res In- tegr Peer Rev. 2019;4:5. Published 2019 Mar 26. doi:10.1186/s41073-019-0064-8 26. Agnihotri S, Mukherji S, Mukherji S. Size-controlled silver nanoparticles synthesized over the range 5–100 nm using the same protocol and their antibacterial efficacy. RSC Adv. 2013;4(8):3974–3983. doi:10.1039/C3RA44507K 27. Baker C, Pradhan A, Pakstis L, Pochan D, Shah S. Synthesis and antibacterial properties of silver nanoparticles. J Nanosci Nanotechnol. 2005;5:244–249. doi:10.1166/jnn.2005.034 28. Liu J, Hurt RH. Ion release kinetics and particle persistence in aqueous nano-silver colloids. Environ Sci Technol. 2010;44(6):2169–2175. doi:10.1021/es9035557 29. Sotiriou GA, Pratsinis SE. Antibacterial activity of nanosilver ions and particles. Environ Sci Technol. 2010;44(14):5649– 5654. doi:10.1021/es101072s 30. Dobias J, Bernier-Latmani R. Silver release from silver nano- particles in natural waters. Environ Sci Technol. 2013;47(9):4140–4146. doi:10.1021/es304023p 31. Morones JR, Elechiguerra JL, Camacho A, Holt K, Kouri JB, Ramírez JT, Yacaman MJ. The bactericidal effect of silver na- noparticles. Nanotechnol. 2005;16(10):2346–2353. doi:10.1088/0957-4484/16/10/059 32. Radzig MA, Nadtochenko VA, Koksharova OA, Kiwi J, Lipasova VA, Khmel IA. Antibacterial effects of silver nano- particles on gram-negative bacteria: influence on the growth and biofilms formation, mechanisms of action. Colloids Surf B Biointerfaces. 2013;102:300–306. doi:10.1016/j.colsurfb.2012.07.039 33. Bélteky P, Rónavári A, Zakupszky D, Boka E, Igaz N, Szerencsés B, Pfeiffer I, Vágvölgyi C, Kiricsi M, Kónya Z. Are smaller nanoparticles always better? Understanding the bio- logical effect of size-dependent silver nanoparticle aggrega- tion under biorelevant conditions. Int J Nanomed. 2021;16:3021–3040. doi:10.2147/IJN.S304138 34. Levard C, Hotze EM, Lowry GV, Brown GE. Environmental transformations of silver nanoparticles: impact on stability and toxicity. Environ Sci Technol. 2012;46(13):6900–6914. doi:10.1021/es2037405 35. Ringe E, Zhang J, Langille M, Mirkin C, Marks L, Duyne R. Correlating the structure and localized surface plasmon reso- nance of single silver right bipyramids. Nanotechnol. 2012;23:444005. doi:10.1088/0957-4484/23/44/444005 36. Liu G, Eichelsdoerfer DJ, Rasin B, Zhou Y, Brown KA, Liao X, Mirkin CA. Delineating the pathways for the site-directed synthesis of individual nanoparticles on surfaces. Proceed Nat Acad Sci. 2013;110(3):887–891. doi:10.1073/pnas.1220689110 37. Wiley B, Sun Y, Xia Y. Synthesis of silver nanostructures with controlled shapes and properties. Acc Chem Res. 2007;40(10):1067–1076. doi:10.1021/ar7000974 38. Pal S, Tak YK, Song JM. Does the antibacterial activity of sil- ver nanoparticles depend on the shape of the nanoparticle? A Study of the gram-negative bacterium escherichia coli. Appl Environ Microbiol. 2007;73(6):1712–1720. doi:10.1128/AEM.02218-06 39. Cheon JY, Kim SJ, Rhee YH, Kwon OH, Park WH. Shape- dependent antimicrobial activities of silver nanoparticles. Int J Nanomed. 2019;14:2773–2780. doi:10.2147/IJN.S196472 40. Helmlinger J, Sengstock C, Groß-Heitfeld C, Mayer C, Schild- hauer TA, Köller M, Epple M. Silver nanoparticles with dif- ferent size and shape: equal cytotoxicity, but different anti- bacterial effects. RSC Adv. 2016;6(22):18490–18501. doi:10.1039/C5RA27836H 41. Alshareef A, Laird K, Cross RBM. Shape-dependent antibacte- rial activity of silver nanoparticles on escherichia coli and en- terococcus faecium bacterium. Appl Surf Sci. 2017;424:310– 315. doi:10.1016/j.apsusc.2017.03.176 42. Lee SH, Jun BH. Silver Nanoparticles: Synthesis and Applica- tion for Nanomedicine. Int J Mol Sci. 2019;20(4):865. doi:10.3390/ijms20040865 43. Zhang C, Hu Z, Deng B. Silver nanoparticles in aquatic envi- ronments: physiochemical behavior and antimicrobial mech- anisms. Water Res. 2016;88:403–427. doi:10.1016/j.watres.2015.10.025 44. Mackay ME, Dao TT, Tuteja A, Ho DL, van Horn B, Kim H-C, Hawker CJ. Nanoscale effects leading to non-einstein-like de- crease in viscosity. Nat Mater. 2003;2(11):762–766. doi:10.1038/nmat999 45. Zhang W, Yao Y, Li K, Huang Y, Chen Y. Influence of dissolved oxygen on aggregation kinetics of citrate-coated silver nano- particles. Environ Pollut. 2011;159(12):3757–3762. doi:10.1016/j.envpol.2011.07.013 46. Panáček A, Kvítek L, Prucek R, Kolář M, Večeřová R, Pizúrová N, Sharma VK, Nevěčná T, Zbořil R. Silver colloid nanoparti- cles: synthesis, characterization, and their antibacterial ac- tivity. J Phys Chem B. 2006;110(33):16248–16253. doi:10.1021/jp063826h 47. Shu M, He F, Li Z, Zhu X, Ma Y, Zhou Z, Yang Z, Gao F, Zeng M. Biosynthesis and antibacterial activity of silver nanoparti- cles using yeast extract as reducing and capping agents. Na- noscale Res Lett. 2020;15(1):14. doi:10.1186/s11671-019-3244-z 48. Ugwoke E, Aisida SO, Mirbahar AA, Arshad M, Ahmad I, Zhao T, Ezema FI. Concentration induced properties of silver na- noparticles and their antibacterial study. Surf Interfaces. 2020;18:100419. doi:10.1016/j.surfin.2019.100419 49. Ajitha B, Reddy YAK, Reddy PS, Jeon H-J, Ahn CW. Role of capping agents in controlling silver nanoparticles size, anti- bacterial activity and potential application as optical hydro- gen peroxide sensor. RSC Adv. 2016;6(42):36171–36179. doi:10.1039/C6RA03766F 50. Li Y, Zhang W, Niu J, Chen Y. Surface-coating-dependent dis- solution, aggregation, and reactive oxygen species (ROS) generation of silver nanoparticles under different irradiation conditions. Environ Sci Technol. 