UMYU Journal of Microbiology Research

E-ISSN: 2814 – 1822; P-ISSN: 2616 – 0668

REVIEW ARTICLE

Recent Advances in the Delivery, Mechanism of Action and Antibacterial Activity of Silver Nanoparticles

*1Nazeef Idris Usman, 2Ahmed Faruk Umar, 3Mahmud Yerima Iliyasu and 4Amina Isa Kobbe

1Department of Microbiology, Bauchi State University, Bauchi, Nigeria

2Department of Biological Sciences, Abubakar Tafawa Balewa University, Bauchi, Nigeria

3Department of Biological Sciences, Abubakar Tafawa Balewa University, Bauchi, Nigeria

4Department of Science Laboratory Technology, Federal Polytechnic Bauchi, Nigeria

Abstract

Nanoparticles, especially silver nanoparticles (AgNPs), have revolutionized various fields like microbiology, biotechnology, pharmacy, and medicine owing to their distinct properties. This research delves into the significant potential of AgNPs in antimicrobial therapy, focusing on recent advancements in their delivery mechanisms, mechanisms of action, and antibacterial efficacy. The effective targeted delivery of AgNPs to specific body sites remains a challenge, leading to innovative approaches in nanotechnology. Nanogels, liposomes, and polymer-based nanoparticles have emerged as promising delivery systems, enhancing the stability, bioavailability, and controlled release of AgNPs. The antimicrobial activity of AgNPs is rooted in their unique physicochemical properties, such as high surface area and reactivity. They disrupt bacterial cell membranes, increasing permeability, causing cell death, and interfering with intracellular components. Additionally, AgNPs have shown potential in inhibiting biofilm formation, a common defense mechanism of bacteria against antibiotics. Despite their promise, addressing issues related to cytotoxicity and refining delivery methods remains imperative. This review comprehensively addresses the challenges associated with the delivery of AgNPs, their cytotoxic effects, and their efficacy against antibiotic-resistant bacteria, highlighting their mechanism of action in bacterial eradication and biofilm inhibition.

Keywords: Anti-microbial resistance, Delivery, Mechanism of action, silver nanoparticles

INTRODUCTION

A thorough literature search focusing on the delivery, mechanism of action, and antibacterial activity of silver nanoparticles published within the last 5-10 years was carried out. Online databases such as PubMed, Scopus, Web of Science, and Google Scholar were utilized. Keywords such as "silver nanoparticles", "delivery", "mechanism of action", "antibacterial activity", "nanotechnology", and "nanomedicine" were used to refine the search results and select pertinent articles for inclusion in the review.

ANTIBACTERIAL MECHANISM OF SILVER NANOPARTICLES

Much research has shown that AgNPs possess antimicrobial capabilities against a range of resistant bacteria. These include Klebsiella pneumoniae (Fontoura et al., 2023), hypervirulent Klebsiella pneumoniae (Lan et al., 2022), Streptococcus pyogenes (Avire et al., 2021), ampicillin-resistant E. coli (Ehsan et al., 2023), multidrug-resistant P. aeruginosa, Listeria monocytogenes, Methicillin-resistant Staphylococcus aureus, erythromycin-resistant, and K. aerogenes (Dove et al.,2023). The diversity of antibacterial activity of AgNPs cuts across many bacteria, and the action can be bactericidal and/or bacteriostatic (Ahmed et al., 2023; Ernest et al., 2014a). The mechanisms employed by AgNPs to induce bactericidal and bacteriostatic action against bacteria include but are not limited to the following:

Generation of Reactive Oxygen species (ROS)

The process by which AgNPs induce the generation of ROS in bacteria starts with penetration and release of silver ions. These silver ions are chemically reactive and can interact with various cellular components (Li et al.,2021). One of the primary mechanisms for ROS generation involves the impairment of the bacterial electron transport chain (ETC) (Li et al.,2020). This interruption can occur through several processes such as cell membrane damage, DNA damage, lipid peroxidation, deactivation of superoxide dismutase, induction of reactive stress, and generation of reactive oxygen species. Silver ions can interact with proteins and enzymes involved in the electron transport chain. This interaction can disrupt the flow of electrons along the chain (Faúndez et al.,2023). The disruption of the ETC can lead to the leakage of electrons from the chain. These leaked electrons can then react with oxygen (O2) to form superoxide radicals. Silver ions can also directly interact with oxygen molecules to form superoxide radicals and other ROS (Dryden, 2018). As highlighted in the equation below:

Ag+ + O2 → Ag + O2•−

The combined effects of disrupted ETC components and direct interaction with oxygen result in the accumulation of ROS within the bacterial cytoplasm. The increased levels of ROS overwhelm the bacterial natural antioxidant defense systems, which include enzymes like superoxide dismutase and catalase. This imbalance results in oxidative stress. ROS are highly volatile and can impair various cellular components, including lipids, proteins, and DNA, leading to protein oxidation, lipid peroxidation, and DNA strand breaks. The accumulation of oxidative damage disrupts essential cellular functions and processes, ultimately causing cell dysfunction and death (Nallanthighal et al.,2017).

Technically, ROS generation by silver nanoparticles leads to almost all antibacterial activity secondary to ROS production, which can result in cell membrane damage, cytoplasm leakage, DNA damage, oxidative stress, lipid peroxidation, and deactivation of ATP production (Mikhailova, 2020).

Oxidative Stress

Silver nanoparticles trigger harmful effects by producing ROS and initiating oxidative stress. Various mechanisms have been suggested to explain how metal nanoparticles exert their antibacterial effects. Among these, two frequently mentioned mechanisms involve the internal toxicity caused by released metal ions and the promotion of oxidative stress through elevated ROS. Another mechanism of oxidative stress induced by AgNPs in bacteria involves the generation of ROS and disruption of the cellular antioxidant defense systems (Mammari et al.,2022). Membrane damage and cytoplasmic leakage, secondary to the production of ROS, disrupt antioxidant activity by interfering with the superoxide dismutase (SOD) enzyme, thereby inducing oxidative stress and damaging all cell organelles within the cytoplasm. Ultimately, the buildup of ROS results in oxidative stress, which ultimately leads to the demise of the cell (Ameh et al.,2022).

Triclosan-Like Antibacterial lipid peroxidation

Triclosan functions by inhibiting lipid biosynthesis through its binding to the enoylacyl carrier protein reductase enzyme (ENR). This binding effectively hampers the microbe's ability to produce the necessary fatty acids for lipid production (Bibens et al.,2023). RNA sequencing data revealed that silver nanoparticles (AgNPs) triggered a bactericidal mechanism similar to triclosan, obstructing type II fatty acid biosynthesis. Additionally, the silver ion induced oxidative stress both inside and outside of K. pneumoniae cells (Pareek et al.,2021). Binding to ENR also leads to direct lipid peroxidation by initiating ferroptosis, which targets oxidant lipids. Uncontrolled lipid peroxidation results in the generation of hydroperoxides, lipid peroxyl radicals, and various oxidation byproducts, ultimately causing membrane rupture and cell death (Endale et al.,2023).

Cell membrane Damage

Silver ions (Ag+) released adhere to the bacterial cell wall and cytoplasmic membrane (More et al.,2023a). Multiple studies propose that they mainly target the cell membrane (Yin et al.,2020). In gram-negative bacteria, AgNPs primarily damage the cell wall at the plasma membrane, while the outer membrane is not impacted by the nanoparticles. The accumulation of silver nanoparticles denatures the cell membrane (More et al.,2023b). Additionally, due to their size, AgNPs can penetrate the cell walls, altering the structure of the cell membrane (Fernandes et al.,2023). This membrane denaturation can result in the disruption of subcellular organelles and even lead to cell destruction. Moreover, AgNPs can influence bacterial signal transduction that relies on phosphate transfer to protein substrates, and nanoparticles can remove phosphate from tyrosine residues on these peptide substrates. This disruption of signal transduction can trigger cell apoptosis and halt the multiplication of cells (Yin et al.,2020).

This mechanism that led to the destruction of bacterial membranes was observed in Gram-negative bacteria, where membrane proteins facilitate the uptake of silver ions into the cell. For instance, intracellular synthesis of AgNPs has been confirmed in Enterobacter cloacae (Mikhailova, 2020). Similar findings have been reported for Pseudomonas stutzeri (Rajora et al.,2016). This mechanism was also detected in Corynebacterium species and various other bacteria belonging to the Streptomyces genus. Among fungal representatives, Verticillium species are notable for their ability to synthesize AgNPs. Additionally, certain microorganisms can carry out AgNP biosynthesis both inside and outside the cell, including Bacillus strain CS 11 and Proteus mirabilis (Mikhailova, 2020).

Cytoplasmic leakage

The leakage of bacterial cytoplasmic contents may result from both extracellular and intracellular effects, primarily damage to the cellular membrane by nanoparticles (Yao et al.,2023). Several studies have demonstrated that nanoparticles can disrupt the structural integrity of microbes by interacting with outer cell wall components, leading to cell leakage and eventual cell death. Nanoparticles have the capacity to interfere with protein structures, causing damage to the cellular membrane and resulting in cell leakage and death. The presence of nanoparticles can induce ROS generation at the bacterial cell interface, further stressing the cell membrane, causing cytoplasmic leakage, and ultimately leading to bacterial cell death (Lohans et al.,2019). Sahoo (Sahoo et al.,2023) reported that qualitative and quantitative measurements of bacterial cytoplasmic content indicated its induction by nanoparticles. Furthermore, advanced microscopic analyses conducted by Modi (Modi et al.,2023) demonstrated increased polarization, as well as changes in protein and membrane fluidity leading to leakage.

