Metabolites 13 00281 v2
Metabolites 13 00281 v2
Metabolites 13 00281 v2
OH
OH
metabolites
Article
Breynia cernua: Chemical Profiling of Volatile Compounds in
the Stem Extract and Its Antioxidant, Antibacterial,
Antiplasmodial and Anticancer Activity In Vitro and In Silico
Hesti Lina Wiraswati 1,2,3,4, * , Nisa Fauziah 1,2,3 , Gita Widya Pradini 1,3 , Dikdik Kurnia 5 , Reza Abdul Kodir 6 ,
Afiat Berbudi 1,2,3 , Annisa Retno Arimdayu 4 , Amila Laelalugina 4 , Supandi 7,8 and Ilma Fauziah Ma’ruf 4,9
1. Introduction
From a historical point of view, medicinal plants have been proven to have therapeutic
properties to cure various diseases, and they still represent an important source of novel
drugs today [1,2]. More than 50% of drugs in modern therapeutics are represented by
plant-derived drugs, either in their natural or in their derivative form [3,4]. For exam-
ple, successful plant-derived drugs are aspirin from Filipendula ulmaria (L.) Maxim and
artemisinin from Artemisia annua L. [5,6]. Indonesia is a country with high plant biodiver-
sity and has very diverse plant resources (approximately 30,000–40,000 plant species), 20%
of which are either wild or cultivated medicinal plants [7].
On the other hand, metabolite profiling and the exploration of medicinal plants in
Indonesia are still rarely conducted. For those reasons, the metabolite profile of uninvesti-
gated plants in Indonesia is interesting to explore since they are a promising source of new
plant-derived drugs. Therefore, this research aimed to unveil the metabolite component of
one herbaceous plant as a reservoir of a particular bioactive compound that can potentially
be used as a drug in the future.
Various Breynia species have been traditionally applied as medicine for numerous
diseases [8]. One such species, Breynia cernua (local name: Katuk Hutan), has been used
in Amuntai, Hulu Sungai Utara, and Kalimantan Selatan as herbal medicine for smallpox
and wounds [9], as well as in Papua as a traditional medicine for cervical and breast
cancer [10]. B. cernua has the following anatomical characteristics: semilunar vascular
bundle, paracyitic and anomocytic stomata, irregular shape of the lower epidermis with
a wavy anticlinal wall, polygonal shape of the upper epidermis with a straight anticlinal
wall, single unicellular trichome on the lower epidermis, one-layer upper epidermis with a
regularly shaped cell, one-layer lower epidermis with a papillated cell, one-layer elongated
palisade tissue, loose arrangement of the spongy tissue, druse calcium oxalate on vascular
bundle parenchyma, and hypodermis in the area near leaf bone [11]. It was found that the
ethanol extract of B. cernua has cytotoxic activity according to the brine shrimp lethality
test and MTT assay against MCF-7 [10]. The ethyl acetate or butanol extract of B. cernua
(stem hardwood) and methanol extract of B. cernua (stem bark) exhibited broad-spectrum
antibacterial activities against Micrococcus luteus, Micrococcus roseus, Bacillus coagulans,
Bacillus cereus, Bacillus megaterium, Bacillus subtilis, Citrobacter freundii, Lactobacillus casei,
Staphylococcus albus, Staphylococcus aureus, Staphylococcus epidermidis, Agrobacterium tume-
faciens, Streptococcus faecalis, Streptococcus pneumoniae, Klebsiella pneumoniae, Eschericia coli,
Proteus vulgaris, Proteus mirabili, Neisseria gonorrhoeae, Pseudomonas aeruginosa, Enterobacter
aerogenes, Salmonella Typhi, Salmonella Typhimurium, and Serratia marcescens. Furthermore,
the butanol or methanol extract of B. cernua (root bark) demonstrated antifungal activities
against Aspergillus niger, Aspergillus versicolour, Aspergillus vitis, Candida albican, Candida trop-
icalis, Cladosporium cladosporides, Penicillium notatum, Trichophyton mentagrophyte, Tricophyton
rubrum, and Tricophyton tonsurans [12]. The ethanol extract of B. cernua also was found to
have antiparkinsonism activity in vitro and in vivo [13]. Considering its multiple in vivo
and in vitro bioactivity, B. cernua is a promising source of novel plant-derived drugs. There-
fore, identification of its chemical composition and activity prediction of each metabolite
represent interesting research pathways.
Traditional medicinal plant research has gradually risen worldwide in recent years,
owing to the natural sources and variety of such plants, which allow them to complement
current pharmaceutical treatments [14,15]. The bioactivities of medicinal plants are corre-
lated with the phytochemicals contained in them such as alkaloids, flavonoids, saponins,
terpenoids, tannins, and quinonens [16]. Metabolite profiling through GC–MS using the
silyl derivatization method has been widely applied to identify the composition of plant
extracts [17,18]. In addition, bioinformatics tools aid drug discovery by using all primary
data gathered from in vivo and in vitro studies in a short time and at a lower cost. In silico
molecular docking is one of the most effective ways to find novel ligands for proteins with
known structures, thus playing an essential role in structure-based drug development [19].
A preliminary study (GC–MS) showed that the stem extract of B. cernua contained more
Metabolites 2023, 13, 281 3 of 28
compounds than the leaf extract. Moreover, the stem extract exhibited better antioxidant
and bactericidal activity than the leaf extract (manuscript in preparation). Therefore, in this
report, we performed metabolite profiling, characterized the in vitro antioxidant, antibacte-
rial, antiplasmodial, and anticancer activity, and conducted virtual drug screening through
molecular docking, drug likeness, and pharmacokinetic analyses of the methanol extract of
B. cernua (stem).
amount of enzyme required to inhibit nitro blue tetrazolium degradation by 50% in 1 min
was measured as U/mg protein [22]. The following equation was used to calculate the
percentage inhibition: %h = (Ab – As)/Ab × 100%, where %h is the percentage inhibition
(free-radical inhibition), Ab is the absorbance blank, and As is the sample absorbance. The
value of 50% free-radical inhibition concentration (IC50 ) was calculated using the regression
equation y = ax + b.
not detected in this extract. Therefore, this study focused on the volatile compounds of
B. cernua (stem).
Table 1. Metabolite profiling of B. cernua methanol extract using gas chromatography/mass spec-
trometry (GC–MS) method.
Retention Time
No Compound Name Molecular Formula m/z
(minute)
1 Methane, dichloronitro- 3322–3386 CHCl2 NO2 43, 54, 68, 82, 90, 134
2 Ether, hexyl pentyl 4135–4187 C11 H24 O 42, 84, 127
3 1,5-Heptadien-4-ol, 3,3,6-trimethyl- 7410–7453 C10 H18 O 42, 56, 70, 82, 97
Sulfurous acid, 2-ethylhexyl
4 7574–7612 C14 H30 O3 S 42, 56, 82, 90, 112
isohexyl ester
5 Sulfurous acid, hexyl nonyl ester 8008–8046 C15 H32 O3 S 42, 56, 76, 82, 94
6 Decane, 2,4-dimethyl- 8119–8156 C12 H26 43, 84, 112, 126
7 Octane, 3,4,5,6-tetramethyl- 8213–8269 C12 H26 42, 56, 70, 82, 90, 112, 126
8 Oxalic acid, cyclohexyl nonyl ester 8558–8593 C17 H30 O4 42, 56, 68, 82, 94, 110
9 Octane, 2,4,6-trimethyl- 8857–8903 C11 H24 42, 56, 70, 84, 90, 112, 126
N-[β-Hydroxy-β-[4-[1-adamantyl-6,8-
10 9383–9.400 C27 H33 Cl2 NO 43, 54, 69, 97, 118, 134, 149
dichloro]quinolyl]ethyl]piperidine
11 1-Chloroundecane 10,582–10,614 C11 H23 Cl 42, 56, 68, 84, 104, 118, 134, 149
12 Dodecane, 1-chloro- 10,628–10,660 C12 H25 Cl 42, 56, 70, 84, 104, 134
13 Decane, 1-chloro- 10,760–10,803 C10 H21 Cl 42, 56, 68, 84, 104, 134, 149
Propanoic acid, 2,2-dimethyl-,
14 10,897–10,948 C15 H22 O2 43, 56, 78, 90, 106, 118, 134, 149
2-(1,1-dimethylethyl) phenyl ester
15 Sulfurous acid, decyl hexyl ester 11,347–11,377 C16 H34 O3 S 42, 84, 98, 112, 134, 154
16 Tetradecane, 1-chloro- 11,436–11,479 C14 H29 Cl 42, 56, 70, 84, 104, 118, 134, 149
17 Dodecane, 4,6-dimethyl- 11,522–11,557 C14 H30 43, 71, 98, 126, 154
18 Cyclohexene, 2-ethenyl-1,3,3-trimethyl- 1894–11,975 C14 H30 42, 56, 70, 90, 104, 118, 134, 149
42, 56, 70, 84, 104, 118, 134,
19 Pentane, 2,3,3-trimethyl- 12,112–12,134 C8 H18
149, 168
20 Hexadecane 12,182–12,220 C16 H34 42, 56, 84, 98, 112, 126, 140, 154
Benzenepropanoic acid, α-hydroxy-,
21 12,953–13,021 C10 H12 O3 43, 64, 76, 90, 102, 120, 161
methyl ester
22 1,3-Phenylene, bis(3-phenylpropenoate) 13,177–13,231 C24 H18 O4 43, 56, 76, 90, 102, 130, 161
23 1,2:4,5:9,10-Triepoxydecane 13,705–13,754 C10 H16 O3 43, 56, 68, 80, 92, 118, 206
1,3-Cyclohexadiene,
40, 56, 70, 84, 92, 104, 118,
24 5-(1,5-dimethyl-4-hexenyl)-2-methyl-, 14,150–14,20 C15 H24
131, 203
[S-(R*,S*)]-
25 1-Octadecanesulphonyl chloride 14,341–14,379 C18 H37 ClO2 S 42, 70, 76, 92, 118, 140, 154, 201
26 Oxalic acid, allyl pentadecyl ester 15,079–15,125 C20 H36 O4 42, 56, 70, 84, 110
27 2-Piperidinone, N-[4-bromo-n-butyl]- 15,472–15,510 C9 H16 BrNO 43, 56, 70, 90, 104, 122, 206
28 2-Bromotetradecane 16,892–16,959 C14 H29 Br 42, 71, 98, 126, 154, 206
29 Oxalic acid, 6-ethyloct-3-yl heptyl ester 17,021–17,059 C19 H36 O4 42, 56, 84, 98, 110, 126, 206
42, 56, 70, 76, 84, 98, 112, 141,
30 2-Bromo dodecane 17,409–17,504 C12 H25 Br
168, 206
Metabolites 2023, 13, 281 7 of 28
Table 1. Cont.
