This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
1. Introduction
Phytological origin is the main source for many flavonoids and corresponding flavonoid O-glycosides (FOGs) [1]; these FOGs are proven antioxidant [2], antimicrobial [3], anticancer [4], antiobesity [5], and medicinal agents [6]. The collective advances of FOGs concerning nanotechnology have emerged as a new arena that captivating medicinal researchers to pursue research in it [7, 8]. As acclaimed in vivo oxidation being identified as a vital process that spawns the ample energy for the proper execution of biological processes in all organisms, often, it causes the overproduction of free radicals in turn for the cell damage and in turn for the metabolic diseases like diabetes, cardiovascular diseases, cancers, and neurological disorders [9, 10]. In its counter administration, antioxidants inhibit the reactive free radicals by neutralizing and arrest the structural disruption of biomolecules in cells [11].
During so, the distinguished biosynthesized phytological nanoparticles (NPs) are identified more remarkable than their plant extracts in exhibiting potential activity [12]. This has fascinated nanotechnology in embodiment with scientific results that abridged the gap between with atomic/molecular structures and bulk materials and accelerated the chemotherapeutic potency in treating various diseases [13, 14]. Structurally, the surface-to-volume ratio of NPs is contrarily proportional to their sizes, [15] more precisely the inherent potential of silver nanoparticles (AgNPs) grows reciprocally with an escalation in the specific surface area owing to high surface energy and catalytic reactivity 16. The preference and advancement of green chemistry over conventional are due to eco-friendliness, cost-effectiveness, and feasibility for large-scale synthesis [16, 17]. The extensive array of AgNPs with medicinal efficacies like anticancer, [18, 19], antioxidants [20], and antimicrobial [21] abilities is derived from various plant origins like Rhinacanthus nasutus [22], Trigonella foenum-graecum [23], Ocimum bacillicum [24], Vitex negundo L [25], Hypnea musciformis (Wulfen) JV lamouroux [26], Terminalia chebula [27], Raphanus sativus var. aegyptiacus [28], Citrus sinensis [29], Cassia roxburghii [30], Eurocyma longifolia [31], Annona muricata [32], and Eriobotrya japonica [33].
It is profound that excessive usage of antibiotics results in dissemination and emergence of multidrug-resistant strains of several types of microorganisms [31, 34]. In this scenario, the needs and demands to discover new medicinal agents are increasing, and nanotechnology paves ways to synthesize NPs to substitute current antibiotics and other synthetic agents. In such, Luffa acutangula (LA), a traditional perennial flowering climber plant, ordinarily stated as ridge gourd regards to Cucurbitaceae family and is enriched with medicinal properties [35] like antioxidant, antidiabetic, antiproliferative, antiangiogenic, anticataleptic, analgesic, antiulcer, and antimicrobial activities [36, 37].
As LA plant parts are enriched with a large number of pharmacologically active phytochemicals like flavonoids, proteins, saponin triterpene, anthraquinones, fatty acids, and other phytoconstituents, it is ethnopharmacologically used to treat hemorrhoids, leprosy, splenitis, and ringworm infections by topical administration of pulverized leaves of LA [38]. Hence, we synthesised LAAgNPs from the leaf extract of LA and succeeded in synthesising AgNPs. The flavonoids present in leaves viz., Cosmosioside (1, Apigenin-7-glucoside), Cynaroside (2, Luteolin-7-glucoside) with potential antioxidant, antibacterial, and anticancer activities are FOGs originated from O-glycosidic linkage of (2ξ)-β-D-arabino-Hexopyranose with 7-OH group of Apigenin and O-glycosidic linkage of β-D-glucopyranose with 7-OH group of Luteolin (Figure 1).
[figure omitted; refer to PDF]
In this connection, previous studies showed similar colour changes to form dark brown colour [21, 46, 47]. It was confirmed that concentrations of plant extracts are one of the significant aspects that influence the rate of synthesis of silver nanoparticles. Higher intensity of colour was spotted as the concentrations increased from 1.0, 2.5, and 5.0%. This could be a result of higher content of the biomolecules that reacted as reducing agents in silver reduction process. Uniform results had also been noticed in the leaves of Luffa acutangula in synthesising silver nanoparticles [22].
3.1.2. UV-Vis Spectral Studies of LA Silver Nanoparticles
Time interval to measure the absorption peak was 30-150 sec. At 150 sec, the highest peak was observed for all the different concentrations, and all the high adsorption peaks were in the range of standard adsorption of silver nanoparticles. The spectral peaks were recorded at 417, 432, and 448 nm for different concentrations of biologically synthesised nanoparticles at 1.0, 2.5, and 5.0%, respectively (Figure 3).
[figure omitted; refer to PDF]
There are no qualms that silver nanoparticles achieved the highest peak as cause of Surface Plasmon Resonance (SPR) adsorption band. Free electrons of biologically synthesised silver nanoparticles promote the generation of SPR band through coalescing the vibrations of electrons in resonance with the light wave [48]. The aspects like size and shape of the nanoparticles, type of biomolecules existing in the plant extracts, silver nitrate concentration, and amount of extracts have influenced the SPR banding patterns.
3.1.3. XRD Analysis
X-ray diffraction analysis is an advanced method to figure out the crystalline nature of metallic nanoparticles. As shown in Figure 4, the peaks at
[figure omitted; refer to PDF]
The present data is in accordance with the result reported for the biosynthesised AgNPs from aqueous leaf extracts of Terminalia mellueri, Terminalia catappa, Terminalia bellerica, and Terminalia bentazoe showed high DPPH free radical scavenging activity (more than 80%) compared to leaf extracts in the range of 60%-70% [56]. The values represented are the
3.2.2. ABTS Antioxidant Assay
In the present study, ABTS free scavenging test was analysed on AgNPs and leaf extract of LA. ABTS+· is considered as protonated radical which could readily accept electron from antioxidant compound and transfer its colour from blue to pink which was detected at 734 nm [58]. The leaf extract of LA showed the potential to scavenge the free radicals was found to be 43.8-82.9% at concentrations from 50-300 μg/mL, respectively, whereas biologically synthesised AgNPs showed the activity as 47.9-85.2% at different concentrations from 50-300 μg/mL, respectively (Figure 8). The values represented are the
[figure omitted; refer to PDF]
The present study proved that gram-negative bacteria, E. coli, were more sensitive to the action of biologically synthesised silver nanoparticles compared to gram-positive bacteria. This is in accordance with the result stated by Kim and the coworkers [63]. Literature denoted the inhibitory effects of silver nanoparticles could be associated with characteristics of specific bacterial species. Naturally, gram-positive and gram-positive grouped bacteria have dissimilar membrane structure, especially the difference in thickness of peptidoglycan layer. The mild antibacterial features of synthesised silver nanoparticles in contradiction of gram-positive bacteria could be due to membrane structure [63]. The antibacterial mechanism of action of metallic nanoparticles is still not exactly explained and being unverified. However, several theories and possible mechanism(s) of actions of biologically and chemically synthesised silver nanoparticles have been reported with basic information [64]. The graphic representation (Figure 10) depicts the penetration of silver nanoparticles (AgNPs) into the cell and their different mode of antibacterial mechanisms. The reactivity begins with synthesis of silver nanoparticles using silver nitrate and selected plant extracts.
[figure omitted; refer to PDF]3.4. Anticancer Activity
Plants contain several types of bioactive compounds that are ideally favorable for the drug development in anticancer therapy. Nowadays, researchers found that the plant-based medicines or drugs are safer and cost-effective when compared to the synthetic drugs [65]. LA is one of the herbal plants which belongs to a family of Cucurbitaceae and widely cultivated in Asia, India, Brazil, and USA [66]. Previously, itself isolated five major components of LA, a bioactive component among them named 1,8 dihydroxy-4-methylanthracene 9,10-dione (DHMA) was reported as potential antiproliferative agent against nonsmall cell lung cancer cells (NCI-H460). DHMA showed promising anticancer activities through inhibition of cell growth, generation of reactive oxygen species (ROS), and induction of p53-mediated apoptotic pathway against human nonsmall cell lung cancer cell line (NCI-H460) [67, 68]. Another study reported on the potential anticancer effect of LA on human colon cancer cell line HT29 cells [69]. LA seeds consist of ribosome inactivating proteins which were reported, and the study revealed the potential anticancer activity of luffaculin 1 and luffaculinin in human leukemia K562 cells [70]. Anticancer effects of AgNPs have been demonstrated in various cell models. It observed a dose-dependent cytotoxic effect of biosynthesized AgNPs from Piper longum extract in MCF-7 breast cancer cells [71]. Cytotoxic effects of AgNPs from other plant extracts such as Iresine herbstii and Vitex negundo Linn were demonstrated in HeLa (cervical) and HCT15 (colorectal) cancer cells, respectively [9, 25]. In the present study, LAAgNPs were tested against four human cancer cell lines, MCF-7, MDA-MB-231, DBTRG, and U87. The synthesised silver nanoparticles by LA leaf extract triggered a dose-dependent reduction in the cell proliferation with IC50 values ranging from 35-42 μg/ml (Figure 11). There are several anticancer mechanisms that have been suggested based on previous studies. AgNPs tend to generate reactive oxidative species (ROS) intracellularly that results excess oxidative stress [72]. High oxidative stress inhibits chromosome inhibition and eventually damage cell cycle of tumor cells [73, 74]. Size independent property of AgNPs enhances cytotoxic effect against drug-resistant cancer cells [75]. In addition, cytotoxic effect can be as the result of poor angiogenesis and programmed cell death by AgNPs [76]. Further studies are needed to interpret the anticancer mechanism(s) of the biosynthesized AgNPs.
