Next Article in Journal
Biodiesel Production Using Wild Apricot (Prunus aitchisonii) Seed Oil via Heterogeneous Catalysts
Next Article in Special Issue
Bimetallic Au–Ag Nanoparticles: Advanced Nanotechnology for Tackling Antimicrobial Resistance
Previous Article in Journal
Tungsten-Based Nanocatalysts: Research Progress and Future Prospects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biological Synthesis of Silver Nanoparticles and Prospects in Plant Disease Management

1
Department of Botany, Lords University, Alwar 301028, India
2
Section of Plant Pathology and Nematology, Aligarh Muslim University, Aligarh 202002, India
3
School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2022, 27(15), 4754; https://doi.org/10.3390/molecules27154754
Submission received: 26 May 2022 / Revised: 18 July 2022 / Accepted: 22 July 2022 / Published: 25 July 2022

Abstract

:
Exploration of nanoparticles (NPs) for various biological and environmental applications has become one of the most important attributes of nanotechnology. Due to remarkable physicochemical properties, silver nanoparticles (AgNPs) are the most explored and used NPs in wide-ranging applications. Also, they have proven to be of high commercial use since they possess great chemical stability, conductivity, catalytic activity, and antimicrobial potential. Though several methods including chemical and physical methods have been devised, biological approaches using organisms such as bacteria, fungi, and plants have emerged as economical, safe, and effective alternatives for the biosynthesis of AgNPs. Recent studies highlight the potential of AgNPs in modern agricultural practices to control the growth and spread of infectious pathogenic microorganisms since the introduction of AgNPs effectively reduces plant diseases caused by a spectrum of bacteria and fungi. In this review, we highlight the biosynthesis of AgNPs and discuss their applications in plant disease management with recent examples. It is proposed that AgNPs are prospective NPs for the successful inhibition of pathogen growth and plant disease management. This review gives a better understanding of new biological approaches for AgNP synthesis and modes of their optimized applications that could contribute to sustainable agriculture.

1. Introduction

Phytopathogenic microbes and plant pests are natural habitants of the plant’s surrounding environment and cause huge crop losses with a worldwide estimated reduction in crop production of 20–40% on an annual basis [1]. Applications of pesticides including fungicides and insecticides are currently being used [2] on a very large scale and are considered a key to pest management. The use of chemical pesticides is a rapid killing strategy against pathogenic microorganisms; however, their overuse and long-term persistence in soils unwantedly reduce soil fertility [3] and disrupt the natural micro and macro soil biota. Among them, many plant-beneficial microbes are either killed or their natural ecology is compromised. This necessitates the development of alternative strategies to control plant diseases. In this regard, nanotechnology seems to be an alternative; it has also progressed significantly in the field of medicine, e.g., as alternative antimicrobials [4,5,6] and pharmacology, e.g., for drug delivery. However, it has attracted relatively less support in precision agriculture [7]. We have seen significant progress in fabrication of NPs with unique functions and dimension-based physicochemical characteristics [8]. Owing to their astonishing properties, NPs with sizes 20–100 nm have received greater attention [9]. The sector of nano-enabled products tries, at their scale, to re-evaluate the biomass assets of previously reported anti-pathogenic substances to alter or inhibit the deadly effects of plant pathogens. Appropriate stimulants seem to be NPs with polymers, metals, or porcelain that can show different biological use depending on their intensification.
NPs play a pivotal role in crop development and improving the nutritive benefits in the agricultural field. The NPs can be used as components of novel fertilizers in agriculture [10], substances for crop protection [11], herbicides [12], and preparations for prolonging cut flower durability [13]. NPs and nanomaterials have recently been proposed as potential biostimulators that could help plants propagate and grow faster [14,15] and develop plant resistance against oxidative stress [16]. The use of NPs could provide several advantages to agriculture and horticulture, but it may pose significant hazards due to their unrecognized environmental consequences [17]. However, detection of optimized concentrations of AgNPs for plant disease management without losing the efficiency and while keeping the non-toxicity of AgNPs to other non-target organisms can be achieved. This can be done through rigorous research on AgNPs selection for particular plant or pathogen species and the local conditions. The development and continuous improvement of AgNPs to regulate plant growth, soil quality, diseases, agrochemical usage, and environmental degradation have substantially strengthened soil quality, crop yield, and ecosystem security, which contribute greatly to sustainable farming and agro-ecosystems [18]. The utilization of agricultural inputs to speed up the site-specific monitored supply of nutrients enhances the crop productivity while assuring the minimization of the use of agri-input. This can be done with the help of engineered AgNPs; that is, with or without other agents that have their physicochemical properties. Along with the help for plant security solutions they offer, agro-nanotechnology has been actively improved, and this may ensure increased plant growth. Indeed, in the current and future crop promotion programs, nanotechnology has incredible potential to control pests and diseases.
Different kinds of metal-based NPs have flourished, including copper (Cu), zinc (Zn), titanium (Ti), magnesium (Mg), gold (Au), and silver (Ag). Variously structured AgNPs have been widely utilized for antimicrobial applications and disease prevention for hundreds of years. The physicochemical and structural heterogeneity of AgNPs is generally achieved by manipulating three important parameters: concentrations of reactants, temperature, and pH [19]. In the majority of studies reporting biological synthesis of AgNPs, silver nitrate (AgNO3) has been used as the source of silver ions, at 1 mM concentration [20,21]. However, some studies have reported the production of smaller-sized AgNPs when moderate [22] to lower [23] concentrations of AgNO3 were used. The temperature is yet another decisive factor that has a significant influence on the time consumed, rate of synthesis, and the stability of the size and properties of AgNPs [24,25]. The pH also control the size and shape of AgNPs, where acidic pH of the reaction medium yields larger-sized AgNPs (Binupriya et al., 2010) than the alkaline pH, as reported for Epicoccum nigrum cell filtrate-mediated synthesis of AgNPs [26]. Another example of pH-induced size variability of AgNPs is Penicillium oxalicum-mediated fabrication of AgNPs over a pH range of 5–13, where alkaline pH (12) produced lower size (4 nm) of AgNPs, consuming less time for synthesis [27]. This suggests higher and longer-term stability in AgNPs, which could be due to the electrostatic repulsion of ions [27]. Similarly, reaction of AgNO3 (1 mM) with Capsicum annum extract at pH 9 showed the maximum yield of biologically capped AgNPs [28].
The antimicrobial actions of AgNPs to plant pathogens can be defined as their properties to hinder or eliminate the growth of plant pathogens without damaging neighboring plant tissues [29]. AgNPs appear to be the most intriguing of metal NPs that exhibit adequate biological activity [30]. They have a wide range of beneficial effects on plants at certain concentrations [31] and arrest the growth of associated pathogenic fungi [32]. AgNPs have been shown to boost germination [33], growth exultation [34], vegetative growth [14], shoot induction, proliferation [14], and pigmentation [35]. Generally, three processes including physical, chemical, and biological processes are employed to prepare AgNPs. Physical and chemical approaches are typically found to be costly and environmentally unfriendly. However, biologically produced AgNPs exhibit great yield, good solubility, and excellent stability for prolonged periods. Out of these three, the biological approach has remained as the simplest, environmentally far less toxic, commercially viable and mostly a single step approach that does not require thermal gradients, pressures, forces, or synthetic chemicals [36]. Biosynthesis of AgNPs through plant parts (i.e., leaf or bark or root or stem, or flower extracts) and microorganisms (i.e., from extra or intracellular molecules of fungi, algae, bacteria) is thus a great tool for simplifying their use in agriculture. This review summarizes the recent progress made in the biosynthesis of AgNPs and discusses the potential of AgNPs in various antibacterial, antifungal, antinematic, and antiviral applications from a plant disease perspective.

2. Biosynthesis of AgNPs

There are two primary approaches for the synthesis of NPs, (i) the top-down approach and (ii) the bottom-up approach, which are graphically presented in Figure 1. In the top-down approach, techniques like mechanical milling, etching, electro explosion, sputtering, and laser ablation are used. The bottom-up approach involves the use of superficial fluid synthesis, plasma spraying synthesis, aerosol process, laser pyrolysis, and green synthesis. In green biological methods, plants and microbial constituents are used as essential bio-reducing agents, which also provide versatility in AgNPs synthesis. Biological methods are considered more effective, inexpensive, sustainable, and environmentally non-toxic compared to chemical and physical methods since the biological materials are easily available and require no or less prior processing [37]. Generally, plant and microbially biosynthesized AgNPs are used as a source that can either be processed extracellularly or intracellularly [38]. Moreover, algae, fungi, and beneficial bacteria that can tolerate toxic materials, e.g., heavy metals and organic chemicals, while retaining the capacity of producing organic and inorganic reducing agents may be the biological solution for AgNPs synthesis.
Based on the available literature, a general mechanism of AgNPs formation and growth can be suggested. Functional groups of biomolecules, such as those from polyphenols, release electrons in the reaction medium that interact with the molar concentrations of metal salt (generally AgNO3), and reduction of silver ions (Ag+) occur. As a result of reduction, particle formation begins and surface plasmon resonance (SPR) increases with a gradual to sharp intensification of dark color of the medium. At some point, the increase in SPR does not increase further and further reduction of Ag+ ions does not occur, which could be due to two major reasons (i) reduction of all available Ag+ ions by reducing agents and/or (ii) lack of adequate electrons in the medium, i.e., low concentration of biomolecules. The reduced atoms of silver (Ag0) then start to form aggregates that could be limited to a certain size if the capping agent is present in the reaction medium; otherwise, smaller aggregates further grow and form larger aggregates. The capping of Ag0 generally involves Van der Waals forces, electrostatic attraction, or covalent bond formation between capping agents and Ag0 atoms, and the AgNPs particle or crystal attains its characteristic size and shape.

2.1. Biological Synthesis of AgNPs by Plant Parts

For the synthesis of AgNPs, plant parts such as leaves, fruits, stems, roots, seeds, and flowers are used due to their abundant quantity and availability, and their biochemical composition is also generally considered non-toxic to humans or other non-target organisms since they are known for producing beneficial phytochemicals. Several plants such as Acacia nilotica, Ocimum sanctum, Coleus aromaticus, Lantana camara, and Moringa oleifera have been used for AgNPs biosynthesis. Plant-derived AgNPs are characterized by various techniques [28], and based on their properties and need, are implemented in agricultural applications (Table 1 and Figure 2). Plants are widely available as the source of many rich metabolites that serve as bio-agents to reduce the Ag+ ions [39]. Biomolecules from several plant species may also be used for the coating of AgNPs in order to reduce the need of chemical reagents and hazardous substances or other approaches for purification [40]. Eucalyptus globulus [19], Azadirachta indica [41], Cissus quadrangularis [42], Lantana camara flower [43], Rubus illecebrosus leaf [44], Nephelium lappaceum peel [45], Ricinus communis [46], Calotropis gigantea are yet other species used in the green synthesis of AgNPs [47]. Reducing and stabilizing agents in plants help shape biocompatible AgNPs. Available secondary compounds including terpenoids, flavonoids, phenols, alkaloids, proteins, and carbohydrates in the extract function as reduction agents [48,49,50,51].

2.2. Biological Synthesis of AgNPs by Bacteria

Biological preparation of AgNPs through microbes has been recognized as a rapidly growing field of research in green nanotechnology. Different microbial entities have constantly been engaged in the synthesis of AgNPs, which are seen as alternatives to the traditional chemical and physical strategies. Many bacterial species such as Serratia nematodiphila, Bacillus stearothermophilus, Escherichia coli, Morganella morganii RP42, Pseudomonas proteolytica, P. meridiana, B. brevis, etc. were used in AgNPs biosynthesis (Table 2 and Figure 2). Moreover, several other microorganisms, including B. methylotrophicus, Brevibacterium frigoritolerans, and Novosphingobium sp., have recently been found effective for the production of AgNPs. Mechanistically, bacteria are responsible for secreting different enzymes in broth culture that include several reductases that perform AgNPs biosynthesis [59]. In the process, bacterial cells act as reducing entities because they extracellularly induce the nucleation and capping of NPs by reducing metal ions (Ag+) to metal NPs (AgNPs) [60]. Bacteria can offer comprehensive opportunities for the synthesis of AgNPs because of their reasonably easy cultivation process and quick replication efficiency. Pseudomonas stutzeri (AG259) was the first bacteria to be used for the synthesis of AgNPs [59]. The AgNPs were also synthesized using Bacillus species like B. megaterium (NCIM 2326), B. cereus, B. licheniformis, B. amyloliquefaciens, B. marisflavi, B. flexus, and B. subtilis [61].

2.3. Biological Synthesis of AgNPs by Algae and Fungi

Among other organisms, algae, a group of marine or freshwater microorganisms, are widely considered as bio-nano factories as both live and dead dried biomasses have been used to synthesize NPs [68]. In addition to algae, fungi also play a significant role in the green synthesis of AgNPs. Algae and fungi serve as reducing agents and function in an environmentally friendly way for sustainable and non-toxic fabrication of AgNPs. The majority of the algae used for this purpose are from the species of Sargassum plagiophyllum, Chlorococcum humicola, Amphora-46, Caulerpa racemose, Padina pavonica, and Chaetomorpha linum etc., and mycelium of fungi, i.e., Aspergillus fumigates, Penicillium fellutanum, A. flavus, Fusarium semitectum, and Alternaria alternata (Table 3 and Figure 2). Fungal molecules based AgNPs are unique over bacteria in terms of providing higher rates of growth of organism, easy cultivation methods and high levels of extracellular enzyme secretions, increased NPs tolerance (protein coating), and excellent stability with high concentrations of NPs [69]. Massive fungal flora have recently had major research involvement in synthesizing suitable biocompatible AgNPs including Trichoderma resei, A. niger, F. oxysporum, and Phytophthora infesatans [70]. For instance, owing to its biologically active substances that have antimicrobial antioxidant, immunomodulating, and anti-cancer properties, Penicillium oxalicum have certain benefits in biogenic synthesis [70]. Other fungi like F. oxysporum, F. semitectum, F. acuminatum, F. solani, Cladosporium cladosporioides, and T. asperellum can also synthesize AgNPs with a broad range of applications [71].

3. Determining the Size, Shape, and Yield of Biological AgNPs

Determining the shape and size of biologically synthesized AgNPs is the most relevant aspect of AgNPs synthesis since it also determines the interfacial reactions with organisms and intracellular penetration of AgNPs. This can be fine-tuned by optimizing the hydrogen ion concentration of the medium. For example, Aloe vera extract mixed at variable pH with solvent resulted in variously sized and shaped Au–Ag core NPs [83]. In most studies, AgNPs have been successfully synthesized at room temperature [84,85], while a few studies also report the control of their shapes by temperature [86]. Regarding shapes, low reaction temperature yielded the formation of nano-triangle AgNPs while high temperature produced spherical AgNPs using Cymbopogon flexuosus extract as reducing and capping agent [87]. When plant biomass is used, some physical and chemical parameters control the crystal growth and structure of NPs [88], for instance, the amount/percentage of the metal salt solution. In fact, for near-perfect control over the size and shape of AgNPs, reaction parameters such as concentrations of metal ions and extracts, duration of the reaction, temperature, and pH are adjusted in a series of different reactions [89,90]. Moreover, the biological production of AgNPs is also governed by enzymatic reaction. For example, in a study, Korbekandi et al. showed pre-incubation of Fusarium oxysporum with 0.1 mM AgNO3 for the activation of reducing enzymes. Additionally, glucose added as electron donor helped in the reduction of AgNPs. The optimized values for four parameters were: 0.1 mM AgNO3 as inducer of enzymes, 5 mM AgNO3 as substrate, amount of biomass (4.96 g dry weight/L), and 56 mM glucose as electron donor. The use of microbes, algae, and plants for biological synthesis of AgNPs is widely known and has been optimized with several species [91]. However, the green synthesis method is faster and more feasible compared to algae mediated or microbial synthesis. This is possibly due to the easy availability of plant materials that do not require maintenance as in the case of microbes. Plant materials are available in plenty, and since plants have a bigger genome size and more complex physiology, the biomolecular composition of plants is far better in terms of their variety and quicker production of AgNPs. AgNPs produced through green routes are generally more biocompatible and stable [92,93].