The ability to induce biofilm formation, hinder growth processes, cause intracellular material release, increase membrane permeability, and stimulate the generation of reactive oxygen species may be associated with its smaller dimensions (5.7 nm), uniformity, zeta potential, and electrical conductivity (Kora & Sashidhar, 2015). This is most effective on gram-negative bacteria, specifically active against Klebsiella pneumoniae in antibiofilm formation (Scandorieiro et al.,2023).

Protein Denaturation

Nanoparticles can denature bacterial proteins through various mechanisms, often involving direct interactions between the nanoparticles and the proteins (Ozdal & Gurkok, 2022). Silver nanoparticles can interact with the surface of bacterial proteins, disrupting their native conformation. This interaction can lead to changes in the protein's secondary, tertiary, and quaternary structures, ultimately rendering the protein nonfunctional (More et al.,2023b). Electrical conductivity or electrostatic interactions of silver nanoparticles with bacterial cells may attract and bind to oppositely charged regions on bacterial proteins, distorting the protein's structure and destabilizing its functional state (Ozdal & Gurkok, 2022). Additionally, silver nanoparticles with hydrophobic surfaces can bind to hydrophobic regions on bacterial proteins, leading to protein unfolding and exposure of the protein's hydrophobic core, disrupting its natural structure and causing denaturation (More et al.,2023b). Bacterial proteins often contain disulfide bonds that contribute to their stability. Nanoparticles can interfere with these bonds, leading to the breakage of disulfide linkages and subsequent protein denaturation (Habibi et al.,2022). Furthermore, silver nanoparticles can physically interact with proteins, exerting mechanical forces that result in protein unfolding and denaturation (Park et al.,2023). This unfolding can occur through shear forces, compression, or stretching induced by the physical interactions of silver nanoparticles synthesized from Cissus quadrangularis extract with egg albumin (Kanimozhi et al.,2022). Most notably, silver nanoparticles and some other nanoparticles can generate ROS when interacting with bacterial cells. ROS can indirectly contribute to protein denaturation by causing oxidative stress and damaging the protein's structure (Endale et al.,2023; Li et al.,2021).

DNA Damage

Virtually all interactions between silver nanoparticles and bacteria affect multiple organelles within the bacteria, primarily triggered by the generation of reactive oxygen species (ROS) (Faúndez et al.,2023). AgNPs can induce the production of reactive oxygen species like O2 and H2O2, which are highly reactive and capable of damaging cellular components, including DNA. ROS can result in the formation of oxidative DNA lesions such as 8-hydroxy-2'-deoxyguanosine (8-OHdG) (Dryden, 2018). The direct binding of AgNPs to DNA molecules can cause physical disruption or destabilization of the DNA double helix, interfering with repair processes, DNA replication, and transcription (More et al.,2023b). Subsequent ROS generation due to AgNPs activity can lead to oxidative stress, physical interactions with DNA, or interference with DNA repair mechanisms, resulting in the breakage of both single and double-stranded DNA (Shukla et al.,2021). DNA repair pathways like base excision repair (BER) and homologous recombination (HR) are crucial for maintaining genomic integrity. Disruption of these pathways can lead to the accumulation of DNA damage (Nallanthighal et al.,2017). According to Chen et al.,(2022), AgNPs might induce epigenetic modifications that can alter gene expression patterns. Changes in gene expression can indirectly impact DNA integrity and repair mechanisms, highlighting the diverse factors that can influence how AgNPs damage bacterial DNA.

Alteration of Cell Division

Silver nanoparticles (AgNPs) can disrupt the normal process of cell division through various mechanisms, including mitotic arrest (Kah et al.,2023; Takáč et al.,2023), chromosomal aberrations (El-Ansary et al.,2023), spindle fiber dysfunction (Daphedar & Taranath, 2018), cytokinesis inhibition (Mikhailova, 2020; Takáč et al.,2023), DNA damage (Nallanthighal et al.,2017), and reactive oxygen species production (Frei et al.,2023). These disruptions can have significant implications for cellular health and overall organism well-being. The progression of the cell cycle is a crucial mechanism ensuring DNA replication, cell growth, division, and organism rejuvenation. However, overexpression of growth factors, gene mutations, or impaired regulation of tumor suppressor proteins can lead to cells losing the ability to exit the cell cycle, resulting in uncontrolled cell division (Takáč et al.,2023).

Deactivation of ATP synthesis

When bacteria are exposed to different levels of Silver nanoparticle treatment, the expression levels of key genes associated with responses that facilitate ATP synthesis are increased (Xu et al.,2023). Silver nanoparticles deactivate ATP synthesis by disrupting several metabolic pathways and the electron transport chain (ETC) responsible for ATP synthesis (Mikhailova, 2020).

Silver nanoparticles are typically small enough to breach the bacterial cell membrane. Once inside the bacterial cell, can release silver ions (Ag+). These ions are chemically reactive and can readily interact with cellular components (Anees Ahmad et al.,2020). The bacterial ETC is responsible for generating a proton gradient across the inner membrane, which is essential for ATP synthesis. Silver ions disrupt the ETC by binding to key proteins involved in electron transfer. This binding interferes with the flow of electrons along the ETC (More et al.,2023b). In normal bacterial respiration, the ETC pumps protons (H+) across the inner membrane to create a proton gradient (Yin et al.,2020). This gradient drives the ATP synthase complex, allowing it to produce ATP. Silver ions can inhibit the proton-pumping function of the ETC, preventing the establishment of the proton gradient. Silver ions can also directly interact with the ATP synthase enzyme, which is responsible for synthesizing ATP from adenosine diphosphate (ADP) and inorganic phosphate (Pi). Inhibition of ATP synthase by silver ions further reduces ATP production. As a result of these multiple interferences, the bacterial cell experiences a significant reduction in ATP production Since ATP is the primary energy currency of the cell, its depletion severely affects the cell's ability to carry out essential metabolic processes and eventually leads to bacterial deactivation (McNeilly et al.,2021).

The stepwise antibacterial mechanism responsible for generating ROS is presented in Figure 1 below

Spectrum of bacteria susceptible to silver nanoparticles

Several research studies have confirmed the antimicrobial effectiveness of AgNPs against a range of resistant bacteria. These bacteria include Streptococcus pyogenes resistant to erythromycin, E. coli resistant to ampicillin, multidrug-resistant Pseudomonas aeruginosa, Listeria monocytogenes, Klebsiella pneumoniae, and K. aerogenes (Dove et al.,2023). Gram-negative bacteria tend to be more vulnerable to the action of silver nanoparticles because their cell walls are thinner compared to those of gram-positive strains (Yin et al.,2020). These nanoparticles demonstrated their effectiveness against other bacterial pathogens such as Hemophilus influenzae and Streptococcus pneumoniae as evidenced by their minimum inhibitory concentrations (MICs): 109.4, 120.60, and 138.80 μg/ml for AgNPs, and 105.80, 114.40, and 129.06 μg/ml for AgNPs, respectively. Furthermore, these nanoparticles inhibited biofilm formation in S. pneumoniae, H. influenzae, and N. meningitidis by 73.14%, 71.89%, and 64.81%, respectively. These results validate the potential of synthesized AgNPs in combating multidrug-resistant (MDR) microbes that cause meningitis and form biofilms (Dove et al.,2023). The measured minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values of the surface-stabilized silver nanoparticles indicate their efficacy as potent antibacterial agents even when used in small quantities.

Figure 1: Stepwise Mechanism for the Generation of Reactive Oxygen Species by Silver Nanoparticles (Ameh et al., 2022; Frei et al., 2023; Guo et al., 2016; N. Li et al., 2023; Nakamura et al., 2016)

ANTIMICROBIAL PROPERTIES OF SILVER NANO PARTICLES

Silver nanoparticles exhibit antimicrobial properties, and their potential toxicity can be evaluated by determining several parameters, including Minimum Inhibitory Concentration (MIC) (Bano et al.,2023), Minimum Bactericidal Concentration (MBC) (Kim et al.,2023), Host Cell Inhibitory Concentration (IL50), Host Cell Lethal Concentration (LD50) (Dutt et al.,2023), and Minimum Cytotoxic Concentration (MCC) (Noga et al.,2023). However, it is crucial to acknowledge that various factors can significantly impact MIC, MBC, IC50, LD50, and MCC values. These factors include nanoparticle size, the presence of stabilizing agents, conjugation with other substances, electrical conductivity (EC), pH levels, Total Dissolved Solids (TDS), and the concentration of dissolved oxygen (DO) in the solution where the nanoparticles are present. Recent advancements in determining the MIC and MBC of these nanoparticles are detailed in Table 1 below.

