Next Article in Journal
Pathological Process of Prompt Connection between Host and Donor Tissue Vasculature Causing Rapid Perfusion of the Engineered Donor Tissue after Transplantation
Next Article in Special Issue
RNA Sequencing Reveals Differential Gene Expression of Cerrena Unicolor in Response to Variable Lighting Conditions
Previous Article in Journal
TCDD Toxicity Mediated by Epigenetic Mechanisms
Previous Article in Special Issue
Uridine Diphosphate-Dependent Glycosyltransferases from Bacillus subtilis ATCC 6633 Catalyze the 15-O-Glycosylation of Ganoderic Acid A
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biosynthesis of Metal Nanoparticles via Microbial Enzymes: A Mechanistic Approach

1
CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
2
University of Chinese Academy of Sciences, Beijing 100049, China
3
Department of Eastern Medicine and Surgery, Qarshi University, Lahore 54000, Pakistan
4
Department of Pharmacy, University of Malakand, Khyber Pakhtunkhwa (KPK), Chakdara 18000, Pakistan
5
Department of Life sciences, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia
6
Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
7
Department of Bioengineering, Rice University, Houston, TX 77030, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2018, 19(12), 4100; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19124100
Submission received: 30 November 2018 / Revised: 8 December 2018 / Accepted: 17 December 2018 / Published: 18 December 2018
(This article belongs to the Special Issue Microbial Enzymes)

Abstract

:
During the last decade, metal nanoparticles (MtNPs) have gained immense popularity due to their characteristic physicochemical properties, as well as containing antimicrobial, anti-cancer, catalyzing, optical, electronic and magnetic properties. Primarily, these MtNPs have been synthesized through different physical and chemical methods. However, these conventional methods have various drawbacks, such as high energy consumption, high cost and the involvement of toxic chemical substances. Microbial flora has provided an alternative platform for the biological synthesis of MtNPs in an eco-friendly and cost effective way. In this article we have focused on various microorganisms used for the synthesis of different MtNPs. We also have elaborated on the intracellular and extracellular mechanisms of MtNP synthesis in microorganisms, and have highlighted their advantages along with their challenges. Moreover, due to several advantages over chemically synthesized nanoparticles, the microbial MtNPs, with their exclusive and dynamic characteristics, can be used in different sectors like the agriculture, medicine, cosmetics and biotechnology industries in the near future.

Graphical Abstract

1. Background and Role of Microbial Enzymes in Metal Nanoparticle (MtNP) Biosynthesis

Nanotechnology has accomplished enormous development during the last decades due to the synthesis and varied applications of metal nanoparticles (MtNPs) in different areas, such as biology, food, agriculture, engineering, electronics, cosmetics, medicine, and in food and biomedical devices. MtNPs have reached a momentous position due to their specific physicochemical characteristics and significant biotechnological applications [1,2]. According to a market report, the worldwide production of MtNPs is currently valued at 13.7 billon US dollars, which is expected to reach 50 billon US dollars by 2026. The wide-ranging use of MtNPs has significantly contributed to robust development in the macroeconomic industry and the demand for MtNPs will continue to be high in three regions of the world: North America, Western Europe and the Asia-Pacific region [3].
During the last decade, many researchers have synthesized MtNPs by orthodox physical and chemical methods. The disadvantages of the physical method include expensive synthesis and little yield. Similarly, the chemical methods are unsafe due to the involvement of hazardous chemical substances that are attached to the surface of MtNPs, which possess detrimental side effects in biomedical applications. Keeping in view the aforementioned concerns, research has shifted towards the synthesis of MtNPs using biological constituents which are economical, biocompatible, non-toxic and eco-friendly [4,5,6,7]. The biological synthesis of MtNPs is mostly carried out by utilizing different types of plants and microorganisms. The phytogenic process of MtNP synthesis is economical and relatively simple. However, this process generates polydispersed nanoparticles, due to its diverse photochemistry [8,9]. In contrast, the microorganisms are considered as prospective bio-factories for the green synthesis of MtNPs, something that has gained vast consideration in recent times, as they are indispensable and of technological importance. This is due to the broad range of microorganisms that react in a different way with the metal ions for MtNP synthesis. Microbial MtNPs of different sizes and shapes have been reported in various species of bacteria, fungi and yeasts due to their enhanced growth rate, easy cultivation and their capability to grow under established conditions of temperature, pH and pressure [10,11]. The biosynthesis of MtNPs and their alloys (gold, silver, gold-silver alloy, selenium, tellurium, platinum, palladium, silica, titania, zirconia, quantum dots, magnetite and uraninite) has been reported in different microorganisms, including bacteria, actinomycetes, fungi, yeasts and viruses.
Different microorganisms have the ability to synthesize inorganic materials via both intracellular and extracellular methods. In the intracellular approach, the microbial cell is comprised of an amazing ion transport system. Due to electrostatic interaction, the bacterial cell wall which is negatively charged attracts the positively charged metal ions. Additionally, the bacterial cell wall contains enzymes that reduce the metal ions to their respective nanoparticles. Whereas in extracellular method, the microbial cell secretes reductases used in the bioreduction of metal ions into the corresponding MtNPs [12]. The following sections of this article will provide an overview on the green synthesis of MtNPs by using potential microbial flora, including bacteria, cyanobacteria, microalgae, actinomycetes, yeasts, fungi, viruses and diatoms. Furthermore, the intracellular and extracellular mechanisms that are adopted by the microorganism and the microbial enzymes for MtNP synthesis will be elaborated upon, along with the contemporary challenges and future prospects. Table 1 highlights the recent literature about the microbe assisted synthesis of MtNPs and their applications.

2. Biosynthesis of MtNPs by Microorganisms

Synthesizing uniform, ultrafine, well-dispersed functional nanoparticles under normal conditions through a controlled manner remains a great challenge [13,14]. This has caused a major surge in looking for alternative means of synthesizing nanoparticles that are devoid of such disadvantages. In recent years, biological resources have been frequently explored for the biosynthesis of metal or metal-based nanoparticles. These biological resources usually provide a versatile, economical and eco-friendly method to fabricate metal nanoparticles [14,15] that exhibit interesting physical, chemical and biological properties. Furthermore, other advantages such as the ease of production and scaling, well defined morphologies and enhanced biocompatibility (relative to the physiochemical based nanoparticles) have attracted many scientists to use such resources as nanofactories [16,17]. The bio-based methods for the synthesis of metal nanoparticles are based on the systematic use of plant extracts and microorganisms, like bacteria, yeasts, and fungi [18,19,20,21,22]. In biological synthesis, no capping or stabilizing agents are added as the biomolecules can perform this function themselves [23,24,25]. The properties of these nanoparticles (such as shape, size, etc.), are governed by these biomolecules [13]. These biomolecules also functionalize the nanoparticles, making it more effective relative to nanoparticles synthesized through nonbiological means [17].
The use of plants for biosynthesis has been discussed in detail previously by our own group as well as other scientists across the world [19,20,26,27]. The phytogenic and microgenic biosynthesis of nanoparticles both have their own sets of advantages and disadvantages. Phytogenic synthesis is time economical and relatively simple but usually leads to polydispersed nanoparticles, due to the involvement of diverse phytochemicals like phenols, flavonoids, terpenoids, etc. [8,9]. Moreover, seasonal variations can alter the phytochemical profile of the extracts used for biosynthesis [28]. On the contrary, microbial synthesis is devoid of such disadvantages, however, it requires the maintainance of a sterilized environment and culture conditions, making it relatively complex [29]. Herein, we have discussed the potential of using the microbial world as nanofactories for the biosynthesis of metal nanoparticles or metal based composite materials, both from functional and mechanistic perspectives. Microorganisms hold an exciting amount of potential for the biosynthesis of nanoparticles as the synthesis is eco-friendly and is devoid of the use of hazardous chemicals. Microorganisms are cost effective and do not have high energy requirements. In addition, they can accumulate and detoxify heavy metals through reductase enzymes, which reduce metal salts to their corresponding metal nanoparticle with less polydispersity and a narrow size range [16].

3. Bacterial and Cyanobacterial Biosynthesis of MtNPs

Bacterial cells have been readily employed as nanofactories for the synthesis of various metal nanoparticles. Both the extracellular and intracellular approaches have been demonstrated. Extracellular biosynthesis occurs outside the bacterial cell after applying diverse techniques, such as (a) using of the bacterial biomass, (b) using the supernatant of bacterial cultures and (c) using cell free extracts. Extracellular synthesis is preferred over intracellular synthesis because it is devoid of of complex downstream processing [28]. These nanoparticles have been used in a wide array of applications, however mostly for biomedical uses. Recently, silver nanoparticles (AgNPs) that were synthesized using Bacillus brevis showed excellent antimicrobial activities against the multidrug resistant strains of Staphylococcus aureus and Salmonella typhi [30]. Pseudomonas stutzeri is another bacterial strain which was found capable of accumulating AgNPs using an intracellular mechanism [31]. The Bacillus sp. was also found to synthesize silver nanoparticles in the intracellular periplasmic space [32]. In one study, two different isolated strains of Pseudomonas aeruginosa were used for the biosynthesis of gold nanoparticles (AuNPs), generating different sizes of AuNPs [33]. Spherical (10–50 nm) and triangular plate (50–400 nm) AuNPs were produced using Rhodopseudomonas capsulate [34]. Serratia ureilytica mediated the as-synthesized ZnO nanoflowers that were used on cotton fabrics to provide antimicrobial effects against S. aureus and E. coli [35]. Lactobacillus plantarum was also reported for the biosynthesis of ZnO nanoparticles [36]. Aeromonas hydrophila is a gram-negative bacterial strain that is used for the biosynthesis and antimicrobial applications of ZnO nanoparticles [37]. Recently, triangular CuO nanoparticles were produced using Halomonas elongate, and their antimicrobial activity was confirmed against E. coli and S. aureus [38]. In another recent report, super paramagnetic iron oxide nanoparticles (29 nm) were produced using Bacillus cereus and they were reported for their anti-cancer effects against the MCF-7 and 3T3 cell lines in a dose-dependent manner [39]. Furthermore, bimetallic Ag-Au nanostructures have also been demonstrated using bacterial strains [40]
Numerous researchers have used cyanobacteria for the biosynthesis of nanoparticles [41]. In one study, eight cyanobacterial strains were screened to investigate their potential for the biosynthesis of AgNPs. The results indicated that seven cyanobacterial strains were capable of producing AgNPs under light, however, only three were capable of producing AgNPs in the dark [42]. The ammonia-sensing potential of the cyanobacterial synthesized silver nanoparticles was also reported [43]. Highly monodispersed AgNPs (5–6.5 nm) were reported through the marine cyanobacterium Phormidium fragile [44]. The Nostoc species was also used for the biosynthesis of AgNPs, which indicated significant cytotoxic activity against MCF-7 anti-cancer cell lines, as well as good antimicrobial activities [45]. The intracellular synthesis of AuNPs was performed using Lyngbya majuscule, which was isolated from the Arabian Gulf. The authors also revealed the interesting role of the use of AuNPs in combination with Lyngbya majuscule as an anti-myocardial infraction agent [46].

