Skip to main content
Erschienen in: BMC Complementary Medicine and Therapies 1/2024

Open Access 01.12.2024 | Research

Secondary metabolic profiling, antioxidant potential, enzyme inhibitory activities and in silico and ADME studies: a multifunctional approach to reveal medicinal and industrial potential of Tanacetum falconeri

verfasst von: Muhammad Imran Tousif, Zaheer Abbas, Mamona Nazir, Muhammad Saleem, Saba Tauseef, Areeba Hassan, Sajid Ali, Maqsood Ahmed, Jallat Khan, Gokhan Zengin, Abeer Hashem, Khalid F. Almutairi, Graciela Dolores Avila-Quezada, Elsayed Fathi Abd-Allah

Erschienen in: BMC Complementary Medicine and Therapies | Ausgabe 1/2024

Abstract

Tanacetum falconeri is a significant flowering plant that possesses cytotoxic, insecticidal, antibacterial, and phytotoxic properties. Its chemodiversity and bioactivities, however, have not been thoroughly investigated. In this work, several extracts from various parts of T. falconeri were assessed for their chemical profile, antioxidant activity, and potential for enzyme inhibition. The total phenolic contents of T. falconeri varied from 40.28 ± 0.47 mg GAE/g to 11.92 ± 0.22 mg GAE/g in various extracts, while flavonoid contents were found highest in TFFM (36.79 ± 0.36 mg QE/g extract) and lowest (11.08 ± 0.22 mg QE/g extract) in TFSC (chloroform extract of stem) in similar pattern as found in total phenolic contents. Highest DPPH inhibition was observed for TFFC (49.58 ± 0.11 mg TE/g extract) and TFSM (46.33 ± 0.10 mg TE/g extract), whereas, TFSM was also potentially active against (98.95 ± 0.57 mg TE/g) ABTS radical. In addition, TFSM was also most active in metal reducing assays: CUPRAC (151.76 ± 1.59 mg TE/g extract) and FRAP (101.30 ± 0.32 mg TE/g extract). In phosphomolybdenum assay, the highest activity was found for TFFE (1.71 ± 0.03 mg TE/g extract), TFSM (1.64 ± 0.035 mg TE/g extract), TFSH (1.60 ± 0.033 mg TE/g extract) and TFFH (1.58 ± 0.08 mg TE/g extract), while highest metal chelating activity was recorded for TFSH (25.93 ± 0.79 mg EDTAE/g extract), TFSE (22.90 ± 1.12 mg EDTAE/g extract) and TFSC (19.31 ± 0.50 mg EDTAE/g extract). In biological screening, all extracts had stronger inhibitory capacity against AChE while in case of BChE the chloroform extract of flower (TFFC) and stem (TFSC) showed the highest activities with inhibitory values of 2.57 ± 0.24 and 2.10 ± 0.18 respectively. Similarly, TFFC and TFSC had stronger inhibitory capacity (1.09 ± 0.015 and 1.08 ± 0.002 mmol ACAE/g extract) against α-Amylase and (0.50 ± 0.02 and 0.55 ± 0.02 mmol ACAE/g extract) α-Glucosidase. UHPLC-MS study of methanolic extract revealed the presence of 133 components including sterols, triterpenes, flavonoids, alkaloids, and coumarins. The total phenolic contents were substantially linked with all antioxidant assays in multivariate analysis. These findings were validated by docking investigations, which revealed that the selected compounds exhibited high binding free energy with the enzymes tested. Finally, it was found that T. falconeri is a viable industrial crop with potential use in the production of functional goods and nutraceuticals.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Tanacetum falconeri is an important flowering herb belong to plant family Asteraceae [1]. It is mostly found in the rocky talus, near the lakes, valley plains or grassy ridges in different parts of Pakistan. Locally, the powdered leaves and extract of leaves of T. falconeri is used against various abdominal problems, whereas, its flowers and buds are beneficial in treating asthma, jaundice and blood pressure problems [2, 3]. Different plant parts are utilised to treat joint discomfort after being dried in the shade [4]. The habitants of Kallaway Indians and the Andes mountains used these plants for back ache, abdominal pain and gastric trouble [5]. The Mexican people used it as a tonic to regulate menstruation and as an antispasmodic. In Venezuela, it's used to cure earaches [6]. A scanty work on chemodiversity and biological potential of T. falconeri has been reported in literature, however, other Tanacetum plants are rich in terpenes mostly as essential oils [713], sterols [1416], phenolic acids and flavonoids [17]. Due to the presence of variety of bioactive compounds, Tanacetum plants extracts have shown various biological activities like anti-inflammatory, antiviral, antifungal, antibacterial and antioxidant [18], edema [19], antibacterial [14, 20], fungicidal activity [21], antioxidant [22], anti-inflammatory [23], anthelmintic, Anticoagulant and antifibrinolytic, insecticidal [14], and anti-ulcer [24, 25] and antitumor [26]. Tanacetum plants have also showed anti-Leishmanial, antibacterial [27], antimalarial [28] activities. Despite of the lack of phytochemical investigation, Tanacetum plants has received recognition as a potential nematocidal, insecticidal, antibacterial, cytotoxic, and phytotoxic herbs [29]. Therefore, the diverse chemical profiling of Tanacetum plants, and their medicinal uses prompted us to investigate T. falconeri for its chemodiversity and biological potential. The goal of this study was to evaluate the traditional therapeutic applications of T. falconeri by evaluating the various extracts for their total bioactive content, full secondary metabolic profile, and bioactivities. In vitro tests were conducted to evaluate the anti-oxidant (DPPH, ABTS, FRAP, CUPRAC, phosphomolybdenum, ferrous chelating) and enzyme inhibitory capabilities of all extracts against various enzymes linked to skin, neurodegenerative, and diabetic illnesses. Additionally, multivariate analysis and docking investigations were carried out.

Experimental procedures

Collection of the plant material and identification

The plant material was collected from Shigar District, Gilgit-Baltistan, Pakistan and was identified by Dr. Zaheer Abbas, a taxonomist at the University of Education, DG Khan Campus, Dera Ghazi Khan, where a voucher specimen No. BT-0063 has been deposited in the herbarium of same university.

Preparation of the extracts

The collected plant material was divided into flower (TFF) and stem with leaves (TFS) parts, which were then dried under shade for one week. Each part (600 and 800 g, respectively) was divided into four parts, which were then extracted separately through maceration using n-hexane, chloroform, ethyl acetate and methanol to get crude extracts of the stem: TFSM: methanolic extract of stem; TFSH: hexane extract of stem; TFSE: ethyl acetate extract of stem; TFSC: chloroform extract of stem and flowers extracts: TFFM: methanolic extract of flowers; TFFH: hexane extract of flowers; TFFE: ethyl acetate extract of flowers; TFFC: chloroform extract of flowers. All these extracts were then studied for their phenolic and flavonoid contents, antioxidant and enzyme inhibition studies and chemodiversity.

Estimation of Total phenolic (TPC) and Total flavonoid (TFC) contents

The Estimation of total phenolic (TPC) and total flavonoid (TFC) contents were done through same methods as we reported previously [3033]. The results of total phenolic contents (TPC) were presented in milligrams of gallic acid equivalent per grams of extract (mg GAE/g extract). The total flavonoid contents (TFC) results were reported in milligrams of rutin equivalent per grams of extract (mg RE/g extract).

Antioxidant activities assays

The antioxidant activities of extracts were measured by following pre-established protocols as we reported previously [3033]. For FRAP, ABTS, DPPH, CUPRAC, and total antioxidant capacity, trolox equivalent was utilized as standard and results were expressed as mg TE/g extract; while for metal chelating assays, ethylene diamine tetraacetic acid (EDTA) was the standard and results were expressed as mg TE/g extract.

Enzyme inhibition assays

The α-amylase, α-glucosidase, BChE, tyrosinase, and AChE enzyme inhibitory assays were conducted using previously published methods [3033]. Acarbose (mmol ACAE/g extract) was used as a standard to measure the inhibitory activity of α-amylase and α-glucosidase. Galantamine (mg GALAE/g extract) was used to measure the inhibitory activity of AChE and BChE, and kojic acid (mmol KAE/g extract) was used to measure the inhibitory activity of tyrosinase.

UHPLC-MS analysis

UHPLC-MS (ultra-high performance liquid chromatography mass spectrometry) analysis was used to profile secondary metabolites using an Agilent 1290 Infinity UHPLC system coupled to an Agilent 6520 Accurate-Mass Q-TOF mass spectrometer with dual ESI source, as we previously reported [3032]. The column was an Agilent Zorbax Eclipse XDB-C18 with 3.5 m in thickness and 2.1 × 150 mm in diameter. A 0.1% formic acid solution in water served as mobile phase A, while a 0.1% formic acid solution in acetonitrile served as mobile phase B. A consistent flow rate of 0.5 millilitres per minute was maintained. One microliter of methanolic extract was given for twenty-five minutes, and then there was a five-minute post-run period. The secondary metabolites were found using the METLIN database.

Statistical analysis

The experiments were performed in triplicate, and differences between the extracts were compared using an ANOVA and Tukey's test. Pearson correlation analysis was used to establish the link between total bioactive components and biological activity assays. Graph Pad Prism (version 9.2) was used for the analysis. To assess the degree of similarity or difference between the extracts, a PCA was carried out using SIMCA (version 14.0).

Docking study methodology

The chemical structures of the five enzymes with the highest resolution were downloaded from the protein data bank in PDB format. Discovery Studio (DS 2021Client) software was employed to formulate protein molecules. Attached chemical moieties (water molecules and other ligand) were removed from macromolecules. Afterward, they were transferred onto the PyRx program (version 0.8) for docking purposes in pdbqt file that contains a protein structure with hydrogens in all polar residues. The structures of selected ligands were acquired from the Pubchem as 3D SDF formats. The software specification and procedure of docking were followed as described by Ahmed et al., [34]. The enzyme molecules were loaded into PyRx and converted to macromolecules by using autodock embedded in PyRx software. Then the ligands were attached using the open babel tool, and energy was minimized to obtain the stable structure; then, ligands were converted to pdbqt format. The docking site on the protein target was defined by establishing a grid box, which was maximized using “maximize” option for better coverage of active site and exhaustiveness was 8. The other settings of the software were used as “default”. The best conformation with the lowest docked energy was chosen after the docking search was completed. The molecular docking result for each compound was visualized as an output pdbqt file by using the molecular graphics laboratory (mgltool) tool. Interactions were finally visualized in discovery studio by using mgltool, to determine some specific contacts between the atoms of the test compounds and amino acids residues of the studied protein molecules [35].

Results and discussion

Total phenolic (TPC) and flavonoid (TFC) contents of T. falconeri

Phenolic compounds are important component of nutraceuticals and functional foods because of their antioxidant properties. The antioxidant properties of phenolics are usually attributed to the presence of hydroxyl group(s) on the benzene ring, which goes about an electron donor and consequently and straight forwardly includes in quenching free radicals. In the present study, several solvent-based crude stem and flower extracts of T. falconeri were screened for their total phenolic and flavonoid contents. Total phenolic contents (TPC) observed in the methanol extract of flower (TFFM) were high (40.28 ± 0.47 mg GAE/g extract), followed by the TFFH extract (33.00 ± 0.67 mg GAE/g extract). Although the same trend of TPC for stem extracts was seen in TFSM (22.21 ± 0.17 mg GAE/g extract) and TFSH (24.34 ± 0.49 mg GAE/g extract) but were lower than those of respective extracts of the flower. Ethyl acetate extracts from both the sources were next in line (Table 1). Similarly, total flavonoids contents (TFC) were also observed high in flower extracts (TFFM 36.79 ± 0.36 mg QE/g extract and TFFH 32.80 ± 0.80 mg QE/g extract), followed by stem extracts (TFSM 17.68 ± 0.32 mg QE/g extract and TFSE 23.38 ± 0.17 mg QE/g extract). In both the cases, lowest phenolic and flavonoid contents were calculated in chloroform extracts (Table 1). Usually phenolics and flavonoids are relatively polar compounds; therefore, their high concentration in methanolic or ethyl acetate extracts seems reasonable, however, in case of flower extracts (TFFH and TFSH), the high amount of phenolic contents could be attributed to the possible presence of esters of phenolic acids in the extracts. Literature reports also substantiate our findings where methanolic extracts of Tanacetum plants have been reported to be rich in phenolic contents [36]. Another report describes that Tanacetum species produce high level of vanillic acid, and caffeic acid, catechin and quercetin [37] and other phenolics and flavonoids [38, 39].; The presence of caffeoylquinic acids in Tanacetum species [40, 41] substantiates our deduction that phenolic acid esters are present in T. falconeri, which are extracted in low polar solvents, and thus TFFH and TFSH also afforded high amount of phenolic contents.
Table 1
Total bioactive contents of T. falconeri
Test Samples
TPC (mg GAE/g extract)
TFC (mg QE/g extract)
TFFM
40.28 ± 0.47a
36.79 ± 0.36a
TFFH
33.00 ± 0.67b
32.80 ± 0.80b
TFFE
17.37 ± 0.23e
11.65 ± 0.09f
TFFC
11.92 ± 0.22f
3.22 ± 0.05g
TFSM
22.21 ± 0.17d
17.68 ± 0.32d
TFSH
24.34 ± 0.49c
13.39 ± 0.30e
TFSE
22.18 ± 0.33d
23.38 ± 0.17c
TFSC
18.07 ± 0.18e
11.08 ± 0.22f
TFFM: methanolic extract of flower of T. falconeri; TFFH: hexane extract of flower of T. falconeri; TFFE: ethyl acetate extract of flower of T. falconeri; TFFC: chloroform extract of flower of T. falconeri; TFSM: methanolic extract of stem of T. falconeri; TFSH: hexane extract of stem of T. falconeri; TFSE: ethyl acetate extract of stem of T. falconeri; TFSC: chloroform extract of stem of T. falconeri. Different letters in same column indicate significant differences in the tested extracts (p < 0.05)