Retention Time
No Compound Name Molecular Formula m/z
(minute)
Ethylamine,
31 17,991–18,037 C13 H23 N 43, 54, 68, 82, 106, 134, 206
2-(adamantan-1-yl)-1-methyl-
32 Pentadecanal- 18,452–18,511 C15 H30 O 43, 56, 68, 81, 108, 206
33 Di-n-decylsulfone 18,662–18,735 C20 H42 O2 S 43, 56, 70, 84, 98, 206
42, 70, 98, 112, 126, 154,
34 Dodecane, 1-iodo- 19,675–19,756 C12 H25 I
183, 206
42, 54, 73, 86, 96, 110,142, 170,
35 Hexadecanoic acid, methyl ester 20,033–20,093 C17 H34 O2
206, 227, 270
Benzenepropanoic acid, 43, 56, 68, 90, 104, 116, 134,
36 20,136–20,200 C18 H28 O3
3,5-bis(1,1-dimethylethyl)-4-hydroxy-, 146, 206
43, 54, 66, 78, 90, 104, 116, 132,
37 Methyl 3-bromo-1-adamantaneacetate 21,087–21,184 C13 H19 BrO2
156, 190, 206, 256, 280
43, 56, 68, 81, 95, 109, 132, 190,
38 1-Cyclohexylnonene 21,375–21,456 C15 H28
206, 280
4-Methyl-2,4-bis(p-
43, 54, 81, 104, 132, 158,
39 hydroxyphenyl)pent-1-ene, 21,765–21,811 C24 H36 O2 Si2
206, 280
2 TMS derivative
40, 54, 66, 81, 95, 109, 123, 146,
40 Cyclohexene, 1-nonyl- 21,930–21,978 C15 H28
190, 206, 280
41 Cyclotrisiloxane, hexamethyl- 22,121–22,194 C6 H18 O3 Si3 43, 70, 84, 95, 114, 132, 190, 280
43, 68, 84, 95, 118, 132, 146, 190,
42 1,4-Bis(trimethylsilyl)benzene 22,245–22,309 C12 H22 Si2
206, 280
9,12,15-Octadecatrienoic acid, methyl 43, 54, 66, 78, 94, 107, 120, 134,
43 22,323–22,366 C19 H32 O2
ester, (Z,Z,Z)- 148, 190, 280
42, 70, 85, 112, 126, 155, 190,
44 Eicosane, 1-iodo- 22,501–22,608 C20 H41 I
206, 238, 280
43, 54, 74, 104, 132, 162, 190,
45 Arsenous acid, tris(trimethylsilyl) ester 24,536–24,585 C9 H27 AsO3 Si3
206, 236, 280, 314
* Note: TMS = trimethylsilyl.
Medicinal plants are a promising reservoir of bioactive metabolites that can support
the discovery of novel drug molecules. GC–MS analysis was conducted on the B. cernua
(stem) methanol extract and revealed 45 bioactive compounds. Studies in the literature
have revealed the isolation of several metabolites, regardless of whether their specific func-
tion was demonstrated or not. Meanwhile, the origin or bioactivity of a few metabolites
has not been documented. Dichloronitromethane, as the dominant compound in ethyl
acetate fraction of endophytic fungus Alternaria alternata AE1, exhibited antimicrobial prop-
erties [34]. 3,3,6-Trimethyl-1,5-heptadien-4-ol identified, as one of the main compounds
of Artemisia austro-yunnanensis essential oil, possessed antioxidant activity [35]. Sulfurous
acid, 2-ethylhexyl isohexyl ester, contained in the methanol extract of Gracilaria corticata and
Gracilaria edulis, showed antioxidant and antibacterial activity [36]. Sulfurous acid, hexyl
nonyl ester was identified in the ethanol extract of Eclipta prostrata and showed antimicro-
bial activity [37]. Decane, 2,4-dimethyl- was identified in the endophytic bacterium Pantoea
sp. strain Dez632 and possessed antibacterial activity [38]. Octane, 3,4,5,6-tetramethyl-,
contained in the endophytic bacterium Pseudomonas sp. strain Bt851, exhibited antibacterial
activity [38]. Oxalic acid, cyclohexyl nonyl ester, isolated from the methanol extract of
Rheum ribes, showed an inhibitory effect on Escherichia coli [39]. Octane, 2,4,6-trimethyl- was
identified in the essential oil of Rumex hastatus, which exhibited inhibitory and antioxidant
Metabolites 2023, 13, 281 8 of 28
activity [40]. 1-Chloroundecane, contained in the extract of Streptomyces sp., exerted antibac-
terial activity [41]. Dodecane, 1-chloro- was identified in the extract of Spongia officinalis
var ceylonensis and possessed antiproliferative activity against three cancer cell lines [42].
Decane, 1-chloro-, identified in the ethyl acetate fraction of Artocarpus anisophyllus Miq stem
bark, exerted antioxidant activity [43]. Sulfurous acid, decyl hexyl ester was identified in
rambutan seed fat [44] and as a volatile organic compound in olive tree [45]. Tetradecane, 1-
chloro- was identified in the crude extract of medicinal herbs used to treat kidney stones [46].
Dodecane, 4,6-dimethyl-, contained in the essential oil of Ephedra pachyclada, possessed an-
tioxidant and antimicrobial activity [47]. Cyclohexene, 2-ethenyl-1,3,3-trimethyl-, extracted
from the ethyl acetate fraction of Streptomyces sp. Al-Dhabi-90 metabolites, demonstrated
broad-spectrum activity against pathogenic bacteria [48]. Hexadecane was identified as a
major component of Monochaetia kansensis chloroform extract, and this compound is known
to have antioxidant and antibacterial activity [49]. Benzenepropanoic acid, α-hydroxy-,
methyl ester (synonym: lactic acid, 3-phenyl-, methyl ester), contained in Lactobacillus plan-
tarum culture supernatants, possessed potential antipathogenic and chronic skin wound-
healing properties [50]. 1,3-Phenylene, bis(3-phenylpropenoate), identified in the acetone
extract of Jasminum annamense subsp. annamense leaves, exerted antioxidant and antibacte-
rial activity [51]. 1,2:4,5:9,10-Triepoxydecane was extracted from the ethyl acetate fraction
of Alysicarpus glumaceus, which had therapeutic properties such as stimulatory, diuretic, an-
tipsychotic, anti-inflammatory, antidiarrheal, abortifacient, antitussive, and anti-asthmatic
activity [52]. 1,3-Cyclohexadiene, 5-(1,5-dimethyl-4-hexenyl)-2-methyl-, [S-(R*,S*)]- was
identified in the methanol extract of Zingiber officinale, and this compound had pharmaco-
logical properties such as antioxidant, anti-inflammatory, and antinociceptive activities [53].
1-Octadecanesulphonyl chloride was previously isolated from the volatile floral extract
of Acacia auriculiformis, which exhibited antioxidant and antifungal activity [54]. Oxalic
acid, allyl pentadecyl ester was identified in the methanol extract of red alga Laurencia bran-
denii, which showed anti-pest, antimicrobial, termiticidal, and maggoticidal activity [55].
2-Piperidinone, N-[4-bromo-n-butyl]- was contained in various organic solvent extracts
of pomegranate peel and demonstrated antimicrobial activities against P. aeruginosa, P.
mirabilis, and C. albicans [56]. 2-Bromotetradecane, as one of the major constituents of
ethyl acetate fraction of Haematocarpus validus Bakh. F. Ex Forman, exhibited antimicrobial
and antioxidant activity [57]. Oxalic acid, 6-ethyloct-3-yl heptyl ester, as one of the major
constituents of Ricinus communis seed oil, exhibited anti-inflammatory, antibacterial, antiox-
idant, anthelmintic, antidiabetic, anticancer, mosquitocidal, and insecticidal activity [58].
2-Bromododecane was identified in the n-hexane extract of Moringa oleifera root bark and
exhibited antibacterial activity [59]. Ethylamine, 2-(adamantan-1-yl)-1-methyl- was con-
tained in the essential oil from Cinnamomum tamala dried leaves, and this plant has been
reported to have therapeutic activities to treat anorexia, shore throat, skin diseases, colds,
cough, colic, and diarrhea [60]. Pentadecanal, isolated from Pseudoalteromonas haloplanktis
TAC125 culture supernatant, exhibited inhibitory activity toward the biofilm formation of
S. epidermidis [61]. Di-n-decylsulfone was identified in the acetone extract of Spatholobus
purpureus root, and this plant has been used to cure animal hemorrhagic septicemia [62].
Dodecane-1-iodo- was present in Rhazya stricta chloroform extract, which exhibited antidi-
abetic activity [63]. Hexadecanoic acid, methyl ester, contained in the FAME extract of
Sesuvium portulacastrum, possessed antibacterial, anticandidal, and antifungal activity [64].
