Next Article in Journal
Active Enhancement of Slow Light Based on Plasmon-Induced Transparency with Gain Materials
Previous Article in Journal
Sol-Gel Hydrothermal Synthesis and Visible Light Photocatalytic Degradation Performance of Fe/N Codoped TiO2 Catalysts
 
 
Correction published on 8 March 2019, see Materials 2019, 12(5), 806.
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Secondary Metabolites in the Green Synthesis of Metallic Nanoparticles

by
Gregory Marslin
1,†,
Karthik Siram
2,†,
Qaisar Maqbool
3,
Rajendran Kamalabai Selvakesavan
3,
Dariusz Kruszka
3,
Piotr Kachlicki
3 and
Gregory Franklin
3,*
1
Ratnam Institute of Pharmacy and Research, Nellore 524346, India
2
Department of Pharmaceutics, PSG College of Pharmacy, Coimbatore 641004, India
3
Institute of Plant Genetics of the Polish Academy of Sciences, Poznan 60479, Poland
*
Author to whom correspondence should be addressed.
Equal contribution.
Materials 2018, 11(6), 940; https://doi.org/10.3390/ma11060940
Submission received: 29 April 2018 / Revised: 25 May 2018 / Accepted: 30 May 2018 / Published: 3 June 2018

Abstract

:
The ability of organisms and organic compounds to reduce metal ions and stabilize them into nanoparticles (NPs) forms the basis of green synthesis. To date, synthesis of NPs from various metal ions using a diverse array of plant extracts has been reported. However, a clear understanding of the mechanism of green synthesis of NPs is lacking. Although most studies have neglected to analyze the green-synthesized NPs (GNPs) for the presence of compounds derived from the extract, several studies have demonstrated the conjugation of sugars, secondary metabolites, and proteins in these biogenic NPs. Despite several reports on the bioactivities (antimicrobial, antioxidant, cytotoxic, catalytic, etc.) of GNPs, only a handful of studies have compared these activities with their chemically synthesized counterparts. These comparisons have demonstrated that GNPs possess better bioactivities than NPs synthesized by other methods, which might be attributed to the presence of plant-derived compounds in these NPs. The ability of NPs to bind with organic compounds to form a stable complex has huge potential in the harvesting of precious molecules and for drug discovery, if harnessed meticulously. A thorough understanding of the mechanisms of green synthesis and high-throughput screening of stabilizing/capping agents on the physico-chemical properties of GNPs is warranted to realize the full potential of green nanotechnology.

1. Introduction

Nanoparticles (NPs) are particles that range 1–100 nm in size. Owing to the large surface area to volume ratio, NPs exhibit enhanced catalytic reactivity, biological activities, thermal conductivity, non-linear optical performance, and chemical steadiness in comparison to their bulk form. As a result of these properties, NPs are used in health, food, feed, space, chemical, cosmetic industries, and in agriculture [1,2,3]. NPs can be synthesized by several methods, including physical and chemical processes (Figure 1). However, as a result of the difficulties in scaling-up the physical processes and the usage of toxic synthetic chemicals that could be carried over by the NPs in chemical processes, alternative methods, such as green synthesis, have been developed. Although green chemistry methods have been practiced in agriculture, consumer products, and medicines for many years, the application of green chemistry to synthesize nanoparticles (NPs) is relatively recent.
Green synthesis is based on redox reaction, in which metal ions are reduced to stable NPs by the components of an organism or its extract. Although living organisms, such as algae, fungi, bacteria and plants, could synthesize NPs in vivo [4,5,6,7,8,9,10,11,12], plant extract-mediated, in vitro green synthesis of NPs has gained popularity due to its simplicity, low cost, eco-friendly nature, and easiness to scale-up [13,14]. Although plant extract-mediated green synthesis is a recent technology, a contrasting process has been used for the preparation of Bhasma (ash of metal) in Ayurveda, a traditional Indian medicine practiced for more than 2000 years. In the former, metal ions are reduced and stabilized as NPs by the components of the extract (bottom-up approach); by contrast, in the latter, NPs are produced by the calcination of metal sheets (top-down approach), which are macerated with medicinal plant extracts to obtain therapeutic potential.
Currently, the number of reports on green-synthesized nanoparticles (GNPs) is increasing exponentially. Extracts derived from diverse plant species, their organs, and isolated compounds are being successfully used in the green synthesis of NPs. In addition to being eco-friendly, NPs can be synthesized using agricultural and industrial waste to make the process more sustainable [15,16]. GNPs often possess better bioactivities [17,18,19,20] and catalytic characteristics compared to their counterparts, which are synthesized by other methods [21,22]; this is probably a result of the compounds attached to their surface [14]. The ability of plant secondary metabolites to bind or conjugate with NPs upon green synthesis could be exploited for the purification of compounds and drug discovery. Although the widespread occurrence of phenolic compounds in the plant kingdom could explain the ability of plant extracts to reduce metal ions, the mechanism of green synthesis is not fully understood. In this article, the current status and potential applications of the green synthesis of NPs with special emphasize on secondary metabolites participating in this process are discussed.

2. Mechanism of Green Synthesis of NPs by Plants

A process known as bioaccumulation, which provides plants with the ability to detoxify metal ions, can explain the mechanism of NP synthesis by plants. When plants absorb metal ions at a rate faster than that of their removal by catabolism, the excess metal ions accumulate in the plant tissues. The presence of metals at toxic levels can induce excessive reactive oxygen species (ROS) production in the cells and damage cellular macromolecules resulting in serious morphological, metabolic, and physiological irregularities in plants. To counteract the metal toxicity, plants are equipped with sophisticated chelation mechanisms to detoxify metals [23]. Cysteine-rich oligopeptides, phytochelatins, and low molecular weight proteins named metallothioneins can form complexes with metals and eventually remove the metal ions via vacuolar sequestration [24].
To maintain the ROS homeostasis upon metal toxicity, cellular enzymatic antioxidant systems are activated [25]. In addition to the enzymatic antioxidant mechanisms, plant secondary metabolites, such as phenolic compounds, can assist the cells in the maintenance of ROS homeostasis [26,27]. Induction of plant secondary metabolism in response to metal stress has been frequently reported. Stimulation of key enzymes of the phenylpropanoid pathway, such as phenylalanine ammonia-lyase (PAL) and chalcone synthase (CHS), has been observed in plants upon exposure to Cu, Cd, Al, Pb, and Ni [28]. Pteris vittata, a hyperaccumulator, produces high concentrations of chlorogenic acid derivatives and a-type procyanidin upon Cu stress [29]. Similarly, elevation of phenolic content in maize exposed to Al [30] and Phaseolus vulgaris exposed to Cu [31] has been reported. The enhanced biosynthesis of phenolic compounds by the plants under metal stress might be due to the high tendency of these compounds to chelate metals [32]. Several flavonoids are capable of chelating metal ions by forming stable complex through their multiple hydroxyl (–OH) groups and the carbonyl moiety. For instance, quercetin, a flavonoid that possesses three potential bidentate binding sites, namely α-hydroxy-carbonyl, β-hydroxy-carbonyl or catechol having two –OH groups in ortho positions, can form stable complex with metal cations such as Mo(VI), Fe(II)/Fe(III), Cu(II), Zn(II), Al(III), Tb(III), Pb(II), Co(II) by quercetin has also been reported [33].
When serving as antioxidants in a concentration-dependent manner to scavenge ROS, phenolic compounds are toxic to the cell due to their high chemical reactivity and protein-denaturing properties [34]. Hence, plants transfer these compounds to compartments with lower biosynthetic activity, such as the extracellular space and the vacuole. Flavonoids are synthesized in the cytosol and transported into the vacuole for storage [35]. The multidrug and toxic efflux transporter TT12, which is localized in the vacuolar membrane, mediates flavonoid transport by an H+-antiport mechanism into the vacuole [36]. Thus, the accumulation of metal ions and antioxidant phenolic compounds in the same cellular compartment might facilitate the formation of NPs (Figure 2).
Many plant species have the ability to synthesize and store NPs in their cells. For example, Medicago sativa (alfalfa) plants grown on an AuCl4 rich environment accumulated 4 nm size Au NPs in their tissues [7]. Similarly, when grown on an Ag ion rich medium, M. sativa accumulated Ag NPs [8]. M. sativa plants could accumulate Ag NPs of 50 nm size to about 13.6% of their fresh weight when grown on a solution containing AgNO3 [9]. Under the same conditions, Brassica juncea accumulated similar NPs to about 12.4% of fresh weight [9]. The uptake of AgNO3, sodium dithiosulfatoargentate [Na3Ag(S2O3)2], and diamine silver nitrate [Ag(NH3)2NO3] by hydroponically grown B. juncea and the conversion of these salts to silver metal NPs has been quantified [10]. Brassica napus, which was cultured on medium containing AgNO3, accumulated Ag NPs in the regenerated leaves [11]. Spatial distribution of Ag NPs in the chloroplast, cytoplasmic spaces, vacuolar, and nucleolar regions was confirmed by transmission electron microscopic analysis of tissues obtained from Vigna radiata plants exposed to AgNO3 [12]. Interestingly, the ability of Phragmites australis and Iris pseudacorus to transform Cu ions into Cu NPs in and around their roots has been revealed when grown in Cu-contaminated soil in the natural environment [37].

3. Mechanism of Green Synthesis of NPs by Plant Extracts

A similar mechanism as discussed above could be operating during green synthesis of NPs by plant extracts in vitro. Figure 3 schematically describes the formation of metallic NPs from the corresponding metal ions. When metallic salt dissociates into cation and anion, cations will be saturated to form hydroxyl complexes. Immediately after the supersaturation of hydroxyl complexes, crystillite growth of metal with oxygen species starts to originate. This results in the formation of crystalline planes with different energy levels. Heat plays a key role in providing energy to the reaction system. The process continues until activation of the capping agent from the plant extracts, which will ultimately arrest the growth of high-energy atomic growth planes. This results in the formation of specific type NPs. Generally, during the synthesis, the reducing agents donate electrons to the metal ions and convert them to NPs. These NPs exist at a high-surface energy state and tend to convert to their low-surface energy conformations by aggregating against each other. Thus, the presence of higher amounts of reducing agents and stabilizing agents prevents the aggregation of nanoparticles and promotes production of smaller NPs. Additionally, proteins can trap metal ions on their surface and convert them to their corresponding nuclei, which could further aggregate and, consequently, form NPs [38]. Amine groups of proteins, hydroxyl and carboxyl groups of polyphenols and amino acids, hydroxyl groups of polysaccharides, and carboxyl groups of organic acids chelate metal ions and suppress the superoxide-driven Fenton reaction (which is believed to be the most important source of ROS), catalyzing the formation of metallic NPs.
Although it is essential to form a protein-metal ion complex for the vacuolar sequestration of metal ions during in vivo accumulation of NPs, the role of proteins upon in vitro green synthesis is not clear. Interestingly, plant extracts possess the capacity to reduce metal ions and produce NPs even after boiling [39,40,41,42]. Although boiling could denature proteins by altering their secondary and tertiary structures, the peptide bonds of the primary structure between the amino acids are left intact. Because all structural levels of the protein determine its function, the denatured protein can no longer be fully functional. It has been stated that the protein can bind to Au NPs, either through free amino groups or cysteine residues; the surface-bound protein lead to the stabilization of the NPs [43].

4. Secondary Metabolites in Plant Extract-Mediated Green Synthesis of NPs

Synthesis of NPs using plant extracts has been reported in several plant species (Table 1). A wide range of molecules, ranging from proteins to various low molecular weight compounds such as terpenoids, alkaloids, amino acids, alcoholic compounds, polyphenols (catechin, flavones, taxifolin, procyanidins of various chain lengths formed by catechin and epicatechin units, and phenolic acids), glutathiones, polysaccharides, antioxidants, organic acids (ascorbic, oxalic, malic, tartaric, and protocatechuic acid), quinones etc., have been reported to play a role in the green synthesis of NPs. The participation of sugars, terpenoids, polyphenols, alkaloids, phenolic acids, and proteins in the reduction of metal ions into NPs and in supporting their subsequent stability has also been postulated [38]. As seen in Table 1, flavonoids have been the compounds most commonly reported/ predicted to participate in the green synthesis.
Although researchers face significant challenges in identifying the elements participating in the green synthesis of NPs, Fourier-transform infrared spectroscopy (FT-IR) analyses have been used to obtain clues on the biomolecules possibly involved in the reduction of the metal ions and capping. A FT-IR analysis of Ag NPs synthesized using Acacia mearnsii bark extract suggested that compounds with hydroxyl, alkyne, carboxyl, and amide groups of the monoterpenoids, sesquiterpenes, and phytols might have participated in these processes [140]. Similar analysis of Au NPs green-synthesized using Suaeda monoica leaf extract indicated that the biomolecules with carboxyl, amine, and hydroxyl functional groups were involved in the reduction of Au ions [91]. Isolated flavonoids, terpenoids, chlorogenic acid, etc. have been successfully used in the green synthesis of NPs. Highly monodispersed (18.24 nm) spherical Au NPs could be synthesized using kaempferol as the in situ reductant and stabilizer [141]. The ability of terpenoid fractions isolated from Andrographis paniculata leaves for the green synthesis of ZnO NPs has also been demonstrated and confirmed by the presence of C=O functional group in the NPs [142]. Au NPs were synthesized using chlorogenic acid as reductant, and the corresponding FT-IR spectra indicated that an –OH functional group was likely involved in the synthesis [143].