Antioxidant activities of the extracts of T. falconeri

Research showed that the antioxidant activity of a plant extract is usually associated to the phenolic contents, i.e. higher the phenolic contents, higher will be the activity [42]. However, in the present study, the highest DPPH free radical scavenging activity (TFFM; 49.58 ± 0.11 mg TE/g extract) was associated to the methanolic flower extract, which is followed by n-hexane flower extract (TFFH; 47.91 ± 0.17 mg TE/g extract), whereas, methanolic stem extract (TFSM) also showed nearly similar inhibition (43.75 ± 0.41 mg TE/g extract). It is already predicted that the presence of phenolic contents in low polar solvents could be of the nature of phenolic acid esters. Literature search revealed that phenolic acid esters are potent antioxidants [43], therefore, the activity of TFFM could be attributed to such kind of compounds and other metabolites. On the other hand, the higher DPPH free radical inhibitory potential of TFSM could be attested for its high phenolic contents (Table 2). TFSM and TFFH were also significantly active, while other extracts were found inactive (Table 2). In case of ABTS free radical activity same pattern was observed as in DPPH and TFFM exhibited highest inhibition value (112.61 ± 0.15 mg TE/g extract). The next in line were TFFH, TFSM and TFSE (Table 2) with values of 84.60 ± 0.57, 73.43 ± 2.77 and 62.51 ± 0.97 respectively. In metal ion reducing power assays, again the TFFM was highly active (CUPRAC: 160.48 ± 6.59 mg TE/g extract; FRAP: 102.58 ± 2.62 mg TE/g extract), followed by the methanolic extract of stem (TFSM). All other extracts also exhibited significant and comparable metal reducing power (Table 2). TFSE was most active in phosphomolybdenum with the value of 1.71 ± 0.03 mg TE/g extract, whereas, TFFH and TFSH were also significantly active with the values of 1.64 ± 0.00 and 1.58 ± 0.08 mg TE/g extract, respectively. Highest metal chelating activity was found for stem extracts, since TFSC and TFSM displayed chelating power as 18.06 ± 0.61 and 15.57 ± 0.22 mg TE/g extract, whereas, flower extracts were found weak chelators (Table 2). It if further noticed that more polar extracts were also weak chelators, however, overall the present study revealed that T. falconeri is a potential antioxidant plant to be considered for its uses in health promoting formulations.
Table 2
Antioxidant activities of the extracts of T. falconeri
Test Samples
DPPH (mg TE/g extract)
ABTS (mg TE/g extract)
CUPRAC (mg TE/g extract)
FRAP (mg TE/g extract)
Phosphomolybdenum (mg TE/g extract)
metal chelating (mg EDTAE/g extract)
TFFM
49.58 ± 0.11a
112.61 ± 0.15a
160.48 ± 6.59a
102.58 ± 2.62a
1.12 ± 0.01 cd
13.02 ± 0.39c
TFFH
47.91 ± 0.17b
84.60 ± 0.57b
123.27 ± 2.38b
67.17 ± 0.21b
1.64 ± 0.00a
10.05 ± 0.60d
TFFE
5.87 ± 0.45f
36.60 ± 1.15f
49.58 ± 2.90d
17.93 ± 0.05f
1.25 ± 0.02bc
8.73 ± 0.29d
TFFC
Not active
15.95 ± 1.20 g
35.17 ± 0.94e
15.23 ± 0.48f
1.00 ± 0.02d
14.03 ± 0.28bc
TFSM
43.75 ± 0.41c
73.43 ± 2.77c
82.73 ± 1.59c
50.64 ± 0.22c
1.10 ± 0.02d
15.57 ± 0.22b
TFSH
19.20 ± 0.37e
47.23 ± 1.11e
77.22 ± 1.98c
40.40 ± 1.36d
1.58 ± 0.08a
9.31 ± 0.70d
TFSE
22.42 ± 0.38d
62.51 ± 0.97d
77.13 ± 0.88c
50.20 ± 0.82c
1.71 ± 0.03a
9.04 ± 0.69d
TFSC
5.95 ± 0.61f
38.33 ± 2.11f
51.75 ± 0.52d
23.93 ± 0.13e
1.36 ± 0.03b
18.06 ± 0.61a
TFFM: methanolic extract of flower of T. falconeri; TFFH: hexane extract of flower of T. falconeri; TFFE: ethyl acetate extract of flower of T. falconeri; TFFC: chloroform extract of flower of T. falconeri; TFSM: methanolic extract of stem of T. falconeri; TFSH: hexane extract of stem of T. falconeri; TFSE: ethyl acetate extract of stem of T. falconeri; TFSC: methanolic extract of stem of T. falconeri. Different letters in same column indicate significant differences in the tested extracts (p < 0.05)

Enzyme inhibition activities of the extracts of T. falconeri

AChE and BChE enzyme inhibition activities

Alzheimer's disease (AD), a noncommunicable disease (NCDs) has been identified as a largely increasing health challenge worldwide. It is an irreversible, progressive form of dementia, associated with an ongoing decline of brain functioning [44] and thus causes memory loss. The World Health Organization (WHO) has reported that more than 30 million people are afflicted by AD and this number is expected to become double every two decades to reach ~ 115 million by 2050. This problem is thus expected to weaken the social and economic development and may affect the social services [45]. Acetylcholine (ACh) and buytrylcholine (BCh) are important neurotransmitters requires for proper brain, memory and body functioning Therefore, low levels of cholines lead to memory issues and muscle disorders. Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) are the enzymes that hydrolyse acetycholine and butyrylcholine, respectively [46]. The inhibitors of these two enzymes results into accumulation of the neurotransmitter acetylcholine and enhanced stimulation of postsynaptic cholinergic receptors [47, 48]. Natural products have already proven to be promising sources of useful acetylcholinesterase (AChE) inhibitors [49]. The currently approved drugs for AD, galantamine and rivastigmine, are plant-derived alkaloids, which offer symptomatic relief from AD [50]. These facts suggest to investigate the use of medicinal plants and their formulations to prevent and treat neurodegenerative disease [51].
In this study, all the extracts of T. falconeri were evaluated against AChE and BChE enzymes. Methanolic (TFFM) and ethyl acetate (TFFE) extracts f flowers, while ethyl acetate (TFSE) and chloroform (TFSC) extracts of stem were most but equally active with inhibitory values as 4.09 ± 0.09, 4.53 ± 0.13, 4.00 ± 0.23 and 4.03 ± 0.22 mg GALAE/g extract, respectively, against AChE, whereas, TFFE, TFFC and TFSC were most active against BChE with values as 2.09 ± 0.18, 2.57 ± 0.24 and 2.10 ± 0.18 mg GALAE/g extract. These observations revealed that ethyl acetate and chloroform extracts are more active against these enzymes.

Tyrosinase enzyme inhibition activities

Browning of raw food and hyperpigmentation of human skin are two undesirable processes caused a group of copper-containing enzyme tyrosinase (EC 1.14. 18.1). Hyper activity of tyrosinase causes results in a less attractive appearance and loss in nutritional quality of food and blackening of human skin [52]. Further, over production of melanin in human skin causes several skin disorders such as melasma, senile lentigines and freckles and thus exert a considerable psychosomatic effect on affected patients [53]. These problems can be controlled by using tyrosinase inhibitors. Presently available tyrosinase inhibitors like hydroquinone, arbutin, kojic acid, ascorbic acid, ellagic acid and others have different problems either in their use or the bioavailability [54].Therefore, there is a great need to discover and develop new but safer tyrosinase inhibitors. For his purpose, the medicinal plant extracts are the main agents being researched and used as tyrosinase inhibitors in these days. In the present work various extracts of T. falconeri were evaluated for their tyrosinase inhibitory activity. Methanolic extracts of both flowers (TFFM) and stem TFSM) of T. falconeri were the most active with inhibition values as 35.53 ± 0.35 and 35.30 ± 0.70 mg KAE/g extract, respectively followed by the hexane extracts (29.96 ± 0.10 and 32.41 ± 1.91 mg KAE/g extract, respectively. All other extracts exhibited equal but significant inhibitory potential (Table 3), which disclosed that T. falconeri can be a potential ingredient in cosmetic and food industry.
Table 3
Enzyme inhibition activities of the extracts of T. falconeri
Test Samples
AChE (mg GALAE/g extract)
BChE (mg GALAE/g extract)
Tyrosinase (mg KAE/g extract)
α-Amylase (mg ACAE/g extract)
α-Glucosidase (mg ACAE/g extract)
TFFM
4.09 ± 0.09ab
0.47 ± 0.08cd
35.53 ± 0.35a
0.38 ± 0.009c
0.46 ± 0.002d
TFFH
3.40 ± 0.28 cd
0.38 ± 0.06d
29.96 ± 0.10bc
0.48 ± 0.02b
0.86 ± 0.001c
TFFE
4.53 ± 0.13a
2.09 ± 0.18a
27.00 ± 0.90c
0.55 ± 0.02ab
1.06 ± 0.002a
TFFC
4.14 ± 0.08ab
2.57 ± 0.24a
27.25 ± 1.29c
0.50 ± 0.02ab
1.09 ± 0.015a
TFSM
3.24 ± 0.12d
0.94 ± 0.09bc
35.30 ± 0.70a
0.41 ± 0.01c
0.91 ± 0.003bc
TFSH
3.67 ± 0.12bcd
1.46 ± 0.10b
32.4 1 ± 1.91ab
0.53 ± 0.01ab
0.87 ± 0.015c
TFSE
4.00 ± 0.23abc
1.16 ± 0.17b
26.79 ± 0.63c
0.53 ± 0.01ab
1.02 ± 0.107ab
TFSC
4.03 ± 0.22ab
2.10 ± 0.18a
27.50 ± 1.50c
0.55 ± 0.02a
1.08 ± 0.002a
TFFM: methanolic extract of flower of T. falconeri; TFFH: hexane extract of flower of T. falconeri; TFFE: ethyl acetate extract of flower of T. falconeri; TFFC: chloroform extract of flower of T. falconeri; TFSM: methanolic extract of stem of T. falconeri; TFSH: hexane extract of stem of T. falconeri; TFSE: ethyl acetate extract of stem of T. falconeri; TFSC: methanolic extract of stem of T. falconeri. Different letters in same column indicate significant differences in the tested extracts (p < 0.05)

α-Amylase and α-glucosidase enzyme inhibition activities

Diabetes mellitus is another major non-communicable metabolic disease that has high impact on health and economy. A published report revealed that only in 2014, 4.9 million deaths were recorded due to diabetes [55]. In diabetic patients usually the blood glucose level increases after taking meal and thus causes postprandial hyperglycemia [56]. The glycosidic linkage in carbohydrates is broken by α-amylase to produce oligosaccharides, which are then degraded to glucose by α-glucosidase [57]. Since both the enzymes digest the carbohydrates and cause diabetes [55]; inhibition of the activity of these enzymes can delay the increase in blood glucose level and reduce the risk of developing diabetes [58]. Among current inhibitors, only acarbose inhibits both α-amylase and α-glucosidase, whereas, miglitol and voglibose inhibit only α-glucosidase [59, 60]. Literature search revealed that some of the plant extracts or pure phytochemicals were found effective against both enzymes [6164], which leads to conclude that medicinal plants can serve as potential antidiabetic agents. Therefore, in the present study, the flower and stem extracts of T. falconeri were evaluated for their inhibitory potential against α-amylase and α-glucosidase. Against amylase, all the extracts exhibited significant activity with inhibitory values in the range of 0.38 ± 0.009 to 0.55 ± 0.002 mg ACAE/g extract, while against α-glucosidase the inhibitory values were observed between 0.46 ± 0.002 to 1.09 ± 0.015 mg ACAE/g extract, with lowest potential in both the cases was observed for methanolic extracts (Table 3). It is reported that the plant extracts exhibit antidiabetic properties due to the combined effect of biologically active compounds like polyphenols, carotenoids, lignans, coumarins, glucosinolates, etc. [65]. These plant metabolites as a combined effect, improve the performance of pancreatic tissue by increasing insulin secretions or by reducing the intestinal absorption of glucose [66]. Therefore, the anti-amylase and anti-glucosidase activities of the extracts of T. falconeri could be attributed to the presence of such metabolites. Literature reports revealed that most of the plant extracts and pure compounds exhibit selective inhibition against either α-amylase or α-glucosidase; while only fewer have been found active against both the enzymes [55]. Overall the crude extracts of T. falconeri were found significantly active against both the enzymes; therefore, it can be a potential component of crude antidiabetic drugs.