4. Silver Nanoparticles in Plant Disease Management

4.1. AgNPs in Bacterial Disease Management

The antimicrobial properties of AgNPs are significant for a wide range of microorganisms including bacteria, fungi, and viruses [94]. AgNPs measuring 10–100 nm in size have a strong antibacterial effect against several Gram-positive and Gram-negative bacteria [95]. AgNPs are well-known to be effective antimicrobials against multidrug-resistant (MDR) bacteria, such as methicillin-resistant Staphylococcus aureus, erythromycin-resistant Streptococcus pyogens, ampicillin-resistant E. coli, and vancomycin-resistant S. aureus [96]. In a study, AgNPs prepared using Azadirachta indica extract inhibited the growth of phytopathogens Penicillium sp., Aspergillus sp., Fusarium sp., and Ralstonia solanacearum [32]. Silver ions prevent bacterial DNA replication by linking and causing denaturation of the DNA, which involves silver ion interaction with thiol proteins accompanied by DNA condensation, which leads to apoptosis [97]. The antibacterial activity of AgNPs is largely due to the release of silver cations, which can bind bacterial proteins specifically to thiol (-SH) groups and interrupt their physiological functions [98], leading to necrobiosis. AgNPs carry out their antibacterial activities via a Trojan horse mechanism: their preliminary attachment to the surface of the cell leads to modification and degradation of cell membrane, and release of intracellular metal silver ions exerts toxicity [99]. AgNPs showed pronounced inhibition of growth of E. coli, P. aeruginosa, and S. aureus [100]. The antibacterial effects of AgNPs are mostly due to their broad surface layers, which allow for greater contact between the NPs and microbial cells, suppressing cell development at very low concentrations present in surroundings [101]. The weaker susceptibility to NPs of Gram-negative bacteria (i.e., E. coli) can even be clarified due to the presence of an external lipopolysaccharide (LPS) coating on their surface that restricts the penetration of compounds. However, elevated concentrations of antibacterial agents are therefore required to achieve the same effect on Gram-positive bacteria [102]. AgNPs rely mostly on size, shape, surface functionalization, and ability to display certain antimicrobial properties [103]. The bactericidal impact is also thought to result from the AgNP-mediated generation of reactive oxygen species (ROS) [104]. Two potential AgNP toxicity mechanisms have been explored [105]. One is that AgNPs have a large bacterial surface contact that can enable the cellular membranes to bind to the particles and penetrate effectively into the bacteria. Another mechanism is the interference of AgNPs or dissolved ions with the respiratory chain of cell membrane. Much evidence demonstrates that the antibacterial activities of AgNPs include silver ions [106,107].
To date, some processes have been demonstrated, including protein disruption [108], ROS production and antioxidant degradation, membrane disability work (e.g., membrane disruption, lack of membrane permeability), mutagenicity, and transport protein gene expression downregulation [109]. Membrane fluidity and metabolic activities are disrupted as a result of the morphological changes of cellular membranes impacted by AgNPs and membrane depolarization, which eventually leads to cell component damage and even death. The leakage of molecules inside the cell, such as enzymes, proteins, metabolites, and DNA, is caused by the breakdown of cellular structures. Furthermore, AgNP can form unusual holes in microbial cell walls, allowing NPs to penetrate extracellular and intracellular areas [110]. TEM may be used to study the behavior of AgNP-induced membrane damage and perforation of cell membranes [98]. In recent studies, TEM shows extensive cell surface disintegration, nuclear and cytoplasmic material leaking, and enlargement of cells [111,112]. Moreover, the antibacterial mechanism of AgNPs can follow a dual mode of action of particles and silver ions [113,114]. Some recent examples of inhibition of growth of bacterial pathogens are listed in Table 4.

4.2. AgNPs in Viral Disease Management

In agriculture, AgNPs can be used as antivirals. Viruses are assumed to be involved in almost 50% of all evolving plant diseases, which seem to be difficult to manage; there is no cure in some situations. Viral infections necessitate appropriate and effective management. The viral multiplication cycle includes viral replication into cells, genomic and viral biosynthesis, viral component assembly, and emission of virus particles from the host cell [126]. AgNP’s potential antiviral mechanisms include interactions with gp120, antagonistic impact on virus attachment to the cell, destruction of virus particles before entry, and interaction of AgNPs with double-stranded DNA [127]. AgNPs can also interact with the polypeptide coats of tomato mosaic virus (ToMV) and potato virus Y (PVY) [128]. Resulting from interactions with envelope glycoproteins, AgNPs directly inactivate TMV, causing immediate damage to TMV shell proteins, agglomeration, and even disintegration. Elbeshehy et al. [129], studied AgNPs fabricated from Bacillus persicus, B. pumilus, and B. licheniformis; the AgNPs had an negative influence on the yellow bean mosaic virus. A post-infection treatment given 24 h after virus exposure inhibited the virus from causing any detrimental manifestations. Plants treated with AgNPs and inoculated simultaneously showed only mild BYMV symptoms, and a 72-h pre-infection treatment had almost no influence on viral concentration or disease severity. When AgNPs were applied on Chenopodium amaranticolor crops 24 h after treatment, they displayed the most substantial antiviral impact against tomato spot wilt virus (TSWV). Plants sprayed after inoculation had a poor infection, but plants treated before inoculation had a weak inhibition [130]. El-shazly et al. [131] reported that potato plants sprayed with AgNPs 24 h after being inoculated with tomato bushy stunt virus (TBSV) showed a decrease in virus concentration and disease percentage [131]. Cymopsis tetragonaloba leaves sprayed with AgNPs and sun hemp rosette virus (SHRV) showed full disease reduction by suppressing virus replication [132]. When compared to individual AgNPs and salicylic acid (SA) treatments, a combination of AgNPs and SA administered before 3 and 7 days of virus infection had a synergistically antiviral impact against TBSV [131]. Because the NPs attached to the virus and hindered viral replication, spraying AgNPs on infected tomatoes for 7 days before treatment with ToMV and PVY reduced disease severity and virus concentration [128].
The expression of antioxidant enzymes POD (peroxidase) and polyphenol peroxidase (PPO) in tomato plants treated with AgNPs and infected with ToMV or PVY increased significantly [128]. Mahfouze et al. [133] discovered that banana plants infected with BBTV and treated with 50 ppm AgNPs did not exhibit any outward symptoms, even though the infection rate was 36%. On the other hand, banana plants treated with 50 ppm AgNPs following viral infection showed a considerable improvement in dry weight and leaf area when compared to BBTV-infected banana plants (viral control). When compared to the healthy control, all plants treated with 50 ppm AgNPs after virus inoculation had considerably higher levels of phenol, proline, and oxidative enzymes. Elbeshehy et al. [129] studied the effect of biosynthesized AgNPs on BYMV-infected fava bean leaves and displayed significant symptoms such as yellow mosaic, mottling, crinkling, size reduction, and deformation, which were not present in non-infected leaves. According to Jain and Kothari [132], AgNPs sprayed over the bean leaves showed 100% suppression of the sun-hemp rosette virus. Cai et al. [134] asserted that antiviral activity of NPs in plants is due not only to inhibition of virus replication, but is also responsible for the stimulation of defense mechanisms in plants that contribute to plant immunity and growth response. For example, AgNPs stimulated a phytohormone cytokinin in Capsicum annuum L. [135]. Tripathi et al. [136] suggested that AgNPs adsorption in plants might hinder apoplastic transportation of virus particles by obstructing cell connections, the plasmodesmata, and preventing the apoplastic movement of liquid and nutrients. Hassan et al. [137] employed RAPD and direct amplification of minisatellite-region DNA (DAMD) approaches to detect DNA modification in olive plants exposed to varying concentrations of silver or selenium nanoparticles. They observed that 5 mg L−1 AgNPs had a positive effect on in vitro performance. However, 10 mg L−1 AgNPs and 2.5 and 5 mg L−1 SeNPs induced changes on genomic DNA level. In addition, RAPD and DAMD assays can be successfully applied as a sensitive method of detecting DNA stability and genotoxicity in olive plants treated with different concentrations of selenium and silver NPs. Mahfouze et al. [133] demonstrated that after BBTV infection, the banana plants sprayed with different concentrations of AgNPs showed a few changes at the genomic DNA level, whereas both random amplified polymorphic DNA (RAPD) and sequence-related amplified polymorphism (SRAP) markers scored nearly the same polymorphism in the banana plants (Table 5).

4.3. AgNPs in Fungal Disease Management

The utilization of AgNPs as an alternative to synthetic fungicides has been a relatively new approach with more efficacy. AgNPs may be used for the control of plant diseases as part of their implementation. A few studies have been performed so far on the implementation of AgNPs against phytopathogenic fungi [140]. The latest anti-fungal agents are often in demand because of the development of resistance in pathogenic fungi [141]. There have been cases of antifungal resistance globally, which favor the emergence of resistant mutants due to frequent applications of agrochemicals that also alters the fungal population [142,143]. AgNPs synthesized using A. flavus inhibited the growth of Trichoderma spp. [144]. The AgNPs also showed antifungal properties against Candida albicans and C. tropicalis [145] and F. oxysporum [146]. Due to enhanced permeability and retention effects, the AgNPs seem to be very attractive and fascinating for diverse antifungal applications. Recently, more work has been directed towards developing safe management strategies, which placed humans and animals at lower risk and concentrate on resolving synthetic fungicide vulnerabilities. Nanotechnology represents a way of developing new antifungal secondary metabolites to reduce the symptoms caused by emerging resistant fungi [147]. AgNPs have several antifungal action mechanisms, involving linkages to DNA phosphates groups [148] and plasma–membrane interactions, that contribute to proton dispersion and cell mortality, sulfhydryl protein and enzyme groups, degradation of the electron transportation chain, and disturbance of membrane proton motive force and phosphate groups [149].
AgNPs have been shown to have a wide range of antifungal properties, including against aggressive fungal infections [150] such as C. albicans, C. tropicalis, C. parapsilosis, C. glabrata [151], Trichophyton rubrum [152], Trichosporona sahii [153], A. niger, Rhizoctonia solani, Curvularia lunata, Colletotrichum sp., and Fusarium sp. [154,155]. DNA impairs the fungal culture’s ability to proliferate after it has been treated with silver crystals (AgCs) [156]; this may result in decreased protein expression in the subunit as well as faulty enzymes and cellular proteins required for ATP generation [157]. Several other plant fungi, such as Alternaria alternate and Trichosporona sahii, have shown similar effects with AgNPs. The production of reactive oxygen species (ROS) can cause NP toxicity [153]. Free radicals can damage cell walls, proteins, lipids, and DNA, among other biological components. Deletions, mutations, single strand rupture, double-strand breakage, and protein crosslinking are all examples of DNA damage [158] (Table 6).

4.4. AgNPs in Nematode Disease Management

Various crops are annually damaged by more than 4100 species of plant pathogenic nematodes [171]. Therefore, the development of new nematicides with lessened side effects on non-target organisms and the environment is critical. Recently, evidence has shown that AgNPs have effective nematicidal potential [172,173]. According to different studies, AgNPs can decrease the nematode infestation through the pretreatment of plant seedlings. Researchers also suggest that AgNPs have a direct toxic effect on nematodes and may result in reproduction inhibition [174]. For this to occur, generation of excessive oxidative stress may be an important mechanism in AgNPs toxicity to nematodes [175]. AgNPs exposure in Caenorhabditis elegans decreased reproductive potential and increased ROS formation, expression of the phosphomevalonate kinase (PMK-1/p38), MAPK (Mitogen-Activated Protein Kinases), hypoxia-inducible factor (HIF-1), and glutathione S-transferase (GST) enzyme activity [174]. Degradation in the cell wall of Meloidogyne incognita second-stage larvae (J2) was also observed due to AgNP toxicity under lab conditions [176].
A study on micro-crystalline cellulose-embedded AgNPs reported that a dose of 20–40 ppm caused deflection during the growth stages and influenced embryogenesis eggs’ hatchability and survival in J2 of M. incognita [173]. In another study, AgNPs synthesized from Artemisia Judaica extracts caused an increase in juvenile mortality and egg hatch inhibition in comparison to crude extracts [177]. Furthermore, AgNPs showed antinematic properties against M. graminicola in rice without adverse effects on seed germination or plant growth [172]. The anti-nematode primary mode of action of produced AgB-NPs inhibits several physiological pathways in nematode cells such as membrane permeability, ATP organization, and reaction to oxidative stress [174,178]. Structural immunization may be significantly affected by different characteristics such as disparities in cell wall composition and the production of phytochemicals and pathogenesis-related proteins [179]. Furthermore, phytoalexin organization is frequently connected to the resulting resistance stages of produced AgB-NPs [179]. As a response to nanoparticle stimuli, some plants produce a wide range of secondary metabolites, including phenol and other phytochemicals. They may be an important part of the plant’s defense mechanism against insects such as root-knot nematodes [180]. Some studies on AgNPs effects on nematodes are summarized in Table 7.

4.5. Mechanisms of AgNPs Mediated Inhibition or Killing of Phytopathogens

Despite plenty of reports on pathogen inhibition by AgNPs, the precise mode of antibacterial/antimicrobial action of AgNPs against phytopathogens is not completely understood. Here, we attempted to gather the available information on common modes of antimicrobial action of AgNPs. The AgNPs show their effects mainly by interacting with the cells as (i) particles and/or (ii) Ag+ ions dissolved from applied AgNPs. Some studies have shown that AgNPs produce excessive intracellular ROS, induce an antioxidant response, and interact with essential macromolecules. For instance, Hajipour et al. [108] reported protein disruption by AgNPs, while Lemire et al. [109] and Xia et al. [153] detected ROS induction by AgNPs and its degradation by cellular antioxidant machinery, which followed destability of membrane, genotoxicity, and inhibition of transporter gene expression. Free radicals (a form of ROS and also a part of free radical chain reaction) disrupts chemical bonding and structure of cell walls and biological molecules, including nucleic acids (i.e., DNA), fats, and proteins. Some of the examples of DNA damage are mutations, deletions, and single/double strand breaks [158]. The toxic outcome of AgNP treatment could bea combination of these effects against phytopathogens, since AgNPs may attack multiple cellular targets at once. This has been explained in many other studies [32,192,193,194]. In the process, AgNPs adhere to the cell membranes through electrostatic attraction, affect the morphology of membranes, and cause membrane depolarization, which makes the membrane more permeable. The AgNPs also inhibit cellular respiration and metabolic activity, leading to direct and/or collateral damage to cell components and induction of cell death. In doing so, the AgNPs overwhelm the microbes and the extensive breakdown of cellular components induces the leakage of essential cell fluids or substances (i.e., essential metabolites, proteins, and enzymes) within and/or outside the cell. The creation holes in membranes and walls of microbes by AgNPs facilitate the release of cell content into intracellular spaces and extracellular environment [110]. This kind of hole or perforation in membrane and on cell surface could be observed by transmission electron microscopy (TEM), and the leakage of cytoplasmic material and swelling was also confirmed by TEM. TEM exhibits severe disruption of cell walls, leakage of nuclear and cytoplasmic material, and swelling [111,112]. Similar effects were observed for the AgNP-treated fungi Trichosporon asahii and Alternaria alternata [153]. In a bacterium Azotobacter vinelandii, AgNPs caused cell membrane damage and were present inside the cells as observed by TEM. These AgNPs induced the formation of hydroxyl radicals in A. vinelandii cells that were detected by electron spin resonance [195]. Summarily, the sequence of events of AgNP interactions with phytopathogens can be described as (i) adherence of AgNPs to bacterial cell surfaces based on their charge and size of NPs, (ii) entry of AgNPs to cell interior or dissolution of Ag+ ions from AgNPs, (iii) ROS generation by AgNPs in the inner membrane, (iv) excessive ROS causing oxidative burst and activating cellular components that further induce damage to membranes, (v) subsequent leakage of DNA and other cellular materials leading to cell death.