5. Flavonoids are the Major Contributors of Green Synthesis of NPs

High performance liquid chromatography (HPLC) analysis of Ag NPs green synthesized using Withania somnifera leaf extract revealed that several phenolic compounds present in the extract were selectively trapped in the Ag NPs [14]. On the basis of their characteristic UV-vis spectra and further confirmation by co-elution with pure standards, these compounds were identified as catechin, p-coumaric acid, and luteolin-7-glucoside. In addition, a major compound that appeared in the NPs was not found in the original extract, which might be a derivative resulting from the interaction of some withanolide derivatives with Ag ions.
Jain and Mehata [58] were able to green synthesize Ag NPs using both leaf extract of Ocimum sanctum and a flavonoid (quercetin) present in the extract separately. Their results revealed that both Ag NPs showed similar optical, morphological, and antibacterial characteristics, demonstrating that quercetin was responsible for the NP synthesis. Other flavonoids such as hesperidin, naringin, and diosmin have also been shown to be involved in the green synthesis of Ag NPs, with the size and shape distribution varied between the compounds. These authors concluded that the –OH group was involved in the reduction of Ag ions into Ag NPs [144].
Flavonoids are a family of natural polyphenolic compounds that include flavone, flavonol, flavanone, flavanonol, and isoflavone derivatives. The skeleton of flavonoids consists of two phenyl rings (A and B), connected by an oxygenated heterocycle ring C, and is hydroxylated in several positions. These compounds have important roles in plants because they participate in the response to biotic and abiotic stresses [145]. Much attention has been paid to the chelating properties and antioxidative activities of natural flavonoids, as they are important for plant physiology and desirable for human health. A flavonoid-metal complex may be a target for ROS and free radicals. However, this complex can be a catalytic center for Fenton reaction and the ligand moiety may act as an acceptor for hydroxyl radical. It has been reported that the antioxidant activity of flavonoid-metal complexes is higher than that of free ligands [146,147].
The number of hydroxyl groups and the structure of flavonoids are important for metal-binding activity (Figure 4). Simple aglycones, such as chrysin, apigenin and genistein, may accept metal ion in one coordination pocket between the 4-carbonyl group and 5-hydroxyl group. Divalent metal ions Cu2+, Co2+, and Ni2+ are bound by two chrysin ligands [148]. Moreover, genistein and biochanin a chelate Cu2+ and Fe3+ with a 1:2 (metal:ligand) stoichiometry. Complexes of isoflavones with Cu have higher antioxidant potential than the free ligands, as revealed via 2,2-diphenyl-1-picrylhydrazyl (DPPH) reduction assays. Fe chelates were shown to have prooxidant potential [149]. In another report, apigenin was found to bind Al3+ and Fe2+ with a 1:3 (metal:ligand) stoichiometry in a dioxan water solution. In this case, the 4′-hydroxyl group was the coordination site [150]. Apigenin, naringenin, and hesperitin can chelate Cu ions and create coordination pocket between 4-carbonyl and 5-hydroxyl groups. Such complexes have a strong DNA-binding properties and cytotoxicity [151].
Bicalein is a highly bioactive flavonoid that is characterized by three possible bidentate binding sites (4-carbonyl and three hydroxyl groups at carbons number 5, 6, 7). In this case, metal:bicalein complexes can be created by using only one binding site. Either hydroxyls at 5- and 6- carbon atoms or 6-hydroxyl-7-hydroxyl are potentially the binding sites of Fe2+ and Fe3+ ions, with a 1:1 and 1:2 stoichiometry. The Fe-bicalein complexes have high antioxidant properties due to inhibition of the Fenton reaction [152,153]. Similarly, two potential bidentate binding sites are present in the luteolin structure (5-hydroxyl-4-carbonyl and 3′4′-hydroxyl groups of the ring B) and both sites can bind Al3+ ions in the molar ratio 2:1 (metal:ligand). The separation of the coordination pocket is important for increasing the chelating properties [154]. Quercetin has three potential bidendate binding places. The stable complexes of quercetin were reported for a large number of metal ions, such as Fe2+, Fe3+, Cu2+, Zn2+, Co2+, Pb2+, Al3+ [155]. 3-hydroxyl and 4-carbonyl groups of quercetin can chelate Fe2+ with a 1:2 (metal:ligand) stoichiometry. Quercetin-iron complex is characterised by high free radical scavenging, DNA binding and antibacterial activities [156].
Cherrak, et al. [33] reported on the chelating properties of quercetin, O-methylated quercetins, rutin, and catechin. The most stable complexes with Fe3+, Zn2+, and Cu2+ were observed for catechin, quercetin, and rutin compared to O-methylated analogues. Moreover, spectrophotometric studies of O-methylated quercetins showed the binding sequence with the iron ion: 3′4′-OH > 3-OH >> 5-OH [33]. In summary, the modification of free hydroxyl groups in the flavonoids structure causes changes to both chelating and antioxidant properties. O-methylated flavonoids are weaker chelators than the free ligands. The aglycones have stronger antioxidant activities in comparison to the glycosides, e.g., baicalein-baicalin or luteolin-luteolin-4′-O-glucoside [147].

6. Green-Synthesized NPs are Highly Bioactive and Biocompatible

As discussed above, GNPs are often capped with secondary metabolites [14]. In addition to providing stability to the GNPs as capping agents or stabilizers, the presence of these compounds might enhance the bioactivities of these NPs (Table 2). The scavenging activities of Ag NPs prepared using three different natural polyphenols, epigallocatechin-3-gallate (EGCG), resveratrol (RSV), and fisetin, were highly correlated with their secondary metabolite content [157]. Green-synthesized Ag NPs using white rot fungi Pycnoporus broth showed better antimicrobial activity against the pathogenic bacteria in comparison to the chemically synthesized ones [158]. Microbiological tests performed using varying concentrations of green (aloe extract) and chemical ZnO NPs showed that green ZnO nanoparticles had enhanced biocidal activity against various pathogens compared to the chemical ZnO NPs [17]. It was observed that the green-synthesized Ag NPs using Salvadora persica root extract exhibited comparable or better antibacterial activities than the chemically obtained Ag NPs [18]. A bioactivity comparison of Ni NPs prepared via the chemical and green routes (Desmodium gangeticum aqueous root extract) suggested that NPs prepared by the green route had better antioxidant and antibacterial activity, without any toxicity towards epithelial cell line and Wistar rats [19].
Interestingly, green-synthesized ZnO NPs, which had been stabilized by plant metabolites, varied in their anti-diabetic activity based on their size in streptozotocin (STZ)-induced diabetic mice [20]. Au NPs synthesized using methanolic extract of Azolla microphylla showed excellent antioxidant activity [159]. Au NPs produced from Hypoxis hemerocallidea exhibited antibacterial activity against Staphylococcus aureus, Staphylococcus epidermidis, Escherichia coli, and Pseudomonas aeruginosa, whereas Au NPs produced from Galenia africana only exhibited antibacterial activity against P. aeruginosa [94]. The Fe3O4 NPs synthesized using agro-waste extracts exhibited higher removal (>90%) of antibiotics than Fe3O4 NPs synthesized by a conventional method [15]. The Cd NPs synthesized using marigold petal extracts showed better larvicidal activity against mosquito larvae compared to the Cd NPs synthesized using rose petal extracts [120]. An Azadirachta indica extract-mediated reduction of Ag ions resulted in the formation of different sizes of NPs (4.74 nm, 8.17 nm, 14.23 nm, and 18.98) when the aqueous extract was not boiled [160], whereas the NPs prepared using boiled extract were of an average size of 34 nm [46]. Moreover, although the latter showed antibacterial activities against both gram positive and gram-negative bacteria, the former did not show activity against gram-positive bacteria.

7. Potential Applications of Green-Synthesized NPs

In addition to the several bioactivities listed above, GNPs also have been found to possess several industrial applications due to the presence of plant compounds (Table 3). For instance, enzymes are important biocatalysts in modern biotechnology but are highly unstable in nature. The thermal, pH, and storage stability of α-amylase could be improved by immobilization with naringin-functionalized magnetite nanoparticles [210]. Ag NPs synthesized using O. sanctum and Chenopodium aristatum showed good catalytic activity in degradation of 4-nitrophenol [21,22]. Similarly, Ag NPs synthesized using Thuja occidentalis extract could be used as a soil conditioner and plant growth promoter [211]. Cu NPs synthesized using Lawsonia inermis extract could be used to prepare electrical-conducting nanobiocomposites [123]. CuO NPs green-synthesized using Ocimum tenuiflorum extract could be used as non-enzymatic glucose biosensor [212].
The ability of plant secondary metabolites to chelate metal ions in the production of stable complexes and their potential to conjugate with NPs has opened a new window for NP use in harvesting these natural products. The ability to form a NP-secondary metabolite complex upon green synthesis provides us an opportunity to establish a relationship between NP type and compound classes, which would have impact on the nanoharvesting of compounds from live plants or tissues. During nanoharvesting, the metabolites are adsorbed onto the NPs and extrudes from plant cells to the medium and the metabolites can be separated through elution and magnetization [217]. Nanoharvesting eliminates the use of organic solvents, allows for the spectral identification of the isolated compounds, and provides new avenues for the use of nanomaterials for coupled isolation and testing of bioactive properties of plant-synthesized compounds.
Kurepa, et al. [213] showed that TiO2 NPs enter Arabidopsis thaliana plant cells, conjugate enediol and catechol group-rich flavonoids in situ, and exit plant cells as flavonoid-nanoparticle conjugates. The compound adsorption capacities of NPs could be further improved by functionalization [217]. For instance, adsorption capacity of SiO2 NPs towards quercetin was higher upon TiO2 functionalization in comparison to non-functionalized and decyl group functionalized SiO2 NPs due to possible binding of quercetin to the metal oxide [218]. This adsorption capacity increased linearly with surface coverage of TiO2, emphasizing the correlation between functional surface and quercetin adsorption. Similar to in vivo nanotrapping, in vitro green synthesis of NPs using plant extracts can be extended further to develop high throughput tools to extract specific classes of compounds from crude extracts (Figure 5).
This NPs-secondary metabolite conjugation property can also be used in different fields of industrial biotechnology. Nanoparticle-mediated delivery of medicinally important flavonoids and other biomolecules will increase their therapeutic efficacy. As a potential drug delivery system, hesperitin-conjugated gold nanoparticles enhanced the treatment of hepatocellular carcinoma by minimizing the side effects and reduced the dose of chemotherapy drug [222]. Recently, several reports have addressed the potential risk of NPs to both human health and the environment, which necessitates a method to detect them in food and other samples. A green, facile, and rapid method using a flavonoid-assisted method was standardized to extract and detect the TiO2 NPs in food samples [214]. It was also applicable in identifying flavonoid traces in biological samples, such as urine and blood [215].

8. Conclusions

Although the ability of extracts from diverse plant species to synthesize NPs could be explained by the widespread occurrence of polyphenolic compounds in the plant kingdom, a precise understanding of the green synthesis process is needed to realize the full potential of this process in medical and industrial applications. In spite of the facile synthesis of NPs via a green method, obtaining homogeneously dispersed NPs is a huge challenge, as several parameters including temperature, pH of the system, nature of the capping agent, concentration of active compounds, etc. might play vital roles in defining the size and morphology [225]. Au NPs synthesized using Cinnamomum zeylanicum leaf broth differed in their shape, which was based on the concentration of the extract [39]. At lower concentrations of the extract, the formation of prism-shaped NPs dominated and, at higher concentrations however, mostly spherical NPs were formed [39]. Hence, reducing, capping, or stabilizing agents participating in the green synthesis need to be further analyzed to specify the NP structural relationship.
A high-throughput analysis of plant extracts with diverse metal ions would provide clues as to whether there is any correlation between the specific compounds and the NPs generated. This would have a huge potential in the trapping of compounds and proteins using metal ions or NPs, which are difficult or expensive to purify by other means. NPs green-synthesized using medicinal plant extracts should be tested for various bioactivities in comparison with their chemically or physically synthesized counterparts to understand whether the bioactivities observed could be attributed to the presence of capping agents in the NPs. Considering that NPs bioactivities also differ between their size shape and zeta potential, similar NPs needs to be compared. For example, in spite of excellent antibacterial activities reported against antibiotic resistant strains, it is not clear whether this is a result of the NPs, the compounds attached to them, both, or, conversely, a result of the other compounds present in the extract; such comparisons require further study.
In the light of above discussion, it has been extensively reported that GNPs surface chemistry is helpful in the advancement of biomedical applications. However, most studies have shown that GNPs are subjected to calcinations using a blast furnace at ultra high temperatures [225,226,227] to attain the highest level of crystalline morphology before they can be used. This will surely provide a pure-phase crystalline structure, but the surface-attached phytochemicals as capping agents will be decomposed. Accordingly, to achieve GNPs with functionalized surfaces and sticky capping agents for advanced biological applications, we must improve the synthesis modes.

Funding

This research was funded by Narodowym Centrum Nauki (NCN), Poland grant number UMO-2016/21/B/NZ9/01980 and the APC was funded by European Union’s 7th Framework Programme for research, technological development, and demonstration under grant agreement No. 621321 and co-financed by funds allocated for education through project No. W26/7.PR/2015 [GA 3413/7.PR/2015/2] for the years 2015–2019.