UHPLC-MS Analysis

UHPLC-MS Analysis of methanolic extract (Fig. 1) of flowers result in identification of 133 compounds (Table 4) of various secondary metabolites class mainly phenolic, flavonoids, Alkaloids, terpenoids and steroids. The presence of important phenolic and flavonoids compounds like 6-Caffeoylsucrose, 3-O-Feruloylquinic acid, Brosimacutin C, Quinic acid, Rutacultin, Castamollissin, Kaempferol 3-p-coumarate and Methylsyringin may be responsible for antioxidant activities [67]. These results demonstrated that T. falconeri is not limited to a specific class of secondary metabolites and can create a broad variety of compounds. Chemodiversity makes T. falconeri a valuable herb with a broad range of bioactivities.
Table 4
UHPLC-MS analysis of methanolic extract of flower of T. falconeri
Sr. No
Analyte Peak Name
Retention Time
Area / Height
Molecular formula
Compound name
Mass
Class of compound
1
381.0730 (M + K+)
1.51
3.93
C19H18O6
1,5,8-Trihydroxy-3-methyl-2-prenylxanthone
342.3
Xanthone
2
317.0928
1.48
4.01
C13H16O9
Ginnalin B
316.0794
Phenolic acid
3
429.2146
1.49
4.97
C23H28N2O6
Carapanaubine
428.1947
Indolizine
4
319.0642
1.50
4.77
C18H10N2O4
Prekinamycin
318.0641
Alkaloids
5
439.1663
1.51
3.80
C21H26O10
sec-o-Glucosylhamaudol
438.1526
Chromene
6
321.0363
1.50
3.45
C18H8O6
Erosnin
320.0321
Coumestan
7
295.0925
1.56
3.65
C18H14O4
7-Acetyloxy-2-methylisoflavone
294.0892
Isoflavone
8
367.1182
1.66
3.96
C21H18O6
Isoglycyrol
366.1103
Phyto
9
353.0613
1.70
3.64
C19H12O7
Daphnoretin
352.0583
Coumarin
10
685.2285
1.73
4.65
C37H36N2O11
Citbismine C
684.2319
Acridine
11
684.2245 (M + NH4+)
1.73
4.39
C30H34O17
Peonidin acetyl 3,5-diglucoside
666.1796
Anthocyanidine glycoside
12
505.1489
1.74
4.67
C21H28O14
6-Caffeoylsucrose
504.1479
Cinnamic acid glycoside
13
244.0595
1.77
3.26
C13H9NO4
Maculine
243.0532
Phyto
14
458.1606
1.81
4.17
C20H27NO11
Amygdalin
457.1584
Cyanogen glycoside
15
367.0863
1.82
5.49
C20H14O7
12a-Methoxydolineone
366.0740
Rotenoid flavonoid
16
306.1160
2.32
6.76
C19H15NO3
N-Acetyldehydroanonaine
305.1052
Isoquinoline alkaloid
17
431.1402
2.45
4.00
C22H22O9
Catechin 3-O-(1,6-dihydroxy-2-cyclohexene-1-carboxylate)
430.1264
Flavonoid
18
419.1085
2.49
2.96
C20H18O10
Kaempferol 3-xyloside
418.0900
Falvonoid
19
312.1150
2.75
8.56
C14H17NO7
Zierin
311.1005
Cyanogen glycoside
20
345.1150
3.03
7.13
C22H16O4
Sanaganone
344.1049
Extended flavonoid
21
378.1217 (M + Na+)
3.14
8.18
C20H21NO5
Promucosine
355.1420
Alkaloid
22
307.1367
3.57
3.03
C20H18O3
Lonchocarpin
306.1256
Chalcone
23
203.0709
3.70
13.48
C12H10O3
5-Hydroxy-2,3-dimethyl-1,4-naphthoquinone
202.0630
Naphthaquinone
24
397.1801
3.66
5.28
C20H28O8
4,5-Dihydroniveusin A
396.1784
Germacranolide
25
305.0988
3.76
9.90
C16H16O6
4'-O-Methylcatechin
304.0947
Catechin
26
329.0808
3.87
5.95
C14H16O9
BERGENIN
328.0794
Trihydroxybenzoic acid derivative
27
270.0759
3.87
5.71
C15H11NO4
Evoxanthidine
269.0688
Acridine
28
319.1141 (M + Na+)
4.06
8.67
C17H18O6
Bryaflavan
318.1103
Isoflavane
29
279.1226
4.06
7.83
C15H18O5
Artecanin
278.1154
Sesquiterpene lactone
30
372.1787 (M + NH4+)
4.36
8.11
C21H22O5
Mundulea flavanone B
354.1467
Flavanone
31
303.0833
4.26
7.97
C16H14O6
Alysifolinone
302.0790
Flavanone
32
293.1148
4.38
6.39
C19H16O3
Purpuritenin A
292.1099
Chalcone
33
325.1269 (M + CH3OH + H+)
4.37
9.25
C15H16O6
Dihydromikanolide
292.0947
Lactone
34
372.1803 (M + NH4+)
4.36
7.46
C21H22O5
Flemistrictin D
354.1467
Chalcone
35
369.1127
4.54
7.78
C17H20O9
3-O-Feruloylquinic acid
368.1107
Quinic acid derivative
36
360.1805 (M + CH3OH + H+)
4.54
5.57
C19H21NO4
Norcorydine
327.1471
Alkaloid
37
323.1477
4.52
6.18
C17H22O6
Tetraneurin A
322.1416
Sesquiterpene lactone
38
360.1891 (M + NH4+)
4.53
4.99
C20H22O5
Brosimacutin C
342.1467
Flavanone
39
239.1290
4.68
6.88
C13H18O4
1-(3-Ethyl-2,4-dihydroxy-6-methoxyphenyl)-1-butanone
238.1205
Aromatic ketone
40
416.1874 (M + NH4+)
4.75
6.14
C19H26O9
Methyl 3,4-dihydroxy-5-prenylbenzoate 3-glucoside
398.1577
Tannin
41
369.1516
4.84
6.55
C18H24O8
4-Hydroxy-3-prenylbenzoic acid glucoside
368.1471
Phenolic glycoside
42
281.1386
4.83
5.90
C15H20O5
8-Deoxy-11,13-dihydroxygrosheimin
280.1311
Sesquiterpene lactone
43
332.1685 (M + NH4+)
4.83
5.55
C18H20NO4
Litcubinine
314.1392
Alkaloids
44
279.1227
4.88
5.27
C15H18O5
Artecanin (Tanacetum parthenium)
278.1154
Sesquiterpene lactone
45
191.0735
4.89
5.74
C11H10O3
7-Hydroxy-2,5-dimethyl-4H-1-benzopyran-4-one
190.0630
Coumarin
46
332.1687
4.83
5.56
C22H21NO2
Melochinone
331.1572
Quinoline
47
444.1823
5.04
9.40
C23H29N3O2S2
Thiothixene
443.1701
Thioxanthene
48
404.2139 (M + CH3OH + H+)
4.91
5.32
C21H25NO5
Capaurine
371.1733
Alkaloid
49
549.2671
5.05
4.12
C29H40O10
Archangelolide
548.2621
Sesquiterpene lactone
50
533.2355
5.03
4.87
C28H36O10
Nomilinic acid
532.2308
Steroidal lactone
51
382.1971
5.05
4.80
C19H27NO7
Petasitenine
381.1788
Spiro epoxide
52
285.1748
5.10
5.18
C18H22NO2
6,7-Dihydro-4-(hydroxymethyl)-2-(p-hydroxyphenethyl)-7-methyl-5H-2-pyrindinium
284.1651
Phenol
53
283.1690
5.10
6.42
C19H22O2
Miltirone
282.1620
Diterpenoid
54
461.2160
5.08
6.37
C25H32O8
Aspidin
460.2097
Phloroglucinol
55
285.1595 (M + NH4+)
5.10
5.38
C17H17NO2
Assoanine
267.1259
Phenanthridine
56
251.1291
5.14
5.79
C14H18O4
Helinorbisabone
250.1205
Terpenoids
57
285.1329
5.14
5.55
C14H20O6
2-Phenylethyl beta-D-glucopyranoside
284.1260
Glycoside
58
193.0680
5.21
3.75
C7H12O6
Quinic acid
192.0634
Cyclitol carboxylic acid
59
452.2076
5.28
7.11
C26H29NO6
Piscerythramine
451.1995
Flavonoids
60
335.1238
5.36
4.55
C21H18O4
Calopogoniumisoflavone A
334.1205
Flavonoids
61
404.2241 (M + NH4+)
5.33
5.30
C19H30O8
Citroside A
386.1941
Glycoside
62
331.1079
5.38
5.34
C18H18O6
7-Hydroxy-5,8,2'-trimethoxyflavanone
330.1103
Flavonoids
63
267.1232
5.37
4.96
C14H18O5
Sapidolide A
266.1154
Lactone
64
317.0991 (M + CH3OH + H+)
5.37
10.44
C16H12O5
6-Methylapigenin
284.0685
Flavonoids
65
346.2001 (M + NH4+)
5.48
7.36
C20H24O4
Sclareapinone
328.1675
Quinone
66
411.1374
5.52
4.10
C23H22O7
Pongapinone A
410.1366
Coumarin
67
367.1725
5.55
4.93
C19H26O7
Orizabin
366.1679
Terpenoids
68
575.1614
5.46
1.36
C35H26O8
Viniferal
574.1628
Benzofuran
69
323.1089
5.67
5.72
C19H16NO4
Berberrubine
322.1079
Alkaloids
70
372.1986 (M + NH4+)
5.68
9.08
C23H27NO2
Murrayazolinine
349.2042
Alkaloids
71
374.1769 (M + NH4+)
5.75
9.07
C13H24O11
Galactopinitol A
356.1319
Glycoside
72
519.2568
5.74
9.43
C24H38O12
Cinnamoside
518.2363
Phenolic amide
73
535.2872
5.75
4.63
C29H42O9
Corchoroside A
534.2829
Cardenolide glycoside
74
418.2217 (M + CH3OH + H+)
5.87
7.80
C22H27NO5
O-Methylandrocymbine
385.1889
Alkaloids
75
335.1238
5.83
5.35
C16H18N2O6
Cappariloside A
334.1165
Indoles
76
561.1657
5.85
7.37
C27H28O13
3'-Deoxymaysin
560.1530
Flavonoids
77
408.2128
5.97
5.41
C25H29NO4
Ancistrocladine
407.2097
Nephthalenes
78
275.1255
5.89
9.18
C16H18O4
Rutacultin
274.1205
Coumarin
79
282.1697 (M + NH4+)
5.89
5.37
C15H20O4
Sequiterpene Lactone 326
264.1362
Terpenoids
80
353.1920
5.91
3.92
C21H24N2O3
Vobasine
352.1787
Alkaloids
81
563.1818
5.99
10.84
C27H30O13
Rhamnellaflavoside A
562.1686
Flavanoid
82
369.1882
5.93
4.82
C21H24N2O4
Baloxine
368.1736
Alkaloid ester
83
469.1053
6.12
2.62
C20H20O13
Castamollissin
468.0904
Phenolic
84
439.2165
6.10
4.62
C26H30O6
Kanzonol G
438.2042
Flavonoids
85
318.0725
6.11
4.48
C19H11NO4
Lettowianthine
317.0688
Alkaloids
86
606.3224
6.16
5.56
C33H43N5O6
Amphibine H
605.3213
Peptide
87
368.1673
6.16
6.28
C18H25NO7
Isatidine
367.1631
Alkaloids
88
342.1887 (M + NH4+)
6.35
6.34
C17H24O6
Chamissonolide
324.1573
Terpenoids
89
293.0782
6.19
4.71
C18H12O4
Karanjin
292.0736
Flavonoids
90
507.3096
6.30
4.33
C29H38N4O4
Mucronine A
506.2893
Peptide
91
293.1751
6.42
4.04
C17H24O4
9-Acetoxyfukinanolide
292.1675
Terpenoids
92
365.1555 (M + CH3OH + H+)
6.45
13.62
C18H20O6
3,3'-Dihydroxy-4',5,7-trimethoxyflavan
332.1260
Flavonoids
93
375.1748 (M + Na+)
6.45
7.96
C21H24N2O3
Ajmalicine
352.1787
Alkaloid
94
551.2326
6.51
3.47
C31H34O9
Lappaol B
550.2203
Phenylpropanoids
95
369.2056
6.60
10.05
C23H28O4
Quercetol B
368.1988
Phenylpropanoids
96
365.1331 (M + K+)
6.64
9.65
C19H22N2O3
Alkaloid AQC2
326.1630
Alkaloids
97
369.1878
6.59
8.25
C21H24N2O4
Uncarine A
368.1736
Alkaloids
98
315.1191
6.67
5.06
C18H18O5
Matteucinol
314.1154
Flavonoids
99
597.2793 (M + Na+)
6.69
5.96
C31H42O10
Asclepin
574.2778
Cardenolide glycoside
100
405.1265 (M + K+)
6.65
5.97
C21H22N2O4
Apodine
366.1580
Alkaloids
101
402.2446 (M + NH4+)
6.75
9.74
C20H32O7
Cinnzeylanol
384.2148
Terpenoids
102
455.0687 (M + Na+)
6.79
4.80
C24H16O8
Kaempferol 3-p-coumarate
432.0845
Phenolics
103
399.2339
6.85
4.83
C23H30N2O4
Desacetoxyvindoline
398.2206
Alkaloids
104
446.2690 (M + NH4+)
6.93
7.66
C24H32N2O5
Aspidoalbine
428.2311
Alkaloids
105
371.2226 (M + CH3OH + H+)
6.92
5.00
C22H26O3
5,7-Dimethoxy-8-prenylflavan
338.1882
Flavonoids
106
384.1988 (M + NH4+)
6.98
10.99
C19H26O7
Orizabin
366.1679
Terpenoids
107
435.1731
8.36
5.91
C26H26O6
Cycloartocarpin A
434.1729
Flavonoids
108
309.1449
8.49
8.04
C20H20O3
Isocordoin
308.1412
Phenolics
109
647.3895
8.74
11.05
C36H54O10
Gypsogenin 3-O-b-D-glucuronide
646.3717
Terpenoids
110
354.1438 (M + Na+)
8.96
5.61
C22H21NO2
Melochinone
331.1572
Alkaloids
111
301.0489
9.00
5.91
C12H12O9
Mumefural
300.0481
Furans
112
423.2096
9.20
6.47
C26H30O5
Alopecurone G
422.2093
Flavonoids
113
415.1712
9.27
5.81
C23H26O7
Heteroflavanone C
414.1679
Flavonoids
114
686.3632
9.39
9.61
C36H51N3O10
Avadharidine
685.3574
Terpenoids
115
415.2072
9.47
5.83
C24H30O6
Armillaripin
414.2042
Terpenoids
116
387.1727
9.63
6.36
C18H26O9
Methylsyringin
386.1577
Terpenoids
117
409.1384 (M + Na+)
9.63
7.39
C25H22O4
Fulvinervin B
386.1518
Flavonoids
118
229.1800
10.40
7.86
C13H24O3
Menthone 1,2-glyceryl ketal
228.1725
Monoterpenoid
119
289.1581
10.51
7.16
C16H20N2O3
( ±)-Rollipyrrole
288.1474
Alkaloids
120
423.2464
10.56
6.34
C23H34O7
Picrasin C
422.2305
Terpenoids
121
281.2062
10.56
4.42
C19H24N2
N-Methylaspidospermatidine
280.1939
Alkaloids
122
512.3634 (M + NH4+)
10.67
9.87
C33H42N4
Auricularine
494.3409
Alkaloids
123
389.1716
10.62
7.30
C25H24O4
Kanzonol E
388.1675
Flavonoids
124
531.3973 (M + NH4+)
10.80
9.25
C18H23NO4
Pandamarilactonine A
317.1627
Alkaloids
125
322.2130
10.78
6.69
C22H27NO
Phenazocine
321.2093
Alkaloids
126
368.2188 (M + NH4+)
10.89
5.09
C16H30O6
L-Citronellol glucoside
318.2042
Terpenoids
127
673.6284
10.96
14.58
C23H37NO5
Cammaconine
407.2672
Terpenoids
128
397.2128
11.06
4.91
C23H28N2O4
Echitovenine
396.2049
Alkaloids
129
600.4545 (M + NH4+)
11.22
10.79
C37H58O5
Hericene B
582.4284
Terpenoids
130
277.1950
11.27
5.58
C16H24N2O2
Carolinianine
276.1838
Alkaloids
131
694.3685 (M + NH4+)
11.29
5.20
C36H52O12
Cucurbitacin I 2-glucoside
676.3459
Terpenoids
132
275.2372
11.44
8.66
C19H30O
4,5-(methanoxy-2-methylethano)isolongifol-4-ene
274.2297
Sesquiterpenoid
133
513.3911
11.29
9.54
C33H52O4
Methyl 3b-hydroxy-13(18)-oleanen-28-oate
512.3866
Triterpenoid

Data analysis

Multivariate analysis provides a bridge between diverse parameters and their interactions. This makes it a fundamental tool in phytochemical studies to gain more information on the relationship between the chemical components and biological activities of plant extracts. For this purpose, we conducted a multivariate analysis of the extracts tested. Initially, we assessed the correlation between the total bioactive compounds and the biological activities. As illustrated in Fig. 2A, the radical quenching and reducing potentials were strongly associated with these compounds. However, metal chelation and phosphomolybdenum capacities had no association with the total of phenolic and flavonoid components. This can be elucidated by the presence of non-phenolic substances like terpenoids or peptides. In agreement with our findings, several researchers highlighted a signficant relationship between the total bioactive constituents and antioxidant properties [68, 69]. In terms of enzyme inhibitory characteristics, no relationship was found with the total bioactive components. Principal component analysis was employed to demonstrate the similarity/dissimilarity among the tested samples and R2 and O2 that shows the fitness and predictive ability of the model were found as 0.98 and 0.82, respectively (Fig. 2B). In Fig. 2C, we observed a loading scatter plot of the tested variables, and the total bioactive components and antioxidant properties were the same in the plot. However, the enzyme inhibitory effects were classified in another plot. According to Fig. 2B and 2D, the tested extracts were classified into five groups. In comparison to the other extracts, the methanol extracts from both parts showed significantly stronger antioxidant activity, thereby setting them apart from the other extracts. At the same time, the chloroform extracts had a greater enzyme inhibition effect, thus leading them to be classified in the same group. It is clear that the plant parts and extraction solvents used influenced the distribution of extracts. Our findings can be utilized for further applications involving T. falconeri.