4. Mycosynthesis of Nanoparticles

Myco-nanotechnological approaches have been successfully applied for the biosynthesis of different metal nanoparticles. Likewise, in bacteria/cyanobacteria, the biosynthesis can be intracellular or extracellular. In intracellular synthesis, metal salts are converted into a less toxic form in the mycelia, which can be used by the fungi [47]. Extracellular biosynthesis includes the use of fungal extracts [48]. Fungi are relatively more resourceful then bacteria in the biosynthesis of nanoparticles due to the presence of a number of bioactive metabolites, high accumulation and enhanced production [16,49,50]. Recently, different filamentous fungi were reported to be proficient in the biosynthesis of AuNPs. This study employed different methods for the biosynthesis of AuNPs. The authors proposed that the fungal compounds and fungal media components potentially played a role in stabilizing the nanoparticles [47]. In another research article, three different strains of fungi (namely Aureobasidium pullulans, Fusarium oxysporum and the Fusarium sp.) were used to biosynthesize AuNPs. The authors indicated that the biosynthesis occurred in fungi vacuoles and that reducing sugars were involved to tailor spherical AuNPs. They also established the role of specific fungal proteins in the capping of the AuNPs [51]. A recent study indicated that Rhizopus stolonifera extracts mediated the synthesis of monodispersed AgNPs (9.4 nm), while condition optimization resulted in 2.86 nm AgNPs [52]. The extracellular synthesis of AgNPs using Candida glabrata indicated good antimicrobial potential [53]. Aspergillus niger mediated ZnO nanoparticles indicated excellent antibacterial potential, while they were also able to degrade the Bismarck brown dye by up to 90% [54]. Recently, cobalt oxide nanoparticles were produced using Aspergillus nidulans [55]. The nanoparticles were characterized via x-ray diffraction analysis (XRD), transmission electron microscopy (TEM), Fourier transform infrared spectroscopy (FTIR) and energy dispersive x-ray analysis (EDX) techniques, which revealed their spherical shape and average size of 20.29 nm. Interestingly, biogenic cobalt oxide nanoparticles have the potential to be applied in energy storage, lithium-ion batteries and in gas sensing, as well as in medicine.

5. Algae as Biosynthesis Factories

The use of algae is also increasingly popular for the biosynthesis of nanoparticles. Sargassum muticum was used for the biosynthesis of ZnO nanoparticles and was reported to decrease angiogenesis along with apoptotic effects in HepG2 cells [56]. AgNPs that were biosynthesized using Gelidium amansii indicated excellent antimicrobial properties by forming a diverse biofilm to combat bacterial strains [14]. Sargassum crassifolium, a macroalgae and sea weed, has been used in the biosynthesis of AuNPs. Furthermore, the authors observed a blue shift in the UV absorption after increasing the concentration of S. crassifolium, which was attributed to a decreased size due to increased nucleation centers in the reductant [57]. Cystoseira trinodis was used for the biosynthesis of CuO nanoparticles (7 nm) and was reported to have enhanced antibacterial activities and possess significant potential as an antioxidant, degrading the methylene blue [58]. Aluminum oxide nanoparticles (~20 nm) were produced using Sargassum ilicifolium [59]. Different algae strains were reported for the biosynthesis of gold nanoparticles, namely Turbinaria conoides [60], Sargassum tenerrimum [61], Acanthophora spicifera [62], Laminaria japonica [63], etc. These have been used for the biosynthesis of AuNPs. Novel core (Au)-shell (Ag) nanoparticle synthesis has also been reported using Spirulina platensis [64].

6. Mechanisms of MtNP Synthesis by Microorganisms

The synthesis of nanosized substances using microbial cells is an emerging trend in the field of nanotechnology. Microbes including bacteria, fungi, viruses, actinomycetes and yeasts act as potential biofactories for the reduction of silver, gold, gold-silver alloy, cadmium, selenium, magnetite, silica, platinum, titania, palladium and other metals to their subsequent nanoparticles for biological applications [109]. Microbes synthesize these nanoparticles either extracellularly or intracellularly using various bio-reduction processes (Table 2).

7. Extracellular Enzymes

Extracellular microbial enzymes are known to play a significant role as reducing agents in the production of MtNPs [110] (Figure 1). Studies suggest that cofactors such as nicotinamide adenine dinucleotide (NADH) and reduced form of Nicotinamide adenine dinucleotide phosphate (NADPH) dependent enzymes both play vital roles as reducing agents via the transfer of the electron from NADH by NADH-reliant enzymes, which act electron carriers [111]. The extracellular synthesis of AuNPs by the bacterium Rhodopseudomonas capsulata is mediated via the secretion of NADH and NADH-reliant enzymes. The bioreduction of gold is initiated via electron transfer from NADH by NADH-reliant reductase enzymes present in R. capsulata. Consequently, gold ions accept electrons and get reduced (Au3+ to Au0), leading to the formation of gold nanoparticles [34]. Several other factors, including the concentration of the predecessor, the pH, the temperature and the duration of reaction are limiting factors in controlling the size of MtNPs. Beside these enzymes, several compounds, including naphthoquinones, anthraquinones and hydroquinones are involved in the production of MtNPs [19]. Microbes utilize various mechanisms for the synthesis of NPs, including changes in solubility, biosorption, metal complexation, extracellular precipitation, toxicity via oxidation-reduction, the absence of specific transporters and efflux pumps [19,112].
Several fungi produce extracellular enzymes like acetyl xylan esterase, cellobiohydrolase D, glucosidase and β-glucosidase, which are known to play a significant role in the biosynthesis of MtNPs [15]. One mechanism involved in the extracellular synthesis of AgNPs is the use of nitrate reductase which is secreted by fungi, which helps in the bioreduction and synthesis of MtNPs. Several studies reported the involvement of nitrate reductase in the extracellular synthesis of MtNPs [113,114]. Studies involving the use of commercially available nitrate reductase disks revealed that these NADH-reliant reductase enzymes were involved in the reduction of Ag+ ions to Ag(0) and the subsequent formation of silver nanoparticles [115,116]. Fusarium oxysporum was used as source of reducing agents for the synthesis of gold and silver NPs. Results showed that extracellular reductases produced by the fungi caused the reduction of Au3+ and Ag1+ to Au–Ag alloy NPs. Moreover, nitrate-reliant reductases and shuttle quinone obtained from various species of this fungi were utilized in the extracellular synthesis of NPs [117]. However, some species like Fusarium moniliforme failed to generate AgNPs, even upon the release of the reductase, indicating the Ag1+ reduction via the involvement of conjugated oxidation-reduction reactions of electron carriers involving NADP-reliant nitrate reductase [116]. Furthermore, nitrate reductase from F. oxysporum was used in an in vitro study in oxygen free conditions in the presence of a cofactor (NADPH), a stabilizer protein (phytochelatin) and an electron carrier (4-hydroxyquinoline) in order to synthesize AgNPs. This fungi exhibited good extracellular production of AgNPs and can be considered an excellent candidate for the extracellular synthesis of other MtNPs [113,118]. Yet in other studies, F. oxysporum was used for the extracellular synthesis of semiconductor CdS nanoparticles, where extremely luminescent CdSe nanoparticles were synthesized using the reductase enzyme of the fungi [114,119]. Enzymes from other fungal stains, including Fusarium semitectum and Fusarium solani, were used for the extracellular production of AgNPs. The results of the study revealed that specific proteins might be responsible for the reduction of Ag+, thus forming AgNPs [120,121]. Cladosporium cladosporioides and Coriolus versicolor were effectively used for the extracellular synthesis of AgNPs that involved fungal proteins, organic acids and polysaccharides which effect the growth and shape of the nanocrystals [122]. Subsequent to incubation of Aspergillus niger in a AgNO3 solution, the extracellular production of AgNPs was stabilized by fungal proteins [123]. Likewise, Aspergillus fumigatus extracellularly produced AgNPs in exceptionally less time (10 min), as compared to other physical and chemical techniques [124]. Thus, A. fumigatus is an ideal candidate for industrial scale production of a variety of NPs. Penicillium fellutanum was also observed to reduce Ag1+ ions in a very short amount of time (10 min). Further studies revealed that a protein of nitrate reductase was responsible for the reduction of Ag1+ ions [125]. Penicillium brevicompactum was reported to cause the reduction of Ag1+ ions via the liberation of NADH-reliant enzyme nitrate reductases [126].
Nanotechnology was also applied to a new group of the plant kingdom known as algae. Among the algae, Sargassum wightii greville was reported to rapidly reduce Au3+ ions to form AuNPs 8–12 nm in size [127]. Another filamentous algae, Chlorella vulgaris, was used in the biosynthesis of gold nanoparticles, resulting in the formation of Au and Au+1S nanoparticles [128].

8. Intracellular Enzymes

In the intracellular mechanism of metal bioreduction, bacterial and fungal cells along with sugars molecules play a significant role. Mainly, the interactions of intracellular enzymes and positively charged groups are utilized in the gripping of metallic ions from the medium and the subsequent reduction inside the cell [150,151]. When observed microscopically, MtNPs accumulated in the periplasmic space, the cytoplasmic membrane and the cell wall. This was due to the diffusion of metal ions across the membranes and enzymatic reduction resulting in MtNPs.
Among the actinomycetes, alkalo-tolerant (Rhodococcus sp.) and alkalo-thermophilic (Thermomonospora sp.) actinomycetes were used for the intracellular synthesis of AuNPs [148,152]. The intracellular production of AuNPs with uniform dimensions was carried out by reacting the Rhodococcus species with an aqueous solution of AuCl4− ions. Reductions of Au3+ were effectively mediated by enzymes at the surface of the mycelia and the cytoplasmic membrane. The exposure of a Verticillium biomass to a Ag+ ionic solution resulted in intracellular reduction and the subsequent formation of AgNPs. Visualization via electron microscopy revealed that the AgNPs were formed under the surface of cell wall as a result of enzymatic bioreduction, which is non-toxic to the fungi [153]. The same procedure was adopted for the synthesis of AuNPs using the fungus Verticillium as the source of reducing enzymes. AuNPs were entrapped in the cytoplasmic membrane and the cell wall of the fungi, indicating that Au3+ was bio-reduced by reductase enzymes that were present there [154]. Southam and Beveridge reported that AuNPs were formed and precipitated inside bacterial cells after the incubation of the bacterial cells in a Au3+ ionic solution [155]. Pseudomonas stutzeri (AG259) when exposed to concentrated AgNO3 solution has reduced Ag1+ ions, with the subsequent formation of AgNPs in the bacterial periplasmic space [31]. A filamentous cyanobacterium (Plectonema boryanum) treated with AuCl4− and Au(S2O3)23- solutions resulted in the formation of AuNPs at the membrane level and gold sulfide residing intracellularly [128]. In another study, Phanerochaete chrysosporium incubation in an ionic Au3+ solution resulted in the formation of AuNPs 10–100 nm in particle size. The Laccase enzyme was used as an extracellular reducing agent, whereas ligninase was found to be responsible for the intracellular reduction of Au3+ ions [156]. Other factors, including the incubation age of the fungi, the concentration of the AuCl4− solution and the incubation temperature demonstrated significant effects on the shape of AuNPs. The mesophilic bacterium Shewanella algae proved to be an efficient bioreducer of AuCl4− ions to elemental gold. Au nanoparticles were found in the bacterial periplasmic space, mediated via intracellular enzymes [157]. Brevibacterium casei treated with aqueous solutions of Au3+ and Ag+ ions were reduced to intracellular enzymes, mediated by spherically shaped AuNPs and AgNPs respectively [147].