2013;47(18):10293–10301. doi:10.1021/es400945v https://doi.org/10.3390/nano11020259 https://doi.org/10.3389/fbioe.2019.00287 https://doi.org/10.3390/catal11080902 https://doi.org/10.1016/j.ibiod.2017.07.015 https://doi.org/10.3390/molecules16108894 https://doi.org/10.3390/molecules24234246 https://doi.org/10.2147/IJN.S32391 https://doi.org/10.1038/s41565-017-0013-y https://doi.org/10.1111/j.1574-6968.2012.02622.x https://doi.org/10.1186/s41073-019-0064-8 https://doi.org/10.1039/C3RA44507K https://doi.org/10.1166/jnn.2005.034 https://doi.org/10.1021/es9035557 https://doi.org/10.1021/es101072s https://doi.org/10.1021/es304023p https://doi.org/10.1088/0957-4484/16/10/059 https://doi.org/10.1016/j.colsurfb.2012.07.039 https://doi.org/10.2147/IJN.S304138 https://doi.org/10.1021/es2037405 https://doi.org/10.1088/0957-4484/23/44/444005 https://doi.org/10.1073/pnas.1220689110 https://doi.org/10.1021/ar7000974 https://doi.org/10.1128/AEM.02218-06 https://doi.org/10.2147/IJN.S196472 https://doi.org/10.1039/C5RA27836H https://doi.org/10.1016/j.apsusc.2017.03.176 https://doi.org/10.3390/ijms20040865 https://doi.org/10.1016/j.watres.2015.10.025 https://doi.org/10.1038/nmat999 https://doi.org/10.1016/j.envpol.2011.07.013 https://doi.org/10.1021/jp063826h https://doi.org/10.1186/s11671-019-3244-z https://doi.org/10.1016/j.surfin.2019.100419 https://doi.org/10.1039/C6RA03766F https://doi.org/10.1021/es400945v Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 16 of 20 51. Kim KD, Han D, Kim H. Optimization of experimental condi- tions based on the taguchi robust design for the formation of nano-sized silver particles by chemical reduction method. 2004. doi:10.1016/J.CEJ.2004.08.003 52. Sharma VK, Siskova KM, Zboril R, Gardea-Torresdey JL. Or- ganic-coated silver nanoparticles in biological and environ- mental conditions: fate, stability and toxicity. Adv Colloid In- terface Sci. 2014;204:15–34. doi:10.1016/j.cis.2013.12.002 53. Loza K, Diendorf J, Sengstock C, Ruiz-Gonzalez L, Gonzalez- Calbet JM, Vallet-Regi M, Köller M, Epple M. The dissolution and biological effects of silver nanoparticles in biological me- dia. J Mater Chem B. 2014;2(12):1634–1643. doi:10.1039/C3TB21569E 54. Mitra C, Gummadidala M, Afshinnia K, Merrifield RC, Baalousha M, Lead JR, Chanda A. Citrate-coated silver nano- particles growth-independently inhibit aflatoxin synthesis in aspergillus parasiticus. Environ Sci Technol. 2017;51(14):8085–8093. doi:10.1021/acs.est.7b01230 55. Shrivas K, Sahu B, Deb MK, Thakur SS, Sahu S, Kurrey R, Kant T, Patle TK, Jangde R. Colorimetric and paper-based de- tection of lead using pva capped silver nanoparticles: exper- imental and theoretical approach. Microchem J. 2019;150. doi:10.1016/j.microc.2019.104156 56. Rezaei H, Rahimpour E, Khoubnasabjafari M, Jouyban- Gharamaleki V, Jouyban A. A colorimetric nanoprobe based on dynamic aggregation of sds-capped silver nanoparticles for tobramycin determination in exhaled breath condensate. Microchim Acta. 2020;187. doi:10.1007/s00604-020-4162-6 57. Kamarudin D, Hashim NA, Ong BH, Che Hassan CR, Abdul Manaf N. Synthesis of silver nanoparticles stabilised by pvp for polymeric membrane application: a comparative study. Mat Technol. 2022;37(5):289–301. doi:10.1080/10667857.2021.1908768 58. Kubo A-L, Capjak I, Vrček IV, Bondarenko OM, Kurvet I, Vija H, Ivask A, Kasemets K, Kahru A. Antimicrobial potency of differently coated 10 and 50 nm silver nanoparticles against clinically relevant bacteria escherichia coli and staphylococ- cus aureus. Colloids Surf B Biointerfaces. 2018;170:401–410. doi:10.1016/j.colsurfb.2018.06.027 59. Leo BF, Chen S, Kyo Y, Herpoldt KL, Terrill NJ, Dunlop IE, McPhail DS, Shaffer MS, Schwander S, Gow A, Zhang J, Chung KF, Tetley TD, Porter AE, Ryan MP. The stability of silver na- noparticles in a model of pulmonary surfactant. Environ Sci Technol. 2013;47(19):11232–11240. doi:10.1021/es403377p 60. Kvítek L, Panáček A, Soukupová J, Kolář M, Večeřová R, Prucek R, Holecová M, Zbořil R. Effect of surfactants and pol- ymers on stability and antibacterial activity of silver nano- particles (NPs). J Phys Chem C. 2008;112(15):5825–5834. doi:10.1021/jp711616v 61. Roy A, Bulut O, Some S, Mandal AK, Yilmaz MD. Green syn- thesis of silver nanoparticles: biomolecule-nanoparticle or- ganizations targeting antimicrobial activity. RSC Adv. 2019;9(5):2673–2702. doi:10.1039/C8RA08982E 62. Tripathi RM, Chung SJ. Biogenic nanomaterials: synthesis, characterization, growth mechanism, and biomedical applica- tions. J Microbiol Method. 2019;157:65–80. doi:10.1016/j.mimet.2018.12.008 63. Hamouda RA, Hussein MH, Abo-elmagd RA, Bawazir SS. Syn- thesis and biological characterization of silver nanoparticles derived from the cyanobacterium oscillatoria limnetica. Sci Rep. 2019;9(1):13071. doi:10.1038/s41598-019-49444-y 64. Siddiqi KS, Husen A, Rao RAK. A review on biosynthesis of silver nanoparticles and their biocidal properties. J Nanobi- otechnol. 2018;16(1):14. doi:10.1186/s12951-018-0334-5 65. Begum NA, Mondal S, Basu S, Laskar RA, Mandal D. Biogenic synthesis of Au and Ag nanoparticles using aqueous solutions of black tea leaf extracts. Colloids Surf B Biointerfaces. 2009;71(1):113–118. doi:10.1016/j.colsurfb.2009.01.012 66. Loo, Y. Y, Chieng, B. W, Nishibuchi, M, Radu, S. Synthesis of silver nanoparticles by using tea leaf extract from camellia sinensis. Int J Nanomed. 2012;7:4263–4267. doi:10.2147/IJN.S33344 67. Rai M, Bonde S, Golinska P, et al. Fusarium as a Novel Fungus for the Synthesis of Nanoparticles: Mechanism and Applica- tions. J Fungi (Basel). 2021;7(2):13915. doi:10.3390/jof7020139 68. Konappa N, Udayashankar AC, Dhamodaran N, Krishna- murthy S, Jagannath S, Uzma F, Pradeep CK, De Britto S, Chowdappa S, Jogaiah S. Ameliorated antibacterial and anti- oxidant properties by trichoderma harzianum mediated green synthesis of silver nanoparticles. Biomol. 2021;11(4):535. doi:10.3390/biom11040535 69. Cheng F, Betts JW, Kelly SM, Hector AL. Green Synthesis of highly concentrated aqueous colloidal solutions of large starch-stabilised silver nanoplatelets. Mater Sci Eng C Mater Biol Appl. 2015;46:530–537. doi:10.1016/j.msec.2014.10.041 70. Skiba MI, Vorobyova VI, Pivovarov A, Makarshenko NP. Green synthesis of silver nanoparticles in the presence of polysac- charide: optimization and characterization. J Nanomat. 2020;e3051308. doi:10.1155/2020/3051308 71. Gopinath V, MubarakAli D, Vadivelu J, Manjunath Kamath S, Syed A, Elgorban AM. Synthesis of biocompatible chitosan decorated silver nanoparticles biocomposites for enhanced antimicrobial and anticancer property. Process Biochem. 2020;99:348–356. doi:10.1016/j.procbio.2020.09.011 72. Muhammad Tahir H, Saleem F, Ali S, Ain Q, Fazal A, Summer M, Mushtaq R, Tariq Zahid M, Liaqat I, Murtaza G. Synthesis of sericin-conjugated silver nanoparticles and their potential antimicrobial activity. J Basic Microbiol. 2020;60(5):458– 467. doi:10.1002/jobm.201900567 73. Azócar MI, Alarcón R, Castillo A, Blamey JM, Walter M, Paez M. Capping of silver nanoparticles by anti-inflammatory lig- ands: antibacterial activity and superoxide anion generation. J Photochem Photobiol B. 2019;193:100–108. doi:10.1016/j.jphotobiol.2019.02.005 74. Lee, K. J, Browning, L. M, Nallathamby, P. D, Xu, X.