Post dock analysis

Among the docked compounds against acetylcholinesterase enzyme the ligand N-acetyldehydroanonaine and kanzonol E showed the highest binding affinity due to the lowest binding energies (-10.0 kcal/mol) compared to standard inhibitor (galantamine; -7.0 kcal/mol) (Fig. 3). Other ligands showed binding energies in the range -9.3 to -6.1 kcal/mol. While three ligands (quinic acid; -6.1, Rutacultin; -6.5, and Methylsyringin; -6.7 kcal/mol) showed binding affinity weaker than the standard (Table 5).
Table 5
Binding energies of identified compounds after docking with enzymes
Sr.No
Compounds
AChE
BChE
ɑ-Amylase
ɑ-glucosidase
Tyrosinase
1
6-Caffeoylsucrose
-7.8
-8.8
-7.3
-8.1
-7.7
2
N-Acetyldehydroanonaine
-10.0
-10.9
-9.4
-8.7
-8.3
3
Bryaflavan
-8.0
-7.7
-8.0
-7.0
-7.2
4
Purpuritenin A
-8.6
-8.8
-7.9
-7.6
-6.7
5
3-O-Feruloylquinic acid
-7.5
-8.5
-7.8
-8.2
-6.9
6
Brosimacutin C
-8.5
-9.3
-8.7
-7.9
-7.5
7
Quinic acid
-6.1
-5.9
-5.8
-5.8
-5.3
8
Murrayazolinine
-9.0
-10.6
-9.0
-9,4
-8.1
9
Rutacultin
-6.5
-8.0
-7.0
-6.8
-6.6
10
Castamollissin
-9.2
-9.5
-8.6
-8.6
-8.1
11
9-Acetoxyfukinanolide
-7.7
-8.6
-7.3
-7.6
-6.2
12
Matteucinol
-9.3
8.1
-7.8
-7.4
-7.2
13
Kaempferol 3-p-coumarate
-8.7
-10.4
-8.5
-8.3
-9.0
14
Isocordoin
-8.9
-9.0
-8.4
-7.8
-7.3
15
Methylsyringin
-6.7
-7.0
-7.3
-7.3
-6.3
16
Kanzonol E
-10.0
-9.8
-9.6
-9.1
-8.7
17
Standard
-7.0a
-8.8a
-7.6b
-8.3b
-5.4c
Standards: Galantamine (a), acarbose (b), and kojic acid (c). While AChE and BChE represent the acetylcholinesterase and butyrylcholinesterase respectively
N-Acetyldehydroanonaine also showed the highest binding affinity among the docked ligands against butyrylcholinesterase enzyme (-10.9 kcal mol) (Fig. 4). Herein, murrayazolinine, kaempferol 3-p-coumarate, kanzonol, E castamollissin, brosimacutin C, isocordoin (binding energies; -10.6, -10.4, -9.8, -9.5, -9.3, and -9.3 kcal/mol respectively) exhibited their higher binding affinities towards the enzyme compared to galantamine (-8.8 kcal/mol). While two compounds (6-caffeoylsucrose and purpuritenin A) were showing the binding affinity similar to the standard drug.
Eleven of the docked ligands showed ɑ-amylase inhibitory properties due to their lower binding energies compared to acarbose (standard; -7.6 kcal/mol). Kanzonol E showed the highest binding affinity to bind with the enzyme compared to all other docked ligands due to its lowest biding energy (-9.6 kcal/mol) (Fig. 5). While, five ligands expressed more binding energies than standard drug and showed represent their less contribution in the inhibition of the enzyme.
Four compounds exhibited higher binding affinities when docked against the ɑ-glucosidase enzyme, which attributes their contribution to the inhibitory effects of the plant extract. Murrayazolinine showed the highest binding affinity due to -9.4 kcal binding energy compared to the standard inhibitor (acarbose; -8.3 kcal/mol) (Fig. 6), while the compounds kanzonol E (-9.1), N-acetyldehydroanonaine (-8.7), and castamollissin (-8.6) also showed better binding than acarbose. Moreover, kaempferol 3-p-coumarate depicted similar binding affinity to the standard and all the remaining compounds showed lesser affinity due to their higher binding energies (-5.8 to -8.2 kcal/mol) than the standard drug.
For tyrosinase inhibition only quinic acid (-5.3 kcal/mol) showed lesser binding compared to the kojic acid (-5.4 kcal/mol) used as standard tyrosinase inhibitor. While, all the other docked ligands exhibited lower binding energies (-6.2 to -9.0 kcal/mol) showing their possible contribution in the tyrosinase inhibitory properties of the extract. The results further showed that kaempferol 3-p-coumarate has highest binding affinity towards tyrosinase enzyme due to its lowest binding energy (-9.0 kcal/mol) (Fig. 7).