Acknowledgments

Qaisar Maqbool and Dariusz Kruszka are supported by PhD grants from NCN projects UMO-2016/23/B/NZ9/02677 and UMO-2016/21/B/NZ9/01980 respectively. Rajendran Kamalabai Selvakesavan is a recipient of postdoc grant from UMO-2016/21/B/NZ9/01980.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Agarwal, H.; Venkat Kumar, S.; Rajeshkumar, S. A review on green synthesis of zinc oxide nanoparticles—An eco-friendly approach. Resour. Effic. Technol. 2017, 3, 406–413. [Google Scholar] [CrossRef]
  2. Prasad, R.; Bhattacharyya, A.; Nguyen, Q.D. Nanotechnology in sustainable agriculture: Recent developments, challenges, and perspectives. Front. Microbiol. 2017, 8, 1014. [Google Scholar] [CrossRef] [PubMed]
  3. Dimkpa, C.O.; Bindraban, P.S. Nanofertilizers: New Products for the Industry? J. Agric. Food Chem. 2017. [Google Scholar] [CrossRef] [PubMed]
  4. Fayaz, A.M.; Balaji, K.; Girilal, M.; Yadav, R.; Kalaichelvan, P.T.; Venketesan, R. Biogenic synthesis of silver nanoparticles and their synergistic effect with antibiotics: A study against gram-positive and gram-negative bacteria. Nanomed. Nanotechnol. Biol. Med. 2010, 6, 103–109. [Google Scholar] [CrossRef] [PubMed]
  5. Gómez-Graña, S.; Perez-Ameneiro, M.; Vecino, X.; Pastoriza-Santos, I.; Perez-Juste, J.; Cruz, J.; Moldes, A. Biogenic synthesis of metal nanoparticles using a biosurfactant extracted from corn and their antimicrobial properties. Nanomaterials 2017, 7, 139. [Google Scholar] [CrossRef] [PubMed]
  6. Narayanan, K.B.; Sakthivel, N. Biological synthesis of metal nanoparticles by microbes. Adv. Colloid Interface Sci. 2010, 156, 1–13. [Google Scholar] [CrossRef] [PubMed]
  7. Gardea-Torresdey, J.L.; Parsons, J.G.; Gomez, E.; Peralta-Videa, J.; Troiani, H.E.; Santiago, P.; Yacaman, M.J. Formation and growth of Au nanoparticles inside live alfalfa plants. Nano Lett. 2002, 2, 397–401. [Google Scholar] [CrossRef]
  8. Gardea-Torresdey, J.L.; Gomez, E.; Peralta-Videa, J.R.; Parsons, J.G.; Troiani, H.; Jose-Yacaman, M. Alfalfa sprouts:  A natural source for the synthesis of silver nanoparticles. Langmuir 2003, 19, 1357–1361. [Google Scholar] [CrossRef]
  9. Harris, A.T.; Bali, R. On the formation and extent of uptake of silver nanoparticles by live plants. J. Nanopart. Res. 2008, 10, 691–695. [Google Scholar] [CrossRef]
  10. Haverkamp, R.G.; Marshall, A.T. The mechanism of metal nanoparticle formation in plants: Limits on accumulation. J. Nanopart. Res. 2009, 11, 1453–1463. [Google Scholar] [CrossRef]
  11. Roh, K.-H.; Kwak, B.-K.; Kim, J.-B.; Lee, K.-R.; Kim, H.-U.; Kim, S.-H. The influence of silver thiosulfate and thidiazuron on shoot regeneration from cotyledon explants of Brassica napus. J. Plant Biotechnol. 2012, 39, 133–139. [Google Scholar] [CrossRef]
  12. Kumari, R.; Singh, J.S.; Singh, D.P. Biogenic synthesis and spatial distribution of silver nanoparticles in the legume mungbean plant (Vigna radiata L.). Plant Physiol. Biochem. 2017, 110, 158–166. [Google Scholar] [CrossRef] [PubMed]
  13. Vineet, K.; Kumar, Y.S. Plant-mediated synthesis of silver and gold nanoparticles and their applications. J. Chem. Technol. Biotechnol. 2009, 84, 151–157. [Google Scholar]
  14. Marslin, G.; Selvakesavan, R.K.; Franklin, G.; Sarmento, B.; Dias, A.C. Antimicrobial activity of cream incorporated with silver nanoparticles biosynthesized from Withania somnifera. Int. J. Nanomed. 2015, 10, 5955–5963. [Google Scholar]
  15. Stan, M.; Lung, I.; Soran, M.-L.; Leostean, C.; Popa, A.; Stefan, M.; Lazar, M.D.; Opris, O.; Silipas, T.-D.; Porav, A.S. Removal of antibiotics from aqueous solutions by green synthesized magnetite nanoparticles with selected agro-waste extracts. Proc. Saf. Environ. Prot. 2017, 107, 357–372. [Google Scholar] [CrossRef]
  16. Kanchi, S.; Kumar, G.; Lo, A.-Y.; Tseng, C.-M.; Chen, S.-K.; Lin, C.-Y.; Chin, T.-S. Exploitation of de-oiled jatropha waste for gold nanoparticles synthesis: A green approach. Arab. J. Chem. 2018, 11, 247–255. [Google Scholar] [CrossRef]
  17. Gunalan, S.; Sivaraj, R.; Rajendran, V. Green synthesized ZnO nanoparticles against bacterial and fungal pathogens. Prog. Nat. Sci. Mater. Int. 2012, 22, 693–700. [Google Scholar] [CrossRef]
  18. Shaik, M.; Albalawi, G.; Khan, S.; Khan, M.; Adil, S.; Kuniyil, M.; Al-Warthan, A.; Siddiqui, M.; Alkhathlan, H.; Khan, M. “Miswak” based green synthesis of silver nanoparticles: Evaluation and comparison of their microbicidal activities with the chemical synthesis. Molecules 2016, 21, 1478. [Google Scholar] [CrossRef] [PubMed]
  19. Sudhasree, S.; Shakila Banu, A.; Brindha, P.; Kurian, G.A. Synthesis of nickel nanoparticles by chemical and green route and their comparison in respect to biological effect and toxicity. Toxicol. Environ. Chem. 2014, 96, 743–754. [Google Scholar] [CrossRef]
  20. Bala, N.; Saha, S.; Chakraborty, M.; Maiti, M.; Das, S.; Basu, R.; Nandy, P. Green synthesis of zinc oxide nanoparticles using Hibiscus subdariffa leaf extract: Effect of temperature on synthesis, anti-bacterial activity and anti-diabetic activity. RSC Adv. 2015, 5, 4993–5003. [Google Scholar] [CrossRef]
  21. Singh, J.; Mehta, A.; Rawat, M.; Basu, S. Green synthesis of silver nanoparticles using sun dried tulsi leaves and its catalytic application for 4-Nitrophenol reduction. J. Environ. Chem. Eng. 2018, 6, 1468–1474. [Google Scholar] [CrossRef]
  22. Yuan, C.-G.; Huo, C.; Gui, B.; Liu, P.; Zhang, C. Green synthesis of silver nanoparticles using Chenopodium aristatum L. stem extract and their catalytic/antibacterial activities. J. Clust. Sci. 2017, 28, 1319–1333. [Google Scholar] [CrossRef]
  23. Anjum, N.A.; Hasanuzzaman, M.; Hossain, M.A.; Thangavel, P.; Roychoudhury, A.; Gill, S.S.; Rodrigo, M.A.M.; Adam, V.; Fujita, M.; Kizek, R.; et al. Jacks of metal/metalloid chelation trade in plants—An overview. Front. Plant Sci. 2015, 6, 192. [Google Scholar] [CrossRef] [PubMed]
  24. Hasan, M.K.; Cheng, Y.; Kanwar, M.K.; Chu, X.-Y.; Ahammed, G.J.; Qi, Z.-Y. Responses of Plant Proteins to Heavy Metal Stress—A Review. Front. Plant Sci. 2017, 8, 1492. [Google Scholar] [CrossRef] [PubMed]
  25. Simão, B.N.; Cristiano, S.; Alexandra, S.; Viviana, M.; Manuel, A.; Hernâni, G.; Fernanda, F. An efficient antioxidant system and heavy metal exclusion from leaves make Solanum cheesmaniae more tolerant to Cu than its cultivated counterpart. Food Energy Secur. 2017, 6, 123–133. [Google Scholar]
  26. Marslin, G.; Sheeba, C.J.; Franklin, G. Nanoparticles alter secondary metabolism in plants via ROS burst. Front. Plant Sci. 2017, 8, 832. [Google Scholar] [CrossRef] [PubMed]
  27. Franklin, G.; Conceição, L.F.R.; Kombrink, E.; Dias, A.C.P. Xanthone biosynthesis in Hypericum perforatum cells provides antioxidant and antimicrobial protection upon biotic stress. Phytochemistry 2009, 70, 60–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Singh, S.; Parihar, P.; Singh, R.; Singh, V.P.; Prasad, S.M. Heavy metal tolerance in plants: Role of transcriptomics, proteomics, metabolomics, and ionomics. Front. Plant Sci. 2016, 6, 1143. [Google Scholar] [CrossRef] [PubMed]
  29. Pham, H.N.; Michalet, S.; Bodillis, J.; Nguyen, T.D.; Nguyen, T.K.O.; Le, T.P.Q.; Haddad, M.; Nazaret, S.; Dijoux-Franca, M.-G. Impact of metal stress on the production of secondary metabolites in Pteris vittata L. and associated rhizosphere bacterial communities. Environ. Sci. Pollut. Res. 2017, 24, 16735–16750. [Google Scholar] [CrossRef] [PubMed]
  30. Winkel-Shirley, B. Biosynthesis of flavonoids and effects of stress. Curr. Opin. Plant Biol. 2002, 5, 218–223. [Google Scholar] [CrossRef]
  31. Dı́az, J.; Bernal, A.; Pomar, F.; Merino, F. Induction of shikimate dehydrogenase and peroxidase in pepper (Capsicum annuum L.) seedlings in response to copper stress and its relation to lignification. Plant Sci. 2001, 161, 179–188. [Google Scholar] [CrossRef]
  32. Jun, M.; Fu, H.Y.; Hong, J.; Wan, X.; Yang, C.S.; Ho, C.T. Comparison of antioxidant activities of isoflavones from kudzu root (Pueraria lobata Ohwi). J. Food Sci. 2003, 68, 2117–2122. [Google Scholar] [CrossRef]
  33. Cherrak, S.A.; Mokhtari-Soulimane, N.; Berroukeche, F.; Bensenane, B.; Cherbonnel, A.; Merzouk, H.; Elhabiri, M. In vitro antioxidant versus metal ion chelating properties of flavonoids: A structure-activity investigation. PLoS ONE 2016, 11, e0165575. [Google Scholar] [CrossRef] [PubMed]
  34. Matile, P. Das toxische Kompartiment der Pflanzenzelle. Naturwissenschaften 1984, 71, 18–24. [Google Scholar] [CrossRef]
  35. Zhao, J. Flavonoid transport mechanisms: How to go, and with whom. Trends Plant Sci. 2015, 20, 576–585. [Google Scholar] [CrossRef] [PubMed]
  36. Marinova, K.; Pourcel, L.; Weder, B.; Schwarz, M.; Barron, D.; Routaboul, J.-M.; Debeaujon, I.; Klein, M. The Arabidopsis MATE transporter TT12 acts as a vacuolar flavonoid/H+-antiporter active in proanthocyanidin-accumulating cells of the seed coat. Plant Cell 2007, 19, 2023. [Google Scholar] [CrossRef] [PubMed]
  37. Manceau, A.; Nagy, K.L.; Marcus, M.A.; Lanson, M.; Geoffroy, N.; Jacquet, T.; Kirpichtchikova, T. Formation of metallic copper nanoparticles at the soil−root interface. Environ. Sci. Technol. 2008, 42, 1766–1772. [Google Scholar] [CrossRef] [PubMed]
  38. Makarov, V.V.; Love, A.J.; Sinitsyna, O.V.; Makarova, S.S.; Yaminsky, I.V.; Taliansky, M.E.; Kalinina, N.O. “Green” nanotechnologies: Synthesis of metal nanoparticles dsing plants. Acta Nat. 2014, 6, 35–44. [Google Scholar]
  39. Smitha, S.L.; Philip, D.; Gopchandran, K.G. Green synthesis of gold nanoparticles using Cinnamomum zeylanicum leaf broth. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2009, 74, 735–739. [Google Scholar] [CrossRef] [PubMed]
  40. Khalil, M.M.H.; Ismail, E.H.; El-Magdoub, F. Biosynthesis of Au nanoparticles using olive leaf extract: 1st Nano Updates. Arab. J. Chem. 2012, 5, 431–437. [Google Scholar] [CrossRef]
  41. MubarakAli, D.; Thajuddin, N.; Jeganathan, K.; Gunasekaran, M. Plant extract mediated synthesis of silver and gold nanoparticles and its antibacterial activity against clinically isolated pathogens. Colloids Surf. B Biointerfaces 2011, 85, 360–365. [Google Scholar] [CrossRef] [PubMed]
  42. Suman, T.Y.; Radhika Rajasree, S.R.; Ramkumar, R.; Rajthilak, C.; Perumal, P. The Green synthesis of gold nanoparticles using an aqueous root extract of Morinda citrifolia L. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2014, 118, 11–16. [Google Scholar] [CrossRef] [PubMed]
  43. Gole, A.; Dash, C.; Ramakrishnan, V.; Sainkar, S.R.; Mandale, A.B.; Rao, M.; Sastry, M. Pepsin-gold colloid conjugates: Preparation, characterization, and enzymatic activity. Langmuir 2001, 17, 1674–1679. [Google Scholar] [CrossRef]
  44. Vanaja, M.; Annadurai, G. Coleus aromaticus leaf extract mediated synthesis of silver nanoparticles and its bactericidal activity. Appl. Nanosci. 2013, 3, 217–223. [Google Scholar] [CrossRef]
  45. Prasad, R.; Swamy, V.S. Antibacterial activity of silver nanoparticles synthesized by bark extract of Syzygium cumini. J. Nanoparticles 2013, 2013, 6. [Google Scholar] [CrossRef]
  46. Ahmed, S.; Saifullah; Ahmad, M.; Swami, B.L.; Ikram, S. Green synthesis of silver nanoparticles using Azadirachta indica aqueous leaf extract. J. Radiat. Res. Appl. Sci. 2016, 9, 1–7. [Google Scholar] [CrossRef]
  47. Kaviya, S.; Santhanalakshmi, J.; Viswanathan, B.; Muthumary, J.; Srinivasan, K. Biosynthesis of silver nanoparticles using Citrus sinensis peel extract and its antibacterial activity. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2011, 79, 594–598. [Google Scholar] [CrossRef] [PubMed]
  48. Velmurugan, P.; Anbalagan, K.; Manosathyadevan, M.; Lee, K.J.; Cho, M.; Lee, S.M.; Park, J.H.; Oh, S.G.; Bang, K.S.; Oh, B.T. Green synthesis of silver and gold nanoparticles using Zingiber officinale root extract and antibacterial activity of silver nanoparticles against food pathogens. Bioprocess Biosyst. Eng. 2014, 37, 1935–1943. [Google Scholar] [CrossRef] [PubMed]