[figure omitted; refer to PDF]3.5. Molecular Docking Studies
The obtained in vitro antioxidant, antibacterial, and anticancer activity of FOGs have been additionally supported by investigating of ligand-protein binding interactions against the selected enzymatic proteins viz., 3NM8-Chain A (DPPH radical scavenging activity), 1DNU-Chain A (ABTS radical scavenging activity), 5FGK-Chain A (gram-positive bacterial activity), 1AB4-Chain A (gram-negative bacterial activity), 4GBD-Chain A (MCF-7 anticancer activity), 5FI2-Chain C (MDA-MB-231 anticancer activity), 1D5R-Chain A (U87 anticancer activity), and 5TIJ-Chain B (DBTRG anticancer activity); and docking postures and binding interactions were bestowed in Tables 1–12.
Table 1
Potential ligand-protein molecular docking bindings of Cosmosioside (1) with identified proteins.
Cosmosioside (1, Apigenin-7-glucoside, C21H20O10) | 3NM8 (chain A)-oxidoreductase-tyrosinase | 1DNU (chain A)-oxidoreductase-myeloperoxidase-thiocyanate complex |
5FGK (chain A)-A Transferase Enzymatic Protein-CDK8-associated CycC | 1AB4 (chain A)-A Topoisomerase Enzymatic Protein-59 KDA fragment of gyrase A | 4GBD (chain A)–Lyase-adenosine deaminase |
5FI2 (chain C)-Hydrolase Inhibitor Enzymatic Protein -GAC in complex UPGL 00009 inhibitor | 1D5R (chain A)-A Hydrolase Enzymatic Protein-PTEN tumor suppressor | 5TIJ (chain B)-A Lyase Enzymatic Protein-human enolase 2 |
Table 2
Potential ligand-protein molecular docking interactions of Cosmosioside (1) with identified proteins.
Enzymatic protein | Cluster number | Cluster element | BE (KCal/mol) | HB | HBL atoms | HBR atoms | Binding interactions | BL (A°) | BA (°) | H.B. type |
3NM8 (chain A) - oxidoreductase - tyrosinase | 0 | 7 | -8.2618 | 3 | 2 | 3 | Ligand[O(4)-H(10)]-------ASP55(OCOH) | 2.0704 | 54.6126## | Acceptor |
Ligand[O(4)-H(10)]-----ASP55[O(H)CO] | 2.2912 | Acceptor | ||||||||
Ligand[C(2)-O(1)]-----------ARG209(HN) | 2.8998 | — | Donor | |||||||
1DNU (chain A) - oxidoreductase - myeloperoxidase-thiocyanate complex | 30 | 7 | -7.9463 | 2 | 2 | 2 | Ligand[O(7)-H(13)]-----------ASP172(OC) | 2.4855 | — | Acceptor |
Ligand[O(8)-H(18)]----------TYR316(OH) | 2.2322 | — | Acceptor | |||||||
5FGK (chain A) - transferase - CDK8 associated CycC | 0 | 0 | -9.1645 | 3 | 3 | 3 | Ligand[O(4)-H(10)]------ASP103(OCOH) | 1.9270 | — | Acceptor |
Ligand[O(6)-H(12)]------------VAL27(OC) | 2.4636 | — | Acceptor | |||||||
Ligand[O(8)-H(18)]-------GLU66(OCOH) | 2.0588 | — | Acceptor | |||||||
1AB4 (chain A) - topoisomerase -59KDA fragment of gyrase A | 0 | 0 | -8.2294 | 4 | 4 | 4 | Ligand[O(4)-H(10)]-------ASP87(OCOH) | 2.5627 | — | Acceptor |
Ligand[O(7)-H(13)]-----GLN94(OCNH2) | 2.2952 | — | Acceptor | |||||||
Ligand[O(8)-H(18)]------------PHE96(OC) | 1.9022 | — | Acceptor | |||||||
Ligand[H(18)-O(8)]----------GLN267(HN) | 2.1500 | — | Donor | |||||||
4GBD (chain A) - Lyase - adenosine deaminase | 8 | 1 | -7.7955 | 3 | 3 | 3 | Ligand[O(4)-H(10)]-------ASP36(OCOH) | 2.1348 | — | Acceptor |
Ligand[H(13)-O(7)]---------ARG411(HN) | 2.0486 | — | Donor | |||||||
Ligand[C(2)-O(1)]-----------ARG149(HN) | 2.5263 | — | Donor | |||||||
5FI2 (chain C) - hydrolase inhibitor - GAC in complex UPGL 00009 inhibitor | 0 | 0 | -8.8921 | 7 | 6 | 7 | Ligand[O(8)-H(18)]------ASP446(OCOH) | 1.8467 | — | Acceptor |
Ligand[O(4)-H(10)]----------TYR248(OH) | 2.1513 | — | Acceptor | |||||||
Ligand[O(6)-H(12)]----------GLU380(OH) | 1.7125 | — | Acceptor | |||||||
Ligand[H(18)-O(8)]------ASN318(HNOC) | 2.2940 | — | Donor | |||||||
Ligand[C(9)-O(3)-C(13)]-ASN334(HNOC) | 2.6215 | — | Donor | |||||||
Ligand[H(11)-O(5)]----------ASN387(HN) | 2.1575 | 106.6382@ | Donor | |||||||
Ligand[H(11)-O(5)]----------TYR413(HO) | 2.4479 | Donor | ||||||||
1D5R (chain A) - hydrolase - PTEN tumor suppressor | 0 | 0 | -8.7975 | 4 | 4 | 3 | Ligand[O(4)-H(10)]------ASP324(OCOH) | 2.2363 | 102.4531$ | Acceptor |
Ligand[O(6)-H(12)]------ASP324(OCOH) | 2.1290 | Acceptor | ||||||||
Ligand[H(13)-O(7)]----------ARG172(HN) | 2.2084 | — | Donor | |||||||
Ligand[H(19)-O(9)]----------ARG173(HN) | 2.1463 | — | Donor | |||||||
5TIJ (chain B) - lyase - human enolase 2 | 5 | 7 | -7.9044 | 4 | 3 | 3 | Ligand[O(8)-H(18)]------ASP282(OCOH) | 1.7981 | — | Acceptor |
Ligand[O(7)-H(13)]---GLU219(O(H)OC) | 2.5990 | 56.2469# | Acceptor | |||||||
Ligand[O(7)-H(13)]-----GLU219(OCOH) | 1.9638 | Acceptor | ||||||||
69.7145 | ||||||||||
Ligand[O(6)-H(12)]-----GLU219(OCOH) | 1.7856 | Acceptor |
$Ligand H(10)-ASP324(OCOH)-Ligand H(12);
Table 3
Potential ligand-protein molecular docking bindings of Cynaroside (2) with identified proteins.
Cynaroside (2, luteolin-7-glucoside), C21H20O11) | 3NM8 (chain A) - oxidoreductase - tyrosinase | 1DNU (chain A) - oxidoreductase - myeloperoxiase-thiocyanate complex |
5FGK (chain A) - A Transferase Enzymatic Protein - CDK8 associated CycC | 1AB4 (chain A) - A Topoisomerase Enzymatic Protein -59KDA fragment of gyrase A | 4GBD (chain A) – lyase - adenosine deaminase |
5FI2 (chain C) - Hydrolase Inhibitor Enzymatic Protein - GAC in complex UPGL 00009 inhibitor | 1D5R (chain A) - A Hydrolase Enzymatic Protein - PTEN tumor suppressor | 5TIJ (chain B) - A Lyase Enzymatic Protein -human enolase 2 |
Table 4
Potential ligand-protein molecular docking interactions of Cynaroside (2) with identified proteins.