Benzenepropanoic acid, 3,5-bis(1,1-dimethylethyl)-4-hydroxy-, methyl ester, present in the
hexane, ethyl acetate, and methanol extracts of Ficus bhotanica, had bioactivities such as
antifungal and antioxidant properties [65]. Methyl 3-bromo-1-adamantaneacetate, as the
major component of Caulerpa racemosa, possessed antibacterial and larvicidal activity [66].
1-Cyclohexylnonene was identified in the n-butanol extract of Ehretia serrata and exhibited
broad-spectrum antimicrobial activity [67]. 4-Methyl-2,4-bis(p-hydroxyphenyl)pent-1-ene,
2TMS derivative was identified in the aqueous extract of Luffa acutangula peel [68], and this
compound has been known to have anticancer activity by inducing apoptosis of pancreatic
β-cells [69]. Cyclohexene, 1-nonyl- was identified as a volatile compound in Piper guineense
Metabolites 2023, 13, 281 9 of 28
leaves and seeds, and this spice exerts medicinal functions such as for the treatment of
rheumatism, bronchitis, cough, and intestinal disease. Moreover, the leaf extract exhib-
ited antimicrobial properties [70]. Cyclotrisiloxane, hexamethyl-, mainly contained in the
acetone extract of Turbinaria decurrens, exhibited antioxidant and antidiabetic activity [71].
1,4-Bis(trimethylsilyl)benzene was found in the volatile oil of Glycosmis pentaphylla, and this
plant has in vivo bioactivities such as antidiabetic, antipyretic, antioxidant, antibacterial, an-
thelmintic, antinociceptive, and hepatoprotective properties [72]. 9,12,15-Octadecatrienoic
acid, methyl ester, (Z,Z,Z)- was found in the n-hexane extract of Archidium ohioense (Schimp.
ex Mull), and this compound has been known to have cardioprotective, antioxidant, anti-
cancer, antipyretic, antibacterial, antiarthritic, and antiandrogenic activities [73]. Eicosane,
1-iodo-, contained in the ethyl acetate and petroleum ether extracts of Alnus cremastogyne
pods, exhibited antioxidant activity [74]. Arsenous acid, tris(trimethylsilyl) ester is one of
the major compounds of Momordica cymbalaria methanol extract, and this plant is commonly
used to cure rheumatism, skin disease, diabetes mellitus, diarrhea, and ulcers [75]. From
the above evidence, we can conclude that the B. cernua extract (stem) contains numerous
compounds with various potential bioactivities.
100
120
Cell population (%)
80 100
60 80
Cell population (%)
40 60
40
20
20
0
0
Figure 1. Quantitative analysis of the impact of B. cernua extract addition to malarial parasite‐in‐
Figure 1. Quantitative analysis of the impact of B. cernua extract addition to malarial parasite-
Figure 1. Quantitative analysis of the impact of B. cernua extract addition to malarial parasite‐in‐
fected erythrocytes.
fected
infectederythrocytes. Control
erythrocytes.Control
Control(−):
(−): ):erythrocytes;
(−erythrocytes; control
control
erythrocytes; (+):
control (+): erythrocytes
erythrocytes
(+): + P.
erythrocytes + P. falciparum;
falciparum;
+ P. falciparum; treatment:
treatment:
treatment:
erythrocytes + P. falciparum + methanol extracts at various concentrations (600 ppm, 300 ppm, 150
erythrocytes + P. falciparum + methanol extracts at various concentrations (600 ppm, 300 ppm, 150
erythrocytes + P. falciparum + methanol extracts at various concentrations (600 ppm, 300 ppm, 150 ppm,
ppm, 100 ppm, 50 ppm, 20 ppm, 10 ppm, 5 ppm, 2 ppm, or 1 ppm). All of the data except for 1 and
ppm, 100 ppm, 50 ppm, 20 ppm, 10 ppm, 5 ppm, 2 ppm, or 1 ppm). All of the data except for 1 and
100 ppm, 50 ppm, 20 ppm, 10 ppm, 5 ppm, 2 ppm, or 1 ppm). All of the data except for 1 and 2 ppm
2 ppm extract were considered significantly differ compared to the control (−) (p <0.05).
2 ppm extract were considered significantly differ compared to the control (−) (p <0.05).
extract were considered significantly differ compared to the control (−) (p <0.05).
Figure 2. Qualitative analysis of the impact of B. cernua extract addition to malarial parasite‐infected
Figure 2. Qualitative analysis of the impact of B. cernua extract addition to malarial parasite-infected
erythrocytes. Control (−): erythrocytes; control (+): erythrocytes + P. falciparum; treatment: erythro‐
erythrocytes. Control (−): erythrocytes; control (+): erythrocytes + P. falciparum; treatment: erythro-
cytes + P. falciparum + methanol extracts at various concentrations (600 ppm, 300 ppm, 150 ppm, 100
cytes + P. falciparum + methanol extracts at various concentrations (600 ppm, 300 ppm, 150 ppm,
ppm, 50 ppm, 20 ppm, 10 ppm, 5 ppm, 2 ppm, or 1 ppm).
Figure 2. Qualitative analysis of the impact of B. cernua extract addition to malarial parasite‐infected
100 ppm, 50 ppm, 20 ppm, 10 ppm, 5 ppm, 2 ppm, or 1 ppm).
erythrocytes. Control (−): erythrocytes; control (+): erythrocytes + P. falciparum; treatment: erythro‐
cytes + P. falciparum + methanol extracts at various concentrations (600 ppm, 300 ppm, 150 ppm, 100
ppm, 50 ppm, 20 ppm, 10 ppm, 5 ppm, 2 ppm, or 1 ppm).
Metabolites 2023, 13, x FOR PEER REVIEW
Metabolites 2023, 13, 281 11 11
of of27
28
Figure 3. The impact of B. cernua extract addition to the malarial parasite‐infected erythrocytes
(Giemsa staining). Control (−): erythrocytes; control (+): erythrocytes + P. falciparum; treatment:
erythrocytes + P. falciparum + methanol extract at 5 ppm. Red arrow: infected erythrocytes at the ring
formation stage; blue arrow: intact cells following extract treatment.
3.4. Anticancer Test Result
Previous research stated that n‐hexane fraction of B. cernua had the highest cytotoxic
toward MCF‐7 breast cancer cells compared to ethyl acetate and water fractions, with an
IC50 value of 165.65 ppm. On the basis of these data, a test was conducted on the methanol
extract of B. cernua using the MCF‐7 cell line. The test was conducted at various concen‐
trations to determine the extract’s activity in killing cancer cells. The result showed that
the extract started killing cancer cells within 3 h of treatment at concentrations of 1600
Figure 3.3. The
Figure The impact
impact of of B.
B. cernua
cernua extract
extract addition to the
addition to the malarial
malarial parasite‐infected
parasite-infected erythrocytes
erythrocytes
ppm and 800 ppm. Concentrations under 800 ppm did not show any effects on the cells.
(Giemsa −): erythrocytes; P.falciparum;
falciparum;treatment:
(Giemsa staining).
staining). Control
Control ((−): erythrocytes;control
control(+):
(+):erythrocytes
erythrocytes+ +P. treatment:
(Figure 4). Further morphology analysis showed that B. cernua (1600 ppm, 3 h) induced
erythrocytes + P. falciparum + methanol extract at 5 ppm. Red arrow: infected erythrocytes at the ring
erythrocytes + P. falciparum + methanol extract at 5 ppm. Red arrow: infected erythrocytes at the ring
stage; blue arrow: intact cells following extract treatment.
cell shrinkage and apoptotic body formation, which are hallmarks of apoptosis (Figure 5).
formation stage; blue arrow: intact cells following extract treatment.
formation
3.4. Anticancer Test Result
45% Previous research stated that n‐hexane fraction of B. cernua had the highest cytotoxic
40% toward MCF‐7 breast cancer cells compared to ethyl acetate and water fractions, with an
3h
35% IC50 value of 165.65 ppm. On the basis of these data, a test was conducted on the methanol
30% 6h
extract of B. cernua using the MCF‐7 cell line. The test was conducted at various concen‐
Cell death
trations to determine the extract’s activity in killing cancer cells. The result showed that
25% 8h
the extract started killing cancer cells within 3 h of treatment at concentrations of 1600
20% ppm and 800 ppm. Concentrations under 800 ppm did not show any effects on the cells.
24h
(Figure 4). Further morphology analysis showed that B. cernua (1600 ppm, 3 h) induced
15%
cell shrinkage and apoptotic body formation, which are hallmarks of apoptosis (Figure 5).
10%
5% 45%
0% 40% 3h
35%Control 1600 ppm 800 ppm 400 ppm 200 ppm
30% 6h
Cell death
25% Concentration 8h
20%
24h
15% Figure 4. Brenia cernua induced the death of MCF‐7 cancer cells (dose and time dependency). The
Figure 4. Brenia cernua induced the death of MCF-7 cancer cells (dose and time dependency). The cell
10% cell viability was estimated by measuring the absorbance at λ570 nm using a Quant ELISA plate
viability was estimated by measuring the absorbance at λ570 nm using a Quant ELISA plate reader
5% reader (Agilent BioTek Instruments). Control: cancer cells treated with solvent (DMSO 0.5% v/v).
(Agilent BioTek Instruments). Control: cancer cells treated with solvent (DMSO 0.5% v/v).
