Next Article in Journal
Assimilation of Cholesterol by Monascus purpureus
Next Article in Special Issue
Nanohybrid Antifungals for Control of Plant Diseases: Current Status and Future Perspectives
Previous Article in Journal
Comparison of Three Skin Sampling Methods and Two Media for Culturing Malassezia Yeast
Previous Article in Special Issue
Biosynthesis of Silver Nanoparticles Using Onion Endophytic Bacterium and Its Antifungal Activity against Rice Pathogen Magnaporthe oryzae
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pleurotus Macrofungi-Assisted Nanoparticle Synthesis and Its Potential Applications: A Review

1
School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India
2
Advance School of Chemical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India
3
School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, India
4
Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic
5
Biology Department, College of Sciences, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
6
Electron Microscopy and Nanoscience Laboratory, Punjab Agricultural University, Ludhiana 141004, India
7
School Bioengineering and Food Technology, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India
8
Agricultural Research Center (ARC), Plant Pathology Research Institute, Giza 12619, Egypt
*
Authors to whom correspondence should be addressed.
Submission received: 19 November 2020 / Revised: 6 December 2020 / Accepted: 7 December 2020 / Published: 9 December 2020
(This article belongs to the Special Issue Fungal Nanotechnology)

Abstract

:
Research and innovation in nanoparticles (NPs) synthesis derived from biomaterials have gained much attention due to their unique characteristics, such as low-cost, easy synthesis methods, high water solubility, and eco-friendly nature. NPs derived from macrofungi, including various mushroom species, such as Agaricus bisporus, Pleurotus spp., Lentinus spp., and Ganoderma spp. are well known to possess high nutritional, immune-modulatory, antimicrobial (antibacterial, antifungal and antiviral), antioxidant, and anticancerous properties. Fungi have intracellular metal uptake ability and maximum wall binding capacity; because of which, they have high metal tolerance and bioaccumulation ability. Primarily, two methods have been comprehended in the literature to synthesize metal NPs from macrofungi, i.e., the intracellular method, which refers to NP synthesis inside fungal cells by transportation of ions in the presence of enzymes; and the extracellular method, which refers to the treatment of fungal biomolecules aqueous filtrate with a metal precursor. Pleurotus derived metal NPs are known to inhibit the growth of numerous foodborne pathogenic bacteria and fungi. To the best of our knowledge, there is no such review article reported in the literature describing the synthesis and complete application and mechanism of NPs derived from macrofungi. Herein, we intend to summarize the progressive research on macrofungi derived NPs regarding their synthesis as well as applications in the area of antimicrobial (antibacterial & antifungal), anticancer, antioxidant, catalytic and food preservation. Additionally, the challenges associated with NPs synthesis will also be discussed.

1. Introduction

The enormous impact of nanobiotechnology on almost all life forms has intrigued researchers globally [1]. In 1959, Richard Feynman introduced the theoretical concept of miniaturization, and, for the first time, provided hidden hints on nanotechnology (directing towards technology, using materials that have dimensions of approximately 1–100 nm) [2]. Nowadays, microorganisms (bacterium, fungi, including mushroom, yeast) and green plants are used for green synthesis of metallic nanoparticles [3].
Pleurotus mushrooms, commonly known as oyster mushrooms, belong to the family of genus Pleurotus, and they are edible and nutritious in nature [4]. Oyster mushrooms are readily available and naturally grow in nearly all latitudes, tropical and subtropical forestry, except Antarctica [5]. The worldwide geographical distribution of different Pleurotus species, with optimum growth temperature, is shown in Table 1. The primary role of the fruiting bodies of oyster mushrooms is to absorb amino acids, proteins, vitamin B (niacin, thiamine, and riboflavin), vitamin D, carbohydrates, and mineral salts (iron, calcium, and phosphorus) [6,7]. Additionally, they show important antifungal, anti-inflammatory, antibacterial, and immunomodulatory activities.
Moreover, they have the ability to reduce sugar and cholesterol levels in the blood [11]. P. ostreatus contains β-1,3-D-glucan and pectin, which are water-soluble gel-forming substances, having the ability to bind with bile acids. They inhibit cholesterol-bile micelle formation, cholesterol absorption, and endogenous synthesis, while increase the removal of plasma cholesterol by reducing the production and secretion of very low density lipoproteins (VLDL) [12]. The content of the nutrients depends on the nature, age, and size of the fungus, as well as their growing conditions [13]. Several nutrients, such as carbohydrates (40–46%), protein (20–25%), fiber (10–21%), and amino acids (20–41%) are present in considerable amounts, while the content of fat is very low, ranging from 10–20% of the dry matter. Fungi fruiting bodies are a rich source of micro- and macro-elements, such as sodium, magnesium, phosphorus, calcium, manganese, potassium, iron, copper, and zinc. Mushrooms of the genus Pleurotus have their own importance, more than the commercially employed basidiomycetes, because they possess better superior nutritional, gastronomic, and medical properties than mushrooms (which can be easily cultivated on a broad range of substrates) [14]. Nowadays, mushrooms show significant potential in metal nanoparticle (NP) synthesis and multifaceted applications [15].
In recent decades, several reports on mycogenesis derived NPs have been published [11]. However, the precise mechanisms of synthesis of mycogenic nanomaterials with variable size dimensions and topologies are not well understood (yet). Members of the fungi kingdom include various heterotrophic multicellular eukaryotic organisms. These microbes play an essential role in diverse ecosystems, particularly in the nutrient cycling paradigms. Fungi can be reproduced by both processes, i.e., sexually as well as asexually, and have shown symbiotic relations with bacteria and plants. Fungi groups mostly consist of mildew, mold, rust, yeast, and mushrooms [16].
The benefits and relevant use of fungal cells, such as NP factories, are attributed to the release of high amounts of extracellular enzymes that can serve as bio-reducing (as well as stabilizing) agents for NP synthesis. Moreover, fungal-derived NPs are much better than the bacteria-derived NPs. Constituents, such as enzymes and metabolites secreted by fungal cells, play an important role in synthesizing metal NPs, which reduce the toxicity of substances [15]. Ions are often liable for toxicity. When metal ions in solution are exposed to bacterial cells, they become uniformly distributed in the environment surrounding the bacterial cell, with no specific localization. In contrast, NPs that interact with the bacterial cell wall produce a focal source of ions through continuous release of ions, and cause enhanced toxicity to the cells [17]. Positively charged metal ions can easily bind with the fungal cell surface containing negative charge through electrostatic and cell receptor-specific interactions. Both types of intracellular (as well as extracellular) fungi could be used to synthesize NPs. Fungi exhibit high metal-binding capabilities in comparison to bacteria, and, hence, fungal biomass has gained the attention of researchers, for the production of NPs, at a large-scale. Various metal NPs, such as PdNPs, AgNPs, AuNPs, CuNPs, FeNPs, ZnNPs, TiNPs, and PtNPs could be synthesized using their oxides, nitrides, sulfides, and fungal biomasses [18].
NPs derived from fungal biomasses exhibit distinct optical, physical, and chemical properties, such as high quantum yield, excellent biocompatibility, high photostability, and adequate near-infrared (NIR) light-absorbing capacity, owing to which, they can be used in various chemical and medical fields, such as sensing, medicine, catalysis, and food packaging [19,20]. Oyster mushrooms mediate myco NPs by using spent mushroom substrate (SMS), and have medical importance towards many pathogenic microorganisms, as reported for the first time in 2007 [3].
To the best of our knowledge, many review articles, based on the synthesis of metal NPs derived from diverse sources and their applications, have been reported. Still, few review articles have discussed the synthesis of distinct metal nanoparticles (MNPs) and several applications of oyster mushroom derived NPs, and we attempted to fill the gap. In this article, we intend to discuss NP synthesis derived from oyster mushrooms, using various techniques and different applications. First section discuses the synthesis of nanoparticles by using intra-and extra-cellular methods. Later, different applications involving antioxidant, anticancer, antibacterial, and catalysis, with possible mechanisms of action, have also been discussed.

2. Green Synthesis of Metal-Based Nanoparticles Mediated by Genus Pleurotus

Fungal exploration and implications in the area of nanotechnology are very significant. In previous literature, it was reported that microorganisms, including bacteria, fungi, and yeast, could be used for the synthesis of metal NPs (metal = calcium, gold, silicon, iron, silver, lead, and gypsum) [21]. We observed that fungi have received immense attention owing to their metal bioaccumulation properties, to produce metal NPs [22]. The fungal material includes mycelia, polysaccharides, and proteins are used in the formation of metal nanoparticles; metal NPs of oyster mushroom species were synthesized using mineral salts [23]. Fungi have intracellular metal uptake capabilities and maximum wall binding abilities because they have high metal tolerance plus bioaccumulation abilities [24,25,26,27]. In comparison with other plants and microbes, the mycelia of fungi provides effective hold ability in the bioreactor, as well as in agitation and high flow pressure [28]. Moreover, fungi secrete extracellular enzymes in high amounts, leading to the massive production of enzymes [29]. Reduction of the enzyme, using both intracellular and extracellular ways, help in metal NP synthesis, nanostructure, and biomimetic mineralization [30,31].
During synthesis, fungi extracts serve the function of capping and reducing agents. At the same time, the fungal mycelium exposed to the metal precursor induces fungus to liberate metabolites and enzymes for its survival [11]. Both the fruiting bodies and mycelium of the mushrooms can be utilized for the synthesis of NPs. It has been reported that the Pleurotus species, such as P. ostreatus, are capable of synthesizing NPs, both intracellularly and extracellularly, while other species, such as P. florida, P. cornucopiae var. citrinopileatus, P. platypus, P. ostreatus, P. sajor-caju, P. eous, and P. djamor var. roseus synthesize NPs extracellularly [32,33,34,35,36,37]. Synthesis of Pleurotus derived metal NPs is shown in Table 2 and Figure 1.

2.1. Intracellular Method

This method includes synthesis of NPs inside the fungal cells by transporting ions during the exposure of enzymes [62,63]. First, the mycelia cultures are treated with a metal precursor and then they are incubated in the dark for 24 h. For intracellular identification, mycelia are resuspended in phosphate buffer saline (PBS, pH 7.4) and homogenized with a sonicator. NPs formed by the intracellular technique have a smaller size when compared with the NPs fabricated by the extracellular method [64,65]. Nucleation of particles inside the fungus could be the cause behind the variation in sizes. This technique is slower when compared with the extracellular method for synthesizing metal NPs [64]. As the NPs synthesis starts within the cell, their downstream processing becomes complicated, increasing cost of synthesizing NPs [66,67,68]. However, this type of synthesis technique is suitable for making composite films [69].