Enzymatic protein | Cluster number | Cluster element | BE (KCal/mol) | HB | HBL atoms | HBR atoms | Binding interactions | BL(A°) | BA (°) | HB type |
3NM8 (chain A) - oxidoreductase - tyrosinase | 19 | 3 | -7.9386 | 2 | 2 | 2 | Ligand[O(7)-H(13)]----ASP324(OCOH) | 2.1516 | — | Acceptor |
Ligand[C(2)-O(1)]--------ARG209(HN) | 2.3601 | — | Donor | |||||||
1DNU (chain A) - oxidoreductase - myeloperoxidase-thiocyanate complex | 0 | 3 | -8.3238 | 3 | 3 | 3 | Ligand[O(10)-H(19)]---ASP324(OCOH) | 1.9449 | — | Acceptor |
Ligand[O(7)-H(13)]---------ARG27(HN) | 2.0962 | — | Acceptor | |||||||
Ligand[O(6)-H(12)]----------ALA24(OC) | 2.2144 | — | Acceptor | |||||||
5FGK (chain A) - transferase - CDK8 associated CycC | 18 | 12 | -8.8514 | 5 | 5 | 5 | Ligand[O(5)-H(11)]----ASP173(OCOH) | 2.0107 | — | Acceptor |
Ligand[C(14)-O(4)]--------ASP173(HN) | 2.2884 | — | Donor | |||||||
Ligand[C(10)-O(7)]----------LYS52(HN) | 2.2874 | — | Donor | |||||||
Ligand[H(14)-O(8)]---------LYS52(HN) | 2.4930 | Donor | ||||||||
Ligand[O(8)-H(14)]---------ASP98(OC) | 2.7157 | Acceptor | ||||||||
1AB4 (chain A) - topoisomerase -59KDA fragment of gyrase A | 1 | 3 | -8.2753 | 7 | 7 | 6 | Ligand[O(5)-H(11)]----ASP113(OCOH) | 2.0095 | 49.2269 | Acceptor |
Ligand[O(7)-H(13)]- --ASP113(OCOH) | 2.6325 | Acceptor | ||||||||
Ligand[O(4)-H(10)]----ASP115(OCOH) | 2.0341 | — | Acceptor | |||||||
Ligand[O(10)-H(19)]-------ASP299(OC) | 2.7050 | — | Acceptor | |||||||
Ligand[C(10)-O(7)]--------LYS270(HN) | 2.0314 | — | Donor | |||||||
Ligand[C(6)-O(2)]---------LYS298(HN) | 2.4233 | — | Donor | |||||||
Ligand[O(9)-H(18)]-------HSD262(OC) | 2.3361 | — | Acceptor | |||||||
4GBD (chain A) – lyase - adenosine deaminase | 4 | 4 | -7.8306 | 4 | 4 | 4 | Ligand[O(10)-H(19)]--ASP215(OCOH) | 2.1032 | — | Acceptor |
Ligand[O(6)-H(12)]------LYS182(OC) | 2.5865 | — | Acceptor | |||||||
Ligand[O(4)-H(10)]--------LYS184(OC) | 1.8875 | — | Acceptor | |||||||
Ligand[H(19)-O(9)]---------ILE189(HN) | 2.2567 | — | Donor | |||||||
5FI2 (Chain C) - hydrolase inhibitor - GAC in complex UPGL 00009 inhibitor | 0 | 0 | -9.1290 | 5 | 5 | 5 | Ligand[O(10)-H(19)]---ASP446(OCOH) | 1.9065 | — | Acceptor |
Ligand[C(11)-O(6)]-------ASN330(HN) | 2.0675 | — | Donor | |||||||
Ligand[O(5)-H(11)]--------CYS462(O’C) | 2.5840 | — | Acceptor | |||||||
Ligand[O(7)-H(13)]---------CYS462(OC) | 1.9953 | — | Acceptor | |||||||
Ligand[O(4)-H(10)]---------SER313(OC) | 2.5769 | — | Acceptor | |||||||
1D5R (chain A) - hydrolase - PTEN tumor suppressor | 0 | 0 | -9.2690 | 7 | 7 | 5 | Ligand[O(4)-H(10)]----ASP324(OCOH) | 1.9343 | 90.6238$ | Acceptor |
Ligand[O(6)-H(12)]----ASP324(OCOH) | 1.8831 | Acceptor | ||||||||
Ligand[C(18)-O(9)]---------LYS183(HN) | 2.4417 | 74.4086# | Donor | |||||||
Ligand[C(17)-O(10)]--------LYS183(HN) | 2.2624 | Donor | ||||||||
Ligand[O(10)-H(19)]--------TYR176(HN) | 2.3215 | — | Acceptor | |||||||
Ligand[C(4)-O(8)]---------ARG173(HN) | 2.2435 | — | Donor | |||||||
Ligand[O(7)-H(13)]--------ARG172(HN) | 2.2234 | — | Donor | |||||||
5TIJ (chain B) -lyase - human enolase 2 | 24 | 1 | -8.7736 | 5 | 4 | 5 | Ligand[O(5)-H(11)]----ASP142(OCOH) | 1.9126 | — | Acceptor |
Ligand[O(7)-H(13)]---------ASP142(OC) | 2.4464 | — | Acceptor | |||||||
Ligand[C(10)-O(7)]---------ASP142(HN) | 2.1296 | 48.5632@ | Donor | |||||||
Ligand[C(10)-O(7)]---------SER141(HN) | 2.3549 | Donor | ||||||||
Ligand[C(14)-O(4)]---------SER432(HO) | 2.2870 | — | Donor |
Table 5
Potential ligand-protein molecular docking bindings of ascorbic acid (3) with identified proteins.
Ascorbic acid (3, hex-1-enofuranos-3-ulose, C6H8O6) | 3NM8 (chain A) - oxidoreductase - tyrosinase | 1DNU (chain A) - oxidoreductase - myeloperoxidase-thiocyanate complex |
Table 6
Potential ligand-protein molecular docking interactions of ascorbic acid (3) with identified proteins.
Enzymatic protein | Cluster number | Cluster element | BE (KCal/mol) | No. of HB | No. of HBL atoms | No. of HBR atoms | Binding interactions | BL (A°) | BA (°) | HB type |
3NM8 (chain A) - oxidoreductase - tyrosinase | 1 | 4 | -6.4837 | 3 | 3 | 3 | Ligand[O(2)-H(4)]---ASP140(C=O) | 2.1328 | — | Acceptor |
Ligand[C-O(1)]--------LYS47(NH) | 2.8116 | — | Donor | |||||||
Ligand[O(5)-H(3)]---PRO219(C=O) | 1.9298 | — | Acceptor | |||||||
1DNU (chain A) - oxidoreductase - myeloperoxidase-thiocyanate complex | 31 | 2 | -6.6112 | 4 | 3 | 3 | Ligand[O(4)-H(6)]------ASP5(O=C) | 2.3826 | 78.7795 | Acceptor |
Ligand[O(4)-H(6)]----ASP5(OCOH) | 1.9709 | Acceptor | ||||||||
61.7009# | ||||||||||
Ligand[O(5)-H(7)]---ASP5(OCOH) | 1.7722 | Acceptor | ||||||||
Ligand[H(7)-O(5)]-----ARG17(NH) | 2.7187 | — | Donor |
Table 7
Potential ligand-protein molecular docking bindings of ampicillin (4) with identified proteins.
Ampicillin (4, aminobenzylpenicillin, C16H19N3O4S) | 5FGK (chain A) - A Transferase Enzymatic Protein - CDK8 associated CycC | 1AB4 (chain A) - A Topoisomerase Enzymatic Protein -59KDA fragment of gyrase A |
Table 8
Potential ligand-protein molecular docking interactions of ampicillin (4) with identified proteins.
Enzymatic protein | Cluster number | Cluster element | BE (KCal/mol) | No. of HB | No. of HBL atoms | No. of HBR atoms | Binding interactions | BL (A°) | BA (°) | HB type |
5FGK (chain A) - transferase - CDK8 associated CycC | 5 | 0 | -8.4767 | 3 | 3 | 3 | Ligand[N(2)-H(17)]--ASP173(OCOH) | 2.1290 | — | Acceptor |
Ligand[C(8)-O(3)]----LYS52(HNCH2) | 2.1803 | — | Donor | |||||||
Ligand[O(1)-H(3)]--------VAL27(OC) | 2.3334 | — | Acceptor | |||||||
1AB4 (chain A) - topoisomerase -59KDA fragment of gyrase A | 1 | 4 | -7.8486 | 3 | 3 | 3 | Ligand[O(1)-H(3)]---ASP104(OCOH) | 2.1401 | — | Acceptor |
Ligand[N(2)-H(18)]--ASP515(OCOH) | 2.4497 | — | Acceptor | |||||||
Ligand[C(5)-O]-----------LYS129(HN) | 2.1821 | — | Donor |
Table 9
Potential ligand-protein molecular docking bindings of tamoxifen (5) with identified proteins.
Tamoxifen (5, 1-p-β-dimethylaminoethoxyphenyl-trans-1,2-diphenylbut-1-ene, C26H29NO) | 4GBD (chain A) – lyase - adenosine deaminase | 5FI2 (chain C) - Hydrolase Inhibitor Enzymatic Protein -GAC in complex UPGL 00009 inhibitor |
Table 10
Potential ligand-protein molecular docking interactions of tamoxifen (5) with identified proteins.