0%
3.5. Virtual Screening with Blind Docking Approach
Control 1600 ppm 800 ppm 400 ppm 200 ppm
Virtual screening was used to estimate each compound’s affinity to selected target
Concentration
proteins: 3VSL as an antibacterial protein model, 3EUB as an antioxidant protein model,
3QS1 as an antiplasmodial protein model, and 3GD4 as an anticancer protein model. Blind
docking performed using Pyrx software revealed that 44 of 45 compounds were successfully
Figure 4. Brenia cernua induced the death of MCF‐7 cancer cells (dose and time dependency). The
docked into the target proteins. However, the docking analysis could not be performed
cell viability was estimated by measuring the absorbance at λ570 nm using a Quant ELISA plate
reader (Agilent BioTek Instruments). Control: cancer cells treated with solvent (DMSO 0.5% v/v).
using arsenous acid, tris(trimethylsilyl) because the arsenic atom could not be recognized
by AutoDock Vina software (Table 2).
Metabolites 2023, 13, 281
Metabolites 2023, 13, x FOR PEER REVIEW 1212
of of
28 27
Figure 5. The application of Brenia cernua to MCF‐7 cell line. Trypan blue staining was used to meas‐
Figure 5. The application of Brenia cernua to MCF-7 cell line. Trypan blue staining was used to
ure the level of cell death. Control: cancer cells treated with solvent (DMSO 0.5% v/v). The cell mor‐
measure the level of cell death. Control: cancer cells treated with solvent (DMSO 0.5% v/v). The cell
phology was observed using inverted microscopy (Agilent). Apoptotic bodies were characterized
morphology was observed using inverted microscopy (Agilent). Apoptotic bodies were characterized
by cell shrinkage.
by cell shrinkage.
3.5. Virtual Screening with Blind Docking Approach
Table 2. Binding affinity of identified compounds to selected target proteins. Nd: not detected.
Virtual screening was used to estimate each compound’s affinity to selected target
No Name of Compounds Binding Affinity between Ligands and Target Proteins (kcal/mol)
proteins: 3VSL as an antibacterial protein model, 3EUB as an antioxidant protein model,
Antibacterial Antioxidant Antiplasmodial Anticancer
3QS1 as an antiplasmodial protein model, and 3GD4 as an anticancer protein model. Blind
(3VSL) (3EUB) (3QS1) (3GD4)
docking performed using Pyrx software revealed that 44 of 45 compounds were success‐
1 Methane, dichloronitro- −3.9
fully docked into the target proteins. −4.4 the docking
However, −analysis
3.7 could −not
4.2 be per‐
2 Ether, hexyl pentyl: formed using arsenous acid, tris(trimethylsilyl) because the arsenic atom could not be rec‐
−4.2 −4.8 −4.5 −4.9
3 ognized by AutoDock Vina software (Table 2).
1,5-Heptadien-4-ol, 3,3,6-trimethyl- −5.4 −5.3 −5.0 −5.0
4 Sulfurous acid, 2-ethylhexyl isohexyl ester −4.3 −4.9 −4.7 −4.5
Table 2. Binding affinity of identified compounds to selected target proteins. Nd: not detected.
5 Sulfurous acid, hexyl nonyl ester −4.6 −5.3 −4.4 −4.7
6 Decane, 2,4-dimethyl-
Binding Affinity between Ligands and Target Proteins
−4.5 −4.4 −4.6 −4.7
No Name of Compounds
(kcal/mol)
7 Octane, 3,4,5,6-tetramethyl- −5.2 −5.3 −4.7 −4.7
Antibacterial Antioxidant Antiplasmodial Anticancer
8 Oxalic acid, cyclohexyl nonyl ester −5.4 −5.1 −4.0 −52
(3VSL) (3EUB) (3QS1) (3GD4)
9 Octane, 2,4,6-trimethyl- −5.3 −5.1 −4.7 −5.3
1 Methane, dichloronitro‐ −3.9 −4.4 −3.7 −4.2
2 N-[β-Hydroxy-β-[4-[1-adamantyl-6,8-
Ether, hexyl pentyl:
10 −−4.2
9.2 −4.8
−10.8 −9.0−4.5 −9.3−4.9
dichloro]quinolyl]ethyl]piperidine
3 1,5‐Heptadien‐4‐ol, 3,3,6‐trimethyl‐ −5.4 −5.3 −5.0 −5.0
11 1-Chloroundecane −4.9 −5.2 −4.7 −5.1
4 Sulfurous acid, 2‐ethylhexyl isohexyl ester −4.3 −4.9 −4.7 −4.5
12 Dodecane, 1-chloro- −3.8 −5.0 −4.7 −5.4
5 Sulfurous acid, hexyl nonyl ester −4.6 −5.3 −4.4 −4.7
6 13 Decane, 1-chloro-
Decane, 2,4‐dimethyl‐ −−4.5
4.3 −−4.4
4.8 −3.1−4.6 −4.4−4.7
7 Propanoic acid, 2,2-dimethyl-,
Octane, 3,4,5,6‐tetramethyl‐
14 −−5.2
6.3 −−5.3
6.6 −6.0−4.7 −7.9−4.7
2-(1,1-dimethylethyl)phenyl ester
8 Oxalic acid, cyclohexyl nonyl ester −5.4 −5.1 −4.0 −52
15 Sulfurous acid, decyl hexyl ester −4.9 −4.7 −3.9 −4.6
9 Octane, 2,4,6‐trimethyl‐ −5.3 −5.1 −4.7 −5.3
N‐[β‐Hydroxy‐β‐[4‐[1‐adamantyl‐6,8‐
10 −9.2 −10.8 −9.0 −9.3
dichloro]quinolyl]ethyl]piperidine
11 1‐Chloroundecane −4.9 −5.2 −4.7 −5.1
12 Dodecane, 1‐chloro‐ −3.8 −5.0 −4.7 −5.4
Metabolites 2023, 13, 281 13 of 28
Table 2. Cont.
No Name of Compounds Binding Affinity between Ligands and Target Proteins (kcal/mol)
Antibacterial Antioxidant Antiplasmodial Anticancer
(3VSL) (3EUB) (3QS1) (3GD4)
16 Tetradecane, 1-chloro- −4.2 −4.8 −3.7 −4.1
17 Dodecane, 4,6-dimethyl- −4.3 −5.7 −4.6 −5.6
18 Cyclohexene, 2-ethenyl-1,3,3-trimethyl- −5.3 −5.7 −5.2 −5.5
19 Pentane, 2,3,3-trimethyl- −4.9 −4.7 −4.3 −4.8
20 Hexadecane −4.0 −5.3 −3.8 −4.5
21 Benzenepropanoic acid,α-hydroxy-, methyl ester −6.2 −6.6 −5.6 −5.9
22 1,3-Phenylene, bis(3-phenylpropenoate) −9.4 −8.4 −8.7 −8.3
23 1,2:4,5:9,10-Triepoxydecane −4.5 −5.7 −4.3 −5.5
1,3-Cyclohexadiene,
24 −5.7 −6.4 −7.4 −5.0
5-(1,5-dimethyl-4-hexenyl)-2-methyl-, [S-(R*,S*)]-
25 1-Octadecanesulphonyl chloride −5.4 −4.5 −4.3 −6.6
26 Oxalic acid, allyl pentadecyl ester −5.4 −4.8 −4.2 −5.2
27 2-Piperidinone, N-[4-bromo-n-butyl]- −4.8 −4.9 −3.7 −5.7
28 2-Bromotetradecane −4.0 −5.2 −4.2 −4.2
29 Oxalic acid, 6-ethyloct-3-yl heptyl ester −5.3 −5.9 −5.4 −7.0
30 2-Bromo dodecane −3.8 −5.3 −3.8 −4.4
31 Ethylamine, 2-(adamantan-1-yl)-1-methyl- −6.0 −7.0 −6.6 −7.3
32 Pentadecanal- −3.8 −5.3 −4.5 −5.0
33 Di-n-decylsulfone −4.6 −4.6 −4.5 −5.0
34 Dodecane, 1-iodo- −3.9 −4.8 −3.2 −5.0
35 Hexadecanoic acid, methyl ester −5.0 −5.4 −3.7 −6.2
Benzenepropanoic acid,
36 3,5-bis(1,1-dimethylethyl)-4-hydroxy-, −6.6 −6.5 −7.0 −6.6
methyl ester
37 Methyl 3-bromo-1-adamantaneacetate −6.3 −6.1 −6.8 −7.4
38 1-Cyclohexylnonene −5.5 −6.2 −3.8 −6.6
4-Methyl-2,4-bis(p-hydroxyphenyl)pent-1-ene,
39 −6.9 −6.5 −7.1 −6.7
2TMS derivative
40 Cyclohexene, 1-nonyl- −5.0 −5.1 −4.7 −5.4
41 Cyclotrisiloxane, hexamethyl- −4.8 −4.9 −4.4 −5.6
42 1,4-Bis(trimethylsilyl)benzene −5.1 −5.7 −4.8 −4.9
9,12,15-Octadecatrienoic acid, methyl
43 −5.3 −6.7 −4.7 −7.6
ester, (Z,Z,Z)-
44 Eicosane, 1-iodo- −4.8 −5.0 −2.9 −4.4
45 Arsenous acid, tris(trimethylsilyl) ester ND ND ND ND
in vivo evidence, four protein models were chosen as molecular docking targets, as they
were previously crystallized and/or used for in silico studies to investigate the binding
affinity of particular compounds: 3EUB as an antioxidant target protein [77], 3VSL as an
antibacterial target protein [78], 3QS1 as an antiplasmodial target protein [79], and 3GD4 as
an anticancer target protein [80]. According to the virtual screening
Metabolites 2023, 13, x FOR PEER REVIEW results
14 of 27 (Table 2), N-
[β-hydroxy-β-[4-[1-adamantyl-6,8-dichloro]quinolyl]ethyl]piperidine and 1,3-phenylene,
bis(3-phenylpropenoate) exhibited the best binding affinity toward all protein models.