-H. N. Study of charge-dependent transport and toxicity of peptide- functionalized silver nanoparticles using zebrafish embryos and single nanoparticle plasmonic spectroscopy. Chem Res Toxicol. 2013;26(6):904–917. doi:10.1021/tx400087d 75. Abbaszadegan A, Ghahramani Y, Gholami A, Hemmateenejad B, Dorostkar S, Nabavizadeh M, Sharghi H. The effect of charge at the surface of silver nanoparticles on antimicrobial activity against gram-positive and gram-negative bacteria: a preliminary study. J Nanomat. 2015;e720654. doi:10.1155/2015/720654 76. Silhavy TJ, Kahne D, Walker S. The bacterial cell envelope. cold spring harbor perspectives in biology. 2010;2(5). doi:10.1101/cshperspect.a000414 77. El Badawy AM, Silva RG, Morris B, Scheckel KG, Suidan MT, Tolaymat TM. Surface charge-dependent toxicity of silver na- noparticles. Environ Sci Technol. 2011;45(1):283–287. doi:10.1021/es1034188 78. Qiao Z, Yao Y, Song S, Yin M, Luo J. Silver nanoparticles with ph induced surface charge switchable properties for antibac- terial and antibiofilm applications. J Mater Chem B. 2019;7(5):830–840. doi:10.1039/C8TB02917B 79. Stebounova LV, Guio E, Grassian VH. Silver nanoparticles in simulated biological media: a study of aggregation, sedimen- tation, and dissolution. J Nanoparticle Res. 2011;13:233–244. doi:10.1007/s11051-010-0022-3 80. Zhou W, Liu Y-L, Stallworth AM, Ye C, Lenhart JJ. Effects of pH, electrolyte, humic acid, and light exposure on the long- term fate of silver nanoparticles. Environ Sci Technol. 2016;50(22):12214–12224. doi:10.1021/acs.est.6b03237 81. McShan D, Ray PC, Yu H. Molecular toxicity mechanism of nanosilver. J Food Drug Anal. 2014;22(1):116–127. doi:10.1016/j.jfda.2014.01.010 82. Bélteky P, Rónavári A, Igaz N, Szerencsés B, Tóth IY, Pfeiffer I, Kiricsi M, Kónya Z. Silver nanoparticles: aggregation be- havior in biorelevant conditions and its impact on biological activity. Int J Nanomed. 2019;14:667–687. doi:10.2147/IJN.S185965 https://doi.org/10.1016/J.CEJ.2004.08.003 https://doi.org/10.1016/j.cis.2013.12.002 https://doi.org/10.1039/C3TB21569E https://doi.org/10.1021/acs.est.7b01230 https://doi.org/10.1016/j.microc.2019.104156 https://doi.org/10.1007/s00604-020-4162-6 https://doi.org/10.1080/10667857.2021.1908768 https://doi.org/10.1016/j.colsurfb.2018.06.027 https://doi.org/10.1021/es403377p https://doi.org/10.1021/jp711616v https://doi.org/10.1039/C8RA08982E https://doi.org/10.1016/j.mimet.2018.12.008 https://doi.org/10.1038/s41598-019-49444-y https://doi.org/10.1186/s12951-018-0334-5 https://doi.org/10.1016/j.colsurfb.2009.01.012 https://doi.org/10.2147/IJN.S33344 https://doi.org/10.3390/jof7020139 https://doi.org/10.3390/biom11040535 https://doi.org/10.1016/j.msec.2014.10.041 https://doi.org/10.1155/2020/3051308 https://doi.org/10.1016/j.procbio.2020.09.011 https://doi.org/10.1002/jobm.201900567 https://doi.org/10.1016/j.jphotobiol.2019.02.005 https://doi.org/10.1021/tx400087d https://doi.org/10.1155/2015/720654 https://doi.org/10.1101/cshperspect.a000414 https://doi.org/10.1021/es1034188 https://doi.org/10.1039/C8TB02917B https://doi.org/10.1007/s11051-010-0022-3 https://doi.org/10.1021/acs.est.6b03237 https://doi.org/10.1016/j.jfda.2014.01.010 https://doi.org/10.2147/IJN.S185965 Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 17 of 20 83. Gorshkov V, Bubis JA, Solovyeva EM, Gorshkov MV, Kjeldsen F. Protein corona formed on silver nanoparticles in blood plasma is highly selective and resistant to physicochemical changes of the solution. Environ Sci Nano. 2019;6(4):1089– 1098. doi:10.1039/C8EN01054D 84. Schöttler S, Landfester K, Mailänder V. Controlling the stealth effect of nanocarriers through understanding the pro- tein corona. Angew Chem Int Ed Engl. 2016;55(31):8806– 8815. doi:10.1002/anie.201602233 85. Lundqvist M, Stigler J, Elia G, Lynch I, Cedervall T, Dawson KA. Nanoparticle size and surface properties determine the protein corona with possible implications for biological im- pacts. Proc Nat Acad Sci USA. 2008;105(38):14265–14270. doi:10.1073/pnas.0805135105 86. Tenzer S, Docter D, Rosfa S, Wlodarski A, Kuharev J, Rekik A, Knauer SK, Bantz C, Nawroth T, BierC, Sirirattanapan J, Mann W, Treuel L, Zellner R, Maskos M, Schild H, Stauber RH. Nanoparticle size is a critical physicochemical determi- nant of the human blood plasma corona: a comprehensive quantitative proteomic analysis. ACS Nano. 2011;5(9):7155– 7167. doi:10.1021/nn201950e 87. Tai J-T, Lai C-S, Ho H-C, Yeh Y-S, Wang H-F, Ho R-M, Tsai D- H. Protein-silver nanoparticle interactions to colloidal stabil- ity in acidic environments. Langmuir. 2014;30(43):12755– 12764. doi:10.1021/la5033465 88. Alarcon E, Bueno-Alejo C, Noel C, Stamplecoskie K, Pacioni N, Poblete H, Scaiano JC. Human serum albumin as protecting agent of silver nanoparticles: role of the protein confor- mation and amine groups in the nanoparticle stabilization. J Nanoparticle Res. 2013;15:1–14. doi:10.1007/s11051-012-1374-7 89. Bhargava A, Dev A, Mohanbhai SJ, Pareek V, Jain N, Choudhury SR, Panwar J, Karmakar S. Pre-coating of protein modulate patterns of corona formation, physiological stabil- ity and cytotoxicity of silver nanoparticles. Sci Total Environ. 2021;772:144797. doi:10.1016/j.scitotenv.2020.144797 90. Walkey CD, Olsen JB, Song F, Liu R, Guo H, Olsen DWH, Co- hen Y, Emili A, Chan WCW. Protein corona fingerprinting predicts the cellular interaction of gold and silver nanoparti- cles. ACS Nano. 2014;8(3):2439–2455. doi:10.1021/nn406018q 91. Fernando I, Zhou Y. Impact of pH on the stability, dissolution and aggregation kinetics of silver nanoparticles. Chemo- sphere. 2019;216:297–305. doi:10.1016/j.chemosphere.2018.10.122 92. Sivera M, Kvitek L, Soukupova J, Panacek A, Prucek R, Vecerova R, Zboril R. Silver nanoparticles modified by gelatin with extraordinary pH stability and long-term antibacterial activity. PLOS ONE. 2014;9(8):e103675. doi:10.1371/journal.pone.0103675 93. Choi O, Clevenger TE, Deng B, Surampalli RY, Ross L, Hu Z. Role of sulfide and ligand strength in controlling nanosilver toxicity. Water Res. 2009;43(7):1879–1886. doi:10.1016/j.watres.2009.01.029 94. Zhang Y, Chen Y, Westerhoff P, Crittenden J. Impact of natu- ral organic matter and divalent cations on the stability of aqueous nanoparticles. Water Res. 2009;43(17):4249–4257. doi:10.1016/j.watres.2009.06.005 95. Baalousha M, Nur Y, Römer I, Tejamaya M, Lead JR. Effect of monovalent and divalent cations, anions and fulvic acid on aggregation of citrate-coated silver nanoparticles. Sci Total Environ. 