ADME Analysis

The information anticipated for the medicinal chemistry, pharmacokinetics, lipophilicity, physicochemical properties, solubility, and drug resemblance of compounds assessed by SwissADME [70] is provided in Table 6. The molecular weights of the docked compounds were found within the range of 200–600 Da and to be 192.17–504.44, based on Lipinski's rule of five. The logP values were between -0.12 to 4.42, which were less than 5. All of the compound's products had an HBA number of 2 to 9 except for two compounds 6-caffeoylsucrose and castamollissin which can accept 14 and 13 HBA respectively. While the same trend was observed for HBD numbers the two compounds have HBD numbers 9 and 8 respectively while the remaining compounds have HBD numbers ≤ 5 [71]. The graphical model known as the Brain Or IntestinaL EstimateD Permeation (BOILED-Egg) technique determines the polarity and lipophilicity of small compounds. Concerning the possibility of oral absorption of medication candidates, this prediction offers a visual cue for the synthesis design of novel compounds [72]. Figure 8 displays a graphic estimate of these selectively docked compounds' gastrointestinal absorption and blood–brain barrier (BBB) penetration. The compounds N-Acetyldehydroanonaine, Purpuritenin A, Murrayazolinine, Rutacultin, 9-Acetoxyfukinanolide and Matteucinol were found in the BBB, while Bryaflavan, Brosimacutin C, Methylsyringin and Kanzonol E were found in white region. The white region contains those compounds which have good potential to be absorbed through the gastrointestinal tract. The compounds 3-O-Feruloylquinic acid, Quinic acid and Kaempferol 3-p-coumarate as indicated by the BOILED-Egg plot were presented in gray region. The gray region is designated for poor intestine absorption. The two compounds 6-caffeoylsucrose and castamollissin violated Lipinski's rule and were not shown in BOILED-Egg The compounds showed with blue spot, was discovered to be indicative of their high bioavailability. The compounds Bryaflavan, Brosimacutin C, Methylsyringin and Kanzonol E show great promise as gastrointestinal tract absorbers since they do not cross the blood–brain barrier. These substances have no adverse effects on central nervous system depression or sleepiness because they cannot penetrate the blood–brain barrier.
Table 6
Medicinal and Drug-like properties of selected compounds predicted using SwissADME
Compounds
Physicochemical Properties
Lipophilicity
Water Solubility
Pharmacokinetics
Drug likeness
Medicinal Chemistry
6-Caffeoylsucrose
Formula: C21H28O14
Molecular weight: 504.44 g/mol
Num. heavy atoms: 35
Num. arom. heavy atoms: 6
Fraction Csp3: 0.57
Num. rotatable bonds: 9
Num. H-bond acceptors: 14
Num. H-bond donors:9
Molar Refractivity: 111.55
TPSA: 236.06
Log Po/w (iLOGP): 1.77
Log Po/w (XLOGP3): -2.30
Log Po/w (WLOGP): -3.83
Log Po/w (MLOGP): -3.59
Log Po/w (SILICOS-IT): -2.65
Consensus Log Po/w: -2.12
Log S (ESOL): -1.05
Solubility: 4.48e + 01 mg/ml; 8.88e-02 mol/l
Class: Very soluble
Log S (Ali): -2.12
Solubility: 3.81e + 00 mg/ml; 7.56e-03 mol/l
Class: Soluble
Log S (SILICOS-IT): 2.10
Solubility: 6.39e + 04 mg/ml; 1.27e + 02 mol/l
Class: Soluble
GI absorption: Low
BBB permeant: No
P-gp substrate: No
CYP1A2 inhibitor: No
CYP2C19 inhibitor: No
CYP2C9 inhibitor: No
CYP2D6 inhibitor: No
CYP3A4 inhibitor: No
Log Kp (skin permeation): -11.01 cm/s
Lipinski: No; 3 violations: MW > 500, NorO > 10, NHorOH > 5
Ghose: No; 2 violations: MW > 480, WLOGP < -0.4
Veber: No; 1 violation: TPSA > 140
Egan: No; 1 violation: TPSA > 131.6
Muegge: No; 4 violations: XLOGP3 < -2, TPSA > 150, H-acc > 10, H-don > 5
Bioavailability Score: 0.17
PAINS: 1 alert: catechol_A
Brenk: 2 alerts: catechol, michael_acceptor_1
Leadlikeness: No; 2 violations: MW > 350, Rotors > 7
Synthetic accessibility: 5.56
N-Acetyldehydroanonaine
Formula: C19H15NO3
SMolecular weight: 305.33 g/mol
Num. heavy atoms: 23
Num. arom. heavy atoms: 14
Fraction Csp3: 0.21
Num. rotatable bonds: 1
Num. H-bond acceptors: 3
Num. H-bond donors: 0
Molar Refractivity: 92.52
TPSA: 38.77
Log Po/w (iLOGP): 2.93
Log Po/w (XLOGP3): 3.63
Log Po/w (WLOGP): 3.25
Log Po/w (MLOGP): 2.89
Log Po/w (SILICOS-IT): 3.88
Consensus Log Po/w: 3.32
Log S (ESOL): -4.40
Solubility: 1.20e-02 mg/ml; 3.94e-05 mol/l
Class: Moderately soluble
Log S (Ali): -4.13
Solubility: 2.25e-02 mg/ml; 7.38e-05 mol/l
Class: Moderately soluble
Log S (SILICOS-IT): -5.71
Solubility: 5.94e-04 mg/ml; 1.95e-06 mol/l
Class: Moderately soluble
GI absorption: High
BBB permeant: Yes
P-gp substrate: Yes
CYP1A2 inhibitor: Yes
CYP2C19 inhibitor: Yes
CYP2C9 inhibitor: Yes
CYP2D6 inhibitor: No
CYP3A4 inhibitor: Yes
Log Kp (skin permeation): -5.59 cm/s
Lipinski: Yes; 0 violation
Ghose: Yes
Veber: Yes
Egan: Yes
Muegge: Yes
Bioavailability Score: 0.55
PAINS: 0 alert
Brenk: 1 alert: polycyclic_aromatic_hydrocarbon_3
Leadlikeness: No; 1 violation: XLOGP3 > 3.5
Synthetic accessibility: 2.53
Bryaflavan
Formula: C17H18O6
Molecular weight: 318.32 g/mol
Num. heavy atoms: 23
Num. arom. heavy atoms: 12
Fraction Csp3: 0.29
Num. rotatable bonds: 3
Num. H-bond acceptors: 6
Num. H-bond donors:3
Molar Refractivity: 84.13
TPSA: 88.38
Log Po/w (iLOGP): 2.43
Log Po/w (XLOGP3): 2.55
Log Po/w (WLOGP): 2.54
Log Po/w (MLOGP): 1.00
Log Po/w (SILICOS-IT): 2.44
Consensus Log Po/w: 2.19
Log S (ESOL): -3.61
Solubility: 7.85e-02 mg/ml; 2.47e-04 mol/l
Class: Soluble
Log S (Ali): -4.05
Solubility: 2.82e-02 mg/ml; 8.84e-05 mol/l
Class: Moderately soluble
Log S (SILICOS-IT): -3.77
Solubility: 5.47e-02 mg/ml; 1.72e-04 mol/l
Class: Soluble
GI absorption: High
BBB permeant: No
P-gp substrate: Yes
CYP1A2 inhibitor: Yes
CYP2C19 inhibitor: No
CYP2C9 inhibitor: No
CYP2D6 inhibitor: Yes
CYP3A4 inhibitor: Yes
Log Kp (skin permeation): -6.43 cm/s
Lipinski: Yes; 0 violation
Ghose: Yes
Veber: Yes
Egan: Yes
Muegge: Yes
Bioavailability Score: 0.55
PAINS: 1 alert: catechol_A
Brenk: 1 alert: catechol
Leadlikeness: Yes
Synthetic accessibility: 3.41
Purpuritenin A
Formula: C19H16O3
Molecular weight: 292.33 g/mol
Num. heavy atoms: 22
Num. arom. heavy atoms: 15
Fraction Csp3: 0.11
Num. rotatable bonds: 4
Num. H-bond acceptors: 3
Num. H-bond donors: 0
Molar Refractivity: 87.48
TPSA: 39.44
Log Po/w (iLOGP): 3.14
Log Po/w (XLOGP3): 4.43
Log Po/w (WLOGP): 4.54
Log Po/w (MLOGP): 2.59
Log Po/w (SILICOS-IT): 4.94
Consensus Log Po/w: 3.93
Log S (ESOL): -4.68
Solubility: 6.05e-03 mg/ml; 2.07e-05 mol/l
Class: Moderately soluble
Log S (Ali): -4.98
Solubility: 3.09e-03 mg/ml; 1.06e-05 mol/l
Class: Moderately soluble
Log S (SILICOS-IT): -6.35
Solubility: 1.31e-04 mg/ml; 4.47e-07 mol/l
Class: Poorly soluble
GI absorption: High
BBB permeant: Yes
P-gp substrate: No
CYP1A2 inhibitor: Yes
CYP2C19 inhibitor: Yes
CYP2C9 inhibitor: Yes
CYP2D6 inhibitor: No
CYP3A4 inhibitor: No
Log Kp (skin permeation): -4.94 cm/s
Lipinski: Yes; 0 violation
Ghose: Yes
Veber: Yes
Egan: Yes
Muegge: Yes
Bioavailability Score: 0.55
PAINS: 0 alert
Brenk: 1 alert: michael_acceptor_1
Leadlikeness: No; 1 violation: XLOGP3 > 3.5
Synthetic accessibility: 3.13
3-O-Feruloylquinic acid
Formula: C17H20O9
Molecular weight: 368.34 g/mol
Num. heavy atoms: 26
Num. arom. heavy atoms: 6
Fraction Csp3: 0.41
Num. rotatable bonds: 6
Num. H-bond acceptors: 9
Num. H-bond donors: 5
Molar Refractivity: 87.97
TPSA: 153.75
Log Po/w (iLOGP): 1.47
Log Po/w (XLOGP3): -0.10
Log Po/w (WLOGP): -0.45
Log Po/w (MLOGP): -0.81
Log Po/w (SILICOS-IT): -0.07
Consensus Log Po/w: 0.01:
Log S (ESOL): -1.84
Solubility: 5.38e + 00 mg/ml; 1.46e-02 mol/l
Class: Very soluble
Log S (Ali): -2.68
Solubility: 7.76e-01 mg/ml; 2.11e-03 mol/l
Class: Soluble
Log S (SILICOS-IT): -0.29
Solubility: 1.89e + 02 mg/ml; 5.13e-01 mol/l
Class: Soluble
GI absorption: Low
BBB permeant: No
P-gp substrate: No
CYP1A2 inhibitor: No
CYP2C19 inhibitor: No
CYP2C9 inhibitor: No
CYP2D6 inhibitor: No
CYP3A4 inhibitor: No
Log Kp (skin permeation): -8.62 cm/s
Lipinski: Yes; 0 violation
Ghose: No; 1 violation: WLOGP < -0.4
Veber: No; 1 violation: TPSA > 140
Egan: No; 1 violation: TPSA > 131.6
Muegge: No; 1 violation: TPSA > 150
Bioavailability Score: 0.11
PAINS: 0 alert
Brenk: 1 alert: michael_acceptor_1
Leadlikeness: No; 1 violation: MW > 350
Synthetic accessibility: 4.25
Brosimacutin C
Formula: C20H22O5
Molecular weight: 342.39 g/mol
Num. heavy atoms: 25
Num. arom. heavy atoms: 12
Fraction Csp3: 0.35
Num. rotatable bonds: 4
Num. H-bond acceptors: 5
Num. H-bond donors: 3
Molar Refractivity: 94.94
TPSA: 86.99
Log Po/w (iLOGP): 2.62
Log Po/w (XLOGP3): 2.75
Log Po/w (WLOGP): 3.18
Log Po/w (MLOGP): 1.63
Log Po/w (SILICOS-IT): 3.62
Consensus Log Po/w: 2.76
Log S (ESOL): -3.79
Solubility: 5.60e-02 mg/ml; 1.63e-04 mol/l
Class: Soluble
Log S (Ali): -4.23
Solubility: 2.01e-02 mg/ml; 5.87e-05 mol/l
Class: Moderately soluble
Log S (SILICOS-IT): -5.02
Solubility: 3.30e-03 mg/ml; 9.64e-06 mol/l
Class: Moderately soluble
GI absorption: High
BBB permeant: No
P-gp substrate: Yes
CYP1A2 inhibitor: No
CYP2C19 inhibitor: No
CYP2C9 inhibitor: No
CYP2D6 inhibitor: Yes
CYP3A4 inhibitor: No
Log Kp (skin permeation): -6.44 cm/s
Lipinski: Yes; 0 violation
Ghose: Yes
Veber: Yes
Egan: Yes
Muegge: Yes
Bioavailability Score: 0.55
PAINS: 0 alert
Brenk: 0 alert
Leadlikeness: Yes
Synthetic accessibility: 3.61
Quinic acid
Formula: C7H12O6
Molecular weight: 192.17 g/mol
Num. heavy atoms: 13
Num. arom. heavy atoms: 0
Fraction Csp3: 0.86
Num. rotatable bonds: 1
Num. H-bond acceptors: 6
Num. H-bond donors: 5
Molar Refractivity: 40.11
TPSA: 118.22
Log Po/w (iLOGP): -0.12
Log Po/w (XLOGP3): -2.37
Log Po/w (WLOGP): -2.32
Log Po/w (MLOGP): -2.14
Log Po/w (SILICOS-IT): -1.82
Consensus Log Po/w: -1.75
Log S (ESOL): 0.53
Solubility: 6.48e + 02 mg/ml; 3.37e + 00 mol/l
Class: Highly soluble
Log S (Ali): 0.43
Solubility: 5.12e + 02 mg/ml; 2.66e + 00 mol/l
Class: Highly soluble
Log S (SILICOS-IT): 2.08
Solubility: 2.30e + 04 mg/ml; 1.20e + 02 mol/l
Class: Soluble
GI absorption: Low
BBB permeant: No
P-gp substrate: Yes
CYP1A2 inhibitor: No
CYP2C19 inhibitor: No
CYP2C9 inhibitor: No
CYP2D6 inhibitor: No
CYP3A4 inhibitor: No
Log Kp (skin permeation): -9.15 cm/s
Lipinski: Yes; 0 violation