5. Biological AgNPs against Plant Pathogens: Possible Implications in Lab to Land Transfer

Several attempts are being made worldwide by nanotechnologists and microbiologist to improve traditional agriculture via the sustainable use of biologically synthesized AgNPs. The control over size, shape, and yield of AgNPs has been made more advanced due to technological innovations in synthesis and characterization processes of AgNPs. The AgNPs may aid the agricultural industry by combating pathogens and avoiding the plant disease incidences in a somewhat regulated and site-specific manner. In general, application of AgNPs to agriculture have a significant potential to improve agricultural productivity [35]. However, the impacts and fate of applied concentrations of AgNPs on agricultural systems as a whole are not fully defined. Therefore, greater public interest in adopting AgNP-based agricultural products for farming and knowledge of their benefits and issues associated with nano-enabled agricultural products will more likely trigger the awareness and acceptability. Since, nanotechnology is gaining impetus at a faster pace, it may become a crucial and most useful component of agriculture industry, the phototoxicity of synthesized AgNPs could be repressed by coating the powdered AgNPs with biocompatible materials such as polyvinyl pyrrole [196]. The toxicity and use cases of Ag-based nanomaterials and AgNPs have been well-established by the Environmental Protection Agency (EPA) [6,197]. In a report, it was claimed that the formulations of >100 pesticides contain AgNPs, which are incorporated as antimicrobial agents. However, there is a lack of information on their trophic transfer and toxicity to humans. Not all types of AgNPs are expected to be toxic. For example, a citrate-coated “colloidal” AgNP showed no photo-, cyto-, or geno-toxicity to human cells, but its “powder” form was toxic [198]. This could be due to the chemical conversion of powder AgNPs silver oxides or release of silver ions. By using biocompatible materials [198], alternatives can be explored and researched to suppress the unwanted toxicity of AgNPs, and this could be accomplished by engineering AgNPs with biocompatible materials during their growth in the media or after the synthesis. Such biocompatible coatings or materials would certainly increase their effects, such as enhanced germination of seeds and plant growth. Sometimes, environmental factors also affect the morphology and physicochemical characteristics of NPs and may show different effects on different plant species in real soil environment or field conditions [199]. Therefore, one type of AgNPs cannot be the solution for all pathogens and safe for all host plant species. Maximum effectiveness from lab setups (controlled hydroponic, pot media, or soil) to land or field environments could only be achieved if one type of AgNP is optimized for use with one plant species, or a group of related species that face a similar pathogen attack and disease symptoms.

6. Environmental Relevance and Possible Toxicity of Biological AgNPs

Despite the well-defined anti-phytopathogenic effects of AgNPs, the harmful impacts or toxicity of AgNPs to non-target organisms or beneficial microbes of the eco-system cannot be denied while they are being used to regulate growth of causal organisms of plant diseases [11,98]. The interaction of AgNPs with organisms may vastly vary in each method of application, i.e., either foliar or soil application. Though the amount of AgNPs applied is low, there is the possibility that they may disperse from the agricultural soil to water and air. The dispersion modes can be surface run-off by rain or leaching to ground water [200]. Therefore, in agricultural applications of AgNPs, the prior knowledge of AgNPs toxicity to other organisms, especially the beneficial rhizoplane/rhizosphere and phylloplane microflora, should be obtained. Additionally, AgNPs have shown toxicity to some edible crop plants [199]. Therefore, only concentrations of AgNPs that are biologically specific to certain phytopathogens, while being safe for other organisms (especially the host plant), should be used.

7. Conclusions

Several strategies have been adopted over the last decade to improve the biological synthesis of AgNPs. Increased understanding and utilization of biological materials viz. plant parts, bacteria, and fungi have resulted in advantages over conventional approaches. The many reasons for this include eco-acceptability, cost-effectiveness, safety, and pharmaceutical compatibility without the involvement of hazardous chemicals/processes, high pressure, or energy. Biologically synthesized AgNPs provide significant nano-enabled features for broad-spectrum agricultural applications, specifically in plant pathogen inhibition, because they exert antibacterial, antifungal, and antinematic potential. The current review also highlights that AgNPs originating from green routes may also accumulate in the ecological niche; however, their expected risk to the microbiome and plants is variable.
Various green methods have been established for the production of anti-plant pathogen AgNPs with high yield, purity, and control over their shapes and sizes. Further research should be directed towards the selection of threshold concentrations of AgNPs that efficiently minimize disease instance or severity in plants without exhibiting detrimental impact on the non-target organisms and soil fertility. The physicochemical nature of produced AgNPs can be specific to one crop species and local environmental conditions.