2.2. Extracellular Method

The extracellular synthesis method is a facile and cost-effective approach that involves the treatment of fungal biomolecule aqueous filtrate with a metal precursor, where these metal ions are adsorbed on the surface of the cells [31,70,71,72,73]. In this technique, downstream processing is not required, which makes this approach more effective in comparison to the intracellular method. Therefore, the extracellular approach is predominantly used for NP synthesis [74]. Extracellular metabolites synthesized by fungi play a crucial function in their survival when exposed to various environmental stresses, such as temperature variations, toxic materials (e.g., metallic ions), and predators [75]. Moreover, this synthesis method shows the capability of immobilization of metallic ions in a suitable carrier [69].
The accepted mechanism for the metallic NP synthesis is the enzymatic reduction via enzyme reductase, within the fungal cell or on the cell membrane [76]. This probable mechanism proposes fungus-mediated NP synthesis, i.e., the action of electron shuttle quinones, nitrate reductase, or by both. It is observed that, in bacteria and fungi, mainly two forms of enzymes: (1) nitrate reductase, and (2) α-NADPH-dependent reductases, are responsible for the metal and metal oxide NP synthesis [69]. Extracellularly synthesized NPs were stabilized by the enzymes and proteins formed by the fungi. Moreover, it has been observed that high molecular weight protein is associated with the synthesis of NPs, such as NADH-dependent reductase [69]. Furthermore, the phytochemicals found in plants play a vital role in the bioreduction of NPs [76]. In the mushroom extract of Pleurotus spp., phytochemicals, including alkaloids, saponins, anthraquinones, flavonoids, tannins, and steroids are present [14].

3. Different Types of Nanoparticles Derived from Oyster Mushroom

3.1. Silver Nanoparticles (AgNPs)

AgNPs play a significant character in the areas of biological and medical sciences. These NPs could be synthesized by various methods, such as physical, chemical, ionizing radiation methods, etc. [70]. However, all of these methods possess potential drawbacks; particularly, the chemicals utilized in AgNP synthesis through wet chemistry routes are less eco-friendly, expensive, and have high toxicity [46,77,78]. However, fabrication of AgNPs by green synthesis methods can be a better alternative as it is cost effective, non-toxic, and ecologically safe than the other synthesis methods [79]. Various studies on the biosynthesis of AgNPs using powdered basidiocarps and mycelia of different oyster mushroom species, such as P. ostreatus, P. sajor-caju, P. florida, P. cornucopiae var. citrinopileatus, P. giganteus, P. platypus, and P. eous have been reported. These basidiocarps and mycelia were soaked in distilled water, boiled, and then filtered [23,24,25,26,27,28,29,30,31,32,33]. The filtrate was freeze-dried to prepare aqueous extract. Various concentrations of this aqueous extract were incubated with AgNO3 solution to synthesize AgNPs by the reduction of Ag+ ions to Ag° (metal). Unboiled mycelia extract of Pleurotus has also been used to synthesize AgNPs [38]. In their report, they crushed the fruiting bodies and mixed them with deionized water. The content was filtered with filter paper, and then the filtrate was used to synthesize AgNPs, with a size of 6–10 nm, with a spherical shape. Moreover, the synthesized AgNPs were further assessed for antibacterial potential against Escherichia coli and Staphylococcus aureus.
Synthesis of AgNPs was carried using P. tuber-regium mushroom extract and 1 mM AgNO3 solution. The mixture of solutions was stirred at 90 °C for 2 h. Cubical and spherical shaped AgNPs, with an average size of 50 nm, were obtained as a black powder [61]. Debnath et al. synthesized spherical shaped AgNPs with the help of aqueous extract of mushroom (5 mL) and mixed with 95 mL silver nitrate (1 mM, AgNO3) solution to reduce Ag+ to Ago. This solution was kept in an incubator for 3 days at 37 °C, resulting in color change from light yellow to yellowish-brown. The obtained AgNPs were crystalline with a size ranging from 5 to 25 nm. Authors evaluated the antibacterial activity of AgNPs against E. coli, B. subtilis, P. aeruginosa, and S. aureus [51]. Similarly, the synthesis of predominantly spherical shaped AgNPs with a size ranging from 2 to 100 nm was carried by various researchers using mushroom extract and AgNO3 solution [34,37,52,57,80].

3.2. Gold Nanoparticles (AuNPs)

AuNPs synthesis was performed by using edible P. florida mushroom by the photo-irradiation method, and evaluated for anticancer potential against A-549, HeLa, K-562, and MDA-MB cell lines. Initially, 5 g of fresh biomass of P. florida mushroom was washed with deionized water and then cut to small pieces. Later, the chopped pieces were added in 500 mL of double-deionized water, under stirring, for half an hour. These contents were then incubated overnight. That content was then filtered via filter paper. Later, the filtrate of mushroom was used to reduce Au+ into Au° in the presence of bright sunlight to form spherical to triangular-shaped AuNPs in the range of 10–50 nm [48].

3.3. Zinc Sulfide Nanoparticles (ZnS) and Zinc Oxide Nanoparticles (ZnO)

ZnS NPs were fabricated using P. ostreatus extract, ZnCl2, and Na2S solution as the precursor material [55]. Firstly, small pieces of mushrooms were boiled and filtered. Then, different concentrations of the resultant filtrate were mixed with aqueous solutions of ZnCl2 and Na2S solution, and resulting solutions were dried at 120 °C for 2 h. Here, the resultant filtrate was used as a stabilizing (as well as a capping) agent for the fabrication of spherical shaped ZnS NPs. Obtained ZnS NPs was highly crystalline with sizes varying from 2.30 nm to 4.04 nm. The author observed that the diameter of those spherical ZnS NPs was decreasing with the increase in extract amount [55]. ZnONPs were synthesized by using P. djamor extract, 20 mL of mushroom extract added into 80 mL of Zn (NO3)2. The 5H2O (5 mM) solution was continuously mixed for 24 h at room temperature until the color transformed into light pink, which confirmed the synthesis of ZnONPs [43].

3.4. Cadmium Sulfide Nanoparticles Quantum Dots (CdS QDs)

In contrast to traditional fluorescent organic dyes and green fluorescent proteins, CdS QDs seem to be superior as they overcame the limitations associated with different factors, such as spectral overlapping, weak signal intensity, and photobleaching [81]. The multiple characteristics of QDs are high photostability, symmetric, slow decay rates, fine emission spectra, wide absorption cross-sections, and broad absorption spectra. The emission color of QDs depends upon their size and surface chemistry; chemical composition used can be altered from the UV to visible or near NIR wavelengths. The increasing interest in the use of CdS QDs is because they act as luminescent probes and labels for biological imaging, disease diagnosis, and molecular histopathology. The studies revealed that the QDs derived from plants did not aggregate [82]. Borovaya et al. synthesized CdS NPs with the help of aqueous extract of roots of Linaria maroccana, CdSO4, and Na2S. First, the mixture solution was incubated for 4 days at 28 °C resulting in the formation of the clear homogeneous solution with a bright yellow color. This indicates the formation of CdS NPs, which are water-soluble and spherical, with sizes of 5–7 nm [82]. In 2015, again biosynthesis of luminescent CdS NPs using mycelium of P. ostreatus, CdSO4 and Na2S. In brief, CdSO4 solution was mixed with mycelium followed by the incubation for 10 days at 26 °C, followed by the addition of Na2S solution. Obtained NPs were spherically shaped, having the size in the range of 4 to 7 nm. In particular, cadmium sulfide QDs are highly useful in investigating the biomolecules interaction and cellular signaling pathway with the help of fluorescent microscopy [81].

3.5. Titanium Dioxide Nanoparticles (TiO2)

TiO2 NPs were synthesized by using edible P. djamor mushroom and evaluated for anticancer potential against A-549 (human lung carcinoma) cell lines, as well as for larvicidal and bactericidal activity. Initially, 10 g of fresh biomass of P. djamor mushroom was washed with deionized water for 10 min and then cut to small pieces. Later, the chopped pieces were added in 100 mL of double- deionized water, boiled at 60 °C for 15 min, and then filtered. Then, 20 mL of filtrate was added to 80 mL of TiCl4 (5 mM) solution, stirred for 2 h, and kept to room temperature for 20 min until the color changed to brown. The intensity of the color of the extract was determined at the wavelength of 345 nm. The synthesized TiO2 NPs formed, spherical in shape, with sizes of 31 nm [44].

3.6. Synthesis of Other Nanoparticles

3.6.1. Iron Nanoparticles (FeNPs)

FeNPs were intracellularly synthesized by using hypha of Pleurotus sp. The reduction process is involved in uptake of FeNPs via the fungal cell membrane, in which reduction of ferric ion (Fe+3) to ferrous ion (Fe+2) takes place. The reduction process is involved during the iron uptake by fungi [83].

3.6.2. Selenium Nanoparticle (SeNP)

SeNPs were synthesized via mushroom polysaccharide-protein complexes (PSPs) isolated from P. tuber-regium sclerotia. These NPs have anticancer activity, excellent bioavailability, and low toxicity. SeNPs have been recorded for inhibiting the proliferation of human breast carcinoma MCF-7 cells by apoptosis; results obtained from the study revealed that cytotoxicity was cancer-specific [84]. PSP–SeNPs have the efficiency to enhance the reactive oxygen species (ROS) generation, and inhibit dose dependently the growth of MCF-7 human breast carcinoma cells, through induction of apoptosis, with the involvement of Poly (ADP-ribose) polymerase (PARP) cleavage and caspase activation. The size of PSP–SeNPs with an average diameter < 50 nm spherical in shape [85]. The synthesis of SeNPs from P. ostreatus extract have been reported for in vitro anticancer activity [86].

3.6.3. Copper Nanoparticles (CuNPs)

Monodispersed copper nanoparticles (CuNPs) were synthesized from aqueous fermented fenugreek powder (FFP), polysaccharides, such as chitosan, sodium alginate, citrus, and pectin, with the help of fungal strains of P. ostreatus, under the exposure of gamma radiation. The CuNPs synthesized have size ranges from 25.0 to 36.0 nm. Because of the stability and the minute sizes, these synthesized CuNPs show antioxidant and antimicrobial activity, and were found beneficial in cosmetics, medical, pharmaceutical, and industrial applications [87].