Hence, the detailed interactions between the two phenolic compounds and the target
anticancer target protein [80]. According to the virtual screening results (Table 2), N‐[β‐
hydroxy‐β‐[4‐[1‐adamantyl‐6,8‐dichloro]quinolyl]ethyl]piperidine
proteins were further investigated. and 1,3‐phenylene,
bis(3‐phenylpropenoate) exhibited the best binding affinity toward all protein models.
Xanthine oxidase (pdb:3EUB) was selected as an antioxidant protein target since
Hence, the detailed interactions between the two phenolic compounds and the target pro‐
the methanol extract of B. cernua stem possessed inhibitory activity against the enzyme
teins were further investigated.
according to the SOD assay.The binding interaction between 3EUB and N-[β-hydroxy-β-[4-
Xanthine oxidase (pdb:3EUB) was selected as an antioxidant protein target since the
[1-adamantyl-6,8-dichloro]quinolyl]ethyl]piperidine revealed one conventional hydrogen
methanol extract of B. cernua stem possessed inhibitory activity against the enzyme ac‐
cording to the SOD assay.The binding interaction between 3EUB and N‐[β‐hydroxy‐β‐[4‐
bond formed between Leu257 and the oxygen atom of the ligand. One electrostatic in-
[1‐adamantyl‐6,8‐dichloro]quinolyl]ethyl]piperidine
teraction was formed between the ligand and revealed
Lys256 one on
conventional hydro‐
3EUB (π–cation). Furthermore,
gen bond formed between Leu257 and the oxygen atom of the ligand. One electrostatic
there were 16 hydrophobic interactions involving Ile264 (alkyl),
interaction was formed between the ligand and Lys256 on 3EUB (π–cation). Furthermore,
Leu257, Ile353, Lys395,
Leu398 (π–alkyl), and 11 other amino acids including Arg394 (van
there were 16 hydrophobic interactions involving Ile264 (alkyl), Leu257, Ile353, Lys395, der Waals) (Figure 6).
The binding interaction between 3EUB and 1,3-phenylene, bis(3-phenylpropenoate) re-
Leu398 (π–alkyl), and 11 other amino acids including Arg394 (van der Waals) (Figure 6).
The binding
vealed twointeraction between
conventional 3EUB and bonds
hydrogen 1,3‐phenylene,
formed bis(3‐phenylpropenoate)
between Leu257 and re‐ Val259 and the
vealed
carboxyltwo group
conventional
of thehydrogen bonds formed abetween
ligand. Meanwhile, van derLeu257
Waalsand Val259 and
interaction wasthe formed between
carboxyl group of the ligand. Meanwhile, a van der Waals interaction was formed be‐
Lys256 and 1,3-phenylene, bis(3-phenylpropenoate). Moreover, there were 16 hydrophobic
tween Lys256 and 1,3‐phenylene, bis(3‐phenylpropenoate). Moreover, there were 16 hy‐
interactions involving Ala301 (π–alkyl), Leu257 (π–sigma), and 14 other amino acids (van
drophobic interactions involving Ala301 (π–alkyl), Leu257 (π–sigma), and 14 other amino
der Waals)
acids (van der (Figure S1). Arg880
Waals) (Figure andand
S1). Arg880 Glu1261 are
Glu1261 active
are active residues
residues of of xanthine oxidase [26].
xanthine
oxidase [26]. Both plant‐derived molecules could be antioxidant drugs by binding to xan‐
Both plant-derived molecules could be antioxidant drugs by binding to xanthine oxidase in
thine oxidase in a competitive manner, leading to the reduction in reactive oxygen species
a competitive manner, leading to the reduction in reactive oxygen species in the cell.
in the cell.
(a)
(b) (c)
(a)
(b) (c)
lows: 200 Da ≤ molecular weight ≤ 400 Da, −2 ≤ log P ≤ 5, TPSA ≤ 150 A2 , <5 hydrogen
bond donors, <10 hydrogen bond acceptors, <15 rotatable bonds, <7 rings, and at least four
carbon atoms and one heteroatom. Solubility was measured using the ESOL model, where
a log S value ≤−6 indicates a poorly soluble compound, while a log S value ≤−10 indicates
an insoluble compound. Additionally, a compound with 0.55 ≤ bioavailability score ≤ 0.85
and 0.25 ≤ Csp3 (degree of flexibility) score ≤1 is qualified as orally bioavailable [82].
Compounds with no more than one violation are promising drug candidates. According to
Lipinski’s criteria, all of the identified molecules in the methanol extract of B. cernua (stem)
can be considered oral drug candidates. According to Ghose’s criteria, almost all molecules
can be considered drug candidates except for compounds 1 and 10. The filter process using
Veber and Egan criteria showed that all molecules qualified as drug candidates. On the
basis of Muegge’s criteria, only compounds 4, 5, 8, 10, 14, 21, 22, 23, 27, 35, 36, 37, 39,
and 41 qualified as drug candidates. The bioavailability and Csp3 score revealed that all
compounds were orally bioavailable except compound 22.
Binding Energy
with N-[β- Binding Energy
Binding Energy Hydroxy-β-[4-[1- with 1,3-Phenylene,
No Native Ligand with Native adamantyl-6,8- bis(3-
Ligand (kcal/mol) dichloro]quinolyl] phenylpropenoate)
ethyl]piperidine (kcal/mol)
(kcal/mol)
Antibacterial protein
1 Cefotaxime −7.6 −7.8 −8.3
target: 3VSL
Flavin adenine
−13.0 −10.1 −10.6
dinucleotide (FAD)
Phosphonic acidmono-(2-
Antioxidant protein
2 amino-5,6-dimercapto-4-oxo-
target: 3EUB
3,7,8A,9,10,10A-hexahydro-4H-
−9.9 −3.6 −7.8
8-oxa-1,3,9,10-tetraaza-
anthracen-7-
ylmethyl)ester (MTE)
(4R)-3-[(2S,3S)-3-{[(2,6-
Dimethylphenoxy)acetyl]amino}-
2-hydroxy-4-phenylbutanoyl]-
Antiplasmodial N-[(1S,2R)-2-hydroxy-2,3-
3 −10.5 −10.6 −9.2
protein target: 3QS1 dihydro-1H-inden-1-yl]-5,5-
dimethyl-1,3-thiazolidine-4-
carboxamide
(KNI-10006)
Flavin adenine
−14.6 −9.1 −10.1
dinucleotide (FAD)
Anticancer protein
4 target: 3GD4 Nicotinamide adenine
−10.5 −11.1 −10.5
dinucleotide (NAD)
have been using particular strategies such as encapsulating drugs in lipid formulations [84].
The pharmacokinetic analysis also revealed that half of the compounds exhibited low pene-
tration into the BBB and half of the compounds exhibited high penetration into the BBB.
Generally, a low penetration of drugs into the BBB is required to minimize the side-effects
on the central nervous system (CNS). On the other hand, drugs with high permeability
across the BBB are needed to cure cancer patients with brain metastases [85]. The prediction
of the interactions with pharmacokinetic proteins revealed that almost all compounds
had a low interaction with CYP enzymes, thus indicating minimal drug–drug interactions.
Moreover, all compounds had easy to moderate synthetic accessibility. In view of this
thorough analysis, these compounds can be good drug leads of natural origin. Further
studies on the purification or synthesis of these can be conducted to reveal the mechanisms
underlying their antioxidant, antibacterial, antiplasmodial, and anticancer activities.
Metabolites 2023, 13, 281 19 of 28
Table 4. Drug likeness results of the identified compounds from B. cernua. Nd: not detected.
Table 4. Cont.
Table 4. Cont.
Table 5. Cont.
4. Conclusions
As a country with huge biodiversity, Indonesia has abundant plant resources to be
used as herbal medicine or as metabolite reservoirs for drug discovery. Amongst them,
B. cernua is a potential medicinal plant to be explored since it has been traditionally used to
treat various diseases. We provided in vitro evidence that the methanol extract of B. cernua
(stem) exhibits antioxidant, antibacterial, antiplasmodial, and anticancer activities. In
addition, the plant has been traditionally demonstrated to have antiviral activity. Further
analysis revealed that the extract induced apoptosis in the MCF-7 breast cancer cell line
and protected erythrocytes from the ring formation stage of P. falciparum. Then, to iden-
tify important phytoconstituents, the extract’s metabolites were profiled using GC–MS
analysis, which revealed 45 compounds that could contribute to the plant’s therapeutic
functions. Bioinformatics was used for drug discovery to save time and chemical reagents.
Therefore, blind docking was performed to identify the binding affinity of the metabo-
lites toward target proteins, which revealed that N-[β-hydroxy-β-[4-[1-adamantyl-6,8-
dichloro]quinolyl]ethyl]piperidine and 1,3-phenylene, bis(3-phenylpropenoate) exhibited
the best results. Further analysis showed that the two molecules were predicted to have an
inhibition effect toward antioxidant, antiplasmodial, and anticancer protein models in a
competitive manner. Meanwhile, the two molecules were predicted to have an inhibitory
effect toward the antibacterial protein model in a noncompetitive manner. Site-specific
docking analysis showed that the two compounds exhibited similar binding energy to
the native ligands. Interestingly, their bioactivities have not yet been documented. The
drug likeness and pharmacokinetic analyses revealed that almost all identified compounds
can be considered promising drugs, showing good oral bioavailability, high absorption,
and good synthetic availability. Our results clearly indicate that the in silico studies were
supported by in vitro studies.