9. Challenges and Limitation of MtNP Synthesis by Microorganisms: A Possible Solution

Nanotechnology is an interdisciplinary field which involves close coordination between engineers, chemists and biologists to design and develop potential candidates for therapeutic applications. Research on nanoparticle biosynthesis using microbes has gained significant interest, owing to its simplicity, low cost of production and eco-friendliness. MtNPs have emerged as excellent agents for biomedical applications due to their unique physicochemical, optical, electronic and biological properties [158]. Expanding interest on the application of MtNPs in biology and healthcare has raised a need for the development of cheap, easy and eco-friendly methods for MtNP preparation, such as the bioinspired approaches [159,160]. The biosynthesis of MtNPs using microbes is well investigated by researchers globally, where the reduction of metal ions to MtNPs occurs as part of the heavy metal-resistance mechanism exhibited by microorganisms [161]. The primary challenge involved in microbial inspired synthesis is the selection of the best candidates, based on the intrinsic properties of microbes, such as growth rate, replication, biochemical activity etc. Controlling the shape, size and monodispersity of the biosynthesized MtNPs is another important criterion for achieving the desired therapeutic effects, which might be modulated by varying the concentration of biomolecules, the reduction time, the temperature and other factors. Another important challenge involved in biosynthesis is the identification of the key components among the large pool of biomolecules obtained from microbial resources responsible for the reduction and stabilization of MtNPs from precursor salts. Maintaining the optimal conditions needed for the microbial growth and enzymatic activity is critical as these act as biocatalysts, playing a major role in the preparation of MtNPs. Optimizing the conditions, such as providing essential nutrients for growth, the size of inoculum, the temperature, the pH and the amount of light greatly enhance the efficiency of MtNP biosynthesis using microbes [162]. Yield of the final product and rate of biosynthesis of MtNPs using microorganisms are another set of important parameters to be considered to translate the measure of production to a large scale industrial level. In general, the large scale production of biosynthesized nanoparticles using microbes has several challenges in scaling up, due to medium to low yield compared to chemically synthesized nanoparticles. In general, the yield of the biosynthesis methods are 1/3rd of the yield obtained from the chemical synthesis methods. Also, we have to keep in mind that the laboratory scale bottom-up or top-down approaches for the synthesis of MtNPs varies significantly from industrial manufacturing. However, due to large availability, easy access and the high growth rate of microbes, the overall cost of the synthesis will be reduced at a large extent. Moreover, the cost of the biosynthesis gets further reduced as there is no requirement of organic solvents, chemical stabilizing agents, thermal heat or any fancy technique during these synthesis methods. Hence, it can be safely mentioned that, if the right techniques are properly utilized, the overall production cost of the NPs can be reduced by almost 1/10th compared to the chemical synthesis methods in the long run. Nevertheless, we strongly feel that during the initial stages of the development of NPs at the lab scale, researchers must consider and deploy appropriate methods useful for large-scale industrial manufacturing [163]. An advantage of this method is that the MtNPs produced using the biosynthesis route are more stable without any agglomeration, even at room temperatures for long periods of time when compared with MtNPs produced using chemical synthetic methods [164,165]. This stability might be due to the in situ capping of the microbial proteins or biomolecules over the MtNP surface during the biosynthesis process. Understanding the source and mechanism of biosynthesis is essential, as sometimes the utility of toxic microorganisms as a bioresource might impart pathogenicity to the prepared MtNPs. Above all, the protocols designed at the laboratory scale might yield well-characterized highly stable MtNPs. Scaling up this process to the industrial level is mandatory to fulfill the unmet medical challenges, which mainly depend upon optimizing the growth rate of the microbes, inoculation of the seed into the biomass, harvesting cells and effective MtNP preparation through reduction, as well as recovery of the final product with good yield [12,166,167]. Taken together, there are several important challenges and technical aspects that must be addressed to successfully translate this bioinspired method of MtNP synthesis for large scale industrial production.

10. Conclusions and Future Prospects

For the time being, it can be safely concluded that there is a bright future for the microbial synthesis of metal nanoparticles and their wide variety of applications in biomedical nanotechnology when compared with chemically assisted NPs, due to their low cost and eco-friendliness. The huge pool of bioresources (microbes and microbial enzymes), if appropriately exploited, could help biosynthesized NPs to turn out to be a potential game changer in the imminent future. However, several challenges must be dealt with before industrial scale production and wide use, which will take a little more time (5–10 years). Nevertheless, there is immense potential for microbial assisted metal nanoparticles as they are low in toxicity and cost and high in degradability and are useful in numerous therapeutic applications. It is also encouraging that several groups are concentrating on decoding the detailed mechanistic aspects of microbial biosynthesis, which will ultimately lead to better understanding and wiser applicability. Even though the mechanisms behind these nanoparticles’ uptake, diffusion, long term toxicity and excretion remain uncertain, the potential biological applications make them a potential candidate to extensively substitute chemically synthesized NPs in the coming years.

Author Contributions

Conceptualization, M.O. and S.M.; writing-original draft preparation, A.T.K., M.A., I.A., S.K.N. and S.M.; writing-review and editing, M.O., S.M.; funding acquisition, S.M.

Funding

This research received no external funding. The APC was funded by Fondren Library, Rice University, Houston, TX, United States.

Acknowledgments

Sudip Mukherjee acknowledges Fondren Library, Rice University, Houston, TX, United States for financial support for the APC of this work. He also acknowledges Omid Veiseh (supervisor/mentor) for providing continuous support for his postdoctoral research.

Conflicts of Interest

There is no conflict of interest. The authors have no other pertinent affiliations or financial connection with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