Ghose: No; 1 violation: WLOGP < -0.4
Veber: Yes
Egan: Yes
Muegge: No; 2 violations: MW < 200, XLOGP3 < -2
Bioavailability Score: 0.56
PAINS: 0 alert
Brenk: 0 alert
Leadlikeness: No; 1 violation: MW < 250
Synthetic accessibility: 3.34
Murrayazolinine
Formula: C23H27NO2 Molecular weight: 349.47 g/mol
Num. heavy atoms: 26
Num. arom. heavy atoms: 13
Fraction Csp3: 0.48
Num. rotatable bonds: 1
Num. H-bond acceptors: 2
Num. H-bond donors: 2
Molar Refractivity: 107.69
TPSA: 45.25
Log Po/w (iLOGP): 3.42
Log Po/w (XLOGP3): 4.84
Log Po/w (WLOGP): 5.44
Log Po/w (MLOGP): 3.93
Log Po/w (SILICOS-IT): 5.46
Consensus Log Po/w: 4.62
Log S (ESOL): -5.36
Solubility: 1.53e-03 mg/ml; 4.37e-06 mol/l
Class: Moderately soluble
Log S (Ali): -5.52
Solubility: 1.05e-03 mg/ml; 2.99e-06 mol/l
Class: Moderately soluble
Log S (SILICOS-IT): -6.87
Solubility: 4.71e-05 mg/ml; 1.35e-07 mol/l
Class: Poorly soluble
GI absorption: High
BBB permeant: Yes
P-gp substrate: Yes
CYP1A2 inhibitor: No
CYP2C19 inhibitor: No
CYP2C9 inhibitor: No
CYP2D6 inhibitor: Yes
CYP3A4 inhibitor: No
Log Kp (skin permeation): -5.00 cm/s
Lipinski: Yes; 0 violation
Ghose: Yes
Veber: Yes
Egan: Yes
Muegge: Yes
Bioavailability Score: 0.55
PAINS: 0 alert
Brenk: 0 alert
Leadlikeness: No; 1 violation: XLOGP3 > 3.5
Synthetic accessibility: 4.53
Rutacultin
Formula: C16H18O4
Molecular weight: 274.31 g/mol
Num. heavy atoms: 20
Num. arom. heavy atoms: 10
Fraction Csp3: 0.31
Num. rotatable bonds: 4
Num. H-bond acceptors: 4
Num. H-bond donors: 0
Molar Refractivity: 79.07
TPSA: 48.67
Log Po/w (iLOGP): 3.25
Log Po/w (XLOGP3): 3.81
Log Po/w (WLOGP): 3.27
Log Po/w (MLOGP): 2.29
Log Po/w (SILICOS-IT): 4.09
Consensus Log Po/w: 3.34
Log S (ESOL): -4.05
Solubility: 2.46e-02 mg/ml; 8.97e-05 mol/l
Class: Moderately soluble
Log S (Ali): -4.53
Solubility: 8.15e-03 mg/ml; 2.97e-05 mol/l
Class: Moderately soluble
Log S (SILICOS-IT): -5.13
Solubility: 2.01e-03 mg/ml; 7.33e-06 mol/l
Class: Moderately soluble
GI absorption: High
BBB permeant: Yes
P-gp substrate: No
CYP1A2 inhibitor: Yes
CYP2C19 inhibitor: Yes
CYP2C9 inhibitor: Yes
CYP2D6 inhibitor: No
CYP3A4 inhibitor: No
Log Kp (skin permeation): -5.27 cm/s
Lipinski: Yes; 0 violation
Ghose: Yes
Veber: Yes
Egan: Yes
Muegge: Yes
Bioavailability Score: 0.55
PAINS: 0 alert
Brenk: 2 alerts: cumarine, isolated_alkene
Leadlikeness: No; 1 violation: XLOGP3 > 3.5
Synthetic accessibility: 3.27
Castamollissin
Formula: C20H20O13
Molecular weight: 468.37 g/mol
Num. heavy atoms: 33
Num. arom. heavy atoms: 12
Fraction Csp3: 0.30
Num. rotatable bonds: 7
Num. H-bond acceptors: 13
Num. H-bond donors: 8
Molar Refractivity: 105.73
TPSA: 223.67 Å2
Log Po/w (iLOGP): 0.93
Log Po/w (XLOGP3): -0.87
Log Po/w (WLOGP): -0.93
Log Po/w (MLOGP): -2.37
Log Po/w (SILICOS-IT): -1.34
Consensus Log Po/w: -0.92
Log S (ESOL): -2.00
Solubility: 4.65e + 00 mg/ml; 9.93e-03 mol/l
Class: Soluble
Log S (Ali): -3.35
Solubility: 2.11e-01 mg/ml; 4.51e-04 mol/l
Class: Soluble
Log S (SILICOS-IT): 0.07
Solubility: 5.49e + 02 mg/ml; 1.17e + 00 mol/l
Class: Soluble
GI absorption: Low
BBB permeant: No
P-gp substrate: No
CYP1A2 inhibitor: No
CYP2C19 inhibitor: No
CYP2C9 inhibitor: No
CYP2D6 inhibitor: No
CYP3A4 inhibitor: No
Log Kp (skin permeation): -9.77 cm/s
Lipinski: No; 2 violations: NorO > 10, NHorOH > 5
Ghose: No; 1 violation: WLOGP < -0.4
Veber: No; 1 violation: TPSA > 140
Egan: No; 1 violation: TPSA > 131.6
Muegge: No; 3 violations: TPSA > 150, H-acc > 10, H-don > 5
Bioavailability Score: 0.17
PAINS: 1 alert: catechol_A
Brenk: 2 alerts: aldehyde, catechol
Leadlikeness: No; 1 violation: MW > 350
Synthetic accessibility: 4.88
9-Acetoxyfukinanolide
Formula: C17H24O4
Molecular weight: 292.37 g/mol
Num. heavy atoms: 21
Num. arom. heavy atoms: 0
Fraction Csp3: 0.76
Num. rotatable bonds: 2
Num. H-bond acceptors: 4
Num. H-bond donors: 0
Molar Refractivity: 79.07
TPSA: 52.60 Å2
Log Po/w (iLOGP): 2.48
Log Po/w (XLOGP3): 2.92
Log Po/w (WLOGP): 2.86
Log Po/w (MLOGP): 2.92
Log Po/w (SILICOS-IT): 3.18
Consensus Log Po/w: 2.87
Log S (ESOL): -3.36
Solubility: 1.28e-01 mg/ml; 4.36e-04 mol/l
Class: Soluble
Log S (Ali): -3.69
Solubility: 6.03e-02 mg/ml; 2.06e-04 mol/l
Class: Soluble
Log S (SILICOS-IT): -3.35
Solubility: 1.30e-01 mg/ml; 4.45e-04 mol/l
Class: Soluble
GI absorption: High
BBB permeant: Yes
P-gp substrate: No
CYP1A2 inhibitor: No
CYP2C19 inhibitor: No
CYP2C9 inhibitor: No
CYP2D6 inhibitor: No
CYP3A4 inhibitor: No
Log Kp (skin permeation): -6.01 cm/s
Lipinski: Yes; 0 violation
Ghose: Yes
Veber: Yes
Egan: Yes
Muegge: Yes
Bioavailability Score: 0.55
PAINS: 0 alert
Brenk: 2 alerts: isolated_alkene, more_than_2_esters
Leadlikeness: Yes
Synthetic accessibility: 4.88
Matteucinol
Formula: C18H18O5
Molecular weight: 314.33 g/mol
Num. heavy atoms: 23
Num. arom. heavy atoms: 12
Fraction Csp3: 0.28
Num. rotatable bonds: 2
Num. H-bond acceptors: 5
Num. H-bond donors: 2
Molar Refractivity: 85.97
TPSA: 75.99 Å2
Log Po/w (iLOGP): 2.95
Log Po/w (XLOGP3): 3.45
Log Po/w (WLOGP): 3.11
Log Po/w (MLOGP): 1.44
Log Po/w (SILICOS-IT): 3.58
Consensus Log Po/w: 2.91
Log S (ESOL): -4.22
Solubility: 1.91e-02 mg/ml; 6.08e-05 mol/l
Class: Moderately soluble
Log S (Ali): -4.73
Solubility: 5.89e-03 mg/ml; 1.87e-05 mol/l
Class: Moderately soluble
Log S (SILICOS-IT): -4.88
Solubility: 4.16e-03 mg/ml; 1.32e-05 mol/l
Class: Moderately soluble
GI absorption: High
BBB permeant: Yes
P-gp substrate: No
CYP1A2 inhibitor: Yes
CYP2C19 inhibitor: Yes
CYP2C9 inhibitor: Yes
CYP2D6 inhibitor: Yes
CYP3A4 inhibitor: Yes
Log Kp (skin permeation): -5.77 cm/s
Lipinski: Yes; 0 violation
Ghose: Yes
Veber: Yes
Egan: Yes
Muegge: Yes
Bioavailability Score: 0.55
PAINS: 0 alert
Brenk: 0 alert
Leadlikeness: Yes
Synthetic accessibility: 3.37
Kaempferol 3-p-coumarate
Formula: C24H16O8
Molecular weight: 432.38 g/mol
Num. heavy atoms: 32
Num. arom. heavy atoms: 22
Fraction Csp3: 0.00
Num. rotatable bonds: 5
Num. H-bond acceptors: 8
Num. H-bond donors: 4
Molar Refractivity: 117.12
TPSA: 137.43 Å2
Log Po/w (iLOGP): 2.67
Log Po/w (XLOGP3): 4.36
Log Po/w (WLOGP): 3.79
Log Po/w (MLOGP): 1.17
Log Po/w (SILICOS-IT): 3.65
Consensus Log Po/w: 3.13
Log S (ESOL): -5.45
Solubility: 1.55e-03 mg/ml; 3.58e-06 mol/l
Class: Moderately soluble
Log S (Ali): -6.96
Solubility: 4.72e-05 mg/ml; 1.09e-07 mol/l
Class: Poorly soluble
Log S (SILICOS-IT): -6.02
Solubility: 4.16e-04 mg/ml; 9.62e-07 mol/l
Class: Poorly soluble
GI absorption: Low
BBB permeant: No
P-gp substrate: No
CYP1A2 inhibitor: No
CYP2C19 inhibitor: No
CYP2C9 inhibitor: Yes
CYP2D6 inhibitor: No
CYP3A4 inhibitor: No
Log Kp (skin permeation): -5.84 cm/s
Lipinski: Yes; 0 violation
Ghose: Yes
Veber: Yes
Egan: No; 1 violation: TPSA > 131.6
Muegge: Yes
Bioavailability Score: 0.55
PAINS: 0 alert
Brenk: 1 alert: michael_acceptor_1
Leadlikeness: No; 2 violations: MW > 350, XLOGP3 > 3.5
Synthetic accessibility: 3.79
Isocordoin
Formula: C20H20O3
Molecular weight: 308.37 g/mol
Num. heavy atoms: 23
Num. arom. heavy atoms: 12
Fraction Csp3: 0.15
Num. rotatable bonds: 5
Num. H-bond acceptors: 3
Num. H-bond donors: 2
Molar Refractivity: 94.01
TPSA: 57.53 Å22
Log Po/w (iLOGP): 3.39
Log Po/w (XLOGP3): 5.46
Log Po/w (WLOGP): 4.39
Log Po/w (MLOGP): 3.28
Log Po/w (SILICOS-IT): 4.70
Consensus Log Po/w: 4.24
Log S (ESOL): -5.25
Solubility: 1.74e-03 mg/ml; 5.65e-06 mol/l
Class: Moderately soluble
Log S (Ali): -6.43
Solubility: 1.16e-04 mg/ml; 3.76e-07 mol/l
Class: Poorly soluble
Log S (SILICOS-IT): -5.06
Solubility: 2.70e-03 mg/ml; 8.77e-06 mol/l
Class: Moderately soluble
GI absorption: High
BBB permeant: Yes
P-gp substrate: No
CYP1A2 inhibitor: Yes
CYP2C19 inhibitor: Yes
CYP2C9 inhibitor: Yes
CYP2D6 inhibitor: No
CYP3A4 inhibitor: Yes
Log Kp (skin permeation): -4.30 cm/s
Lipinski: Yes; 0 violation
Ghose: Yes
Veber: Yes
Egan: Yes
Muegge: No; 1 violation: XLOGP3 > 5
Bioavailability Score: 0.55
PAINS: 0 alert
Brenk: 2 alerts: isolated_alkene, michael_acceptor_1
Leadlikeness: No; 1 violation: XLOGP3 > 3.5
Synthetic accessibility: 2.99
Methylsyringin
Formula: C18H26O9
Molecular weight: 386.39 g/mol
Num. heavy atoms: 27
Num. arom. heavy atoms: 6
Fraction Csp3: 0.56
Num. rotatable bonds: 8
Num. H-bond acceptors: 9
Num. H-bond donors: 4
Molar Refractivity: 94.36
TPSA: 127.07 Å2
Log Po/w (iLOGP): 2.46
Log Po/w (XLOGP3): -0.77
Log Po/w (WLOGP): -0.57
Log Po/w (MLOGP): -1.35
Log Po/w (SILICOS-IT): 0.42
Consensus Log Po/w: 0.04
Log S (ESOL): -1.39
Solubility: 1.59e + 01 mg/ml; 4.10e-02 mol/l
Class: Very soluble
Log S (Ali): -1.42
Solubility: 1.47e + 01 mg/ml; 3.80e-02 mol/l
Class: Very soluble
Log S (SILICOS-IT): -1.03
Solubility: 3.61e + 01 mg/ml; 9.33e-02 mol/l
Class: Soluble
GI absorption: High
BBB permeant: No
P-gp substrate: Yes
CYP1A2 inhibitor: No
CYP2C19 inhibitor: No
CYP2C9 inhibitor: No
CYP2D6 inhibitor: No
CYP3A4 inhibitor: No
Log Kp (skin permeation): -9.20 cm/s
Lipinski: Yes; 0 violation
Ghose: No; 1 violation: WLOGP < -0.4
Veber: Yes
Egan: Yes
Muegge: Yes
Bioavailability Score: 0.55
PAINS: 0 alert
Brenk: 0 alert
Leadlikeness: No; 2 violations: MW > 350, Rotors > 7
Synthetic accessibility: 4.88
Kanzonol E
Formula: C25H24O4
Molecular weight: 388.46 g/mol
Num. heavy atoms: 29
Num. arom. heavy atoms: 16
Fraction Csp3: 0.24
Num. rotatable bonds: 3
Num. H-bond acceptors: 4
Num. H-bond donors: 1
Molar Refractivity: 117.78
TPSA: 59.67 Å2
Log Po/w (iLOGP): 4.24
Log Po/w (XLOGP3): 5.67
Log Po/w (WLOGP): 5.75
Log Po/w (MLOGP): 3.20
Log Po/w (SILICOS-IT): 6.14
Consensus Log Po/w: 5.00
Log S (ESOL): -6.03
Solubility: 3.62e-04 mg/ml; 9.32e-07 mol/l
Class: Poorly soluble
Log S (Ali): -6.69
Solubility: 7.97e-05 mg/ml; 2.05e-07 mol/l
Class: Poorly soluble
Log S (SILICOS-IT): -7.62
Solubility: 9.34e-06 mg/ml; 2.40e-08 mol/l
Class: Poorly soluble
GI absorption: High
BBB permeant: No
P-gp substrate: No
CYP1A2 inhibitor: No
CYP2C19 inhibitor: Yes
CYP2C9 inhibitor: Yes
CYP2D6 inhibitor: No
CYP3A4 inhibitor: No
Log Kp (skin permeation): -4.64 cm/s
Lipinski: Yes; 0 violation
Ghose: No; 1 violation: WLOGP > 5.6
Veber: Yes
Egan: Yes
Muegge: No; 1 violation: XLOGP3 > 5
Bioavailability Score: 0.55
PAINS: 0 alert
Brenk: 1 alert: isolated_alkene
Leadlikeness: No; 2 violations: MW > 350, XLOGP3 > 3.5
Synthetic accessibility: 4.14