Enzymatic protein | Cluster number | Cluster element | BE (KCal/mol) | HB | HBL atoms | HBR atoms | Binding interactions | BL (A°) | BA (°) | HB type |
4GBD (chain A) - lyase - adenosine deaminase | 0 | 3 | -7.7235 | 3 | 1 | 3 | Ligand[C-N]-------ASP316(OCOH) | 4.2864 | 39.9133 | Acceptor |
Ligand[C-N]----------ASN314(O=C) | 4.4192 | Acceptor | ||||||||
40.7597$ | ||||||||||
Ligand[C-N]----------GLU282(OCOH) | 3.2491 | Acceptor | ||||||||
5FI2 (Chain C) - hydrolase inhibitor - GAC in complex UPGL 00009 inhibitor | 4 | 0 | -7.5313 | 2 | 2 | 2 | Ligand[C(2)Ph-O]----------ASP466(HN) | 4.0253 | 104. 5635# | Donor |
Ligand[C(2)Ph-O]---------ASN318(HN) | 3.5771 | Donor |
Table 11
Potential ligand-protein molecular docking bindings of gefitinib (6) with identified proteins.
Gefitinib (6, N-(3-chloro-4-fluorophenyl)-7-methoxy-6-(3-morpholinopropoxy) quinazolin-4-amine, C22H24ClFN4O3) | 1D5R (chain A) - A Hydrolase Enzymatic Protein - PTEN tumor suppressor | 5TIJ (chain B) - A Lyase Enzymatic Protein - human enolase 2 |
Table 12
Potential ligand-protein molecular docking interactions of gefitinib (6) with identified proteins.
Enzymatic protein | Cluster number | Cluster element | BE (KCal/mol) | HB | HBL atoms | HBR atoms | Binding interactions | BL (A°) | BA (°) | HB type |
1D5R (chain A) - hydrolase - PTEN tumor suppressor | 0 | 0 | -8.0338 | 5 | 3 | 4 | Ligand[N(2)-H(3)]------ASP324(OCOH) | 2.3863 | 54.6314$ | Acceptor |
Ligand[N(2)-H(3)]------ASP324(OCOH) | 2.1188 | Acceptor | ||||||||
Ligand[C(5)-O(2)]-------ARG173(HN=C) | 2.2728 | 71.0798# | Donor | |||||||
Ligand[C(4)-O]---------ARG173(HN=C) | 2.3931 | Donor | ||||||||
50.4066 | ||||||||||
Ligand[C(4)-O]--------ARG173(HN-CH) | 2.8653 | Donor | ||||||||
5TIJ (chain B) -lyase - human enolase 2 | 13 | 3 | -7.8985 | 2 | 1 | 2 | Ligand[C(12)-O(1)]----------ASP142(HN) | 2.4308 | 52.5638$ | Donor |
Ligand[C(12)-O(1)]----------SER141(HN) | 2.4781 | Donor |
$ASP324(HOCO)-Ligand [H(3)]-ASP324(O(H)OC);
In view of antioxidant activity, the hydroxy groups (-OH) of FOGs bound with carbonyl groups (O=C) of aspartic acid and amino groups (-NH) of arginine in 3NM8 (Chain A) are responsible for DPPH radical scavenging activity; and binding of -OH of FOGs bound with O=C of aspartic acid and alanine, -NH of arginine, and -OH of tyrosine in 1DNU (Chain A) is responsible for ABTS radical scavenging activity. Concerning the antibacterial activity, the -OH of FOGs bound with C=O of aspartic acid, valine and glutamic acid, and -NH of aspartic acid and lysine in 5FGK (Chain A) is responsible for gram-positive bacterial activity; and binding of -OH in FOGs with C=O of aspartic acid, glutamine, histidine and phenyl alanine, and -NH of glutamine and lysine in 1AB4 (Chain A) is responsible for gram-negative bacterial activity. In relation to MCF-7 anticancer inhibition, the -OH of FOGs bound with C=O of aspartic acid and lysine, and -NH of arginine and isoleucine in 4GBD (Chain A) were identified as responsible. Coming to MDA-MB-231 anticancer inhibition, the -OH of FOGs bound with C=O of aspartic acid, cysteine and serine, -OH of tyrosine and glutamic acid, and -NH of asparagine in 5FI2 (Chain C) was identified as responsible.
Concerning the U87 anticancer inhibition, the -OH of FOGs bound with C=O of aspartic acid, and -NH of lysine, and tyrosine and arginine in 1D5R (chain A was identified as responsible). In aspects of DBTRG anticancer inhibition, the -OH of FOGs bound with C=O of aspartic acid and glutamic acid, -OH of glutamic acid and serine, and -NH of aspartic acid and serine in 5TIJ (chain B) was identified as responsible. The binding specificity studies have affirmed the promising ligand-protein binding interactions in between the hydroxy groups of the FOGs and aspartic acid of the concerned enzymatic proteins with a binding energy in the range of -9.2690 to -7.7955 KCal/mol.
3.6. ADMET Properties
The study of ADMET properties of the interested analytes under investigation helps to realize their physicochemical interactions [77]. The potentiality of a drug depends on its degree of absorption and in turn on its inherent bioavailability properties. Once a potential drug be absorbed and self-distributed in to muscles and organs by circulation through extracellular sites and hence lowers its plasma concentration individually, therefore, metabolizes in vivo, then, such metabolites will be distributed by the action of reduction and oxidation reactions by the enzymatic action and work potentially on cellular systems, and ultimately the inert metabolites will be automatically excreted from kidneys. Such analysis of ADMET properties (Table 13) inferred us that the two FOGs are with 0.0373 and 0.0336 of BBB penetration potentiality confirms their CNS significance and esteems their superior permeability and their in vivo distribution. Further, it is supported on the ground of the in vitro Caco-2 cell permeability held with 7.2167 and 4.8722 nm/sec, respectively, which enables their robust permeability to bind to plasma proteins and to penetrate in to the BBB system. The in vitro PPB efficiency with 73.43 and 73.27 respective percentages approves their robust binding capability to plasma proteins. The in vitro MDCK cell permeability with 0.6424 and 0.7567 nm/sec empowers their strong permeability. The %HIA with 47.1059 and 25.1651 supports their interactions with targeted domains of the cells. The negative magnitudes of the toxicity calculations that designate FOGs are nontoxic and with safer drug properties. In ultimate, ADMET analysis of the two FOGs has greatly manifested their potential physicochemical interactions and drug-likeness.
Table 13
ADMET properties the compounds 1-6.
Compounds | In vivo blood-brain barrier penetration (C. brain/C. blood)a | In vitro Caco-2 cell permeability | In vitro plasma protein binding | In vitro MDCK cell permeability | Human intestinal absorption (HIA, %)e | Toxicityf |
Apigenin-7-glucoside (1) | 0.0373 | 7.2167 | 73.4332 | 0.6424 | 47.1059 | Negative |
Luteolin-7-glucoside (2) | 0.0336 | 4.8722 | 73.2796 | 0.7567 | 25.1651 | Negative |
Ampicillin (3) | 0.0588 | 0.6307 | 36.1547 | 0.9376 | 81.4785 | Negative |
Ascorbic acid (4) | 0.1173 | 2.4836 | 5.3035 | 0.8819 | 33.1572 | Negative |
Gefitinib (5) | 0.0476 | 54.1474 | 80.7309 | 0.07737 | 96.6375 | Negative |
Tamoxifen (6) | 14.1639 | 49.5448 | 94.7448 | 69.8462 | 100 | Negative |
a
3.7. QSAR Studies
QSAR results (Table 14) indicate that FOGs under study with molecular weights 432.38 and 448.38 (less than 500 Daltons) have confirmed their greater permeability via cell membranes with log
Table 14
QSAR properties of the compounds 1-6.