MIC and MBC of Silver Nanoparticles

The minimum inhibitory concentration (MIC) for AgNPs was shown to be effective against both gram-positive and gram-negative nosocomial pathogens using various methods, with the U-bottom, 96-well microtiter plate incubation method being particularly notable. Enterococcus faecalis demonstrated inhibition at concentrations ranging from 1.05 μg/mL to 2.5 μg/mL, as reported by Dutt et al.,(2023), with a MBC of 10 μg/mL according to Tverezovska et al.,2022). P. aeruginosa and S. aureus were inhibited at concentrations of 1 μg/mL and 2 μg/mL, respectively, as documented by Liao et al., in 2019. Conversely, E. coli, S. aureus, and S. multivorum exhibited inhibition at 0.003 μM, 0.003 μM, and 0.25 μM, respectively, and were killed at 0.031 μM, 0.015 μM, and 0.25 μM when AgNPs were stabilized with cetyltrimethylammonium bromide (CTAB), per Ameh et al.,(2023). Dove et al.,(2023) reported an E. coli with an MBC of 6.2 μg/mL, while the MIC was 3.1 μg/mL. MICs of 1 µg/mL against P. aeruginosa and 2 µg/mL against S. aureus were also observed for AgNPs. The antibacterial effect of AgNPs was found to be dosage-dependent based on the results of the cell viability assay, with MICs of 0.8 μg/mL against P. melaninogenica and 1.0 μg/mL against A. pyogenes. Additionally, MBC values of AgNPs against P. melaninogenica and A. pyogenes were determined to be 1.0 μg/mL and 1.5 μg/mL, respectively (Liao et al.,2019). Klebsiella pneumoniae and Proteus mirabilis showed inhibition at concentrations of 25 µg/mL, 50 µg/mL, and 75 µg/mL when exposed to morphologically altered AgNPs, with effective elimination occurring at a concentration of 100 µg/mL, as reported by Rajivgandhi et al.,2023).

The effectiveness of silver nanoparticles in inhibiting microbial and human cells can be assessed through the measurement of half maximal inhibitory concentration (IC50). For instance, in a study by Manivasagan et al.. (2013), HeLa cells were inhibited at concentrations of 200 μg/mL. Another study by Hublikar et al.. (2023) found that silver nanoparticles synthesized using Lantana camara leaf extract exhibited an IC50 value of 49.52 g/mL when tested with Cell Proliferation Kit I. At a dose of 50 μg/mL, silver nanoparticles affected mouse embryonic cells, while 1 µg/mL induced cytotoxicity and abnormal cell morphology in the human hepatoma cell line (HepG2), leading to DNA damage, production of reactive oxygen species (ROS), increased oxidative stress, and ultimately apoptosis (Noga et al., 2023). Additionally, synthesized silver nanoparticles demonstrated anticancer activity against Michigan Cancer Foundation breast cell lines (MCF-7) with IC50 values of 2.41 µg·mL−1 (Nguyen et al., 2023). In studies by Bano et al.. (2023), the IC50 values of silver nanoparticles for N. meningitidis, S. pneumoniae, and H. influenzae were found to be 82.90, 87.20, and 99.50 μg/ml, respectively. Furthermore, HCT-116 colon carcinoma cells were inhibited at a concentration of 744.23 µg/mL (Dutt et al., 2023).

In experiments involving a mouse model, Alfatemi et al.. (2020) determined that the lethal concentration 50 (LC50) of synthesized nonchelating silver nanoparticles sized between 20nm-35nm ranged from 250mg/kg to 350mg/kg. In a study by Mao et al.. (2018) using the drosophila model, the range of lethal concentration of silver nanoparticles was observed to start from 20-50µl/ml, with the size of the drosophila decreasing as the concentration of the silver nanoparticles increased.

Table 1: The Minimum Inhibitory and Bactericidal Concentration of AgNPs on Some Nosocomial Pathogens

Some prevalent Nosocomial Pathogen Forms of AgNPs

AgNPs

Size

MIC

(µg/Ml)

MBC (µg/mL) Ref.
E.coli Free AgNPs 9.26 to 31.18 nm 85 127.5 (Selem et al.,2022)
Cedecea capped AgNPs 10-100nm 12.5 (More et al.,2023b)
Serum Protein coated AgNPs 2–20 nm 16.0 32.0 (Du et al.,2023)
K.pneumoniae Imipenem-AgNPs Conjugate 20nm 0.015 0.5 (Fontoura et al.,2023)
Amikacin-AgNPs Conjigate 17 nm 3.1 6.9 (Dove et al.,2023)
AgNp 20-65nm 62.5 125 (Siddique et al.,2020)
P.aeraginosa Cedecea capped AgNPs 10-100nm 6.25 12.5 (More et al.,2023b)
Free AgNPs 11nm 15.6 31.25 (Al-Momani et al.,2023)
Free AgNPs 20nm 10.0 15.6 (Abeer Mohammed et al.,2022)
E.feacalis Free AgNPs 40-50nm 2.5 10 (More et al.,2023a)
Free AgNPs 1-100nm 300 300 (Kaukab et al.,2023)
AgNPs –Vancomycin Conjugate 392nm 0.12 1.0 (Veriato et al.,2023)
S.aureus AgNPs –Vancomycin Conjugate 392nm 0.25 1.0 (Veriato et al.,2023)
AgNPs 1-100nm 30 60 (da Cunha et al.,2023)

Minimum Cytotoxic Concentration of Silver Nanoparticles

In in-vitro cell culture experiments, it has been observed that silver nanoparticles (AgNPs) can exhibit toxicity towards various human cell lines (Shi et al.,2018). While silver nanoparticles (AgNPs) possess various biological properties with extensive applications, our knowledge regarding their potential effect on human health and the environment remains incomplete. The safety of administering AgNPs, whether intravenously or intradermally, is not fully comprehended. Consequently, there is an ongoing concern regarding their potential effects on heart, liver, and kidney functions (Olugbodi et al.,2023). Extensive research has been conducted on the toxicity of AgNPs in various models, including animals, cells, and humans, primarily due to concerns about their potential bioaccumulation. AgNPs predominantly enter cells through diffusion and endocytosis, whether through inhalation, ingestion, or skin contact, and tend to amass in different organs, including the heart, liver, and kidneys. In the liver, the presence of AgNPs triggers the generation of ROS (Faúndez et al.,2023) and oxidative stress, which, in turn, activate inflammation by stimulating the production of chemokines and cytokines. This inflammatory response can further aggravate damage to the liver (Olugbodi et al.,2023). Due to this bioaccumulation, even minute concentrations of AgNPs may be cytotoxic to both microbial and human cells. The cytotoxicity caused by AgNPs may vary depending on their concentration, size, and exposure duration, with a notable emphasis on nanoparticles with sizes equal to or less than 10 nanometers as reported by (Liao et al.,2019).

Various studies have investigated the effects of silver nanoparticles (AgNPs) on different cell types. In one study by Dilshad et al. (2020), human breast and liver cell lines were exposed to maleic acid capped AgNPs at a concentration of 50µM. Kakian et al. (2024) studied the impact of AgNPs on Proteus mirabilis bacteria and Fibroblast L929 cells, using a concentration of 7.8 µg/ml. Halkai et al. (2019) examined human gingival fibroblast cells treated with AgNPs at a concentration of 260 μg/ml. Bhatia et al. (2021) investigated the effects of AgNPs on C2C12 skeletal muscle cell line at a concentration of 5.45 μg/ml. Klein et al. (2023) studied human oral fibroblast cell line exposed to AgNPs at a concentration of 0.015 mg/mL. Lastly, Kadir et al. (2024) looked at the impact of AgNPs on African Green Monkey Kidney, Vero cells at a concentration of 500 µg/mL as presented in the Table 2 below:

Table 2: Minimum cytotoxic concentration of silver nanoparticles

Cell type Forms of AgNp MCC References
Human breast and liver cell lines maleic acid capped AgNps 50µM (Dilshad et al., 2020)

-Bacteria-Proteus mirabilis

-Fibroblast L929 cells

AgNPs 7.8 µg/ml (Kakian et al., 2024)
human gingival fibroblast AgNPs 260 μg/ml (Halkai et al., 2019)
C2C12 skeletal muscle cell line AgNPs 5.45 μg/ml (Bhatia et al., 2021)
human oral fibroblast cell line AgNPs 0.015 mg/mL. (Klein et al., 2023)
African Green Monkey Kidney, Vero cells AgNPs 500 µg/mL (Kadir et al., 2024)

MCC = Minimum Cytotoxic Concentration

DELIVERY AND TRANSPORTATION OF SILVER NANOPARTICLE

The impact of silver nanoparticles (AgNPs) is contingent on the delivery method, the combination of compounds involved, as well as their intrinsic characteristics like shape and size, which are significantly shaped by the manufacturing process. Keeping these factors into consideration, silver nanoparticles can serve as a transporter and can be delivered coated using other biomolecules. There are various methods that can be used for the delivery of silver nanoparticles, depending on the intended use.