Acknowledgements

The authors would like to extend their sincere appreciation to the Researchers Supporting Project Number (RSP2024R356), King Saud University, Riyadh, Saudi Arabia.

Code availability

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Tsevegsuren N, Fujimoto K, Christie WW, Endo Y. Occurrence of a novel cis, cis, cis-octadeca-3,9,12-trienoic (Z,Z,Z-octadeca-3,9,12-trienoic) acid in Chrysanthemum (tanacetum) zawadskii herb seed oil. Lipids. 2003;38(5):573–8.PubMedCrossRef Tsevegsuren N, Fujimoto K, Christie WW, Endo Y. Occurrence of a novel cis, cis, cis-octadeca-3,9,12-trienoic (Z,Z,Z-octadeca-3,9,12-trienoic) acid in Chrysanthemum (tanacetum) zawadskii herb seed oil. Lipids. 2003;38(5):573–8.PubMedCrossRef
2.
Zurück zum Zitat Bano A, Ahmad M, Hadda TB, Saboor A, Sultana S, Zafar M, Khan MPZ, Arshad M, Ashraf MA. Quantitative ethnomedicinal study of plants used in the skardu valley at high altitude of Karakoram-Himalayan range, Pakistan Pakistan. J Ethnobiol Ethnomed. 2014;10(1):1–18.CrossRef Bano A, Ahmad M, Hadda TB, Saboor A, Sultana S, Zafar M, Khan MPZ, Arshad M, Ashraf MA. Quantitative ethnomedicinal study of plants used in the skardu valley at high altitude of Karakoram-Himalayan range, Pakistan Pakistan. J Ethnobiol Ethnomed. 2014;10(1):1–18.CrossRef
3.
Zurück zum Zitat Khan SW, Abbas Q, Hassan SN, Khan H, Hussain A. Medicinal Plants of Turmic Valley (Central Karakoram National Park), Gilgit-Baltistan, Pakistan. J Biores Manage. 2015;2(2):11. Khan SW, Abbas Q, Hassan SN, Khan H, Hussain A. Medicinal Plants of Turmic Valley (Central Karakoram National Park), Gilgit-Baltistan, Pakistan. J Biores Manage. 2015;2(2):11.
4.
Zurück zum Zitat Chawla A, Rajkumar S, Singh K, Lal B, Singh R, Thukral A. Plant species diversity along an altitudinal gradient of Bhabha Valley in western Himalaya. J Mt Sci. 2008;5(2):157–77.CrossRef Chawla A, Rajkumar S, Singh K, Lal B, Singh R, Thukral A. Plant species diversity along an altitudinal gradient of Bhabha Valley in western Himalaya. J Mt Sci. 2008;5(2):157–77.CrossRef
5.
Zurück zum Zitat Abbas Z, Khan SM, Alam J, Khan SW, Abbasi AM. Medicinal plants used by inhabitants of the Shigar Valley Baltistan region of Karakorum range-Pakistan. J Ethnobiol Ethnomed. 2017;13(1):1–15.CrossRef Abbas Z, Khan SM, Alam J, Khan SW, Abbasi AM. Medicinal plants used by inhabitants of the Shigar Valley Baltistan region of Karakorum range-Pakistan. J Ethnobiol Ethnomed. 2017;13(1):1–15.CrossRef
6.
Zurück zum Zitat Molina-Torres J, Garcia-Chavez A, Ramirez-Chavez E. Antimicrobial properties of alkamides present in flavouring plants traditionally used in Mesoamerica: affinin and capsaicin. J Ethnopharmacol. 1999;64(3):241–8.PubMedCrossRef Molina-Torres J, Garcia-Chavez A, Ramirez-Chavez E. Antimicrobial properties of alkamides present in flavouring plants traditionally used in Mesoamerica: affinin and capsaicin. J Ethnopharmacol. 1999;64(3):241–8.PubMedCrossRef
7.
Zurück zum Zitat Shahhoseini R, Azizi M, Asili J, Moshtaghi N, Samiei L. Comprehensive assessment of phytochemical potential of Tanacetum parthenium (L.): phenolic compounds, antioxidant activity, essential oil and parthenolide. J Essential Oil Bearing Plants. 2019;22(3):614–29.CrossRef Shahhoseini R, Azizi M, Asili J, Moshtaghi N, Samiei L. Comprehensive assessment of phytochemical potential of Tanacetum parthenium (L.): phenolic compounds, antioxidant activity, essential oil and parthenolide. J Essential Oil Bearing Plants. 2019;22(3):614–29.CrossRef
8.
Zurück zum Zitat Ali Ş, Kürkçüoğlu M, Bitiş L, Doğan A, Başer K. Essential oil composition of different parts of Tanacetum cilicicum (Boiss.) Grierson. Nat Volatiles Essential Oils. 2020;7(3):18–28. Ali Ş, Kürkçüoğlu M, Bitiş L, Doğan A, Başer K. Essential oil composition of different parts of Tanacetum cilicicum (Boiss.) Grierson. Nat Volatiles Essential Oils. 2020;7(3):18–28.
9.
Zurück zum Zitat Abad M, Bermejo P, Villar A. An approach to the genus Tanacetum L. (Compositae): phytochemical and pharmacological review. Phytother Res. 1995;9(2):79–92.CrossRef Abad M, Bermejo P, Villar A. An approach to the genus Tanacetum L. (Compositae): phytochemical and pharmacological review. Phytother Res. 1995;9(2):79–92.CrossRef
10.
Zurück zum Zitat Holland R, Hendriks JH, Vebeek AL, Mravunac M, Schuurmans Stekhoven JH. Extent, distribution, and mammographic/histological correlations of breast ductal carcinoma in situ. Lancet. 1990;335(8688):519–22.PubMedCrossRef Holland R, Hendriks JH, Vebeek AL, Mravunac M, Schuurmans Stekhoven JH. Extent, distribution, and mammographic/histological correlations of breast ductal carcinoma in situ. Lancet. 1990;335(8688):519–22.PubMedCrossRef
11.
Zurück zum Zitat Hitmi A, Coudret A, Barthomeuf C. The production of pyrethrins by plant cell and tissue cultures of Chrysanthemum cinerariaefolium and Tagetes species. Crit Rev Biochem Mol Biol. 2000;35(5):317–37.PubMedCrossRef Hitmi A, Coudret A, Barthomeuf C. The production of pyrethrins by plant cell and tissue cultures of Chrysanthemum cinerariaefolium and Tagetes species. Crit Rev Biochem Mol Biol. 2000;35(5):317–37.PubMedCrossRef
12.
Zurück zum Zitat Sadique J, Chandra T, Thenmozhi V, Elango V. The anti-inflammatory activity of Enicostemma littorale and Mollugo cerviana. Biochem Med Metab Biol. 1987;37(2):167–76.PubMedCrossRef Sadique J, Chandra T, Thenmozhi V, Elango V. The anti-inflammatory activity of Enicostemma littorale and Mollugo cerviana. Biochem Med Metab Biol. 1987;37(2):167–76.PubMedCrossRef
13.
Zurück zum Zitat Mladenova K, Tsankova E, van Hung D. New sesquiterpenoids from Chrysanthemum indicum var. tuneful. Planta Med. 1988;54(6):553–5.PubMedCrossRef Mladenova K, Tsankova E, van Hung D. New sesquiterpenoids from Chrysanthemum indicum var. tuneful. Planta Med. 1988;54(6):553–5.PubMedCrossRef
14.
Zurück zum Zitat Kumar V, Tyagi D. Chemical composition and biological activities of essential oils of genus Tanacetum-a review. J Pharmacognosy Phytochem. 2013;2(3):155–9. Kumar V, Tyagi D. Chemical composition and biological activities of essential oils of genus Tanacetum-a review. J Pharmacognosy Phytochem. 2013;2(3):155–9.
15.
16.
Zurück zum Zitat Chavez F, Strutton P, Friederich G, Feely R, Feldman G, Foley D, McPhaden M. Biological and chemical response of the equatorial Pacific Ocean to the 1997–98 El Niño. Science. 1999;286(5447):2126–31.PubMedCrossRef Chavez F, Strutton P, Friederich G, Feely R, Feldman G, Foley D, McPhaden M. Biological and chemical response of the equatorial Pacific Ocean to the 1997–98 El Niño. Science. 1999;286(5447):2126–31.PubMedCrossRef
17.
Zurück zum Zitat Hwang SH, Kim HY, Quispe YNG, Wang Z, Zuo G, Lim SS. Aldose Reductase, Protein Glycation inhibitory and antioxidant of Peruvian medicinal plants: the case of Tanacetum parthenium L. and its constituents. Molecules. 2019;24(10):2010.PubMedPubMedCentralCrossRef Hwang SH, Kim HY, Quispe YNG, Wang Z, Zuo G, Lim SS. Aldose Reductase, Protein Glycation inhibitory and antioxidant of Peruvian medicinal plants: the case of Tanacetum parthenium L. and its constituents. Molecules. 2019;24(10):2010.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Schinella GR, Giner RM, Recio MC, Mordujovich de Buschiazzo P, Rios JL, Manez S. Anti-inflammatory effects of South American Tanacetum vulgare. J Pharm Pharmacol. 1998;50(9):1069–74.PubMedCrossRef Schinella GR, Giner RM, Recio MC, Mordujovich de Buschiazzo P, Rios JL, Manez S. Anti-inflammatory effects of South American Tanacetum vulgare. J Pharm Pharmacol. 1998;50(9):1069–74.PubMedCrossRef
19.
Zurück zum Zitat Mordujovich-Buschiazzo P, Balsa E, Buschiazzo H, Mandrile E, Rosella M. Anti-inflammatory activity of Tanacetum vulgare. Fitoterapia. 1996;67(4):319–22. Mordujovich-Buschiazzo P, Balsa E, Buschiazzo H, Mandrile E, Rosella M. Anti-inflammatory activity of Tanacetum vulgare. Fitoterapia. 1996;67(4):319–22.
20.
Zurück zum Zitat Bagci E, Kursat M, Kocak A, Gur S. Composition and antimicrobial activity of the essential oils of Tanacetum balsamita L. subsp balsamita and T. chiliophyllum (Fisch. et Mey.) Schultz Bip var chiliophyllum (Asteraceae) from Turkey. J Essential Oil Bearing Plants. 2008;11(5):476–84.CrossRef Bagci E, Kursat M, Kocak A, Gur S. Composition and antimicrobial activity of the essential oils of Tanacetum balsamita L. subsp balsamita and T. chiliophyllum (Fisch. et Mey.) Schultz Bip var chiliophyllum (Asteraceae) from Turkey. J Essential Oil Bearing Plants. 2008;11(5):476–84.CrossRef
21.
Zurück zum Zitat Keskitalo M, Pehu E, Simon JE. Variation in volatile compounds from tansy (Tanacetum vulgare L.) related to genetic and morphological differences of genotypes. Biochem Syst Ecol. 2001;29(3):267–85.PubMedCrossRef Keskitalo M, Pehu E, Simon JE. Variation in volatile compounds from tansy (Tanacetum vulgare L.) related to genetic and morphological differences of genotypes. Biochem Syst Ecol. 2001;29(3):267–85.PubMedCrossRef
22.
Zurück zum Zitat Bandonien D, Pukalskas A, Venskutonis P, Gruzdien D. Preliminary screening of antioxidant activity of some plant extracts in rapeseed oil. Food Res Int. 2000;33(9):785–91.CrossRef Bandonien D, Pukalskas A, Venskutonis P, Gruzdien D. Preliminary screening of antioxidant activity of some plant extracts in rapeseed oil. Food Res Int. 2000;33(9):785–91.CrossRef
23.
Zurück zum Zitat Petrovic SD, Dobric S, Bokonjic D, Niketic M, Garcia-Pineres A, Merfort I. Evaluation of Tanacetum larvatum for an anti-inflammatory activity and for the protection against indomethacin-induced ulcerogenesis in rats. J Ethnopharmacol. 2003;87(1):109–13.PubMedCrossRef Petrovic SD, Dobric S, Bokonjic D, Niketic M, Garcia-Pineres A, Merfort I. Evaluation of Tanacetum larvatum for an anti-inflammatory activity and for the protection against indomethacin-induced ulcerogenesis in rats. J Ethnopharmacol. 2003;87(1):109–13.PubMedCrossRef
24.
Zurück zum Zitat Tournier H, Schinella G, de Balsa EM, Buschiazzo H, Manez S, Mordujovich de Buschiazzo P. Effect of the chloroform extract of Tanacetum vulgare and one of its active principles, parthenolide, on experimental gastric ulcer in rats. J Pharm Pharmacol. 1999;51(2):215–9.PubMedCrossRef Tournier H, Schinella G, de Balsa EM, Buschiazzo H, Manez S, Mordujovich de Buschiazzo P. Effect of the chloroform extract of Tanacetum vulgare and one of its active principles, parthenolide, on experimental gastric ulcer in rats. J Pharm Pharmacol. 1999;51(2):215–9.PubMedCrossRef
25.
Zurück zum Zitat Kuusik A, Tartes U, Harak M, Hiiesaar K, Metspalu L. Developmental changes during metamorphosis in Tenebrio molitor (Coleoptera: Tenebrionidae) studied by calorimetric thermography. EJE. 2013;91(3):297–305. Kuusik A, Tartes U, Harak M, Hiiesaar K, Metspalu L. Developmental changes during metamorphosis in Tenebrio molitor (Coleoptera: Tenebrionidae) studied by calorimetric thermography. EJE. 2013;91(3):297–305.
26.
Zurück zum Zitat Vukic MD, Vukovic NL, Obradovic AD, Galovičová L, Čmiková N, Kačániová M, Matic MM. Chemical composition and biological activity of Tanacetum balsamita essential oils obtained from different plant organs. Plants. 2022;11(24):3474.PubMedPubMedCentralCrossRef Vukic MD, Vukovic NL, Obradovic AD, Galovičová L, Čmiková N, Kačániová M, Matic MM. Chemical composition and biological activity of Tanacetum balsamita essential oils obtained from different plant organs. Plants. 2022;11(24):3474.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Tiuman TS, Ueda-Nakamura T, Garcia Cortez DA, Dias Filho BP, Morgado-Diaz JA, de Souza W, Nakamura CV. Antileishmanial activity of parthenolide, a sesquiterpene lactone isolated from Tanacetum parthenium. Antimicrob Agents Chemother. 2005;49(1):176–82.PubMedPubMedCentralCrossRef Tiuman TS, Ueda-Nakamura T, Garcia Cortez DA, Dias Filho BP, Morgado-Diaz JA, de Souza W, Nakamura CV. Antileishmanial activity of parthenolide, a sesquiterpene lactone isolated from Tanacetum parthenium. Antimicrob Agents Chemother. 2005;49(1):176–82.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Pillay P, Maharaj VJ, Smith PJ. Investigating South African plants as a source of new antimalarial drugs. J Ethnopharmacol. 2008;119(3):438–54.PubMedCrossRef Pillay P, Maharaj VJ, Smith PJ. Investigating South African plants as a source of new antimalarial drugs. J Ethnopharmacol. 2008;119(3):438–54.PubMedCrossRef
29.
Zurück zum Zitat Ismail M, Kowsar A, Javed S, Choudhary MI, Khan SW, Abbas Q, Tang Y, Wang W. The Antibacterial, Insecticidal and Nematocidal Activities and Toxicity Studies of Tanacetum falconeri Hook. F. Turk J Pharm Sci. 2021;18(6):744–51.PubMedPubMedCentralCrossRef Ismail M, Kowsar A, Javed S, Choudhary MI, Khan SW, Abbas Q, Tang Y, Wang W. The Antibacterial, Insecticidal and Nematocidal Activities and Toxicity Studies of Tanacetum falconeri Hook. F. Turk J Pharm Sci. 2021;18(6):744–51.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Shazmeen N, Nazir M, Riaz N, Saleem M, Tousif MI, Tauseef S, Uddin R, Mukhtar M, Zengin G, Mollica A. In vitro antioxidant and enzyme inhibitory studies, computational analysis and chemodiversity of an emergency food plant Caralluma edulis (Edgew.) Benth. ex Hook. f: A multifunctional approach to provide new ingredients for nutraceuticals and functional foods. Food Bioscience. 2022;50:102097.CrossRef Shazmeen N, Nazir M, Riaz N, Saleem M, Tousif MI, Tauseef S, Uddin R, Mukhtar M, Zengin G, Mollica A. In vitro antioxidant and enzyme inhibitory studies, computational analysis and chemodiversity of an emergency food plant Caralluma edulis (Edgew.) Benth. ex Hook. f: A multifunctional approach to provide new ingredients for nutraceuticals and functional foods. Food Bioscience. 2022;50:102097.CrossRef
31.
Zurück zum Zitat Khan J, Tousif MI, Saleem M, Nazir M, Touseef S, Saleem K, Asim S, Khan A, Asghar MA, Zengin G. Insight into the phytochemical composition, biological activities and docking studies of Moringa oleifera Lam. to authenticate its use in biopharmaceutical industries. Industr Crops Prod. 2021;172:114042.CrossRef Khan J, Tousif MI, Saleem M, Nazir M, Touseef S, Saleem K, Asim S, Khan A, Asghar MA, Zengin G. Insight into the phytochemical composition, biological activities and docking studies of Moringa oleifera Lam. to authenticate its use in biopharmaceutical industries. Industr Crops Prod. 2021;172:114042.CrossRef
32.
Zurück zum Zitat Saleem M, Shazmeen N, Nazir M, Riaz N, Zengin G, Ataullah HM, Qurat Ul A, Nisar F, Mukhtar M, Tousif MI. Investigation on the phytochemical composition, antioxidant and enzyme inhibition potential of Polygonum Plebeium R. Br: a comprehensive approach to disclose new nutraceutical and functional food ingredients. Chem Biodivers. 2021;18(12):2100706.CrossRef Saleem M, Shazmeen N, Nazir M, Riaz N, Zengin G, Ataullah HM, Qurat Ul A, Nisar F, Mukhtar M, Tousif MI. Investigation on the phytochemical composition, antioxidant and enzyme inhibition potential of Polygonum Plebeium R. Br: a comprehensive approach to disclose new nutraceutical and functional food ingredients. Chem Biodivers. 2021;18(12):2100706.CrossRef
33.
Zurück zum Zitat Tousif MI, Nazir M, Saleem M, Tauseef S, Uddin R, Altaf M, Zengin G, Ak G, Ozturk RB, Mahomoodally MF. Exploring the industrial importance of a miracle herb Withania somnifera (L.) Dunal: Authentication through chemical profiling, in vitro studies and computational analyses. Process Biochem. 2022;121:514–28.CrossRef Tousif MI, Nazir M, Saleem M, Tauseef S, Uddin R, Altaf M, Zengin G, Ak G, Ozturk RB, Mahomoodally MF. Exploring the industrial importance of a miracle herb Withania somnifera (L.) Dunal: Authentication through chemical profiling, in vitro studies and computational analyses. Process Biochem. 2022;121:514–28.CrossRef
34.
Zurück zum Zitat Ahmed M, Ahmad S, Aati HY, Sherif AE, Ashkan MF, Alrahimi J, Motwali EA, Tousif MI, Khan MA, Hussain M. Phytochemical, antioxidant, enzyme inhibitory, thrombolytic, antibacterial, antiviral and in silico studies of Acacia jacquemontii leaves. Arab J Chem. 2022;15(12):104345.CrossRef Ahmed M, Ahmad S, Aati HY, Sherif AE, Ashkan MF, Alrahimi J, Motwali EA, Tousif MI, Khan MA, Hussain M. Phytochemical, antioxidant, enzyme inhibitory, thrombolytic, antibacterial, antiviral and in silico studies of Acacia jacquemontii leaves. Arab J Chem. 2022;15(12):104345.CrossRef
35.