2013;454–455:119–131. doi:10.1016/j.scitotenv.2013.02.093 96. Harmon AR, Kennedy AJ, Poda AR, Bednar AJ, Chappell MA, Steevens JA. Determination of nanosilver dissolution kinetics and toxicity in an environmentally relevant aqueous medium. Environ Toxicol Chem. 2014;33(8):1783–1791. doi:10.1002/etc.2616 97. Fan X, Yahia L, Sacher E. Antimicrobial properties of the ag, cu nanoparticle system. Biol. 2021;10(2):137. doi:10.3390/biology10020137 98. Salleh A, Naomi R, Utami ND, Mohammad AW, Mahmoudi E, Mustafa N, Fauzi MB. The potential of silver nanoparticles for antiviral and antibacterial applications: a mechanism of ac- tion. Nanomater. 2020;10(8):1566. doi:10.3390/nano10081566 99. Vazquez-Muñoz R, Meza-Villezcas A, Fournier PGJ, Soria- Castro E, Juarez-Moreno K, Gallego-Hernández AL, Bog- danchikova N, Vazquez-Duhalt R, Huerta-Saquero A. En- hancement of antibiotics antimicrobial activity due to the sil- ver nanoparticles impact on the cell membrane. PLoS One. 2019;14(11):e0224904. doi:10.1371/journal.pone.0224904 100. Li W-R, Xie X-B, Shi Q-S, Zeng H-Y, Ou-Yang Y-S, Chen Y-B. Antibacterial Activity and Mechanism of Silver Nanoparticles on Escherichia Coli. Appl Microbiol Biotechnol 2010, 85 (4), 1115–1122. doi:10.1007/s00253-009-2159-5 101. Akter M, Sikder MdT, Rahman MdM, Ullah AKMA, Hossain KFB, Banik S, Hosokawa T, Saito T, Kurasaki M. A systematic review on silver nanoparticles-induced cytotoxicity: physico- chemical properties and perspectives. J Adv Res. 2017;9:1–16. doi:10.1016/j.jare.2017.10.008 102. Qing Y, Cheng L, Ruiyan L, Liu G, Zhang Y, Tang X, Wang J, Liu H, Qin Y. Potential antibacterial mechanism of silver na- noparticles and the optimization of orthopedic implants by advanced modification technologies. Int J Nanomed. 2018;13. doi:10.2147/IJN.S165125 103. Dakal TC, Kumar A, Majumdar RS, Yadav V. Mechanistic basis of antimicrobial actions of silver nanoparticles. Front Micro- biol. 2016;7. 104. Kowalczyk P, Szymczak M, Maciejewska M, Laskowski Ł, Laskowska M, Ostaszewski R, Skiba G, Franiak-Pietryga I. All that glitters is not silver—a new look at microbiological and medical applications of silver nanoparticles. Int J Mol Sci. 2021;22:854. doi:10.3390/ijms22020854 105. Fanoro OT, Oluwafemi OS. Bactericidal antibacterial mecha- nism of plant synthesized silver, gold and bimetallic nanopar- ticles. Pharm. 2020;12(11):1044. doi:10.3390/pharmaceutics12111044 106. Lara H, Ayala-Nunez V, Ixtepan Turrent L, Rodríguez-Padilla C. Bactericidal effect of agnps against multidrug-resistant bacteria. World J Microbiol Biotechnol. 2009;26:615–621. doi:10.1007/s11274-009-0211-3 107. Lee MJ, Lee SJ, Yun SJ, Jang J-Y, Kang H, Kim K, Choi I-H, Park S. Silver nanoparticles affect glucose metabolism in he- patoma cells through production of reactive oxygen species. IJN. 2015;11:55–68. doi:10.2147/IJN.S94907 108. Yin IX, Zhang J, Zhao IS, Mei ML, Li Q, Chu CH. The antibac- terial mechanism of silver nanoparticles and its application in dentistry. IJN. 2020;15:2555–2562. doi:10.2147/IJN.S246764 109. Tang S, Zheng J. Antibacterial activity of silver nanoparticles: structural effects. Adv Health Mater. 2018;7(13):e1701503. doi:10.1002/adhm.201701503 110. Velusamy P, Su C-H, Venkat Kumar G, Adhikary S, Pandian K, Gopinath SCB, Chen Y, Anbu P. Biopolymers regulate silver nanoparticle under microwave irradiation for effective anti- bacterial and antibiofilm activities. PLoS One. 2016;11(6):e0157612. doi:10.1371/journal.pone.0157612 111. Ivask A, ElBadawy A, Kaweeteerawat C, Boren D, Fischer H, Ji Z, Chang CH, Liu R, Tolaymat T, Telesca D, Zink JI, Cohen Y, Holden PA, Godwin HA. Toxicity mechanisms in escherichia coli vary for silver nanoparticles and differ from ionic silver. ACS Nano. 2014;8(1):374–386. doi:10.1021/nn4044047 112. Gibała A, Żeliszewska P, Gosiewski T, Krawczyk A, Dura- czyńska D, Szaleniec J, Szaleniec M, Oćwieja M. Antibacterial and antifungal properties of silver nanoparticles—effect of a surface-stabilizing agent. Biomol. 2021;11(10):1481. doi:10.3390/biom11101481 113. Haytham MM Ibrahim. Green synthesis and characterization of silver nanoparticles using banana peel extract and their antimicrobial activity against representative microorgan- isms. J Radiation Res Appl Sci. 2015;8(3):265–275. doi:10.1016/j.jrras.2015.01.007 https://doi.org/10.1039/C8EN01054D https://doi.org/10.1002/anie.201602233 https://doi.org/10.1073/pnas.0805135105 https://doi.org/10.1021/nn201950e https://doi.org/10.1021/la5033465 https://doi.org/10.1007/s11051-012-1374-7 https://doi.org/10.1016/j.scitotenv.2020.144797 https://doi.org/10.1021/nn406018q https://doi.org/10.1016/j.chemosphere.2018.10.122 https://doi.org/10.1371/journal.pone.0103675 https://doi.org/10.1016/j.watres.2009.01.029 https://doi.org/10.1016/j.watres.2009.06.005 https://doi.org/10.1016/j.scitotenv.2013.02.093 https://doi.org/10.1002/etc.2616 https://doi.org/10.3390/biology10020137 https://doi.org/10.3390/nano10081566 https://doi.org/10.1371/journal.pone.0224904 https://doi.org/10.1007/s00253-009-2159-5 https://doi.org/10.1016/j.jare.2017.10.008 https://doi.org/10.2147/IJN.S165125 https://doi.org/10.3390/ijms22020854 https://doi.org/10.3390/pharmaceutics12111044 https://doi.org/10.1007/s11274-009-0211-3 https://doi.org/10.2147/IJN.S94907 https://doi.org/10.2147/IJN.S246764 https://doi.org/10.1002/adhm.201701503 https://doi.org/10.1371/journal.pone.0157612 https://doi.org/10.1021/nn4044047 https://doi.org/10.3390/biom11101481 Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 18 of 20 114. Lv X, Wang P, Bai R, Cong Y, Suo S, Ren X, Chen C. Inhibitory effect of silver nanomaterials on transmissible virus-induced host cell infections. Biomater. 2014;35(13):4195–4203. doi:10.1016/j.biomaterials.2014.01.054 115. Jeremiah SS, Miyakawa K, Morita T, Yamaoka Y, Ryo A. Po- tent antiviral effect of silver nanoparticles on SARS-CoV-2. Biochem Biophys Res Commun. 2020;533(1):195–200. doi:10.1016/j.bbrc.2020.09.018 116. Sun L, Singh AK, Vig K, Pillai SR, Singh SR. Silver nanoparti- cles inhibit replication of respiratory syncytial virus. J Bio- med Nanotechnol. 