Author Contributions

Conceptualization, M.T., K.N.M. and M.A.S.; writing—original draft preparation, M.T., K.N.M., B.A. and M.A.S.; writing—review and editing, B.A., J.L., and M.A.S.; visualization, M.T. and B.A.; supervision, M.A.S.; funding acquisition, J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the National Research Foundation of Korea (NRF) grant funded by the Korean Government (MSIT) (No. 2021R1G1A1094698) and Priority Research Centers Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2014R1A6A1031189).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The author M.T. is thankful to Department of Botany, Lords University, Alwar for providing necessary facilities to carry out this research work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Savary, S.; Ficke, A.; Aubertot, J.-N.; Hollier, C. Crop Losses Due to Diseases and Their Implications for Global Food Production Losses and Food Security. Food Secur. 2012, 4, 519–537. [Google Scholar] [CrossRef]
  2. Shahid, M.; Ahmed, B.; Zaidi, A.; Khan, M.S. Toxicity of Fungicides to: Pisum Sativum: A Study of Oxidative Damage, Growth Suppression, Cellular Death and Morpho-Anatomical Changes. RSC Adv. 2018, 8, 38483–38498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Shahid, M.; Ahmed, B.; Khan, M.S. Evaluation of Microbiological Management Strategy of Herbicide Toxicity to Greengram Plants. Biocatal. Agric. Biotechnol. 2018, 14, 96–108. [Google Scholar] [CrossRef]
  4. Rani, P.; Ahmed, B.; Singh, J.; Kaur, J.; Rawat, M.; Kaur, N.; Matharu, A.S.; AlKahtanie, M.; Alhomaidif, E.A.H.; Lee, J. Silver Nanostructures Prepared via Novel Green Approach as an Effective Platform for Biological and Environmental Applications. Saudi J. Biol. Sci. 2022, 29, 103296. [Google Scholar] [CrossRef]
  5. Kaur, K.; Ahmed, B.; Singh, J.; Rawat, M.; Kaur, G.; AlKahtani, M.; Alhomaidi, E.A.H.; Lee, J. Bryonia Laciniosa Linn Mediated Synthesized Green Au NPs for Catalytic and Antimicrobial Applications. J. King Saud Univ. 2022, 34, 102022. [Google Scholar] [CrossRef]
  6. Janani, B.; Syed, A.; Raju, L.L.; Bahkali, A.H.; Al-Rashed, S.; Elgorban, A.M.; Ahmed, B.; Thomas, A.M.; Khan, S.S. Designing Intimate Porous Al2O3 Decorated 2D CdO Nano-Heterojunction as Enhanced White Light Driven Photocatalyst and Antibacterial Agent. J. Alloys Compd. 2022, 896, 162807. [Google Scholar] [CrossRef]
  7. Sinha, K.; Ghosh, J.; Sil, P.C. New Pesticides: A Cutting-Edge View of Contributions from Nanotechnology for the Development of Sustainable Agricultural Pest Control. In New Pesticides and Soil Sensors; Elsevier: Amsterdam, The Netherlands, 2017; pp. 47–79. [Google Scholar]
  8. Arya, S.S.; Sharma, M.M.; Das, R.K.; Rookes, J.; Cahill, D.; Lenka, S.K. Vanillin Mediated Green Synthesis and Application of Gold Nanoparticles for Reversal of Antimicrobial Resistance in Pseudomonas Aeruginosa Clinical Isolates. Heliyon 2019, 5, e02021. [Google Scholar] [CrossRef] [Green Version]
  9. Jiang, B.; Li, C.; Dag, Ö.; Abe, H.; Takei, T.; Imai, T.; Hossain, M.; Shahriar, A.; Islam, M.; Wood, K. Mesoporous Metallic Rhodium Nanoparticles. Nat. Commun. 2017, 8, 15581. [Google Scholar] [CrossRef]
  10. Elmer, W.H.; White, J.C. The Use of Metallic Oxide Nanoparticles to Enhance Growth of Tomatoes and Eggplants in Disease Infested Soil or Soilless Medium. Environ. Sci. Nano 2016, 3, 1072–1079. [Google Scholar] [CrossRef]
  11. Worrall, E.A.; Hamid, A.; Mody, K.T.; Mitter, N.; Pappu, H.R. Nanotechnology for Plant Disease Management. Agronomy 2018, 8, 285. [Google Scholar] [CrossRef] [Green Version]
  12. Maruyama, C.R.; Guilger, M.; Pascoli, M.; Bileshy-José, N.; Abhilash, P.C.; Fraceto, L.F.; De Lima, R. Nanoparticles Based on Chitosan as Carriers for the Combined Herbicides Imazapic and Imazapyr. Sci. Rep. 2016, 6, 19768. [Google Scholar] [CrossRef]
  13. He, Y.; Qian, L.; Liu, X.; Hu, R.; Huang, M.; Liu, Y.; Chen, G.; Losic, D.; Zhu, H. Graphene Oxide as an Antimicrobial Agent Can Extend the Vase Life of Cut Flowers. Nano Res. 2018, 11, 6010–6022. [Google Scholar] [CrossRef]
  14. Saha, N.; Dutta Gupta, S. Promotion of Shoot Regeneration of Swertia Chirata by Biosynthesized Silver Nanoparticles and Their Involvement in Ethylene Interceptions and Activation of Antioxidant Activity. Plant Cell Tissue Organ Cult. 2018, 134, 289–300. [Google Scholar] [CrossRef]
  15. Thangavelu, R.M.; Gunasekaran, D.; Jesse, M.I.; SU, M.R.; Sundarajan, D.; Krishnan, K. Nanobiotechnology Approach Using Plant Rooting Hormone Synthesized Silver Nanoparticle as “Nanobullets” for the Dynamic Applications in Horticulture—An In Vitro and Ex Vitro Study. Arab. J. Chem. 2018, 11, 48–61. [Google Scholar] [CrossRef] [Green Version]
  16. Avestan, S.; Ghasemnezhad, M.; Esfahani, M.; Byrt, C.S. Application of Nano-Silicon Dioxide Improves Salt Stress Tolerance in Strawberry Plants. Agronomy 2019, 9, 246. [Google Scholar] [CrossRef] [Green Version]
  17. Feregrino-Perez, A.A.; Magaña-López, E.; Guzmán, C.; Esquivel, K. A General Overview of the Benefits and Possible Negative Effects of the Nanotechnology in Horticulture. Sci. Hortic. 2018, 238, 126–137. [Google Scholar] [CrossRef]
  18. Chen, Y.W.; Lee, H.V.; Juan, J.C.; Phang, S.-M. Production of New Cellulose Nanomaterial from Red Algae Marine Biomass Gelidium Elegans. Carbohydr. Polym. 2016, 151, 1210–1219. [Google Scholar] [CrossRef]
  19. Ali, K.; Ahmed, B.; Dwivedi, S.; Saquib, Q.; Al-Khedhairy, A.A.; Musarrat, J. Microwave Accelerated Green Synthesis of Stable Silver Nanoparticles with Eucalyptus Globulus Leaf Extract and Their Antibacterial and Antibiofilm Activity on Clinical Isolates. PLoS ONE 2015, 10, e0131178. [Google Scholar] [CrossRef]
  20. Rahman, S.; Rahman, L.; Khalil, A.T.; Ali, N.; Zia, D.; Ali, M.; Shinwari, Z.K. Endophyte-Mediated Synthesis of Silver Nanoparticles and Their Biological Applications. Appl. Microbiol. Biotechnol. 2019, 103, 2551–2569. [Google Scholar] [CrossRef]
  21. Flieger, J.; Franus, W.; Panek, R.; Szymańska-Chargot, M.; Flieger, W.; Flieger, M.; Kołodziej, P. Green Synthesis of Silver Nanoparticles Using Natural Extracts with Proven Antioxidant Activity. Molecules 2021, 26, 4986. [Google Scholar] [CrossRef]
  22. AbdelRahim, K.; Mahmoud, S.Y.; Ali, A.M.; Almaary, K.S.; Mustafa, A.E.-Z.M.A.; Husseiny, S.M. Extracellular Biosynthesis of Silver Nanoparticles Using Rhizopus Stolonifer. Saudi J. Biol. Sci. 2017, 24, 208–216. [Google Scholar] [CrossRef] [Green Version]
  23. Phanjom, P.; Ahmed, G. Effect of Different Physicochemical Conditions on the Synthesis of Silver Nanoparticles Using Fungal Cell Filtrate of Aspergillus Oryzae (MTCC No. 1846) and Their Antibacterial Effect. Adv. Nat. Sci. Nanosci. Nanotechnol. 2017, 8, 45016. [Google Scholar] [CrossRef] [Green Version]
  24. Elamawi, R.M.; Al-Harbi, R.E.; Hendi, A.A. Biosynthesis and Characterization of Silver Nanoparticles Using Trichoderma Longibrachiatum and Their Effect on Phytopathogenic Fungi. Egypt. J. Biol. Pest Control 2018, 28, 28. [Google Scholar] [CrossRef] [Green Version]
  25. Jiang, X.C.; Chen, W.M.; Chen, C.Y.; Xiong, S.X.; Yu, A.B. Role of Temperature in the Growth of Silver Nanoparticles through a Synergetic Reduction Approach. Nanoscale Res. Lett. 2011, 6, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Qian, Y.; Yu, H.; He, D.; Yang, H.; Wang, W.; Wan, X.; Wang, L. Biosynthesis of Silver Nanoparticles by the Endophytic Fungus Epicoccum Nigrum and Their Activity against Pathogenic Fungi. Bioprocess Biosyst. Eng. 2013, 36, 1613–1619. [Google Scholar] [CrossRef] [PubMed]
  27. Du, L.; Xu, Q.; Huang, M.; Xian, L.; Feng, J.-X. Synthesis of Small Silver Nanoparticles under Light Radiation by Fungus Penicillium Oxalicum and Its Application for the Catalytic Reduction of Methylene Blue. Mater. Chem. Phys. 2015, 160, 40–47. [Google Scholar] [CrossRef]
  28. Ahmed, B.; Hashmi, A.; Khan, M.S.; Musarrat, J. ROS Mediated Destruction of Cell Membrane, Growth and Biofilms of Human Bacterial Pathogens by Stable Metallic AgNPs Functionalized from Bell Pepper Extract and Quercetin. Adv. Powder Technol. 2018, 29, 1601–1616. [Google Scholar] [CrossRef]
  29. Panja, A.; Mishra, A.K.; Dash, M.; Pandey, N.K.; Singh, S.K.; Kumar, B. Silver Nanoparticles–A Review. Eurasian J. Med. Oncol. 2021, 5, 95. [Google Scholar] [CrossRef]
  30. Zhang, X.-F.; Liu, Z.-G.; Shen, W.; Gurunathan, S. Silver Nanoparticles: Synthesis, Characterization, Properties, Applications, and Therapeutic Approaches. Int. J. Mol. Sci. 2016, 17, 1534. [Google Scholar] [CrossRef]
  31. Mehmood, A. Brief Overview of the Application of Silver Nanoparticles to Improve Growth of Crop Plants. IET Nanobiotechnol. 2018, 12, 701–705. [Google Scholar] [CrossRef]
  32. Haroon, M.; Zaidi, A.; Ahmed, B.; Rizvi, A.; Khan, M.S.M.S.; Musarrat, J. Effective Inhibition of Phytopathogenic Microbes by Eco-Friendly Leaf Extract Mediated Silver Nanoparticles (AgNPs). Indian J. Microbiol. 2019, 59, 273–287. [Google Scholar] [CrossRef]
  33. Parveen, A.; Rao, S. Effect of Nanosilver on Seed Germination and Seedling Growth in Pennisetum Glaucum. J. Clust. Sci. 2015, 26, 693–701. [Google Scholar] [CrossRef]
  34. Jasim, B.; Thomas, R.; Mathew, J.; Radhakrishnan, E.K. Plant Growth and Diosgenin Enhancement Effect of Silver Nanoparticles in Fenugreek (Trigonella Foenum-Graecum L.). Saudi Pharm. J. 2017, 25, 443–447. [Google Scholar] [CrossRef]
  35. Latif, H.H.; Ghareib, M.; Tahon, M.A. Phytosynthesis of Silver Nanoparticles Using Leaf Extracts from Ocimum Basilicum and Mangifira Indica and Their Effect on Some Biochemical Attributes of Triticum Aestivum. Gesunde Pflanz. 2017, 69, 39–46. [Google Scholar] [CrossRef]
  36. Ge, L.; Li, Q.; Wang, M.; Ouyang, J.; Li, X.; Xing, M.M.Q. Nanosilver Particles in Medical Applications: Synthesis, Performance, and Toxicity. Int. J. Nanomed. 2014, 9, 2399. [Google Scholar]
  37. Biradar, D.; Lingappa, K.; Dayanand, A. Antibacterial Activity of Nano Goldparticles Synthesized by Bacillus Sps. J. Ecobiotechnol. 2012, 4, 43–45. [Google Scholar]
  38. Arokiyaraj, S.; Vincent, S.; Saravanan, M.; Lee, Y.; Oh, Y.K.; Kim, K.H. Green Synthesis of Silver Nanoparticles Using Rheum Palmatum Root Extract and Their Antibacterial Activity against Staphylococcus Aureus and Pseudomonas Aeruginosa. Artif. Cells Nanomed. Biotechnol. 2017, 45, 372–379. [Google Scholar] [CrossRef] [Green Version]
  39. Sumathi, S.; Thomas, A. Eco-Friendly and Antibacterial Finishes of Organic Fabrics Using Herbal Composite Microencapsules. Int. J. Pharma Bio Sci. 2017, 8, 310–321. [Google Scholar]
  40. Joseph, S.; Mathew, B. Microwave Assisted Biosynthesis of Silver Nanoparticles Using the Rhizome Extract of Alpinia Galanga and Evaluation of Their Catalytic and Antimicrobial Activities. J. Nanopart. 2014, 2014, 967802. [Google Scholar] [CrossRef] [Green Version]
  41. Ahmed, S.; Saifullah; Ahmad, M.; Swami, B.L.; Ikram, S. Green Synthesis of Silver Nanoparticles Using Azadirachta Indica Aqueous Leaf Extract. J. Radiat. Res. Appl. Sci. 2016, 9, 1–7. [Google Scholar] [CrossRef] [Green Version]
  42. Dhand, V.; Soumya, L.; Bharadwaj, S.; Chakra, S.; Bhatt, D.; Sreedhar, B. Green Synthesis of Silver Nanoparticles Using Coffea Arabica Seed Extract and Its Antibacterial Activity. Mater. Sci. Eng. C 2016, 58, 36–43. [Google Scholar] [CrossRef]
  43. Fatimah, I.; Indriani, N. Silver Nanoparticles Synthesized Using Lantana Camara Flower Extract by Reflux, Microwave and Ultrasound Methods. Chem. J. Mold. 2018, 13, 95–102. [Google Scholar] [CrossRef]
  44. Kumar, B.; Smita, K.; Cumbal, L.; Debut, A. Green Synthesis of Silver Nanoparticles Using Andean Blackberry Fruit Extract. Saudi J. Biol. Sci. 2017, 24, 45–50. [Google Scholar] [CrossRef] [Green Version]
  45. Kumar, B.; Smita, K.; Cumbal, L.; Angulo, Y. Fabrication of Silver Nanoplates Using Nephelium Lappaceum (Rambutan) Peel: A Sustainable Approach. J. Mol. Liq. 2015, 211, 476–480. [Google Scholar] [CrossRef]
  46. Ojha, S.; Sett, A.; Bora, U. Green Synthesis of Silver Nanoparticles by Ricinus Communis Var. Carmencita Leaf Extract and Its Antibacterial Study. Adv. Nat. Sci. Nanosci. Nanotechnol. 2017, 8, 35009. [Google Scholar] [CrossRef]
  47. Joshi, P.S.; Ramesh, G.; Packiyam, J.E.; Jayanna, S.K. Green Synthesis and Evaluation of Silver Nanoparticles Using Leaf Extract from Calotropis Gigantean. Int. J. Curr. Biotechnol. 2007, 5, 1–5. [Google Scholar]
  48. Ahmed, B.; Syed, A.; Ali, K.; Elgorban, A.M.; Khan, A.; Lee, J.; AL-Shwaiman, H.A. Synthesis of Gallotannin Capped Iron Oxide Nanoparticles and Their Broad Spectrum Biological Applications. RSC Adv. 2021, 11, 9880–9893. [Google Scholar] [CrossRef]
  49. Ali, K.; Saquib, Q.; Ahmed, B.; Siddiqui, M.A.; Ahmad, J.; Al-Shaeri, M.; Al-Khedhairy, A.A.; Musarrat, J. Bio-Functionalized CuO Nanoparticles Induced Apoptotic Activities in Human Breast Carcinoma Cells and Toxicity against Aspergillus Flavus: An in Vitro Approach. Process Biochem. 2020, 91, 387–397. [Google Scholar] [CrossRef]
  50. Ahmed, B.; Solanki, B.; Zaidi, A.; Khan, M.S.; Musarrat, J. Bacterial Toxicity of Biomimetic Green Zinc Oxide Nanoantibiotic: Insights into ZnONP Uptake and Nanocolloid-Bacteria Interface. Toxicol. Res. 2019, 8, 246–261. [Google Scholar] [CrossRef] [Green Version]