  49. Ramteke, C.; Chakrabarti, T.; Sarangi, B.K.; Pandey, R.-A. Synthesis of silver nanoparticles from the aqueous extract of leaves of Ocimum sanctum for enhanced antibacterial activity. J. Chem. 2013, 2013, 7. [Google Scholar] [CrossRef]
  50. Dinesh, S.; Karthikeyan, S.; Arumugam, P. Biosynthesis of silver nanoparticles from Glycyrrhiza glabra root extract. Arch Appl. Sci. Res. 2012, 4, 178–187. [Google Scholar]
  51. Chahardoli, A.; Karimi, N.; Fattahi, A. Biosynthesis, characterization, antimicrobial and cytotoxic effects of silver nanoparticles using Nigella arvensis seed extract. Iran. J. Pharm. Res. 2017, 16, 1167–1175. [Google Scholar] [PubMed]
  52. Ghosh, S.; Patil, S.; Ahire, M.; Kitture, R.; Kale, S.; Pardesi, K.; Cameotra, S.S.; Bellare, J.; Dhavale, D.D.; Jabgunde, A.; et al. Synthesis of silver nanoparticles using Dioscorea bulbifera tuber extract and evaluation of its synergistic potential in combination with antimicrobial agents. Int. J. Nanomed. 2012, 7, 483–496. [Google Scholar]
  53. Ajitha, B.; Ashok Kumar Reddy, Y.; Shameer, S.; Rajesh, K.M.; Suneetha, Y.; Sreedhara Reddy, P. Lantana camara leaf extract mediated silver nanoparticles: Antibacterial, green catalyst. J. Photochem. Photobiol. B 2015, 149, 84–92. [Google Scholar] [CrossRef] [PubMed]
  54. Thakore, S.; Rathore, P.S.; Jadeja, R.N.; Thounaojam, M.; Devkar, R.V. Sunflower oil mediated biomimetic synthesis and cytotoxicity of monodisperse hexagonal silver nanoparticles. Mater. Sci. Eng. C 2014, 44, 209–215. [Google Scholar] [CrossRef] [PubMed]
  55. Ghaedi, M.; Yousefinejad, M.; Safarpoor, M.; Khafri, H.Z.; Purkait, M.K. Rosmarinus officinalis leaf extract mediated green synthesis of silver nanoparticles and investigation of its antimicrobial properties. J. Ind. Eng. Chem. 2015, 31, 167–172. [Google Scholar] [CrossRef]
  56. Kiran Kumar, H.A.; Mandal, B.K.; Mohan Kumar, K.; Maddinedi, S. b.; Sai Kumar, T.; Madhiyazhagan, P.; Ghosh, A.R. Antimicrobial and antioxidant activities of Mimusops elengi seed extract mediated isotropic silver nanoparticles. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2014, 130, 13–18. [Google Scholar] [CrossRef] [PubMed]
  57. Kumar, V.; Yadav, S.C.; Yadav, S.K. Syzygium cumini leaf and seed extract mediated biosynthesis of silver nanoparticles and their characterization. J. Chem. Technol. Biotechnol. 2010, 85, 1301–1309. [Google Scholar] [CrossRef]
  58. Jain, S.; Mehata, M.S. Medicinal plant leaf extract and pure flavonoid mediated green synthesis of silver nanoparticles and their enhanced antibacterial property. Sci. Rep. 2017, 7, 15867. [Google Scholar] [CrossRef] [PubMed]
  59. Krishnaraj, C.; Jagan, E.G.; Rajasekar, S.; Selvakumar, P.; Kalaichelvan, P.T.; Mohan, N. Synthesis of silver nanoparticles using Acalypha indica leaf extracts and its antibacterial activity against water borne pathogens. Colloids Surf. B Biointerfaces 2010, 76, 50–56. [Google Scholar] [CrossRef] [PubMed]
  60. LÜ, F.; Gao, Y.; Huang, J.; Sun, D.; Li, Q. Roles of biomolecules in the biosynthesis of silver nanoparticles: Case of Gardenia jasminoides extract. Chin. J. Chem. Eng. 2014, 22, 706–712. [Google Scholar] [CrossRef]
  61. Cruz, D.; Fale, P.L.; Mourato, A.; Vaz, P.D.; Serralheiro, M.L.; Lino, A.R. Preparation and physicochemical characterization of Ag nanoparticles biosynthesized by Lippia citriodora (Lemon Verbena). Colloids Surf. B Biointerfaces 2010, 81, 67–73. [Google Scholar] [CrossRef] [PubMed]
  62. Rashmi, V.; Sanjay, K.R. Green synthesis, characterisation and bioactivity of plant-mediated silver nanoparticles using Decalepis hamiltonii root extract. IET Nanobiotechnol. 2017, 11, 247–254. [Google Scholar] [CrossRef] [PubMed]
  63. Elumalai, D.; Kaleena, P.K.; Ashok, K.; Suresh, A.; Hemavathi, M. Green synthesis of silver nanoparticle using Achyranthes aspera and its larvicidal activity against three major mosquito vectors. Eng. Agric. Environ. Food 2016, 9, 1–8. [Google Scholar] [CrossRef]
  64. Suman, T.Y.; Rajasree, S.R.R.; Jayaseelan, C.; Mary, R.R.; Gayathri, S.; Aranganathan, L.; Remya, R.R. GC-MS analysis of bioactive components and biosynthesis of silver nanoparticles using Hybanthus enneaspermus at room temperature evaluation of their stability and its larvicidal activity. Environ. Sci. Pollut. Res. 2016, 23, 2705–2714. [Google Scholar] [CrossRef] [PubMed]
  65. Ahmad, N.; Sharma, S.; Singh, V.N.; Shamsi, S.F.; Fatma, A.; Mehta, B.R. Biosynthesis of Silver Nanoparticles from Desmodium triflorum: A Novel Approach Towards Weed Utilization. Biotechnol. Res. Int. 2011, 2011, 454090. [Google Scholar] [CrossRef] [PubMed]
  66. Nabikhan, A.; Kandasamy, K.; Raj, A.; Alikunhi, N.M. Synthesis of antimicrobial silver nanoparticles by callus and leaf extracts from saltmarsh plant, Sesuvium portulacastrum L. Colloids Surf. B Biointerfaces 2010, 79, 488–493. [Google Scholar] [CrossRef] [PubMed]
  67. Sengottaiyan, A.; Mythili, R.; Selvankumar, T.; Aravinthan, A.; Kamala-Kannan, S.; Manoharan, K.; Thiyagarajan, P.; Govarthanan, M.; Kim, J.-H. Green synthesis of silver nanoparticles using Solanum indicum L. and their antibacterial, splenocyte cytotoxic potentials. Res. Chem. Int. 2016, 42, 3095–3103. [Google Scholar] [CrossRef]
  68. Mukunthan, K.S.; Balaji, S. Cashew apple juice (Anacardium occidentale L.) speeds up the synthesis of silver nanoparticles. Int. J. Green Nanotechnol. 2012, 4, 71–79. [Google Scholar] [CrossRef]
  69. Dipankar, C.; Murugan, S. The green synthesis, characterization and evaluation of the biological activities of silver nanoparticles synthesized from Iresine herbstii leaf aqueous extracts. Colloids Surf. B Biointerfaces 2012, 98, 112–119. [Google Scholar] [CrossRef] [PubMed]
  70. Geethalakshmi, R.; Sarada, D.V. Synthesis of plant-mediated silver nanoparticles using Trianthema decandra extract and evaluation of their antimicrobial activities. Int. J. Eng. Sci. Technol. 2010, 2, 970–975. [Google Scholar]
  71. Kumar, K.R.; Nattuthurai, N.; Gopinath, P.; Mariappan, T. Synthesis of eco-friendly silver nanoparticles from Morinda tinctoria leaf extract and its larvicidal activity against Culex quinquefasciatus. Parasitol. Res. 2015, 114, 411–417. [Google Scholar] [CrossRef] [PubMed]
  72. Banala, R.R.; Nagati, V.B.; Karnati, P.R. Green synthesis and characterization of Carica papaya leaf extract coated silver nanoparticles through X-ray diffraction, electron microscopy and evaluation of bactericidal properties. Saudi J. Biol. Sci. 2015, 22, 637–644. [Google Scholar] [CrossRef] [PubMed]
  73. Vivek, R.; Thangam, R.; Muthuchelian, K.; Gunasekaran, P.; Kaveri, K.; Kannan, S. Green biosynthesis of silver nanoparticles from Annona squamosa leaf extract and its in vitro cytotoxic effect on MCF-7 cells. Process Biochem. 2012, 47, 2405–2410. [Google Scholar] [CrossRef]
  74. Geethalakshmi, R.; Sarada, D.V.L. Characterization and antimicrobial activity of gold and silver nanoparticles synthesized using saponin isolated from Trianthema decandra L. Ind. Crops Products 2013, 51, 107–115. [Google Scholar]
  75. Jagajjanani Rao, K.; Paria, S. Green synthesis of silver nanoparticles from aqueous Aegle marmelos leaf extract. Mater. Res. Bull. 2013, 48, 628–634. [Google Scholar] [CrossRef]
  76. Dubey, S.P.; Lahtinen, M.; Sillanpää, M. Green synthesis and characterizations of silver and gold nanoparticles using leaf extract of Rosa rugosa. Colloids Surf. A Physicochem. Eng. Asp. 2010, 364, 34–41. [Google Scholar] [CrossRef]
  77. Nayak, D.; Ashe, S.; Rauta, P.R.; Nayak, B. Biosynthesis, characterisation and antimicrobial activity of silver nanoparticles using Hibiscus rosa-sinensis petals extracts. IET Nanobiotechnol. 2015, 9, 288–293. [Google Scholar] [CrossRef] [PubMed]
  78. Im, A.R.; Han, L.; Kim, E.R.; Kim, J.; Kim, Y.S.; Park, Y. Enhanced antibacterial activities of Leonuri herba extracts containing silver nanoparticles. Phytother. Res. 2012, 26, 1249–1255. [Google Scholar] [CrossRef] [PubMed]
  79. Zhou, Y.; Tang, R.-C. Facile and eco-friendly fabrication of AgNPs coated silk for antibacterial and antioxidant textiles using honeysuckle extract. J. Photochem. Photobiol. B Biol. 2018, 178, 463–471. [Google Scholar] [CrossRef] [PubMed]
  80. Yang, N.; Li, W.-H. Mango peel extract mediated novel route for synthesis of silver nanoparticles and antibacterial application of silver nanoparticles loaded onto non-woven fabrics. Ind. Crops Prod. 2013, 48, 81–88. [Google Scholar] [CrossRef]
  81. Liu, Y.; Jin, X.; Chen, Z. The formation of iron nanoparticles by Eucalyptus leaf extract and used to remove Cr(VI). Sci. Total Environ. 2018, 627, 470–479. [Google Scholar] [CrossRef] [PubMed]
  82. Niraimathi, K.L.; Sudha, V.; Lavanya, R.; Brindha, P. Biosynthesis of silver nanoparticles using Alternanthera sessilis (Linn.) extract and their antimicrobial, antioxidant activities. Colloids Surf. B Biointerfaces 2013, 102, 288–291. [Google Scholar] [CrossRef] [PubMed]
  83. Kora, A.J.; Sashidhar, R.B.; Arunachalam, J. Aqueous extract of gum olibanum (Boswellia serrata): A reductant and stabilizer for the biosynthesis of antibacterial silver nanoparticles. Process Biochem. 2012, 47, 1516–1520. [Google Scholar] [CrossRef]
  84. Lagashetty, A.N. Green synthesis and characterization of silver nanoparticles using Piper betel leaf extract. Bull. Adv. Sci. Res. 2015, 1, 136–138. [Google Scholar]
  85. Patil, C.D.; Patil, S.V.; Borase, H.P.; Salunke, B.K.; Salunkhe, R.B. Larvicidal activity of silver nanoparticles synthesized using Plumeria rubra plant latex against Aedes aegypti and Anopheles stephensi. Parasitol. Res. 2012, 110, 1815–1822. [Google Scholar] [CrossRef] [PubMed]
  86. Bar, H.; Bhui, D.K.; Sahoo, G.P.; Sarkar, P.; De, S.P.; Misra, A. Green synthesis of silver nanoparticles using latex of Jatropha curcas. Colloids Surf. A Physicochem. Eng. Asp. 2009, 339, 134–139. [Google Scholar] [CrossRef]
  87. Yasmin, A.; Ramesh, K.; Rajeshkumar, S. Optimization and stabilization of gold nanoparticles by using herbal plant extract with microwave heating. Nano Converg. 2014, 1, 12. [Google Scholar] [CrossRef] [PubMed]
  88. Khalil, M. Biosynthesis of gold nanoparticles using extract of grape (Vitis vinifera) leaves and seeds. Prog. Nanotechnol. Nanomater. 2014, 3, 1–12. [Google Scholar]
  89. Philip, D. Rapid green synthesis of spherical gold nanoparticles using Mangifera indica leaf. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2010, 77, 807–810. [Google Scholar] [CrossRef] [PubMed]
  90. Mata, R.; Nakkala, J.R.; Sadras, S.R. Polyphenol stabilized colloidal gold nanoparticles from Abutilon indicum leaf extract induce apoptosis in HT-29 colon cancer cells. Colloids Surf. B Biointerfaces 2016, 143, 499–510. [Google Scholar] [CrossRef] [PubMed]
  91. Rajathi, F.A.A.; Arumugam, R.; Saravanan, S.; Anantharaman, P. Phytofabrication of gold nanoparticles assisted by leaves of Suaeda monoica and its free radical scavenging property. J. Photochem. Photobiol. B Biol. 2014, 135, 75–80. [Google Scholar] [CrossRef] [PubMed]
  92. Das, J.; Velusamy, P. Catalytic reduction of methylene blue using biogenic gold nanoparticles from Sesbania grandiflora L. J. Taiwan Inst. Chem. Eng. 2014, 45, 2280–2285. [Google Scholar] [CrossRef]
  93. Yu, J.; Xu, D.; Guan, H.N.; Wang, C.; Huang, L.K.; Chi, D.F. Facile one-step green synthesis of gold nanoparticles using Citrus maxima aqueous extracts and its catalytic activity. Mater. Lett. 2016, 166, 110–112. [Google Scholar] [CrossRef]
  94. Elbagory, A.M.; Meyer, M.; Cupido, C.N.; Hussein, A.A. Inhibition of bacteria associated with wound infection by biocompatible green synthesized gold nanoparticles from South African plant extracts. Nanomaterials 2017, 7, 417. [Google Scholar] [CrossRef] [PubMed]
  95. Chahardoli, A.; Karimi, N.; Sadeghi, F.; Fattahi, A. Green approach for synthesis of gold nanoparticles from Nigella arvensis leaf extract and evaluation of their antibacterial, antioxidant, cytotoxicity and catalytic activities. Artif. Cells Nanomed. Biotechnol. 2018, 46, 579–588. [Google Scholar] [CrossRef] [PubMed]