2008;4(2):149–158. doi:10.1166/jbn.2008.012 117. Lu L, Sun R, Chen R, Hui C-K, Ho C-M, Luk J, Lau G, Che C-M. Silver nanoparticles inhibit hepatitis B virus replication. An- tiviral Therapy. 2008;13:253–262. 118. Galdiero S, Falanga A, Vitiello M, Cantisani M, Marra V, Galdiero M. Silver nanoparticles as potential antiviral agents. Molecules. 2011;16(10):8894-8918. doi:10.3390/molecules16108894 119. Kumar S, Nyodu R, Maurya VK, Saxena SK. Morphology, Ge- nome Organization, Replication, and Pathogenesis of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Coronavirus Disease 2019 (COVID-19). 2020 Apr 30:23–31. doi:10.1007/978-981-15-4814-7_3 120. Gaikwad S, Ingle A, Gade A, Rai M, Falanga A, Incoronato N, Galdiero S, Galdiero M. Antiviral activity of mycosynthesized silver nanoparticles against herpes simplex virus and human parainfluenza virus type 3. Int J Nanomed. 2013;8:4303– 4314. doi:10.2147/IJN.S50070 121. Chen L, Liang J. An overview of functional nanoparticles as novel emerging antiviral therapeutic agents. Mater Sci Eng C Mater Biol Appl. 2020;112:110924. doi:10.1016/j.msec.2020.110924 122. Orlowski P, Tomaszewska E, Gniadek M, Baska P, Nowakow- ska J, Sokolowska J, Nowak Z, Donten M, Celichowski G, Gro- belny J, Krzyzowska M. Tannic acid modified silver nanopar- ticles show antiviral activity in herpes simplex virus type 2 infection. PLOS ONE. 2014;9(8):e104113. doi:10.1371/journal.pone.0104113 123. Mehrbod P, Motamed N, Tabatabaian M, Estyar R, Amini E, Shahidi M, Kheiri M. In vitro antiviral effect of “nanosilver” on influenza virus. DARU J Pharm Sci. 2009;17. 124. Lin Z, Li Y, Guo M, Xu T, Wang C, Zhao M, Wang H, Chen T, Zhu B. The inhibition of h1n1 influenza virus-induced apopto- sis by silver nanoparticles functionalized with zanamivir. RSC Adv. 2017;7(2):742–750. doi:10.1039/C6RA25010F 125. Nakamura S, Sato M, Sato Y, Ando N, Takayama T, Fujita M, Ishihara M. Synthesis and application of silver nanoparticles (Ag NPs) for the prevention of infection in healthcare work- ers. Int J Mol Sci. 2019;20(15):3620. doi:10.3390/ijms20153620 126. Elechiguerra JL, Burt JL, Morones JR, Camacho-Bragado A, Gao X, Lara HH, Yacaman MJ. Interaction of silver nanoparti- cles with HIV-1. J Nanobiotechnol. 2005;3(1):6. doi:10.1186/1477-3155-3-6 127. Lara HH, Ixtepan-Turrent L, Garza-Treviño EN, Rodriguez- Padilla C. PVP-coated silver nanoparticles block the transmis- sion of cell-free and cell-associated HIV-1 in human cervical culture. J Nanobiotechnol. 2010;8(1):15. doi:10.1186/1477-3155-8-15 128. Speshock JL, Murdock RC, Braydich-Stolle LK, Schrand AM, Hussain SM. Interaction of silver nanoparticles with tacaribe Virus. J Nanobiotechnol. 2010;8(1):19. doi:10.1186/1477-3155-8-19 129. Rogers JV, Parkinson CV, Choi YW, Speshock JL, Hussain SM. A preliminary assessment of silver nanoparticle inhibition of monkeypox virus plaque formation. Nanoscale Res Lett. 2008;3(4):129–133. doi:10.1007/s11671-008-9128-2 130. El-Sheekh MM, El-Kassas HY. Algal production of nano-silver and gold: their antimicrobial and cytotoxic activities: a re- view. J Genetic Eng Biotechnol. 2016;14(2):299–310. doi:10.1016/j.jgeb.2016.09.008 131. Almatroudi A. Silver nanoparticles: synthesis, characterisa- tion and biomedical applications. Open Life Sci. 2020;15(1):819–839. doi:10.1515/biol-2020-0094 132. Rai M, Kon K, Ingle A, Duran N, Galdiero S, Galdiero M. Broad-spectrum bioactivities of silver nanoparticles: the emerging trends and future prospects. Appl Microbiol Bio- technol. 2014;98(5):1951–1961. doi:10.1007/s00253-013-5473-x 133. Żarowska B, Koźlecki T, Piegza M, Jaros-Koźlecka K, Robak M. New look on antifungal activity of silver nanoparticles (AgNPs). Pol J Microbiol. 2019;68(4):515–525. doi:10.33073/pjm-2019-051 134. Kim K-J, Sung WS, Moon S-K, Choi J-S, Kim JG, Lee DG. Anti- fungal effect of silver nanoparticles on dermatophytes. J Mi- crobiol Biotechnol. 2008;18(8):1482–1484. 135. Suwan T, Khongkhunthian S, Okonogi S. Antifungal activity of polymeric micelles of silver nanoparticles prepared from psidium guajava aqueous extract. Drug Discoveries Thera- peutics. 2019;13:62–69. doi:10.5582/ddt.2019.01024 136. Lotfali E, Toreyhi H, Makhdoomi Sharabiani K, Fattahi A, Soheili A, Ghasemi R, Keymaram M, Rezaee Y, Iranpanah S. Comparison of antifungal properties of gold, silver, and sele- nium nanoparticles against amphotericin b-resistant candida glabrata clinical isolates. Avicenna J Medical Biotechnol. 2020;13(1):47–50. doi:10.18502/ajmb.v13i1.4578 137. Monteiro D, Silva S, Negri M, Gorup L, Camargo E, Oliveira R, Barros Barbosa D, Henriques M. Silver nanoparticles: influ- ence of stabilizing agent and diameter on antifungal activity against candida albicans and candida glabrata biofilms. Lett Appl Microbiol. 2012;54:383–391. doi:10.1111/j.1472-765X.2012.03219.x 138. Radhakrishnan VS, Reddy Mudiam MK, Kumar M, Dwivedi SP, Singh SP, Prasad T. Silver nanoparticles induced altera- tions in multiple cellular targets, which are critical for drug susceptibilities and pathogenicity in fungal pathogen (Can- dida albicans). Int J Nanomedicine. 2018;13:2647-2663. doi:10.2147/IJN.S150648 139. Rónavári A, Igaz N, Gopisetty MK, et al. Biosynthesized silver and gold nanoparticles are potent antimycotics against op- portunistic pathogenic yeasts and dermatophytes. Int J Na- nomedicine. 2018;13:695-703. doi:10.2147/IJN.S152010 140. Xu Y, Gao C, Li X, He Y, Zhou L, PangbG, Sun S. In Vitro anti- fungal activity of silver nanoparticles against ocular patho- genic filamentous fungi. J Ocul Pharmacol Ther. 2013;29(2):270–274. doi:10.1089/jop.2012.0155 141. Mussin JE, Roldán MV, Rojas F, Sosa M de los Á, Pellegri N, Giusiano G. Antifungal activity of silver nanoparticles in combination with ketoconazole against malassezia furfur. AMB Express. 2019;9:131. doi:10.1186/s13568-019-0857-7 142. Dilshad E, Bibi M, Sheikh NA, Tamrin KF, Mansoor Q, Maqbool Q, Nawaz M. Synthesis of functional silver nanopar- ticles and microparticles with modifiers and evaluation of their antimicrobial, anticancer, and antioxidant activity. J Funct Biomater. 2020;11(4):76. doi:10.3390/jfb11040076 143. Farrag HMM, Mostafa FAAM, Mohamed ME, Huseein EAM. Green biosynthesis of silver nanoparticles by aspergillus ni- ger and its antiamoebic effect against allovahlkampfia spe- laea trophozoite and cyst. Exp Parasitol. 