Entry | Lipinski parameters | Veber parameters | Other parameters | ||||||||||
MW | HB Don | HB | logP (o/w) | MR | Lip. Vio. | TPSA | No. of | Veb. | No. of H | V. Volume | Solubility | ||
Apigenin-7-glucoside (1) | 432.38 | 10 | 6 | 0.68 | 107.46 | 0 | 170.05 | 4 | 0 | 20 | 356.17 | 1.642 | -2.74 |
Luteolin-7-glucoside (2) | 448.38 | 11 | 7 | 0.19 | 109.27 | 0 | 190.28 | 4 | 0 | 20 | 364.19 | 1.713 | -2.45 |
Ampicillin (3) | 349.41 | 7 | 4 | -0.87 | 89.37 | 0 | 112.73 | 4 | 0 | 19 | 298.87 | 1.453 | -1.57 |
Ascorbic acid (4) | 176.12 | 6 | 4 | -1.40 | 36.61 | 0 | 107.22 | 2 | 0 | 8 | 139.71 | 1.954 | -0.35 |
Gefitinib (5) | 446.91 | 7 | 1 | 4.19 | 118.38 | 0 | 68.75 | 8 | 0 | 24 | 385.07 | 1.322 | -5.06 |
Tamoxifen (6) | 371.52 | 2 | 0 | 6.06 | 119.87 | 0 | 12.47 | 8 | 0 | 29 | 376.13 | 1.042 | -4.40 |
MW: molecular weight; HB Don: hydrogen bond donors; HB Acc: hydrogen bond acceptors; logP: octanol to water partition coefficient; MR: molecular refractivity (cm3/mol); Lip Vio: Lipinski violations; TPSA: total polar surface area; No. of RB: number of rotatable bonds; Veb Vio: Veber violations; No. of “H”: number of hydrophobic atoms; V. Volume: Van der Waals volume;
3.8. Bioactivity and Toxicity Risk Studies
The bioactivity and toxicity risk exploration studies of the FOGs have shown their bioactivity properties viz., GPCR ligand property, ion channel modulator, kinase inhibitor, nuclear receptor ligand interactions, protease inhibitor, and enzyme inhibitor interactions; and the drug properties like drug-likeness and drug score and established as potential nontoxic drugs (Table 15). This molinspiration exploration comprehensively assists us to explore the cheminformatics of the molecules under investigation by correlating with the in vitro and in vivo results database of the recognized drugs basing on the functional group similarities in mutual. The drug property exploration of the two FOGs has evidenced for their safer drug properties as they are with no risks of tumorigenicity, irritant effects, mutagenicity, and shown no effect on reproductive system. The positive magnitude of the drug-likeness value represents that the scrutinized molecule comprises the significant fragments that are present in the established commercial drugs [45]. Drug-likeness is an significant factor which helps in understanding the kinesis of a molecule from the site of administration to the bloodstream, hence, its good solubility accounts for good absorption and assures the drug-likeness [80]. Similarly, drug score is also a complementary parameter of the drug-likeness and helps to assure to decide molecule’s drug potentiality. Hence, the present investigation reveals that all the properties of the bioactivity and toxicity risk studies are up to the potential limits of the safe drugs and ascertains the FOGs as the drug-like compounds.
Table 15
Bioactivity scores, drug properties, and toxicity risks of the compounds 1-6.
Compounds | Structure | Bioactivity | Drug properties | ||||||
GPCRL | ICM | KI | NRL | PI | EI | Drug-likeness | Toxicity risks | ||
Apigenin-7-glucoside (1) | 0.10 | -0.01 | 0.14 | 0.31 | 0.02 | 0.43 | 2.29 | 0.44 | |
Luteolin-7-glucoside (2) | 0.09 | -0.02 | 0.15 | 0.27 | -0.01 | 0.42 | 1.79 | 0.45 | |
Ampicillin (3) | 0.04 | -0.47 | -0.71 | -0.61 | 0.87 | 0.25 | 10.72 | 0.91 | |
Ascorbic acid (4) | -0.53 | -0.24 | -1.09 | -1.01 | -0.81 | 0.20 | 0.02 | 0.74 | |
Gefitinib (5) | 0.12 | -0.04 | 0.66 | -0.21 | 0.30 | 0.03 | -2.62 | 0.28 | |
Tamoxifen (6) | 0.30 | 0.002 | -0.01 | 0.57 | 0.04 | 0.32 | 6.3 | 0.35 |
GPCRL: G protein-coupled receptor ligand; ICM: ion channel modulator; KI: kinase inhibitor; NRL: nuclear receptor ligand; PI: protease inhibitor; EI: enzyme inhibitor.
4. Conclusions
Luffa acutangula is one of the regularly used plants with various secondary metabolites such as polyphenols and flavonoids, which possesses biological and pharmacological activities. Here in this study, the aim is to test the biogenically synthesised nanoparticles for their biological activities including antibacterial, antioxidant, and anticancer activities. The results revealed that the silver nanoparticles of Luffa acutangula leaf extract enriched with its inherent flavonoid O-glycosides (FOGs, viz., Cosmosioside (1, Apigenin-7-glucoside) and from -9.2690 to -7.8306 for Cynaroside (2, Luteolin-7-glucoside)) prepared by green biosynthetic approach. The biogenically synthesised silver nanoparticles found to be significant against bacteria and cancer cell lines which clearly show antibacterial and anticancer activities. Antioxidants play an important in reducing the oxidative stress and diminishing the growth of the cancerous cell. The results showed that AgNPs showed potential antioxidant activity. The profound studies performed based on the molecular docking analysis have revealed that the FOGs are identified as antagonists of aspartic acid receptor of enzymatic proteins referenced based on the microorganisms, cell lines, and oxidizing agents considered for the in vitro studies. Furthermore, QSAR, ADMET properties showed them as prospective drugs. The results validated that AgNPs could be potential agents to treat various types of cancers and boosting the immune system functions. Nevertheless, future studies with in vivo toxicological studies with clear mechanism of action and the pharmacodynamics studies of LAAgNPs would shed the light more thoroughly to show the possible mechanisms for anticancer activities.
Acknowledgments
The corresponding author Dr. P. V. R. acknowledges Universiti Malaysia Kelantan and Universiti Sains Malaysia for providing the facilities. This research was funded by Fundamental Research Grant Scheme (FRGS), Malaysia (Grant number: R/FRGS/A07.00/00295A/002/2014/000183), Research Acculturation Collaborative Effort (RACE), Malaysia (Grant number: R/RACE/A07.00/01147A/001/2015/000237), and Transdisciplinary Research University (RUT) of Universiti Sains Malaysia (USM), Malaysia (Grant number: 1001/PPSP/853002). The corresponding author thanks Universiti Malaysia Sabah for providing the funding support.
[1] E. Corradini, P. Foglia, P. Giansanti, R. Gubbiotti, R. Samperi, A. Laganà, "Flavonoids: chemical properties and analytical methodologies of identification and quantitation in foods and plants," Natural Product Research, vol. 25 no. 5, pp. 469-495, DOI: 10.1080/14786419.2010.482054, 2011.
[2] P. G. Pietta, "Flavonoids as antioxidants," Journal of Natural Products, vol. 63 no. 7, pp. 1035-1042, DOI: 10.1021/np9904509, 2000.
[3] T. P. T. Cushnie, A. J. Lamb, "Antimicrobial activity of flavonoids," International Journal of Antimicrobial Agents, vol. 26 no. 5, pp. 343-356, DOI: 10.1016/j.ijantimicag.2005.09.002, 2005.
[4] H. Khan, M. Saeedi, S. M. Nabavi, M. S. Mubarak, A. Bishayee, "Glycosides from medicinal plants as potential anticancer agents: emerging trends towards future drugs," Current Medicinal Chemistry, vol. 26 no. 13, pp. 2389-2406, DOI: 10.2174/0929867325666180403145137, 2019.
[5] J. Y. Park, C. S. Kim, K. M. Park, P. S. Chang, "Inhibitory characteristics of flavonol-3- O -glycosides from Polygonum aviculare L. (common knotgrass) against porcine pancreatic lipase," Scientific Reports, vol. 9 no. 1, article 18080,DOI: 10.1038/s41598-019-54546-8, 2019.
[6] K. Niaz, F. Khan, "Analysis of polyphenolics," Recent Advances in Natural Products Analysis,DOI: 10.1016/B978-0-12-816455-6.00003-2, 2020.
[7] F. J. Osonga, A. Akgul, I. Yazgan, A. Akgul, R. Ontman, V. M. Kariuki, G. B. Eshun, O. A. Sadik, "Flavonoid-derived anisotropic silver nanoparticles inhibit growth and change the expression of virulence genes inEscherichia coliSM10," RSC Advances, vol. 8 no. 9, pp. 4649-4661, DOI: 10.1039/C7RA13480K, 2018.
[8] J. Kurepa, R. Nakabayashi, T. Paunesku, M. Suzuki, K. Saito, G. E. Woloschak, J. A. Smalle, "Direct isolation of flavonoids from plants using ultra-small anataseTiO2nanoparticles," The Plant Journal, vol. 77 no. 3, pp. 443-453, DOI: 10.1111/tpj.12361, 2014.
[9] C. Dipankar, S. Murugan, "The green synthesis, characterization and evaluation of the biological activities of silver nanoparticles synthesized from Iresine herbstii leaf aqueous extracts," Colloids and Surfaces B: Biointerfaces, vol. 98 no. 9, pp. 112-119, DOI: 10.1016/j.colsurfb.2012.04.006, 2012.
[10] P. V. Rao, D. Nallappan, K. Madhavi, S. Rahman, L. Jun Wei, S. H. Gan, "Phytochemicals and biogenic metallic nanoparticles as anticancer agents," Oxidative Medicine and Cellular Longevity, vol. 2016,DOI: 10.1155/2016/3685671, 2016.
[11] C. A. Santos, F. A. Almeida, B. X. V. Quecán, P. A. P. Pereira, K. M. B. Gandra, L. R. Cunha, U. M. Pinto, "Bioactive properties of Syzygium cumini (L.) skeels pulp and seed phenolic extracts," Frontiers in Microbiology, vol. 11,DOI: 10.3389/fmicb.2020.00990, 2020.