Topical application of AgNPs in Cream, Gels, Spray, and Nanogels

Silver nanoparticles have been extensively studied for their broad-spectrum antimicrobial properties and have been recently integrated into creams and ointments for topical application in wound healing (Paladini & Pollini, 2019). AgNPs can also be delivered topically through bandages, sutures, plasters, and various other creams and ointments (Parveen et al.,2018). Recently, AgNPs have been included in hydrogels, sponges, and electrospun nanofibers. An ointment containing Chloroxine-conjugated Silver nanoparticles has shown effectiveness in wound treatment compared to Calendula flower oil verseline (Nandhini et al.,2023). Additionally, a silver nanoparticle-based electrospun spray, combined with polycaprolactone/chitosan, has been tested to create a nanofibrous dressing for wound healing (Saghafi et al.,2023). Utilizing AgNPs immobilized in other medical sprays can enhance the treatment of topical bacterial infections.

These nanogels are hydrophilic polymer nanoparticles sized between 20 to 200 nm, which are cross-linked. They can be administered orally, topically, vaginally, via ocular routes, or through other methods. Their size and flexible membranes facilitate controlled drug release, enhanced skin penetration, and swelling through diffusion. Importantly, nanogels are highly biocompatible and can effectively deliver a wide array of hydrophilic medications (Mali & Patil, 2023). Other biocompatible compound drug delivery for antimicrobial purposes includes liposomes, niosomes, microemulsion, nano-emulsion, metal nanoparticles, polymeric nanoparticles, quantum dots, colloidal gold, nano diamonds, dendrimers, nanocrystals, carbon nanotubes (Jayachandran et al.,2023).

One of the advantages of topical application of AgNPs, either in composite or conjugate form, is enhanced tissue repair by approximately 99%, achieved through increased angiogenesis, collagen deposition, and accelerated re-epithelialization. The combined action of AgNPs conjugates also leads to an increase in the expression of growth factors and a decrease in the expression of pro-inflammatory factors, particularly IL-6 and TNF-α. These results suggest that with optimized AgNP concentrations, designed antibacterial, cytocompatible, and nanoporous materials can be used as effective wound dressings (Rybka et al., 2022). However, a drawback of these particles is their small size, which allows them to penetrate natural barriers like the skin, potentially resulting in immediate and long-term toxic effects (Ferdous & Nemmar, 2020).

Oral Delivery of AgNPs

For the safe and efficient delivery of silver nanoparticles to host cells, various approaches have been proposed involving the coating, conjugation, and immobilization of AgNPs with different biomolecules. Naturally occurring polymers play multiple roles, such as serving as carriers, providing specific binding surfaces, and offering protective coatings. These polymers can also incorporate biologically active molecules with properties like anti-cancer, antimicrobial, or anticoagulant effects, making them useful as therapeutic agents or natural medications. Polymers derived from plants or microbes, such as poly(saccharides) and poly(phenols), can provide structural support and a means to connect and transport medications (Veerapandian et al.,2023). This concept can also be applied to AgNPs. Recent methods for the safe delivery of AgNPs and other nanomaterials include the Red Blood Cell Membrane Camouflaged Nanoparticle (RBCMCN) (Dai et al.,2021), originally designed for treating tumor cells but may also be effective against bacteria, the Hyaluronic Acid-based Drug Nano Carrier (HABDNC) (Mauri & Scialla, 2023), the Polymer Lipid Hybrid nanocarrier (PLHN) (de Araujo et al.,2023), and various other approaches that can be incorporated into tablets and capsules (Edis et al.,2021).

The oral route takes advantage of the highly absorptive intestinal epithelium, which provides a substantial surface area for drug absorption in the gastrointestinal (GI) tract. Every section of the gastrointestinal tract has distinct functions, including protection, secretion, absorption, and digestion (Mathur et al., 2018). Oral administration of silver nanoparticles has several disadvantages, such as the use of preparation techniques that employ toxic organic solvents, which can be harmful to biological systems. Additionally, there is a tendency for the nanoparticles to aggregate and the drugs to be expelled due to crystallization during storage (Lotfipour et al., 2021).

Intravenous Delivery of AgNPs

Using various animal models, intravenous administration of AgNPs has been shown to result in their accumulation in multiple organs (Ferdous & Nemmar, 2020). This method offers a rapid response and precise control over the rate of drug delivery into the body, making it suitable for medications that cannot be absorbed through the gastrointestinal tract or administered via intramuscular or other tissue injections. Importantly, intravenous delivery bypasses first-pass metabolism (Chandrakala et al.,2022). However, AgNPs have the potential to disrupt the endothelial barrier by reducing the interaction of VE-cadherin in endothelial cells, leading to increased permeability and subsequent peripheral inflammation in organs like the liver, kidney, and lungs (Guo et al.,2016). A study by Wieler et al.,(2023) investigated the intravenous application of AgNPs in patients with moderate-severe and severe COVID-19 pneumonia, revealing its safety and efficacy. This approach could serve as a cost-effective and readily available adjunctive therapy for severe COVID-19 pneumonia cases, potentially reducing mortality rates and the need for additional oxygen regardless of the specific SARS-CoV-2 variants. Additionally, AgNPs may hold promise for addressing other respiratory infections, particularly in light of the increasing resistance of pathogens to existing anti-infective agents (Wieler et al.,2023).

The advantage of biocompatible AgNPs in intravenous delivery systems allows for precise targeting, resulting in regulated drug release and a sustained impact. These systems offer benefits in cancer treatment, such as improved medication targeting and penetration into tumor cells, enhancing therapeutic efficacy (Nikolova et al., 2023). However, concerns regarding toxicity and immune system recognition pose potential risks that need to be addressed. Extensive research is crucial to improve these systems and overcome their limitations for safe and effective clinical use. Conflicting mechanisms of action, complex engineering processes, high manufacturing costs, and limited practicality for widespread clinical adoption present additional challenges. Additionally, the potential toxicity of polymeric nanoparticles and their elimination from the body create obstacles for their clinical application (Elumalai et al., 2024).

Nebulized formulation of AgNPs for delivery to the airways

The optimal size of nanoparticles formulated for delivery to the airways ranges between 3-7nm with a minimal bactericidal concentration of 10μg/ml and a calculated tissue deposition concentration of 5μg/ml aerosol droplets as reported by Zachar, (2022). Other trials also utilize 3-7nm AgNPs formulation for airway delivery and a minimum antibacterial concentration of 10-25μg/ml with 5μg/ml aerosol droplets in a cumulative volume of 0.25cc from a source with a concentration of 100ppm (μg/ml) in the bronchial tree, and delivering a total volume of 1cc from a source with a concentration of 250ppm (μg/ml) into the alveoli of the lungs also reported by Zachar, (2020). Silver nanoparticles-OCT mixture encapsulated with alginate to create a carrier called AgNPs-OCT-Alg, designed for drug delivery via nebulization. Each formulation had a total dosage of 1.27 mg/kg, and 50 mL of each formulation was nebulized at a rate of 5 mL/h. In the case of alginate-coated AgNPs-OCT resulting in AgNPs-OCT-Ag, this straightforward delivery carrier was effectively employed for nebulization. The formulation was successfully delivered and internalized in healthy lung tissues through the nebulization method in the animal model (Abdellatif et al.,2022).

The nebulized formulation of agnps presents challenges such as rapid drug clearance from the nasal cavity and potential enzymatic degradation, which warrant further investigation. The nasal mucus, nasal epithelium, and lamina propria collectively form the natural anatomical barrier for drug delivery from the nose to the brain. Safety concerns include potential toxicity to nasal cilia or various nasal cells, local inflammation, and systemic effects (Chen et al., 2024). The advantages of intranasal administration of silver nanoparticles include increased loading capacity, protection against degradation, enhanced stability, precise targeting, potential dosage reduction, and the ability to improve affinity for Aβ proteins (Dighe et al., 2024).

Intramuscular or subcutaneous injection/delivery of silver nanoparticles

Silver nanoparticles taken orally can be absorbed through paracellular transport, transcytosis, and M-cell uptake within the gastrointestinal tract. Conversely, macrophages and lymphatic uptake primarily absorb nanoparticles through subcutaneous, intramuscular, or inhaled routes (Mahmod & Al-Jumaili, 2022; Mathur et al.,2018). The most suitable infection model for evaluating the effectiveness of nanomedicine in animal-based studies to combat infections is a model involving skin and subcutaneous region infections. In this context, silver nanoparticles (AgNPs) were administered intravenously to mice to address subcutaneous abscesses while exposing them to near-infrared (NIR) irradiation. The application of NIR induced a hyperthermal effect, resulting in increased release of Ag and enhanced eradication of MRSA, ultimately promoting the healing of abscesses and wound acceleration (Yeh et al.,2020).

The advantages of administering silver nanoparticles intramuscularly include rapid and uniform absorption, allowing for larger drug volumes to be injected compared to subcutaneous delivery. However, there are potential drawbacks to this method, such as injection site discomfort, dosage dependence on muscle mass, the risk of hematoma formation, and the potential for unintentional drug entry into the bloodstream from accidental vessel rupture (Sultana et al., 2022).

STRATEGY FOR SILVER NANOPARTICLE CONJUGATION/IMMOBILIZATION

Many different delivery strategies for AgNPs can assist in safe delivery and prevent the induction of cytotoxicity.