4. Applications of Pleurotus Derived Nanoparticles

Different types of metal NPs synthesized from oyster mushrooms are discussed above. Recently, they have been considered as valuable in various fields of medicine and industries. Schematic representation of different applications of metal NPs derived from Pleurotus is summarized in Figure 2.

4.1. Antimicrobial Activity

In the past few decades, the microbial infection has become a major health issue across the world, due to its continuously evolving nature and ability to develop resistance against the existing regime [64]. Therefore, search for a strong alternative candidate that can kill or inhibit multidrug resistance microbes is needed [88]. Metal nanoparticles (MNPs) are known to possess potent antimicrobial activity against a wide variety of microbes, including bacteria (Gram-negative and Gram-positive) and fungi, via their photodynamic effects and strong oxidative stress [89]. Furthermore, the direct physical contact of MNPs to bacterial membrane results in the release of intercellular material, loss of cell membrane integrity, and cell death [88]. In literature, several reports are offered on MNPs’ (AgNPs, AuNPs, ZnSNPs) exhibiting bactericidal activity. However, only a few reports focus on the support of their antifungal and antibacterial activity with appropriate mechanisms [88,89,90,91]. Moreover, many reports available in literature that favor the mechanism involved in bacterial and fungi cells are almost similar.
In the year 2009, Nithya and Ragunathan synthesized AgNPs (5–50 nm, spherical) derived from Pleurotus sajor-caju fungi as the starting material and evaluated their bactericidal activity against the both Gram-negative (Pseudomonas aeruginosa, E. coli) and Gram-positive (S. aureus) bacteria. Authors claimed that the inhibitory action of AgNPs was attributed to the generation of Ag+ ions by NPs that resulted in DNA damage, protein denaturation, enzymes inhibitions [32]. Two years later, in 2011, Bhat et al. also fabricated AgNPs (20 ± 5 nm, spherical) using P. florida mushroom, and studied their antibacterial activity against S. aureus, Salmonella typhi, Providencia alcalifaciens, and Proteus mirabilis. The prepared NPs show higher activity against the Gram-positive microbes than Gram-negative, especially in the case of P. mirabilis [33]. A similar kind of study was presented by Shivashankar et al. (2013) using AgNPs derived from P. pulmonarius, P. djamor, and Hypsizygus (pleurotus) ulmarius as the precursors [40,41].
Yehia and Sheikh (2014) synthesized AgNPs (4–15 nm, spherical) derived from P. ostreatus extracted via the green synthesis route and evaluated their antifungal activity toward the various Candida species, i.e., C. tropicalis, C. albicans, C. parapsilosis, C. krusei, and C. glabrata. The (minimum inhibitory concentration) MIC (IC80) results demonstrated that AgNPs showed higher toxicity against all of the candida species (5–28 μg/mL) than the amphotericin B (5–8 μg/mL) and fluconazole (13–33 μg/mL) [36]. The tiny size and capping ability of the bioactive white NPs derived from P. tuber-regium extract and silver nitrate showed higher therapeutic efficacy against the various diseases and disorders [61]. Devi and Joshi (2015) synthesized AgNPs derived from three different endophytic fungi, i.e., Aspergillus niger, Aspergillus tamarii, and Penicillium ochrochloron isolated from the ethnomedicinal plant Potentilla fulgens leaves via the green synthesis method. The electron microscopy results revealed that all of the AgNPs derived from different fungi were spherically shaped. However, NPs synthesized from A. tamarii showed the smallest size (~3.5 nm) than A. niger (~8.7 nm) and P. ochrochloron (~7.7 nm), respectively [74]. In the year 2018, Bawadekji et al. fabricated Au NPs (~22.9 nm, spherical) from P. ostreatus extract and evaluated their antimicrobial activity toward the bacteria Enterococcus faecalis, E. coli, Klebsiella pneumonia, S. aureus, P. aeruginosa, and C. albicans. The results demonstrated that synthesized NPs showed significant toxicity against C. albicans, P. aeruginosa, and S. aureus. In contrast, no toxicity was observed in the case of E. faecalis, E. coli, and K. pneumonia [54].
Acay and Baran (2019) reported the green synthesis of AgNPs derived from Pleurotus eryngii (PE) extract and their antimicrobial activity against the various human pathogen microorganisms, such as E. coli, S. aureus, Streptococcus pyogenes, P. aeruginosa, and C. albicans. The authors used drug vancomycin, colistin, and fluconazole as the control over the gram-positive, gram-negative, and fungus microorganisms. The observed MIC values for S. aureus, E. coli, S. pyogenes, C. albicans, and P. aeruginosa were 0.035, 0.07, 0.018, 0.07, and 0.035 mg/L, respectively. Authors claimed that AgNPs derived from (PE) Pleurotus eryngii extract could be used as a better alternative, as an antibiotic, compared to the other silver nitrates and antibiotics [45]. After that, Debnath et al. (2019) also fabricated AgNPs from Pleurotus giganteus and analyzed their antibacterial activity [51]. The TiO2 NPs mediated from extract of P. djamor exhibited significant bactericidal activity against human pathogenic bacteria with maximum zone of inhibition P. fluorescens (33 ± 0.2 mm), Corynebacterium diphtheria (32 ± 0.1 mm), S. aureus (32 ± 0.4 mm), and showed higher levels of the inhibitory effect [44]. The P. djamor ZnONPs showed a maximum zone of inhibition against C. diphtheriae (28.6 ± 0.3 mm), P. fluorescens (27 ± 0.5 mm), and S. aureus (26.6 ± 1.5 mm) [43]. The general mechanism of microbial cell death is summarized, as below and in Figure 3.

4.1.1. Antimicrobial Mechanisms

Physical Destruction

In this case, positively charged metal NPs can easily bind to the negatively charged components (i.e., porins, peptidoglycans, and proteins) of the cell membrane via electrostatic interaction, leading to damage of bacterial/fungi membrane, intercellular leakage and, finally, cell inhibition [92].

Oxidative Stress

Another primary mechanism for the antimicrobial activity is based upon the occurrence of oxidative stress, either in the presence of light or under dark conditions. In the microbial cell, the metal NPs can generate ROS, such as •OH and •O2, leading to protein denaturation, DNA damage, enzyme activation, ribosome disassemble and, finally, cell death [92,93]. Furthermore, metal NPs can also act as photoabsorber material upon excitation of light (most often NIR), resulting in cell death. The photothermal effect comes in origin when the emitted electrons from a higher energy state returns to a low energy state, and release their energy in the form of heat and vibrational energy [94].

4.2. Antioxidant Activity

In the human body, excessive reactive free radicals are formed from various sources, such as low diet, mental stress, smoking, and other ailments [95]. Metal NPs exhibited profound antioxidant activities in both intracellular and extracellular environments, as summarized in Figure 4.
In the year 2012, Adebayo et al. synthesized metabolite derived from P. pulmonarius extract and evaluated their radical scavenging ability via α, α-diphenyl-β-picrylhydrazyl (DPPH) radical scavenging assay and the β-carotene-linoleate model method. The metabolite derived from P. pulmonarius extract showed dose-dependent radical scavenging activity. It was found that the existence of glutathione, ascorbic acid, cysteine, tocopherol, polyhydroxy compounds, and aromatic amines in metabolite reduces and decolorizes the violet color of DPPH via hydrogen transferability. Authors claimed that, at a concentration of 2 mg/mL, metabolite showed butylated hydroxyanisole (BHA) (75%), LAU 09 (80%), and α-tocopherol (90%) of inhibition, which attributes to of the presence of phenolic compounds in the extract [14]. A few years later, in 2017, Madhanraj et al. synthesized gold (Au) and silver (Ag) nanoparticles derived from edible mushroom (basidiomycetes) and studied their antioxidant activity via various radical scavenging assays. Both the prepared NPs (Au & Ag) showed significant antioxidant activity in a cell-free system [39]. Acay and Baran (2020), synthesized P. eryngii AgNPs and evaluated their radical scavenging ability via DPPH, chelation of ferrous ions reducing power, and the β-carotene-linoleate model method, and found that, at a concentration of 10 mg/mL, antioxidant activities were 85%, 82%, and 77%, respectively [96]. Zinc plays a role in protecting cells from oxidative stress and acts as an antioxidant. The ZnONPs derived with the help of P. djamor possess strong antioxidant properties (DPPH 59%, H2O2 59.65%, and ABTS 59.30%), with IC50 values of 428.35 µg/mL, 417.22 lg/mL, and 500 lg/mL), respectively [43].
Two possible primary mechanisms for the antioxidant activity are; (i) hydrogen atom transfer, and (ii) single electron transfer [92]. Excessive free radicals could be neutralized or terminated via donating a hydrogen atom that includes total oxyradical scavenging capacity assay, inhibition of induced low-density lipoprotein oxidation, oxygen radical absorbance capability, and radical-trapping antioxidant parameters [78]. On the other hand, the single-electron transfer involves the reduction of compounds, such as radicals, metals and carbonyls by transferring one electron, including change in the color when the compound is reduced, such as Ferric Reducing Antioxidant Potential (FRAP), 2,2-diphenyl-1-picrylhydrazyl radical (DPPH) [92]. However, in the intracellular level, metal NPs enter inside the cells via endocytosis and decrease the ROS levels generated by any probe; for example, 2, 7′-dichlorodihydrofluorescein diacetate (DCFDA) [92].