  96. 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]
  97. Bogireddy, N.K.R.; Hoskote Anand, K.K.; Mandal, B.K. Gold nanoparticles—Synthesis by Sterculia acuminata extract and its catalytic efficiency in alleviating different organic dyes. J. Mol. Liq. 2015, 211, 868–875. [Google Scholar] [CrossRef]
  98. Mohan Kumar, K.; Mandal, B.K.; Kiran Kumar, H.A.; Maddinedi, S.B. Green synthesis of size controllable gold nanoparticles. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2013, 116, 539–545. [Google Scholar] [CrossRef] [PubMed]
  99. Ankamwar, B. Biosynthesis of gold nanoparticles (Green-gold) using leaf extract of Terminalia catappa. E J. Chem. 2010, 7, 1334–1339. [Google Scholar] [CrossRef]
  100. Koperuncholan, M. Bioreduction of chloroauric acid (HAuCl4) for the synthesis of gold nanoparticles (GNPs): A special empathies of pharmacological activity. Int. J. Phytopharm. 2015, 5, 72–80. [Google Scholar]
  101. Venkatachalam, M.; Govindaraju, K.; Mohamed Sadiq, A.; Tamilselvan, S.; Ganesh Kumar, V.; Singaravelu, G. Functionalization of gold nanoparticles as antidiabetic nanomaterial. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2013, 116, 331–338. [Google Scholar] [CrossRef] [PubMed]
  102. Choi, Y.; Kang, S.; Cha, S.H.; Kim, H.S.; Song, K.; Lee, Y.J.; Kim, K.; Kim, Y.S.; Cho, S.; Park, Y. Platycodon saponins from Platycodi Radix (Platycodon grandiflorum) for the green synthesis of gold and silver nanoparticles. Nanoscale Res. Lett. 2018, 13, 018–2436. [Google Scholar] [CrossRef] [PubMed]
  103. Zayed, M.F.; Eisa, W.H. Phoenix dactylifera L. leaf extract phytosynthesized gold nanoparticles; controlled synthesis and catalytic activity. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2014, 121, 238–244. [Google Scholar] [CrossRef] [PubMed]
  104. Shankar, S.; Jaiswal, L.; Aparna, R.S.L.; Prasad, R.G.S.V. Synthesis, characterization, in vitro biocompatibility, and antimicrobial activity of gold, silver and gold silver alloy nanoparticles prepared from Lansium domesticum fruit peel extract. Mater. Lett. 2014, 137, 75–78. [Google Scholar] [CrossRef]
  105. Islam, N.U.; Jalil, K.; Shahid, M.; Rauf, A.; Muhammad, N.; Khan, A.; Shah, M.R.; Khan, M.A. Green synthesis and biological activities of gold nanoparticles functionalized with Salix alba. Arab. J. Chem. 2015. [Google Scholar] [CrossRef]
  106. Francis, G.; Thombre, R.; Parekh, F.; Leksminarayan, P. Bioinspired Synthesis of Gold Nanoparticles Using Ficus benghalensis (Indian Banyan) Leaf Extract. Chem. Sci. Trans. 2014, 2014, 470–474. [Google Scholar]
  107. Arunachalam, K.D.; Arun, L.B.; Annamalai, S.K.; Arunachalam, A.M. Biofunctionalized gold nanoparticles synthesis from gymnema sylvestre and its preliminary anticancer activity. Int. J. Pharm. Pharm. Sci. 2014, 6, 423–430. [Google Scholar]
  108. Gopinath, K.; Venkatesh, K.S.; Ilangovan, R.; Sankaranarayanan, K.; Arumugam, A. Green synthesis of gold nanoparticles from leaf extract of Terminalia arjuna, for the enhanced mitotic cell division and pollen germination activity. Ind. Crops Prod. 2013, 50, 737–742. [Google Scholar] [CrossRef]
  109. Ahmad, T.; Bustam, M.A.; Irfan, M.; Moniruzzaman, M.; Anwaar Asghar, H.M.; Bhattacharjee, S. Green synthesis of stabilized spherical shaped gold nanoparticles using novel aqueous Elaeis guineensis (oil palm) leaves extract. J. Mol. Struct. 2018, 1159, 167–173. [Google Scholar] [CrossRef]
  110. Varun, S.; Sivakumar, S.; Rafiqkhan, M.; Vijayakumar, S. Green synthesis of gold nanoparticles using Argemone mexicana L. Leaf extract and its characterization. Int. J. Pharm. Sci. Rev. Res. 2015, 32, 42–44. [Google Scholar]
  111. Correa, S.N.; Naranjo, A.M.; Herrera, A.P. Biosynthesis and characterization of gold nanoparticles using extracts of Tamarindus indica L. Leaves. J. Phys. Conf. Ser. 2016, 687, 012082. [Google Scholar] [CrossRef]
  112. Isaac, R.S.R.; Sakthivel, G.; Murthy, C. Green synthesis of gold and silver nanoparticles using Averrhoa bilimbi fruit extract. J. Nanotechnol. 2013, 2013, 6. [Google Scholar] [CrossRef]
  113. Sathishkumar, G.; Jha, P.K.; Vignesh, V.; Rajkuberan, C.; Jeyaraj, M.; Selvakumar, M.; Jha, R.; Sivaramakrishnan, S. Cannonball fruit (Couroupita guianensis, Aubl.) extract mediated synthesis of gold nanoparticles and evaluation of its antioxidant activity. J. Mol. Liq. 2016, 215, 229–236. [Google Scholar]
  114. Dutta, P.P.; Bordoloi, M.; Gogoi, K.; Roy, S.; Narzary, B.; Bhattacharyya, D.R.; Mohapatra, P.K.; Mazumder, B. Antimalarial silver and gold nanoparticles: Green synthesis, characterization and in vitro study. Biomed. Pharmacother. 2017, 91, 567–580. [Google Scholar] [CrossRef] [PubMed]
  115. Zarzuela, R.; Luna, M.J.; Gil, M.L.A.; Ortega, M.J.; Palacios-Santander, J.M.; Naranjo-Rodríguez, I.; Delgado, J.J.; Cubillana-Aguilera, L.M. Analytical determination of the reducing and stabilization agents present in different Zostera noltii extracts used for the biosynthesis of gold nanoparticles. J. Photochem. Photobiol. B Biol. 2018, 179, 32–38. [Google Scholar] [CrossRef] [PubMed]
  116. Abbasi, T.; Anuradha, J.; Ganaie, S.U.; Abbasi, S.A. Gainful utilization of the highly intransigent weed ipomoea in the synthesis of gold nanoparticles. J. King Saud Univ. Sci. 2015, 27, 15–22. [Google Scholar] [CrossRef]
  117. Vankar, P.S.; Bajpai, D. Preparation of gold nanoparticles from Mirabilis jalapa flowers. Indian J. Biochem. Biophys. 2010, 47, 157–160. [Google Scholar] [PubMed]
  118. Jimenez Perez, Z.E.; Mathiyalagan, R.; Markus, J.; Kim, Y.J.; Kang, H.M.; Abbai, R.; Seo, K.H.; Wang, D.; Soshnikova, V.; Yang, D.C. Ginseng-berry-mediated gold and silver nanoparticle synthesis and evaluation of their in vitro antioxidant, antimicrobial, and cytotoxicity effects on human dermal fibroblast and murine melanoma skin cell lines. Int. J. Nanomed. 2017, 12, 709–723. [Google Scholar] [CrossRef] [PubMed]
  119. Abdel-Raouf, N.; Al-Enazi, N.M.; Ibraheem, I.B.M. Green biosynthesis of gold nanoparticles using Galaxaura elongata and characterization of their antibacterial activity. Arab. J. Chem. 2017, 10, S3029–S3039. [Google Scholar] [CrossRef]
  120. Hajra, A.; Dutta, S.; Mondal, N.K. Mosquito larvicidal activity of cadmium nanoparticles synthesized from petal extracts of marigold (Tagetes sp.) and rose (Rosa sp.) flower. J. Parasit. Dis. Off. Organ Indian Soc. Parasitol. 2016, 40, 1519–1527. [Google Scholar] [CrossRef] [PubMed]
  121. Nazar, N.; Bibi, I.; Kamal, S.; Iqbal, M.; Nouren, S.; Jilani, K.; Umair, M.; Ata, S. Cu nanoparticles synthesis using biological molecule of P. granatum seeds extract as reducing and capping agent: Growth mechanism and photo-catalytic activity. Int. J. Biol. Macromol. 2018, 106, 1203–1210. [Google Scholar] [CrossRef] [PubMed]
  122. Brumbaugh, A.D.; Cohen, K.A.; St. Angelo, S.K. Ultrasmall copper nanoparticles synthesized with a plant eea reducing agent. ACS Sustain. Chem. Eng. 2014, 2, 1933–1939. [Google Scholar] [CrossRef]
  123. Cheirmadurai, K.; Biswas, S.; Murali, R.; Thanikaivelan, P. Green synthesis of copper nanoparticles and conducting nanobiocomposites using plant and animal sources. RSC Adv. 2014, 4, 19507–19511. [Google Scholar] [CrossRef]
  124. Nasrollahzadeh, M.; Mohammad Sajadi, S. Pd nanoparticles synthesized in situ with the use of Euphorbia granulate leaf extract: Catalytic properties of the resulting particles. J. Colloid Interface Sci. 2016, 462, 243–251. [Google Scholar] [CrossRef] [PubMed]
  125. Nasrollahzadeh, M.; Sajadi, S.M.; Maham, M. Green synthesis of palladium nanoparticles using Hippophae rhamnoides Linn leaf extract and their catalytic activity for the Suzuki–Miyaura coupling in water. J. Mol. Catal. A Chem. 2015, 396, 297–303. [Google Scholar] [CrossRef]
  126. Dauthal, P.; Mukhopadhyay, M. Biosynthesis of palladium nanoparticles using Delonix regia leaf extract and its catalytic activity for nitro-aromatics hydrogenation. Ind. Eng. Chem. Res. 2013, 52, 18131–18139. [Google Scholar] [CrossRef]
  127. Zheng, B.; Kong, T.; Jing, X.; Odoom-Wubah, T.; Li, X.; Sun, D.; Lu, F.; Zheng, Y.; Huang, J.; Li, Q. Plant-mediated synthesis of platinum nanoparticles and its bioreductive mechanism. J. Colloid Interface Sci. 2013, 396, 138–145. [Google Scholar] [CrossRef] [PubMed]
  128. Song, J.Y.; Kwon, E.Y.; Kim, B.S. Biological synthesis of platinum nanoparticles using Diopyros kaki leaf extract. Bioprocess Biosyst. Eng. 2010, 33, 159–164. [Google Scholar] [CrossRef] [PubMed]
  129. Ghosh, S.; Nitnavare, R.; Dewle, A.; Tomar, G.B.; Chippalkatti, R.; More, P.; Kitture, R.; Kale, S.; Bellare, J.; Chopade, B.A. Novel platinum-palladium bimetallic nanoparticles synthesized by Dioscorea bulbifera: Anticancer and antioxidant activities. Int. J. Nanomed. 2015, 10, 7477–7790. [Google Scholar]
  130. Suresh, D.; Nethravathi, P.C.; Udayabhanu; Rajanaika, H.; Nagabhushana, H; Sharma, S.C. Green synthesis of multifunctional zinc oxide (ZnO) nanoparticles using Cassia fistula plant extract and their photodegradative, antioxidant and antibacterial activities. Mater. Sci. Semicond. Process. 2015, 31, 446–454. [Google Scholar] [CrossRef]
  131. Elumalai, K.; Velmurugan, S. Green synthesis, characterization and antimicrobial activities of zinc oxide nanoparticles from the leaf extract of Azadirachta indica (L.). Appl. Surf. Sci. 2015, 345, 329–336. [Google Scholar] [CrossRef]
  132. Jafarirad, S.; Mehrabi, M.; Divband, B.; Kosari-Nasab, M. Biofabrication of zinc oxide nanoparticles using fruit extract of Rosa canina and their toxic potential against bacteria: A mechanistic approach. Mater. Sci. Eng. C 2016, 59, 296–302. [Google Scholar] [CrossRef] [PubMed]
  133. Ali, K.; Dwivedi, S.; Azam, A.; Saquib, Q.; Al-Said, M.S.; Alkhedhairy, A.A.; Musarrat, J. Aloe vera extract functionalized zinc oxide nanoparticles as nanoantibiotics against multi-drug resistant clinical bacterial isolates. J. Colloid Interface Sci. 2016, 472, 145–156. [Google Scholar] [CrossRef] [PubMed]
  134. Thema, F.T.; Manikandan, E.; Dhlamini, M.S.; Maaza, M. Green synthesis of ZnO nanoparticles via Agathosma betulina natural extract. Mater. Lett. 2015, 161, 124–127. [Google Scholar] [CrossRef]
  135. Dobrucka, R.; Dlugaszewska, J. Biosynthesis and antibacterial activity of ZnO nanoparticles using Trifolium pratense flower extract. Saudi J. Biol. Sci. 2016, 23, 517–523. [Google Scholar] [CrossRef] [PubMed]
  136. Rajiv, P.; Rajeshwari, S.; Venckatesh, R. Bio-fabrication of zinc oxide nanoparticles using leaf extract of Parthenium hysterophorus L. and its size-dependent antifungal activity against plant fungal pathogens. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2013, 112, 384–387. [Google Scholar] [CrossRef] [PubMed]
  137. Anbuvannan, M.; Ramesh, M.; Viruthagiri, G.; Shanmugam, N.; Kannadasan, N. Anisochilus carnosus leaf extract mediated synthesis of zinc oxide nanoparticles for antibacterial and photocatalytic activities. Mater. Sci. Semicond. Process. 2015, 39, 621–628. [Google Scholar] [CrossRef]
  138. Nagajyothi, P.C.; Sreekanth, T.V.M.; Tettey, C.O.; Jun, Y.I.; Mook, S.H. Characterization, antibacterial, antioxidant, and cytotoxic activities of ZnO nanoparticles using Coptidis Rhizoma. Bioorgan. Med. Chem. Lett. 2014, 24, 4298–4303. [Google Scholar] [CrossRef] [PubMed]
  139. Gawade, V.V.; Gavade, N.L.; Shinde, H.M.; Babar, S.B.; Kadam, A.N.; Garadkar, K.M. Green synthesis of ZnO nanoparticles by using Calotropis procera leaves for the photodegradation of methyl orange. J. Mater. Sci. Mater. Electron. 2017, 28, 14033–14039. [Google Scholar] [CrossRef]
  140. Avoseh, O.N.; Oyedeji, O.O.; Aremu, O.; Nkeh-Chungag, B.N.; Songca, S.P.; Oyedeji, A.O.; Mohan, S.; Oluwafemi, O.S. Biosynthesis of silver nanoparticles from Acacia mearnsii De Wild stem bark and its antinociceptive properties. Green Chem. Lett. Rev. 2017, 10, 59–68. [Google Scholar] [CrossRef]