Zurück zum Zitat Ahmed M, Khan K-U-R, Ahmad S, Aati HY, Ovatlarnporn C, Rehman MS-U, Javed T, Khursheed A, Ghalloo BA, Dilshad R. Comprehensive phytochemical profiling, biological activities, and molecular docking studies of Pleurospermum candollei: An insight into potential for natural products development. Molecules. 2022;27(13):4113.PubMedPubMedCentralCrossRef Ahmed M, Khan K-U-R, Ahmad S, Aati HY, Ovatlarnporn C, Rehman MS-U, Javed T, Khursheed A, Ghalloo BA, Dilshad R. Comprehensive phytochemical profiling, biological activities, and molecular docking studies of Pleurospermum candollei: An insight into potential for natural products development. Molecules. 2022;27(13):4113.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Tepe B, Sokmen A. Screening of the antioxidative properties and total phenolic contents of three endemic Tanacetum subspecies from Turkish flora. Bioresour Technol. 2007;98(16):3076–9.PubMedCrossRef Tepe B, Sokmen A. Screening of the antioxidative properties and total phenolic contents of three endemic Tanacetum subspecies from Turkish flora. Bioresour Technol. 2007;98(16):3076–9.PubMedCrossRef
37.
Zurück zum Zitat Emre I. The biochemical content and antioxidant capacities of endemic Tanacetum densum (Lab.) Schultz Bip. Subsp. laxum, and Tanacetum densum (Lab.) Schultz Bip. Subsp. amani Heywood growing in Turkey. Braz J Biol. 2021;81(4):1106–14.PubMedCrossRef Emre I. The biochemical content and antioxidant capacities of endemic Tanacetum densum (Lab.) Schultz Bip. Subsp. laxum, and Tanacetum densum (Lab.) Schultz Bip. Subsp. amani Heywood growing in Turkey. Braz J Biol. 2021;81(4):1106–14.PubMedCrossRef
38.
Zurück zum Zitat Babich O, Larina V, Krol O, Ulrikh E, Sukhikh S, Gureev MA, Prosekov A, Ivanova S. In vitro study of biological activity of Tanacetum vulgare Extracts. Pharmaceutics. 2023;15(2):616.PubMedPubMedCentralCrossRef Babich O, Larina V, Krol O, Ulrikh E, Sukhikh S, Gureev MA, Prosekov A, Ivanova S. In vitro study of biological activity of Tanacetum vulgare Extracts. Pharmaceutics. 2023;15(2):616.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Gevrenova R, Zengin G, Sinan KI, Zheleva-Dimitrova D, Balabanova V, Kolmayer M, Voynikov Y, Joubert O. An in-depth study of metabolite profile and biological potential of Tanacetum balsamita L. (Costmary). Plants (Basel). 2022;12(1):22.PubMedCrossRef Gevrenova R, Zengin G, Sinan KI, Zheleva-Dimitrova D, Balabanova V, Kolmayer M, Voynikov Y, Joubert O. An in-depth study of metabolite profile and biological potential of Tanacetum balsamita L. (Costmary). Plants (Basel). 2022;12(1):22.PubMedCrossRef
40.
Zurück zum Zitat Wu C, Chen F, Wang X, Wu Y, Dong M, He G, Galyean RD, He L, Huang G. Identification of antioxidant phenolic compounds in feverfew (Tanacetum parthenium) by HPLC-ESI-MS/MS and NMR. Phytochem Anal. 2007;18(5):401–10.PubMedCrossRef Wu C, Chen F, Wang X, Wu Y, Dong M, He G, Galyean RD, He L, Huang G. Identification of antioxidant phenolic compounds in feverfew (Tanacetum parthenium) by HPLC-ESI-MS/MS and NMR. Phytochem Anal. 2007;18(5):401–10.PubMedCrossRef
41.
Zurück zum Zitat Devrnja N, Krstic-Milosevic D, Janosevic D, Tesevic V, Vinterhalter B, Savic J, Calic D. In vitro cultivation of tansy (Tanacetum vulgare L.): a tool for the production of potent pharmaceutical agents. Protoplasma. 2021;258(3):587–99.PubMedCrossRef Devrnja N, Krstic-Milosevic D, Janosevic D, Tesevic V, Vinterhalter B, Savic J, Calic D. In vitro cultivation of tansy (Tanacetum vulgare L.): a tool for the production of potent pharmaceutical agents. Protoplasma. 2021;258(3):587–99.PubMedCrossRef
42.
Zurück zum Zitat Chen J, Yang J, Ma L, Li J, Shahzad N, Kim CK. Structure-antioxidant activity relationship of methoxy, phenolic hydroxyl, and carboxylic acid groups of phenolic acids. Sci Rep. 2020;10(1):2611.PubMedPubMedCentralCrossRef Chen J, Yang J, Ma L, Li J, Shahzad N, Kim CK. Structure-antioxidant activity relationship of methoxy, phenolic hydroxyl, and carboxylic acid groups of phenolic acids. Sci Rep. 2020;10(1):2611.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Cos P, Rajan P, Vedernikova I, Calomme M, Pieters L, Vlietinck AJ, Augustyns K, Haemers A, Vanden Berghe D. In vitro antioxidant profile of phenolic acid derivatives. Free Radic Res. 2002;36(6):711–6.PubMedCrossRef Cos P, Rajan P, Vedernikova I, Calomme M, Pieters L, Vlietinck AJ, Augustyns K, Haemers A, Vanden Berghe D. In vitro antioxidant profile of phenolic acid derivatives. Free Radic Res. 2002;36(6):711–6.PubMedCrossRef
44.
Zurück zum Zitat Stanciu GD, Luca A, Rusu RN, Bild V, Beschea Chiriac SI, Solcan C, Bild W, Ababei DC. Alzheimer’s disease pharmacotherapy in relation to cholinergic system involvement. Biomolecules. 2019;10(1):40.PubMedPubMedCentralCrossRef Stanciu GD, Luca A, Rusu RN, Bild V, Beschea Chiriac SI, Solcan C, Bild W, Ababei DC. Alzheimer’s disease pharmacotherapy in relation to cholinergic system involvement. Biomolecules. 2019;10(1):40.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Patterson C. World alzheimer report 2018, Alzheimer’s Disease International. London: ADI); 2018. Patterson C. World alzheimer report 2018, Alzheimer’s Disease International. London: ADI); 2018.
46.
Zurück zum Zitat Geula C, Darvesh S. Butyrylcholinesterase, cholinergic neurotransmission and the pathology of Alzheimer’s disease. Drugs Today (Barc). 2004;40(8):711–21.PubMedCrossRef Geula C, Darvesh S. Butyrylcholinesterase, cholinergic neurotransmission and the pathology of Alzheimer’s disease. Drugs Today (Barc). 2004;40(8):711–21.PubMedCrossRef
47.
Zurück zum Zitat Pope C, Karanth S, Liu J. Pharmacology and toxicology of cholinesterase inhibitors: uses and misuses of a common mechanism of action. Environ Toxicol Pharmacol. 2005;19(3):433–46.PubMedCrossRef Pope C, Karanth S, Liu J. Pharmacology and toxicology of cholinesterase inhibitors: uses and misuses of a common mechanism of action. Environ Toxicol Pharmacol. 2005;19(3):433–46.PubMedCrossRef
48.
Zurück zum Zitat Rees TM, Brimijoin S. The role of acetylcholinesterase in the pathogenesis of Alzheimer’s disease. Drugs Today (Barc). 2003;39(1):75–83.PubMedCrossRef Rees TM, Brimijoin S. The role of acetylcholinesterase in the pathogenesis of Alzheimer’s disease. Drugs Today (Barc). 2003;39(1):75–83.PubMedCrossRef
49.
Zurück zum Zitat Moodie LWK, Sepcic K, Turk T, Frange ZR, Svenson J. Natural cholinesterase inhibitors from marine organisms. Nat Prod Rep. 2019;36(8):1053–92.PubMedCrossRef Moodie LWK, Sepcic K, Turk T, Frange ZR, Svenson J. Natural cholinesterase inhibitors from marine organisms. Nat Prod Rep. 2019;36(8):1053–92.PubMedCrossRef
50.
Zurück zum Zitat Uddin MJ, Russo D, Rahman MM, Uddin SB, Halim MA, Zidorn C, Milella L. Anticholinesterase activity of eight medicinal plant species: in vitro and in silico studies in the search for therapeutic agents against Alzheimer’s disease. Evid-Based Complement Altern Med. 2021;2021:9995614.CrossRef Uddin MJ, Russo D, Rahman MM, Uddin SB, Halim MA, Zidorn C, Milella L. Anticholinesterase activity of eight medicinal plant species: in vitro and in silico studies in the search for therapeutic agents against Alzheimer’s disease. Evid-Based Complement Altern Med. 2021;2021:9995614.CrossRef
51.
Zurück zum Zitat Ahmed S, Khan ST, Zargaham MK, Khan AU, Khan S, Hussain A, Uddin J, Khan A, Al-Harrasi A. Potential therapeutic natural products against Alzheimer’s disease with reference of Acetylcholinesterase. Biomed Pharmacother. 2021;139:111609.PubMedCrossRef Ahmed S, Khan ST, Zargaham MK, Khan AU, Khan S, Hussain A, Uddin J, Khan A, Al-Harrasi A. Potential therapeutic natural products against Alzheimer’s disease with reference of Acetylcholinesterase. Biomed Pharmacother. 2021;139:111609.PubMedCrossRef
52.
Zurück zum Zitat Loizzo MR, Tundis R, Menichini F. Natural and synthetic Tyrosinase inhibitors as antibrowning agents: an update. Comprehensive Rev Food Sci Food Safety. 2012;11(4):378–98.CrossRef Loizzo MR, Tundis R, Menichini F. Natural and synthetic Tyrosinase inhibitors as antibrowning agents: an update. Comprehensive Rev Food Sci Food Safety. 2012;11(4):378–98.CrossRef
53.
Zurück zum Zitat Zaidi KU, Ali SA, Ali A, Naaz I. Natural Tyrosinase inhibitors: role of herbals in the treatment of Hyperpigmentary disorders. Mini Rev Med Chem. 2019;19(10):796–808.PubMedCrossRef Zaidi KU, Ali SA, Ali A, Naaz I. Natural Tyrosinase inhibitors: role of herbals in the treatment of Hyperpigmentary disorders. Mini Rev Med Chem. 2019;19(10):796–808.PubMedCrossRef
54.
Zurück zum Zitat Pillaiyar T, Manickam M, Namasivayam V. Skin whitening agents: medicinal chemistry perspective of tyrosinase inhibitors. J Enzyme Inhib Med Chem. 2017;32(1):403–25.PubMedPubMedCentralCrossRef Pillaiyar T, Manickam M, Namasivayam V. Skin whitening agents: medicinal chemistry perspective of tyrosinase inhibitors. J Enzyme Inhib Med Chem. 2017;32(1):403–25.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Poovitha S, Parani M. In vitro and in vivo alpha-amylase and alpha-glucosidase inhibiting activities of the protein extracts from two varieties of bitter gourd (Momordica charantia L.). BMC Complement Altern Med. 2016;16(Suppl 1):185.PubMedPubMedCentralCrossRef Poovitha S, Parani M. In vitro and in vivo alpha-amylase and alpha-glucosidase inhibiting activities of the protein extracts from two varieties of bitter gourd (Momordica charantia L.). BMC Complement Altern Med. 2016;16(Suppl 1):185.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Gin H, Rigalleau V. Post-prandial hyperglycemia post-prandial hyperglycemia and diabetes. Diabetes Metab. 2000;26(4):265–72.PubMed Gin H, Rigalleau V. Post-prandial hyperglycemia post-prandial hyperglycemia and diabetes. Diabetes Metab. 2000;26(4):265–72.PubMed
57.
Zurück zum Zitat Lordan S, Smyth TJ, Soler-Vila A, Stanton C, Ross RP. The alpha-amylase and alpha-glucosidase inhibitory effects of Irish seaweed extracts. Food Chem. 2013;141(3):2170–6.PubMedCrossRef Lordan S, Smyth TJ, Soler-Vila A, Stanton C, Ross RP. The alpha-amylase and alpha-glucosidase inhibitory effects of Irish seaweed extracts. Food Chem. 2013;141(3):2170–6.PubMedCrossRef
58.
Zurück zum Zitat Lebovitz HE. alpha-Glucosidase inhibitors. Endocrinol Metab Clin North Am. 1997;26(3):539–51.PubMedCrossRef Lebovitz HE. alpha-Glucosidase inhibitors. Endocrinol Metab Clin North Am. 1997;26(3):539–51.PubMedCrossRef
60.
Zurück zum Zitat Etxeberria U, de la Garza AL, Campion J, Martinez JA, Milagro FI. Antidiabetic effects of natural plant extracts via inhibition of carbohydrate hydrolysis enzymes with emphasis on pancreatic alpha amylase. Expert Opin Ther Targets. 2012;16(3):269–97.PubMedCrossRef Etxeberria U, de la Garza AL, Campion J, Martinez JA, Milagro FI. Antidiabetic effects of natural plant extracts via inhibition of carbohydrate hydrolysis enzymes with emphasis on pancreatic alpha amylase. Expert Opin Ther Targets. 2012;16(3):269–97.PubMedCrossRef
61.
Zurück zum Zitat Mohamed EA, Siddiqui MJ, Ang LF, Sadikun A, Chan SH, Tan SC, Asmawi MZ, Yam MF. Potent alpha-glucosidase and alpha-amylase inhibitory activities of standardized 50% ethanolic extracts and sinensetin from Orthosiphon stamineus Benth as anti-diabetic mechanism. BMC Complement Altern Med. 2012;12:176.PubMedPubMedCentralCrossRef Mohamed EA, Siddiqui MJ, Ang LF, Sadikun A, Chan SH, Tan SC, Asmawi MZ, Yam MF. Potent alpha-glucosidase and alpha-amylase inhibitory activities of standardized 50% ethanolic extracts and sinensetin from Orthosiphon stamineus Benth as anti-diabetic mechanism. BMC Complement Altern Med. 2012;12:176.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Perez-Gutierrez RM, Damian-Guzman M. Meliacinolin: a potent alpha-glucosidase and alpha-amylase inhibitor isolated from Azadirachta indica leaves and in vivo antidiabetic property in streptozotocin-nicotinamide-induced type 2 diabetes in mice. Biol Pharm Bull. 2012;35(9):1516–24.PubMedCrossRef Perez-Gutierrez RM, Damian-Guzman M. Meliacinolin: a potent alpha-glucosidase and alpha-amylase inhibitor isolated from Azadirachta indica leaves and in vivo antidiabetic property in streptozotocin-nicotinamide-induced type 2 diabetes in mice. Biol Pharm Bull. 2012;35(9):1516–24.PubMedCrossRef
63.
Zurück zum Zitat Ali RB, Atangwho IJ, Kuar N, Ahmad M, Mahmud R, Asmawi MZ. In vitro and in vivo effects of standardized extract and fractions of Phaleria macrocarpa fruits pericarp on lead carbohydrate digesting enzymes. BMC Complement Altern Med. 2013;13:39.PubMedPubMedCentralCrossRef Ali RB, Atangwho IJ, Kuar N, Ahmad M, Mahmud R, Asmawi MZ. In vitro and in vivo effects of standardized extract and fractions of Phaleria macrocarpa fruits pericarp on lead carbohydrate digesting enzymes. BMC Complement Altern Med. 2013;13:39.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Kim K-T, Rioux L-E, Turgeon SL. Alpha-amylase and alpha-glucosidase inhibition is differentially modulated by fucoidan obtained from Fucus vesiculosus and Ascophyllum nodosum. Phytochemistry. 2014;98:27–33.PubMedCrossRef Kim K-T, Rioux L-E, Turgeon SL. Alpha-amylase and alpha-glucosidase inhibition is differentially modulated by fucoidan obtained from Fucus vesiculosus and Ascophyllum nodosum. Phytochemistry. 2014;98:27–33.PubMedCrossRef
65.
Zurück zum Zitat Durazzo A, D’Addezio L, Camilli E, Piccinelli R, Turrini A, Marletta L, Marconi S, Lucarini M, Lisciani S, Gabrielli P, Gambelli L, Aguzzi A, Sette S. From plant compounds to botanicals and back: a current snapshot. Molecules. 2018;23(8):1844.PubMedPubMedCentralCrossRef Durazzo A, D’Addezio L, Camilli E, Piccinelli R, Turrini A, Marletta L, Marconi S, Lucarini M, Lisciani S, Gabrielli P, Gambelli L, Aguzzi A, Sette S. From plant compounds to botanicals and back: a current snapshot. Molecules. 2018;23(8):1844.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Salehi B, Ata A, Anil Kumar NV, Sharopov F, Ramírez-Alarcón K, Ruiz-Ortega A, Abdulmajid Ayatollahi S, Valere Tsouh Fokou P, Kobarfard F, Amiruddin Zakaria Z. Antidiabetic potential of medicinal plants and their active components. Biomolecules. 2019;9(10):551.PubMedPubMedCentralCrossRef Salehi B, Ata A, Anil Kumar NV, Sharopov F, Ramírez-Alarcón K, Ruiz-Ortega A, Abdulmajid Ayatollahi S, Valere Tsouh Fokou P, Kobarfard F, Amiruddin Zakaria Z. Antidiabetic potential of medicinal plants and their active components. Biomolecules. 2019;9(10):551.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Diwan R, Shinde A, Malpathak N. Phytochemical composition and antioxidant potential of Ruta graveolens L. in vitro culture lines. J Bot. 2012;(2012):1–6. Diwan R, Shinde A, Malpathak N. Phytochemical composition and antioxidant potential of Ruta graveolens L. in vitro culture lines. J Bot. 2012;(2012):1–6.
68.
Zurück zum Zitat Abdelouhab K, Guemmaz T, Karamac M, Kati DE, Amarowicz R, Arrar L. Phenolic composition and correlation with antioxidant properties of various organic fractions from Hertia cheirifolia extracts. J Pharm Biomed Anal. 2023;235:115673.PubMedCrossRef Abdelouhab K, Guemmaz T, Karamac M, Kati DE, Amarowicz R, Arrar L. Phenolic composition and correlation with antioxidant properties of various organic fractions from Hertia cheirifolia extracts. J Pharm Biomed Anal. 2023;235:115673.PubMedCrossRef
69.
Zurück zum Zitat Zhang B, Quan H, Cai Y, Han X, Kang H, Lu Y, Cheng H, Xiang N, Lan X, Guo X. Comparative study of browning, phenolic profiles, antioxidant and antiproliferative activities in hot air and vacuum drying of lily (Lilium lancifolium Thunb.) bulbs. LWT. 2023;115015:184. Zhang B, Quan H, Cai Y, Han X, Kang H, Lu Y, Cheng H, Xiang N, Lan X, Guo X. Comparative study of browning, phenolic profiles, antioxidant and antiproliferative activities in hot air and vacuum drying of lily (Lilium lancifolium Thunb.) bulbs. LWT. 2023;115015:184.
70.
Zurück zum Zitat Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017;7(1):42717.PubMedPubMedCentralCrossRef Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017;7(1):42717.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Mishra S, Dahima R. In vitro ADME studies of TUG-891, a GPR-120 inhibitor using SWISS ADME predictor. J Drug Deliver Ther. 2019;9(2-s):366–9. Mishra S, Dahima R. In vitro ADME studies of TUG-891, a GPR-120 inhibitor using SWISS ADME predictor. J Drug Deliver Ther. 2019;9(2-s):366–9.
72.
Zurück zum Zitat Daina A, Zoete V. A boiled-egg to predict gastrointestinal absorption and brain penetration of small molecules. ChemMedChem. 2016;11(11):1117–21.PubMedPubMedCentralCrossRef Daina A, Zoete V. A boiled-egg to predict gastrointestinal absorption and brain penetration of small molecules. ChemMedChem. 2016;11(11):1117–21.PubMedPubMedCentralCrossRef
Metadaten
Titel
Secondary metabolic profiling, antioxidant potential, enzyme inhibitory activities and in silico and ADME studies: a multifunctional approach to reveal medicinal and industrial potential of Tanacetum falconeri
verfasst von
Muhammad Imran Tousif
Zaheer Abbas
Mamona Nazir
Muhammad Saleem
Saba Tauseef
Areeba Hassan
Sajid Ali
Maqsood Ahmed
Jallat Khan
Gokhan Zengin
Abeer Hashem
Khalid F. Almutairi
Graciela Dolores Avila-Quezada
Elsayed Fathi Abd-Allah
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
BMC Complementary Medicine and Therapies / Ausgabe 1/2024
Elektronische ISSN: 2662-7671
DOI
https://doi.org/10.1186/s12906-024-04459-5

Weitere Artikel der Ausgabe 1/2024

BMC Complementary Medicine and Therapies 1/2024 Zur Ausgabe