4.3. Anticancer Activity

In addition to antibacterial and antioxidant activity, metal NPs derived from fungi and other sources have been known to possess outstanding anticancer activity because of their profound ROS generation ability under the dark and light exposure [92,97,98,99]. Sankar et al. (2013) studied the anticancer activity of AgNPs (~136 nm) derived from Origanum vulgare extract against the human lung epithelial cells (A549 cells). AgNPs exhibited dose-dependent toxicity against the A549 cells by 85% inhibition at the dose of 500 μg/mL [100]. Bhat et al. (2013) fabricated Au NPs (12–15 nm, spherical) derived from P. florida mushroom extract via the photo-irradiated method and evaluated their anticancer activity against the A-549, MDA-MB, HeLa, and K-562 cell lines. The prepared AuNPs showed concentration-dependent activity against all cell lines in between 10 and 30 μg/mL [48].
Gliga et al. (2014) attempted to understand the coating and size-dependent toxicity of the AgNPs toward the human lung cells (BEAS-2B cells) with an appropriate mechanism. The results confirmed that prepared NPs with size <10 nm showed the highest toxicity against the BEAS-2B cells, which attributes for its aggregation in cell medium, intracellular localization, cellular uptake, and formation of Ag ions intracellularly. However, it is confirmed that all of the AgNPs showed toxicity against BEAS-2B cells via an increase in overall DNA damage within 24 h [101]. In the same year, Yehia and Sheikh (2014) used P. ostreatus derived AgNPs (4–15 nm, spherical) as an anticancer agent against the MCF-7 cells. The prepared AgNPs showed dose-dependent cell inhibition ranging from 5% to 78% at concentration 10 to 640 μg/mL [36].
Similarly, in the year 2015, Ismail et al., fabricated AgNPs derived from P. ostreatus extract and studied their anticancer effect against the HepG2 and MCF-7 adenocarcinoma cancer cell lines. The authors claimed that NPs induced cytotoxicity toward cancer cells attributes for the formation of ROS species, apoptosis, necrosis, and cell death. ROS are the highly reactive species that result in oxidative damage of proteins, DNA, and induce mitochondrial dysfunction, as summarized in Figure 3 [102]. Similarly, Raman et al. (2015) used P. djamor var. roseus derived AgNPs as anticancer agent toward the human prostrate carcinoma PC3 cells [42]. In the year 2013 and 2014, Priyaragini and Kim et al. demonstrated that metal NPs are harmless at a lower concentration and may be lethal at a higher dose toward normal healthy cells. Many reports revealed the biosynthetic routes to synthesize AgNPs as an anticancer agent against various cell lines. However, AgNPs synthesized using green methods also showed a sort of cytotoxicity against tumor cells [103,104]. Even the extensive use of artificial AgNPs has been already reported, but still, there are limited studies to regulate the cytotoxic effects of AgNPs [36]. Studies on P. eryngii (PE) AgNPs showed cytotoxic activity of HeLa with maximum inhibitory effect 73.46% at 60 μg/mL concentration, PC-3 99.02% at 10 μg/mL and MCF-7 cells 93.89% at 20 μg/mL concentration with IC50 values of 46.594, 2.185, and 6.169 μg/mL, respectively, during a 24-h incubation period [90]. Chaturvedi et al. (2020) studied cytotoxic activity and revealed that the AgNPs and AuNPs mediated from P. sajor-caju extract (PS) showed effective results against HCT-116 cancer cell line. HCT-116 cancer cells viability showed inhibition by P. sajor-caju extract, Au NPs as well as Ag NPs showing IC50 value of 60, 80, and 50 µg/mL respectively. The study revealed that the green synthesized AgNPs showed high antiproliferative activity in contrast to other PS extract and Au NPs, and the reason behind the mechanism was due to the generation of more ROS, leading to oxidative stress, resulting in undeviated damage of protein functionality and integrity [60]. The anticancer activity of TiO2 NPs showed potential toxic effect against human lung cancer (A549) cell lines with maximum inhibited growth of 64% at concentration of 100 µg/mL, after 24 h of exposure [44]. The anticancer activity evaluated from P. djamor ZnONPs showed potent inhibitory on A549 cancerous cells with (LC50(Lethal concentration required to kill 50% of population) value as 42.26 µg/mL) in a dose-dependent manner [43].

4.4. Histopathological Study and Larvicidal Activity

The histopathological profile of TiO2NPs mediated from a P. djamor extract treated mosquito (Aedes aegypti and Culex quinquefasciatus) resulted in the complete collapse of caeca, digestive tract, and desertion of the cuticle and epithelial layer, with harsh damage to the mid-and hind-gut, muscles, as well as nerve ganglia of the brush border. The treating of TiO2 NPs on IVth instar larvae of Ae. aegypti and Cx. quinquefasciatus resulted in larvicidal activity with LC50 (5.88 and 4.84 µg/L) and LC90 (22.80 and 19.33 µg/L) [44]. The Ae. aegypti larvae treated with ZnONPs showed morphological alteration in the digestive tract, wrecked membrane, midgut, and severe damaging of the brush border, cortex with hyperplasia of gut epithelial cells, and variations in the cytoplasmic masses. The larvae of Cx. quinquefasciatus showed the complete putrefaction of abdominal parts, specifically in the caeca, mid-gut, and epithelial layer [43].

4.5. Antidiabetic Activity

The antidiabetic activity was investigated in vitro through the inhibition of α- amylase, an enzyme that digests starch. AgNPs synthesized from P. giganteus possess good α-amylase inhibition activity, which helps in making diabetic drugs; inhibition percentage can be increased with increasing concentration of biosynthesized AgNPs [51].

4.6. Removal of Dyes

El-Batal et al. (2014) extracted fungal laccase derived from P. ostreatus via solid fermentation. The authors demonstrated that this enzyme could be used to decolorize/degrade numerous dyes, i.e., methyl orange, trypan blue, ramazol brilliant red, and ramazol brilliant yellow with more than fifty percent decolorization in their color within 3 h, confirming the laccase degrading ability. The highest reduction was observed for the methyl orange and trypan blue. Furthermore, laccase enzyme was used to synthesize gold NPs, proving that laccase obtained from P. ostreatus had strong potential in many significant industrial applications, for example, in biological pretreatment processes [105,106].

4.7. Catalytic Activity

The recent use of the 4-nitrophenol and derivatives in the manufacturing of insecticides, herbicides, and dyestuffs cause harm to the environment as common wastewater pollutants. Because of their high toxicity, it is challenging to eliminate these pollutants, which is a primary environmental concern. In the year 2007, Panigrahi et al. prepared citrate-capped negatively charged Au NPs (8−55 nm, spherical) for the catalytic degradation of aromatic nitro compounds. The authors claimed that the rate of the reaction rose with the rise in the loading of the catalyst, and decreased in particles size, clearly reflecting the catalytic behavior of gold nanoparticles against aromatic compounds, resulting in amino-compounds [107]. Similarly, Lim et al. (2016) prepared gold nanoparticles (AC-Au NPs, 16.88 ± 5.47~29.93 ± 9.80 nm, spherical) from Agrostis capillaris extract, and studied their catalytic efficacy in the presence of NaBH4 against the 4-nitrophenol. They demonstrated that particle size falls with the rise in extract concentration during the synthesis process. It was observed that the catalytic degradation of 4-nitrophenol rises as the particles size decreases [108].
In the same year, 2016, Rostami-Vartooni et al. developed AgNPs (8–35 nm, spherical) loaded on perlite (sheet-like) using Hamamelis virginiana leaf extract and evaluated their catalytic activity against the 4-nitrophenol and Congo red (CR) dye. The authors demonstrated that, with the rise in the concentration of NaBH4 and AgNPs/perlite, the degradation time of 4-nitrophenol decreases, respectively. The AgNPs supported on the surface of perlite facilitate the electron relay from BH4- to 4-nitrophenol as well as CR dye. Furthermore, they claimed that AgNPs/perlite showed high stability and could be used up to 4 times with significant degradation efficacy [109]. Later, Gopalakrishnan et al. (2017) reported the catalytic degradation of 4-nitrophenol to 4-aminophenol via NaBH4 in the presence of PdNPs (<20 nm, spherical) derived from seed extract of Silybum marianum. The total reduction action was attained within 27 min and is attributed to the relay of electrons from BH4- to 4-nitrophenol, resulting in 4-aminophenol. However, the authors claimed that no reduction was detected in the case of bare NaBH4 [110]. A similar kind of 4-nitrophenol reduction was also performed by Sen et al. (2013) using P. florida derived AuNPs and NaBH4 [49].

4.8. Food Packaging and Preservation

Biocompatible fabricated zinc NPs might be efficiently applied in the biomedical and food packaging fields. The potential antimicrobial action of mushroom against many foodborne bacteria, such as Escherichia coli, Streptococcus faecalis, Bacillus subtilis, Micrococcus luteus, and Listeria innocua could be considered as a boon for the food industry since, by using metal NPs, the contamination of foodstuffs can be avoided, besides for long-time preservation [55,111].

5. Conclusions

Nanomaterials derived from oyster mushrooms have been found to possess great potential over a wide range of applications, especially in the biomedical field. In the present review, we discussed the progress of research, to date, on metal nanoparticles and other nanomaterials derived from oyster mushrooms, regarding their synthesis and applications, particularly in the areas of antimicrobial, larvicidal, antioxidant, anticancer, and catalysis. Generally, AgNPs derived from Pleurotus spp. have a higher synthesis and biomedical applications among mushrooms. The importance of derived nanoparticles is due to their unique characteristics, such as cost effective, crystalline nature, nanosize, and non-hazardous nature. Mainly there are two well-known methods of synthesis, i.e., intracellular and extracellular. It is noticed that nanomaterials derived from oyster mushrooms showed profound applications in the areas of biomedicine and catalysis, but some areas of research are needed to be addressed, which are as follows:
  • To date, oyster mushroom derived NPs are not directly applied to the live samples. Hence, the progress can be made in this direction.
  • Available literature provides evidence that considerable work has been carried out for ascertaining the efficacy of oyster mushroom derived NPs under in vitro conditions against the various cancer cell lines. As less information is available regarding in vivo studies, there is a need for further exploration.
  • More studies are needed to define oyster mushrooms that can be genetically engineered to produce more enzymes primarily involved in NP synthesis, and to expand the knowledge and functions of nanomaterial, so that significant achievements could be attained in the fields of medicine, electronics, cosmetics, agriculture, the environment, and many more.

Author Contributions

Conceptualization, P.B., K.A.A.-E., and K.K.; manuscript writing, K.B., A.S. (Anirudh Sharma); manuscript editing, N.T., S.B., A.K. (Anu Kalia), A.K. (Anil Kumar), A.S. (Ashwag Shami). Critical revising, P.B., K.A.A.-E., E.N., and K.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the University of Hradec Kralove (Faculty of Science VT2019-2021) and Excellence project UHK.