2020;219:108031. doi:10.1016/j.exppara.2020.108031 144. Allahverdiyev AM, Abamor ES, Bagirova M, Ustundag CB, Kaya C, Kaya F, Rafailovich M. Antileishmanial effect of silver nanoparticles and their enhanced antiparasitic activity under ultraviolet light. Int J Nanomed. 2011;6:2705–2714. doi:10.2147/IJN.S23883 145. Fanti JR, Tomiotto-Pellissier F, Miranda-Sapla MM, et al. Biogenic silver nanoparticles inducing Leishmania ama- zonensis promastigote and amastigote death in vitro. Acta Trop. 2018;178:46-54. doi:10.1016/j.actatropica.2017.10.027 146. Mayelifar K, Taheri AR, Sazgarnia A. Ultraviolet B efficacy in improving antileishmanial effects of silver nanoparticles. Iran J Basic Medical Sci. 2015;18:677–683. doi:10.1016/j.actatropica.2017.10.027 https://doi.org/10.1016/j.biomaterials.2014.01.054 https://doi.org/10.1016/j.bbrc.2020.09.018 https://doi.org/10.1166/jbn.2008.012 https://doi.org/10.3390/molecules16108894 https://doi.org/10.1007/978-981-15-4814-7_3 https://doi.org/10.2147/IJN.S50070 https://doi.org/10.1016/j.msec.2020.110924 https://doi.org/10.1371/journal.pone.0104113 https://doi.org/10.1039/C6RA25010F https://doi.org/10.3390/ijms20153620 https://doi.org/10.1186/1477-3155-3-6 https://doi.org/10.1186/1477-3155-8-15 https://doi.org/10.1186/1477-3155-8-19 https://doi.org/10.1007/s11671-008-9128-2 https://doi.org/10.1016/j.jgeb.2016.09.008 https://doi.org/10.1515/biol-2020-0094 https://doi.org/10.1007/s00253-013-5473-x https://doi.org/10.33073/pjm-2019-051 https://doi.org/10.5582/ddt.2019.01024 https://doi.org/10.18502/ajmb.v13i1.4578 https://doi.org/10.1111/j.1472-765X.2012.03219.x https://doi.org/10.2147/IJN.S150648 https://doi.org/10.2147/IJN.S152010 https://doi.org/10.1089/jop.2012.0155 https://doi.org/10.1186/s13568-019-0857-7 https://doi.org/10.3390/jfb11040076 https://doi.org/10.1016/j.exppara.2020.108031 https://doi.org/10.2147/IJN.S23883 https://doi.org/10.1016/j.actatropica.2017.10.027 https://doi.org/10.1016/j.actatropica.2017.10.027 Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 19 of 20 147. Vergara-Duque D, Cifuentes-Yepes L, Hincapie-Riaño T, Clavijo-Acosta F, Juez-Castillo G, Valencia-Vidal B. Effect of Silver Nanoparticles on the Morphology of Toxoplasma gondii and Salmonella braenderup. J Nanotechnol. 2020;2020:9483428. doi:10.1155/2020/9483428 148. Norouzi R, Ataei A, Hejazy M, Noreddin A, El Zowalaty ME. Scolicidal Effects of Nanoparticles Against Hydatid Cyst Pro- toscolices in vitro. Int J Nanomed. 2020;15:1095-1100. doi:10.2147/IJN.S228538 149. Azzam A, Saad A, Soliman M, Bayoumy A. Antiparasitic activi- ty of silver and copper oxide nanoparticles against entamoe- ba histolytica and cryptosporidium parvum cysts. J Egyp Soc Parasitol. 2015;45:593–602. doi:10.12816/0017920 150. Costa IN, Ribeiro M, Silva Franco P, da Silva RJ, de Araújo TE, Milián ICB, Luz LC, Guirelli PM, Nakazato G, Mineo JR, Mineo TWP, Barbosa BF, Ferro EAV. Biogenic silver nanoparticles can control toxoplasma gondii infection in both human trophoblast cells and villous explants. Front Microbiol. 2020;11:623947. doi:10.3389/fmicb.2020.623947 151. Machado LF, Sanfelice RA, Bosqui LR, Assolini JP, Scando- rieiro S, Navarro IT, Depieri Cataneo AH, Wowk PF, Nakazato G, Bordignon J, Pavanelli WR, Conchon-Costa I, Costa IN. Bio- genic silver nanoparticles reduce adherence, infection, and proliferation of toxoplasma gondii rh strain in hela cells without inflammatory mediators induction. Exp Parasitol. 2020;211:107853. doi:10.1016/j.exppara.2020.107853 152. Gaafar MR, Mady RF, Diab RG, Shalaby TI. Chitosan and sil- ver nanoparticles: promising anti-toxoplasma agents. Exp Parasitol. 2014;143:30–38. doi:10.1016/j.exppara.2014.05.005 153. Adeyemi OS, Murata Y, Sugi T, Kato K. Inorganic nanoparti- cles kill toxoplasma gondii via changes in redox status and mitochondrial membrane potential. Int J Nanomed. 2017;12:1647–1661. doi:10.2147/IJN.S122178 154. Younis MS, Abououf EA, Ali AE, Abdelhady SM, Wassef RM. In Vitro effect of silver nanoparticles on blastocystis hominis. Int J Nanomed. 2020;15:8167–8173. doi:10.2147/IJN.S272532 155. Albalawi AE, Alanazi AD, Baharvand P, Sepahvand M, Mahmoudvand H. High potency of organic and inorganic na- noparticles to treat cystic echinococcosis: an evidence-based review. Nanomater. 2020;10(12):E2538. doi:10.3390/nano10122538 156. Nassef NE, Saad A-GE, Harba NM, Beshay EVN, Gouda MA, Shendi SS, Mohamed ASED. Evaluation of the therapeutic ef- ficacy of albendazole-loaded silver nanoparticles against echinococcus granulosus infection in experimental mice. J Parasit Dis. 2019;43(4):658–671. doi:10.1007/s12639-019-01145-z 157. Said DE, Elsamad LM, Gohar YM. Validity of silver, chitosan, and curcumin nanoparticles as anti-giardia agents. Parasitol Res. 2012;111(2):545–554. doi:10.1007/s00436-012-2866-1 158. Mnkandhla D, Marwijk J, Hoppe H, Wilhelmi BS, Whiteley CG. In vivo; in vitro interaction of silver nanoparticles with leucine aminopeptidase from human and plasmodium falci- parum. J Nanosci Nanotechnol. 2018;18(2):865–871. doi:10.1166/jnn.2018.13966 159. Hendiger EB, Padzik M, Sifaoui I, Reyes-Batlle M, López- Arencibia A, Rizo-Liendo A, Bethencourt-Estrella CJ, Nicolás- Hernández DS, Chiboub O, Rodríguez-Expósito RL, Grodzik M, Pietruczuk-Padzik A, Stępień K, Olędzka G, Chomicz L, Piñero JE, Lorenzo-Morales J. Silver nanoparticles as a novel potential preventive agent against acanthamoeba keratitis. Pathogen. 2020;9(5):E350. doi:10.3390/pathogens9050350 160. Xie Y, Bagby TR, Cohen MS, Forrest ML. Drug delivery to the lymphatic system: importance in future cancer diagnosis and therapies. Expert Opin Drug Deliv. 2009;6(8):785–792. doi:10.1517/17425240903085128 161. Mathur P, Jha S, Ramteke S, Jain NK. Pharmaceutical aspects of silver nanoparticles. Artif Cells Nanomed Biotechnol. 2018;46:115–126. doi:10.1080/21691401.2017.1414825 162. De Matteis V, Cascione M, Toma CC, Leporatti S. Silver nano- particles: synthetic routes, in vitro toxicity and theranostic applications for cancer disease. Nanomater. 2018;8(5):319. doi:10.3390/nano8050319 163. Dejean L, Martinez-Caballero S, Kinnally K. Is MAC the knife that cuts cytochrome c from mitochondria during apoptosis? Cell Death Differ. 