Supplementary Materials: The following supporting information can be downloaded at: https://
www.mdpi.com/article/10.3390/metabo13020281/s1, Figure S1. Binding interaction of1,3-Phenylene,
bis(3-phenylpropenoate) compound with antioxidant target protein (3EUB) based on the binding
energy generated by PyRx program.A; Figure S2: Binding interaction of N-[.beta.-Hydroxy-.beta.-
[4-[1-adamantyl-6,8-dichloro]quinolyl]ethyl] piperidine compound with antibacterial target protein
(3VSL) based on the binding energy generated by PyRx program; Figure S3: Binding interaction of
N-[.beta.-Hydroxy-.beta.-[4-[1-adamantyl-6,8-dichloro]quinolyl]ethyl] piperidine compound with an-
tiplasmodial target protein (3QS1) based on the binding energy generated by PyRx pro-gram; Figure
S4: Binding interaction of1,3-Phenylene, bis(3-phenylpropenoate) compound with an-tiplasmodial
target protein (3QS1) based on the binding energy generated by PyRx program; Figure S5: Bind-
ing interaction of N-[.beta.-Hydroxy-.beta.-[4-[1-adamantyl-6,8-dichloro]quinolyl]ethyl] piperidine
compound with anticancer target protein (3GD4) based on the binding energy gener-ated by PyRx
program; Figure S6: Binding interaction of 1,3-Phenylene, bis(3-phenylpropenoate) compound with
an-ti-cancer target protein (3GD4) based on the binding energy generated by PyRx program; Figure
S7: 2D interaction of Antioxidant protein target (3EUB) and ligands; Figure S8: 2D interaction of
Antibacterial protein target (3VSL) and ligands; Figure S9: 2D interaction of Antioxidant protein
target (3EUB) and ligands; Figure S10: 2D interaction of Antiplasmodial protein target (3QS1) and
ligands; Figure S11: 2D interaction of Anticancer protein target (3GD4) and ligands; Figure S12: 2D
interaction of Anticancer protein target (3GD4) and ligands.
Author Contributions: Conceptualization, H.L.W. and S.; methodology, H.L.W. and D.K.; software,
H.L.W. and I.F.M.; validation, G.W.P. and A.R.A.; formal analysis, A.L.; investigation, A.R.A. and A.L.;
resources, S; data curation, H.L.W.; writing—original draft preparation, H.L.W.; writing—review and
editing, H.L.W., N.F., G.W.P., D.K., R.A.K., A.B. and I.F.M.; visualization, A.L. and I.F.M.; supervision,
H.L.W., N.F., G.W.P., D.K., R.A.K. and A.B.; project administration, A.L.; funding acquisition, H.L.W.
All authors have read and agreed to the published version of the manuscript.
Funding: The APC was funded by Directorate of Research and Community Engagement Universi-
tas Padjadjaran.
Institutional Review Board Statement: Not applicable.
Metabolites 2023, 13, 281 25 of 28
References
1. Abdel-Razek, A.S.; El-Naggar, M.E.; Allam, A.; Morsy, O.M.; Othman, S.I. Microbial Natural Products in Drug Discovery. Processes
2020, 8, 470. [CrossRef]
2. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Supuran, C.T. Natural Products in Drug Discovery: Advances and Opportunities.
Nat. Rev. Drug Discov. 2021, 20, 200–216. [CrossRef]
3. Pan, S.; Zhou, S.; Gao, S.; Yu, Z.; Zhang, S.; Tang, M.; Sun, J.; Ma, D.; Han, Y.; Fong, W.; et al. New Perspectives on How to
Discover Drugs from Herbal Medicines: CAM’s Outstanding Contribution to Modern Therapeutics. eCAM 2013, 2013, 627375.
[CrossRef]
4. Li, C.Q.; Lei, H.M.; Hu, Q.Y.; Li, G.H.; Zhao, P.J. Recent Advances in the Synthetic Biology of Natural Drugs. Front. Bioeng.
Biotechnol. 2021, 9, 691152. [CrossRef] [PubMed]
5. Anand, U.; Jacobo-Herrera, N.; Altemimi, A.; Lakhssassi, N.A. Comprehensive Review on Medicinal Plants as Antimicrobial
Therapeutics: Potential Avenues of Biocompatible Drug Discovery. Metabolites 2019, 9, 258. [CrossRef]
6. Kumar, A.; Kumar, S.; Komal; Ramchiary, N.; Singh, P. Role of Traditional Ethnobotanical Knowledge and Indigenous Communi-
ties in Achieving Sustainable Development Goals. Sustainability 2021, 13, 3062. [CrossRef]
7. Cahyaningsih, R.; Brehm, J.M.; Maxted, N. Gap Analysis of Indonesian Priority Medicinal Plant Species as Part of Their
Conservation Planning. Glob. Ecol. Conserv. 2021, 26, e01459. [CrossRef]
8. Saadullah, M.; Asif, M.; Arif, S.; Kanwal, B. A Comprehensive Review on Traditional Uses, Chemical Constituents, and Diverse
Pharmacological Importance of the Genus Breynia. Rec. Nat Prod. 2022, 6, 538–549. [CrossRef]
9. Fitriyanti, F.; Wathan, N.; Gunawan, G. Kajian Farmakognostik Tumbuhan Sugi-Sugi (Breynia cernua Muel. Arg.) Asal Amuntai
Kalimantan Selatan. J. Pharma. Sci. 2016, 3, 43–48. [CrossRef]
10. Dirgantara, S.; Tanjung, S.H.R.; Maury, H.K.; Meiyanto, E. Cytotoxic Activity and Phytochemical Analysis of Breynia cernua from
Papua. IJPST 2018, 1, 31–36. [CrossRef]
11. Wulansari, T.Y.I.; Dewi, A.P. Anatomical Structure of Phyllanthaceae Leaf from Banggai Kepulauan. Al-kauniyah: J. Biologi 2021,
14, 29–41. [CrossRef]
12. Khan, M.R.; Omoloso, A.D. Antibacterial and Antifungal Activities of Breynia cernua. Fitoterapia 2008, 79, 370–373. [CrossRef]
13. Saadullah, M.; Arif, S.; Hussain, L.; Asif, M.; Khurshid, U. Dose Dependent Effects of Breynia cernua Against the Paraquat
Induced Parkinsonism like Symptoms in Animals’ Model: In Vitro, In Vivo and Mechanistic Studies. Dose-Response 2022, 20, 1–16.
[CrossRef]
14. Salmerón-Manzano, E.; Garrido-Cardenas, J.A.; Manzano-Agugliaro, F. Worldwide Research Trends on Medicinal Plants. Int. J.
Environ. Res. Public. Health. 2020, 17, 3376. [CrossRef] [PubMed]
15. Konappa, N.; Udayashankar, A.C.; Krishnamurthy, S.; Pradeep, C.K.; Chowdappa, S.; Jogaiah, S. GC–MS Analysis of Phytocon-
stituentsfrom Amomum nilgiricum and Molecular Docking Interactions of Bioactive Serverogenin Acetate With Target Proteins.
Sci. Rep. 2020, 10, 16438. [CrossRef]
16. Azzam, M.H.; Fauziah, N.; Wiraswati, H.L. The Anticancer Effect of Phytochemicals and Potential of Breynia cernua: An overview.
Biomed. Pharmacol. J. 2022, 15, 2259–2278. [CrossRef]
17. Tava, A.; Pecetti, L. Chemical Investigation of Saponins from Twelve Annual Medicago species and Their Bioassay with The Brine
Shrimp Artemia salina. Nat. Prod. Commun. 2012, 7, 837–840. [CrossRef] [PubMed]
18. Lingwan, M.; Masakapalli, S.K. A Robust Method of Extraction and GC-MS Analysis of Monophenols Exhibited UV-B Mediated
Accumulation in Arabidopsis. Physiol. Mol. Biol. Plants 2022, 28, 533–543. [CrossRef] [PubMed]
19. Halder, S.K.; Ahmad, I.; Shathi, J.F.; Mim, M.M.; Hassan, R.; Jewel, J.I.; Dey, P.; Islam, S.; Patel, H.; Morshed, R.; et al. A
Comprehensive Study to Unleash the Putative Inhibitors of Serotype2 of Dengue Virus: Insights from an In Silico Structure-Based
Drug Discovery. Mol. Biotechnol. 2022, 2022, 1–14. [CrossRef]
20. Supandi; Saputra, Y.H.E.; Anwar, C.; Kinanto; Kodir, R.A.; Kurnia, D.; Fauziah, N.; Laelalugina, A.; Wiraswati, H.L. Potential of
Reclamation Area of Coal Mining Sites in Medical Field. IJARET 2020, 11, 714–720.
21. Pinu, F.R.; Edwards, P.J.B.; Jouanneau, S.; Kilmartin, P.A.; Gardner, R.C.; Villas-Boas, S.G. Sauvignon Blanc Metabolomics: Grape
Juice Metabolites Affecting the Development of Varietal Thiols and Other Aroma Compounds in Wines. Metabolomics 2014, 10,
556–573. [CrossRef]
22. Giao, M.S.; Pestana, D.; Faria, A.; Guimaraes, J.T.; Pintado, M.E.; Calhau, C.; Azevedo, I.; Malcatal, F.X. Effects of Extracts of
Selected Medicinal Plants upon Hepatic Oxidative Stress. J. Med. Food. 2010, 13, 131–136. [CrossRef]
Metabolites 2023, 13, 281 26 of 28
23. Balouiri, M.; Sadiki, M.; Ibnsouda, S.K. Methods for in-vitro Evaluating Antimicrobial Activity: A Review. J. Pharm. Anal. 2016, 6,
71–79. [CrossRef] [PubMed]
24. Chiang, Y.S.; Huang, Y.F.; Midha, M.K.; Chen, T.H.; Shiau, H.C.; Chiu, K.P. Single Cell Transcriptome Analysis of MCF-7 Reveals
Consistently and Inconsistently Expressed Gene Groups Each Associated with Distinct Cellular Localization and Functions. PLoS
ONE 2018, 13, e0199471. [CrossRef]
25. Berman, H.M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T.N.; Weissig, H.; Shindyalov, I.N.; Bourne, P.E. The Protein Data Bank.