Acknowledgments

We acknowledge the University of Hradec Kralove (Faculty of Science, VT2019-2021) and Excellence project UHK.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bhardwaj, K.; Dhanjal, D.S.; Sharma, A.; Nepovimova, E.; Kalia, A.; Thakur, S.; Bhardwaj, S.; Chopra, C.; Singh, R.; Verma, R.; et al. Conifer-derived metallic nanoparticles: Green synthesis and biological applications. Int. J. Mol. Sci. 2020, 21, 9028. [Google Scholar] [CrossRef]
  2. Kumar, H.; Bhardwaj, K.; Kuča, K.; Kalia, A.; Nepovimova, E.; Verma, R.; Kumar, D. Flower-Based Green Synthesis of Metallic Nanoparticles: Applications beyond Fragrance. Nanomaterials 2020, 10, 766. [Google Scholar] [CrossRef] [Green Version]
  3. Owaid, M.N. Biomedical Applications of Nanoparticles Synthesized from Mushrooms. In Green Nanoparticles; Patra, J., Fraceto, L., Das, G., Campos, E., Eds.; Springer: Cham, Switzerland, 2020; pp. 289–303. [Google Scholar]
  4. Xu, H.; Yao, L.; Sun, H.; Wu, Y. Chemical composition and antitumor activity of different polysaccharides from the roots of Actinidia eriantha. Carbohydr. Polym. 2009, 78, 316–322. [Google Scholar] [CrossRef]
  5. Sykes, E.A.; Chen, J.; Zheng, G.; Chan, W.C.W. Investigating the impact of nanoparticle size on active and passive tumor targeting efficiency. ACS Nano 2014, 8, 5696–5706. [Google Scholar] [CrossRef] [PubMed]
  6. Shankar, S.S.; Rai, A.; Ankamwar, B.; Singh, A.; Ahmad, A.; Sastry, M. Biological synthesis of triangular gold nanoprisms. Nat. Mater. 2004, 3, 482–488. [Google Scholar] [CrossRef] [PubMed]
  7. Philip, D. Biosynthesis of Au, Ag and Au-Ag nanoparticles using edible mushroom extract. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2009, 73, 374–381. [Google Scholar] [CrossRef] [PubMed]
  8. Maftoun, P.; Johari, H.; Soltani, M.; Malik, R.; Othman, N.Z.; El Enshasy, H.A. The edible mushroom Pleurotus spp.: I. Biodiversity and nutritional values. Int. J. Biotechnol. Wellness Ind. 2015, 4, 67–83. [Google Scholar]
  9. Raman, J.; Jang, K.-Y.; Oh, Y.-L.; Oh, M.; Im, J.-H.; Lakshmanan, H.; Sabaratnam, V. Cultivation and Nutritional Value of Prominent Pleurotus spp.: An Overview. Mycobiology 2020, 1–14. [Google Scholar] [CrossRef]
  10. Kumla, J.; Suwannarach, N.; Jaiyasen, A.; Bussaban, B.; Lumyong, S. Development of an edible wild strain of Thai oyster mushroom for economic mushroom production. Chiang Mai J. Sci. 2013, 40, 161–172. [Google Scholar]
  11. Mirunalini, S.; Arulmozhi, V.; Deepalakshmi, K.; Krishnaveni, M. Intracellular biosynthesis and antibacterial activity of silver nanoparticles using edible mushrooms. Not. Sci. Biol. 2012, 4, 55–61. [Google Scholar] [CrossRef]
  12. Mowsurni, F.R.; Chowdhury, M.B.K. Oyster mushroom: Biochemical and medicinal prospects. Bangladesh J. Med. Biochem. 2010, 3, 23–28. [Google Scholar] [CrossRef]
  13. Lok, C.N.; Ho, C.M.; Chen, R.; He, Q.Y.; Yu, W.Y.; Sun, H.; Tam, P.K.H.; Chiu, J.F.; Che, C.M. Silver nanoparticles: Partial oxidation and antibacterial activities. J. Biol. Inorg. Chem. 2007, 12, 527–534. [Google Scholar] [CrossRef] [PubMed]
  14. Adebayo, E.A.; Oloke, J.K.; Ayandele, A.A.; Adegunlola, C. Phytochemical, antioxidant and antimicrobial assay of mushroom metabolite from Pleurotus pulmonarius—LAU 09 (JF736658). J. Microbiol. Biotechnol. Res. 2012, 2, 366–374. [Google Scholar]
  15. Owaid, M.N.; Ibraheem, I.J. Mycosynthesis of nanoparticles using edible and medicinal mushrooms. Eur. J. Nanomed. 2017, 9, 5–23. [Google Scholar] [CrossRef]
  16. Duhan, J.S.; Kumar, R.; Kumar, N.; Kaur, P.; Nehra, K.; Duhan, S. Nanotechnology: The new perspective in precision agriculture. Biotechnol. Rep. 2017, 15, 11–23. [Google Scholar] [CrossRef] [PubMed]
  17. McQuillan, J.S.; Groenaga Infante, H.; Stokes, E.; Shaw, A.M. Silver nanoparticle enhanced silver ion stress response in Escherichia coli K12. Nanotoxicology 2012, 6, 857–866. [Google Scholar] [CrossRef]
  18. Gade, A.; Ingle, A.; Whiteley, C.; Rai, M. Mycogenic metal nanoparticles: Progress and applications. Biotechnol. Lett. 2010, 32, 593–600. [Google Scholar] [CrossRef]
  19. Kreibig, U.; Vollmer, M. Optical Properties of Metal Clusters; Springer Series in Materials Science; Springer: Berlin/Heidelberg, Germany, 1995; Volume 25, ISBN 978-3-642-08191-0. [Google Scholar]
  20. Narayanan, R.; El-Sayed, M.A. Effect of Catalytic Activity on the Metallic Nanoparticle Size Distribution: Electron-Transfer Reaction between Fe(CN)6 and Thiosulfate Ions Catalyzed by PVP—Platinum Nanoparticles. J. Phys. Chem. B 2003, 107, 12416–12424. [Google Scholar] [CrossRef]
  21. Asmathunisha, N.; Kathiresan, K. A review on biosynthesis of nanoparticles by marine organisms. Colloids Surf. B Biointerfaces 2013, 103, 283–287. [Google Scholar] [CrossRef]
  22. Sastry, M.; Ahmad, A.; Khan, M.I.; Kumar, R. Biosynthesis of metal nanoparticles using fungi and actinomycete. Curr. Sci. 2003, 85, 162–170. [Google Scholar]
  23. Owaid, M.N. Green synthesis of silver nanoparticles by Pleurotus (oyster mushroom) and their bioactivity: Review. Environ. Nanotechnol. Monit. Manag. 2019, 12, 100256. [Google Scholar] [CrossRef]
  24. Naveen, H.K.; Kumar, G.; Rao, B.K. Extracellular biosynthesis of silver nanoparticles using the filamentous fungus Penicillium sp. Arch. Appl. Sci. Res. 2010, 2, 161–167. [Google Scholar]
  25. Ingle, A.; Gade, A.; Pierrat, S.; Sonnichsen, C.; Rai, M. Mycosynthesis of Silver Nanoparticles Using the Fungus Fusarium acuminatum and Its Activity against Some Human Pathogenic Bacteria. Curr. Nanosci. 2008, 4, 141–144. [Google Scholar] [CrossRef]
  26. Jain, N.; Bhargava, A.; Majumdar, S.; Tarafdar, J.C.; Panwar, J. Extracellular biosynthesis and characterization of silver nanoparticles using Aspergillus flavus NJP08: A mechanism perspective. Nanoscale 2011, 3, 635–641. [Google Scholar] [CrossRef]
  27. Sanghi, R.; Verma, P. Biomimetic synthesis and characterisation of protein capped silver nanoparticles. Bioresour. Technol. 2009, 100, 501–504. [Google Scholar] [CrossRef]
  28. Soni, N.; Prakash, S. Synthesis of gold nanoparticles by the fungus Aspergillus niger and its efficacy against mosquito larvae. Rep. Parasitol. 2012, 2, 7. [Google Scholar] [CrossRef] [Green Version]
  29. Castro-Longoria, E.; Moreno-Velásquez, S.D.; Vilchis-Nestor, A.R.; Arenas-Berumen, E.; Avalos-Borja, M. Production of platinum nanoparticles and nanoaggregates using Neurospora crassa. J. Microbiol. Biotechnol. 2012, 22, 1000–1004. [Google Scholar] [CrossRef] [Green Version]
  30. Ahmad, A.; Senapati, S.; Khan, M.I.; Kumar, R.; Ramani, R.; Srinivas, V.; Sastry, M. Intracellular synthesis of gold nanoparticles by a novel alkalotolerant actinomycete, Rhodococcus species. Nanotechnology 2003, 14, 824–828. [Google Scholar] [CrossRef]
  31. Durán, N.; Marcato, P.D.; Alves, O.L.; De Souza, G.I.H.; Esposito, E. Mechanistic aspects of biosynthesis of silver nanoparticles by several Fusarium oxysporum strains. J. Nanobiotechnol. 2005, 3, 8. [Google Scholar] [CrossRef] [Green Version]
  32. Nithya, R.; Ragunathan, R. Synthesis of Silver Nanoparticle using Pleurotus Sajor Caju and Its Antimicrobial Study. Dig. J. Nanomater. Biostruct. 2009, 4, 623–629. [Google Scholar]
  33. Bhat, R.; Deshpande, R.; Ganachari, S.V.; Huh, D.S.; Venkataraman, A. Photo-irradiated biosynthesis of silver nanoparticles using edible mushroom Pleurotus florida and their antibacterial activity studies. Bioinorg. Chem. Appl. 2011, 2011, 650979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sujatha, S.; Tamilselvi, S.; Subha, K.; Panneerselvam, A. Studies on biosynthesis of silver nanoparticles using mushroom and its antibacterial activities. Int. J. Curr. Microbiol. Appl. Sci. 2013, 2, 605–614. [Google Scholar]
  35. Owaid, M.N. Testing Efficiency of Different Agriculture Media in Growth and Production of Four Species of Oyster Mushroom Pleurotus and Evaluation the Bioactivity of Tested Species. Ph.D. Thesis, College of Science, University of Anbar, Ramadi, Iraq, 2013. [Google Scholar]
  36. Yehia, R.S.; Al-Sheikh, H. Biosynthesis and characterization of silver nanoparticles produced by pleurotus ostreatus and their anticandidal and anticancer activities. World J. Microbiol. Biotechnol. 2014, 30, 2797–2803. [Google Scholar] [CrossRef] [PubMed]