  141. Halder, A.; Das, S.; Bera, T.; Mukherjee, A. Rapid synthesis for monodispersed gold nanoparticles in kaempferol and anti-leishmanial efficacy against wild and drug resistant strains. RSC Adv. 2017, 7, 14159–14167. [Google Scholar] [CrossRef] [Green Version]
  142. Kavitha, S.; Dhamodaran, M.; Prasad, R.; Ganesan, M. Synthesis and characterisation of zinc oxide nanoparticles using terpenoid fractions of Andrographis paniculata leaves. Int. Nano Lett. 2017, 7, 141–147. [Google Scholar] [CrossRef]
  143. Hwang, S.J.; Jun, S.H.; Park, Y.; Cha, S.-H.; Yoon, M.; Cho, S.; Lee, H.-J.; Park, Y. Green synthesis of gold nanoparticles using chlorogenic acid and their enhanced performance for inflammation. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 1677–1688. [Google Scholar] [CrossRef] [PubMed]
  144. Sahu, N.; Soni, D.; Chandrashekhar, B.; Satpute, D.B.; Saravanadevi, S.; Sarangi, B.K.; Pandey, R.A. Synthesis of silver nanoparticles using flavonoids: Hesperidin, naringin and diosmin, and their antibacterial effects and cytotoxicity. Int. Nano Lett. 2016, 6, 173–181. [Google Scholar] [CrossRef]
  145. Kachlicki, P.; Piasecka, A.; Stobiecki, M.; Marczak, Ł. Structural Characterization of Flavonoid Glycoconjugates and Their Derivatives with Mass Spectrometric Techniques. Molecules 2016, 21, 1494. [Google Scholar] [CrossRef] [PubMed]
  146. Kostyuk, V.A.; Potapovich, A.I.; Kostyuk, T.V.; Cherian, M.G. Metal complexes of dietary flavonoids: Evaluation of radical scavenger properties and protective activity against oxidative stress in vivo. Cell Mol. Biol. 2007, 53, 62–69. [Google Scholar] [PubMed]
  147. Samsonowicz, M.; Regulska, E.; Kalinowska, M. Hydroxyflavone metal complexes—Molecular structure, antioxidant activity and biological effects. Chem. Biol. Interact. 2017, 273, 245–256. [Google Scholar] [CrossRef] [PubMed]
  148. Pusz, J.; Nitka, B. Synthesis and physicochemical properties of the complexes of Co(II), Ni(II), and Cu(II) with chrysin. Microchem. J. 1997, 56, 373–381. [Google Scholar] [CrossRef]
  149. Dowling, S.; Regan, F.; Hughes, H. The characterisation of structural and antioxidant properties of isoflavone metal chelates. J. Inorg. Biochem. 2010, 104, 1091–1098. [Google Scholar] [CrossRef] [PubMed]
  150. Erdogan, G.; Karadag, R.; Eler, A. Aluminium(III), Fe(II) complexes and dyeing properties of apigenin(5,7,4′-trihydroxy flavone). Rev. Anal. Chem. 2010, 29, 211–232. [Google Scholar] [CrossRef]
  151. Tan, M.; Zhu, J.; Pan, Y.; Chen, Z.; Liang, H.; Liu, H.; Wang, H. Synthesis, cytotoxic activity, and DNA binding properties of Copper (II) complexes with hesperetin, naringenin, and apigenin. Bioinorg. Chem. Appl. 2009, 2009, 347872. [Google Scholar] [CrossRef] [PubMed]
  152. Perez, C.A.; Wei, Y.; Guo, M. Iron-binding and anti-Fenton properties of baicalein and baicalin. J. Inorg. Biochem. 2009, 103, 326–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Dimitrić Marković, J.M.; Marković, Z.S.; Brdarić, T.P.; Pavelkić, V.M.; Jadranin, M.B. Iron complexes of dietary flavonoids: Combined spectroscopic and mechanistic study of their free radical scavenging activity. Food Chem. 2011, 129, 1567–1577. [Google Scholar] [CrossRef]
  154. Rygula, A.; Wrobel, T.P.; Szklarzewicz, J.; Baranska, M. Raman and UV–vis spectroscopy studies on luteolin–Al(III) complexes. Vib. Spectrosc. 2013, 64, 21–26. [Google Scholar] [CrossRef]
  155. Leopoldini, M.; Russo, N.; Toscano, M. The molecular basis of working mechanism of natural polyphenolic antioxidants. Food Chem. 2011, 125, 288–306. [Google Scholar] [CrossRef]
  156. Raza, A.; Xu, X.; Xia, L.; Xia, C.; Tang, J.; Ouyang, Z. Quercetin-Iron complex: Synthesis, characterization, antioxidant, DNA binding, DNA cleavage, and antibacterial activity studies. J. Fluoresc. 2016, 26, 2023–2031. [Google Scholar] [CrossRef] [PubMed]
  157. Sanna, V.; Pala, N.; Dessi, G.; Manconi, P.; Mariani, A.; Dedola, S.; Rassu, M.; Crosio, C.; Iaccarino, C.; Sechi, M. Single-step green synthesis and characterization of gold-conjugated polyphenol nanoparticles with antioxidant and biological activities. Int. J. Nanomed. 2014, 9, 4935–4951. [Google Scholar]
  158. Gudikandula, K.; Charya Maringanti, S. Synthesis of silver nanoparticles by chemical and biological methods and their antimicrobial properties. J. Exp. Nanosci. 2016, 11, 714–721. [Google Scholar] [CrossRef]
  159. Kunjiappan, S.; Chowdhury, R.; Bhattacharjee, C. A green chemistry approach for the synthesis and characterization of bioactive gold nanoparticles using Azolla microphylla methanol extract. Front. Mater. Sci. 2014, 8, 123–135. [Google Scholar] [CrossRef]
  160. Nazeruddin, G.M.; Prasad, N.R.; Waghmare, S.R.; Garadkar, K.M.; Mulla, I.S. Extracellular biosynthesis of silver nanoparticle using Azadirachta indica leaf extract and its anti-microbial activity. J. Alloys Compd. 2014, 583, 272–277. [Google Scholar] [CrossRef]
  161. Bagherzade, G.; Tavakoli, M.M.; Namaei, M.H. Green synthesis of silver nanoparticles using aqueous extract of saffron (Crocus sativus L.) wastages and its antibacterial activity against six bacteria. Asian Pac. J. Trop. Biomed. 2017, 7, 227–233. [Google Scholar] [CrossRef]
  162. Gomathi, M.; Rajkumar, P.V.; Prakasam, A.; Ravichandran, K. Green synthesis of silver nanoparticles using Datura stramonium leaf extract and assessment of their antibacterial activity. Resour. Eff. Technol. 2017, 3, 280–284. [Google Scholar] [CrossRef]
  163. Abdelmoteleb, A.; Valdez-Salas, B.; Ceceña-Duran, C.; Tzintzun-Camacho, O.; Gutiérrez-Miceli, F.; Grimaldo-Juarez, O.; González-Mendoza, D. Silver nanoparticles from Prosopis glandulosa and their potential application as biocontrol of Acinetobacter calcoaceticus and Bacillus cereus. Chem. Speciat. Bioavailab. 2017, 29, 1–5. [Google Scholar] [CrossRef]
  164. Muthulakshmi, L.; Rajini, N.; Nellaiah, H.; Kathiresan, T.; Jawaid, M.; Rajulu, A.V. Preparation and properties of cellulose nanocomposite films with in situ generated copper nanoparticles using Terminalia catappa leaf extract. Int. J. Biol. Macromol. 2017, 95, 1064–1071. [Google Scholar] [CrossRef] [PubMed]
  165. Tareq, F.K.; Fayzunnesa, M.; Kabir, M.S.; Nuzat, M. Mechanism of bio molecule stabilized selenium nanoparticles against oxidation process and Clostridium Botulinum. Microb. Pathog. 2018, 115, 68–73. [Google Scholar] [CrossRef] [PubMed]
  166. Tahir, K.; Nazir, S.; Ahmad, A.; Li, B.; Khan, A.U.; Khan, Z.U.; Khan, F.U.; Khan, Q.U.; Khan, A.; Rahman, A.U. Facile and green synthesis of phytochemicals capped platinum nanoparticles and in vitro their superior antibacterial activity. J. Photochem. Photobiol. B 2017, 166, 246–251. [Google Scholar] [CrossRef] [PubMed]
  167. Subhapriya, S.; Gomathipriya, P. Green synthesis of titanium dioxide (TiO2) nanoparticles by Trigonella foenum-graecum extract and its antimicrobial properties. Microb. Pathog. 2018, 116, 215–220. [Google Scholar] [CrossRef] [PubMed]
  168. Manikandan, V.; Velmurugan, P.; Park, J.-H.; Chang, W.-S.; Park, Y.-J.; Jayanthi, P.; Cho, M.; Oh, B.-T. Green synthesis of silver oxide nanoparticles and its antibacterial activity against dental pathogens. 3 Biotech 2017, 7, 72. [Google Scholar] [CrossRef] [PubMed]
  169. Baskaran, X.; Geo Vigila, A.V.; Parimelazhagan, T.; Muralidhara-Rao, D.; Zhang, S. Biosynthesis, characterization, and evaluation of bioactivities of leaf extract-mediated biocompatible silver nanoparticles from an early tracheophyte, Pteris tripartita Sw. Int. J. Nanomed. 2016, 11, 5789–5806. [Google Scholar] [CrossRef] [PubMed]
  170. Singh, K.; Panghal, M.; Kadyan, S.; Chaudhary, U.; Yadav, J.P. Green silver nanoparticles of Phyllanthus amarus: As an antibacterial agent against multi drug resistant clinical isolates of Pseudomonas aeruginosa. J. Nanobiotechnol. 2014, 12, 40. [Google Scholar] [CrossRef] [PubMed]
  171. Kumar, S.S.D.; Houreld, N.N.; Kroukamp, E.M.; Abrahamse, H. Cellular imaging and bactericidal mechanism of green-synthesized silver nanoparticles against human pathogenic bacteria. J. Photochem. Photobiol. B Biol. 2018, 178, 259–269. [Google Scholar] [CrossRef] [PubMed]
  172. Yasir, M.; Singh, J.; Tripathi, M.K.; Singh, P.; Shrivastava, R. Green synthesis of silver nanoparticles using leaf extract of common arrowhead houseplant and its anticandidal activity. Pharmacogn. Mag. 2018, 13 (Suppl. 4), S840–S844. [Google Scholar] [PubMed]
  173. Zahir, A.A.; Rahuman, A.A. Evaluation of different extracts and synthesised silver nanoparticles from leaves of Euphorbia prostrata against Haemaphysalis bispinosa and Hippobosca maculata. Vet. Parasitol. 2012, 187, 511–520. [Google Scholar] [CrossRef] [PubMed]
  174. Rajakumar, G.; Abdul Rahuman, A. Larvicidal activity of synthesized silver nanoparticles using Eclipta prostrata leaf extract against filariasis and malaria vectors. Acta Tropica 2011, 118, 196–203. [Google Scholar] [CrossRef] [PubMed]
  175. Kumar, D.; Kumar, G.; Das, R.; Agrawal, V. Strong larvicidal potential of silver nanoparticles (AgNPs) synthesized using Holarrhena antidysenterica (L.) Wall. bark extract against malarial vector, Anopheles stephensi Liston. Process Saf. Environ. Prot. 2018, 116, 137–148. [Google Scholar] [CrossRef]
  176. Jayaseelan, C.; Rahuman, A.A.; Rajakumar, G.; Vishnu Kirthi, A.; Santhoshkumar, T.; Marimuthu, S.; Bagavan, A.; Kamaraj, C.; Zahir, A.A.; Elango, G. Synthesis of pediculocidal and larvicidal silver nanoparticles by leaf extract from heartleaf moonseed plant, Tinospora cordifolia Miers. Parasitol. Res. 2011, 109, 185–194. [Google Scholar] [CrossRef] [PubMed]
  177. Ghramh, H.A.; Al-Ghamdi, K.M.; Mahyoub, J.A.; Ibrahim, E.H. Chrysanthemum extract and extract prepared silver nanoparticles as biocides to control Aedes aegypti (L.), the vector of dengue fever. J. Asia Pac. Entomol. 2018, 21, 205–210. [Google Scholar] [CrossRef]
  178. Chai, S.H.; Wang, Y.; Qiao, Y.; Wang, P.; Li, Q.; Xia, C.; Ju, M. Bio fabrication of silver nanoparticles as an effective wound healing agent in the wound care after anorectal surgery. J. Photochem. Photobiol. B 2018, 178, 457–462. [Google Scholar] [CrossRef] [PubMed]
  179. Kanjikar, A.P.; Hugar, A.L.; Londonkar, R.L. Characterization of phyto-nanoparticles from Ficus krishnae for their antibacterial and anticancer activities. Drug Dev. Ind. Pharm. 2018, 44, 377–384. [Google Scholar] [CrossRef] [PubMed]
  180. Suriyakalaa, U.; Antony, J.J.; Suganya, S.; Siva, D.; Sukirtha, R.; Kamalakkannan, S.; Pichiah, P.B.; Achiraman, S. Hepatocurative activity of biosynthesized silver nanoparticles fabricated using Andrographis paniculata. Colloids Surf. B Biointerfaces 2013, 102, 189–194. [Google Scholar] [CrossRef] [PubMed]
  181. Sudha, A.; Jeyakanthan, J.; Srinivasan, P. Green synthesis of silver nanoparticles using Lippia nodiflora aerial extract and evaluation of their antioxidant, antibacterial and cytotoxic effects. Resour. Eff. Technol. 2017, 3, 506–515. [Google Scholar] [CrossRef]
  182. Velu, V.; Das, M.; Dua, K.; Malipeddi, H. Evaluation of in vitro and in vivo anti-urolithiatic activity of silver nanoparticles containing aqueous leaf extract of Tragia involucrata. Drug Deliv. Transl. Res. 2017, 7, 439–449. [Google Scholar] [CrossRef] [PubMed]
  183. Sukhwal, A.; Jain, D.; Joshi, A.; Rawal, P.; Kushwaha, H.S. Biosynthesised silver nanoparticles using aqueous leaf extract of Tagetes patula L. and evaluation of their antifungal activity against phytopathogenic fungi. IET Nanobiotechnol. 2017, 11, 531–537. [Google Scholar] [CrossRef] [PubMed]
  184. Swain, S.; Barik, S.K.; Behera, T.; Nayak, S.K.; Sahoo, S.K.; Mishra, S.S.; Swain, P. Green synthesis of gold nanoparticles using root and leaf extracts of Vetiveria zizanioides and Cannabis sativa and its antifungal activities. BioNanoScience 2016, 6, 205–213. [Google Scholar] [CrossRef]