Nucleic. Acids. Res. 2000, 28, 235–242. [CrossRef]
26. Pauff, J.M.; Cao, H.; Hille, R. Substrate Orientation and Catalysis at the Molybdenum Site in Xanthine Oxidase: Crystal Strcuture
in Complex with xanthine and Lumazine. J. Biol. Chem. 2019, 284, 8760–8767. [CrossRef] [PubMed]
27. Yoshida, H.; Kawai, F.; Obayashi, E.; Akashi, S.; Roper, D.I.; Tame, J.R.; Park, S.Y. Crystal Structures of Penicillin-binding Protein
3 (PBP3) from Methicillin-resistant Staphylococcus aureus in The Apo and Cefotaxime-bound Forms. J. Mol. Biol. 2012, 423,
351–364. [CrossRef]
28. Bhaumik, P.; Horimoto, Y.; Xiao, H.; Miura, T.; Hidaka, K.; Kiso, Y.; Wlodawer, A.; Yada, R.Y.; Gustchina, A. Crystal Structures of
The Free and Inhibited Forms of Plasmepsin I (PMI) from Plasmodium falciparum. J. Struct. Biol. 2011, 175, 73–84. [CrossRef]
29. Sevrioukova, I.F. Redox-linked Conformational Dynamics in Apoptosis-inducing Factor. J. Mol. Biol. 2019, 390, 924–938.
[CrossRef]
30. Pettersen, E.F.; Goddard, T.D.; Huang, C.C.; Couch, G.S.; Greenblatt, D.M.; Meng, E.C.; Ferrin, T.E. UCSF Chimeraa Visualization
System for Exploratory Research and Analysis. J. Comput. Chem. 2004, 25, 1605–1612. [CrossRef]
31. Dallakyan, S.; Olson, A.J. Small-molecule Library Screening by Docking with PyRx. Methods Mol. Biol. 2015, 1263, 243–250.
[CrossRef]
32. Trott, O.; Olson, A.J. AutoDock Vina: Improving the Speed and Accuracy of Docking with A New Scoring Function, Efficient
Optimization and Multithreading. J. Comp. Chem. 2010, 31, 455–461. [CrossRef] [PubMed]
33. Eberhardt, J.; Santos-Martins, D.; Tillack, A.F.; Forli, S. AutoDock Vina 1.2.0: New Docking Methods, Expanded Force Field, and
Python Bindings. J. Chem. Inf. Model. 2021, 1, 3891–3898. [CrossRef] [PubMed]
34. Chatterjee, S.; Ghosh, R.; Mandal, N.C. Production of Bioactive Compounds with Bactericidal and Antioxidant Potential by
Endophytic Fungus Alternaria alternata AE1 Isolated from Azadirachta indica A. Juss. PLoS ONE 2019, 14, e0214744. [CrossRef]
[PubMed]
35. Chen-xing, Z.; Mi, Z.; Jing, H.; Yafang, D.; BaoCai, L. Chemical Composition and Antioxidant Activity of The Essential Oil from
The Flowers of Artemisia austro-yunnanensis. J. Chem. Pharm. Res. 2014, 6, 1583–1587.
36. Arulkumar, A.; Rosemary, T.; Paramasivam, S.; Rajendran, R.B. Phytochemical Composition, In Vitro Antioxidant, Antibacterial
Potential and GC-MS Analysis of Red Seaweeds (Gracilaria corticata and Gracilaria edulis) from Palk Bay, India. Biocatal. Agric.
Biotechnol. 2018, 15, 63–71. [CrossRef]
37. Nalini, D.D.; Mohamed, S.G.A. Phytochemical Screening, GC_MS Analysis, In Vitro Antibacterial Activity and Antioxidant
Property of Eclypta prostrata L. Int. J. Pharm. Biol. Sci. 2018, 8, 2321–3272.
38. Safara, S.; Harighi, B.; Bahramnejad, B.; Ahmadi, S. Antibacterial Activity of Endophytic Bacteria Against Sugar Beet Root Rot
Agent by Volatile Organic Compound Production and Induction of Systemic Resistance. Front. Microbiol. 2022, 13, 921762.
[CrossRef]
39. Obaid, H.H.; Tawfeeq, H.Q.; Khalaf, Z.Z.; Shafeeq, Z.S. Inhibitory Effect of Rhizomes Methanolic Extracts of Rheum ribes and
Tio2 NPs on Escherichia coli. IJSBAR 2016, 30, 265–275.
40. Ahmad, S.; Ullah, F.; Sadiq, A.; Ayaz, M.; Imran, M.; Ali, I.; Zeb, A.; Ullah, F.; Shah, M.R. Chemical Composition, Antioxidant
and Anticholinesterase Potentials of Essential Oil of Rumex hastatus D. Don Collected From the Northwest of Pakistan. BMC
Complement. Altern. Med. 2016, 16, 29. [CrossRef]
41. Rubaye, T.S.; Risan, M.H.; Rubaye, D. Gas Chromatography-mass- spectroscopy Analysis of Bioactive Compounds from
Streptomyces spp. Isolated from Tigris river Sediments in Baghdad City. J. Biotech. Res. Center. 2020, 14, 63–71. [CrossRef]
42. Athira Krishnan, K.A.; Keerthi, T.R. Cultivable Bacterial Symbionts from the Marine Sponge with Biological Activity. BJSTR 2021,
36, 28635–28646. [CrossRef]
43. Erwin, E.; Pratiwi, D.R.; Saputra, I.; Alimuddin, A. Antioxidant Assay with Scavenging DPPH Radical of Artocarpus anisophyllus
Miq Stem bark extracts and Chemical compositions and Toxicity Evaluation for the Most Active Fraction. Res. J. Pharm. Technol.
2021, 14, 2819–2833. [CrossRef]
44. Khairy, H.; Saadoon, A.F.; Zzaman, W.; Yang, T.A.; Easa, A.M. Identification of Flavor Compounds in Rambutan Seed Fat and Its
Mixture with Cocoa Butter Determined by SPME-GCMS. JKSUS 2017, 30, 316–323. [CrossRef]
45. Dini, I.; Marra, R.; Cavallo, P.; Pironti, A.; Sepe, I.; Troisi, J.; Scala, G.; Lombari, P.; Vinale, F. Trichoderma Strains and Metabolites
Selectively Increase the Production of Volatile Organic Compounds (VOCs) in Olive Trees. Metabolites 2021, 11, 213. [CrossRef]
46. Rajadurai, M.V.; Maithili, R.A.; Yogesh, V. Phytochemical Profiling of Medically Significant Crude Extract using GC-MS Analysis.
Int. J. Curr. Pharm. Res. 2021, 10, 16–20. [CrossRef]
47. Khanal, L.N.; Sharma, K.R.; Pokharel, Y.R.; Kalauni, S.K. Characterization of Essential Oil, Estimation of Phenolic and Flavonod
Content and Biological Activities of Ephedra pachyclada BOISS. JIST 2018, 27, 27–35. [CrossRef]
48. Al-Dhabi, N.A.; Mohammed Ghilan, A.; Esmail, G.A.; Valan Arasu, M.; Duraipandiyan, V.; Ponmurugan, K. Bioactivity
Assessment of The Saudi Arabian Marine Streptomyces sp. Al-Dhabi-90, Metabolic Profiling and It’s In Vitro Inhibitory Property
Metabolites 2023, 13, 281 27 of 28
Against Multidrug Resistant and Extended-spectrum Beta-lactamase Clinical Bacterial Pathogens. J. Infect. Public Health. 2019, 12,
549–556. [CrossRef]
49. Yogeswari, S.; Ramalakshmi, S.; Neelavathy, R.; Muthumary, J. Identification and Comparative Studies of Different Volatile
Fractions from Monochaetia kansensis by GCMS. Glob. J. Pharmacol. 2012, 6, 65–71.
50. Ramos, A.N.; Sesto Cabral, M.E.; Arena, M.E.; Arrighi, C.F.; Arroyo Aguilar, A.A.; Valdéz, J.C. Compounds from Lactobacillus
plantarum Culture Supernatants with Potential Pro-healing and Anti-pathogenic Psroperties in Skin Chronic Wounds. Pharm.
Biol. 2015, 53, 350–358. [CrossRef] [PubMed]
51. Van, H.T.; Ngo, N.H.; Nguyen, N.T.; Nguyen, N.A.; Pham, T.V.; Nguyen, H.T.D.; Thieu, G.L.; Dang, T.H.T.; Nguyen, Q.H.; Le,
T.T.; et al. Chemical composition, antibacterial and antioxidant activities of acetone extract from the branches and leaves of
Jasminum annamense subsp. annamense (Oleaceae). J. Phytol. 2022, 14, 17–23. [CrossRef]
52. Khan, F.; Magaji, M.G.; Aguye, I.A.; Hussaini, I.M.; Hamza, A.; Olorukooba, A.B.; Sani, M.A.; Maje, I.M. Phytochemical Profiling
of The Bioactive Principles of Alyscarpus glumaceus (Vahl) DC. Aerial Parts. Istanbul J. Pharm. 2021, 51, 228–238. [CrossRef]
53. Shareef, H.K.; Muhammed, H.J.; Hussein, H.M.; Hameed, I.H. Antibacterial Effect of Ginger (Zingiber officinale) Roscoe and
Bioactive Chemical Analysis using Gas Chromatography Mass Spectrum. Orient J. Chem. 2016, 32, 817–837. [CrossRef]
54. Samling, B.; Umaru1, I.J. Phytochemical Screening, Antioxidant, Antifungal Potentials of Acacia auriculiformis Floral Scent
Composition. J. Anal Pharm. Res. 2018, 7, 646–650. [CrossRef]
55. Manilal, A.; Sujith, S.; Sabarathnam, B.; Kiran, G.S.; Selvin, J.; Shakir, C.; Lipton, A.P. Biological Activity of The Red Alga Laurencia
brandenii. Acta Bot. Croat. 2011, 70, 81–90. [CrossRef]
56. Al-Bahadily, D.C.H.; Shari, F.H.; Najm, M.A.A.; Al-Salman, H.N.K. Antimicrobial Activity of the Compound 2-Piperidinone,
N-[4-Bromo-n-butyl]- Extracted from Pomegranate Peels. Asian J. Pharm. 2019, 13, 46–53. [CrossRef]
57. Sasikumar, R.; Das, D.; Saravanan, C.; Deka, S.C. GC-HRMS Screening of Bioactive Compounds Responsible for Antimicrobial
and Antioxidant Activities of Blood Fruit (Haematocarpus validus Bakh. F. Ex Forman) of North-East India. Arch. Microbiol. 2020,
202, 2643–2654. [CrossRef] [PubMed]
58. Jepkorir, A.K.; Irungu, C.M.; Kendagor, P.B. Chemical composition of Azadirachta indica A. Juss and Ricinus communis Linn.