  37. Owaid, M.N.; Raman, J.; Lakshmanan, H.; Al-Saeedi, S.S.S.; Sabaratnam, V.; Ali Abed, I. Mycosynthesis of silver nanoparticles by Pleurotus cornucopiae var. citrinopileatus and its inhibitory effects against Candida sp. Mater. Lett. 2015, 153, 186–190. [Google Scholar] [CrossRef]
  38. Maurya, S.; Bhardwaj, A.K.; Gupta, K.K.; Agarwal, S.; Kushwaha, A.; Vk, C.; Pathak, R.K.; Gopal, R.; Uttam, K.N.; Singh, A.K. Green synthesis of silver nanoparticles using Pleurotus and its bactericidal activity. Cell. Mol. Biol. 2016, 62, 131. [Google Scholar]
  39. Madhanraj, R.; Eyini, M.; Balaji, P. Antioxidant Assay of Gold and Silver Nanoparticles from Edible Basidiomycetes Mushroom Fungi. Free Radic. Antioxid. 2017, 7, 137–142. [Google Scholar] [CrossRef]
  40. Shivashankar, M.; Premkumari, B.; Chandan, N. Comparative studies on biosynthesis, partial characterization of silver nanoparticles and antimicrobial activities in some edible mushroom. Nano Sci. Nano Technol. Indian J. 2013, 8, 8–14. [Google Scholar]
  41. Shivashankar, M.; Premkumari, B.; Chandan, N. Biosynthesis, partial characterization and antimicrobial activities of silver nanoparticles from pleurotus species. Int. J. Integr. Sci. Innov. Technol. 2013, 2, 13–23. [Google Scholar]
  42. Raman, J.; Reddy, G.R.; Lakshmanan, H.; Selvaraj, V.; Gajendran, B.; Nanjian, R.; Chinnasamy, A.; Sabaratnam, V. Mycosynthesis and characterization of silver nanoparticles from Pleurotus djamor var. roseus and their in vitro cytotoxicity effect on PC3 cells. Process Biochem. 2015, 50, 140–147. [Google Scholar] [CrossRef]
  43. Manimaran, K.; Balasubramani, G.; Ragavendran, C.; Natarajan, D.; Murugesan, S. Biological Applications of Synthesized ZnO Nanoparticles Using Pleurotus Djamor against Mosquito Larvicidal, Histopathology, Antibacterial, Antioxidant and Anticancer Effect. J. Clust. Sci. 2020, 1–13. [Google Scholar] [CrossRef]
  44. Manimaran, K.; Murugesan, S.; Ragavendran, C.; Balasubramani, G.; Natarajan, D.; Ganesan, A.; Seedevi, P. Biosynthesis of TiO2 Nanoparticles Using Edible Mushroom (Pleurotus djamor) Extract: Mosquito Larvicidal, Histopathological, Antibacterial and Anticancer Effect. J. Clust. Sci. 2020, 1–12. [Google Scholar] [CrossRef]
  45. Acay, H.; Baran, M.F. Biosynthesis and characterization of silver nanoparticles using king oyster (Pleurotus eryngii) extract: Effect on some microorganisms. Appl. Ecol. Environ. Res. 2019, 17, 9205–9214. [Google Scholar] [CrossRef]
  46. Manzoor-ul-Haq, R.V.; Patil, S.; Singh, D.; Krishnaveni, R. Isolation and Screening of Mushrooms for Potent Silver Nanoparticles Production from Bandipora District (Jammu and Kashmir) and their characterization. Int. J. Curr. Microbiol. Appl. Sci 2014, 3, 704–714. [Google Scholar]
  47. Kaur, T.; Kapoor, S.; Kalia, A. Synthesis of Silver Nanoparticles from Pleurotus florida, Characterization and Analysis of their Antimicrobial Activity. Int. J. Curr. Microbiol. Appl. Sci 2018, 7, 4085–4095. [Google Scholar] [CrossRef]
  48. Bhat, R.; Sharanabasava, V.G.; Deshpande, R.; Shetti, U.; Sanjeev, G.; Venkataraman, A. Photo-bio-synthesis of irregular shaped functionalized gold nanoparticles using edible mushroom Pleurotus florida and its anticancer evaluation. J. Photochem. Photobiol. B Biol. 2013, 125, 63–69. [Google Scholar] [CrossRef]
  49. Sen, I.K.; Maity, K.; Islam, S.S. Green synthesis of gold nanoparticles using a glucan of an edible mushroom and study of catalytic activity. Carbohydr. Polym. 2013, 91, 518–528. [Google Scholar] [CrossRef]
  50. Gurunathan, S.; Han, J.W.; Park, J.H.; Kim, J.H. A green chemistry approach for synthesizing biocompatible gold nanoparticles. Nanoscale Res. Lett. 2014, 9, 248. [Google Scholar] [CrossRef] [Green Version]
  51. Debnath, G.; Das, P.; Saha, A.K. Green Synthesis of Silver Nanoparticles Using Mushroom Extract of Pleurotus giganteus: Characterization, Antimicrobial, and α-Amylase Inhibitory Activity. Bionanoscience 2019, 9, 611–619. [Google Scholar] [CrossRef]
  52. Al-Bahrani, R.; Raman, J.; Lakshmanan, H.; Hassan, A.A.; Sabaratnam, V. Green synthesis of silver nanoparticles using tree oyster mushroom Pleurotus ostreatus and its inhibitory activity against pathogenic bacteria. Mater. Lett. 2017, 186, 21–25. [Google Scholar] [CrossRef]
  53. Devika, R.; Elumalai, S.; Manikandan, E.; Eswaramoorthy, D. Biosynthesis of Silver Nanoparticles Using the Fungus Pleurotus ostreatus and their Antibacterial Activity. Open Access Sci. Rep. 2012, 1, 557. [Google Scholar] [CrossRef]
  54. Bawadekji, A.; Oueslati, M.H.; Ali, A.; Basha, J. Biosynthesis of Gold Nanoparticles using Pleurotus ostreatus (Jacq. ex. Fr.) Kummer Extract and their Antibacterial and Antifungal Activities. J. Appl. Environ. Biol. Sci. 2018, 8, 142–147. [Google Scholar]
  55. Senapati, U.S.; Sarkar, D. Characterization of biosynthesized zinc sulphide nanoparticles using edible mushroom Pleurotuss ostreatu. Indian J. Phys. 2014, 88, 557–562. [Google Scholar] [CrossRef]
  56. Wu, H.F.; Kailasa, S.K.; Shastri, L. Electrostatically self-assembled azides on zinc sulfide nanoparticles as multifunctional nanoprobes for peptide and protein analysis in MALDI-TOF MS. Talanta 2010, 82, 540–547. [Google Scholar] [CrossRef] [PubMed]
  57. Musa, S.F.; Yeat, T.S.; Kamal, L.Z.M.; Tabana, Y.M.; Ahmed, M.A.; El Ouweini, A.; Lim, V.; Keong, L.C.; Sandai, D. Pleurotus sajor-caju can be used to synthesize silver nanoparticles with antifungal activity against Candida albicans. J. Sci. Food Agric. 2018, 98, 1197–1207. [Google Scholar] [CrossRef] [PubMed]
  58. Devi, M.R.; Krishnakumari, S. Qualitative screening of phytoconstituents of Pleurotus sajor caju (Fries sing) and comparison between hot and cold-aqueous and silver nanoparticles extracts. J. Med. Plants Stud. 2015, 3, 172–176. [Google Scholar]
  59. Vigneshwaran, N.; Kathe, A.A.; Varadarajan, P.V.; Nachane, R.P.; Balasubramanya, R.H. Silver-protein (core-shell) nanoparticle production using spent mushroom substrate. Langmuir 2007, 23, 7113–7117. [Google Scholar] [CrossRef]
  60. Chaturvedi, V.K.; Yadav, N.; Rai, N.K.; Abd Ellah, N.H.; Bohara, R.A.; Rehan, I.F.; Marraiki, N.; Batiha, G.E.S.; Hetta, H.F.; Singh, M.P. Pleurotus sajor-caju-Mediated Synthesis of Silver and Gold Nanoparticles Active against Colon Cancer Cell Lines: A New Era of Herbonanoceutics. Molecules 2020, 25, 3091. [Google Scholar] [CrossRef]
  61. Dandapat, S.; Kumar, M.; Sinha, M.P. Synthesis of White Nanopartials mediate by Pleurotus tuber-regium (Rumph. ex Fr.) Extract and Silver Nitrate. In Proceedings of the Exploring Basic and Applied Sciences (EBAS-2014), Jalandhar, India, 14–15 November 2014; Elsevier: Amsterdam, The Netherlands, 2014; pp. 98–101. [Google Scholar]
  62. Chan, S.; Mashitah, M. Instantaneous biosynthesis of silver nanoparticles by selected macro fungi. Aust. J. Basic Appl. Sci. 2012, 6, 86–88. [Google Scholar]
  63. Vala, A.K.; Shah, S.; Patel, R. Biogenesis of silver nanoparticles by marine-derived fungus Aspergillus flavus from Bhavnagar Coast, Gulf of Khambhat, India. J. Mar. Biol. Ocean. 2014, 3, 1–3. [Google Scholar]
  64. Narayanan, K.B.; Sakthivel, N. Biological synthesis of metal nanoparticles by microbes. Adv. Colloid Interface Sci. 2010, 156, 1–13. [Google Scholar] [CrossRef]
  65. Thakkar, K.N.; Mhatre, S.S.; Parikh, R.Y. Biological synthesis of metallic nanoparticles. Nanomed. Nanotechnol. Biol. Med. 2010, 6, 257–262. [Google Scholar] [CrossRef] [PubMed]
  66. Dhillon, G.S.; Brar, S.K.; Kaur, S.; Verma, M. Green approach for nanoparticle biosynthesis by fungi: Current trends and applications. Crit. Rev. Biotechnol. 2012, 32, 49–73. [Google Scholar] [CrossRef] [PubMed]
  67. Gade, A.K.; Gadge Baba, S.; Ingle, A.; Marcato, P.D.; Duran, N.; Gade, A.K.; Bonde, P.; Ingle, A.P.; Marcato, P.D.; Durán, N.; et al. Exploitation of Aspergillus niger for fabrication of silver nanoparticles. J. Biobased Mater. Bioenergy 2008, 2, 1–5. [Google Scholar] [CrossRef]