2006;13:1387–1395. doi:10.1038/sj.cdd.4401949 164. Franco-Molina MA, Mendoza-Gamboa E, Sierra-Rivera CA, Gómez-Flores RA, Zapata-Benavides P, Castillo-Tello P, Alco- cer-González JM, Miranda-Hernández DF, Tamez-Guerra R. S, Rodríguez-Padilla C. Antitumor activity of colloidal silver on mcf-7 human breast cancer cells. J Exp Clin Cancer Res. 2010;29(1):148. doi:10.1186/1756-9966-29-148 165. Ahamed M, Akhtar MJ, Khan MAM, Alhadlaq HA. A Novel Green Preparation of Ag/RGO Nanocomposites with Highly Effective Anticancer Performance. Polym. 2021;13(19):3350. doi:10.3390/polym13193350 166. Packirisamy G, Gogoi S, Chattopadhyay A, Ghosh S. Implica- tions of silver nanoparticle induced cell apoptosis for in vitro gene therapy. Nanotechnol. 2008;19:075104. doi:10.1088/0957-4484/19/7/075104 167. Abdel-Fattah W, Ghareib W. On the anti-cancer activities of silver nanoparticles. J Appl Biotechnol Bioeng. 2018;5. doi:10.15406/jabb.2018.05.00116 168. AshaRani PV, Low Kah Mun G, Hande MP, Valiyaveettil S. Cytotoxicity and genotoxicity of silver nanoparticles in hu- man cells. ACS Nano. 2009;3(2):279–290. doi:10.1021/nn800596w 169. Ratan ZA, Haidere MF, Nurunnabi Md, Shahriar SMd, Aham- mad AJS, Shim YY, Reaney MJT, Cho JY. Green chemistry syn- thesis of silver nanoparticles and their potential anticancer effects. Cancers. 2020;12(4):855. doi:10.3390/cancers12040855 170. Kemp M, Kumar A, Mousa S, Dyskin E, Yalcin M, Ajayan P, Linhardt R, Mousa S. Gold and silver nanoparticles conjugat- ed with heparin derivative possess anti-angiogenesis proper- ties. Nanotechnol. 2009:20:455104. doi:10.1088/0957-4484/20/45/455104 171. Gurunathan S, Lee K-J, Kalishwaralal K, Sheikpranbabu S, Vaidyanathan R, Eom S. antiangiogenic properties of silver nanoparticles. Biomater. 2009;30:6341–6350. doi:10.1016/j.biomaterials.2009.08.008 172. Yang T, Yao Q, Cao F, Liu Q, Liu B, Wang X-H. Silver nanopar- ticles inhibit the function of hypoxia-inducible factor-1 and target genes: insight into the cytotoxicity and antiangiogene- sis. Int J Nanomed. 2016;11:6679–6692. doi:10.2147/IJN.S109695 173. Sanpui P, Chattopadhyay A, Ghosh S. Induction of apoptosis in cancer cells at low silver nanoparticle concentrations using chitosan nanocarrier. ACS Appl Materi Interfaces. 2011;3:218–228. doi:10.1021/am100840c 174. Barbalinardo M, Caicci F, Cavallini M, Gentili D. Protein co- rona mediated uptake and cytotoxicity of silver nanoparticles in mouse embryonic fibroblast small. Nanomater. 2018;14(34):e1801219. doi:10.1002/smll.201801219 175. Cunningham B, Engstrom AM, Harper BJ, Harper SL, Mackiewicz MR. Silver nanoparticles stable to oxidation and silver ion release show size-dependent toxicity in vivo. Na- nomater. 2021;11(6):1516. doi:10.3390/nano11061516 176. Ferreira LAB, Garcia-Fossa F, Radaic A, Durán N, Fávaro WJ, de Jesus MB. Biogenic silver nanoparticles: in vitro and in vi- vo antitumor activity in bladder cancer. Eur J Pharm Bio- pharm. 2020;151:162–170. doi:10.1016/j.ejpb.2020.04.012 177. Chen Y, Yang T, Chen S, Qi S, Zhang Z, Xu Y. Silver nanoparti- cles regulate autophagy through lysosome injury and cell hy- poxia in prostate cancer cells. J Biochem Molec Toxicol. 2020;34(5):e22474. doi:10.1002/jbt.22474 178. Xiao H, Chen Y, Alnaggar M. Silver nanoparticles induce cell death of colon cancer cells through impairing cytoskeleton and membrane nanostructure. Micron. 2019;126:102750. doi:10.1016/j.micron.2019.102750 179. Huy TQ, Huyen PTM, Le A-T, Tonezzer M. Recent advances of silver nanoparticles in cancer diagnosis and treatment. Anti- https://doi.org/10.1155/2020/9483428 https://doi.org/10.2147/IJN.S228538 https://doi.org/10.12816/0017920 https://doi.org/10.3389/fmicb.2020.623947 https://doi.org/10.1016/j.exppara.2020.107853 https://doi.org/10.1016/j.exppara.2014.05.005 https://doi.org/10.2147/IJN.S122178 https://doi.org/10.2147/IJN.S272532 https://doi.org/10.3390/nano10122538 https://doi.org/10.1007/s12639-019-01145-z https://doi.org/10.1007/s00436-012-2866-1 https://doi.org/10.1166/jnn.2018.13966 https://doi.org/10.3390/pathogens9050350 https://doi.org/10.1517/17425240903085128 https://doi.org/10.1080/21691401.2017.1414825 https://doi.org/10.3390/nano8050319 https://doi.org/10.1038/sj.cdd.4401949 https://doi.org/10.1186/1756-9966-29-148 https://doi.org/10.3390/polym13193350 https://doi.org/10.1088/0957-4484/19/7/075104 https://doi.org/10.15406/jabb.2018.05.00116 https://doi.org/10.1021/nn800596w https://doi.org/10.3390/cancers12040855 https://doi.org/10.1088/0957-4484/20/45/455104 https://doi.org/10.1016/j.biomaterials.2009.08.008 https://doi.org/10.2147/IJN.S109695 https://doi.org/10.1021/am100840c https://doi.org/10.1002/smll.201801219 https://doi.org/10.3390/nano11061516 https://doi.org/10.1016/j.ejpb.2020.04.012 https://doi.org/10.1002/jbt.22474 https://doi.org/10.1016/j.micron.2019.102750 Chimica Techno Acta 2022, vol. 9(4), No. 20229402 REVIEW 20 of 20 cancer Agents Med Chem. 2020;20(11):1276–1287. doi:10.2174/1871520619666190710121727 180. Xu L, Wang YY, Huang J, Chen CY, Wang ZX, Xie H. Silver nanoparticles: Synthesis, medical applications and biosafety. Theranostics. 2020;10(20):8996-9031. doi:10.7150/thno.45413 181. Skonieczna M, Hudy D. Biological Activity of Silver Nanopar- ticles and Their Applications in Anticancer Therapy; IntechOpen, 2018. https://doi.org/10.5772/intechopen.77075. 182. Bahadar H, Maqbool F, Niaz K, Abdollahi M. Toxicity of nano- particles and an overview of current experimental models. Iran Biomed J. 2016;20(1):1–11. doi:10.7508/ibj.2016.01.001 183. Ferdous Z, Nemmar A. Health impact of silver nanoparticles: a review of the biodistribution and toxicity following various routes of exposure. Int J Mol Sci. 2020;21(7):2375. doi:10.3390/ijms21072375 184. Lee JH, Kim YS, Song KS, Ryu HR, Sung JH, Park JD, Park HM, Song NW, Shin BS, Marshak D, Ahn K, Lee JE, Yu IJ. Bioper- sistence of silver nanoparticles in tissues from sprague– dawley rats. Particle Fibre Toxicol. 2013;10(1):36. doi:10.1186/1743-8977-10-36 185. Wen H, Dan M, Yang Y, Lyu J, Shao A, Cheng X, Chen L, Xu L. Acute toxicity and genotoxicity of silver nanoparticle in rats. PLoS One. 2017;12(9):e0185554. doi:10.1371/journal.pone.0185554 186. Kim K-T, Tanguay RL. The role of chorion on toxicity of silver nanoparticles in the embryonic zebrafish assay. Environ Health Toxicol. 