  185. Panja, S.; Chaudhuri, I.; Khanra, K.; Bhattacharyya, N. Biological application of green silver nanoparticle synthesized from leaf extract of Rauvolfia serpentina Benth. Asian Pac. J. Trop. Dis. 2016, 6, 549–556. [Google Scholar] [CrossRef]
  186. Daisy, P.; Saipriya, K. Biochemical analysis of Cassia fistula aqueous extract and phytochemically synthesized gold nanoparticles as hypoglycemic treatment for diabetes mellitus. Int. J. Nanomed. 2012, 7, 1189–1202. [Google Scholar] [CrossRef] [PubMed]
  187. Roy, P.; Saha, S.K.; Gayen, P.; Chowdhury, P.; Sinha Babu, S.P. Exploration of antifilarial activity of gold nanoparticle against human and bovine filarial parasites: A nanomedicinal mechanistic approach. Colloids Surf. B Biointerfaces 2018, 161, 236–243. [Google Scholar] [CrossRef] [PubMed]
  188. Islam, N.U.; Khan, I.; Rauf, A.; Muhammad, N.; Shahid, M.; Shah, M.R. Antinociceptive, muscle relaxant and sedative activities of gold nanoparticles generated by methanolic extract of Euphorbia milii. BMC Complement Altern. Med. 2015, 15, 160. [Google Scholar] [CrossRef] [PubMed]
  189. Kumar, B.; Smita, K.; Cumbal, L.; Debut, A. Green synthesis of silver nanoparticles using Andean blackberry fruit extract. Saudi J. Biol. Sci. 2017, 24, 45–50. [Google Scholar] [CrossRef] [PubMed]
  190. Gubitosa, J.; Rizzi, V.; Lopedota, A.; Fini, P.; Laurenzana, A.; Fibbi, G.; Fanelli, F.; Petrella, A.; Laquintana, V.; Denora, N.; et al. One pot environmental friendly synthesis of gold nanoparticles using Punica Granatum Juice: A novel antioxidant agent for future dermatological and cosmetic applications. J. Colloid Interface Sci. 2018, 521, 50–61. [Google Scholar] [CrossRef] [PubMed]
  191. Singh, A.; Singh, N.B.; Hussain, I.; Singh, H. Effect of biologically synthesized copper oxide nanoparticles on metabolism and antioxidant activity to the crop plants Solanum lycopersicum and Brassica oleracea var. botrytis. J. Biotechnol. 2017, 262, 11–27. [Google Scholar] [CrossRef] [PubMed]
  192. Sulaiman, G.M.; Tawfeeq, A.T.; Jaaffer, M.D. Biogenic synthesis of copper oxide nanoparticles using olea europaea leaf extract and evaluation of their toxicity activities: An in vivo and in vitro study. Biotechnol. Prog. 2018, 34, 218–230. [Google Scholar] [CrossRef] [PubMed]
  193. Ahn, S.; Singh, P.; Jang, M.; Kim, Y.-J.; Castro-Aceituno, V.; Simu, S.Y.; Kim, Y.J.; Yang, D.-C. Gold nanoflowers synthesized using Acanthopanacis cortex extract inhibit inflammatory mediators in LPS-induced RAW264.7 macrophages via NF-κB and AP-1 pathways. Colloids Surf. B Biointerfaces 2018, 162, 398–404. [Google Scholar] [CrossRef] [PubMed]
  194. Ateeq, M.; Shah, M.R.; Ali, H.; Kabir, N.; Khan, A.; Nadeem, S. Hepatoprotective and urease inhibitory activities of garlic conjugated gold nanoparticles. New J. Chem. 2015, 39, 5003–5007. [Google Scholar] [CrossRef]
  195. Aswathy Aromal, S.; Philip, D. Green synthesis of gold nanoparticles using Trigonella foenum-graecum and its size-dependent catalytic activity. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2012, 97, 1–5. [Google Scholar] [CrossRef] [PubMed]
  196. Devipriya, D.; Roopan, S.M. Cissus quadrangularis mediated ecofriendly synthesis of copper oxide nanoparticles and its antifungal studies against Aspergillus niger, Aspergillus flavus. Mater. Sci. Eng. C Mater. Biol. Appl. 2017, 80, 38–44. [Google Scholar] [CrossRef] [PubMed]
  197. Gnanavel, V.; Palanichamy, V.; Roopan, S.M. Biosynthesis and characterization of copper oxide nanoparticles and its anticancer activity on human colon cancer cell lines (HCT-116). J. Photochem. Photobiol. B 2017, 171, 133–138. [Google Scholar] [CrossRef] [PubMed]
  198. Şahin, B.; Aygün, A.; Gündüz, H.; Şahin, K.; Demir, E.; Akocak, S.; Şen, F. Cytotoxic effects of platinum nanoparticles obtained from pomegranate extract by the green synthesis method on the MCF-7 cell line. Colloids Surf. B Biointerfaces 2018, 163, 119–124. [Google Scholar] [CrossRef] [PubMed]
  199. Jayaseelan, C.; Gandhi, P.R.; Rajasree, S.R.R.; Suman, T.Y.; Mary, R.R. Toxicity studies of nanofabricated palladium against filariasis and malaria vectors. Environ. Sci. Pollut. Res. Int. 2018, 25, 324–332. [Google Scholar] [CrossRef] [PubMed]
  200. Li, Y.; Wang, H.; Zhang, R.; Zhang, G.; Yang, Y.; Liu, Z. Biofabrication of polyphenols coated Nano palladium and its in-vitro cytotoxicity against human leukemia cell lines (K562). J. Photochem. Photobiol. B 2017, 175, 173–177. [Google Scholar] [CrossRef] [PubMed]
  201. Mahendra, C.; Murali, M.; Manasa, G.; Ponnamma, P.; Abhilash, M.R.; Lakshmeesha, T.R.; Satish, A.; Amruthesh, K.N.; Sudarshana, M.S. Antibacterial and antimitotic potential of bio-fabricated zinc oxide nanoparticles of Cochlospermum religiosum (L.). Microb. Pathog. 2017, 110, 620–629. [Google Scholar] [CrossRef] [PubMed]
  202. Gandhi, P.R.; Jayaseelan, C.; Mary, R.R.; Mathivanan, D.; Suseem, S.R. Acaricidal, pediculicidal and larvicidal activity of synthesized ZnO nanoparticles using Momordica charantia leaf extract against blood feeding parasites. Exp. Parasitol. 2017, 181, 47–56. [Google Scholar] [CrossRef] [PubMed]
  203. Ishwarya, R.; Vaseeharan, B.; Kalyani, S.; Banumathi, B.; Govindarajan, M.; Alharbi, N.S.; Kadaikunnan, S.; Al-anbr, M.N.; Khaled, J.M.; Benelli, G. Facile green synthesis of zinc oxide nanoparticles using Ulva lactuca seaweed extract and evaluation of their photocatalytic, antibiofilm and insecticidal activity. J. Photochem. Photobiol. B Biol. 2018, 178, 249–258. [Google Scholar] [CrossRef] [PubMed]
  204. Angel Ezhilarasi, A.; Judith Vijaya, J.; Kaviyarasu, K.; John Kennedy, L.; Ramalingam, R.J.; Al-Lohedan, H.A. Green synthesis of NiO nanoparticles using Aegle marmelos leaf extract for the evaluation of in-vitro cytotoxicity, antibacterial and photocatalytic properties. J. Photochem. Photobiol. B 2018, 180, 39–50. [Google Scholar] [CrossRef] [PubMed]
  205. Chen, G.; Xu, Y. Biosynthesis of cerium oxide nanoparticles and their effect on lipopolysaccharide (LPS) induced sepsis mortality and associated hepatic dysfunction in male Sprague Dawley rats. Mater. Sci. Eng. C Mater. Biol. Appl. 2018, 83, 148–153. [Google Scholar] [CrossRef] [PubMed]
  206. Thandapani, K.; Kathiravan, M.; Namasivayam, E.; Padiksan, I.A.; Natesan, G.; Tiwari, M.; Giovanni, B.; Perumal, V. Enhanced larvicidal, antibacterial, and photocatalytic efficacy of TiO2 nanohybrids green synthesized using the aqueous leaf extract of Parthenium hysterophorus. Environ. Sci. Pollut. Res. Int. 2017, 23, 10328–10339. [Google Scholar] [CrossRef] [PubMed]
  207. Khatami, M.; Alijani, H.; Sharifi, I.; Sharifi, F.; Pourseyedi, S.; Kharazi, S.; Lima Nobre, M.A.; Khatami, M. Leishmanicidal activity of biogenic Fe3O4 nanoparticles. Sci. Pharm. 2017, 85, 36. [Google Scholar] [CrossRef] [PubMed]
  208. Sisubalan, N.; Ramkumar, V.S.; Pugazhendhi, A.; Karthikeyan, C.; Indira, K.; Gopinath, K.; Hameed, A.S.H.; Basha, M.H.G. ROS-mediated cytotoxic activity of ZnO and CeO2 nanoparticles synthesized using the Rubia cordifolia L. leaf extract on MG-63 human osteosarcoma cell lines. Environ. Sci. Pollut. Res. Int. 2017, 29, 1–11. [Google Scholar] [CrossRef] [PubMed]
  209. Sowndarya, P.; Ramkumar, G.; Shivakumar, M.S. Green synthesis of selenium nanoparticles conjugated Clausena dentata plant leaf extract and their insecticidal potential against mosquito vectors. Artif. Cells Nanomed. Biotechnol. 2017, 45, 1490–1495. [Google Scholar] [CrossRef] [PubMed]
  210. Defaei, M.; Taheri-Kafrani, A.; Miroliaei, M.; Yaghmaei, P. Improvement of stability and reusability of α-amylase immobilized on naringin functionalized magnetic nanoparticles: A robust nanobiocatalyst. Int. J. Biol. Macromol. 2018, 113, 354–360. [Google Scholar] [CrossRef] [PubMed]
  211. Das, P.; Barua, S.; Sarkar, S.; Karak, N.; Bhattacharyya, P.; Raza, N.; Kim, K.-H.; Bhattacharya, S.S. Plant extract–mediated green silver nanoparticles: Efficacy as soil conditioner and plant growth promoter. J. Hazard. Mater. 2018, 346, 62–72. [Google Scholar] [CrossRef] [PubMed]
  212. Dayakar, T.; Rao, K.V.; Bikshalu, K.; Rajendar, V.; Park, S.H. Novel synthesis and characterization of pristine Cu nanoparticles for the non-enzymatic glucose biosensor. J. Mater. Sci. Mater. Med. 2017, 28, 109. [Google Scholar] [CrossRef] [PubMed]
  213. Kurepa, J.; Nakabayashi, R.; Paunesku, T.; Suzuki, M.; Saito, K.; Woloschak, G.E.; Smalle, J.A. Direct isolation of flavonoids from plants using ultra-small anatase TiO2 nanoparticles. Plant J. 2014, 77, 443–453. [Google Scholar] [CrossRef] [PubMed]
  214. Zhao, B.; Cao, X.; De La Torre-Roche, R.; Tan, C.; Yang, T.; White, J.C.; Xiao, H.; Xing, B.; He, L. A green, facile, and rapid method for microextraction and Raman detection of titanium dioxide nanoparticles from milk powder. RSC Adv. 2017, 7, 21380–21388. [Google Scholar] [CrossRef] [Green Version]
  215. Xiao, D.; Zhang, C.; Yuan, D.; He, J.; Wu, J.; Zhang, K.; Lin, R.; He, H. Magnetic solid-phase extraction based on Fe3O4 nanoparticle retrieval of chitosan for the determination of flavonoids in biological samples coupled with high performance liquid chromatography. RSC Adv. 2014, 4, 64843–64854. [Google Scholar] [CrossRef]
  216. Cao, X.; Ma, C.; Gao, Z.; Zheng, J.; He, L.; McClements, D.J.; Xiao, H. Characterization of the interactions between titanium dioxide nanoparticles and polymethoxyflavones using surface-enhanced Raman spectroscopy. J. Agric. Food Chem. 2016, 64, 9436–9441. [Google Scholar] [CrossRef] [PubMed]
  217. Khan, M.A.; Wallace, W.T.; Islam, S.Z.; Nagpure, S.; Strzalka, J.; Littleton, J.M.; Rankin, S.E.; Knutson, B.L. Adsorption and recovery of polyphenolic flavonoids using TiO2-functionalized mesoporous silica nanoparticles. ACS Appl. Mater. Interfaces 2017, 9, 32114–32125. [Google Scholar] [CrossRef] [PubMed]
  218. Schlipf, D.M.; Jones, C.A.; Armbruster, M.E.; Rushing, E.S.; Wooten, K.C.; Rankin, S.E.; Knutson, B.L. Flavonoid adsorption and stability on titania-functionalized silica nanoparticles. Colloids Surf. A Physicochem. Eng. Asp. 2015, 478, 15–21. [Google Scholar] [CrossRef]
  219. Lee, D.; Ko, W.-K.; Hwang, D.-S.; Heo, D.N.; Lee, S.J.; Heo, M.; Lee, K.-S.; Ahn, J.-Y.; Jo, J.; Kwon, I.K. Use of baicalin-conjugated gold nanoparticles for apoptotic induction of breast cancer cells. Nanoscale Res. Lett. 2016, 11, 381. [Google Scholar] [CrossRef] [PubMed]
  220. Singh, B.; Rani, M.; Singh, J.; Moudgil, L.; Sharma, P.; Kumar, S.; Saini, G.S.S.; Tripathi, S.K.; Singh, G.; Kaura, A. Identifying the preferred interaction mode of naringin with gold nanoparticles through experimental, DFT and TDDFT techniques: Insights into their sensing and biological applications. RSC Adv. 2016, 6, 79470–79484. [Google Scholar] [CrossRef]
  221. Pal, R.; Panigrahi, S.; Bhattacharyya, D.; Chakraborti, A.S. Characterization of citrate capped gold nanoparticle-quercetin complex: Experimental and quantum chemical approach. J. Mol. Struct. 2013, 1046, 153–163. [Google Scholar] [CrossRef]
  222. Krishnan, G.; Subramaniyan, J.; Chengalvarayan Subramani, P.; Muralidharan, B.; Thiruvengadam, D. Hesperetin conjugated PEGylated gold nanoparticles exploring the potential role in anti-inflammation and anti-proliferation during diethylnitrosamine-induced hepatocarcinogenesis in rats. Asian J. Pharm. Sci. 2017, 12, 442–455. [Google Scholar] [CrossRef]
  223. Pak, P.J.; Go, E.B.; Hwang, M.H.; Lee, D.G.; Cho, M.J.; Joo, Y.H.; Chung, N. Evaluation of the cytotoxicity of gold nanoparticle-quercetin complex and its potential as a drug delivery vesicle. J. Appl. Biol. Chem. 2016, 59, 145–147. [Google Scholar] [CrossRef]
  224. Rajesh Kumar, S.; Priyatharshni, S.; Babu, V.N.; Mangalaraj, D.; Viswanathan, C.; Kannan, S.; Ponpandian, N. Quercetin conjugated superparamagnetic magnetite nanoparticles for in-vitro analysis of breast cancer cell lines for chemotherapy applications. J. Colloid Interface Sci. 2014, 436, 234–242. [Google Scholar] [CrossRef] [PubMed]