  68. Zhang, X.; Yan, S.; Tyagi, R.D.; Surampalli, R.Y. Synthesis of nanoparticles by microorganisms and their application in enhancing microbiological reaction rates. Chemosphere 2011, 82, 489–494. [Google Scholar] [CrossRef] [PubMed]
  69. Khandel, P.; Shahi, S.K. Mycogenic nanoparticles and their bio-prospective applications: Current status and future challenges. J. Nanostruct. Chem. 2018, 8, 369–391. [Google Scholar] [CrossRef] [Green Version]
  70. Durán, N.; Marcato, P.D.; De Souz, G.I.; Alves, O.L.; Esposito, E. Antibacterial effect of silver nanoparticles produced by fungal process on textile fabrics and their effluent treatment. J. Biomed. Nanotech. 2007, 3, 203–208. [Google Scholar] [CrossRef] [Green Version]
  71. Mukherjee, P.; Roy, M.; Mandal, B.P.; Dey, G.K.; Mukherjee, P.K.; Ghatak, J.; Tyagi, A.K.; Kale, S.P. Green synthesis of highly stabilized nanocrystalline silver particles by a non-pathogenic and agriculturally important fungus T. asperellum. Nanotechnology 2008, 19, 075103. [Google Scholar] [CrossRef] [PubMed]
  72. Nanda, A.; Majeed, S. Enhanced antibacterial efficacy of biosynthesized AgNPs from Penicillium glabrum (MTCC1985) pooled with different drugs. Int. J. PharmTech Res. 2014, 6, 217–223. [Google Scholar]
  73. Guilger-Casagrande, M.; de Lima, R. Synthesis of Silver Nanoparticles Mediated by Fungi: A Review. Front. Bioeng. Biotechnol. 2019, 7, 287. [Google Scholar] [CrossRef] [Green Version]
  74. Devi, L.; Joshi, S. Ultrastructures of silver nanoparticles biosynthesized using endophytic fungi. J. Microsc. Ultrastruct. 2015, 3, 29. [Google Scholar] [CrossRef] [Green Version]
  75. Mehra, R.K.; Winge, D.R. Metal ion resistance in fungi: Molecular mechanisms and their regulated expression. J. Cell. Biochem. 1991, 45, 30–40. [Google Scholar] [CrossRef] [PubMed]
  76. Singh, J.; Dutta, T.; Kim, K.H.; Rawat, M.; Samddar, P.; Kumar, P. “Green” synthesis of metals and their oxide nanoparticles: Applications for environmental remediation. J. Nanobiotechnol. 2018, 16, 84. [Google Scholar] [CrossRef] [PubMed]
  77. Kumar, H.; Bhardwaj, K.; Sharma, R.; Nepovimova, E.; Kuča, K.; Dhanjal, D.S.; Verma, R.; Bhardwaj, P.; Sharma, S.; Kumar, D. Fruit and Vegetable Peels: Utilization of High Value Horticultural Waste in Novel Industrial Applications. Molecules 2020, 25, 2812. [Google Scholar] [CrossRef]
  78. Kumar, H.; Bhardwaj, K.; Nepovimova, E.; Kuča, K.; Singh Dhanjal, D.; Bhardwaj, S.; Bhatia, S.K.; Verma, R.; Kumar, D. Antioxidant Functionalized Nanoparticles: A Combat against Oxidative Stress. Nanomaterials 2020, 10, 1334. [Google Scholar] [CrossRef]
  79. Kumar, H.; Bhardwaj, K.; Dhanjal, D.S.; Nepovimova, E.; Șen, F.; Regassa, H.; Singh, R.; Verma, R.; Kumar, V.; Kumar, D.; et al. Fruit Extract Mediated Green Synthesis of Metallic Nanoparticles: A New Avenue in Pomology Applications. Int. J. Mol. Sci. 2020, 21, 8458. [Google Scholar] [CrossRef]
  80. Latha, S. Extracellular Biosynthesis, Characterization and In Vitro Antimicrobial Potential of Silver Nanoparticles Using Myconanofactoris. Ph.D. Thesis, Bharathidasan University, Tiruchirappalli, India, 2010. [Google Scholar]
  81. Borovaya, M.; Pirko, Y.; Krupodorova, T.; Naumenko, A.; Blume, Y.; Yemets, A. Biosynthesis of cadmium sulphide quantum dots by using Pleurotus ostreatus (Jacq.) P. Kumm. Biotechnol. Biotechnol. Equip. 2015, 29, 1156–1163. [Google Scholar] [CrossRef]
  82. Borovaya, M.N.; Naumenko, A.P.; Matvieieva, N.A.; Blume, Y.B.; Yemets, A.I. Biosynthesis of luminescent CdS quantum dots using plant hairy root culture. Nanoscale Res. Lett. 2014, 9, 686. [Google Scholar] [CrossRef] [Green Version]
  83. Mazumdar, H.; Haloi, N. A study on Biosynthesis of Iron nanoparticles by Pleurotus sp. J. Microbiol. Biotechnol. Res. Sch. Res. Libr. J. Microbiol. Biotech. Res. 2011, 1, 39–49. [Google Scholar]
  84. Wong, K. Preparation of highly stable selenium nanoparticles with strong anti-tumor activity using tiger milk mushroom. Apoptosis 2012, 134, 253–261. [Google Scholar]
  85. Wu, H.; Li, X.; Liu, W.; Chen, T.; Li, Y.; Zheng, W.; Man, C.W.Y.; Wong, M.K.; Wong, K.H. Surface decoration of selenium nanoparticles by mushroom polysaccharides-protein complexes to achieve enhanced cellular uptake and antiproliferative activity. J. Mater. Chem. 2012, 22, 9602–9610. [Google Scholar] [CrossRef]
  86. Kaur, G.; Kalia, A.; Kapoor, S.; Sodhi, H.S.; Khanna, P.K. Scanning electron microscopy of Pleurotus ostreatus in response to inorganic selenium. In Proceedings of the Indian Mushroom Conference, Ludhiana, India, 16–17 April 2013; pp. 17–18. [Google Scholar]
  87. El-Batal, A.I.; Al-Hazmi, N.E.; Mosallam, F.M.; El-Sayyad, G.S. Biogenic synthesis of copper nanoparticles by natural polysaccharides and Pleurotus ostreatus fermented fenugreek using gamma rays with antioxidant and antimicrobial potential towards some wound pathogens. Microb. Pathog. 2018, 118, 159–169. [Google Scholar] [CrossRef] [PubMed]
  88. Vittal, R.R.; Aswathanarayan, J.B. Nanoparticles and their potential application as antimicrobials. In Science against Microbial Pathogens: Communicating Current Research and Technological Advances; Méndez-Vilas, A., Ed.; Formatex: Badajoz, Spain, 2011; pp. 197–209. [Google Scholar]
  89. Warnes, S.L.; Caves, V.; Keevil, C.W. Mechanism of copper surface toxicity in Escherichia coli O157:H7 and Salmonella involves immediate membrane depolarization followed by slower rate of DNA destruction which differs from that observed for Gram-positive bacteria. Environ. Microbiol. 2012, 14, 1730–1743. [Google Scholar] [CrossRef] [PubMed]
  90. Beyth, N.; Houri-Haddad, Y.; Domb, A.; Khan, W.; Hazan, R. Alternative antimicrobial approach: Nano-antimicrobial materials. Evidence-Based Complement. Altern. Med. 2015, 2015, 246012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Chiriac, V.; Stratulat, D.N.; Calin, G.; Nichitus, S.; Burlui, V.; Stadoleanu, C.; Popa, M.; Popa, I.M. Antimicrobial property of zinc based nanoparticles. In IOP Conference Series: Materials Science and Engineering; IOP Publishing: Bristol, UK, 2016; Volume 133, p. 012055. [Google Scholar]
  92. Wang, L.; Hu, C.; Shao, L. The antimicrobial activity of nanoparticles: Present situation and prospects for the future. Int. J. Nanomed. 2017, 12, 1227–1249. [Google Scholar] [CrossRef] [Green Version]
  93. Mehta, M.; Sharma, P.; Kaur, S.; Dhanjal, D.S.; Singh, B.; Vyas, M.; Gupta, G.; Chellappan, D.K.; Nammi, S.; Singh, T.G.; et al. Plant-based drug delivery systems in respiratory diseases. In Targeting Chronic Inflammatory Lung Diseases Using Advanced Drug Delivery Systems; Elsevier: Amsterdam, The Netherlands, 2020; pp. 517–539. [Google Scholar]
  94. Addae, E.; Dong, X.; McCoy, E.; Yang, C.; Chen, W.; Yang, L. Investigation of antimicrobial activity of photothermal therapeutic gold/copper sulfide core/shell nanoparticles to bacterial spores and cells. J. Biol. Eng. 2014, 8, 11. [Google Scholar] [CrossRef] [Green Version]
  95. Dhanjal, D.S.; Bhardwaj, S.; Sharma, R.; Bhardwaj, K.; Kumar, D.; Chopra, C.; Nepovimova, E.; Singh, R.; Kuca, K. Plant Fortification of the Diet for Anti-Ageing Effects: A Review. Nutrients 2020, 12, 3008. [Google Scholar] [CrossRef]
  96. Acay, H.; Baran, M. Determination of Antioxidant and cytotoxic activities of king oyster mushroom mediated AgNPs synthesized with environmentally friendly methods. Medicine 2020, 9, 760–765. [Google Scholar] [CrossRef]
  97. Sharma, P.; Mehta, M.; Dhanjal, D.S.; Kaur, S.; Gupta, G.; Singh, H.; Thangavelu, L.; Rajeshkumar, S.; Tambuwala, M.; Bakshi, H.A.; et al. Emerging trends in the novel drug delivery approaches for the treatment of lung cancer. Chem. Biol. Interact. 2019, 309, 108720. [Google Scholar] [CrossRef]
  98. Mehta, M.; Dhanjal, D.S.; Paudel, K.R.; Singh, B.; Gupta, G.; Rajeshkumar, S.; Thangavelu, L.; Tambuwala, M.M.; Bakshi, H.A.; Chellappan, D.K.; et al. Cellular signalling pathways mediating the pathogenesis of chronic inflammatory respiratory diseases: An update. Inflammopharmacology 2020, 28, 795–817. [Google Scholar] [CrossRef]
  99. Mehta, M.; Dhanjal, D.S.; Satija, S.; Wadhwa, R.; Paudel, K.R.; Chellappan, D.K.; Mohammad, S.; Haghi, M.; Hansbro, P.M.; Dua, K. Advancing of Cellular Signaling Pathways in Respiratory Diseases Using Nanocarrier Based Drug Delivery Systems. Curr. Pharm. Des. 2020, 26. [Google Scholar] [CrossRef]