2014;29:e2014021. doi:10.5620/eht.e2014021 187. Fewtrell L, Majuru B, Hunter PR. A re-assessment of the safe- ty of silver in household water treatment: rapid systematic review of mammalian in vivo genotoxicity studies. Environ Health. 2017;16(1):66. doi:10.1186/s12940-017-0279-4 188. Li Y, Cummins E. Hazard characterization of silver nanopar- ticles for human exposure routes. J Environ Sci Health A Tox Hazard Subst Environ Eng. 2020;55(6):704-725. doi:10.1080/10934529.2020.1735852 189. Antony JJ, Sivalingam P, Chen B. Toxicological effects of sil- ver nanoparticles. Environ Toxicol Pharmacol. 2015;40(3):729–732. doi:10.1016/j.etap.2015.09.003 190. Zou J, Feng H, Mannerström M, Heinonen T, Pyykkö I. Toxici- ty of silver nanoparticle in rat ear and BALB/c 3T3 cell line. J Nanobiotechnol. 2014;12:52. doi:10.1186/s12951-014-0052-6 191. Almofti MR. Silver ion induces a cyclosporine a-insensitive permeability transition in rat liver mitochondria and release of apoptogenic cytochrome. J Biochem. 2003;134(1):43–49. doi:10.1093/jb/mvg111 192. Cho Y-M, Mizuta Y, Akagi J, Toyoda T, Sone M, Ogawa K. Size- dependent acute toxicity of silver nanoparticles in mice. J Toxicol Pathol. 2018;31(1):73–80. doi:10.1293/tox.2017-0043 193. Sánchez-López E, Gomes D, Esteruelas G, Bonilla L, Lopez- Machado AL, Galindo R, Cano A, Espina M, Ettcheto M, Camins A, Silva AM, Durazzo A, Santini A, Garcia ML, Souto EB. Metal-based nanoparticles as antimicrobial agents: an overview. Nanomater. 2020;10(2):292. doi:10.3390/nano10020292 194. Liao C, Li Y, Tjong SC. Bactericidal and cytotoxic properties of silver nanoparticles. Int J Mol Sci. 2019;20(2):449. doi:10.3390/ijms20020449 195. Huang C-L, Hsiao I-L, Lin H-C, Wang C-F, Huang Y-J, Chuang C-Y. Silver nanoparticles affect on gene expression of in- flammatory and neurodegenerative responses in mouse brain neural cells. Environ Res. 2015;136:253–263. doi:10.1016/j.envres.2014.11.006 196. Lin C-X, Yang S-Y, Gu J-L, Meng J, Xu H-Y, Cao J-M. The acute toxic effects of silver nanoparticles on myocardial trans- membrane potential, INa and IK1 channels and heart rhythm in mice. Nanotoxicol. 2017;11(6):827–837. doi:10.1080/17435390.2017.1367047 197. Sung JH, Ji JH, Yoon JU, Kim DS, Song MY, Jeong J, Han BS, Han JH, Chung YH, Kim J, Kim TS, Chang HK, Lee EJ, Lee JH, Yu IJ. Lung Function changes in sprague-dawley rats after prolonged inhalation exposure to silver nanoparticles. Inhal Toxicol. 2008;20(6):567–574. doi:10.1080/08958370701874671 198. Amreddy N, Babu A, Muralidharan R, Panneerselvam J, Sri- vastava A, Ahmed R, Mehta M, Munshi A, Ramesh R. Recent advances in nanoparticle-based cancer drug and gene deliv- ery. Adv Cancer Res. 2018;137:115–170. doi:10.1016/bs.acr.2017.11.003 199. Levard C, Hotze EM, Colman BP, Dale AL, Truong L, Yang XY, Bone AJ, Brown GE, Tanguay RL, Di Giulio RT, Bernhardt ES, Meyer JN, Wiesner MR, Lowry GV. Sulfidation of silver nano- particles: natural antidote to their toxicity. Environ Sci Tech- nol. 2013;47(23):13440–13448. doi:10.1021/es403527n 200. Abbasi E, Milani M, Fekri Aval S, Kouhi M, Akbarzadeh A, Tayefi Nasrabadi H, Nikasa P, Joo SW, Hanifehpour Y, Nejati- Koshki K, Samiei M. Silver nanoparticles: synthesis methods, bio-applications and properties. Crit Rev Microbiol. 2016;42(2):173–180. doi:10.3109/1040841X.2014.912200 10 Most important cited papers 1. Yin IX, Zhang J, Zhao IS, Mei ML, Li Q, Chu CH. The antibac- terial mechanism of silver nanoparticles and its application in dentistry. Int J Nanomed. 2020;15:2555–2562. doi:10.2147/IJN.S246764 2. Bruna T, Maldonado-Bravo F, Jara P, Caro N. Silver Nanopar- ticles and Their Antibacterial Applications. Int J Mol Sci. 2021;22(13):7202. doi:10.3390/ijms22137202 3. Lee SH, Jun BH. Silver Nanoparticles: Synthesis and Applica- tion for Nanomedicine. Int J Mol Sci. 2019;20(4):865. doi:10.3390/ijms20040865 4. Zhang XF, Liu ZG, Shen W, Gurunathan S. Silver Nanoparti- cles: Synthesis, Characterization, Properties, Applications, and Therapeutic Approaches. Int J Mol Sci. 2016;17(9):1534. doi:10.3390/ijms17091534 5. Xu L, Wang Y-Y, Huang J, Chen C-Y, Wang Z-X, Xie H. Silver nanoparticles: synthesis, medical applications and biosafety. Theranostics. 2020;10(20):8996–9031. doi:10.7150/thno.45413 6. Gherasim O, Puiu RA, Bîrcă AC, Burdușel A-C, Grumezescu AM. An updated review on silver nanoparticles in biomedi- cine. Nanomater. 2020;10(11):2318. doi:10.3390/nano10112318 7. Ahamed M, Alsalhi MS, Siddiqui MK. Silver nanoparticle ap- plications and human health. Clin Chim Acta. 2010;411(23– 24):1841–1848. doi:10.1016/j.cca.2010.08.016 8. Siddiqi KS, Husen A, Rao RAK. A review on biosynthesis of silver nanoparticles and their biocidal properties. J Nanobi- otechnol. 2018;16(1):14. doi:10.1186/s12951-018-0334-5 9. Li Y, Cummins E. Hazard characterization of silver nanopar- ticles for human exposure routes. J Environ Sci Health A Tox Hazard Subst Environ Eng. 2020;55(6):704–725. doi:10.1080/10934529.2020.1735852 10. Zivic F, Grujovic N, Mitrovic S, Ahad IU, Brabazon D. Charac- teristics and applications of silver nanoparticles. In: et al. Commercialization of Nanotechnologies–A Case Study Ap- proach. Springer, Cham. doi:10.1007/978-3-319-56979-6_10 https://doi.org/10.2174/1871520619666190710121727 https://doi.org/10.7150/thno.45413 https://doi.org/10.5772/intechopen.77075 https://doi.org/10.3390/ijms21072375 https://doi.org/10.1186/1743-8977-10-36 https://doi.org/10.1371/journal.pone.0185554 https://doi.org/10.5620/eht.e2014021 https://doi.org/10.1186/s12940-017-0279-4 https://doi.org/10.1080/10934529.2020.1735852 https://doi.org/10.1016/j.etap.2015.09.003 https://doi.org/10.1186/s12951-014-0052-6 https://doi.org/10.1093/jb/mvg111 https://doi.org/10.1293/tox.2017-0043 https://doi.org/10.3390/nano10020292 https://doi.org/10.3390/ijms20020449 https://doi.org/10.1016/j.envres.2014.11.006 https://doi.org/10.1080/17435390.2017.1367047 https://doi.org/10.1080/08958370701874671 https://doi.org/10.1016/bs.acr.2017.11.003 https://doi.org/10.1021/es403527n https://doi.org/10.3109/1040841X.2014.912200 https://doi.org/10.2147/IJN.S246764 https://doi.org/10.3390/ijms22137202 https://doi.org/10.3390/ijms20040865 https://doi.org/10.3390/ijms17091534 https://doi.org/10.7150/thno.45413 https://doi.org/10.3390/nano10112318 https://doi.org/10.1016/j.cca.2010.08.016 https://doi.org/10.1186/s12951-018-0334-5 https://doi.org/10.1080/10934529.2020.1735852 https://doi.org/10.1007/978-3-319-56979-6_10