Seed Oils Growing in Marigat, Baringo County, Kenya. EAJSTI 2020, 1, 1–11. [CrossRef]
59. Aagboke, A.; Aftama, A.A. Bioactive Components and Antibacterial Activities of n-hexane Extract of Moringa oleifera Root Bark
on Clinical Isolates of Methicilin Resistant Staphylococcus aureus. IJCRCPS 2016, 3, 1–9.
60. Chowdhury, J.U.; Shaha, G.C.; Begum, F.; Bhuiyan, M.N.H.; Rahim, M. Essential Oil Composition from Fresh and Dried Leaves of
C. tamala. Bangladesh J. Sci. Ind. Res. 2013, 48, 151–154. [CrossRef]
61. Casillo, A.; Papa, R.; Ricciardelli, A.; Sannino, F.; Ziaco, M.; Tilott, M.; Selan, L.; Marino, G.; Corsaro, M.M.; Tutino, M.L.; et al. Anti-
biofilm Activity of a Long-chain Fatty Aldehyde from Antarctic Pseudoalteromonas haloplanktis TAC125 against Staphylococcus
epidermidis Biofilm. Front. Cell. Infect. Microbiol. 2017, 7, 46. [CrossRef]
62. Deshmukh, O.S. GC-MS Analysis of Root Acetone Extracts of Spatholobus purpureus—A High Ethno-veterinary Medicinal
Value Plant. Int. J. Curr. Res. 2015, 7, 1013–1017.
63. Mahmood, R.; Kayani, W.K.; Ahmed, T.; Malik, F.; Hussain, S.; Ashfaq, M.; Ali, H.; Rubnawaz, S.; Green, B.D.; Calderwood, D.;
et al. Assessment of Antidiabetic Potential and Phytochemical Profiling of Rhazya stricta Root Extracts. BMC Complement. Med.
Ther. 2020, 20, 293. [CrossRef] [PubMed]
64. Chandrasekaran, M.; Senthilkumar, A.; Venkatesalu, V. Antibacterial and Antifungal Efficacy of Fatty Acid Methyl Esters from
The Leaves of Sesuvium portulacastrum L. Eur. Rev. Med. Pharmacol. Sci. 2011, 15, 775–780.
65. Gogoi, D.; Bora, G.; Borgohain, R.; Handique, J.G. Antioxidant Capacity and GC-MS Analysis of Hexane, Ethylacetate and
Methanol Extracts of Ficus bhotanica—A Potential Folklore Medicinal Plant. Int.J. Pharmacog. Phytochem. Res. 2018, 10, 201–212.
66. Nagaraj, S.R.; Osborne, J.W. Bioactive Compounds from Caulerpa racemosa as A Potent Larvicidal and Antibacterial Agent.
Front. Biol. 2014, 9, 300–305. [CrossRef]
67. Waheed, A.; Chohan, M.M.; Ahmed, D.; Ullah, N. The First Report on The In Vitro Antimicrobial Activities of Extracts of Leaves
of Ehretia serrata. Saudi J. Biol. Sci. 2019, 26, 1253–1261. [CrossRef] [PubMed]
68. Ananthalakshmi, R.; Rajarathinam, S.R.X.; Sadiq, A.M.; Poongothai, A. Phytochemical Profiling of Luffa acutangula Peel Extract
using GCMS Study. Res. J. Pharm. Tech. 2019, 12, 6071–6074. [CrossRef]
69. Huang, C.C.; Yang, C.Y.; Su, C.C.; Fang, K.M.; Yen, C.C.; Lin, C.T.; Liu, J.M.; Lee, K.I.; Chen, Y.W.; Liu, S.H.; et al. 4-Methyl-2,4-
bis(4-hydroxyphenyl)pent-1-ene, A Major Active Metabolite of Bisphenol A, Triggers Pancreatic β-Cell Death via a JNK/AMPKα
Activation-Regulated Endoplasmic Reticulum Stress-Mediated Apoptotic Pathway. Int. J. Mol. Sci. 2021, 22, 4379. [CrossRef]
70. Ojinnaka, M.C.; Ubbor, S.C.; Okudo, H.O.; Uga, U. Volatile Compound Analysis of The Leaves and Seeds of Piper guineense
Using Gas Chromatography-mass Spectrometry (GC-MS). Afr. J. Food Sci. 2016, 10, 327–332. [CrossRef]
71. Gehan, I.; Gheda, S.; Abo-Shady, A.; Omnia, A.K. In Vitro Potential Activity of Some Seaweeds as Antioxidants and Inhibitors of
Diabetic Enzymes. Food Sci. Technol. 2019, 40, 681–691. [CrossRef]
72. Prakasia, P.P.; Ashalatha, S.N. Chemical Fingerprint of Essential Oil Components from Fresh Leaves of Glycosmis pentaphylla
(Retz.) Correa. Pharma Innov. 2015, 3, 50–56.
73. Godwin, A.; Akinpelu, B.A.; Makinde, A.M.; Aderogba, M.A.; Oyedapo, O.O. Identification of n-Hexane Fraction Constituents of
Archidium ohioense (Schimp. ex Mull) Extract Using GC-MS Technique. J. Res. Int. 2015, 6, 366–375. [CrossRef]
Metabolites 2023, 13, 281 28 of 28
74. Chen, G.; Pan, F.; Gao, Y.; Li, H.; Qin, X.; Jiang, Y.; Qi, J.; Xie, J.; Jia, S. Analysis of Components and Properties of Extractives from
Alnus cremastogyne Pods from Different Provenances. Molecules 2022, 27, 7802. [CrossRef]
75. Manikandan, G.; Pandiselvi, P.; Sobana, N.; Murugan, M. GC-MS Analysis of Chemical Constituents in The Methanolic Tuber
Extract of Momordica Cymbalaria Hook. F. Int. Res. J. Pharm. 2019, 10, 135–140. [CrossRef]
76. Galati, G.; O’Brien, P.J. Potential Toxicity of Flavonoids and Other Dietary Phenolics: Significance for Their Chemopreventive and
Anticancer Properties. Free Radic. Biol. Med. 2004, 37, 287–303. [CrossRef]
77. Nishiwaki, K.; Ohigashi, K.; Deguchi, T.; Murata, K.; Nakamura, S.; Matsuda, H.; Nakanishi, I. Structure–Activity Relationships
and Docking Studies of Hydroxychavicol and Its Analogs as Xanthine Oxidase Inhibitors. Chem. Pharm. Bull. 2018, 66, 741–747.
[CrossRef]
78. Bin Emran, T.; Rahman, A.; Uddin, M.M.N.; Dash, R.; Hossen, F.; Mohiuddin, M.; Alam, R. Molecular Docking and Inhibition
Studies on The Interactions of Bacopa monnieri’s Potent Phytochemicals Against Pathogenic Staphylococcus aureus. DARU J.
Pharm. Sci. 2015, 23, 26. [CrossRef] [PubMed]
79. Nerdy, N. In Silico Study of Sesquiterpene Lactone Compounds from South Africa leaves (Vernonia amygdalina Del.) as
Antimalarial and Anticancer. Int. J. PharmTech Res. 2015, 7, 47–53.
80. Wiraswati, H.L.; Martoprawiro, M.A.; Akhmaloka.; Warganegara, F.M. Apoptosis Inducing Factor (AIF) Stabilizes Menadione-
Conjugate Product in Programmed Cell Death. Int. J. PharmTech Res. 2017, 10, 237–245. [CrossRef]
81. Wiraswati, H.L.; Hangen, E.; Sanz, A.B.; Lam, N.V.; Reinhardt, C.; Sauvat, A.; Mogha, A.; Ortiz, A.; Kroemer, G.; Modjtahedi,
N. Apoptosis Inducing Factor (AIF) Mediates Lethal Redox Stress Induced by Menadione. Oncotarget 2016, 7, 76496–76507.
[CrossRef]
82. Bogari, H.A.; Rashied, R.M.H.; Abdelfattah, M.A.O.; Malatani, R.T.; Khinkar, R.M.; Hareeri, R.H.; Wink, M.; Sobeh, M. Euclea
divinorum Hiern: Chemical Profiling of the Leaf Extract and Its Antioxidant Activity In Silico, In Vitro and in Caenorhabditis elegans
Model. Metabolites 2022, 12, 1031. [CrossRef] [PubMed]
83. 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, 42717. [CrossRef] [PubMed]
84. Subramanian, N.; Ghosal, S.K. Enhancement of Gastrointestinal Absorption of Poorly Water Soluble Drugs via Lipid based
Systems. Indian J. Exp. Biol. 2004, 42, 1056–1065.
85. Lowery, F.J.; Yu, D. Brain Metastasis: Unique Challenges and Open Opportunities. Biochim. Biophys. Acta Rev. Cancer 2017, 1867,
49–57. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.