  225. Shah, M.; Fawcett, D.; Sharma, S.; Tripathy, S.K.; Poinern, G.E.J. Green synthesis of metallic nanoparticles via biological entities. Materials 2015, 8, 7278–7308. [Google Scholar] [CrossRef]
  226. Shaik, M.; Ali, Z.; Khan, M.; Kuniyil, M.; Assal, M.; Alkhathlan, H.; Al-Warthan, A.; Siddiqui, M.; Khan, M.; Adil, S. Green synthesis and characterization of palladium nanoparticles using Origanum vulgare L. rxtract and their catalytic activity. Molecules 2017, 22, 165. [Google Scholar] [CrossRef] [PubMed]
  227. Rajendran, S.P.; Sengodan, K. Synthesis and characterization of zinc oxide and iron oxide nanoparticles using Sesbania grandiflora leaf extract as reducing agent. J. Nanosci. 2017, 2017, 7. [Google Scholar] [CrossRef]
Figure 1. Schematic presentation of various methods used in the synthesis of metal NPs.
Figure 1. Schematic presentation of various methods used in the synthesis of metal NPs.
Materials 11 00940 g001
Figure 2. A scheme describing the possible mechanism of in vivo green synthesis of NPs.
Figure 2. A scheme describing the possible mechanism of in vivo green synthesis of NPs.
Materials 11 00940 g002
Figure 3. A scheme describing the mechanism of in vitro green synthesis of nanoparticles.
Figure 3. A scheme describing the mechanism of in vitro green synthesis of nanoparticles.
Materials 11 00940 g003
Figure 4. Metal-chelating properties of flavonoids.
Figure 4. Metal-chelating properties of flavonoids.
Materials 11 00940 g004
Figure 5. A scheme showing possible nanotrapping/nanoharvesting and drug discovery applications of green synthesis.
Figure 5. A scheme showing possible nanotrapping/nanoharvesting and drug discovery applications of green synthesis.
Materials 11 00940 g005
Table 1. Plant components possibly involved in the green synthesis of nanoparticles from various plant species.
Table 1. Plant components possibly involved in the green synthesis of nanoparticles from various plant species.
Plant SpeciesNPsMetabolites Identified in the Extract/NPsReference
Coleus aromaticusAgFlavonoids[44]
Syzygium cuminiAgFlavonoids[45]
Azadirachta indicaAgFlavanoids, terpenoids[46]
Citrus sinensisAgFlavonoids, ascorbic acid, volatile oils[47]
Zingiber officinaleAgFlavonoid, alkaloids[48]
Ocimum sanctumAgEugenols, linalool, terpenes[49]
Glycyrrhiza glabraAgFlavonoids, thiamine and terpenoids[50]
Nigella arvensisAgFlavonoids, alkaloids[51]
Dioscorea bulbiferaAgFlavonoids, polyphenols[52]
Lantana camaraAgFlavonoids, glycosides and carbohydrates[53]
Helianthus annuusAgFatty acids, triglycerides, phenolics, tocopherols[54]
Rosmarinus officinalisAgPolyphenols[55]
Mimusops elengiAgPolyphenols[56]
Syzygium cuminiAgPolyphenols[57]
Ocimum sanctumAgQuercetin[58]
Acalypha indicaAgQuercetin, plant pigment[59]
Gardenia jasminoidesAgRutin, gallic acid, chlorogenic acid[60]
Withania somniferaAgCatechin p-coumaric acid, luteolin-7-glucoside, withanolides[14]
Lippia citriodoraAgVerbascoside, isoverbascoside, chrysoeriol-7-O-diglucoronide, luteonin-7-O-diglucoronide[61]
Decalepis hamiltoniiAgPolyols, phenols[62]
Achyranthes asperaAgPolyols[63]
Hybanthus enneaspermusAgSeveral bioactive phytochemical compounds[64]
Desmodium triflorumAgAscorbic acid[65]
Sesuvium portulacastrumAgFlavones, proteins, terpenoids[66]
Solanum xanthocarpumAgAlcohols, phenols, carboxylic anions[67]
Mentha piperitaAgAlkaloids, flavones, steroids, polysaccharides, amino acids, oximes, proteins, menthol[41]
Anacardium occidentaleAgProteins, polyols[68]
Dioscorea bulbiferaAgDiosgenin, ascorbic acid[52]
Iresine herbstiiAgPhenolic compound[69]
Trianthema decandraAgCatechins, hydroxyflavones[70]
Morinda pubescensAgFlavonoids, triterpenoids, polyphenols[71]
Carica papayaAgProteins, alcohols, phenolics[72]
Annona squamosaAgAlkaloids, glycoside, saponins, tannins, phenolic, carbohydrates[73]
Trianthema decandraAgSaponin[74]
Aegle marmelosAgTannin[75]
Rosa rugosaAgCarboxylate, amine groups[76]
Hibiscus rosa- sinensisAgCarboxylate ion groups[77]
Leonuri herbaAgHydroxyl, polyphenols groups[78]
Lonicera japonicaAgPhenolic and hydroxyl groups of chlorogenic acid[79]
Mangifera indicaAgKetone, aldehydes, hydroxyl, carboxyl groups[80]
EucalyptusFeAlcohol, phenols, alkylaldehyde[81]
Alternanthera sessilisAgTannins, carbohydrates, proteins, ascorbic acid[82]
Boswellia serrataAgProteins[83]
Piper betleAgAmide, aromatic amine[84]
Plumeria rubraAgProteins[85]
Jatropha curcasAgCyclic peptides (curcacycline A and curcacycline B)[86]
Hibiscus rosa- sinensisAuFlavonoids[87]
Vitis viniferaAuFlavonoids[88]
Mangifera indicaAuFavonoids, terpenoids, thiamine[89]
Abutilon indicumAuFlavonoids, phenolic compounds[90]
Suaeda monoicaAuFlavonoids, terpenoids, soluble proteins[91]
Sesbania grandifloraAuFlavonoids, polyphenols[92]
Citrus maximaAuFlavonoids, terpenes, vitamins[93]
Hypoxis hemerocallideaAuFlavonoids, terpenoids, phenolic compounds and/or carbohydrates[94]
Galenia africanaAuFlavonoids, terpenoids, phenolic compounds and/or carbohydrates[94]
Nigella arvensisAuFlavonoids, phenolic compounds[95]
Butea monospermaAuPolyphenols[96]
Sterculia acuminataAuPolyphenols[97]
Terminalia arjunaAuPolyphenols[98]
Terminalia catappaAuHydroxyl group of phenols[99]
Hygrophila spinosaAuHydroxyl group[100]
Cassia auriculataAuHydroxyl group[101]
Platycodon grandiflorumAuHydroxyl group[102]
Phoenix dactyliferaAuHydroxyl group[103]
Lansium domesticumAuCarboxylic acid[104]
Salix albaAuProteins, metabolites having functional groups of amines, alcohols, ketones, aldehydes, carboxylic acids (salicin)[105]
Cinnamomum zeylanicumAuProteins[39]
Ficus benghalensisAuProteins[106]
JatrophaAuProteins[16]
Morinda citrifoliaAuProteins[42]
Gymnema sylvestreAuProteins, polypeptides[107]
Olea europaeaAuProteins[40]
Trianthema decandraAuSaponin[74]
Terminalia arjunaAuHydrolyzable tannins[108]
Elaeis guineensisAuPhenolic, carboxylic, amines[109]
Mentha piperitaAuMenthol[41]
Argemone mexicanaAuPhosphorous compounds[110]
Tamarindus indicaAuPhenolic compounds[111]
Averrhoa bilimbiAuPhenols, tertiary amides[112]
Couroupita guianensisAuPhenol group[113]
Syzygium jambosAuSaccharides, phenolics[114]
Zostera noltiiAuFlavone sulfates[115]
Ipomoea carneaAuPolysaccharides, protein[116]
Mirabilis jalapaAuPolysaccharides[117]
Panax ginsengAuPolysaccharides, phenolic compounds[118]
Galaxaura elongataAuGlutamic acid, hexadecanoic acid, oleic acid, 11-eicosenoic acid, stearic acid, gallic acid, epigallocatechin, catechin, epicatechin gallate[119]
Tagetes sp. and Rosa sp.CdAlcoholic, amide, C–C, –OCH3 groups (tannins, flavonoids, alkaloids and carotenoids)[120]
Punica granatumCuFlavonoids, alkaloids, polyphenols[121]
Cymbopogon citratusCuPolyphenols, proteins[122]
Lawsonia inermisCuPhenolic compounds[123]
Euphorbia granulatePdHydroxyflavones, phenolics[124]
Hippophae rhamnoidesPdFlavonoids[125]
Delonix regiaPdPolyphenols[126]
Cacumen platycladiPtFlavonoids, proteins[127]
Diospyros kakiPtTerpenoids[128]
Dioscorea bulbiferaPt-PdHydroxyl group of polyphenolic compounds[129]
Cassia fistulaZnOFlavonoids, polyphenols[130]
Azadirachta indicaZnOFlavonoids, phenolic acid, terpenoids, protein[131]
Rosa caninaZnOPhenolic and carboxylic acids[132]
Aloe barbadensisZnOPhenol, amines, alcohol groups[133]
Agathosma betulinaZnOHydroxyl group[134]
Trifolium pratenseZnOHydroxyl group[135]
Parthenium hysterophorusZnOPhosphorus compound, secondary sulfonamide, monosubstituted alkyne[136]
Anisochilus carnosusZnOPhenol, carboxylic acid[137]
Coptis chinensisZnOAlcohol, carboxylic acid, alkyl halide, alkynes[138]
Calotropis proceraZnOHydroxyl groups, aldehydes, amines, ketones, carboxylic acids[139]
Table 2. Bioactivities of green-synthesized NPs.
Table 2. Bioactivities of green-synthesized NPs.
NPsPlant Species UsedBioactivity ReportedReference
AgWithania somniferaAntibacterial, anticandidal[14]
AgLansium domesticumAntibacterial[104]
AgCrocus sativusAntibacterial[161]
AgDatura stramoniumAntibacterial[162]
AgProsopis glandulosaAntibacterial[163]
AgAzadirachta indicaAntibacterial[46]
AuHypoxis hemerocallideaAntibacterial[94]
AuGalenia africanaAntibacterial[94]
CuTerminalia catappaAntibacterial[164]
SeAzadirachta indicaAntibacterial[165]
PtTaraxacum laevigatumAntibacterial[166]
TiO2Trigonella foenum-graecumAntibacterial[167]
Ag2OFicus benghalensisAntibacterial[168]
AgPteris tripartitaAntibacterial, antifungal, antioxidant, antiinflammatory[169]
AgPhyllanthus amarusAntibacterial[170]
AgAloe arborescensAntibacterial[171]
AgSyngonium podophyllumAnticandidal[172]
AgEuphorbia prostrataAntiplasmodial[173]
AgOcimum sanctumAntibacterial[58]
AgHybanthus enneaspermusLarvicidal[64]
AgEclipta prostrataLarvicidal[174]
CdTagetes sp. and Rosa sp.Larvicidal[120]
AgHolarrhena antidysentericaLarvicidal[175]
AgTinospora cordifoliaLarvicidal[176]
AgChrysanthemumLarvicidal[177]
AgDelonix elataWound healing[178]
AgFicus krishnaeAntibacterial, anticancer[179]
AgAndrographis paniculataHepatocurative[180]
AgLippia nodifloraAntioxidant, antibacterial, cytotoxic[181]
AgTragia involucrataAntiurolithic[182]
AgTagetes patulaAntifungal[183]
AuVetiveria zizanioidesAntifungal[184]
AuCannabis sativaAntifungal[184]
AgRauvolfia serpentinaAntimicrobial, larvicidal and cytotoxic[185]
AuCassia fistulaAntihypoglycemic[186]
AuTerminalia chebulaAntifilarial[187]
AuEuphorbia miliiAntinociceptive, muscle relaxant, sedative[188]
AgRubus glaucusAntioxidant[189]
AuPunica GranatumAntioxidant[190]
AuAzolla microphyllaAntioxidant[159]
CuOMorus albaAntioxidant[191]
CuOOlea europaeaAntioxidant[192]
AuAcanthopanacis cortexAnti-inflammatory[193]
AuAllium sativumHepatoprotective[194]
AuTrigonella foenum-graecumCatalytic[195]
CuOCissus quadrangularisAntifungal[196]
CuOOrmocarpum cochinchinenseAnticancer[197]
PtPunica granatumCytotoxic[198]
PdTinospora cordifoliaAntifilarial, antimalarial[199]
PdPelargonium graveolensCytotoxic[200]
ZnCochlospermum religiosumAntibacterial, antimitotic[201]
ZnMomordica charantiaAcaricidal, pediculicidal, larvicidal[202]
ZnOUlva lactucaInsecticidal[203]
ZnOHibiscus sabdariffaAntibacterial, antidiabetic[20]
ZnOCalotropis proceraPhotocatalytic[139]
NiDesmodium gangeticumAntioxidant, antibacterial[19]
NiOAegle marmelosCytotoxic, antibacterial[204]
CeO2Camellia sinensisHealing of liver sepsis[205]
TiO2Parthenium hysterophorusLarvicidal, antibacterial, photocatalytic[206]
Fe3O4Rosmarinus officinalisLeishmanicidal[207]
CeO2Rubia cordifoliaAnticancer[208]
SeClausena dentataLarvicidal[209]
AuSalix albaAntifungal, antinociceptive, muscle relaxant[105]
Table 3. Secondary metabolites conjugated with NPs.
Table 3. Secondary metabolites conjugated with NPs.
NPSourceCompound(s) Trapped/ConjugatedReference
AgWithania somnifera leaf extractCatechin, p-coumaric acid, and luteolin-7-glucoside and withanolides[14]
TiO2Arabidopsis thaliana seedlingsFlavonoids[213]
TiO2Food samplesMyricetin[214]
Fe3O4Urine and bloodLuteolin, quercetin, kaempferol[215]
TiO2FlavonoidsFlavonoids[216]
TiO2-SiO2Quercetin, rutinQuercetin, rutin[217]
TiO2-SiO2QuercetinQuercetin[218]
AuBaicalinBaicalin[219]
AuNaringinNaringin[220]
AuQuercetinQuercetin[221]
AuHesperetinHesperetin[222]
AuQuercetinQuercetin[223]
Fe3O4QuercetinQuercetin[224]
Fe3O4NaringinNaringin[210]

Share and Cite

MDPI and ACS Style

Marslin, G.; Siram, K.; Maqbool, Q.; Selvakesavan, R.K.; Kruszka, D.; Kachlicki, P.; Franklin, G. Secondary Metabolites in the Green Synthesis of Metallic Nanoparticles. Materials 2018, 11, 940. https://doi.org/10.3390/ma11060940

AMA Style

Marslin G, Siram K, Maqbool Q, Selvakesavan RK, Kruszka D, Kachlicki P, Franklin G. Secondary Metabolites in the Green Synthesis of Metallic Nanoparticles. Materials. 2018; 11(6):940. https://doi.org/10.3390/ma11060940

Chicago/Turabian Style

Marslin, Gregory, Karthik Siram, Qaisar Maqbool, Rajendran Kamalabai Selvakesavan, Dariusz Kruszka, Piotr Kachlicki, and Gregory Franklin. 2018. "Secondary Metabolites in the Green Synthesis of Metallic Nanoparticles" Materials 11, no. 6: 940. https://doi.org/10.3390/ma11060940

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