  51. Ali, K.; Ahmed, B.; Khan, M.S.; Musarrat, J. Differential Surface Contact Killing of Pristine and Low EPS Pseudomonas Aeruginosa with Aloe Vera Capped Hematite (α-Fe2O3) Nanoparticles. J. Photochem. Photobiol. B Biol. 2018, 188, 146–158. [Google Scholar] [CrossRef]
  52. Jebakumar Immanuel Edison, T.N.; Sethuraman, M.G. Electrocatalytic Reduction of Benzyl Chloride by Green Synthesized Silver Nanoparticles Using Pod Extract of Acacia Nilotica. ACS Sustain. Chem. Eng. 2013, 1, 1326–1332. [Google Scholar] [CrossRef]
  53. Mallikarjuna, K.; Narasimha, G.; Dillip, G.R.; Praveen, B.; Shreedhar, B.; Lakshmi, C.S.; Reddy, B.V.S.; Raju, B.D.P. Green Synthesis of Silver Nanoparticles Using Ocimum Leaf Extract and Their Characterization. Dig. J. Nanomater. Biostruct. 2011, 6, 181–186. [Google Scholar]
  54. Vanaja, M.; Annadurai, G. Coleus Aromaticus Leaf Extract Mediated Synthesis of Silver Nanoparticles and Its Bactericidal Activity. Appl. Nanosci. 2013, 3, 217–223. [Google Scholar] [CrossRef] [Green Version]
  55. Ajitha, B.; Reddy, Y.A.K.; Reddy, P.S. Green Synthesis and Characterization of Silver Nanoparticles Using Lantana Camara Leaf Extract. Mater. Sci. Eng. C 2015, 49, 373–381. [Google Scholar] [CrossRef] [PubMed]
  56. Reddy, N.J.; Vali, D.N.; Rani, M.; Rani, S.S. Evaluation of Antioxidant, Antibacterial and Cytotoxic Effects of Green Synthesized Silver Nanoparticles by Piper Longum Fruit. Mater. Sci. Eng. C 2014, 34, 115–122. [Google Scholar] [CrossRef]
  57. Vasanth, K.; Ilango, K.; MohanKumar, R.; Agrawal, A.; Dubey, G.P. Anticancer Activity of Moringa Oleifera Mediated Silver Nanoparticles on Human Cervical Carcinoma Cells by Apoptosis Induction. Colloids Surf. B Biointerfaces 2014, 117, 354–359. [Google Scholar] [CrossRef]
  58. Atale, N.; Saxena, S.; Nirmala, J.G.; Narendhirakannan, R.T.; Mohanty, S.; Rani, V. Synthesis and Characterization of Sygyzium Cumini Nanoparticles for Its Protective Potential in High Glucose-Induced Cardiac Stress: A Green Approach. Appl. Biochem. Biotechnol. 2017, 181, 1140–1154. [Google Scholar] [CrossRef]
  59. Anil Kumar, S.; Abyaneh, M.K.; Gosavi, S.W.; Kulkarni, S.K.; Pasricha, R.; Ahmad, A.; Khan, M.I. Nitrate Reductase-Mediated Synthesis of Silver Nanoparticles from AgNO3. Biotechnol. Lett. 2007, 29, 439–445. [Google Scholar] [CrossRef]
  60. Saratale, R.G.; Karuppusamy, I.; Saratale, G.D.; Pugazhendhi, A.; Kumar, G.; Park, Y.; Ghodake, G.S.; Bharagava, R.N.; Banu, J.R.; Shin, H.S. A Comprehensive Review on Green Nanomaterials Using Biological Systems: Recent Perception and Their Future Applications. Colloids Surf. B Biointerfaces 2018, 170, 20–35. [Google Scholar] [CrossRef]
  61. Malarkodi, C.; Rajeshkumar, S.; Paulkumar, K.; Gnanajobitha, G.; Vanaja, M.; Annadurai, G. Bacterial Synthesis of Silver Nanoparticles by Using Optimized Biomass Growth of Bacillus sp. Nanosci. Nanotechnol. 2013, 3, 26–32. [Google Scholar]
  62. Hallol, M. Studies on Bacterial Synthesis of Silver Nanoparticles Using Gamma Radiation and Their Activity against Some Pathogenic Microbes. 2013. Available online: https://inis.iaea.org/collection/NCLCollectionStore/_Public/46/066/46066330.pdf (accessed on 1 July 2022).
  63. Das, V.L.; Thomas, R.; Varghese, R.T.; Soniya, E.V.; Mathew, J.; Radhakrishnan, E.K. Extracellular Synthesis of Silver Nanoparticles by the Bacillus Strain CS 11 Isolated from Industrialized Area. 3 Biotech 2014, 4, 121–126. [Google Scholar] [CrossRef] [Green Version]
  64. Gandhi, H.; Khan, S. Biological Synthesis of Silver Nanoparticles and Its Antibacterial Activity. J. Nanomed. Nanotechnol. 2016, 7, 1000366. [Google Scholar] [CrossRef] [Green Version]
  65. Parikh, R.Y.; Ramanathan, R.; Coloe, P.J.; Bhargava, S.K.; Patole, M.S.; Shouche, Y.S.; Bansal, V. Genus-Wide Physicochemical Evidence of Extracellular Crystalline Silver Nanoparticles Biosynthesis by Morganella spp. PLoS ONE 2011, 6, e21401. [Google Scholar] [CrossRef]
  66. Shivaji, S.; Madhu, S.; Singh, S. Extracellular Synthesis of Antibacterial Silver Nanoparticles Using Psychrophilic Bacteria. Process Biochem. 2011, 46, 1800–1807. [Google Scholar] [CrossRef]
  67. Saravanan, M.; Barik, S.K.; MubarakAli, D.; Prakash, P.; Pugazhendhi, A. Synthesis of Silver Nanoparticles from Bacillus Brevis (NCIM 2533) and Their Antibacterial Activity against Pathogenic Bacteria. Microb. Pathog. 2018, 116, 221–226. [Google Scholar] [CrossRef]
  68. Mukherjee, A.; Sarkar, D.; Sasmal, S. A Review of Green Synthesis of Metal Nanoparticles Using Algae. Front. Microbiol. 2021, 12, 693899. [Google Scholar] [CrossRef]
  69. Akther, T.; Mathipi, V.; Kumar, N.S.; Davoodbasha, M.; Srinivasan, H. Fungal-Mediated Synthesis of Pharmaceutically Active Silver Nanoparticles and Anticancer Property against A549 Cells through Apoptosis. Environ. Sci. Pollut. Res. 2019, 26, 13649–13657. [Google Scholar] [CrossRef]
  70. Barabadi, H.; Tajani, B.; Moradi, M.; Damavandi Kamali, K.; Meena, R.; Honary, S.; Mahjoub, M.A.; Saravanan, M. Penicillium Family as Emerging Nanofactory for Biosynthesis of Green Nanomaterials: A Journey into the World of Microorganisms. J. Clust. Sci. 2019, 30, 843–856. [Google Scholar] [CrossRef]
  71. Gade, A.; Ingle, A.; Whiteley, C.; Rai, M. Mycogenic Metal Nanoparticles: Progress and Applications. Biotechnol. Lett. 2010, 32, 593–600. [Google Scholar] [CrossRef]
  72. Jena, J.; Pradhan, N.; Dash, B.P.; Sukla, L.B.; Panda, P.K. Biosynthesis and Characterization of Silver Nanoparticles Using Microalga Chlorococcum Humicola and Its Antibacterial Activity. Int. J. Nanomater Biostruct 2013, 3, 1–8. [Google Scholar]
  73. Jena, J.; Pradhan, N.; Dash, B.P.; Panda, P.K.; Mishra, B.K. Pigment Mediated Biogenic Synthesis of Silver Nanoparticles Using Diatom Amphora Sp. and Its Antimicrobial Activity. J. Saudi Chem. Soc. 2015, 19, 661–666. [Google Scholar] [CrossRef] [Green Version]
  74. Kathiraven, T.; Sundaramanickam, A.; Shanmugam, N.; Balasubramanian, T. Green Synthesis of Silver Nanoparticles Using Marine Algae Caulerpa Racemosa and Their Antibacterial Activity against Some Human Pathogens. Appl. Nanosci. 2015, 5, 499–504. [Google Scholar] [CrossRef] [Green Version]
  75. Sahayaraj, K.; Rajesh, S.; Rathi, J.M. Silver nanoparticles biosynthesis using marine alga padina pavonica (linn.) and its microbicidal activity. Dig. J. Nanomater. Biostruct. 2012, 7, 1557–1567. [Google Scholar]
  76. Kannan, R.; Arumugam, R.; Ramya, D.; Manivannan, K.; Anantharaman, P. Green Synthesis of Silver Nanoparticles Using Marine Macroalga Chaetomorpha Linum. Appl. Nanosci. 2013, 3, 229–233. [Google Scholar] [CrossRef] [Green Version]
  77. Pugazhendhi, A.; Prabakar, D.; Jacob, J.M.; Karuppusamy, I.; Saratale, R.G. Synthesis and Characterization of Silver Nanoparticles Using Gelidium Amansii and Its Antimicrobial Property against Various Pathogenic Bacteria. Microb. Pathog. 2018, 114, 41–45. [Google Scholar] [CrossRef] [PubMed]
  78. Bhainsa, K.C.; D’souza, S.F. Extracellular Biosynthesis of Silver Nanoparticles Using the Fungus Aspergillus Fumigatus. Colloids Surf. B Biointerfaces 2006, 47, 160–164. [Google Scholar] [CrossRef] [PubMed]
  79. Kathiresan, K.; Manivannan, S.; Nabeel, M.A.; Dhivya, B. Studies on Silver Nanoparticles Synthesized by a Marine Fungus, Penicillium Fellutanum Isolated from Coastal Mangrove Sediment. Colloids Surf. B Biointerfaces 2009, 71, 133–137. [Google Scholar] [CrossRef] [PubMed]
  80. Vigneshwaran, N.; Ashtaputre, N.M.; Varadarajan, P.V.; Nachane, R.P.; Paralikar, K.M.; Balasubramanya, R.H. Biological Synthesis of Silver Nanoparticles Using the Fungus Aspergillus Flavus. Mater. Lett. 2007, 61, 1413–1418. [Google Scholar] [CrossRef]
  81. Basavaraja, S.; Balaji, S.D.; Lagashetty, A.; Rajasab, A.H.; Venkataraman, A. Extracellular Biosynthesis of Silver Nanoparticles Using the Fungus Fusarium Semitectum. Mater. Res. Bull. 2008, 43, 1164–1170. [Google Scholar] [CrossRef]
  82. Gajbhiye, M.; Kesharwani, J.; Ingle, A.; Gade, A.; Rai, M. Fungus-Mediated Synthesis of Silver Nanoparticles and Their Activity against Pathogenic Fungi in Combination with Fluconazole. Nanomed. Nanotechnol. Biol. Med. 2009, 5, 382–386. [Google Scholar] [CrossRef]
  83. Rashidipour, M.; Heydari, R. Biosynthesis of Silver Nanoparticles Using Extract of Olive Leaf: Synthesis and in Vitro Cytotoxic Effect on MCF-7 Cells. J. Nanostruct. Chem. 2014, 4, 112. [Google Scholar] [CrossRef] [Green Version]
  84. Majeed Khan, M.A.; Kumar, S.; Ahamed, M.; Alrokayan, S.A.; AlSalhi, M.S. Structural and Thermal Studies of Silver Nanoparticles and Electrical Transport Study of Their Thin Films. Nanoscale Res. Lett. 2011, 6, 434. [Google Scholar] [CrossRef] [Green Version]
  85. Njagi, E.C.; Huang, H.; Stafford, L.; Genuino, H.; Galindo, H.M.; Collins, J.B.; Hoag, G.E.; Suib, S.L. Biosynthesis of Iron and Silver Nanoparticles at Room Temperature Using Aqueous Sorghum Bran Extracts. Langmuir 2011, 27, 264–271. [Google Scholar] [CrossRef]
  86. Lukman, A.I.; Gong, B.; Marjo, C.E.; Roessner, U.; Harris, A.T. Facile Synthesis, Stabilization, and Anti-Bacterial Performance of Discrete Ag Nanoparticles Using Medicago Sativa Seed Exudates. J. Colloid Interface Sci. 2011, 353, 433–444. [Google Scholar] [CrossRef]
  87. Raju, D.; Mehta, U.J.; Hazra, S. Synthesis of Gold Nanoparticles by Various Leaf Fractions of Semecarpus Anacardium L. Tree. Trees 2011, 25, 145–151. [Google Scholar] [CrossRef]
  88. Marchiol, L. Synthesis of Metal Nanoparticles in Living Plants. Ital. J. Agron. 2012, 7, e37. [Google Scholar] [CrossRef]
  89. Veerasamy, R.; Xin, T.Z.; Gunasagaran, S.; Xiang, T.F.W.; Yang, E.F.C.; Jeyakumar, N.; Dhanaraj, S.A. Biosynthesis of Silver Nanoparticles Using Mangosteen Leaf Extract and Evaluation of Their Antimicrobial Activities. J. Saudi Chem. Soc. 2011, 15, 113–120. [Google Scholar] [CrossRef] [Green Version]
  90. Krishnaraj, C.; Ramachandran, R.; Mohan, K.; Kalaichelvan, P.T. Optimization for Rapid Synthesis of Silver Nanoparticles and Its Effect on Phytopathogenic Fungi. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2012, 93, 95–99. [Google Scholar] [CrossRef]
  91. Ali, M.; Ahmed, T.; Wu, W.; Hossain, A.; Hafeez, R.; Islam Masum, M.; Wang, Y.; An, Q.; Sun, G.; Li, B. Advancements in Plant and Microbe-Based Synthesis of Metallic Nanoparticles and Their Antimicrobial Activity against Plant Pathogens. Nanomaterials 2020, 10, 1146. [Google Scholar] [CrossRef]
  92. Mittal, A.K.; Chisti, Y.; Banerjee, U.C. Synthesis of Metallic Nanoparticles Using Plant Extracts. Biotechnol. Adv. 2013, 31, 346–356. [Google Scholar] [CrossRef]
  93. Iravani, S. Green Synthesis of Metal Nanoparticles Using Plants. Green Chem. 2011, 13, 2638–2650. [Google Scholar] [CrossRef]
  94. Sre, P.R.R.; Reka, M.; Poovazhagi, R.; Kumar, M.A.; Murugesan, K. Antibacterial and Cytotoxic Effect of Biologically Synthesized Silver Nanoparticles Using Aqueous Root Extract of Erythrina Indica Lam. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2015, 135, 1137–1144. [Google Scholar]
  95. Braithwaite, M.C.; Tyagi, C.; Tomar, L.K.; Kumar, P.; Choonara, Y.E.; Pillay, V. Nutraceutical-Based Therapeutics and Formulation Strategies Augmenting Their Efficiency to Complement Modern Medicine: An Overview. J. Funct. Foods 2014, 6, 82–99. [Google Scholar] [CrossRef]
  96. Bose, D.; Chatterjee, S. Biogenic Synthesis of Silver Nanoparticles Using Guava (Psidium Guajava) Leaf Extract and Its Antibacterial Activity against Pseudomonas Aeruginosa. Appl. Nanosci. 2016, 6, 895–901. [Google Scholar] [CrossRef] [Green Version]
  97. Rai, M.K.; Deshmukh, S.D.; Ingle, A.P.; Gade, A.K. Silver Nanoparticles: The Powerful Nanoweapon against Multidrug-resistant Bacteria. J. Appl. Microbiol. 2012, 112, 841–852. [Google Scholar] [CrossRef]
  98. Ahmed, B.; Ameen, F.; Rizvi, A.; Ali, K.; Sonbol, H.; Zaidi, A.; Khan, M.S.; Musarrat, J. Destruction of Cell Topography, Morphology, Membrane, Inhibition of Respiration, Biofilm Formation, and Bioactive Molecule Production by Nanoparticles of Ag, ZnO, CuO, TiO2, and Al2O3 toward Beneficial Soil Bacteria. ACS Omega 2020, 5, 7861–7876. [Google Scholar] [CrossRef] [Green Version]
  99. Burdușel, A.-C.; Gherasim, O.; Grumezescu, A.M.; Mogoantă, L.; Ficai, A.; Andronescu, E. Biomedical Applications of Silver Nanoparticles: An up-to-Date Overview. Nanomaterials 2018, 8, 681. [Google Scholar] [CrossRef] [Green Version]
  100. Kaviya, S.; Santhanalakshmi, J.; Viswanathan, B.; Muthumary, J.; Srinivasan, K. Biosynthesis of Silver Nanoparticles Using Citrus Sinensis Peel Extract and Its Antibacterial Activity. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2011, 79, 594–598. [Google Scholar] [CrossRef]
  101. Palanisamy, S.; Rajasekar, P.; Vijayaprasath, G.; Ravi, G.; Manikandan, R.; Prabhu, N.M. A Green Route to Synthesis Silver Nanoparticles Using Sargassum Polycystum and Its Antioxidant and Cytotoxic Effects: An in Vitro Analysis. Mater. Lett. 2017, 189, 196–200. [Google Scholar] [CrossRef]
  102. Da Rosa, C.G.; de Melo, A.P.Z.; Sganzerla, W.G.; Machado, M.H.; Nunes, M.R.; de Maciel, M.V.O.B.; Bertoldi, F.C.; Barreto, P.L.M. Application in Situ of Zein Nanocapsules Loaded with Origanum Vulgare Linneus and Thymus Vulgaris as a Preservative in Bread. Food Hydrocoll. 2020, 99, 105339. [Google Scholar] [CrossRef]