[12] A. Singh, P. K. Gautam, A. Verma, V. Singh, P. M. Shivapriya, S. Shivalkar, A. K. Sahoo, S. K. Samanta, "Green synthesis of metallic nanoparticles as effective alternatives to treat antibiotics resistant bacterial infections: a review," Biotechnology Reports, vol. 25, article e00427,DOI: 10.1016/j.btre.2020.e00427, 2020.
[13] M. R. Siddiqui, M. Khan, Khan, Adil, Tahir, W. Tremel, Khathlan, Warthan, "Green synthesis of silver nanoparticles mediated by Pulicaria glutinosa extract," International Journal of Nanomedicine, vol. 8 no. 1, pp. 1507-1616, DOI: 10.2147/IJN.S43309, 2013.
[14] G. Z. Kyzas, K. A. Matis, "Nanoadsorbents for pollutants removal: a review," Journal of Molecular Liquids, vol. 203, pp. 159-168, DOI: 10.1016/j.molliq.2015.01.004, 2015.
[15] P. J. Shiny, A. Mukherjee, N. Chandrasekaran, "DNA damage and mitochondria-mediated apoptosis of A549 lung carcinoma cells induced by biosynthesised silver and platinum nanoparticles," RSC Advances, vol. 6 no. 33, pp. 27775-27787, DOI: 10.1039/C5RA27185A, 2016.
[16] S. Das, A. Das, A. Maji, M. Beg, A. Singha, M. Hossain, "A compact study on impact of multiplicative Streblus asper inspired biogenic silver nanoparticles as effective photocatalyst, good antibacterial agent and interplay upon interaction with human serum albumin," Journal of Molecular Liquids, vol. 259, pp. 18-29, DOI: 10.1016/j.molliq.2018.02.111, 2018.
[17] A. Niaz, A. Bibi, Huma, M. I. Zaman, M. Khan, A. Rahim, "Highly selective and ecofriendly colorimetric method for the detection of iodide using green tea synthesized silver nanoparticles," Journal of Molecular Liquids, vol. 249, pp. 1047-1051, DOI: 10.1016/j.molliq.2017.11.151, 2018.
[18] L. Wei, J. Lu, H. Xu, A. Patel, Z. S. Chen, G. Chen, "Silver nanoparticles: synthesis, properties, and therapeutic applications," Drug Discovery Today, vol. 20 no. 5, pp. 595-601, DOI: 10.1016/j.drudis.2014.11.014, 2015.
[19] D. Ghosh, S. Dhibar, A. Dey, S. Mukherjee, N. Joardar, S. P. S. Babu, B. Dey, "Graphene oxide dispersed supramolecular hydrogel capped benign green silver nanoparticles for anticancer, antimicrobial, cell attachment and intracellular imaging applications," Journal of Molecular Liquids, vol. 282,DOI: 10.1016/j.molliq.2019.03.010, 2019.
[20] M. S. Abdel-Aziz, M. S. Shaheen, A. A. el-Nekeety, M. A. Abdel-Wahhab, "Antioxidant and antibacterial activity of silver nanoparticles biosynthesized using _Chenopodium murale_ leaf extract," Journal of Saudi Chemical Society, vol. 18 no. 4, pp. 356-363, DOI: 10.1016/j.jscs.2013.09.011, 2014.
[21] P. Banerjee, M. Satapathy, A. Mukhopahayay, P. Das, "Leaf extract mediated green synthesis of silver nanoparticles from widely available Indian plants: synthesis, characterization, antimicrobial property and toxicity analysis," Bioresources and Bioprocessing, vol. 1 no. 1, article 3,DOI: 10.1186/s40643-014-0003-y, 2014.
[22] V. R. Pasupuleti, T. N. V. K. V. Prasad, R. A. Sheikh, S. K. Balam, G. Narasimhulu, S. H. Gan, C. Reddy, I. Rahman, "Biogenic silver nanoparticles using Rhinacanthus nasutus leaf extract: synthesis, spectral analysis, and antimicrobial studies," International Journal of Nanomedicine, vol. 8 no. 1, pp. 3355-3564, DOI: 10.2147/IJN.S49000, 2013.
[23] V. K. Vidhu, D. Philip, "Catalytic degradation of organic dyes using biosynthesized silver nanoparticles," Micron, vol. 56, pp. 54-62, DOI: 10.1016/j.micron.2013.10.006, 2014.
[24] S. Pirtarighat, M. Ghannadnia, S. Baghshahi, "Biosynthesis of silver nanoparticles using _Ocimum basilicum_ cultured under controlled conditions for bactericidal application," Materials Science and Engineering: C Materials for Biological Applications, vol. 98, pp. 250-255, DOI: 10.1016/j.msec.2018.12.090, 2019.
[25] D. Prabhu, C. Arulvasu, G. Babu, R. Manikandan, P. Srinivasan, "Biologically synthesized green silver nanoparticles from leaf extract of Vitex negundo L. induce growth-inhibitory effect on human colon cancer cell line HCT15," Process Biochemistry, vol. 48 no. 2, pp. 317-324, DOI: 10.1016/j.procbio.2012.12.013, 2013.
[26] G. Ganapathy Selvam, K. Sivakumar, "Phycosynthesis of silver nanoparticles and photocatalytic degradation of methyl orange dye using silver (Ag) nanoparticles synthesized from Hypnea musciformis (Wulfen) J.V. Lamouroux," Applied Nanoscience, vol. 5 no. 5, pp. 617-622, DOI: 10.1007/s13204-014-0356-8, 2015.
[27] K. Mohan Kumar, M. Sinha, B. K. Mandal, A. R. Ghosh, K. Siva Kumar, P. Sreedhara Reddy, "Green synthesis of silver nanoparticles using Terminalia chebula extract at room temperature and their antimicrobial studies," Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, vol. 91, pp. 228-233, DOI: 10.1016/j.saa.2012.02.001, 2012.
[28] S. M. Ali, N. M. H. Yousef, N. A. Nafady, "Application of biosynthesized silver nanoparticles for the control of land snail eobania vermiculata and some plant pathogenic fungi," Journal of Nanomaterials, vol. 2015,DOI: 10.1155/2015/218904, 2015.
[29] S. Kaviya, J. Santhanalakshmi, B. Viswanathan, J. Muthumary, K. Srinivasan, "Biosynthesis of silver nanoparticles using citrus sinensis peel extract and its antibacterial activity," Spectrochimica Acta, Part A: Molecular and Biomolecular Spectroscopy, vol. 79 no. 3, pp. 594-598, DOI: 10.1016/j.saa.2011.03.040, 2011.
[30] P. Moteriya, H. Padalia, S. Chanda, "Characterization, synergistic antibacterial and free radical scavenging efficacy of silver nanoparticles synthesized using Cassia roxburghii leaf extract," Journal of Genetic Engineering and Biotechnology, vol. 15 no. 2, pp. 505-513, DOI: 10.1016/j.jgeb.2017.06.010, 2017.
[31] D. Nallappan, N. V. K. V. P. Tollamadugu, A. N. Fauzi, N. S. Yaacob, V. R. Pasupuleti, "Biomimetic synthesis and anticancer activity ofEurycoma longifoliabranch extract-mediated silver nanoparticles," IET Nanobiotechnology, vol. 11 no. 7, pp. 889-897, DOI: 10.1049/iet-nbt.2016.0181, 2017.
[32] M. S. Jabir, Y. M. Saleh, G. M. Sulaiman, N. Y. Yaseen, U. I. Sahib, Y. H. Dewir, M. S. Alwahibi, D. A. Soliman, "Green synthesis of silver nanoparticles using Annona muricata extract as an inducer of apoptosis in cancer cells and inhibitor for NLRP3 inflammasome via enhanced autophagy," Nanomaterials, vol. 11 no. 2, article 384,DOI: 10.3390/nano11020384, 2021.
[33] M. S. Jabir, A. A. Hussien, G. M. Sulaiman, N. Y. Yaseen, Y. H. Dewir, M. S. Alwahibi, D. A. Soliman, H. Rizwana, "Green synthesis of silver nanoparticles fromEriobotrya japonicaextract: a promising approach against cancer cells proliferation, inflammation, allergic disorders and phagocytosis induction," Artificial Cells, Nanomedicine, and Biotechnology, vol. 49 no. 1, pp. 48-60, DOI: 10.1080/21691401.2020.1867152, 2021.
[34] H. Harbottle, S. Thakur, S. Zhao, D. G. White, "Genetics of antimicrobial resistance," Animal Biotechnology, vol. 17 no. 2, pp. 111-124, DOI: 10.1080/10495390600957092, 2006.
[35] M. J. Firdhouse, P. Lalitha, "Biosynthesis of silver nanoparticles and its applications," Journal of Nanotechnology, vol. 2015,DOI: 10.1155/2015/829526, 2015.