Nanocarriers

Nanocarriers, such as liposomes, micelles, and polymers, can be utilized to encapsulate AgNPs and shield them from degradation and rapid clearance. These nanocarriers offer the advantage of being functionalized to target specific tissues and cells (Tiwari et al.,2023). They represent a promising strategy for delivering AgNPs to specific tissues and cells, enhancing their stability, circulation time, and targeting precision. Nandhini et al.,(2023) introduced a novel micellar nanocarrier tailored for agnp delivery to wound sites. This nanocarrier was engineered to release AgNPs gradually, thereby fostering the process of wound healing.

Conjugates

AgNPs can be conjugated to various molecules, including antibodies, peptides, and aptamers, to target specific receptors and transporter proteins (Todorova et al.,2023). One significant benefit of AgNP conjugates is their ability to target specific cells or tissues by attaching them to molecules that bind to specific receptors on the target cells or tissues. For instance, AgNPs can be linked to antibodies that target cancer cells or peptides that target bacteria (Veerapandian et al.,2023).

Another advance in nanoparticle conjugation involves the immobilization of silver nanoparticles using enzymes or as capping enzymes. The conjugation of silver nanoparticles with lysozyme and colistin was proven effective in disrupting bacterial biofilms (Meesaragandla et al.,2022). Additionally, the synergistic conjugation of lysozyme with AgNPs increased the antibacterial activity of the conjugates by 86% (Ernest et al.,2014).

External stimuli

External stimuli, such as magnetic fields, ultrasound, and light, can be utilized to regulate the release of AgNPs from nanocarriers or conjugates (Jain et al.,2022). The development of external stimuli-responsive AgNPs has addressed this challenge by enabling the controlled release of drugs in response to specific stimuli like light, magnetic fields, and ultrasound (Kah et al.,2023). This tailored approach facilitates precise and targeted drug delivery to distinct cells and tissues. For example, light-responsive AgNPs have been engineered to release their drug payload upon exposure to near-infrared light, proving effective in delivering drugs to tumors in mice with minimal side effects. Similarly, magnetic-responsive AgNPs have been devised to release drugs in response to an alternating magnetic field, demonstrating efficacy in drug delivery to the brain in rats while minimizing damage to healthy tissue (Sahoo et al.,2023).

CONCLUSION

Effective AgNP antimicrobial activity in the treatment of pathogens, including covirulent multidrug and hypervirulent strains, depends on the delivery method, conjugates, and nanocarriers. The stepwise mechanism of AgNPs' actions and recent advances in delivery methods provide a roadmap for the safe delivery of AgNPs as alternative nanobiotics for treating resistant strains. Various forms of AgNPs in conjugates and capped forms, when applied in sprays and antiseptics, have been shown to be effective against nosocomial pathogens, thus contributing to the prevention of hospital-acquired infections and, consequently, community-acquired infections.

ACKNOWLEDGEMENT

The authors are highly thankful to their families for their motivation, time, and encouragement.

CONFLICT OF INTEREST

No conflict-of-interest record about this study

FINANCIAL SUPPORT

No financial support received for this study

REFERENCES

Abdellatif, A. A. H., Khan, R. A., Alhowail, A. H., Alqasoumi, A., Sajid, S. M., Mohammed, A. M., Alsharidah, M., Rugaie, O. A., & Mousa, A. M. (2022). Octreotide-conjugated silver nanoparticles for active targeting of somatostatin receptors and their application in a nebulized rat model. Nanotechnology Reviews, 11(1), 266–283. [Crossref]

Abeer Mohammed, A. B., Abd Elhamid, M. M., Khalil, M. K. M., Ali, A. S., & Abbas, R. N. (2022). The potential activity of biosynthesized silver nanoparticles of Pseudomonas aeruginosa as an antibacterial agent against multidrug-resistant isolates from intensive care unit and anticancer agent. Environmental Sciences Europe, 34(1), 109. [Crossref]

Ahmed, S., Ahmed, M. Z., Rafique, S., Almasoudi, S. E., Shah, M., Jalil, N. A. C., & Ojha, S. C. (2023). Recent Approaches for Downplaying Antibiotic Resistance: Molecular Mechanisms. BioMed Research International, 2023, e5250040. [Crossref]

Aldakheel, F. M., Sayed, M. M. E., Mohsen, D., Fagir, M. H., & El Dein, D. K. (2023). Green Synthesis of Silver Nanoparticles Loaded Hydrogel for Wound Healing; Systematic Review. Gels, 9(7), 530. [Crossref]

Alfatemi, S., Fallah, F., Armin, S., Hafizi, M., Karimi, A., & Kalanaky, S. (2020). Evaluation of Blood and Liver Cytotoxicity and Apoptosis-necrosis Induced by Nanochelating Based Silver Nanoparticles in Mouse Model. Iranian Journal of Pharmaceutical Research : IJPR, 19, 207–218. [Crossref]

Al-Momani, H., Almasri, M., Al Balawi, D., Hamed, S., Albiss, B. A., Aldabaibeh, N., Ibrahim, L., Albalawi, H., Al Haj Mahmoud, S., Khasawneh, A. I., Kilani, M., Aldhafeeri, M., Bani-Hani, M., Wilcox, M., Pearson, J., & Ward, C. (2023). The efficacy of biosynthesized silver nanoparticles against Pseudomonas aeruginosa isolates from cystic fibrosis patients. Scientific Reports, 13, 8876. [Crossref]

Ameh, T., Gibb, M., Stevens, D., Pradhan, S. H., Braswell, E., & Sayes, C. M. (2022). Silver and Copper Nanoparticles Induce Oxidative Stress in Bacteria and Mammalian Cells. Nanomaterials, 12(14). [Crossref]

Ameh, T., Zarzosa, K., Dickinson, J., Braswell, W. E., & Sayes, C. M. (2023). Nanoparticle surface stabilizing agents influence antibacterial action. Frontiers in Microbiology, 14. www.frontiersin.org

Anees Ahmad, S., Sachi Das, S., Khatoon, A., Tahir Ansari, M., Afzal, Mohd., Saquib Hasnain, M., & Kumar Nayak, A. (2020). Bactericidal activity of silver nanoparticles: A mechanistic review. Materials Science for Energy Technologies, 3, 756–769. [Crossref]

Avire, N. J., Whiley, H., & Ross, K. (2021). A Review of Streptococcus pyogenes: Public Health Risk Factors, Prevention and Control. Pathogens, 10(2), 248. [Crossref]

Bano, N., Iqbal, D., Al Othaim, A., Kamal, M., Albadrani, H. M., Algehainy, N. A., Alyenbaawi, H., Alghofaili, F., Amir, M., & Roohi. (2023). Antibacterial efficacy of synthesized silver nanoparticles of Microbacterium proteolyticum LA2(R) and Streptomyces rochei LA2(O) against biofilm forming meningitis causing microbes. Scientific Reports, 13, 4150. [Crossref]

Bhatia, D., Mittal, A., & Malik, D. K. (2021). Antimicrobial potential and in vitro cytotoxicity study of polyvinyl pyrollidone-stabilised silver nanoparticles synthesised from Lysinibacillus boronitolerans. IET Nanobiotechnology, 15(4), 427–440. [Crossref]

Bibens, L., Becker, J.-P., Dassonville-Klimpt, A., & Sonnet, P. (2023). A Review of Fatty Acid Biosynthesis Enzyme Inhibitors as Promising Antimicrobial Drugs. Pharmaceuticals, 16(3), 425. [Crossref]

Chandrakala, V., Aruna, V., & Angajala, G. (2022). Review on metal nanoparticles as nanocarriers: Current challenges and perspectives in drug delivery systems. Emergent Materials, 5(6), 1593–1615. [Crossref]

Chen, Y., Sheng, F., Wang, X., Zhang, Z., Qi, S., & Chen, L. (2022). Early Epigenetic Responses in the Genomic DNA Methylation Fingerprints in Cells in Response to Sublethal Exposure of Silver Nanoparticles. Frontiers in Bioengineering and Biotechnology, 10, 927036. [Crossref]

Chen, Y., Zhang, C., Huang, Y., Ma, Y., Song, Q., Chen, H., Jiang, G., & Gao, X. (2024). Intranasal drug delivery: The interaction between nanoparticles and the nose-to-brain pathway. Advanced Drug Delivery Reviews, 207, 115196. [Crossref]

da Cunha, K. F., Albernaz, D. T. F., Garcia, M. de O., Allend, S. O., & Hartwig, D. D. (2023). Silver nanoparticles (AgNPs) in the control of Staphylococcus spp. Letters in Applied Microbiology, 76(1), ovac032. [Crossref]

Dai, J., Wu, M., Wang, Q., Ding, S., Dong, X., Xue, L., Zhu, Q., Zhou, J., Xia, F., Wang, S., & Hong, Y. (2021). Red blood cell membrane-camouflaged nanoparticles loaded with AIEgen and Poly(I: C) for enhanced tumoral photodynamic-immunotherapy. National Science Review, 8(6), nwab039. [Crossref]

Daphedar, A., & Taranath, T. C. (2018). Characterization and cytotoxic effect of biogenic silver nanoparticles on mitotic chromosomes of Drimia polyantha (Blatt. & McCann) Stearn. Toxicology Reports, 5, 910–918. [Crossref]

de Araujo, M. M., Borgheti-Cardoso, L. N., Praça, F. G., Marcato, P. D., & Bentley, M. V. L. B. (2023). Solid Lipid–Polymer Hybrid Nanoplatform for Topical Delivery of siRNA: In Vitro Biological Activity and Permeation Studies. Journal of Functional Biomaterials, 14(7), 374. [Crossref]