References

  1. Slavin, Y.N.; Asnis, J.; Häfeli, U.O.; Bach, H. Metal nanoparticles: Understanding the mechanisms behind antibacterial activity. J. Nanobiotechnol. 2017, 15, 65. [Google Scholar] [CrossRef] [PubMed]
  2. Khan, I.; Saeed, K.; Khan, I. Nanoparticles: Properties, applications and toxicities. Arab. J. Chem. 2017. [Google Scholar] [CrossRef]
  3. FutureMarketInsights Global Market for Metal & Metal Oxide Nanoparticles to Surge at More Than 10% CAGR. Available online: http://markets.businessinsider.com/news/stocks/Global-Market-forMetal-Metal-Oxide-Nanoparticles-to-Surge-at-More-Than-10CAGR-1001862836 (accessed on 21 November 2018).
  4. Mukherjee, S.; Vinothkumar, B.; Prashanthi, S.; Bangal, P.R.; Sreedhar, B.; Patra, C.R. Potential therapeutic and diagnostic applications of one-step in situ biosynthesized gold nanoconjugates (2-in-1 system) in cancer treatment. RSC Adv. 2013, 3, 2318–2329. [Google Scholar] [CrossRef]
  5. Patra, S.; Mukherjee, S.; Barui, A.K.; Ganguly, A.; Sreedhar, B.; Patra, C.R. Green synthesis, characterization of gold and silver nanoparticles and their potential application for cancer therapeutics. Mater. Sci. Eng. C 2015, 53, 298–309. [Google Scholar] [CrossRef] [PubMed]
  6. Ovais, M.; Zia, N.; Ahmad, I.; Khalil, A.T.; Raza, A.; Ayaz, M.; Sadiq, A.; Ullah, F.; Shinwari, Z.K. Phyto-Therapeutic and Nanomedicinal Approaches to Cure Alzheimer’s Disease: Present Status and Future Opportunities. Front. Aging Neurosci. 2018, 10. [Google Scholar] [CrossRef] [PubMed]
  7. Mukherjee, S.; Dasari, M.; Priyamvada, S.; Kotcherlakota, R.; Bollu, V.S.; Patra, C.R. A green chemistry approach for the synthesis of gold nanoconjugates that induce the inhibition of cancer cell proliferation through induction of oxidative stress and their in vivo toxicity study. J. Mater. Chem. B 2015, 3, 3820–3830. [Google Scholar] [CrossRef]
  8. Ovais, M.; Khalil, A.T.; Raza, A.; Khan, M.A.; Ahmad, I.; Islam, N.U.; Saravanan, M.; Ubaid, M.F.; Ali, M.; Shinwari, Z.K. Green synthesis of silver nanoparticles via plant extracts: beginning a new era in cancer theranostics. Nanomedicine 2016, 12, 3157–3177. [Google Scholar] [CrossRef] [PubMed]
  9. Salunke, G.R.; Ghosh, S.; Kumar, R.S.; Khade, S.; Vashisth, P.; Kale, T.; Chopade, S.; Pruthi, V.; Kundu, G.; Bellare, J.R. Rapid efficient synthesis and characterization of silver, gold, and bimetallic nanoparticles from the medicinal plant Plumbago zeylanica and their application in biofilm control. Int. J. Nanomed. 2014, 9, 2635. [Google Scholar]
  10. Barabadi, H.; Ovais, M.; Shinwari, Z.K.; Saravanan, M. Anti-cancer green bionanomaterials: present status and future prospects. Green Chem. Lett. Rev. 2017, 10, 285–314. [Google Scholar] [CrossRef]
  11. Emmanuel, R.; Saravanan, M.; Ovais, M.; Padmavathy, S.; Shinwari, Z.K.; Prakash, P. Antimicrobial efficacy of drug blended biosynthesized colloidal gold nanoparticles from Justicia glauca against oral pathogens: a nanoantibiotic approach. Microb. Pathog. 2017, 113, 295–302. [Google Scholar] [CrossRef] [PubMed]
  12. Hulkoti, N.I.; Taranath, T. Biosynthesis of nanoparticles using microbes—A review. Colloids Surf. B 2014, 121, 474–483. [Google Scholar] [CrossRef] [PubMed]
  13. Tang, L.; Shi, J.; Wu, H.; Zhang, S.; Liu, H.; Zou, H.; Wu, Y.; Zhao, J.; Jiang, Z. In situ biosynthesis of ultrafine metal nanoparticles within a metal-organic framework for efficient heterogeneous catalysis. Nanotechnology 2017, 28, 365604. [Google Scholar] [CrossRef] [PubMed]
  14. Ovais, M.; Raza, A.; Naz, S.; Islam, N.U.; Khalil, A.T.; Ali, S.; Khan, M.A.; Shinwari, Z.K. Current state and prospects of the phytosynthesized colloidal gold nanoparticles and their applications in cancer theranostics. Appl. Microbiol. Biotechnol. 2017, 101, 3551–3565. [Google Scholar] [CrossRef] [PubMed]
  15. Ovais, M.; Khalil, A.T.; Islam, N.U.; Ahmad, I.; Ayaz, M.; Saravanan, M.; Shinwari, Z.K.; Mukherjee, S. Role of plant phytochemicals and microbial enzymes in biosynthesis of metallic nanoparticles. Appl. Microbiol. Biotechnol. 2018, 102, 6799–6814. [Google Scholar] [CrossRef] [PubMed]
  16. Singh, P.; Kim, Y.-J.; Zhang, D.; Yang, D.-C. Biological synthesis of nanoparticles from plants and microorganisms. Trends Biotechnol. 2016, 34, 588–599. [Google Scholar] [CrossRef] [PubMed]
  17. Baker, S.; Rakshith, D.; Kavitha, K.S.; Santosh, P.; Kavitha, H.U.; Rao, Y.; Satish, S. Plants: Emerging as nanofactories towards facile route in synthesis of nanoparticles. BioImpacts 2013, 3, 111. [Google Scholar] [PubMed]
  18. Kumari, R.; Barsainya, M.; Singh, D.P. Biogenic synthesis of silver nanoparticle by using secondary metabolites from Pseudomonas aeruginosa DM1 and its anti-algal effect on Chlorella vulgaris and Chlorella pyrenoidosa. Environ. Sci. Pollut. Res. 2017, 24, 4645–4654. [Google Scholar] [CrossRef] [PubMed]
  19. Patra, C.R.; Mukherjee, S.; Kotcherlakota, R. Biosynthesized silver nanoparticles: A step forward for cancer theranostics? Nanomedicine 2014, 9, 1445–1448. [Google Scholar] [CrossRef] [PubMed]
  20. Mukherjee, S.; Nethi, S.K.; Patra, C.R. Green synthesized gold nanoparticles for future biomedical applications. In Particulate Technology for Delivery of Therapeutics; Springer: Singapore, 2017; pp. 359–393. [Google Scholar]
  21. Khalil, A.T.; Ovais, M.; Ullah, I.; Ali, M.; Shinwari, Z.K.; Khamlich, S.; Maaza, M. Sageretia thea (Osbeck.) mediated synthesis of zinc oxide nanoparticles and its biological applications. Nanomedicine 2017, 12, 1767–1789. [Google Scholar] [CrossRef] [PubMed]
  22. Khalil, A.T.; Ovais, M.; Ullah, I.; Ali, M.; Shinwari, Z.K.; Hassan, D.; Maaza, M. Sageretia thea (Osbeck.) modulated biosynthesis of NiO nanoparticles and their in vitro pharmacognostic, antioxidant and cytotoxic potential. Artif. Cells Nanomed. Biotechnol. 2018, 46, 838–852. [Google Scholar] [CrossRef] [PubMed]
  23. Makarov, V.; Love, A.; Sinitsyna, O.; Makarova, S.; Yaminsky, I.; Taliansky, M.; Kalinina, N. “Green” nanotechnologies: Synthesis of metal nanoparticles using plants. Acta Nat. 2014, 6, 35–44. [Google Scholar]
  24. Sintubin, L.; Verstraete, W.; Boon, N. Biologically produced nanosilver: Current state and future perspectives. Biotechnol. Bioeng. 2012, 109, 2422–2436. [Google Scholar] [CrossRef] [PubMed]
  25. Mukherjee, S.; Sushma, V.; Patra, S.; Barui, A.K.; Bhadra, M.P.; Sreedhar, B.; Patra, C.R. Green chemistry approach for the synthesis and stabilization of biocompatible gold nanoparticles and their potential applications in cancer therapy. Nanotechnology 2012, 23, 455103. [Google Scholar] [CrossRef] [PubMed]
  26. Mukherjee, S.; Sau, S.; Madhuri, D.; Bollu, V.S.; Madhusudana, K.; Sreedhar, B.; Banerjee, R.; Patra, C.R. Green synthesis and characterization of monodispersed gold nanoparticles: Toxicity study, delivery of doxorubicin and its bio-distribution in mouse model. J. Biomed. Nanotechnol. 2016, 12, 165–181. [Google Scholar] [CrossRef] [PubMed]
  27. Mukherjee, S.; Chowdhury, D.; Kotcherlakota, R.; Patra, S. Potential theranostics application of bio-synthesized silver nanoparticles (4-in-1 system). Theranostics 2014, 4, 316–335. [Google Scholar] [CrossRef] [PubMed]
  28. Singh, R.; Wagh, P.; Wadhwani, S.; Gaidhani, S.; Kumbhar, A.; Bellare, J.; Chopade, B.A. Synthesis, optimization, and characterization of silver nanoparticles from Acinetobacter calcoaceticus and their enhanced antibacterial activity when combined with antibiotics. Int. J. Nanomed. 2013, 8, 4277. [Google Scholar] [Green Version]
  29. Singh, R.; Shedbalkar, U.U.; Wadhwani, S.A.; Chopade, B.A. Bacteriagenic silver nanoparticles: Synthesis, mechanism, and applications. Appl. Microbiol. Biotechnol. 2015, 99, 4579–4593. [Google Scholar] [CrossRef] [PubMed]
  30. Saravanan, M.; Barik, S.K.; Mubarak Ali, 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] [PubMed]
  31. Klaus, T.; Joerger, R.; Olsson, E.; Granqvist, C.-G. Silver-based crystalline nanoparticles, microbially fabricated. Proc. Natl. Acad. Sci. USA 1999, 96, 13611–13614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Pugazhenthiran, N.; Anandan, S.; Kathiravan, G.; Prakash, N.K.U.; Crawford, S.; Ashokkumar, M. Microbial synthesis of silver nanoparticles by Bacillus sp. J. Nanopart. Res. 2009, 11, 1811. [Google Scholar] [CrossRef]
  33. Husseiny, M.; El-Aziz, M.A.; Badr, Y.; Mahmoud, M. Biosynthesis of gold nanoparticles using Pseudomonas aeruginosa. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2007, 67, 1003–1006. [Google Scholar] [CrossRef] [PubMed]
  34. He, S.; Guo, Z.; Zhang, Y.; Zhang, S.; Wang, J.; Gu, N. Biosynthesis of gold nanoparticles using the bacteria Rhodopseudomonas capsulata. Mater. Lett. 2007, 61, 3984–3987. [Google Scholar] [CrossRef]
  35. Dhandapani, P.; Siddarth, A.S.; Kamalasekaran, S.; Maruthamuthu, S.; Rajagopal, G. Bio-approach: Ureolytic bacteria mediated synthesis of ZnO nanocrystals on cotton fabric and evaluation of their antibacterial properties. Carbohyd. Polym. 2014, 103, 448–455. [Google Scholar] [CrossRef] [PubMed]
  36. Selvarajan, E.; Mohanasrinivasan, V. Biosynthesis and characterization of ZnO nanoparticles using Lactobacillus plantarum VITES07. Mater. Lett. 2013, 112, 180–182. [Google Scholar] [CrossRef]
  37. Jayaseelan, C.; Rahuman, A.A.; Kirthi, A.V.; Marimuthu, S.; Santhoshkumar, T.; Bagavan, A.; Gaurav, K.; Karthik, L.; Rao, K.B. Novel microbial route to synthesize ZnO nanoparticles using Aeromonas hydrophila and their activity against pathogenic bacteria and fungi. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2012, 90, 78–84. [Google Scholar] [CrossRef] [PubMed]