  103. Kang, Y.O.; Lee, T.S.; Park, W.H. Green Synthesis and Antimicrobial Activity of Silver Chloride Nanoparticles Stabilized with Chitosan Oligomer. J. Mater. Sci. Mater. Med. 2014, 25, 2629–2638. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, Y.-W.; Tang, H.; Wu, D.; Liu, D.; Liu, Y.; Cao, A.; Wang, H. Enhanced Bactericidal Toxicity of Silver Nanoparticles by the Antibiotic Gentamicin. Environ. Sci. Nano 2016, 3, 788–798. [Google Scholar] [CrossRef]
  105. Wong, K.K.Y.; Liu, X. Silver Nanoparticles—The Real “Silver Bullet” in Clinical Medicine? Medchemcomm 2010, 1, 125–131. [Google Scholar] [CrossRef]
  106. Wu, J.; Yu, C.; Tan, Y.; Hou, Z.; Li, M.; Shao, F.; Lu, X. Effects of Prenatal Exposure to Silver Nanoparticles on Spatial Cognition and Hippocampal Neurodevelopment in Rats. Environ. Res. 2015, 138, 67–73. [Google Scholar] [CrossRef]
  107. Yuan, Z.; Li, J.; Cui, L.; Xu, B.; Zhang, H.; Yu, C.P. Interaction of Silver Nanoparticles with Pure Nitrifying Bacteria. Chemosphere 2013, 90, 1404–1411. [Google Scholar] [CrossRef]
  108. Hajipour, M.J.; Fromm, K.M.; Ashkarran, A.A.; de Aberasturi, D.J.; de Larramendi, I.R.; Rojo, T.; Serpooshan, V.; Parak, W.J.; Mahmoudi, M. Antibacterial Properties of Nanoparticles. Trends Biotechnol. 2012, 30, 499–511. [Google Scholar] [CrossRef] [Green Version]
  109. Lemire, J.A.; Harrison, J.J.; Turner, R.J. Antimicrobial Activity of Metals: Mechanisms, Molecular Targets and Applications. Nat. Rev. Microbiol. 2013, 11, 371–384. [Google Scholar] [CrossRef]
  110. Alghuthaymi, M.A.; Almoammar, H.; Rai, M.; Said-Galiev, E.; Abd-Elsalam, K.A. Myconanoparticles: Synthesis and Their Role in Phytopathogens Management. Biotechnol. Biotechnol. Equip. 2015, 29, 221–236. [Google Scholar] [CrossRef]
  111. Masum, M.; Islam, M.; Siddiqa, M.; Ali, K.A.; Zhang, Y.; Abdallah, Y.; Ibrahim, E.; Qiu, W.; Yan, C.; Li, B. Biogenic Synthesis of Silver Nanoparticles Using Phyllanthus Emblica Fruit Extract and Its Inhibitory Action against the Pathogen Acidovorax Oryzae Strain RS-2 of Rice Bacterial Brown Stripe. Front. Microbiol. 2019, 10, 820. [Google Scholar] [CrossRef]
  112. Ibrahim, E.; Zhang, M.; Zhang, Y.; Hossain, A.; Qiu, W.; Chen, Y.; Wang, Y.; Wu, W.; Sun, G.; Li, B. Green-Synthesization of Silver Nanoparticles Using Endophytic Bacteria Isolated from Garlic and Its Antifungal Activity against Wheat Fusarium Head Blight Pathogen Fusarium Graminearum. Nanomaterials 2020, 10, 219. [Google Scholar] [CrossRef] [Green Version]
  113. Kędziora, A.; Speruda, M.; Krzyżewska, E.; Rybka, J.; Łukowiak, A.; Bugla-Płoskońska, G. Similarities and Differences between Silver Ions and Silver in Nanoforms as Antibacterial Agents. Int. J. Mol. Sci. 2018, 19, 444. [Google Scholar] [CrossRef] [Green Version]
  114. Qing, Y.; Cheng, L.; Li, R.; Liu, G.; Zhang, Y.; Tang, X.; Wang, J.; Liu, H.; Qin, Y. Potential Antibacterial Mechanism of Silver Nanoparticles and the Optimization of Orthopedic Implants by Advanced Modification Technologies. Int. J. Nanomed. 2018, 13, 3311. [Google Scholar] [CrossRef] [Green Version]
  115. Ibrahim, E.; Fouad, H.; Zhang, M.; Zhang, Y.; Qiu, W.; Yan, C.; Li, B.; Mo, J.; Chen, J. Biosynthesis of Silver Nanoparticles Using Endophytic Bacteria and Their Role in Inhibition of Rice Pathogenic Bacteria and Plant Growth Promotion. RSC Adv. 2019, 9, 29293–29299. [Google Scholar] [CrossRef] [Green Version]
  116. Singh, A.; Gautam, P.K.; Verma, A.; Singh, V.; Shivapriya, P.M.; Shivalkar, S.; Sahoo, A.K.; Samanta, S.K. Green Synthesis of Metallic Nanoparticles as Effective Alternatives to Treat Antibiotics Resistant Bacterial Infections: A Review. Biotechnol. Rep. 2020, 25, e00427. [Google Scholar] [CrossRef]
  117. Tortella, G.; Rubilar, O.; Fincheira, P.; Pieretti, J.C.; Duran, P.; Lourenço, I.M.; Seabra, A.B. Bactericidal and Virucidal Activities of Biogenic Metal-Based Nanoparticles: Advances and Perspectives. Antibiotics 2021, 10, 783. [Google Scholar] [CrossRef]
  118. Andleeb, S.; Tariq, F.; Muneer, A.; Nazir, T.; Shahid, B.; Latif, Z.; Abbasi, S.A.; ul Haq, I.; Majeed, Z.; Khan, S.U.-D. In Vitro Bactericidal, Antidiabetic, Cytotoxic, Anticoagulant, and Hemolytic Effect of Green-Synthesized Silver Nanoparticles Using Allium Sativum Clove Extract Incubated at Various Temperatures. Green Process. Synth. 2020, 9, 538–553. [Google Scholar] [CrossRef]
  119. Rodríguez-Serrano, C.; Guzmán-Moreno, J.; Ángeles-Chávez, C.; Rodríguez-González, V.; Ortega-Sigala, J.J.; Ramírez-Santoyo, R.M.; Vidales-Rodríguez, L.E. Biosynthesis of Silver Nanoparticles by Fusarium Scirpi and Its Potential as Antimicrobial Agent against Uropathogenic Escherichia Coli Biofilms. PLoS ONE 2020, 15, e0230275. [Google Scholar] [CrossRef] [Green Version]
  120. Noshad, A.; Iqbal, M.; Hetherington, C.; Wahab, H. Biogenic AgNPs—a Nano Weapon against Bacterial Canker of Tomato (Bct). Adv. Agric. 2020, 2020, 9630785. [Google Scholar] [CrossRef]
  121. Le Thi, H.; Nguyen, T.P.H.; Le Trong, D.; Vu, T.H.; Le Thi, V.; Hoang, T.G.; Chu, D.H.; Nguyen, T.H.; Nguyen, D.P.; Le Huy, H. Synthesis of Chitosan Stabilized Silver Nanoparticles and Evaluation of the in Vitro Antibacterial Activity Against Xanthomonas Oryzae Pv. Oryzae Causing Blight Disease of Rice. VNU J. Sci. Nat. Sci. Technol. 2021, 6, 65–75. [Google Scholar]
  122. Cheng, H.-J.; Wang, H.; Zhang, J.-Z. Phytofabrication of Silver Nanoparticles Using Three Flower Extracts and Their Antibacterial Activities against Pathogen Ralstonia Solanacearum Strain YY06 of Bacterial Wilt. Front. Microbiol. 2020, 11, 2110. [Google Scholar] [CrossRef]
  123. Xing, Y.; Liao, X.; Liu, X.; Li, W.; Huang, R.; Tang, J.; Xu, Q.; Li, X.; Yu, J. Characterization and Antimicrobial Activity of Silver Nanoparticles Synthesized with the Peel Extract of Mango. Materials 2021, 14, 5878. [Google Scholar] [CrossRef]
  124. Ahmed, T.; Shahid, M.; Noman, M.; Niazi, M.B.K.; Mahmood, F.; Manzoor, I.; Zhang, Y.; Li, B.; Yang, Y.; Yan, C. Silver Nanoparticles Synthesized by Using Bacillus Cereus SZT1 Ameliorated the Damage of Bacterial Leaf Blight Pathogen in Rice. Pathogens 2020, 9, 160. [Google Scholar] [CrossRef] [Green Version]
  125. del Carmen Travieso Novelles, M.; Ortega, A.R.; Pita, B.A.; López, M.C.; Pérez, L.D.; Medina, E.A.; Pérez, O.P. Biosynthesis of Fluorescent Silver Nanoparticles from Leea Coccinea Leaves and Their Antibacterial Potentialities against Xanthomonas Phaseoli Pv Phaseoli. Bioresour. Bioprocess. 2021, 8, 3. [Google Scholar] [CrossRef]
  126. Mazzon, M.; Marsh, M. Targeting Viral Entry as a Strategy for Broad-Spectrum Antivirals. F1000Research 2019, 8, 1–10. [Google Scholar] [CrossRef] [Green Version]
  127. Galdiero, S.; Falanga, A.; Vitiello, M.; Cantisani, M.; Marra, V.; Galdiero, M. Silver Nanoparticles as Potential Antiviral Agents. Molecules 2011, 16, 8894–8918. [Google Scholar] [CrossRef] [Green Version]
  128. Noha, K.; Bondok, A.M.; El-Dougdoug, K.A. Evaluation of Silver Nanoparticles as Antiviral Agent against ToMV and PVY in Tomato Plants. Sciences 2018, 8, 100–111. [Google Scholar]
  129. Elbeshehy, E.K.F.; Elazzazy, A.M.; Aggelis, G. Silver Nanoparticles Synthesis Mediated by New Isolates of Bacillus spp., Nanoparticle Characterization and Their Activity against Bean Yellow Mosaic Virus and Human Pathogens. Front. Microbiol. 2015, 6, 453. [Google Scholar] [CrossRef] [Green Version]
  130. Shafie, R.M.; Salama, A.M.; Farroh, K.Y. Silver Nanoparticles Activity against Tomato Spotted Wilt Virus. Middle East J. Appl. Sci. 2018, 7, 1251–1267. [Google Scholar]
  131. El-Shazly, M.A.; Attia, Y.A.; Kabil, F.F.; Anis, E.; Hazman, M. Inhibitory Effects of Salicylic Acid and Silver Nanoparticles on Potato Virus Y-Infected Potato Plants in Egypt. Middle East J. Agric. Res. 2017, 6, 835–848. [Google Scholar]
  132. Jain, D.; Kothari, S.L. Green Synthesis of Silver Nanoparticles and Their Application in Plant Virus Inhibition. J. Mycol Plant Pathol 2014, 44, 21–24. [Google Scholar]
  133. Mahfouze, H.A.; El-Dougdoug, N.K.; Mahfouze, S.A. Virucidal Activity of Silver Nanoparticles against Banana Bunchy Top Virus (BBTV) in Banana Plants. Bull. Natl. Res. Cent. 2020, 44, 199. [Google Scholar] [CrossRef]
  134. Cai, L.; Cai, L.; Jia, H.; Liu, C.; Wang, D.; Sun, X. Foliar Exposure of Fe3O4 Nanoparticles on Nicotiana Benthamiana: Evidence for Nanoparticles Uptake, Plant Growth Promoter and Defense Response Elicitor against Plant Virus. J. Hazard. Mater. 2020, 393, 122415. [Google Scholar] [CrossRef]
  135. Vinković, T.; Novák, O.; Strnad, M.; Goessler, W.; Jurašin, D.D.; Parađiković, N.; Vrček, I.V. Cytokinin Response in Pepper Plants (Capsicum Annuum L.) Exposed to Silver Nanoparticles. Environ. Res. 2017, 156, 10–18. [Google Scholar] [CrossRef]
  136. Tripathi, D.K.; Tripathi, A.; Shweta; Singh, S.; Singh, Y.; Vishwakarma, K.; Yadav, G.; Sharma, S.; Singh, V.K.; Mishra, R.K.; et al. Uptake, Accumulation and Toxicity of Silver Nanoparticle in Autotrophic Plants, and Heterotrophic Microbes: A Concentric Review. Front. Microbiol. 2017, 8, 7. [Google Scholar] [CrossRef]
  137. Hassan, S.A.M.; Mahfouze, H.A.; Mahfouze, S.A.; Abd-Allatif, A.M. Genotoxicity Assessment of Nano-Particles on Micropropagated Olive (Olea Europaea L.) Plants Using RAPD and DAMD Markers. Plant Arch. 2019, 19, 1985–1994. [Google Scholar]
  138. Elazzazy, A.M.; Elbeshehy, E.K.F.; Betiha, M.A. In Vitro Assessment of Activity of Graphene Silver Composite Sheets against Multidrug-Resistant Bacteria and Tomato Bushy Stunt Virus. Trop. J. Pharm. Res. 2017, 16, 2705–2711. [Google Scholar] [CrossRef]
  139. Wang, Y.; Sun, C.; Xu, C.; Wang, Z.; Zhao, M.; Wang, C.; Liu, L.; Chen, F. Preliminary Experiments on Nano-Silver against Tobacco Mosaic Virus and Its Mechanism. Tob. Sci. Technol. 2016, 49, 22–30. [Google Scholar]
  140. Jo, Y.-K.; Cromwell, W.; Jeong, H.-K.; Thorkelson, J.; Roh, J.-H.; Shin, D.-B. Use of Silver Nanoparticles for Managing Gibberella Fujikuroi on Rice Seedlings. Crop Prot. 2015, 74, 65–69. [Google Scholar] [CrossRef]
  141. Nyilasi, I.; Kocsubé, S.; Krizsán, K.; Galgóczy, L.; Pesti, M.; Papp, T.; Vágvölgyi, C. In Vitro Synergistic Interactions of the Effects of Various Statins and Azoles against Some Clinically Important Fungi. FEMS Microbiol. Lett. 2010, 307, 175–184. [Google Scholar] [CrossRef] [Green Version]
  142. Guilger, M.; Pasquoto-Stigliani, T.; Bilesky-Jose, N.; Grillo, R.; Abhilash, P.C.; Fraceto, L.F.; de Lima, R. Biogenic Silver Nanoparticles Based on Trichoderma Harzianum: Synthesis, Characterization, Toxicity Evaluation and Biological Activity. Sci. Rep. 2017, 7, 44421. [Google Scholar] [CrossRef]
  143. Auyeung, A.; Casillas-Santana, M.A.; Martínez-Castañón, G.A.; Slavin, Y.N.; Zhao, W.; Asnis, J.; Häfeli, U.O.; Bach, H. Effective Control of Molds Using a Combination of Nanoparticles. PLoS ONE 2017, 12, e0169940. [Google Scholar] [CrossRef] [Green Version]
  144. Fatima, F.; Verma, S.R.; Pathak, N.; Bajpai, P. Extracellular Mycosynthesis of Silver Nanoparticles and Their Microbicidal Activity. J. Glob. Antimicrob. Resist. 2016, 7, 88–92. [Google Scholar] [CrossRef]
  145. Mallmann, E.J.J.; Cunha, F.A.; Castro, B.N.M.F.; Maciel, A.M.; Menezes, E.A.; Fechine, P.B.A. Antifungal Activity of Silver Nanoparticles Obtained by Green Synthesis. Rev. Inst. Med. Trop. Sao Paulo 2015, 57, 165–167. [Google Scholar] [CrossRef]
  146. Gopinath, V.; Velusamy, P. Extracellular Biosynthesis of Silver Nanoparticles Using Bacillus sp. GP-23 and Evaluation of Their Antifungal Activity towards Fusarium Oxysporum. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2013, 106, 170–174. [Google Scholar] [CrossRef]
  147. Hahn, M. The Rising Threat of Fungicide Resistance in Plant Pathogenic Fungi: Botrytis as a Case Study. J. Chem. Biol. 2014, 7, 133–141. [Google Scholar] [CrossRef] [Green Version]
  148. Matsumura, Y.; Yoshikata, K.; Kunisaki, S.; Tsuchido, T. Mode of Bactericidal Action of Silver Zeolite and Its Comparison with That of Silver Nitrate. Appl. Environ. Microbiol. 2003, 69, 4278–4281. [Google Scholar] [CrossRef] [Green Version]
  149. Feng, Q.L.; Wu, J.; Chen, G.Q.; Cui, F.Z.; Kim, T.N.; Kim, J.O. A Mechanistic Study of the Antibacterial Effect of Silver Ions on Escherichia Coli and Staphylococcus Aureus. J. Biomed. Mater. Res. 2000, 52, 662–668. [Google Scholar] [CrossRef]
  150. 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. Biotechnol. 2014, 98, 1951–1961. [Google Scholar] [CrossRef]
  151. Al-Thabaiti, S.A.; Khan, Z.; Manzoor, N. Biosynthesis of Silver Nanoparticles and Its Antibacterial and Antifungal Activities towards Gram-Positive, Gram-Negative Bacterial Strains and Different Species of Candida Fungus. Bioprocess Biosyst. Eng. 2015, 38, 1773–1781. [Google Scholar]
  152. Pereira, L.; Dias, N.; Carvalho, J.; Fernandes, S.; Santos, C.; Lima, N. Synthesis, Characterization and Antifungal Activity of Chemically and Fungal-produced Silver Nanoparticles against T Richophyton Rubrum. J. Appl. Microbiol. 2014, 117, 1601–1613. [Google Scholar] [CrossRef] [Green Version]