Dighe, S., Jog, S., Momin, M., Sawarkar, S., & Omri, A. (2024). Intranasal Drug Delivery by Nanotechnology: Advances in and Challenges for Alzheimer’s Disease Management. Pharmaceutics, 16(1), Article 1. [Crossref]

Dilshad, E., Bibi, M., Sheikh, N., Tamrin, K., Mansoor, Q., Maqbool, Q., & Nawaz, M. (2020). Synthesis of Functional Silver Nanoparticles and Microparticles with Modifiers and Evaluation of Their Antimicrobial, Anticancer, and Antioxidant Activity. Journal of Functional Biomaterials, 11, 76. [Crossref]

Dove, A. S., Dzurny, D. I., Dees, W. R., Qin, N., Nunez Rodriguez, C. C., Alt, L. A., Ellward, G. L., Best, J. A., Rudawski, N. G., Fujii, K., & Czyż, D. M. (2023). Silver nanoparticles enhance the efficacy of aminoglycosides against antibiotic-resistant bacteria. Frontiers in Microbiology, 13. www.frontiersin.org

Dryden, M. (2018). Reactive oxygen species: A novel antimicrobial. International Journal of Antimicrobial Agents, 51(3), 299–303. [Crossref]

Du, H., Wang, X., Zhang, H., Chen, H., Deng, X., He, Y., Tang, H., Deng, F., & Ren, Z. (2023). Serum protein coating enhances the antisepsis efficacy of silver nanoparticles against multidrug-resistant Escherichia coli infections in mice. Frontiers in Microbiology, 14. www.frontiersin.org

Dutt, Y., Pandey, R. P., Dutt, M., Gupta, A., Vibhuti, A., Raj, V. S., Chang, C.-M., & Priyadarshini, A. (2023). Silver Nanoparticles Phytofabricated through Azadirachta indica: Anticancer, Apoptotic, and Wound-Healing Properties. Antibiotics, 12(1), 121. [Crossref]

Edis, Z., Wang, J., Waqas, M. K., Ijaz, M., & Ijaz, M. (2021). Nanocarriers-Mediated Drug Delivery Systems for Anticancer Agents: An Overview and Perspectives. International Journal of Nanomedicine, 16, 1313–1330. [Crossref]

Ehsan, B., Haque, A., Qasim, M., Ali, A., & Sarwar, Y. (2023). High prevalence of extensively drug resistant and extended spectrum beta lactamases (ESBLs) producing uropathogenic Escherichia coli isolated from Faisalabad, Pakistan. World Journal of Microbiology & Biotechnology, 39(5), 132. [Crossref]

El-Ansary, A. E., Omran, A. A. A., Mohamed, H. I., & El-Mahdy, O. M. (2023). Green synthesized silver nanoparticles mediated by Fusarium nygamai isolate AJTYC1: Characterizations, antioxidant, antimicrobial, anticancer, and photocatalytic activities and cytogenetic effects. Environmental Science and Pollution Research. [Crossref]

Elmowafy, M., Shalaby, K., Elkomy, M. H., Alsaidan, O. A., Gomaa, H. A. M., Abdelgawad, M. A., & Mostafa, E. M. (2023). Polymeric Nanoparticles for Delivery of Natural Bioactive Agents: Recent Advances and Challenges. Polymers, 15(5), Article 5. [Crossref]

Elumalai, K., Srinivasan, S., & Shanmugam, A. (2024). Review of the efficacy of nanoparticle-based drug delivery systems for cancer treatment. Biomedical Technology, 5, 109–122. [Crossref]

Endale, H. T., Tesfaye, W., & Mengstie, T. A. (2023). ROS induced lipid peroxidation and their role in ferroptosis. Frontiers in Cell and Developmental Biology, 11. www.frontiersin.org

Ernest, V., Gajalakshmi, S., Mukherjee, A., & Chandrasekaran, N. (2014a). Enhanced activity of lysozyme-AgNP conjugate with synergic antibacterial effect without damaging the catalytic site of lysozyme. Artificial Cells, Nanomedicine, and Biotechnology, 42(5), 336–343. [Crossref]

Ernest, V., Gajalakshmi, S., Mukherjee, A., & Chandrasekaran, N. (2014b). Enhanced activity of lysozyme-AgNP conjugate with synergic antibacterial effect without damaging the catalytic site of lysozyme. Artificial Cells, Nanomedicine and Biotechnology, 42(5), 336–343. [Crossref]

Faúndez, X., Báez, M. E., Martínez, J., Zúñiga-López, M. C., Espinoza, J., & Fuentes, E. (2023). Evaluation of the generation of reactive oxygen species and antibacterial activity of honey as a function of its phenolic and mineral composition. Food Chemistry, 426, 136561. [Crossref]

Ferdous, Z., & Nemmar, A. (2020). Health Impact of Silver Nanoparticles: A Review of the Biodistribution and Toxicity Following Various Routes of Exposure. International Journal of Molecular Sciences, 21(7), Article 7. [Crossref]

Fernandes, M., González-Ballesteros, N., da Costa, A., Machado, R., Gomes, A. C., & Rodríguez-Argüelles, M. C. (2023). Antimicrobial and anti-biofilm activity of silver nanoparticles biosynthesized with Cystoseira algae extracts. JBIC Journal of Biological Inorganic Chemistry, 28(4), 439–450. [Crossref]

Fontoura, I., Veriato, T. S., Raniero, L. J., & Castilho, M. L. (2023). Analysis of Capped Silver Nanoparticles Combined with Imipenem against Different Susceptibility Profiles of Klebsiella pneumoniae. Antibiotics, 12(3), Article 3. [Crossref]

Frei, A., Verderosa, A. D., Elliott, A. G., Zuegg, J., & Blaskovich, M. A. T. (2023). Metals to combat antimicrobial resistance. Nature Reviews Chemistry, 7(3), 202–224. [Crossref]

Guo, H., Zhang, J., Boudreau, M., Meng, J., Yin, J., Liu, J., & Xu, H. (2016). Intravenous administration of silver nanoparticles causes organ toxicity through intracellular ROS-related loss of inter-endothelial junction. Particle and Fibre Toxicology, 13. [Crossref]

Habibi, N., Mauser, A., Ko, Y., & Lahann, J. (2022). Protein Nanoparticles: Uniting the Power of Proteins with Engineering Design Approaches. Advanced Science, 9(8), 2104012. [Crossref]

Halkai, K. R., Mudda, J. A., Shivanna, V., Patil, V., Rathod, V., & Halkai, R. (2019). Cytotoxicity evaluation of fungal-derived silver nanoparticles on human gingival fibroblast cell line: An in vitro study. Journal of Conservative Dentistry : JCD, 22(2), 160–163. [Crossref]

Hublikar, L. V., Ganachari, S. V., Patil, V. B., Nandi, S., & Honnad, A. (2023). Anticancer potential of biologically synthesized silver nanoparticles using Lantana camara leaf extract. Progress in Biomaterials, 12, 155–169. [Crossref]

Jain, A., Garrett, N. T., & Malone, Z. P. (2022). Ruthenium‐based Photoactive Metalloantibiotics . Photochemistry and Photobiology, 98(1), 6–16. [Crossref]

Jayachandran, P., Ilango, S., Suseela, V., Nirmaladevi, R., Shaik, M. R., Khan, M., Khan, M., & Shaik, B. (2023). Green Synthesized Silver Nanoparticle-Loaded Liposome-Based Nanoarchitectonics for Cancer Management: In Vitro Drug Release Analysis. Biomedicines, 11(1), 217. [Crossref]

Kadir, N. H. A., Khan, A. A., Kumaresan, T., Khan, A. U., & Alam, M. (2024). The impact of pumpkin seed-derived silver nanoparticles on corrosion and cytotoxicity: A molecular docking study of the simulated AgNPs. Green Chemistry Letters and Reviews. https://www.tandfonline.com/doi/abs/10.1080/17518253.2024.2319246

Kah, G., Chandran, R., & Abrahamse, H. (2023). Biogenic Silver Nanoparticles for Targeted Cancer Therapy and Enhancing Photodynamic Therapy. Cells, 12(15), 2012. [Crossref]

Kakian, F., Mirzaei, E., Moattari, A., Takallu, S., & Bazargani, A. (2024). Determining the cytotoxicity of the Minimum Inhibitory Concentration (MIC) of silver and zinc oxide nanoparticles in ESBL and carbapenemase producing Proteus mirabilis isolated from clinical samples in Shiraz, Southwest Iran. BMC Research Notes, 17(1), 40. [Crossref]

Kanimozhi, S., Durga, R., Sabithasree, M., Kumar, A. V., Sofiavizhimalar, A., Kadam, A. A., Rajagopal, R., Sathya, R., & Azelee, N. I. W. (2022). Biogenic synthesis of silver nanoparticle using Cissus quadrangularis extract and its invitro study. Journal of King Saud University - Science, 34(4), 101930. [Crossref]