[36] B. P. Pimple, P. V. Kadam, M. J. Patil, "Protective effect of Luffa acutangula extracts on gastric ulceration in NIDDM rats: Role of gastric mucosal glycoproteins and antioxidants," Asian Pacific Journal of Tropical Medicine, vol. 5 no. 8, pp. 610-615, DOI: 10.1016/s1995-7645(12)60126-6, 2012.
[37] J. Taruna, J. B. Kaushal, J. Bhatti, P. Kumar, "Green synthesis and physico-chemical study of silver nanoparticles extracted from a natural source Luffa acutangula," Journal of Molecular Liquids, vol. 224 no. A, pp. 991-998, DOI: 10.1016/j.molliq.2016.10.065, 2016.
[38] P. N. Shendge, S. Belemkar, "Therapeutic potential of Luffa acutangula: a review on its traditional uses, phytochemistry, pharmacology and toxicological aspects," Frontiers in Pharmacology, vol. 9, article 1177,DOI: 10.3389/fphar.2018.01177, 2018.
[39] July 2021, http://www.swissdock.ch/docking
[40] E. F. Pettersen, T. D. Goddard, C. C. Huang, G. S. Couch, D. M. Greenblatt, E. C. Meng, T. E. Ferrin, "UCSF Chimera - a visualization system for exploratory research and analysis," Journal of Computational Chemistry, vol. 25 no. 13, pp. 1605-1612, DOI: 10.1002/jcc.20084, 2004.
[41] M. A. Bakht, M. S. Yar, S. G. Abdel-Hamid, S. I. al Qasoumi, A. Samad, "Molecular properties prediction, synthesis and antimicrobial activity of some newer oxadiazole derivatives," European Journal of Medicinal Chemistry, vol. 45 no. 12, pp. 5862-5869, DOI: 10.1016/j.ejmech.2010.07.069, 2010.
[42] July 2021, http://preadmet.bmdrc.kr/
[43] C. A. Lipinski, F. Lombardo, B. W. Dominy, P. J. Feeney, "Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings 1," Advanced Drug Delivery Reviews, vol. 46 no. 1-3,DOI: 10.1016/S0169-409X(00)00129-0, 2001.
[44] July 2021, http://www.molinspiration.com/cgi-bin/properties
[45] July 2021, https://www.organic-chemistry.org/prog/peo/druglikeness.html
[46] S. Ahmed, M. Saifullah, M. Ahmad, B. L. Swami, S. Ikram, "Green synthesis of silver nanoparticles usingAzadirachta indicaaqueous leaf extract," Journal of Radiation Research and Applied Sciences, vol. 9 no. 1,DOI: 10.1016/j.jrras.2015.06.006, 2016.
[47] H. Padalia, P. Moteriya, S. Chanda, "Green synthesis of silver nanoparticles from marigold flower and its synergistic antimicrobial potential," Arabian Journal of Chemistry, vol. 8 no. 5, pp. 732-741, DOI: 10.1016/j.arabjc.2014.11.015, 2015.
[48] N. G. Mlalila, H. S. Swai, A. Hilonga, D. Kadam, "Antimicrobial dependence of silver nanoparticles on surface plasmon resonance bands against Escherichia coli," Nanotechnology, Science and Applications, vol. Volume 10,DOI: 10.2147/NSA.S123681, 2017.
[49] A. M. Awwad, N. M. Salem, A. O. Abdeen, "Green synthesis of silver nanoparticles using carob leaf extract and its antibacterial activity," International Journal of Industrial Chemistry, vol. 4 no. 1,DOI: 10.1186/2228-5547-4-29, 2013.
[50] M. M. H. Khalil, E. H. Ismail, K. Z. el-Baghdady, D. Mohamed, "Green synthesis of silver nanoparticles using olive leaf extract and its antibacterial activity," Arabian Journal of Chemistry, vol. 7 no. 6, pp. 1131-1139, DOI: 10.1016/j.arabjc.2013.04.007, 2014.
[51] C. Ramteke, T. Chakrabarti, B. K. Sarangi, R. A. Pandey, "Synthesis of silver nanoparticles from the aqueous extract of leaves of Ocimum sanctum for enhanced antibacterial activity," Journal of Chemistry, vol. 2013,DOI: 10.1155/2013/278925, 2013.
[52] P. Kumar, M. Govindaraju, S. Senthamilselvi, K. Premkumar, "Photocatalytic degradation of methyl orange dye using silver (Ag) nanoparticles synthesized from Ulva lactuca," Colloids and Surfaces B: Biointerfaces, vol. 103, pp. 658-661, DOI: 10.1016/j.colsurfb.2012.11.022, 2013.
[53] T. Nagao, R. Tanaka, Y. Iwase, H. Hanazono, H. Okabe, "Studies on the constituents of Luffa acutangula Roxb. I. Structures of acutosides A--G, oleanane-type triterpene saponins isolated from the herb," Chemical and Pharmaceutical Bulletin, vol. 39 no. 3, pp. 599-606, DOI: 10.1248/cpb.39.599, 1991.
[54] M. V. K. Reddy, K. Y. Rao, G. Anusha, G. M. Kumar, A. G. Damu, K. R. Reddy, N. P. Shetti, T. M. Aminabhavi, P. V. G. Reddy, "In-vitro evaluation of antioxidant and anticholinesterase activities of novel pyridine, quinoxaline and s -triazine derivatives," Environmental Research, vol. 199, article 111320,DOI: 10.1016/j.envres.2021.111320, 2021.
[55] N. Chandrasekhar, S. P. Vinay, "Yellow colored blooms of Argemone mexicana and Turnera ulmifolia mediated synthesis of silver nanoparticles and study of their antibacterial and antioxidant activity," Applied Nanoscience, vol. 7 no. 8, pp. 851-861, DOI: 10.1007/s13204-017-0624-5, 2017.
[56] H. Mohamed el-Rafie, M. Abdel-Aziz Hamed, "Antioxidant and anti-inflammatory activities of silver nanoparticles biosynthesized from aqueous leaves extracts of fourTerminaliaspecies," Advances in Natural Sciences: Nanoscience and Nanotechnology, vol. 5 no. 3, article 035008,DOI: 10.1088/2043-6262/5/3/035008, 2014.
[57] D. Bose, S. Chatterjee, "Biogenic synthesis of silver nanoparticles using guava (Psidium guajava) leaf extract and its antibacterial activity against Pseudomonas aeruginosa," Applied Nanoscience, vol. 6 no. 6, pp. 895-901, DOI: 10.1007/s13204-015-0496-5, 2016.
[58] M. Gangwar, M. K. Gautam, A. K. Sharma, Y. B. Tripathi, R. K. Goel, G. Nath, "Antioxidant capacity and radical scavenging effect of polyphenol rich Mallotus philippenensis fruit extract on human erythrocytes: an in vitro study," Scientific World Journal, vol. 2014, article 279451,DOI: 10.1155/2014/279451, 2014.
[59] P. Dauthal, M. Mukhopadhyay, "In-vitro free radical scavenging activity of biosynthesized gold and silver nanoparticles using Prunus armeniaca (apricot) fruit extract," The Journal of Nanoparticle Research, vol. 15 no. 1, article 1366,DOI: 10.1007/s11051-012-1366-7, 2013.
[60] A. K. Mittal, J. Bhaumik, S. Kumar, U. C. Banerjee, "Biosynthesis of silver nanoparticles: elucidation of prospective mechanism and therapeutic potential," Journal of Colloid and Interface Science, vol. 415, pp. 39-47, DOI: 10.1016/j.jcis.2013.10.018, 2014.
[61] R. Geethalakshmi, D. V. Sarada, "Gold and silver nanoparticles from Trianthema decandra: synthesis, characterization, and antimicrobial properties," International Journal of Nanomedicine, vol. 7, pp. 5375-5384, DOI: 10.2147/ijn.s36516, 2012.
[62] F. C. Tenover, "Mechanisms of antimicrobial resistance in bacteria," The American Journal of Medicine, vol. 119 no. 6, pp. S3-S10, DOI: 10.1016/j.amjmed.2006.03.011, 2006.
[63] J. S. Kim, E. Kuk, K. N. Yu, J. H. Kim, S. J. Park, H. J. Lee, S. H. Kim, Y. K. Park, Y. H. Park, C. Y. Hwang, Y. K. Kim, Y. S. Lee, D. H. Jeong, M. H. Cho, "Antimicrobial effects of silver nanoparticles," Nanomedicine: Nanotechnology, Biology and Medicine, vol. 3 no. 1, pp. 95-101, DOI: 10.1016/j.nano.2006.12.001, 2007.
[64] H. Soliman, A. Elsayed, A. Dyaa, "Antimicrobial activity of silver nanoparticles biosynthesised byRhodotorulasp.strain ATL72," Egyptian Journal of Basic and Applied Sciences, vol. 5 no. 3, pp. 228-233, DOI: 10.1016/j.ejbas.2018.05.005, 2018.