  153. Xia, Z.-K.; Ma, Q.-H.; Li, S.-Y.; Zhang, D.-Q.; Cong, L.; Tian, Y.-L.; Yang, R.-Y. The Antifungal Effect of Silver Nanoparticles on Trichosporon Asahii. J. Microbiol. Immunol. Infect. 2016, 49, 182–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Bera, R.K.; Mandal, S.M.; Raj, C.R. Antimicrobial Activity of Fluorescent Ag Nanoparticles. Lett. Appl. Microbiol. 2014, 58, 520–526. [Google Scholar] [CrossRef] [PubMed]
  155. Balakumaran, M.D.; Ramachandran, R.; Kalaichelvan, P.T. Exploitation of Endophytic Fungus, Guignardia Mangiferae for Extracellular Synthesis of Silver Nanoparticles and Their in Vitro Biological Activities. Microbiol. Res. 2015, 178, 9–17. [Google Scholar] [CrossRef] [PubMed]
  156. Ma, J.F.; Hiradate, S. Form of Aluminium for Uptake and Translocation in Buckwheat (Fagopyrum Esculentum Moench). Planta 2000, 211, 355–360. [Google Scholar] [CrossRef] [PubMed]
  157. Kim, S.W.; Jung, J.H.; Lamsal, K.; Kim, Y.S.; Min, J.S.; Lee, Y.S. Antifungal Effects of Silver Nanoparticles (AgNPs) against Various Plant Pathogenic Fungi. Mycobiology 2012, 40, 53–58. [Google Scholar] [CrossRef] [Green Version]
  158. Soenen, S.J.; Rivera-Gil, P.; Montenegro, J.-M.; Parak, W.J.; De Smedt, S.C.; Braeckmans, K. Cellular Toxicity of Inorganic Nanoparticles: Common Aspects and Guidelines for Improved Nanotoxicity Evaluation. Nano Today 2011, 6, 446–465. [Google Scholar] [CrossRef]
  159. Al-Otibi, F.; Perveen, K.; Al-Saif, N.A.; Alharbi, R.I.; Bokhari, N.A.; Albasher, G.; Al-Otaibi, R.M.; Al-Mosa, M.A. Biosynthesis of Silver Nanoparticles Using Malva Parviflora and Their Antifungal Activity. Saudi J. Biol. Sci. 2021, 28, 2229–2235. [Google Scholar] [CrossRef]
  160. Yassin, M.A.; Elgorban, A.M.; El-Samawaty, A.E.-R.M.A.; Almunqedhi, B.M.A. Biosynthesis of Silver Nanoparticles Using Penicillium Verrucosum and Analysis of Their Antifungal Activity. Saudi J. Biol. Sci. 2021, 28, 2123–2127. [Google Scholar] [CrossRef]
  161. Nguyen, D.H.; Vo, T.N.N.; Le, N.T.T.; Thi, D.P.N.; Thi, T.T.H. Evaluation of Saponin-Rich/Poor Leaf Extract-Mediated Silver Nanoparticles and Their Antifungal Capacity. Green Process. Synth. 2020, 9, 429–439. [Google Scholar] [CrossRef]
  162. Shafea, T.M.M.; Mahmoud, K.M. Green Synthesis, Characterization and Antimicrobial Activity of Silver Nanoparticles Using Locally Growing Allium Sativum Extract. Zanco J. Pure Appl. Sci. 2021, 33, 35–41. [Google Scholar]
  163. Anum, F.; Jabeen, K.; Javad, S.; Iqbal, S.; Tahir, A.; Javed, Z.; Cruz-Martins, N.; Ayatollahi, S.A.; Sharifi-Rad, J.; Alshehri, M.M. Green Synthesized Silver Nanoparticles as Potent Antifungal Agent against Aspergillus Terreus Thom. J. Nanomater. 2021, 2021, 2992335. [Google Scholar] [CrossRef]
  164. Le, N.T.T.; Nguyen, D.H.; Nguyen, N.H.; Ching, Y.C.; Pham Nguyen, D.Y.; Ngo, C.Q.; Nhat, H.N.T.; Hoang Thi, T.T. Silver Nanoparticles Ecofriendly Synthesized by Achyranthes Aspera and Scoparia Dulcis Leaf Broth as an Effective Fungicide. Appl. Sci. 2020, 10, 2505. [Google Scholar] [CrossRef] [Green Version]
  165. Ghojavand, S.; Madani, M.; Karimi, J. Green Synthesis, Characterization and Antifungal Activity of Silver Nanoparticles Using Stems and Flowers of Felty Germander. J. Inorg. Organomet. Polym. Mater. 2020, 30, 2987–2997. [Google Scholar] [CrossRef]
  166. Bocate, K.P.; Reis, G.F.; de Souza, P.C.; Junior, A.G.O.; Durán, N.; Nakazato, G.; Furlaneto, M.C.; de Almeida, R.S.; Panagio, L.A. Antifungal Activity of Silver Nanoparticles and Simvastatin against Toxigenic Species of Aspergillus. Int. J. Food Microbiol. 2019, 291, 79–86. [Google Scholar] [CrossRef]
  167. Nokkrut, B.; Pisuttipiched, S.; Khantayanuwong, S.; Puangsin, B. Silver Nanoparticle-Based Paper Packaging to Combat Black Anther Disease in Orchid Flowers. Coatings 2019, 9, 40. [Google Scholar] [CrossRef] [Green Version]
  168. Nguyen, D.H.; Vo, T.N.N.; Nguyen, N.T.; Ching, Y.C.; Hoang Thi, T.T. Comparison of Biogenic Silver Nanoparticles Formed by Momordica Charantia and Psidium Guajava Leaf Extract and Antifungal Evaluation. PLoS ONE 2020, 15, e0239360. [Google Scholar] [CrossRef]
  169. Mansoor, S.; Zahoor, I.; Baba, T.R.; Padder, S.A.; Bhat, Z.A.; Koul, A.M.; Jiang, L. Fabrication of Silver Nanoparticles against Fungal Pathogens. Front. Nanotechnol. 2021, 3, 679358. [Google Scholar] [CrossRef]
  170. Vrandečić, K.; Ćosić, J.; Ilić, J.; Ravnjak, B.; Selmani, A.; Galić, E.; Pem, B.; Barbir, R.; Vinković Vrček, I.; Vinković, T. Antifungal Activities of Silver and Selenium Nanoparticles Stabilized with Different Surface Coating Agents. Pest Manag. Sci. 2020, 76, 2021–2029. [Google Scholar] [CrossRef]
  171. Nicol, J.M.; Turner, S.J.; Coyne, D.L.; den Nijs, L.; Hockland, S.; Maafi, Z.T. Current Nematode Threats to World Agriculture. In Genomics and Molecular Genetics of Plant-Nematode Interactions; Springer: Berlin, Germany, 2011; pp. 21–43. [Google Scholar]
  172. Baronia, R.; Kumar, P.; Singh, S.P.; Walia, R.K. Silver Nanoparticles as a Potential Nematicide against Meloidogyne Graminicola. J. Nematol. 2020, 52, 1–9. [Google Scholar] [CrossRef] [Green Version]
  173. Fouda, M.M.G.; Abdelsalam, N.R.; Gohar, I.M.A.; Hanfy, A.E.M.; Othman, S.I.; Zaitoun, A.F.; Allam, A.A.; Morsy, O.M.; El-Naggar, M. Utilization of High Throughput Microcrystalline Cellulose Decorated Silver Nanoparticles as an Eco-Nematicide on Root-Knot Nematodes. Colloids Surf. B Biointerfaces 2020, 188, 110805. [Google Scholar] [CrossRef]
  174. Lim, D.; Roh, J.; Eom, H.; Choi, J.; Hyun, J.; Choi, J. Oxidative Stress-related PMK-1 P38 MAPK Activation as a Mechanism for Toxicity of Silver Nanoparticles to Reproduction in the Nematode Caenorhabditis Elegans. Environ. Toxicol. Chem. 2012, 31, 585–592. [Google Scholar] [CrossRef]
  175. Miura, N.; Shinohara, Y. Cytotoxic Effect and Apoptosis Induction by Silver Nanoparticles in HeLa Cells. Biochem. Biophys. Res. Commun. 2009, 390, 733–737. [Google Scholar] [CrossRef]
  176. Hassan, M.E.M.; Zawam, H.S.; El-Nahas, S.E.M.; Desoukey, A.F. Comparison Study between Silver Nanoparticles and Two Nematicides against Meloidogyne Incognita on Tomato Seedlings. Plant Pathol. J. 2016, 15, 144–151. [Google Scholar] [CrossRef] [Green Version]
  177. Soliman, B.S.; Abbassy, M.A.; Abdel-Rasoul, M.A.; Nassar, A.M. Efficacy of Silver Nanoparticles of Extractives of Artemisia Judaica against Root-Knot Nematode. J. Environ. Stud. Res 2017, 7, 1–13. [Google Scholar]
  178. Nassar, A.M.K. Effectiveness of Silver Nano-Particles of Extracts of Urtica Urens (Urticaceae) against Root-Knot Nematode Meloidogyne Incognita. Asian J. Nematol. 2016, 5, 14–19. [Google Scholar] [CrossRef]
  179. Walters, D.R.; Newton, A.C.; Lyon, G.D. Induced Resistance for Plant Defense: A Sustainable Approach to Crop Protection; John Wiley & Sons: New York, NY, USA, 2014; ISBN 1118371836. [Google Scholar]
  180. Wuyts, N.; De Waele, D.; Swennen, R. Extraction and Partial Characterization of Polyphenol Oxidase from Banana (Musa Acuminata Grande Naine) Roots. Plant Physiol. Biochem. 2006, 44, 308–314. [Google Scholar] [CrossRef]
  181. Fabiyi, O.; Olatunji, G.; Atolani, O.; Olawuyi, O. Preparation of Bio-Nematicidal Nanoparticles of Eucalyptus Officinalis for the Control of Cyst Nematode (Heterodera Sacchari). J. Anim. Plant Sci. 2020, 30, 1172–1177. [Google Scholar]
  182. Kalaiselvi, D.; Mohankumar, A.; Shanmugam, G.; Nivitha, S.; Sundararaj, P. Green Synthesis of Silver Nanoparticles Using Latex Extract of Euphorbia Tirucalli: A Novel Approach for the Management of Root Knot Nematode, Meloidogyne Incognita. Crop Prot. 2019, 117, 108–114. [Google Scholar] [CrossRef]
  183. Abbassy, M.A.; Abdel-Rasoul, M.A.; Nassar, A.M.K.; Soliman, B.S.M. Nematicidal Activity of Silver Nanoparticles of Botanical Products against Root-Knot Nematode, Meloidogyne Incognita. Arch. Phytopathol. Plant Prot. 2017, 50, 909–926. [Google Scholar] [CrossRef]
  184. El-Deen, A.H.N.; El-Deeb, B.A. Effectiveness of Silver Nanoparticles against Root-Knot Nematode, Meloidogyne Incognita Infecting Tomato under Greenhouse Conditions. J. Agric. Sci. 2018, 10, 148–156. [Google Scholar]
  185. Hamed, S.M.; Hagag, E.S.; El-Raouf, N.A. Green Production of Silver Nanoparticles, Evaluation of Their Nematicidal Activity against Meloidogyne Javanica and Their Impact on Growth of Faba Bean. Beni-Suef Univ. J. Basic Appl. Sci. 2019, 8, 9. [Google Scholar] [CrossRef] [Green Version]
  186. Abdellatif, K.F.; Abdelfattah, R.H.; El-Ansary, M.S.M. Green Nanoparticles Engineering on Root-Knot Nematode Infecting Eggplants and Their Effect on Plant DNA Modification. Iran. J. Biotechnol. 2016, 14, 250. [Google Scholar] [CrossRef] [Green Version]
  187. Cromwell, W.A.; Yang, J.; Starr, J.L.; Jo, Y.-K. Nematicidal Effects of Silver Nanoparticles on Root-Knot Nematode in Bermudagrass. J. Nematol. 2014, 46, 261. [Google Scholar]
  188. Ghareeb, R.Y.; EL-Saedy, M.A.M.; Fathy, M.M.; Hafez, E.E.; Basyony, A.B.A. Green Biosynthesized Silver Nanoparticles Using Macroalgae and Their Usage as a Bioagent on Meloidogyne incognita. Plant Archives. 2020, 20, 9682–9692. [Google Scholar]
  189. Ardakani, A.S. Toxicity of Silver, Titanium and Silicon Nanoparticles on the Root-Knot Nematode, Meloidogyne Incognita, and Growth Parameters of Tomato. Nematology 2013, 15, 671–677. [Google Scholar] [CrossRef]
  190. El-Batal, A.I.; Attia, M.S.; Nofel, M.M.; El-Sayyad, G.S. Potential Nematicidal Properties of Silver Boron Nanoparticles: Synthesis, Characterization, in Vitro and in Vivo Root-Knot Nematode (Meloidogyne Incognita) Treatments. J. Clust. Sci. 2019, 30, 687–705. [Google Scholar] [CrossRef]
  191. Ghareeb, R.Y.; Alfy, H.; Fahmy, A.A.; Ali, H.M.; Abdelsalam, N.R. Utilization of Cladophora Glomerata Extract Nanoparticles as Eco-Nematicide and Enhancing the Defense Responses of Tomato Plants Infected by Meloidogyne Javanica. Sci. Rep. 2020, 10, 19968. [Google Scholar] [CrossRef]
  192. Mikhailova, E.O. Silver Nanoparticles: Mechanism of Action and Probable Bio-Application. J. Funct. Biomater. 2020, 11, 84. [Google Scholar] [CrossRef]
  193. Jian, Y.; Chen, X.; Ahmed, T.; Shang, Q.; Zhang, S.; Ma, Z.; Yin, Y. Toxicity and Action Mechanisms of Silver Nanoparticles against the Mycotoxin-Producing Fungus Fusarium Graminearum. J. Adv. Res. 2022, 38, 1–12. [Google Scholar] [CrossRef]
  194. Ahmad, S.A.; Das, S.S.; Khatoon, A.; Ansari, M.T.; Afzal, M.; Hasnain, M.S.; Nayak, A.K. Bactericidal Activity of Silver Nanoparticles: A Mechanistic Review. Mater. Sci. Energy Technol. 2020, 3, 756–769. [Google Scholar] [CrossRef]
  195. Zhang, L.; Wu, L.; Si, Y.; Shu, K. Size-Dependent Cytotoxicity of Silver Nanoparticles to Azotobacter Vinelandii: Growth Inhibition, Cell Injury, Oxidative Stress and Internalization. PLoS ONE 2018, 13, e0209020. [Google Scholar] [CrossRef] [Green Version]
  196. Karn, B.; Kuiken, T.; Otto, M. Nanotechnology and in situ remediation: A review of the benefits and potential risks. Ciência Saúde Coletiva 2011, 16, 165–178. [Google Scholar] [CrossRef] [Green Version]
  197. Bergeson, L.L. Nanosilver: US EPA’s pesticide office considers how best to proceed. Environ. Qual. Manag. 2010, 19, 79–85. [Google Scholar] [CrossRef]
  198. Gurav, A.S.; Kodas, T.T.; Wang, L.-M.; Kauppinen, E.I.; Joutsensaari, J. Generation of nanometer-size fullerene particles via vapor condensation. Chem. Phys. Lett. 1994, 218, 304–308. [Google Scholar] [CrossRef]
  199. Ahmed, B.; Rizvi, A.; Ali, K.; Lee, J.; Zaidi, A.; Khan, M.S.; Musarrat, J. Nanoparticles in the Soil–Plant System: A Review. Environ. Chem. Lett. 2021, 19, 1545–1609. [Google Scholar] [CrossRef]
  200. Morales-Díaz, A.B.; Ortega-Ortíz, H.; Juárez-Maldonado, A.; Cadenas-Pliego, G.; González-Morales, S.; Benavides-Mendoza, A. Application of Nanoelements in Plant Nutrition and Its Impact in Ecosystems. Adv. Nat. Sci. Nanosci. Nanotechnol. 2017, 8, 13001. [Google Scholar] [CrossRef]
Figure 1. Illustration of two approaches of synthesis of AgNPs in left panel: (i) top-down approaches such as mechanical, chemical, and electro-explosion, etc. In bottom-up approaches, inorganic or organic substances are used as a reducing or capping agents. In bottom-up approaches, various techniques, i.e., superficial fluid synthesis, aerosol process, green synthesis (using bacteria, algae, fungi and plant or their biological products) etc. are used. Right panel shows a few proven examples of microbes and plants for AgNPs synthesis.
Figure 1. Illustration of two approaches of synthesis of AgNPs in left panel: (i) top-down approaches such as mechanical, chemical, and electro-explosion, etc. In bottom-up approaches, inorganic or organic substances are used as a reducing or capping agents. In bottom-up approaches, various techniques, i.e., superficial fluid synthesis, aerosol process, green synthesis (using bacteria, algae, fungi and plant or their biological products) etc. are used. Right panel shows a few proven examples of microbes and plants for AgNPs synthesis.
Molecules 27 04754 g001
Figure 2. Illustration of green synthesis of AgNPs from various extracts such as plant parts (i.e., stem, leaf, flower, barks, seeds, and root) and microorganisms. Prepared extracts of known dilutions are mixed with predefined molarity of silver nitrate (AgNO3) solution and stirred/kept under dark conditions for a fixed time, e.g., 72 h. Following harvest, the AgNPs are physicochemically characterized for size, shape, and surface area, charged by techniques such as transmission electron microscopy (TEM), scanning electron microscopy (SEM), UV-Vis spectroscopy, FTIR, XRD, DLS, and zeta-potential, etc., and applied for several purposes such as removal of bacteria, fungi, viruses, or nematodes.