Kaukab, A., Gaur, S., Agnihotri, R., & Taneja, V. (2023). Silver Nanoparticles as an Intracanal Medicament: A Scoping Review. The Scientific World Journal, 2023, 9451685. [Crossref]

Kim, S.-M., Choi, H.-J., Lim, J.-A., Woo, M.-A., Chang, H.-J., Lee, N., & Lim, M.-C. (2023). Biosynthesis of Silver Nanoparticles from Duchesnea indica Extracts Using Different Solvents and Their Antibacterial Activity. Microorganisms, 11(6), Article 6. [Crossref]

Klein, W., Ismail, E., Maboza, E., Hussein, A. A., & Adam, R. Z. (2023). Green-Synthesized Silver Nanoparticles: Antifungal and Cytotoxic Potential for Further Dental Applications. Journal of Functional Biomaterials, 14(7), 379. [Crossref]

Kora, A. J., & Sashidhar, R. B. (2015). Antibacterial activity of biogenic silver nanoparticles synthesized with gum ghatti and gum olibanum: A comparative study. The Journal of Antibiotics, 68(2), Article 2. [Crossref]

Kumar, L., Bisen, M., Harjai, K., Chhibber, S., Azizov, S., Lalhlenmawia, H., & Kumar, D. (2023). Advances in Nanotechnology for Biofilm Inhibition. ACS Omega, 8(24), 21391–21409. [Crossref]

Lan, Y., Zhou, M., Li, X., Liu, X., Li, J., & Liu, W. (2022). Preliminary Investigation of Iron Acquisition in Hypervirulent Klebsiella pneumoniae Mediated by Outer Membrane Vesicles. Infection and Drug Resistance, 15. [Crossref]

Li, H., Zhou, X., Huang, Y., Liao, B., Cheng, L., & Ren, B. (2021). Reactive Oxygen Species in Pathogen Clearance: The Killing Mechanisms, the Adaption Response, and the Side Effects. Frontiers in Microbiology, 11. www.frontiersin.org

Li, J., Zhang, B., Chang, X., Gan, J., Li, W., Niu, S., Kong, L., Wu, T., Zhang, T., Tang, M., & Xue, Y. (2020). Silver nanoparticles modulate mitochondrial dynamics and biogenesis in HepG2 cells. Environmental Pollution, 256, 113430. [Crossref]

Li, N., Zhu, S., Wen, C., Xu, H., Li, C., Zhu, S., Li, R., Chen, L., & Luo, X. (2023). The uptake, elimination, and toxicity of silver nanoparticles and silver ions in single-species and natural mixed-species bacterial biofilms. Journal of Water Process Engineering, 56, 104256. [Crossref]

Liao, C., Li, Y., & Tjong, S. C. (2019). Bactericidal and Cytotoxic Properties of Silver Nanoparticles. International Journal of Molecular Sciences, 20(2). [Crossref]

Lohans, C. T., Freeman, E. I., Groesen, E. van, Tooke, C. L., Hinchliffe, P., Spencer, J., Brem, J., & Schofield, C. J. (2019). Mechanistic Insights into β-Lactamase-Catalysed Carbapenem Degradation Through Product Characterisation. Scientific Reports, 9(1), Article 1. [Crossref]

Lotfipour, F., Shahi, S., Farjami, A., Salatin, S., Mahmoudian, M., & Dizaj, S. M. (2021). Safety and Toxicity Issues of Therapeutically Used Nanoparticles from the Oral Route. BioMed Research International, 2021, 9322282. [Crossref]

Mahmod, W. S., & Al-Jumaili, E. F. (2022). Pharmaceutical Nano-Delivery Systems: A Review. Indian Journal of Forensic Medicine & Toxicology, 16(2), 433–439. [Crossref]

Mali, R., & Patil, J. (2023). Nanoparticles: A Novel Antifungal Drug Delivery System. Materials Proceedings, 14(1), Article 1. [Crossref]

Malik, S., Muhammad, K., & Waheed, Y. (2023a). Emerging Applications of Nanotechnology in Healthcare and Medicine. Molecules, 28(18), 6624. [Crossref]

Malik, S., Muhammad, K., & Waheed, Y. (2023b). Nanotechnology: A Revolution in Modern Industry. Molecules, 28(2), 661. [Crossref]

Mammari, N., Lamouroux, E., Boudier, A., & Duval, R. E. (2022). Current Knowledge on the Oxidative-Stress-Mediated Antimicrobial Properties of Metal-Based Nanoparticles. Microorganisms, 10(2), Article 2. [Crossref]

Manivasagan, P., Venkatesan, J., Senthilkumar, K., Sivakumar, K., & Kim, S.-K. (2013). Biosynthesis, Antimicrobial and Cytotoxic Effect of Silver Nanoparticles Using a Novel Nocardiopsis sp. MBRC-1. BioMed Research International, 2013, e287638. [Crossref]

Mao, B.-H., Chen, Z.-Y., Wang, Y.-J., & Yan, S.-J. (2018). Silver nanoparticles have lethal and sublethal adverse effects on development and longevity by inducing ROS-mediated stress responses. Scientific Reports, 8, 2445. [Crossref]

Mathur, P., Jha, S., Ramteke, S., & Jain, N. K. (2018). Pharmaceutical aspects of silver nanoparticles. Artificial Cells, Nanomedicine, and Biotechnology, 46(sup1), 115–126. [Crossref]

Mauri, E., & Scialla, S. (2023). Nanogels Based on Hyaluronic Acid as Potential Active Carriers for Dermatological and Cosmetic Applications. Cosmetics, 10(4), Article 4. [Crossref]

McNeilly, O., Mann, R., Hamidian, M., & Gunawan, C. (2021). Emerging Concern for Silver Nanoparticle Resistance in Acinetobacter baumannii and Other Bacteria. Frontiers in Microbiology, 12. www.frontiersin.org

Meesaragandla, B., Hayet, S., Fine, T., Janke, U., Chai, L., & Delcea, M. (2022). Inhibitory Effect of Epigallocatechin Gallate-Silver Nanoparticles and Their Lysozyme Bioconjugates on Biofilm Formation and Cytotoxicity. ACS Applied Bio Materials, 5(9), 4213–4221. [Crossref]

Mikhailova, E. O. (2020). Silver Nanoparticles: Mechanism of Action and Probable Bio-Application. Journal of Functional Biomaterials, 11(4), 84. [Crossref]

Modi, S. K., Gaur, S., Sengupta, M., & Singh, M. S. (2023). Mechanistic insights into nanoparticle surface-bacterial membrane interactions in overcoming antibiotic resistance. Frontiers in Microbiology, 14. www.frontiersin.org

More, P. R., Pandit, S., Filippis, A. D., Franci, G., Mijakovic, I., & Galdiero, M. (2023a). Silver Nanoparticles: Bactericidal and Mechanistic Approach against Drug Resistant Pathogens. Microorganisms, 11(2). [Crossref]

More, P. R., Pandit, S., Filippis, A. D., Franci, G., Mijakovic, I., & Galdiero, M. (2023b). Silver Nanoparticles: Bactericidal and Mechanistic Approach against Drug Resistant Pathogens. Microorganisms, 11(2), Article 2. [Crossref]

Nakamura, Y., Mochida, A., Choyke, P. L., & Kobayashi, H. (2016). Nano-drug delivery: Is the enhanced permeability and retention (EPR) effect sufficient for curing cancer? Bioconjugate Chemistry, 27(10), 2225–2238. [Crossref]

Nallanthighal, S., Chan, C., Murray, T. M., Mosier, A. P., Cady, N. C., & Reliene, R. (2017). Differential effects of silver nanoparticles on DNA damage and DNA repair gene expression in Ogg1-deficient and wild type mice. Nanotoxicology, 11(8), 996–1011. [Crossref]

Nandhini, S. N., Sisubalan, N., Vijayan, A., Karthikeyan, C., Gnanaraj, M., Gideon, D. A. M., Jebastin, T., Varaprasad, K., & Sadiku, R. (2023). Recent advances in green synthesized nanoparticles for bactericidal and wound healing applications. Heliyon, 9(2), e13128. [Crossref]

Nikolova, S., Milusheva, M., Gledacheva, V., Feizi-Dehnayebi, M., Kaynarova, L., Georgieva, D., Delchev, V., Stefanova, I., Tumbarski, Y., Mihaylova, R., Cherneva, E., Stoencheva, S., & Todorova, M. (2023). Drug-Delivery Silver Nanoparticles: A New Perspective for Phenindione as an Anticoagulant. Biomedicines, 11(8), Article 8. [Crossref]

Noga, M., Milan, J., Frydrych, A., & Jurowski, K. (2023). Toxicological Aspects, Safety Assessment, and Green Toxicology of Silver Nanoparticles (AgNPs)—Critical Review: State of the Art. International Journal of Molecular Sciences, 24(6), Article 6. [Crossref]

Olugbodi, J. O., Lawal, B., Bako, G., Onikanni, A. S., Abolenin, S. M., Mohammud, S. S., Ataya, F. S., & Batiha, G. E.-S. (2023). Effect of sub-dermal exposure of silver nanoparticles on hepatic, renal and cardiac functions accompanying oxidative damage in male Wistar rats. Scientific Reports, 13(1), Article 1. [Crossref]