  38. Rad, M.; Taran, M.; Alavi, M. Effect of Incubation Time, CuSO4 and Glucose Concentrations on Biosynthesis of Copper Oxide (CuO) Nanoparticles with Rectangular Shape and Antibacterial Activity: Taguchi Method Approach. Nano Biomed. Eng. 2018, 10, 25–33. [Google Scholar] [CrossRef]
  39. Fatemi, M.; Mollania, N.; Momeni-Moghaddam, M.; Sadeghifar, F. Extracellular biosynthesis of magnetic iron oxide nanoparticles by Bacillus cereus strain HMH1: Characterization and in vitro cytotoxicity analysis on MCF-7 and 3T3 cell lines. J. Biotechnol. 2018, 270, 1–11. [Google Scholar] [CrossRef] [PubMed]
  40. Nair, B.; Pradeep, T. Coalescence of nanoclusters and formation of submicron crystallites assisted by Lactobacillus strains. Cryst. Growth Des. 2002, 2, 293–298. [Google Scholar] [CrossRef]
  41. Lengke, M.F.; Fleet, M.E.; Southam, G. Biosynthesis of silver nanoparticles by filamentous cyanobacteria from a silver (I) nitrate complex. Langmuir 2007, 23, 2694–2699. [Google Scholar] [CrossRef] [PubMed]
  42. Patel, V.; Berthold, D.; Puranik, P.; Gantar, M. Screening of cyanobacteria and microalgae for their ability to synthesize silver nanoparticles with antibacterial activity. Biotechnol. Rep. 2015, 5, 112–119. [Google Scholar] [CrossRef] [PubMed]
  43. Tomer, A.K.; Rahi, T.; Neelam, D.K.; Dadheech, P.K. Cyanobacterial extract-mediated synthesis of silver nanoparticles and their application in ammonia sensing. Int. Microbiol. 2018. [Google Scholar] [CrossRef]
  44. Satapathy, S.; Shukla, S.P. Application of a marine cyanobacterium Phormidium fragile for green synthesis of silver nanoparticles. Indian J. Biotechnol. 2017, 16, 110–113. [Google Scholar]
  45. Sonker, A.S.; Pathak, J.; Kannaujiya, V.; Sinha, R. Characterization and in vitro antitumor, antibacterial and antifungal activities of green synthesized silver nanoparticles using cell extract of Nostoc sp. strain HKAR-2. Can. J. Biotechnol. 2017, 1, 26–37. [Google Scholar] [CrossRef]
  46. Bakir, E.; Younis, N.; Mohamed, M.; El Semary, N. Cyanobacteria as Nanogold Factories: Chemical and Anti-Myocardial Infarction Properties of Gold Nanoparticles Synthesized by Lyngbya majuscula. Mar. Drugs 2018, 16, 217. [Google Scholar] [CrossRef] [PubMed]
  47. Molnár, Z.; Bódai, V.; Szakacs, G.; Erdélyi, B.; Fogarassy, Z.; Sáfrán, G.; Varga, T.; Kónya, Z.; Tóth-Szeles, E.; Szűcs, R. Green synthesis of gold nanoparticles by thermophilic filamentous fungi. Sci. Rep. 2018, 8, 3943. [Google Scholar] [CrossRef] [PubMed]
  48. Zhao, X.; Zhou, L.; Riaz Rajoka, M.S.; Yan, L.; Jiang, C.; Shao, D.; Zhu, J.; Shi, J.; Huang, Q.; Yang, H. Fungal silver nanoparticles: Synthesis, application and challenges. Crit. Rev. Biotechnol. 2018, 38, 817–835. [Google Scholar] [CrossRef] [PubMed]
  49. Alghuthaymi, M.A.; Almoammar, H.; Rai, M.; Said-Galiev, E.; Abd-Elsalam, K.A. Myconanoparticles: Synthesis and their role in phytopathogens management. Biotechnol. Biotechnol. Eq. 2015, 29, 221–236. [Google Scholar] [CrossRef] [PubMed]
  50. Castro-Longoria, E.; Vilchis-Nestor, A.R.; Avalos-Borja, M. Biosynthesis of silver, gold and bimetallic nanoparticles using the filamentous fungus Neurospora crassa. Colloids Surf. B 2011, 83, 42–48. [Google Scholar] [CrossRef] [PubMed]
  51. Zhang, X.; He, X.; Wang, K.; Yang, X. Different active biomolecules involved in biosynthesis of gold nanoparticles by three fungus species. J. Biomed. Nanotechnol. 2011, 7, 245–254. [Google Scholar] [CrossRef] [PubMed]
  52. AbdelRahim, K.; Mahmoud, S.Y.; Ali, A.M.; Almaary, K.S.; Mustafa, A.E.-Z.M.; Husseiny, S.M. Extracellular biosynthesis of silver nanoparticles using Rhizopus stolonifer. Saudi J. Biol. Sci. 2017, 24, 208–216. [Google Scholar] [CrossRef] [PubMed]
  53. Jalal, M.; Ansari, M.; Alzohairy, M.; Ali, S.; Khan, H.; Almatroudi, A.; Raees, K. Biosynthesis of Silver Nanoparticles from Oropharyngeal Candida glabrata Isolates and Their Antimicrobial Activity against Clinical Strains of Bacteria and Fungi. Nanomaterials 2018, 8, 586. [Google Scholar] [CrossRef] [PubMed]
  54. Kalpana, V.; Kataru, B.A.S.; Sravani, N.; Vigneshwari, T.; Panneerselvam, A.; Rajeswari, V.D. Biosynthesis of Zinc oxide nanoparticles using culture filtrates of Aspergillus niger: Antimicrobial textiles and dye degradation studies. OpenNano 2018. [Google Scholar] [CrossRef]
  55. Vijayanandan, A.S.; Balakrishnan, R.M. Biosynthesis of cobalt oxide nanoparticles using endophytic fungus Aspergillus nidulans. J. Environ. Manag. 2018, 218, 442–450. [Google Scholar] [CrossRef] [PubMed]
  56. Sanaeimehr, Z.; Javadi, I.; Namvar, F. Antiangiogenic and antiapoptotic effects of green-synthesized zinc oxide nanoparticles using Sargassum muticum algae extraction. Cancer Nanotechnol. 2018, 9, 3. [Google Scholar] [CrossRef] [PubMed]
  57. Maceda, A.F.; Ouano, J.J.S.; Que, M.C.O.; Basilia, B.A.; Potestas, M.J.; Alguno, A.C. Controlling the Absorption of Gold Nanoparticles via Green Synthesis Using Sargassum crassifolium Extract. In Key Engineering Materials; Trans Tech Publ: Clausthal-Zellerfeld, Germany, 2018; pp. 44–48. [Google Scholar]
  58. Gu, H.; Chen, X.; Chen, F.; Zhou, X.; Parsaee, Z. Ultrasound-assisted biosynthesis of CuO-NPs using brown alga Cystoseira trinodis: Characterization, photocatalytic AOP, DPPH scavenging and antibacterial investigations. Ultrason. Sonochem. 2018, 41, 109–119. [Google Scholar] [CrossRef] [PubMed]
  59. Koopi, H.; Buazar, F. A novel one-pot biosynthesis of pure alpha aluminum oxide nanoparticles using the macroalgae Sargassum ilicifolium: A green marine approach. Ceram. Int. 2018, 44, 8940–8945. [Google Scholar] [CrossRef]
  60. Vijayaraghavan, K.; Mahadevan, A.; Sathishkumar, M.; Pavagadhi, S.; Balasubramanian, R. Biosynthesis of Au (0) from Au (III) via biosorption and bioreduction using brown marine alga Turbinaria conoides. Chem. Eng. J. 2011, 167, 223–227. [Google Scholar] [CrossRef]
  61. Ramakrishna, M.; Babu, D.R.; Gengan, R.M.; Chandra, S.; Rao, G.N. Green synthesis of gold nanoparticles using marine algae and evaluation of their catalytic activity. J. Nanostruct. Chem. 2016, 6, 1–13. [Google Scholar] [CrossRef]
  62. Swaminathan, S.; Murugesan, S.; Damodarkumar, S.; Dhamotharan, R.; Bhuvaneshwari, S. Synthesis and characterization of gold nanoparticles from alga Acanthophora spicifera (VAHL) Boergesen. Int. J. Nanosci. Nanotechnol. 2011, 2, 85–94. [Google Scholar]
  63. Ghodake, G.; Lee, D.S. Biological synthesis of gold nanoparticles using the aqueous extract of the brown algae Laminaria japonica. J. Nanoelectron. Optoelectron. 2011, 6, 268–271. [Google Scholar] [CrossRef]
  64. Govindaraju, K.; Basha, S.K.; Kumar, V.G.; Singaravelu, G. Silver, gold and bimetallic nanoparticles production using single-cell protein (Spirulina platensis) Geitler. J. Mater. Sci. 2008, 43, 5115–5122. [Google Scholar] [CrossRef]
  65. Wypij, M.; Golinska, P.; Dahm, H.; Rai, M. Actinobacterial-mediated synthesis of silver nanoparticles and their activity against pathogenic bacteria. IET Nanobiotechnol. 2016, 11, 336–342. [Google Scholar] [CrossRef] [PubMed]
  66. Wadhwani, S.A.; Shedbalkar, U.U.; Singh, R.; Vashisth, P.; Pruthi, V.; Chopade, B.A. Kinetics of synthesis of gold nanoparticles by Acinetobacter sp. SW30 isolated from environment. Indian J. Microbiol. 2016, 56, 439–444. [Google Scholar] [CrossRef] [PubMed]
  67. Prema, P.; Iniya, P.; Immanuel, G. Microbial mediated synthesis, characterization, antibacterial and synergistic effect of gold nanoparticles using Klebsiella pneumoniae (MTCC-4030). RSC Adv. 2016, 6, 4601–4607. [Google Scholar] [CrossRef]
  68. Manikprabhu, D.; Cheng, J.; Chen, W.; Sunkara, A.K.; Mane, S.B.; Kumar, R.; Hozzein, W.N.; Duan, Y.-Q.; Li, W.-J. Sunlight mediated synthesis of silver nanoparticles by a novel actinobacterium (Sinomonas mesophila MPKL 26) and its antimicrobial activity against multi drug resistant Staphylococcus aureus. J. Photochem. Photobiol. B 2016, 158, 202–205. [Google Scholar] [CrossRef] [PubMed]
  69. Syed, B.; Prasad, N.M.; Satish, S. Endogenic mediated synthesis of gold nanoparticles bearing bactericidal activity. J. Microsc. Ultrastruct. 2016, 4, 162–166. [Google Scholar] [PubMed]
  70. Gan, L.; Zhang, S.; Zhang, Y.; He, S.; Tian, Y. Biosynthesis, characterization and antimicrobial activity of silver nanoparticles by a halotolerant Bacillus endophyticus SCU-L. Prep. Biochem. Biotechnol. 2018. [Google Scholar] [CrossRef] [PubMed]
  71. Vijayabharathi, R.; Sathya, A.; Gopalakrishnan, S. Extracellular biosynthesis of silver nanoparticles using Streptomyces griseoplanus SAI-25 and its antifungal activity against Macrophomina phaseolina, the charcoal rot pathogen of sorghum. Biocatal. Agric. Biotechnol. 2018, 14, 166–171. [Google Scholar] [CrossRef]
  72. Ranjani, A.; Gopinath, P.M.; Ananth, S.; Narchonai, G.; Santhanam, P.; Thajuddin, N.; Dhanasekaran, D. Multidimensional dose–response toxicity exploration of silver nanoparticles from Nocardiopsis flavascens RD30. Appl. Nanosci. 2018, 8, 699–713. [Google Scholar] [CrossRef]
  73. Bing, W.; Sun, H.; Wang, F.; Song, Y.; Ren, J. Hydrogen-producing Hyperthermophilic Bacteria Synthesized Size-controllable Fine Gold Nanoparticles with Excellence for Eradicating Biofilm and Antibacterial Applications. J. Mater. Chem. B 2018. [Google Scholar] [CrossRef]
  74. Lv, Q.; Zhang, B.; Xing, X.; Zhao, Y.; Cai, R.; Wang, W.; Gu, Q. Biosynthesis of copper nanoparticles using Shewanella loihica PV-4 with antibacterial activity: Novel approach and mechanisms investigation. J. Hazard. Mater. 2018, 347, 141–149. [Google Scholar] [CrossRef] [PubMed]
  75. Ahmed, E.; Kalathil, S.; Shi, L.; Alharbi, O.; Wang, P. Synthesis of ultra-small platinum, palladium and gold nanoparticles by Shewanella loihica PV-4 electrochemically active biofilms and their enhanced catalytic activities. J. Saudi Chem. Soc. 2018. [Google Scholar] [CrossRef]