Figure 2. Illustration of green synthesis of AgNPs from various extracts such as plant parts (i.e., stem, leaf, flower, barks, seeds, and root) and microorganisms. Prepared extracts of known dilutions are mixed with predefined molarity of silver nitrate (AgNO3) solution and stirred/kept under dark conditions for a fixed time, e.g., 72 h. Following harvest, the AgNPs are physicochemically characterized for size, shape, and surface area, charged by techniques such as transmission electron microscopy (TEM), scanning electron microscopy (SEM), UV-Vis spectroscopy, FTIR, XRD, DLS, and zeta-potential, etc., and applied for several purposes such as removal of bacteria, fungi, viruses, or nematodes.
Molecules 27 04754 g002
Table 1. Biological synthesis of AgNPs from various plant species.
Table 1. Biological synthesis of AgNPs from various plant species.
PlantsFamilyPlant PartMetabolites and Their StructuresCorresponding Particle Size Refs.
Acacia niloticaFabaceaePod Molecules 27 04754 i001 Molecules 27 04754 i00220–30 nm[52]
Gallic acid (C7H6O5)Ellagic acid (C14H6O8)
Molecules 27 04754 i003 Molecules 27 04754 i004
Epicatechin (C15H14O6)Rutin (C27H30O16)
Ocimum sanctumLamiaceaeFresh leaves Molecules 27 04754 i00515.72 nm[53]
Tannins (tannic acid; C76H52O46)
Molecules 27 04754 i006
Saponin (C58H94O27)
Coleus aromaticusLamiaceaeFresh leaves Molecules 27 04754 i007 Molecules 27 04754 i008 Molecules 27 04754 i00944 nm[54]
Carvacrol (C10H14O)Caryophyllen (C15H24)Patchoulene (C15H24)
Lantana camaraVerbenaceaeFresh leaves Molecules 27 04754 i010 Molecules 27 04754 i01114–27 nm [55]
Phenolic acidTerpenoid
Molecules 27 04754 i012
Lipid
Molecules 27 04754 i013
Carbohydrate
Piper longumPiperaceaeDried fruit Molecules 27 04754 i014 Molecules 27 04754 i015 Molecules 27 04754 i01615–200 nm[56]
Piperidine (C5H11N) Terpinenes (C10H16)Sesamin (C20H18O6)
Moringa oleiferaMoringaceaeFresh stem bark Molecules 27 04754 i017 Molecules 27 04754 i01840 nm[57]
β-sitosterol (C29H50O)Caffeoylquinic acid (C16H18O9)
Syzygium cuminiMyrtaceaeAir dried seeds Molecules 27 04754 i019 Molecules 27 04754 i02040–100 nm[58]
p-coumaric acid (C9H8O3)3,4-dihyroxybenzoic acid (C7H6O4)
Table 2. Biological synthesis of AgNPs by various bacterial strains.
Table 2. Biological synthesis of AgNPs by various bacterial strains.
Bacterial StrainsMetabolitesStructureSizeReferences
Serratia nematodiphila
(a)
Prodigiosin
(b)
Sodorifen
(c)
p-Nitrophenol, etc.
Molecules 27 04754 i021 Molecules 27 04754 i022 Molecules 27 04754 i02365–70 nm[61]
ProdigiosinSodorifenp-Nitrophenol
Bacillus stearothermophilus
(a)
Macrolactin-A
(b)
Bacillibactin, etc.
Molecules 27 04754 i02414 nm[62]
Macrolactin-A
Molecules 27 04754 i025
Bacillibactin
Bacillus strain CS11
(a)
Bacteriocin
(b)
Surfactin, etc.
Molecules 27 04754 i02642–92 nm[63]
Bacteriocin (small)
Molecules 27 04754 i027
Surfactin
Escherichia coli
(a)
13-Tetradecynoic acid
(b)
Hexadecanol, etc.
Molecules 27 04754 i028NA[64]
13-Tetradecynoic acid
Molecules 27 04754 i029
Hexadecanol
Morganella morganii RP42
(a)
Phenyl-β-d-glucoside
(b)
Phenyl ester, etc.
Molecules 27 04754 i030 Molecules 27 04754 i03110–50 nm[65]
Phenyl-β-D-glucosidePhenyl ester
Pseudomonas proteolytica and
Pseudomonas meridiana
(a)
Phenazines
(b)
Hydrogen cyanide
(c)
Pseudomonine, etc.
Molecules 27 04754 i032 Molecules 27 04754 i0334–13 nm[66]
PhenazineHydrogen cyanide
Molecules 27 04754 i034
Pseudomonine
Bacillus brevis
(NCIM 2533)
(a)
Bacilotetrin
(b)
Hetiamacin, etc.
Molecules 27 04754 i035 Molecules 27 04754 i03668 nm[67]
BacilotetrinHetiamacin
Table 3. Biological synthesis of AgNPs by various strains of algae and fungi.
Table 3. Biological synthesis of AgNPs by various strains of algae and fungi.
AlgaeMetabolitesStructureSizeReferences
Sargassum plagiophyllum
(a)
Saponins
(b)
Anthocyanins
(c)
Triterpenes, etc.
Molecules 27 04754 i037 Molecules 27 04754 i03818–42 nm[64]
SaponinAnthocyanin
Molecules 27 04754 i039
Triterpene
Chlorococcum humicola
(a)
Anthraquinone
(b)
Steroids
(c)
1-propene, etc.
Molecules 27 04754 i040 Molecules 27 04754 i0414–6 nm[72]
Anthraquinone1-propene
Molecules 27 04754 i042
Steroid
Amphora-46
(a)
β-carotene
(b)
Catechin
(c)
p-coumaric acid, etc.
Molecules 27 04754 i0435–70 nm[73]
β-carotene
Molecules 27 04754 i044
Catechin
Molecules 27 04754 i045
p-coumaric acid
Caulerpa racemose
(a)
α-tocopherol
(b)
β-sitosterol, etc.
Molecules 27 04754 i0465–25 nm[74]
α-tocopherol
Molecules 27 04754 i047
β-sitosterol
Padina pavonica
(a)
Ferulic acid
(b)
Naringenin
(c)
Kaempferol, etc.
Molecules 27 04754 i048 Molecules 27 04754 i04910–72 nm[75]
Ferulic acidNaringenin
Molecules 27 04754 i050
Kaempferol
Chaetomorpha linum
(a)
Linoleic acid
(b)
Arachidonic acid
(c)
Eicosapentaenoic acid, etc.
Molecules 27 04754 i0513–44 nm[76]
Linoleic acid
Molecules 27 04754 i052
Arachidonic acid
Molecules 27 04754 i053
Eicosapentaenoic acid
Gelidiumamansii
(a)
Lactic acid
(b)
Butyric acid, etc.
Molecules 27 04754 i054 Molecules 27 04754 i05527–54 nm[77]
Lactic acidButyric acid
Fungi
Aspergillus fumigates
(a)
Fumigaclavine
(b)
Fumagillins, etc.
Molecules 27 04754 i0565–25 nm[78]
Fumigaclavine
Molecules 27 04754 i057
Fumagillins
Penicillium fellutanum
(a)
Fellutamide
(b)
Citrinin, etc
Molecules 27 04754 i0585–25 nm[79]
Fellutamide
Molecules 27 04754 i059
Citrinin
Aspergillus flavus
(a)
Aflavarin
(b)
Cladosporin, etc.
Molecules 27 04754 i0608.92 nm[80]
Aflavarin
Molecules 27 04754 i061
Cladosporin
Fusarium semitectum
(a)
Fusapyrone
(b)
Deoxyfusapyrone, etc.
Molecules 27 04754 i06210–60 nm[81]
Fusapyrone
Molecules 27 04754 i063
Deoxyfusapyrone
Alternaria alternata
(a)
Tenuazonic acid
(b)
Maculosin, etc.
Molecules 27 04754 i064 Molecules 27 04754 i06520–60 nm[82]
Tenuazonic acidMaculosin
Table 4. AgNPs in bacterial disease management.
Table 4. AgNPs in bacterial disease management.
AgNP TypesSize (nm)Pathogen(s)Effect(s)References
AgNPs25 to 50Xanthomonas oryzae pv. Oryzae strain LND0005 and Acidovorax oryzae strain RS-1Inhibited bacterial growth, biofilm formation[115]
AgNPs-Bacillus subtilis and Escherichia coliSuppressed the growth of pathogens[116]
AgNPs7 and 25Edwardsiella tarda and E. coliAntibacterial activity[117]
AgNPs27P. aeruginosa and S. marcescens.Antibacterial activity[117]
AgNPs-Klebsiella pneumoniae, P. aeruginosa, S. marcescens, Streptococcus pyogenes, and Staphylococcus aureusIn vitro activity against bacterial pathogens[118]
AgNPs-E. coliSuppressed biofilm formation[119]
AgNPs12Clavibacter michiganensis subsp. michiganensis (Cmm)Inhibited bacterial canker in tomatoes[120]
AgCSs15 to 25X. oryzae pv. oryzaeIn vitro activity against blight disease of rice[121]
AgNPs24.5R. solanacearum strain YY06Negatively affected bacterial growth, biofilm formation, swimming motility, induced cell membrane damage, and reactive oxygen species (ROS) [122]
AgNPs25 to 75E. coli and S. aureusApplied as antibacterial material for fruit and vegetable preservation[123]
AgNPs18 to 39X. oryzae pv. oryzaeIncreased the plant biomass with a decreased levels of cellular ROS[124]
AgNPs470X. phaseoli pv. phaseoliGrowth inhibition[125]
Table 5. AgNPs in viral disease management.
Table 5. AgNPs in viral disease management.
AgNP TypesSize (nm)PlantPathogenEffect(s)References
AgNPs10–20Cymopsis tetragonalobaSunhemp Rosette Virus (SHRV)Complete suppression of the disease[132]
AgNPs77Vicia fabaBean Yellow Mosaic Virus (BYMV)Decrease in virus concentration, percentage of infection and disease severity, reduction in lesions on infected leaves[129]
AgNPs12Solanum tuberosumPotato Virus Y (PVY)Resistance to virus infection[131]
Graphene oxide-silver NPs
(GO-AgNPs)
30–50Lactuca sativaTomato Bushy Stunt Virus (TBSV)Decrease in virus concentration, infection percentage, and disease severity[138]
AgNPs12.6Solanum tuberosum L. cv. SpuntaTomato Spotted Wilt Virus (TSWV)Decrease in TSWV infectivity and produces an inhibitory effect in local lesions[130]
AgNPs-Solanum lycopersicumTomato Mosaic Virus (ToMV) Potato Virus Y (PVY)Reduction in disease severity and virus infection[128]
AgNPs-Autotrophic plantsBanana Bunchy Top Virus (BBTV) Inhibition of apoplastic trafficking by blocking pores and barriers in the cell wall or plasmodesmata[136]
AgNPs S. tuberosumPotato Virus Y (PVY)Induced resistance to virus[138]
Schiff base AgNPs N. tabacumTobacco Mosaic Virus (TMV)Induced resistance to virus by promoting plant immunity[139]
Table 6. AgNPs in fungal disease management.
Table 6. AgNPs in fungal disease management.
NanoparticlesSize (nm)PathogenEffectReferences
AgNPs50.6Helminthosporium rostratum, Fusarium solani, F. oxysporum and Alternaria alternataEffectively mitigated the mycelial growth[159]
AgNPs10–12F. chlamydosporum and Aspergillus flavusSuppressed the growth of pathogens[160]
OT-AgNPs5–61F. oxysporum, A. niger, and A. flavusAntifungal ability[161]
AgNPs15Candida albicansSuppressed the growth of pathogens[162]
AgNPs25.6A. terreus ThomRetardation in fungus growth and biomass[163]
AA.AgNPs and SD.AgNPs8–52 and 5–45A. niger, A. flavus and F. oxysporumHighly antifungal effect against pathogens[164]
AgNPs-F. oxysporumAntifungal activity[165]
AgCSs-Aspergillus sp. Abnormal spore germination and distorted hyphae[166]
AgNPs47Colletotrichum gloeosporioidesControlled black anther infection during storage of cut orchid flowers[167]
MC.AgNPs and PG.AgNPs5–29 and 5–53A. niger, A. flavus and F. oxysporumInhibitory action[168]
AgNPs A. fumigates, A. niger, A. flavus, Trichophyton rubrum, C. albicans, and Penicillium sp.Inhibition of fungal growth and biofilm[169]
AgNPs100M. phaseolina, S. sclerotiorum, and D. longicollaInhibited the growth of fungi[170]
Table 7. AgNPs in nematode disease management.
Table 7. AgNPs in nematode disease management.
NanoparticlesSize (nm)Target NematodeTest CropEffectReferences
AgNPs100Heterodera sacchariOryza sativaDecreased nematode population in the root and soil, improved vegetative development of the rice plant[181]
AgNPs15Meloidogyne incognitaSolanum nigrumApart from nematode movement, impacts on production, embryogenesis, hatchability percentage, and larval stages were evident[173]
Et-AgNPs20–30M. incognitaS. lycopersicumInhibition of J2 worms and prevention of egg hatching (in vitro). In vivo infestation of tomato roots was considerably decreased when a root dip therapy with AgNPs was used[182]
AgNPs30–100M. incognitaS. melongenaInhibition of eggs and 2nd juvenile (J2) stage of M. incognita[183]
AgNPs2M. incognitaArachis hypogeaVegetative growth and fruit weight were increased to varying degrees when the nematode population was diminished[181]
AgNPs50–150M. incognitaS. lycopersicumAntagonistic effect on the nematode eggs and larval stages[177]
AgNPs5–50M. incognitaS. lycopersicumHighest increase in growth parameters, as well as the minimum galls and egg masses[184]
AgNPs20M. graminicolaO. sativaA substantial reduction in the formation of root galls[172]
AgNPs16M. javanicaFaba beanDrastically decreased egg hatching, increased larval mortality, diminished root galling, and J2 population in soils[185]
Green Silver Nanoparticles (GSN)8–19M. javanicaS. melongenaReduced second-stage juveniles (J2s), nematode population in soil, and enhanced growth characteristics[186]
AgNPs-M. incognitaBermuda grassIncreased turfgrass productivity in one year and reduced root gall development in two years without phytotoxicity[187]
AgNPs5–10M. incognitaS. lycopersicumSignificant reduction in the number of galls, egg masses, developmental stage, rate of build up, and nematode population in soil[176]
AgNPs13.09 and 10.51M. javanicaS. lycopersicumIncreased the plant defense gene’s expression (chitinase gene)[188]
AgNPs20M. incognitaS. lycopersicumSecond-stage juvenile immobility and mortality[189]
Ag-BNPs29.55M. incognitaS. lycopersicumReduced the level of second-stage juveniles, females, and developmental stages while improving the host plant’s resistance and immunity[190]
AgNPs25 to 55M. incognitaS. lycopersicumGalls, egg masses, females per root system/plant, and juvenile mortality were all reduced, and the immune system was induced to resist against nematode infection[191]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tariq, M.; Mohammad, K.N.; Ahmed, B.; Siddiqui, M.A.; Lee, J. Biological Synthesis of Silver Nanoparticles and Prospects in Plant Disease Management. Molecules 2022, 27, 4754. https://doi.org/10.3390/molecules27154754

AMA Style

Tariq M, Mohammad KN, Ahmed B, Siddiqui MA, Lee J. Biological Synthesis of Silver Nanoparticles and Prospects in Plant Disease Management. Molecules. 2022; 27(15):4754. https://doi.org/10.3390/molecules27154754

Chicago/Turabian Style

Tariq, Moh, Khan Nazima Mohammad, Bilal Ahmed, Mansoor A. Siddiqui, and Jintae Lee. 2022. "Biological Synthesis of Silver Nanoparticles and Prospects in Plant Disease Management" Molecules 27, no. 15: 4754. https://doi.org/10.3390/molecules27154754

Article Metrics

Back to TopTop