Ozdal, M., & Gurkok, S. (2022). Recent advances in nanoparticles as antibacterial agent. ADMET & DMPK, 10(2), 115. [Crossref]

Paladini, F., & Pollini, M. (2019). Antimicrobial Silver Nanoparticles for Wound Healing Application: Progress and Future Trends. Materials, 12(16), 2540. [Crossref]

Pareek, V., Devineau, S., Sivasankaran, S. K., Bhargava, A., Panwar, J., Srikumar, S., & Fanning, S. (2021). Silver Nanoparticles Induce a Triclosan-Like Antibacterial Action Mechanism in Multi-Drug Resistant Klebsiella pneumoniae. Frontiers in Microbiology, 12, 638640. [Crossref]

Park, H.-Y., Chung, C., Eiken, M. K., Baumgartner, K. V., Fahy, K. M., Leung, K. Q., Bouzos, E., Asuri, P., Wheeler, K. E., & Riley, K. R. (2023). Silver nanoparticle interactions with glycated and non-glycated human serum albumin mediate toxicity. Frontiers in Toxicology, 5. www.frontiersin.org

Parveen, A., Kulkarni, N., Yalagatti, M., Abbaraju, V., & Deshpande, R. (2018). In vivo efficacy of biocompatible silver nanoparticles cream for empirical wound healing. Journal of Tissue Viability, 27(4), 257–261. [Crossref]

Rajivgandhi, G., Chelliah, C. K., Ramachandran, G., Chackaravarthi, G., Maruthupandy, M., Alharbi, N. S., Kadaikunnan, S., Natesan, M., Li, W.-J., & Quero, F. (2023). Morphological modification of silver nanoparticles against multi-drug resistant gram-negative bacteria and cytotoxicity effect in A549 lung cancer cells through in vitro approaches. Archives of Microbiology, 205(8), 282. [Crossref]

Rajora, N., Kaushik, S., Jyoti, A., & Kothari, S. L. (2016). Rapid synthesis of silver nanoparticles by Pseudomonas stutzeri isolated from textile soil under optimised conditions and evaluation of their antimicrobial and cytotoxicity properties. IET Nanobiotechnology, 10(6), 367–373. [Crossref]

Rybka, M., Mazurek, Ł., & Konop, M. (2022). Beneficial Effect of Wound Dressings Containing Silver and Silver Nanoparticles in Wound Healing—From Experimental Studies to Clinical Practice. Life, 13(1), 69. [Crossref]

Saghafi, Y., Baharifar, H., Najmoddin, N., Asefnejad, A., Maleki, H., Sajjadi-Jazi, S. M., Bonkdar, A., Shams, F., & Khoshnevisan, K. (2023). Bromelain- and Silver Nanoparticle-Loaded Polycaprolactone/Chitosan Nanofibrous Dressings for Skin Wound Healing. Gels, 9(8), Article 8. [Crossref]

Sahoo, B., Panigrahi, L. L., Jena, S., Jha, S., & Arakha, M. (2023). Oxidative stress generated due to photocatalytic activity of biosynthesized selenium nanoparticles triggers cytoplasmic leakage leading to bacterial cell death. RSC Advances, 13(17), 11406–11414. [Crossref]

Scandorieiro, S., Teixeira, F. M. M. B., Nogueira, M. C. L., Panagio, L. A., de Oliveira, A. G., Durán, N., Nakazato, G., & Kobayashi, R. K. T. (2023). Antibiofilm Effect of Biogenic Silver Nanoparticles Combined with Oregano Derivatives against Carbapenem-Resistant Klebsiella pneumoniae. Antibiotics, 12(4), Article 4. [Crossref]

Selem, E., Mekky, A. F., Hassanein, W. A., Reda, F. M., & Selim, Y. A. (2022). Antibacterial and antibiofilm effects of silver nanoparticles against the uropathogen Escherichia coli U12. Saudi Journal of Biological Sciences, 29(11), 103457. [Crossref]

Shi, T., Sun, X., & He, Q.-Y. (2018). Cytotoxicity of Silver Nanoparticles Against Bacteria and Tumor Cells. Current Protein & Peptide Science, 19(6), 525–536. [Crossref]

Shukla, R. K., Badiye, A., Vajpayee, K., & Kapoor, N. (2021). Genotoxic Potential of Nanoparticles: Structural and Functional Modifications in DNA. Frontiers in Genetics, 12, 728250. [Crossref]

Siddique, M. H., Aslam, B., Imran, M., Ashraf, A., Nadeem, H., Hayat, S., Khurshid, M., Afzal, M., Malik, I. R., Shahzad, M., Qureshi, U., Khan, Z. U. H., & Muzammil, S. (2020). Effect of Silver Nanoparticles on Biofilm Formation and EPS Production of Multidrug-Resistant Klebsiella pneumoniae. BioMed Research International, 2020, e6398165. [Crossref]

Sultana, A., Zare, M., Thomas, V., Kumar, T. S. S., & Ramakrishna, S. (2022). Nano-based drug delivery systems: Conventional drug delivery routes, recent developments and future prospects. Medicine in Drug Discovery, 15, 100134. [Crossref]

Takáč, P., Michalková, R., Čižmáriková, M., Bedlovičová, Z., Balážová, Ľ., & Takáčová, G. (2023). The Role of Silver Nanoparticles in the Diagnosis and Treatment of Cancer: Are There Any Perspectives for the Future? Life, 13(2), 466. [Crossref]

Tiwari, H., Rai, N., Singh, S., Gupta, P., Verma, A., Singh, A. K., Kajal, Salvi, P., Singh, S. K., & Gautam, V. (2023). Recent Advances in Nanomaterials-Based Targeted Drug Delivery for Preclinical Cancer Diagnosis and Therapeutics. Bioengineering, 10(7), 760. [Crossref]

Todorova, M., Milusheva, M., Kaynarova, L., Georgieva, D., Delchev, V., Simeonova, S., Pilicheva, B., & Nikolova, S. (2023). Drug-Loaded Silver Nanoparticles—A Tool for Delivery of a Mebeverine Precursor in Inflammatory Bowel Diseases Treatment. Biomedicines, 11(6), Article 6. [Crossref]

Tverezovska, O., Holubnycha, V., Banasiuk, R., Husak, Y., Anton, S., & Korniienko, V. (2022). The Effect of Silver Nanoparticles Against Formation of Enterococcus Faecalis Biofilms. 2022 IEEE 12th International Conference Nanomaterials: Applications & Properties (NAP), NRA12-1-NRA12-5. [Crossref]

Veerapandian, M., Ramasundaram, S., Jerome, P., Chellasamy, G., Govindaraju, S., Yun, K., & Oh, T. H. (2023). Drug Delivery Application of Functional Nanomaterials Synthesized Using Natural Sources. Journal of Functional Biomaterials, 14(8), 426. [Crossref]

Veriato, T. S., Fontoura, I., Oliveira, L. D., Raniero, L. J., & Castilho, M. L. (2023). Nano-antibiotic based on silver nanoparticles functionalized to the vancomycin–cysteamine complex for treating Staphylococcus aureus and Enterococcus faecalis. Pharmacological Reports, 75(4), 951–961. [Crossref]

Wasilewska, A., Klekotka, U., Zambrzycka, M., Zambrowski, G., Święcicka, I., & Kalska-Szostko, B. (2023). Physico-chemical properties and antimicrobial activity of silver nanoparticles fabricated by green synthesis. Food Chemistry, 400, 133960. [Crossref]

Wieler, L., Vittos, O., Mukherjee, N., & Sarkar, S. (2023). Reduction in the COVID-19 pneumonia case fatality rate by silver nanoparticles: A randomized case study. Heliyon, 9(3), e14419. [Crossref]

Xu, Y., Li, H., Li, X., & Liu, W. (2023). What happens when nanoparticles encounter bacterial antibiotic resistance? Science of The Total Environment, 876, 162856. [Crossref]

Yao, C., Zhang, D., Wang, H., & Zhang, P. (2023). Recent Advances in Cell Membrane Coated-Nanoparticles as Drug Delivery Systems for Tackling Urological Diseases. Pharmaceutics, 15(7), Article 7. [Crossref]

Yeh, Y.-C., Huang, T.-H., Yang, S.-C., Chen, C.-C., & Fang, J.-Y. (2020). Nano-Based Drug Delivery or Targeting to Eradicate Bacteria for Infection Mitigation: A Review of Recent Advances. Frontiers in Chemistry, 8. www.frontiersin.org

Yin, I. X., Zhang, J., Zhao, I. S., Mei, M. L., Li, Q., & Chu, C. H. (2020). The Antibacterial Mechanism of Silver Nanoparticles and Its Application in Dentistry. International Journal of Nanomedicine, 15, 2555–2562. [Crossref]

Zachar, O. (2020). Formulations for COVID-19 Early Stage Treatment via Silver Nanoparticles Inhalation Delivery at Home and Hospital. ScienceOpen Preprints. [Crossref]

Zachar, O. (2022). Nanomedicine formulations for respiratory infections by inhalation delivery: Covid-19 and beyond. Medical Hypotheses, 159, 110753. [Crossref]