  76. Jafari, M.; Rokhbakhsh-Zamin, F.; Shakibaie, M.; Moshafi, M.H.; Ameri, A.; Rahimi, H.R.; Forootanfar, H. Cytotoxic and antibacterial activities of biologically synthesized gold nanoparticles assisted by Micrococcus yunnanensis strain J2. Biocatal. Agric. Biotechnol. 2018. [Google Scholar] [CrossRef]
  77. Camas, M.; Camas, A.S.; Kyeremeh, K. Extracellular Synthesis and Characterization of Gold Nanoparticles Using Mycobacterium sp. BRS2A-AR2 Isolated from the Aerial Roots of the Ghanaian Mangrove Plant, Rhizophora racemosa. Indian J. Microbiol. 2018, 58, 214–221. [Google Scholar] [CrossRef] [PubMed]
  78. Shah, R.; Oza, G.; Pandey, S.; Sharon, M. Biogenic fabrication of gold nanoparticles using Halomonas salina. J. Microbiol. Biotechnol. 2017, 2, 485–492. [Google Scholar]
  79. Kobashigawa, J.M.; Robles, C.A.; Ricci, M.L.M.; Carmarán, C.C. Influence of strong bases on the synthesis of silver nanoparticles (AgNPs) using the ligninolytic fungi Trametes trogii. Saudi J.Biol. Sci. 2018. [Google Scholar] [CrossRef]
  80. 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]
  81. Tripathi, R.M.; Shrivastav, B.R.; Shrivastav, A. Antibacterial and catalytic activity of biogenic gold nanoparticles synthesised byTrichoderma harzianum. IET Nanobiotechnol. 2018, 12, 509–513. [Google Scholar] [CrossRef] [PubMed]
  82. Ahmed, A.-A.; Hamzah, H.; Maaroof, M. Analyzing formation of silver nanoparticles from the filamentous fungus Fusarium oxysporum and their antimicrobial activity. Turk. J. Biol. 2018, 42, 54–62. [Google Scholar] [CrossRef]
  83. El Domany, E.B.; Essam, T.M.; Ahmed, A.E.; Farghali, A.A. Biosynthesis Physico-Chemical Optimization of Gold Nanoparticles as Anti-Cancer and Synergetic Antimicrobial Activity Using Pleurotus ostreatus Fungus. J. Appl. Pharm. Sci. Vol. 2018, 8, 119–128. [Google Scholar]
  84. Singh, P.S.; Vidyasagar, G. Biosynthesis of antibacterial silver nano-particles from Aspergillus terreus. World News Nat. Sci. 2018, 16, 117–124. [Google Scholar]
  85. Mohanta, Y.K.; Nayak, D.; Biswas, K.; Singdevsachan, S.K.; Abd_Allah, E.F.; Hashem, A.; Alqarawi, A.A.; Yadav, D.; Mohanta, T.K. Silver Nanoparticles Synthesized Using Wild Mushroom Show Potential Antimicrobial Activities against Food Borne Pathogens. Molecules 2018, 23, 655. [Google Scholar] [CrossRef] [PubMed]
  86. Saravanan, M.; Arokiyaraj, S.; Lakshmi, T.; Pugazhendhi, A. Synthesis of silver nanoparticles from Phenerochaete chrysosporium (MTCC-787) and their antibacterial activity against human pathogenic bacteria. Microb. Pathog. 2018, 117, 68–72. [Google Scholar] [CrossRef] [PubMed]
  87. Neethu, S.; Midhun, S.J.; Sunil, M.; Soumya, S.; Radhakrishnan, E.; Jyothis, M. Efficient visible light induced synthesis of silver nanoparticles by Penicillium polonicum ARA 10 isolated from Chetomorpha antennina and its antibacterial efficacy against Salmonella enterica serovar Typhimurium. J. Photochem. Photobiol. B 2018, 180, 175–185. [Google Scholar] [CrossRef] [PubMed]
  88. Spagnoletti, F.N.; Spedalieri, C.; Kronberg, F.; Giacometti, R. Extracellular biosynthesis of bactericidal Ag/AgCl nanoparticles for crop protection using the fungus Macrophomina phaseolina. J. Environ. Manag. 2019, 231, 457–466. [Google Scholar] [CrossRef] [PubMed]
  89. Cunha, F.A.; da CSO Cunha, M.; da Frota, S.M.; Mallmann, E.J.; Freire, T.M.; Costa, L.S.; Paula, A.J.; Menezes, E.A.; Fechine, P.B. Biogenic synthesis of multifunctional silver nanoparticles from Rhodotorula glutinis and Rhodotorula mucilaginosa: Antifungal, catalytic and cytotoxicity activities. World J. Microbiol. Biotechnol. 2018, 34, 127. [Google Scholar] [CrossRef] [PubMed]
  90. Popli, D.; Anil, V.; Subramanyam, A.B.; Rao, S.N.; Rai, R.V.; Govindappa, M. Endophyte fungi, Cladosporium species-mediated synthesis of silver nanoparticles possessing in vitro antioxidant, anti-diabetic and anti-Alzheimer activity. Artif. Cells Nanomed. 2018. [Google Scholar] [CrossRef] [PubMed]
  91. Joshi, C.G.; Danagoudar, A.; Poyya, J.; Kudva, A.K.; Dhananjaya, B. Biogenic synthesis of gold nanoparticles by marine endophytic fungus-Cladosporium cladosporioides isolated from seaweed and evaluation of their antioxidant and antimicrobial properties. Process Biochem. 2017, 63, 137–144. [Google Scholar]
  92. Farsi, M.; Farokhi, S. Biosynthesis of Antibacterial Silver Nanoparticles by Endophytic Fungus Nemania sp. Isolated From Taxus baccata L.(Iranian Yew). Zahedan J. Res. Med. Sci. 2018, 20. [Google Scholar] [CrossRef]
  93. Subramaniyan, S.A.; Sheet, S.; Vinothkannan, M.; Yoo, D.J.; Lee, Y.S.; Belal, S.A.; Shim, K.S. One-Pot Facile Synthesis of Pt Nanoparticles Using Cultural Filtrate of Microgravity Simulated Grown P. chrysogenum and Their Activity on Bacteria and Cancer Cells. J. Nanosci. Nanotechnol. 2018, 18, 3110–3125. [Google Scholar] [CrossRef] [PubMed]
  94. Shen, W.; Qu, Y.; Pei, X.; Li, S.; You, S.; Wang, J.; Zhang, Z.; Zhou, J. Catalytic reduction of 4-nitrophenol using gold nanoparticles biosynthesized by cell-free extracts of Aspergillus sp. WL-Au. J. Hazard. Mater. 2017, 321, 299–306. [Google Scholar] [CrossRef] [PubMed]
  95. Kumaresan, M.; Anand, K.V.; Govindaraju, K.; Tamilselvan, S.; Kumar, V.G. Seaweed Sargassum wightii mediated preparation of zirconia (ZrO2) nanoparticles and their antibacterial activity against gram positive and gram negative bacteria. Microb. Pathog. 2018, 124, 311–315. [Google Scholar] [CrossRef] [PubMed]
  96. Bao, Z.; Lan, C.Q. Mechanism of light-dependent biosynthesis of silver nanoparticles mediated by cell extract of Neochloris oleoabundans. Colloids Surf. B 2018. [Google Scholar] [CrossRef] [PubMed]
  97. González-Ballesteros, N.; Prado-López, S.; Rodriguez-Gonzalez, J.; Lastra, M.; Rodríguez-Argüelles, M. Green synthesis of gold nanoparticles using brown algae cystoseira baccata: Its activity in colon cancer cells. Colloids Surf. B 2017, 153, 190–198. [Google Scholar] [CrossRef] [PubMed]
  98. Pytlik, N.; Kaden, J.; Finger, M.; Naumann, J.; Wanke, S.; Machill, S.; Brunner, E. Biological synthesis of gold nanoparticles by the diatom Stephanopyxis turris and in vivo SERS analyses. Algal Res. 2017, 28, 9–15. [Google Scholar] [CrossRef]
  99. Abdel-Raouf, N.; Al-Enazi, N.M.; Ibraheem, I.B. Green biosynthesis of gold nanoparticles using Galaxaura elongata and characterization of their antibacterial activity. Arab. J. Chem. 2017, 10, S3029–S3039. [Google Scholar] [CrossRef]
  100. Arsiya, F.; Sayadi, M.H.; Sobhani, S. Green synthesis of palladium nanoparticles using Chlorella vulgaris. Mater. Lett. 2017, 186, 113–115. [Google Scholar] [CrossRef]
  101. Ramkumar, V.S.; Pugazhendhi, A.; Gopalakrishnan, K.; Sivagurunathan, P.; Saratale, G.D.; Dung, T.N.B.; Kannapiran, E. Biofabrication and characterization of silver nanoparticles using aqueous extract of seaweed Enteromorpha compressa and its biomedical properties. Biotechnol. Rep. 2017, 14, 1–7. [Google Scholar] [CrossRef] [PubMed]
  102. Vanlalveni, C.; Rajkumari, K.; Biswas, A.; Adhikari, P.P.; Lalfakzuala, R.; Rokhum, L. Green Synthesis of Silver Nanoparticles Using Nostoc linckia and its Antimicrobial Activity: A Novel Biological Approach. Bionanoscience 2018, 8, 624–631. [Google Scholar] [CrossRef]
  103. Zada, S.; Ahmad, A.; Khan, S.; Yu, X.; Chang, K.; Iqbal, A.; Ahmad, A.; Ullah, S.; Raza, M.; Khan, A. Biogenic synthesis of silver nanoparticles using extracts of Leptolyngbya JSC-1 that induce apoptosis in HeLa cell line and exterminate pathogenic bacteria. Artif. Cells Nanomed. Biotechnol. 2018. [Google Scholar] [CrossRef] [PubMed]
  104. Murugesan, S.; Bhuvaneswari, S.; Sivamurugan, V. Green synthesis, characterization of silver nanoparticles of a marine red alga Spyridia fusiformis and their antibacterial activity. Int. J. Pharm. Pharm. Sci. 2017, 9, 192–197. [Google Scholar] [CrossRef]
  105. Zhang, Z.; Chen, J.; Yang, Q.; Lan, K.; Yan, Z.; Chen, J. Eco-friendly intracellular microalgae synthesis of fluorescent CdSe QDs as a sensitive nanoprobe for determination of imatinib. Sens. Actuators B Chem. 2018, 263, 625–633. [Google Scholar] [CrossRef]
  106. Abdel-Raouf, N.; Al-Enazi, N.M.; Ibraheem, I.B.M.; Alharbi, R.M.; Alkhulaifi, M.M. Biosynthesis of silver nanoparticles by using of the marine brown alga Padina pavonia and their characterization. Saudi J. Biol. Sci. 2018. [Google Scholar] [CrossRef]
  107. Sayadi, M.H.; Salmani, N.; Heidari, A.; Rezaei, M.R. Bio-synthesis of palladium nanoparticle using Spirulina platensis alga extract and its application as adsorbent. Surf. Interfaces 2018, 10, 136–143. [Google Scholar] [CrossRef]
  108. Sharma, M.; Behl, K.; Nigam, S.; Joshi, M. TiO2-GO nanocomposite for photocatalysis and environmental applications: A green synthesis approach. Vacuum 2018, 156, 434–439. [Google Scholar] [CrossRef]
  109. Narayanan, K.B.; Sakthivel, N. Biological synthesis of metal nanoparticles by microbes. Adv. Colloids Interface Sci. 2010, 156. [Google Scholar] [CrossRef] [PubMed]
  110. Subbaiya, R.; Saravanan, M.; Priya, A.R.; Shankar, K.R.; Selvam, M.; Ovais, M.; Balajee, R.; Barabadi, H. Biomimetic synthesis of silver nanoparticles from Streptomyces atrovirens and their potential anticancer activity against human breast cancer cells. IET Nanobiotechnol. 2017, 11, 965–972. [Google Scholar] [CrossRef] [PubMed]
  111. 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]
  112. Mukherjee, S.; Patra, C.R. Biologically synthesized metal nanoparticles: Recent advancement and future perspectives in cancer theranostics. Future Sci. 2017. [Google Scholar] [CrossRef] [PubMed]
  113. Kumar, S.A.; Abyaneh, M.K.; Gosavi, S.; Kulkarni, S.K.; Pasricha, R.; Ahmad, A.; Khan, M. Nitrate reductase-mediated synthesis of silver nanoparticles from AgNO3. Biotechnol. Lett. 2007, 29, 439–445. [Google Scholar] [CrossRef] [PubMed]
  114. Kumar, S.A.; Ansary, A.A.; Ahmad, A.; Khan, M. Extracellular biosynthesis of CdSe quantum dots by the fungus, Fusarium oxysporum. J. Biomed. Nanotechnol. 2007, 3, 190–194. [Google Scholar] [CrossRef]
  115. 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]
  116. Durán, N.; Marcato, P.D.; Alves, O.L.; De Souza, G.I.; Esposito, E. Mechanistic aspects of biosynthesis of silver nanoparticles by several Fusarium oxysporum strains. J. Nanobiotechnol. 2005, 3, 8. [Google Scholar] [CrossRef] [PubMed]
  117. Senapati, S.; Ahmad, A.; Khan, M.I.; Sastry, M.; Kumar, R. Extracellular biosynthesis of bimetallic Au–Ag alloy nanoparticles. Small 2005, 1, 517–520. [Google Scholar] [CrossRef] [PubMed]
  118. Karbasian, M.; Atyabi, S.; Siadat, S.; Momen, S.; Norouzian, D. Optimizing nano-silver formation by Fusarium oxysporum PTCC 5115 employing response surface methodology. Am. J. Agric. Biol. Sci. 2008, 3, 433–437. [Google Scholar]
  119. Ahmad, A.; Mukherjee, P.; Mandal, D.; Senapati, S.; Khan, M.I.; Kumar, R.; Sastry, M. Enzyme mediated extracellular synthesis of CdS nanoparticles by the fungus, Fusarium oxysporum. J. Am. Chem. Soc. 2002, 124, 12108–12109. [Google Scholar] [CrossRef] [PubMed]
  120. Basavaraja, S.; Balaji, S.; Lagashetty, A.; Rajasab, A.; Venkataraman, A. Extracellular biosynthesis of silver nanoparticles using the fungus Fusarium semitectum. Mater. Res. Bull. 2008, 43, 1164–1170. [Google Scholar] [CrossRef]
  121. Ingle, A.; Rai, M.; Gade, A.; Bawaskar, M. Fusarium solani: A novel biological agent for the extracellular synthesis of silver nanoparticles. J. Nanopart. Res. 2009, 11, 2079. [Google Scholar] [CrossRef]
  122. Balaji, D.; Basavaraja, S.; Deshpande, R.; Mahesh, D.B.; Prabhakar, B.; Venkataraman, A. Extracellular biosynthesis of functionalized silver nanoparticles by strains of Cladosporium cladosporioides fungus. Colloids Surf. B 2009, 68, 88–92. [Google Scholar] [CrossRef] [PubMed]
  123. Gade, A.; Bonde, P.; Ingle, A.; Marcato, P.; Duran, N.; Rai, M. Exploitation of Aspergillus niger for synthesis of silver nanoparticles. J. Biobased Mater. Biol. 2008, 2, 243–247. [Google Scholar] [CrossRef]
  124. Bhainsa, K.C.; D’souza, S. Extracellular biosynthesis of silver nanoparticles using the fungus Aspergillus fumigatus. Colloids Surf. B 2006, 47, 160–164. [Google Scholar] [CrossRef] [PubMed]
  125. Kathiresan, K.; Manivannan, S.; Nabeel, M.; Dhivya, B. Studies on silver nanoparticles synthesized by a marine fungus, Penicillium fellutanum isolated from coastal mangrove sediment. Colloids Surf. B 2009, 71, 133–137. [Google Scholar] [CrossRef] [PubMed]
  126. Shaligram, N.S.; Bule, M.; Bhambure, R.; Singhal, R.S.; Singh, S.K.; Szakacs, G.; Pandey, A. Biosynthesis of silver nanoparticles using aqueous extract from the compactin producing fungal strain. Process. Biochem. 2009, 44, 939–943. [Google Scholar] [CrossRef]
  127. Singaravelu, G.; Arockiamary, J.; Kumar, V.G.; Govindaraju, K. A novel extracellular synthesis of monodisperse gold nanoparticles using marine alga, Sargassum wightii Greville. Colloids Surf. B 2007, 57, 97–101. [Google Scholar] [CrossRef] [PubMed]
  128. Lengke, M.F.; Fleet, M.E.; Southam, G. Morphology of gold nanoparticles synthesized by filamentous cyanobacteria from gold (I)− thiosulfate and gold (III)− chloride complexes. Langmuir 2006, 22, 2780–2787. [Google Scholar] [CrossRef] [PubMed]
  129. Lloyd, J.R.; Yong, P.; Macaskie, L.E. Enzymatic recovery of elemental palladium by using sulfate-reducing bacteria. Appl. Environ. Microbiol. 1998, 64, 4607–4609. [Google Scholar] [PubMed]
  130. Kashefi, K.; Lovley, D.R. Reduction of Fe (III), Mn (IV), and toxic metals at 100 C by Pyrobaculum islandicum. Appl. Environ. Microbiol. 2000, 66, 1050–1056. [Google Scholar] [CrossRef] [PubMed]
  131. Bao, H.; Lu, Z.; Cui, X.; Qiao, Y.; Guo, J.; Anderson, J.M.; Li, C.M. Extracellular microbial synthesis of biocompatible CdTe quantum dots. Acta Biomater. 2010, 6, 3534–3541. [Google Scholar] [CrossRef] [PubMed]
  132. Du, L.; Jiang, H.; Liu, X.; Wang, E. Biosynthesis of gold nanoparticles assisted by Escherichia coli DH5α and its application on direct electrochemistry of hemoglobin. Electrochem. Commun. 2007, 9, 1165–1170. [Google Scholar] [CrossRef]
  133. Kalimuthu, K.; Babu, R.S.; Venkataraman, D.; Bilal, M.; Gurunathan, S. Biosynthesis of silver nanocrystals by Bacillus licheniformis. Colloids Surf. B 2008, 65, 150–153. [Google Scholar] [CrossRef] [PubMed]
  134. Lee, J.-H.; Han, J.; Choi, H.; Hur, H.-G. Effects of temperature and dissolved oxygen on Se (IV) removal and Se (0) precipitation by Shewanella sp. HN-41. Chemosphere 2007, 68, 1898–1905. [Google Scholar] [CrossRef] [PubMed]
  135. Juibari, M.M.; Abbasalizadeh, S.; Jouzani, G.S.; Noruzi, M. Intensified biosynthesis of silver nanoparticles using a native extremophilic Ureibacillus thermosphaericus strain. Mater. Lett. 2011, 65, 1014–1017. [Google Scholar] [CrossRef]
  136. Sneha, K.; Sathishkumar, M.; Mao, J.; Kwak, I.; Yun, Y.-S. Corynebacterium glutamicum-mediated crystallization of silver ions through sorption and reduction processes. Chem. Eng. J. 2010, 162, 989–996. [Google Scholar] [CrossRef]
  137. Suresh, A.K.; Pelletier, D.A.; Wang, W.; Broich, M.L.; Moon, J.-W.; Gu, B.; Allison, D.P.; Joy, D.C.; Phelps, T.J.; Doktycz, M.J. Biofabrication of discrete spherical gold nanoparticles using the metal-reducing bacterium Shewanella oneidensis. Acta Biomater. 2011, 7, 2148–2152. [Google Scholar] [CrossRef] [PubMed]
  138. Lengke, M.F.; Ravel, B.; Fleet, M.E.; Wanger, G.; Gordon, R.A.; Southam, G. Mechanisms of gold bioaccumulation by filamentous cyanobacteria from gold (III)− chloride complex. Environ. Sci. Technol. 2006, 40, 6304–6309. [Google Scholar] [CrossRef] [PubMed]
  139. Vigneshwaran, N.; Kathe, A.A.; Varadarajan, P.; Nachane, R.P.; Balasubramanya, R. Biomimetics of silver nanoparticles by white rot fungus, Phaenerochaete chrysosporium. Colloids Surf. B 2006, 53, 55–59. [Google Scholar] [CrossRef] [PubMed]
  140. Vigneshwaran, N.; Ashtaputre, N.; Varadarajan, P.; Nachane, R.; Paralikar, K.; Balasubramanya, R. Biological synthesis of silver nanoparticles using the fungus Aspergillus flavus. Mater. Lett. 2007, 61, 1413–1418. [Google Scholar] [CrossRef]
  141. Senapati, S.; Mandal, D.; Ahmad, A. Fungus mediated synthesis of silver nanoparticles: A novel biological approach. Indian J. Phys. A 2004, 78, 101. [Google Scholar]
  142. Fayaz, A.M.; Balaji, K.; Kalaichelvan, P.; Venkatesan, R. Fungal based synthesis of silver nanoparticles—An effect of temperature on the size of particles. Colloids Surf. B 2009, 74, 123–126. [Google Scholar] [CrossRef] [PubMed]
  143. Agnihotri, M.; Joshi, S.; Kumar, A.R.; Zinjarde, S.; Kulkarni, S. Biosynthesis of gold nanoparticles by the tropical marine yeast Yarrowia lipolytica NCIM 3589. Mater. Lett. 2009, 63, 1231–1234. [Google Scholar] [CrossRef]
  144. Konishi, Y.; Ohno, K.; Saitoh, N.; Nomura, T.; Nagamine, S.; Hishida, H.; Takahashi, Y.; Uruga, T. Bioreductive deposition of platinum nanoparticles on the bacterium Shewanella algae. J. Biotechnol. 2007, 128, 648–653. [Google Scholar] [CrossRef] [PubMed]
  145. Sinha, A.; Khare, S.K. Mercury bioaccumulation and simultaneous nanoparticle synthesis by Enterobacter sp. cells. Bioresour. Technol. 2011, 102, 4281–4284. [Google Scholar] [CrossRef] [PubMed]
  146. Babu, M.G.; Gunasekaran, P. Production and structural characterization of crystalline silver nanoparticles from Bacillus cereus isolate. Colloids Surf. B 2009, 74, 191–195. [Google Scholar] [CrossRef] [PubMed]
  147. Kalishwaralal, K.; Deepak, V.; Pandian, S.R.K.; Kottaisamy, M.; BarathManiKanth, S.; Kartikeyan, B.; Gurunathan, S. Biosynthesis of silver and gold nanoparticles using Brevibacterium casei. Colloids Surf. B 2010, 77, 257–262. [Google Scholar] [CrossRef] [PubMed]
  148. 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. [Google Scholar] [CrossRef]
  149. Gericke, M.; Pinches, A. Biological synthesis of metal nanoparticles. Hydrometallurgy 2006, 83, 132–140. [Google Scholar] [CrossRef]
  150. Thakkar, K.N.; Mhatre, S.S.; Parikh, R.Y. Biological synthesis of metallic nanoparticles. Nanomedicine 2010, 6, 257–262. [Google Scholar] [CrossRef] [PubMed]
  151. Dauthal, P.; Mukhopadhyay, M. Noble metal nanoparticles: Plant-mediated synthesis, mechanistic aspects of synthesis, and applications. Ind. Eng. Chem. Res. 2016, 55, 9557–9577. [Google Scholar] [CrossRef]
  152. Ahmad, A.; Senapati, S.; Khan, M.I.; Kumar, R.; Sastry, M. Extracellular biosynthesis of monodisperse gold nanoparticles by a novel extremophilic actinomycete, Thermomonospora sp. Langmuir 2003, 19, 3550–3553. [Google Scholar] [CrossRef]
  153. Mukherjee, P.; Ahmad, A.; Mandal, D.; Senapati, S.; Sainkar, S.R.; Khan, M.I.; Parishcha, R.; Ajaykumar, P.; Alam, M.; Kumar, R. Fungus-mediated synthesis of silver nanoparticles and their immobilization in the mycelial matrix: A novel biological approach to nanoparticle synthesis. Nano Lett. 2001, 1, 515–519. [Google Scholar] [CrossRef]
  154. Mukherjee, P.; Ahmad, A.; Mandal, D.; Senapati, S.; Sainkar, S.R.; Khan, M.I.; Ramani, R.; Parischa, R.; Ajayakumar, P.; Alam, M. Bioreduction of AuCl4−ions by the fungus, Verticillium sp. and surface trapping of the gold nanoparticles formed. Angew. Chem. Int. Ed. 2001, 40, 3585–3588. [Google Scholar] [CrossRef]
  155. Southam, G.; Beveridge, T.J. The occurrence of sulfur and phosphorus within bacterially derived crystalline and pseudocrystalline octahedral gold formed in vitro. Geochim. Cosmochim. Acta 1996, 60, 4369–4376. [Google Scholar] [CrossRef]
  156. Sanghi, R.; Verma, P.; Puri, S. Enzymatic formation of gold nanoparticles using Phanerochaete chrysosporium. Adv. Chem. Eng. Sci. 2011, 1, 154. [Google Scholar] [CrossRef]
  157. Konishi, Y.; Tsukiyama, T.; Tachimi, T.; Saitoh, N.; Nomura, T.; Nagamine, S. Microbial deposition of gold nanoparticles by the metal-reducing bacterium Shewanella algae. Electrochim. Acta 2007, 53, 186–192. [Google Scholar] [CrossRef]
  158. Mody, V.V.; Siwale, R.; Singh, A.; Mody, H.R. Introduction to metallic nanoparticles. J. Pharm. Bioallied Sci. 2010, 2, 282. [Google Scholar] [CrossRef] [PubMed]
  159. Iravani, S. Green synthesis of metal nanoparticles using plants. Green Chem. 2011, 13, 2638–2650. [Google Scholar] [CrossRef]
  160. Nethi, S.K.; Mukherjee, S.; Veeriah, V.; Barui, A.K.; Chatterjee, S.; Patra, C.R. Bioconjugated gold nanoparticles accelerate the growth of new blood vessels through redox signaling. Chem. Commun. 2014, 50, 14367–14370. [Google Scholar] [CrossRef] [PubMed]
  161. Luo, C.-H.; Shanmugam, V.; Yeh, C.-S. Nanoparticle biosynthesis using unicellular and subcellular supports. NPG Asia Mater. 2015, 7, e209. [Google Scholar] [CrossRef]
  162. Korbekandi, H.; Iravani, S.; Abbasi, S. Production of nanoparticles using organisms. Crit. Rev. Biotechnol. 2009, 29, 279–306. [Google Scholar] [CrossRef] [PubMed]
  163. Mukherjee, S.; Patra, C.R. Therapeutic application of anti-angiogenic nanomaterials in cancers. Nanoscale 2016, 8, 12444–12470. [Google Scholar] [CrossRef] [PubMed]
  164. Wen, L.; Lin, Z.; Gu, P.; Zhou, J.; Yao, B.; Chen, G.; Fu, J. Extracellular biosynthesis of monodispersed gold nanoparticles by a SAM capping route. J. Nanopart. Res. 2009, 11, 279–288. [Google Scholar] [CrossRef]
  165. Balakrishnan, S.; Mukherjee, S.; Das, S.; Bhat, F.A.; Raja, S.P.; Patra, C.R.; Arunakaran, J. Gold nanoparticles-conjugated quercetin induces apoptosis via inhibition of EGFR/PI3K/Akt-mediated pathway in breast cancer cell lines (MCF-7 and MDA-MB-231). Cell Biochem. Funct. 2017, 35, 217–231. [Google Scholar] [CrossRef] [PubMed]
  166. Moon, J.-W.; Rawn, C.J.; Rondinone, A.J.; Love, L.J.; Roh, Y.; Everett, S.M.; Lauf, R.J.; Phelps, T.J. Large-scale production of magnetic nanoparticles using bacterial fermentation. J. Ind. Microbiol. Biotechnol. 2010, 37, 1023–1031. [Google Scholar] [CrossRef] [PubMed]
  167. Korbekandi, H.; Iravani, S.; Abbasi, S. Optimization of biological synthesis of silver nanoparticles using Lactobacillus casei subsp. casei. J. Chem. Technol. Biotechnol. 2012, 87, 932–937. [Google Scholar] [CrossRef]
Figure 1. Role of NADH and NADH-dependent microbial enzymes in the synthesis of metal nanoparticles (MtNPs).
Figure 1. Role of NADH and NADH-dependent microbial enzymes in the synthesis of metal nanoparticles (MtNPs).
Ijms 19 04100 g001
Table 1. Studies from 2016 to date, indicating different microorganisms for nanoparticle biosynthesis.
Table 1. Studies from 2016 to date, indicating different microorganisms for nanoparticle biosynthesis.
Serial NumberMicroorganisms Nanoparticle Size/ShapeApplicationReference
Bacteria
1ActinobacterAg13.2 nm/SphericalAntibacterial [65]
2AcinetobacterAu19 nm/Spherical-triangular-polyhedral-[66]
3Klebsiella pneumoniaAu10–15 nm/SphericalAntibacterial[67]
4Sinomonas mesophilaAg4–50 nm/SphericalAntibacterial[68]
5Pseudomonas fluorescensAu5–50 nm/Spherical Antibacterial[69]
6Bacillus endophyticusAg5.1 nm/SphericalAntimicrobial [70]
7Bacillus brevisAg41–68 nm/Spherical Antibacterial[30]
8StreptomycesgriseoplanusAg19.5–20.9 nm/SphericalAntifungal[71]
9Nocardiopsis flavascensAg5 and 50/SphericalCytotoxicity [72]
10Caldicellulosiruptor changbaiensisAu<20 nm/Spherical Antibacterial, Antibiofilm[73]
11Shewanella loihicaCu10–16 nm/SphericalAntibacterial[74]
12Shewanella loihicaPt1–10 nm/SphericalDye degradation[75]
13Shewanella loihicaPd1–12 nm/SphericalDye degradation[75]
14Shewanella loihicaAu2–15 nm/SphericalDye degradation[75]
15Micrococcus yunnanensisAu53.8 nm/Spherical Antibacterial, Anticancer [76]
16Mycobacterium sp.Au5–55 nm/SphericalAnticancer[77]
17Halomonas salinaAu30–100 nm/Spherical -[78]
Fungi
18Aspergillus nigerZnO53–69 nm/SphericalAntibacterial Dye degradation[54]
19Trametes trogiiAg5–65 nm/Spherical- Ellipsoidal -[79]
20Trichoderma longibrachiatumAg10 nm/Spherical Antifungal against phyto-pathogenic fungi[80]
21Trichoderma harzianumAu32–44 nm/SphericalAntibacterial, Dye degradation[81]
22Fusarium oxysporumAg21.3–37 nm/Spherical Antimicrobial [82]
23Pleurotus ostreatusAu10–30 nm/SphericalAntimicrobial, Anticancer[83]
24Aspergillus terreusAg16–57 nm/Spherical Antibacterial [84]
25Ganoderma sessiliformeAg~45 nm/Spherical Antibacterial, Antioxidant, Anticancer[85]
26Phenerochaete chrysosporiumAg34–90 nm/Spherical-OvalAntibacterial[86]
27Penicillium polonicumAg10–15 nm/Spherical Antibacterial[87]
28Candida glabrataAg2–15 nm/SphericalAntibacterial[53]
29Macrophomina phaseolinaAg/AgCl5–30 nm/SphericalAntibacterial[88]
30Aspergillus nidulansCoO20.29 nm/Spinel -[55]
31Rhodotorula glutinisAg15.45 nm/SphericalAntifungal, Dye degradation, Cytotoxicity [89]
32RhodotorulamucilaginosaAg13.70 nm/Spherical Antifungal, Dye degradation, Cytotoxicity[89]
33Cladosporium sp.Ag24 nm/Spherical Antioxidant, Antidiabetic, Anti-Alzheimer [90]
34Cladosporium cladosporioidesAu60 nm/RoundAntioxidant, Antibacterial[91]
35Nemania sp.Ag33.52 nm/SphericalAntibacterial[92]
36Penicillium chrysogenumPt5–40 nm/SphericalCytotoxicity[93]
37Aspergillus sp. Au2.5–6.7 nm/Spherical Nitrophenol reduction [94]
38Rhizopus stoloniferAg2.86 nm/Spherical -[52]
Algae/Cyanobacteria
39Sargassum wightiiZrO218 nm/SphericalAntibacterial [95]
40Neochloris oleoabundansAg40 nm/SphericalAntibacterial[96]
41Cystoseira baccataAu8.4 nm/SphericalAnticancer [97]
42Stephanopyxis turrisAu10–30 nm/Spherical -[98]
43Galaxaura elongateAu3.85–77 nm/Spherical-rods-triangular Antibacterial [99]
44Chlorella vulgarisPd5–20 nm nm/Spherical -[100]
45Enteromorpha compressaAg4–24 nm/Spherical Antimicrobial, Anticancer [101]
46Nostoc linckiaAg5–60 nm/Spherical Antibacterial[102]
47Nostoc spAg51–100 nm/SphericalSpherical [45]
48LeptolyngbyaAg5–50 nm/Spherical Antibacterial, Anticancer[103]
49Spyridia fusiformisAg5–50 nm/Spherical Antibacterial[104]
50Chlorella pyrenoidosaCdSe QD4–5 nmImatinib sensing[105]
52Sargassum ilicifoliumAl2O3 20 nm/Spherical -[59]
53Padina pavoniaAg49.58–86.37 nm/spherical-triangular-rectangle-polyhedral-hexagonal-[106]
53Spirulina platensisPd10–20 nm/Spherical Adsorbent [107]
54Chlorella pyrenoidosaTiO250 nm/Spherical Dye degradation[108]
Table 2. List of extracellular and intracellular bioreducing microbial enzymes and resulting nanoparticles.
Table 2. List of extracellular and intracellular bioreducing microbial enzymes and resulting nanoparticles.
Source of Extracellular Enzymes (Bacterial Species)Nature of OrganismMetals UsedShapeSize (nm)Temperature (°C)Reference
Desulfovibrio desulfuricansG−ive BacteriaPdRound5025[129]
Pyrobaculum islandicumG−ive RodsU, Tc, Cr, Co, MnRoundNA100[130]
Escherichia coliG−ive BacteriaCdTeRound2–3.237[131]
Escherichia coliG−ive BacteriaAuHexagonal, Triangle20–3037[132]
Bacillus licheniformisG+ive mesophilic bacteriaAgNA5037[133]
Shewanella speciesMarine BacteriaSeRound18130[134]
Ureibacillus thermosphaericusG+ive BacteriaAuNA50–7060–80[135]
Corynebacterium glutamicumG+ive BacteriaAgIrregular5–5030[136]
Rhodopseudomonas capsulatePhototrophic BacteriaAuRound10–2030[34]
Pseudomonas aeruginosaG−ive BacteriaAuNA15–3037[33]
Shewanella OneidensisFacultative BacteriaAuRound1230[137]
Fungi and Algae Species
Plectonema boryanum UTEX 485Filamentous FungiAuOctahedral10 nm–6 µm25[138]
Phaenerochaete chrysosporiumFungiAgPyramidal50–20037[139]
Aspergillus flavusFungiAgRound8.9225[140]
YeastFungiAu, AgPolygonal9–2530[131]
Fusarium oxysporumAscomycete fungusAlloy of Au–AgRound8–1425[117]
Sargassum wightiiMacro-algaeAuPlanar8–12NA[127]
Neurospora crassaBread moldAu–Ag, AuRound20–5028[50]
Verticillium sp.FungiAgRound25–3225[141]
Aspergillus fumigatusFungiAgRound5–2525[124]
Trichoderma virideFungiAgNA2–410–40[142]
Yarrowia lipolyticaFungiAuTriangles1530[143]
Source of Intracellular Enzyme (Bacterial Species)Nature of Microb.Metals usedShapeSize(nm)Temperature (°C)Reference
Shewanella algaeG−ive marine bacteriaPtNA525[144]
Enterobacter speciesAnaerobic G−ive BacilliHgRound2–530[145]
Bacillus cereusG+ive BacteriaAgRound4–537[146]
Brevibacterium caseiActinomycetales BacteriaAg, AuRoud10–5037[147]
Rhodococcus sp.ActinobacteriaAuRound8-12NA[148]
Fungi and Algae Species
Plectonema boryanumAlgaeAuCubic<10–2525–100[128]
Neurospora crassaBread moldAu–Ag, AuRound3228[50]
Verticillum luteoalbumAscomycota FungiAuNANA37[149]
Candida utilisFungusAuNANA25[149]
NA: Not available; G+ive: Gram-positive; G−ive: Gram-negative.

Share and Cite

MDPI and ACS Style

Ovais, M.; Khalil, A.T.; Ayaz, M.; Ahmad, I.; Nethi, S.K.; Mukherjee, S. Biosynthesis of Metal Nanoparticles via Microbial Enzymes: A Mechanistic Approach. Int. J. Mol. Sci. 2018, 19, 4100. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19124100

AMA Style

Ovais M, Khalil AT, Ayaz M, Ahmad I, Nethi SK, Mukherjee S. Biosynthesis of Metal Nanoparticles via Microbial Enzymes: A Mechanistic Approach. International Journal of Molecular Sciences. 2018; 19(12):4100. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19124100

Chicago/Turabian Style

Ovais, Muhammad, Ali Talha Khalil, Muhammad Ayaz, Irshad Ahmad, Susheel Kumar Nethi, and Sudip Mukherjee. 2018. "Biosynthesis of Metal Nanoparticles via Microbial Enzymes: A Mechanistic Approach" International Journal of Molecular Sciences 19, no. 12: 4100. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19124100

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