[65] G. M. Sulaiman, E. H. Ali, I. I. Jabbar, A. H. Saleem, "Synthesis, characterization, antibacterial and cytotoxic effects of silver nanoparticles," Digest Journal of Nanomaterials and Biostructures, vol. 9 no. 2, pp. 787-796, 2014.
[66] G. Chomicki, H. Schaefer, S. S. Renner, "Origin and domestication of Cucurbitaceae crops: insights from phylogenies, genomics and archaeology," New Phytologist, vol. 226 no. 5, pp. 1240-1255, DOI: 10.1111/nph.16015, 2020.
[67] R. Vanajothi, P. Srinivasan, "An anthraquinone derivative fromLuffa acutangulainduces apoptosis in human lung cancer cell line NCI-H460 through p53-dependent pathway," Journal of Receptor and Signal Transduction Research, vol. 36 no. 3, pp. 292-302, DOI: 10.3109/10799893.2015.1108335, 2016.
[68] R. Vanajothi, P. Srinivasan, "Bioassay-guided isolation and identification of bioactive compound from aerial parts ofLuffa acutangulaagainst lung cancer cell line NCI-H460," Journal of Receptor and Signal Transduction Research, vol. 35 no. 4, pp. 295-302, DOI: 10.3109/10799893.2014.977451, 2015.
[69] M. Mahadi, A. Jamil, H. B. Mamman, S. Ganeson, "Investigation of anti-cancer proliferation properties of Luffa acutangula on HT-29 using electroporation method," 8th International Conference on Biomedical Engineering and Technology (ICBET) Bali Indonesia, pp. 108-111, DOI: 10.1145/3208955.3208968, .
[70] L. Junkai, M. Chen, X. Jieming, Z. Rong, Y. Xiaoming, S. Xiaoli, W. Ziren, "Purification and characterization of two luffaculins, ribosome-inactivating proteins from seeds of Luffa acutangula," Chinese Journal of Biochemistry and Molecular Biology, vol. 18 no. 5, pp. 609-613, 2002.
[71] K. Venugopal, H. A. Rather, K. Rajagopal, M. P. Shanthi, K. Sheriff, M. Illiyas, R. A. Rather, E. Manikandan, S. Uvarajan, M. Bhaskar, M. Maaza, "Synthesis of silver nanoparticles (Ag NPs) for anticancer activities (MCF 7 breast and A549 lung cell lines) of the crude extract of Syzygium aromaticum," Journal of Photochemistry and Photobiology B: Biology, vol. 167, pp. 282-289, DOI: 10.1016/j.jphotobiol.2016.12.013, 2017.
[72] Z. Yu, Q. Li, J. Wang, Y. Yu, Y. Wang, Q. Zhou, P. Li, "Reactive oxygen species-related nanoparticle toxicity in the biomedical field," Nanoscale Research Letters, vol. 15 no. 1,DOI: 10.1186/s11671-020-03344-7, 2020.
[73] P. D. Nallathamby, X. H. N. Xu, "Study of cytotoxic and therapeutic effects of stable and purified silver nanoparticles on tumor cells," Nanoscale, vol. 2 no. 6, pp. 942-952, DOI: 10.1039/C0NR00080A, 2010.
[74] M. I. Sriram, S. B. M. Kanth, K. Kalishwaralal, "Antitumor activity of silver nanoparticles in Dalton’s lymphoma ascites tumor model," International Journal of Nanomedicine, vol. 5 no. 5, pp. 753-762, DOI: 10.2147/ijn.s11727, 2010.
[75] J. Liu, Y. Zhao, Q. Guo, Z. Wang, H. Wang, Y. Yang, Y. Huang, "TAT-modified nanosilver for combating multidrug-resistant cancer," Biomaterials, vol. 33 no. 26, pp. 6155-6161, DOI: 10.1016/j.biomaterials.2012.05.035, 2012.
[76] P. Sanpui, A. Chattopadhyay, S. S. Ghosh, "Induction of apoptosis in cancer cells at low silver nanoparticle concentrations using chitosan nanocarrier," ACS Applied Materials & Interfaces, vol. 3 no. 2, pp. 218-228, DOI: 10.1021/am100840c, 2011.
[77] T. I. Oprea, H. Matter, "Integrating virtual screening in lead discovery," Current Opinion in Chemical Biology, vol. 8 no. 4, pp. 349-358, DOI: 10.1016/j.cbpa.2004.06.008, 2004.
[78] P. Ertl, B. Rohde, P. Selzer, "Fast calculation of molecular polar surface area as a sum of fragment-based contributions and its application to the prediction of drug transport properties," Journal of Medicinal Chemistry, vol. 43 no. 20, pp. 3714-3717, DOI: 10.1021/jm000942e, 2000.
[79] M. Maximo da Silva, M. Comin, T. Santos Duarte, M. Foglio, J. de Carvalho, M. do Carmo Vieira, A. Nazari Formagio, "Synthesis, antiproliferative activity and molecular properties predictions of galloyl derivatives," Molecules, vol. 20 no. 4, pp. 5360-5373, DOI: 10.3390/molecules20045360, 2015.
[80] A. M. Alafeefy, S. I. Alqasoumi, A. E. Ashour, V. Masand, N. A. al-Jaber, T. Ben Hadda, M. A. Mohamed, "Quinazoline-tyrphostin as a new class of antitumor agents, molecular properties prediction, synthesis and biological testing," European Journal of Medicinal Chemistry, vol. 53, pp. 133-140, DOI: 10.1016/j.ejmech.2012.03.044, 2012.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2021 Devi Nallappan et al. This is an open access article distributed under the Creative Commons Attribution License (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License. https://creativecommons.org/licenses/by/4.0/
Abstract
Studies on green biosynthesis of newly engineered nanoparticles for their prominent medicinal applications are being the torch-bearing concerns of the state-of-the-art research strategies. In this concern, we have engineered the biosynthesized Luffa acutangula silver nanoparticles of flavonoid O-glycosides in the anisotropic form isolated from aqueous leave extracts of Luffa acutangula, a popular traditional and ayurvedic plant in south-east Asian countries. These were structurally confirmed by Ultraviolet-visible (UV-Vis), Fourier transform infrared spectroscopy accessed with attenuated total reflection (FTIR-ATR) spectral analyses followed by the scanning electron microscopic (SEM) and the X-ray diffraction (XRD) crystallographic studies and found them with the face-centered cubic (fcc) structure. Medicinally, we have explored their significant antioxidant (DPPH and ABTS assays), antibacterial (disc diffusion assay on E. coli, S. aureus, B. subtilis, S. fecilis, and S. boydii), and anticancer (MTT assay on MCF-7, MDA-MB-231, U87, and DBTRG cell lines) potentialities which augmented the present investigation. The molecular docking analysis of title compounds against 3NM8 (DPPH) and 1DNU (ABTS) proteins for antioxidant activity; 5FGK (Gram-Positive Bacteria) and 1AB4 (Gram-Negative Bacteria) proteins for antibacterial activity; and 4GBD (MCF-7), 5FI2 (MDA-MB-231), 1D5R (U87), and 5TIJ (DBTRG) proteins for anticancer activity has affirmed the promising ligand-protein binding interactions among the hydroxy groups of the title compounds and aspartic acid of the concerned enzymatic proteins. The binding energy varying from -9.1645 to -7.7955 for Cosmosioside (1, Apigenin-7-glucoside) and from -9.2690 to -7.8306 for Cynaroside (2, Luteolin-7-glucoside) implies the isolated compounds as potential bioactive compounds. In addition, the performed studies like QSAR, ADMET, bioactivity properties, drug scores, and toxicity risks confirmed them as potential drug candidates and aspartic acid receptor antagonists. This research auxiliary augmented the existing array of phytological nanomedicines with new drug candidates that are credible with multiple bioactivities.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details









1 Bioindustrial Technology Program, Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Campus Jeli, 17600 Jeli, Malaysia; Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
2 Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
3 Department of Chemistry, Sri Venkateswara University, -517502, Tirupati, Andhra Pradesh, India
4 Department of Biochemistry, Sri Kadiri Babu Rao College of Agriculture, C.S. Puram, Andhra Pradesh-523112, India
5 Centre for Chemical Sciences and Technology, Institute of Science & Technology, Jawaharlal Nehru Technological University Hyderabad, -500085, Hyderabad, Telangana, India
6 Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha-61421, Saudi Arabia
7 Bioindustrial Technology Program, Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Campus Jeli, 17600 Jeli, Malaysia; Department of Biomedical Science and Therapeutics, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia; Department of Biochemistry, Faculty of Medicine and Health Sciences, Abdurrab University, Jl Riau Ujung No. 73, Pekanbaru, 28292 Riau, Indonesia; Centre for Excellence in Biomaterials Engineering (CoEBE), AIMST University, 08100 Bedong, Kedah, Malaysia