  100. Sankar, R.; Karthik, A.; Prabu, A.; Karthik, S.; Shivashangari, K.S.; Ravikumar, V. Origanum vulgare mediated biosynthesis of silver nanoparticles for its antibacterial and anticancer activity. Colloids Surfaces B Biointerfaces 2013, 108, 80–84. [Google Scholar] [CrossRef] [PubMed]
  101. Gliga, A.R.; Skoglund, S.; Odnevall Wallinder, I.; Fadeel, B.; Karlsson, H.L. Size-dependent cytotoxicity of silver nanoparticles in human lung cells: The role of cellular uptake, agglomeration and Ag release. Part. Fibre Toxicol. 2014, 11, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Ismail, A.F.M.; Ahmed, M.M.; Salem, A.A.M. Biosynthesis of silver nanoparticles using mushroom extracts: Induction of apoptosis in HepG2 and MCF-7 Cells via caspases stimulation and regulation of BAX and Bcl-2 gene expressions. J. Pharm. Biomed. Sci. 2015, 5, 1–9. [Google Scholar]
  103. Singh, P.; Sathishkumar, S.R.; Bhaskararao, K.V. Biosynthesis of Silver Nanoparticles using Actinobacteria and Evaluating Its Antimicrobial and Cytotoxicity Activity. Int. J. Pharm. Pharm. Sci. 2013, 5, 709–712. [Google Scholar]
  104. Kim, J.H.; Lee, Y.; Kim, E.J.; Gu, S.; Sohn, E.J.; Seo, Y.S.; An, H.J.; Chang, Y.S. Exposure of iron nanoparticles to Arabidopsis thaliana enhances root elongation by triggering cell wall loosening. Environ. Sci. Technol. 2014, 48, 3477–3485. [Google Scholar] [CrossRef] [PubMed]
  105. El-Batal, A.I.; Elkenawy, N.M.; Yassin, A.S.; Amin, M.A. Laccase production by Pleurotus ostreatus and its application in synthesis of gold nanoparticles. Biotechnol. Rep. 2015, 5, 31–39. [Google Scholar] [CrossRef] [Green Version]
  106. Bagewadi, Z.K.; Mulla, S.I.; Ninnekar, H.Z. Optimization of laccase production and its application in delignification of biomass. Int. J. Recycl. Org. Waste Agric. 2017, 6, 351–365. [Google Scholar] [CrossRef] [Green Version]
  107. Panigrahi, S.; Basu, S.; Praharaj, S.; Pande, S.; Jana, S.; Pal, A.; Ghosh, S.K.; Pal, T. Synthesis and size-selective catalysis by supported gold nanoparticles: Study on heterogeneous and homogeneous catalytic process. J. Phys. Chem. C 2007, 111, 4596–4605. [Google Scholar] [CrossRef]
  108. Lim, S.H.; Ahn, E.Y.; Park, Y. Green Synthesis and Catalytic Activity of Gold Nanoparticles Synthesized by Artemisia capillaris Water Extract. Nanoscale Res. Lett. 2016, 11, 474. [Google Scholar] [CrossRef] [Green Version]
  109. Rostami-Vartooni, A.; Nasrollahzadeh, M.; Alizadeh, M. Green synthesis of perlite supported silver nanoparticles using Hamamelis virginiana leaf extract and investigation of its catalytic activity for the reduction of 4-nitrophenol and Congo red. J. Alloy. Compd. 2016, 680, 309–314. [Google Scholar] [CrossRef]
  110. Gopalakrishnan, R.; Loganathan, B.; Dinesh, S.; Raghu, K. Strategic Green Synthesis, Characterization and Catalytic Application to 4-Nitrophenol Reduction of Palladium Nanoparticles. J. Clust. Sci. 2017, 28, 2123–2131. [Google Scholar] [CrossRef]
  111. Sharma, D.; Dhanjal, D.S. Bio-Nanotechnology for Active Food Packaging. J. Appl. Pharm. Sci. 2016, 6, 220–226. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Graphical representation of green-synthesis of nanoparticles from Pleurotus (Oyster) mushroom.
Figure 1. Graphical representation of green-synthesis of nanoparticles from Pleurotus (Oyster) mushroom.
Jof 06 00351 g001
Figure 2. Schematic diagram showing the applications of oyster mushroom derived nanoparticles.
Figure 2. Schematic diagram showing the applications of oyster mushroom derived nanoparticles.
Jof 06 00351 g002
Figure 3. Graphical representation of mechanism showing anticancer and antimicrobial activity.
Figure 3. Graphical representation of mechanism showing anticancer and antimicrobial activity.
Jof 06 00351 g003
Figure 4. Schematic diagram of the antioxidant mechanism.
Figure 4. Schematic diagram of the antioxidant mechanism.
Jof 06 00351 g004
Table 1. Geographical Distribution of Pleurotus spp.
Table 1. Geographical Distribution of Pleurotus spp.
SpeciesGeographical DistributionOptimum Growth Temperature °CReference
Pleurotus citrinopileatusRussia, China, Japan21–29[8,9]
P. cornucopiaeEurope, USA, and Mexico>25[8,9]
P. djamorTropical region, Indonesia, Malaysia, Japan, Mexico21–35[8,9]
P. eousSub-tropical part of the world23–28[8,9]
P. eryngiiEurope, Asia, Africa20–25[8,9]
P. flabellatusIndia, Mauritius25–28[8,9]
P. floridaHungary, Kenya20–28[8,9]
P. giganteusThailand, Sri Lanka15–35[8,10]
P. ostreatusWidespread around the world18–22[8,9]
P. sajor-cajuKenya, India, Philippines, Australia, Mauritius20–28[8,9]
P. tuber-regiumAfrica, Australia, Asia25–30[8,9]
P. platypusIndia15–25[8,9]
P. pulmonariusWarm tropical area20–28[8,9]
Table 2. Varieties of Pleurotus spp. and their nanoparticles.
Table 2. Varieties of Pleurotus spp. and their nanoparticles.
SpeciesTypes of Nanoparticles Synthesize and Their Size (nm)Chemical UsedReaction Time (hour)Reducing AgentStabilizing AgentSpecific Temperature
(°C)
MorphologyReferences
Pleurotus citrinopileatusAg, 6–10AgNO324mushroom extract, Nitratemushroom extract60spherical[38]
P. cornucopiae (citrinopileatus)Ag, 20–30AgNO324aqueous extractaqueous extract25spherical[37]
P. cystidiosusAg, 2–100AgNO324aqueous extractaqueous extract25ND[39]
P. cystidiosusAu, NDHAuCl424aqueous extractaqueous extract29ND[39]
P. djamorAg, 5–50AgNO348, 24aqueous extractaqueous extractRTspherical[40,41,42]
P. djamorZnO, 70–80Zn(NO3)2.5H2O24NDNDRTspherical[43]
P. djamorTiO2TiCl420 minaqueous extractaqueous extractRTspherical[44]
P. eryngiiAg, 18.45AgNO35 daysaqueous extractaqueous extractRTspherical[45]
P. flabellatusAg, 2–100AgNO324aqueous extractaqueous extract25ND[39,40,41]
P. flabellatusAu, NDHAuCl424aqueous extractaqueous extract29ND[39]
P. floridaAg, 20AgNO3Overnight; 72aqueous extractaqueous extractRTspherical[33,34,40,41,46,47,48]
P. floridaAu, 2–14HAuCl41.5aqueous extract, glucanglucan70spherical[49]
P. floridaAu, 20HAuCl424aqueous extractaqueous s extractRTspherical[50]
P. giganteusAg, 5–25AgNO33 daysaqueous extractaqueous extract37spherical[51]
P. ostreatusAg, 4,28,50AgNO324; 72; 1aqueous extract; mushroom brothaqueous extract28; 75spherical;[36,39,52,53]
P. ostreatusAu, 22.9HAuCl424 haqueous extractaqueous extract29spherical[39,54]
P. ostreatusZnS, 2–5ZnCl2Over nightmushroommushroom extract70spherical with crystalline[55]
P. ostreatusZn, 15ZnS-N31aqueous extractaqueous extract, sodium azide4uniform[56]
P. platypusAg, 0.56µmAgNO372aqueous extractaqueous extract37spherical[34]
P. pulmonariusAg, 2–100AgNO324aqueous extractaqueous extract25ND[40,41]
P. pulmonariusAu, NDHAuCl424aqueous extractaqueous extract29ND[39]
P. sajor-cajuAg, 5–50AgNO348aqueous extractaqueous extract25spherical[32,57,58,59,60]
P. sajor-cajuAu, 16–18HAuCl4·3H2OOver nightaqueous extractaqueous extractRTspherical[60]
P. tuber-regiumAg, 50AgNO32aqueous extractaqueous extract80spherical and cubical[61]
Note: RT—room temperature; ND—not detected.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bhardwaj, K.; Sharma, A.; Tejwan, N.; Bhardwaj, S.; Bhardwaj, P.; Nepovimova, E.; Shami, A.; Kalia, A.; Kumar, A.; Abd-Elsalam, K.A.; et al. Pleurotus Macrofungi-Assisted Nanoparticle Synthesis and Its Potential Applications: A Review. J. Fungi 2020, 6, 351. https://0-doi-org.brum.beds.ac.uk/10.3390/jof6040351

AMA Style

Bhardwaj K, Sharma A, Tejwan N, Bhardwaj S, Bhardwaj P, Nepovimova E, Shami A, Kalia A, Kumar A, Abd-Elsalam KA, et al. Pleurotus Macrofungi-Assisted Nanoparticle Synthesis and Its Potential Applications: A Review. Journal of Fungi. 2020; 6(4):351. https://0-doi-org.brum.beds.ac.uk/10.3390/jof6040351

Chicago/Turabian Style

Bhardwaj, Kanchan, Anirudh Sharma, Neeraj Tejwan, Sonali Bhardwaj, Prerna Bhardwaj, Eugenie Nepovimova, Ashwag Shami, Anu Kalia, Anil Kumar, Kamel A. Abd-Elsalam, and et al. 2020. "Pleurotus Macrofungi-Assisted Nanoparticle Synthesis and Its Potential Applications: A Review" Journal of Fungi 6, no. 4: 351. https://0-doi-org.brum.beds.ac.uk/10.3390/jof6040351

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop