ArticlePDF Available

Molecular Mechanisms of Cassia fistula against Epithelial Ovarian Cancer Using Network Pharmacology and Molecular Docking Approaches

Authors:

Abstract and Figures

Epithelial ovarian cancer (EOC) is one of the deadliest reproductive tract malignancies that form on the external tissue covering of an ovary. Cassia fistula is popular for its anti-inflammatory and anticarcinogenic properties in conventional medications. Nevertheless, its molecular mechanisms are still unclear. The current study evaluated the potential of C. fistula for the treatment of EOC using network pharmacology approach integrated with molecular docking. Eight active constituents of C. fistula were obtained from two independent databases and the literature, and their targets were retrieved from the SwissTargetPrediction. In total, 1077 EOC associated genes were retrieved from DisGeNET and GeneCardsSuite databases, and 800 potential targets of eight active constituents of C. fistula were mapped to the 1077 EOC targets and intersected targets from two databases. Ultimately, 98 potential targets were found from C. fistula for EOC. Finally, the protein–protein interaction network (PPI) topological interpretation revealed AKT1, CTNNB1, ESR1, and CASP3 as key targets. This is the first time four genes have been found against EOC from C. fistula. The major enriched pathways of these candidate genes were established by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) investigations. To confirm the network pharmacology findings, the molecular docking approach demonstrated that active molecules have higher affinity for binding to putative targets for EOC suppression. More pharmacological and clinical research is required for the development of a drug to treat EOC.
Content may be subject to copyright.
Citation: Kanwal, A.; Azeem, F.;
Nadeem, H.; Ashfaq, U.A.; Aadil,
R.M.; Kober, A.K.M.H.; Rajoka,
M.S.R.; Rasul, I. Molecular
Mechanisms of Cassia fistula against
Epithelial Ovarian Cancer Using
Network Pharmacology and
Molecular Docking Approaches.
Pharmaceutics 2022,14, 1970.
https://doi.org/10.3390/
pharmaceutics14091970
Academic Editor: Tatjana
P. Stanojkovi´c
Received: 29 July 2022
Accepted: 9 September 2022
Published: 19 September 2022
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2022 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
pharmaceutics
Article
Molecular Mechanisms of Cassia fistula against Epithelial
Ovarian Cancer Using Network Pharmacology and Molecular
Docking Approaches
Aqsa Kanwal 1, , Farrukh Azeem 1 ,† , Habibullah Nadeem 1, Usman Ali Ashfaq 1, Rana Muhammad Aadil 2,
A. K. M. Humayun Kober 3, Muhammad Shahid Riaz Rajoka 4and Ijaz Rasul 1, *
1Department of Bioinformatics and Biotechnology, Government College University Faisalabad,
Faisalabad 38000, Pakistan
2National Institute of Food Science and Technology, University of Agriculture Faisalabad,
Faisalabad 38000, Pakistan
3Department of Dairy and Poultry Science, Chittagong Veterinary and Animal Sciences University,
Chittagong 4225, Bangladesh
4Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University,
Sendai 980-8572, Japan
*Correspondence: ijazrasul@gcuf.edu.pk
These authors contributed equally to this work.
Abstract:
Epithelial ovarian cancer (EOC) is one of the deadliest reproductive tract malignancies that
form on the external tissue covering of an ovary. Cassia fistula is popular for its anti-inflammatory and
anticarcinogenic properties in conventional medications. Nevertheless, its molecular mechanisms are
still unclear. The current study evaluated the potential of C. fistula for the treatment of EOC using
network pharmacology approach integrated with molecular docking. Eight active constituents of
C. fistula were obtained from two independent databases and the literature, and their targets were
retrieved from the SwissTargetPrediction. In total, 1077 EOC associated genes were retrieved from
DisGeNET and GeneCardsSuite databases, and 800 potential targets of eight active constituents
of C. fistula were mapped to the 1077 EOC targets and intersected targets from two databases.
Ultimately, 98 potential targets were found from C. fistula for EOC. Finally, the protein–protein
interaction network (PPI) topological interpretation revealed AKT1, CTNNB1, ESR1, and CASP3 as
key targets. This is the first time four genes have been found against EOC from C. fistula. The major
enriched pathways of these candidate genes were established by Gene Ontology (GO) and Kyoto
Encyclopedia of Genes and Genomes (KEGG) investigations. To confirm the network pharmacology
findings, the molecular docking approach demonstrated that active molecules have higher affinity
for binding to putative targets for EOC suppression. More pharmacological and clinical research is
required for the development of a drug to treat EOC.
Keywords:
epithelial ovarian cancer; Cassia fistula; anticarcinogenic; network pharmacology; active
constituents; gene ontology; molecular docking
1. Introduction
Ovarian cancer (OC) is the world’s seventh highest prevalent lethal gynecological
malignancy and accounted for 2.5% of new cancers in women. In 2019, the number of
new cases was predicted to be 222,240, while the death toll was estimated to be 14,170 [
1
].
The most frequent type of OC is epithelial ovarian cancer (EOC), having a 45.6% survival
rate [
2
]. EOC originates from the ovaries, ovarian surface epithelial (OSE), oviduct, and
pelvic epithelium locales. About 75% individuals are examined at later phases because
EOC symptoms are non-specific and typically include abdominal bloating, distention, early
satiety, nausea, changes in bowel function, urinary tract problems, back pain, fatigue, and
weight loss [
3
]. EOCs are listed at different stages based on conformation of the cancer. The
Pharmaceutics 2022,14, 1970. https://doi.org/10.3390/pharmaceutics14091970 https://www.mdpi.com/journal/pharmaceutics
Pharmaceutics 2022,14, 1970 2 of 17
malignancy is restricted to either one or both ovaries at phase I. In phase II, the tumor also
extends to the uterus and other epithelial tissues in the pelvic region. In phase III, the tumor
advances at lymphoid tissue and is confined to the abdominopelvic cavity. In phase IV, both
or one of the ovaries are implicated in remote metastases [
4
]. EOC is not a single disease,
while studies show that it is composed of tumors categorized on the basis of molecular
genetics. EOC composed of subtypes on the basis of closely resembled tissues encompasses
serous, mucinous, endometriosis, clear-cell and transitional cell types. EOC is treated
surgically, followed by chemotherapy [
4
]. The etiology of EOC includes stress-induced
recurrent ovulation [
5
,
6
], elevated levels of estrogen [
7
], high levels of androgens [
7
], and
stromal hyperactivity [
8
]. However, the etiology and pathogenesis of EOC is very complex
and not yet fully understood.
The use of plants and their components to control different diseases in humans has an
age-old history. Several therapeutic plants have been demonstrated to be effective in the
administration of quality of life through antioxidant, anti-inflammation, and antidiabetic
biological effects [
9
11
]. These herbal medicines, composed of hundreds of phytochemicals,
are derived from many herbs, though their effects and molecular mechanisms are not well
known. Network pharmacology, a new drug development technique, was introduced in
Nature Biotechnology by Hopkins in 2007, updated to the present day “one-target-on-one-
drug” approach to the advanced “network targets, multiple-constituent” technique [
12
].
Network pharmacology analyzes the complex biological mechanisms and selects specific
signaling nodes to develop multi-target drug molecules. It highlights the active constituents,
improves therapeutic effects of drugs, reduces toxicity and maps active constituents to
disease gene to seek potential targets to develop PPI network and carry out gene annotation
analysis [
13
,
14
]. It focuses to demonstrate the interaction of active constituents, potential
targets, and disease-related genes by constructing active constituent/potential target sig-
naling pathway networks [
15
]. In this way, it shows how the screened potential targets
work to cure a disease.
C. fistula belongs to the subfamily Caesalpinioideae of the legume family, Fabaceae
popular as Amaltas. C. fistula is indigenous to the Indian subcontinent, Mauritius, Thailand,
China, Brazil, Sri Lanka, and southern Pakistan. It plays a central role in disease preven-
tion because of its valuable constituents such as flavonoids, anthraquinone, chromones,
coumarins, alkaloids, phytosterols, long-chain hydrocarbons, phenolic, and triterpenes.
Former research confirmed that C. fistula showed roles in antifertility [
16
], antimicrobial [
17
],
hepatoprotective [
18
], improved tissue regeneration and wound healing activities [
19
,
20
].
C. fistula also played a wide range of activities including hypoglycemic activities [
21
,
22
],
antipyretic [
23
], larvicidal [
24
], anti-inflammatory [
25
], antioxidant and antitumor activi-
ties [
26
]. C. fistula is prescribed against epidermis, hepatic, and lung problems as well as
hematemesis, chronic itchy skin, hypopigmentation, and diabetes mellitus [27]. The exact
activities of its molecular mechanisms to prevent a disease are yet entirely unknown. In this
investigation, a C. fistula-target EOC network is constructed, from analysis of compound
to the interaction of potential targets, integrated with pathway analysis to investigate the
molecular mechanism of C. fistula in order to treat EOC.
2. Materials and Methods
2.1. Phytochemical Library Construction
Phyto-constituents of C. fistula were acquired from the literature [28,29], KNApSAcK
Family Core System database [
30
] (http://www.knapsackfamily.com/, accessed on 8
August 2022) and Traditional Chinese Medicine System Pharmacology (TCMSP) (https:
//tcmsp-e.com/tcmsp.php, accessed on 8 August 2022), which was performed by using
the plant name Cassia fistula as a search term in kNAPSAcK database [
31
] and literature
mining performed via Google Scholar and PubMed. Their 2D structures in .sdf file format
were collected through PubChem database (https://pubchem.ncbi.nlm.nih.gov/, accessed
on 10 August 2022) to construct phytochemical library. Meanwhile, their SMILES were also
collected in order to investigate the pharmacodynamics attributes. Active constituents of
Pharmaceutics 2022,14, 1970 3 of 17
C. fistula were obtained from admetSAR (http://lmmd.ecust.edu.cn/admetsar2, accessed
on 13 August 2022) and TCMSP database, i.e., OB and DL greater than 30% and 0.18,
respectively [
32
]. OB stands for oral bioavailability of pharmacological components, while
DL stands for drug-component similarity, which might suggest a prospective drug.
2.2. Drug Target Profiles for C. fistula
SwissTargetPrediction tool (http://www.swisstargetprediction.ch/, accessed on 15
August 2022) was used to find out prospective genes targeting active compounds via
providing the SMILES, while Homo sapiens were selected as the species. SwissTargetPre-
diction is extensively used program for reverse screening of chemical compounds and
predicting the bioactivity of the chemicals. Protein IDs were aligned with UniProtKB
(https://www.uniprot.org/help/uniprotkb, accessed on 16 August 2022) [
33
], performed
in order to eliminate duplication.
2.3. Candidate Targets of C. fistula for EOC
EOC-related Human targets were retrieved from DisGeNET (https://www.disgenet.
org/, accessed on 20 August 2022) and GeneCards (https://www.genecards.org/, accessed
on 20 August 2022) using keyword “epithelial ovarian cancer” [
34
,
35
]. A Venn illustration
was built to map drug-targets profiles of active constituents of C. fistula to the EOC-related
targets [
36
] in order to obtain prospective genes of C. fistula to treat EOC. These mapped
genes were chosen to advance the investigations.
2.4. Compound-Target Network
The network of active constituents of C. fistula with associated genes was built and
analyzed through Cytoscape v3.8.2 [
37
]. Cytoscape is a freely available bioinformatics
platform to visualize, incorporate complex networks from different types of information.
Nodes of network formed represent the phyto-constituents and genes. The edges depict the
interrelationship between them. A plug-in “Network Analyzer” was employed to assess
the network’s topology [38]. The network was evaluated based on “degree” [39].
2.5. Protein–Protein Interaction (PPI) Network
To evaluate the interaction between the prospective genes of C. fistula, these were
imported into STRING v11.5 database (https://string-db.org/, accessed on 25 August 2022)
to construct a PPI network [
40
]. The multiple protein option was taken and potential targets
were added using the “Homo sapiens” as the target species. The network was built with a
confidence of 0.4. This network was further analyzed using Cytoscape for visualization
and topological analysis [
37
]. A plug-in “CytoHubba” was used to obtain targets of higher
degree. In fact, higher the degree means they are linked more [
41
]. “Network Analyzer”
was utilized to analyze the topological properties [38].
2.6. Gene Functional Annotation
The gene and pathway enrichment investigation was performed through DAVID
(https://david.ncifcrf.gov/home.jsp, accessed on 26 August 2022). DAVID is a database of
functional annotations that can be accessed online that helps researchers comprehends the
biological meanings of a huge number of genes. The gene enrichment analysis categorized
the gene functions that incorporate Biological Process (BP), Cellular Component (CC) and
Molecular Function (MF). Moreover, enriched pathways were filtered out through KEGG
analysis [
42
]. The potential genes were copied into DAVID, selecting the species as the
Homo sapiens. The probability value was set to p< 0.05 to select enriched pathways. To
illustrate the GO annotation and KEGG pathways, bubble plots were created in R via
ggplot2 package.
The highest 20 enriched pathways were chosen to construct the pathway–target net-
work, which was constructed and examined by using Cytoscape v3.8.2 to understand
interactions of pathways with the potential targets to evaluate the key targets [37].
Pharmaceutics 2022,14, 1970 4 of 17
2.7. Compound–Target–Signaling Pathway Network
The compound–target–signaling pathways network was built by integration of compound–
target and pathway–target networks. The Cytoscape program was used to understand the
interaction of active constituents with prospective targets and signaling pathways in order
to evaluate the primary targets.
2.8. Molecular Docking
Molecular docking makes it easier to figure out how ligands interact with their cor-
responding proteins. Finally, the results of this network pharmacological study were
verified through molecular docking approach. For that, the 3D structures of active com-
ponents were retrieved from the PubChem search in the .sdf format and optimized [
43
].
The Research Collaboratory for Structural Bioinformatics Protein Data Bank (RCSBPDB)
(https://www.rcsb.org/, accessed on 29 August 2022) was used to retrieve receptor protein
data, and their PDB files were downloaded [
44
]. The input protein was preprocessed by the
Chimera to eliminate ligand molecules from the source file in order to fulfill the docking
criteria.
Auto-dock Vina is used to carry out the protein–ligand docking for the prediction
of predominant binding mode of the ligand with a protein [
45
]. The active constituents
acted as ligands, while the targets acted as receptors. Finally, the best docking results were
selected for the visualization using the Chimera. The graphical framework of the network
pharmacology and molecular docking techniques is shown (Figure 1).
Pharmaceutics 2022, 14, x FOR PEER REVIEW 5 of 20
Figure 1. The network pharmacology and molecular docking techniques employed in order to
predict C. fistula’s potential drug targets for the treatment of EOC depicted graphically.
3. Results
3.1. Active Constituents of C. fistula
A phytochemical library was constructed with the help of previous knowledge and
multiple databases. That library contained 78 phytochemicals isolated from different
parts of C. fistula such as leaves, fruit, bark, stem, as well as seeds (Table S1). Eight phy-
tochemicals (Rhein, Ellagic Acid, Quercetin, Kaempferol, Gibberellin A3, Licoisoflavone,
β-Sitosterol and Stigmasterol) were predicted with pharmacokinetic criteria (30% OB
and 0.18 DL. These eight phytochemicals were filtered as effective constituents and
their properties are presented (Table 1).
Figure 1.
The network pharmacology and molecular docking techniques employed in order to predict
C. fistula’s potential drug targets for the treatment of EOC depicted graphically.
3. Results
3.1. Active Constituents of C. fistula
A phytochemical library was constructed with the help of previous knowledge and
multiple databases. That library contained 78 phytochemicals isolated from different parts
Pharmaceutics 2022,14, 1970 5 of 17
of C. fistula such as leaves, fruit, bark, stem, as well as seeds (Table S1). Eight phytochemicals
(Rhein, Ellagic Acid, Quercetin, Kaempferol, Gibberellin A3, Licoisoflavone,
β
-Sitosterol
and Stigmasterol) were predicted with pharmacokinetic criteria (
30% OB and
0.18 DL.
These eight phytochemicals were filtered as effective constituents and their properties are
presented (Table 1).
Table 1. Properties of active phytochemicals C. fistula.
Sr.No. Phytochemicals Molecular
Formula
Molecular
Weight (Dalton)
Drug Likeness
(>0.18)
Oral Bioavailability
(>30%) 2D Structures PubChem
ID
01 Rhein C15H8O6284.22 0.28 47.07
Pharmaceutics 2022, 14, x FOR PEER REVIEW 6 of 20
Table 1. Properties of active phytochemicals C. fistula.
Sr.No.PhytochemicalsMolecular
Formula
Molecular
Weight (Dalton)
Drug
Likeness
(>0.18)
Oral
Bioavailability
(>30%)
2D Structures PubChem
ID
01 Rhein C15H8O6 284.22 0.28 47.07 10168
02 Ellagic Acid C14H6O8 302.19 0.43 43.06 5281855
03 Quercetin C15H10O7 302.23 0.28 46.43 5280343
04 Kaempferol C15H10O6 286.24 0.24 41.88
5280863
05 Gibberellin A3 C19H22O6 346.4 0.53 81.59
6466
06 Licoisoflavone C20H18O6 354.4 0.42 41.61 5281789
07 β-Sitosterol C29H50O 414.7 0.75 36.91
222284
10168
02 Ellagic Acid C14H6O8302.19 0.43 43.06
Pharmaceutics 2022, 14, x FOR PEER REVIEW 6 of 20
Table 1. Properties of active phytochemicals C. fistula.
Sr.No.PhytochemicalsMolecular
Formula
Molecular
Weight (Dalton)
Drug
Likeness
(>0.18)
Oral
Bioavailability
(>30%)
2D Structures PubChem
ID
01 Rhein C15H8O6 284.22 0.28 47.07 10168
02 Ellagic Acid C14H6O8 302.19 0.43 43.06 5281855
03 Quercetin C15H10O7 302.23 0.28 46.43 5280343
04 Kaempferol C15H10O6 286.24 0.24 41.88
5280863
05 Gibberellin A3 C19H22O6 346.4 0.53 81.59
6466
06 Licoisoflavone C20H18O6 354.4 0.42 41.61 5281789
07 β-Sitosterol C29H50O 414.7 0.75 36.91
222284
5281855
03 Quercetin C15 H10O7302.23 0.28 46.43
Pharmaceutics 2022, 14, x FOR PEER REVIEW 6 of 20
Table 1. Properties of active phytochemicals C. fistula.
Sr.No.PhytochemicalsMolecular
Formula
Molecular
Weight (Dalton)
Drug
Likeness
(>0.18)
Oral
Bioavailability
(>30%)
2D Structures PubChem
ID
01 Rhein C15H8O6 284.22 0.28 47.07 10168
02 Ellagic Acid C14H6O8 302.19 0.43 43.06 5281855
03 Quercetin C15H10O7 302.23 0.28 46.43 5280343
04 Kaempferol C15H10O6 286.24 0.24 41.88
5280863
05 Gibberellin A3 C19H22O6 346.4 0.53 81.59
6466
06 Licoisoflavone C20H18O6 354.4 0.42 41.61 5281789
07 β-Sitosterol C29H50O 414.7 0.75 36.91
222284
5280343
04 Kaempferol C15H10 O6286.24 0.24 41.88
Pharmaceutics 2022, 14, x FOR PEER REVIEW 6 of 20
Table 1. Properties of active phytochemicals C. fistula.
Sr.No.PhytochemicalsMolecular
Formula
Molecular
Weight (Dalton)
Drug
Likeness
(>0.18)
Oral
Bioavailability
(>30%)
2D Structures PubChem
ID
01 Rhein C15H8O6 284.22 0.28 47.07 10168
02 Ellagic Acid C14H6O8 302.19 0.43 43.06 5281855
03 Quercetin C15H10O7 302.23 0.28 46.43 5280343
04 Kaempferol C15H10O6 286.24 0.24 41.88
5280863
05 Gibberellin A3 C19H22O6 346.4 0.53 81.59
6466
06 Licoisoflavone C20H18O6 354.4 0.42 41.61 5281789
07 β-Sitosterol C29H50O 414.7 0.75 36.91
222284
5280863
05 Gibberellin A3 C19H22 O6346.4 0.53 81.59
Pharmaceutics 2022, 14, x FOR PEER REVIEW 6 of 20
Table 1. Properties of active phytochemicals C. fistula.
Sr.No.PhytochemicalsMolecular
Formula
Molecular
Weight (Dalton)
Drug
Likeness
(>0.18)
Oral
Bioavailability
(>30%)
2D Structures PubChem
ID
01 Rhein C15H8O6 284.22 0.28 47.07 10168
02 Ellagic Acid C14H6O8 302.19 0.43 43.06 5281855
03 Quercetin C15H10O7 302.23 0.28 46.43 5280343
04 Kaempferol C15H10O6 286.24 0.24 41.88
5280863
05 Gibberellin A3 C19H22O6 346.4 0.53 81.59
6466
06 Licoisoflavone C20H18O6 354.4 0.42 41.61 5281789
07 β-Sitosterol C29H50O 414.7 0.75 36.91
222284
6466
06 Licoisoflavone C20H18 O6354.4 0.42 41.61
Pharmaceutics 2022, 14, x FOR PEER REVIEW 6 of 20
Table 1. Properties of active phytochemicals C. fistula.
Sr.No.PhytochemicalsMolecular
Formula
Molecular
Weight (Dalton)
Drug
Likeness
(>0.18)
Oral
Bioavailability
(>30%)
2D Structures PubChem
ID
01 Rhein C15H8O6 284.22 0.28 47.07 10168
02 Ellagic Acid C14H6O8 302.19 0.43 43.06 5281855
03 Quercetin C15H10O7 302.23 0.28 46.43 5280343
04 Kaempferol C15H10O6 286.24 0.24 41.88
5280863
05 Gibberellin A3 C19H22O6 346.4 0.53 81.59
6466
06 Licoisoflavone C20H18O6 354.4 0.42 41.61 5281789
07 β-Sitosterol C29H50O 414.7 0.75 36.91
222284
5281789
07 β-Sitosterol C29H50 O 414.7 0.75 36.91
222284
08 Stigmasterol C29H48 O 412.7 0.76 43.83
Pharmaceutics 2022, 14, x FOR PEER REVIEW 7 of 20
08 Stigmasterol C29H48O 412.7 0.76 43.83
5280794
3.2. Drug Target Profiling for C. fistula
In total, 800 target genes of corresponding active phyto-constituents of C. fistula
were collected from the SwissTargetPrediction that showed the bioactivity of active con-
stituents of C. fistula. Protein ID’s of the targets were aligned from UniProtKB to remove
the duplicates. In the end, 415 unique targets are selected for further analysis.
3.3. Potential Targets of C. fistula for EOC
A total of 1077 EOC-related targets (Table S2) were acquired by using DisGeNET &
Genecard databases. These were intersected to the active constituent’s target genes. Out
of those, 98 potential targets (Table S3) were predicted for the treatment of EOC (Figure
2).
Figure 2. Venn diagram of potential targets.
3.4. Compound–Target Network
The compound–target network of eight active constituents to corresponding 98 po-
tential targets was constructed by using Cytoscape software. That network had 106
nodes and 217 edges. Each node represented active constituents or potential targets, and
lines showed interaction between them (Figure 3).
5280794
Pharmaceutics 2022,14, 1970 6 of 17
3.2. Drug Target Profiling for C. fistula
In total, 800 target genes of corresponding active phyto-constituents of C. fistula were
collected from the SwissTargetPrediction that showed the bioactivity of active constituents
of C. fistula. Protein ID’s of the targets were aligned from UniProtKB to remove the
duplicates. In the end, 415 unique targets are selected for further analysis.
3.3. Potential Targets of C. fistula for EOC
A total of 1077 EOC-related targets (Table S2) were acquired by using DisGeNET &
Genecard databases. These were intersected to the active constituent’s target genes. Out of
those, 98 potential targets (Table S3) were predicted for the treatment of EOC (Figure 2).
Pharmaceutics 2022, 14, x FOR PEER REVIEW 7 of 20
08 Stigmasterol C29H48O 412.7 0.76 43.83
5280794
3.2. Drug Target Profiling for C. fistula
In total, 800 target genes of corresponding active phyto-constituents of C. fistula
were collected from the SwissTargetPrediction that showed the bioactivity of active con-
stituents of C. fistula. Protein ID’s of the targets were aligned from UniProtKB to remove
the duplicates. In the end, 415 unique targets are selected for further analysis.
3.3. Potential Targets of C. fistula for EOC
A total of 1077 EOC-related targets (Table S2) were acquired by using DisGeNET &
Genecard databases. These were intersected to the active constituent’s target genes. Out
of those, 98 potential targets (Table S3) were predicted for the treatment of EOC (Figure
2).
Figure 2. Venn diagram of potential targets.
3.4. Compound–Target Network
The compound–target network of eight active constituents to corresponding 98 po-
tential targets was constructed by using Cytoscape software. That network had 106
nodes and 217 edges. Each node represented active constituents or potential targets, and
lines showed interaction between them (Figure 3).
Figure 2. Venn diagram of potential targets.
3.4. Compound–Target Network
The compound–target network of eight active constituents to corresponding 98 poten-
tial targets was constructed by using Cytoscape software. That network had 106 nodes and
217 edges. Each node represented active constituents or potential targets, and lines showed
interaction between them (Figure 3).
Topological analysis of network revealed the network characteristics: the density is
0.039, network centralization is 0.271, network heterogeneity is 1.668, and characteristic
path length is 3.042. Furthermore, the active constituents with respective degrees were
found as: Rhein (32), Ellagic Acid (32), Quercetin (30), Kaempferol (29), Gibberellin A3 (28),
Licoisoflavone (26),
β
-Sitosterol (21), and Stigmasterol (19). They interacted with multiple
targets (Table 2).
Table 2. Degrees of 8 active constituents analyzed by the Cytoscape.
Sr. No. Phytochemical Name Categories Degree
01 Rhein Anthraquinone 32
02 Ellagic Acid Polyphenol 32
03 Quercetin Flavonoid 30
04 Kaempferol Flavonoid 29
05 Gibberellin A3 Hormone 28
06 Licoisoflavone Flavonoid 26
07 ß-Sitosterol Phytosterols 21
08 Stigmasterol Phytosterols 19
Pharmaceutics 2022,14, 1970 7 of 17
Pharmaceutics 2022, 14, x FOR PEER REVIEW 8 of 20
Figure 3. Compound–target network of active constituents and potential targets (circle shape
shows active constituents and diamond shape shows potential targets).
Topological analysis of network revealed the network characteristics: the density is
0.039, network centralization is 0.271, network heterogeneity is 1.668, and characteristic
path length is 3.042. Furthermore, the active constituents with respective degrees were
found as: Rhein (32), Ellagic Acid (32), Quercetin (30), Kaempferol (29), Gibberellin A3
(28), Licoisoflavone (26), β-Sitosterol (21), and Stigmasterol (19). They interacted with
multiple targets (Table 2).
Table 2. Degrees of 8 active constituents analyzed by the Cytoscape.
Sr. No. Phytochemical Name Categories Degree
01 Rhein Anthraquinone 32
02 Ellagic Acid Polyphenol 32
03 Quercetin Flavonoid 30
04 Kaempferol Flavonoid 29
05 Gibberellin A3 Hormone 28
06 Licoisoflavone Flavonoid 26
Figure 3.
Compound–target network of active constituents and potential targets (circle shape shows
active constituents and diamond shape shows potential targets).
3.5. Protein–Protein Interaction (PPI) Network
The 98 potential target genes of C. fistula which may be the potential targeting genes
to treat EOC were copied to STRING v11.5to build a PPI network with a score of 0.4
confidence interaction. That was performed to predict the interactions of potential proteins
with Homo sapiens proteins and their physiological functions. In that network, the nodes
are representing the targets, and the lines connecting the nodes are edges that represent
the intermolecular interactions between multiple targets during a disease development
(Figure S1). There were 98 nodes and 1153 edges in the network. In addition, the density,
heterogeneity, network centralization, and path length were 0.243, 0.721, 0.521, and 1.862,
respectively (Figure 4A). Later, the “Network Analyzer” tool was used to examine the
PPI network. The five highest degree targets were AKT1 (73), ALB (65), CTNNB1 (64),
ESR1 (64), and CASP3 (62). The higher degree depicted that the target genes are extremely
connected; hence, these five genes might be important targets (Figure 4B). When the data
were compared to the data obtained from enrichment analysis of 98 potential targets,
specifically four out of those five genes, AKT1, CTNNB1, ESR1, and CASP3, were identified
as the principal anticancerous targets of C. fistula.
Pharmaceutics 2022,14, 1970 8 of 17
Pharmaceutics 2022, 14, x FOR PEER REVIEW 9 of 20
07 ß-Sitosterol Phytosterols 21
08 Stigmasterol Phytosterols 19
3.5. ProteinProtein Interaction (PPI) Network
The 98 potential target genes of C. fistula which may be the potential targeting genes
to treat EOC were copied to STRING v11.5to build a PPI network with a score of 0.4 con-
fidence interaction. That was performed to predict the interactions of potential proteins
with Homo sapiens proteins and their physiological functions. In that network, the nodes
are representing the targets, and the lines connecting the nodes are edges that represent
the intermolecular interactions between multiple targets during a disease development
(Figure S1). There were 98 nodes and 1153 edges in the network. In addition, the density,
heterogeneity, network centralization, and path length were 0.243, 0.721, 0.521, and
1.862, respectively (Figure 4A). Later, the “Network Analyzer” tool was used to examine
the PPI network. The five highest degree targets were AKT1 (73), ALB (65), CTNNB1
(64), ESR1 (64), and CASP3 (62). The higher degree depicted that the target genes are ex-
tremely connected; hence, these five genes might be important targets (Figure 4B). When
the data were compared to the data obtained from enrichment analysis of 98 potential
targets, specifically four out of those five genes, AKT1, CTNNB1, ESR1, and CASP3,
were identified as the principal anticancerous targets of C. fistula.
(A)
Pharmaceutics 2022, 14, x FOR PEER REVIEW 10 of 20
(B)
Figure 4. (A) Analysis and Visualization of PPI network in Cytoscape. (B) Top 10 targets of C. fis-
tula on EOC analyzed by Cytoscape.
3.6. Gene Functional Annotation
The gene and enriched pathways were analyzed through DAVID to predict gene
role and signaling pathways of eight active constituents of C. fistula for the EOC treat-
ment. The GO enrichment analysis contained 324 Biological Processes (BPs), 51 Cellular
Components (CCs) and 85 Molecular Functions (MFs) as conformed screening criteria,
with count 2 and p 0.05. Moreover, 133 enriched pathways were identified on the ba-
sis of criteria p < 0.05. A pathwaytarget network was constructed by using Cytoscape
(Figure 5). GO annotations and KEGG pathway were plotted by using ggplot2 in R lan-
guage (Figures 6 and 7).
Figure 4.
(
A
) Analysis and Visualization of PPI network in Cytoscape. (
B
) Top 10 targets of C. fistula
on EOC analyzed by Cytoscape.
3.6. Gene Functional Annotation
The gene and enriched pathways were analyzed through DAVID to predict gene role
and signaling pathways of eight active constituents of C. fistula for the EOC treatment. The
GO enrichment analysis contained 324 Biological Processes (BPs), 51 Cellular Components
(CCs) and 85 Molecular Functions (MFs) as conformed screening criteria, with count
2
and p
0.05. Moreover, 133 enriched pathways were identified on the basis of criteria
p< 0.05. A pathway–target network was constructed by using Cytoscape (Figure 5). GO
Pharmaceutics 2022,14, 1970 9 of 17
annotations and KEGG pathway were plotted by using ggplot2 in R language (Figures 6
and 7).
Pharmaceutics 2022, 14, x FOR PEER REVIEW 11 of 20
Figure 5. Hub genes enriched in top 20 signaling pathways. Signaling pathways are represented as
hexagons and targets are represented as ellipses.
(A) (B)
Figure 5.
Hub genes enriched in top 20 signaling pathways. Signaling pathways are represented as
hexagons and targets are represented as ellipses.
Pharmaceutics 2022, 14, x FOR PEER REVIEW 11 of 20
Figure 5. Hub genes enriched in top 20 signaling pathways. Signaling pathways are represented as
hexagons and targets are represented as ellipses.
(A) (B)
Figure 6. Cont.
Pharmaceutics 2022,14, 1970 10 of 17
Pharmaceutics 2022, 14, x FOR PEER REVIEW 12 of 20
(C)
Figure 6. GO analysis of C. fistula’s potential targets on EOC. (A) Biological Processes (BP), (B) Cel-
lular Components (CC), and (C) Molecular Functions (MF).
Figure 6.
GO analysis of C. fistula’s potential targets on EOC. (
A
) Biological Processes (BP), (
B
)
Cellular Components (CC), and (C) Molecular Functions (MF).
Pharmaceutics 2022, 14, x FOR PEER REVIEW 12 of 20
(C)
Figure 6. GO analysis of C. fistula’s potential targets on EOC. (A) Biological Processes (BP), (B) Cel-
lular Components (CC), and (C) Molecular Functions (MF).
Figure 7. KEGG pathway enrichment analysis: 20 enriched pathways plotted through R language.
Pharmaceutics 2022,14, 1970 11 of 17
3.7. Compound–Target–Signaling Pathway Network
A compound–target–signaling pathways network was built by integration of compound–
target and pathway–target networks using the Cytoscape. The “Network Analyzer” anal-
ysis showed that it included 126 nodes, 570 edges, 8 active phytochemicals, 98 potential
targets and 20 associated pathways. The targets of active phytochemicals were intercon-
nected with pathways (Figure 8).
Pharmaceutics 2022, 14, x FOR PEER REVIEW 13 of 20
Figure 7. KEGG pathway enrichment analysis: 20 enriched pathways plotted through R language.
3.7. Compound–TargetSignaling Pathway Network
A compound–targetsignaling pathways network was built by integration of com-
pound–target and pathway–target networks using the Cytoscape. The “Network Ana-
lyzer” analysis showed that it included 126 nodes, 570 edges, 8 active phytochemicals, 98
potential targets and 20 associated pathways. The targets of active phytochemicals were
interconnected with pathways (Figure 8).
Figure 8. C. fistula’s compound–target–signaling pathway network to EOC. Candidate active phy-
tochemicals shown as ellipses, potential targets represented as diamonds, and pathways repre-
sented as hexagons.
3.8. Molecular Docking
Molecular docking was used to screen potential targets of components with the ca-
pacity to decrease the prevalence of EOC. The highest four target genes, AKT1,
CTNNB1, ESR1, and CASP3 were selected via the topological examination of PPI net-
work. The three-dimensional (3D) structures of these target proteins (AKT1 (PDB id:
3QKK), CTNNB1 (PDB id: 1jdh), ESR1 (PDB id: 1pcg), CASP3 (PDB id: 3kjf) were re-
trieved through the PDB. These structures were refined by the UCSF Chimera tool with
Figure 8.
C. fistula’s compound–target–signaling pathway network to EOC. Candidate active phyto-
chemicals shown as ellipses, potential targets represented as diamonds, and pathways represented as
hexagons.
3.8. Molecular Docking
Molecular docking was used to screen potential targets of components with the
capacity to decrease the prevalence of EOC. The highest four target genes, AKT1, CTNNB1,
ESR1, and CASP3 were selected via the topological examination of PPI network. The
three-dimensional (3D) structures of these target proteins (AKT1 (PDB id: 3QKK), CTNNB1
(PDB id: 1jdh), ESR1 (PDB id: 1pcg), CASP3 (PDB id: 3kjf) were retrieved through the PDB.
These structures were refined by the UCSF Chimera tool with 1000 decent steps of energy
minimization [
46
]. In order to avoid collisions and erroneous compositions, non-standard
residues were eliminated from the proteins. The docking analysis was used to accurately
predict the significant binding affinity between active constituents and four target protein’s’
binding pockets. The eight active components of C. fistula were docked with the four EOC
potential targets (Figure 9). All compounds and targets showed binding scores ranging
from
5.8 to
9.2 kcal/mole. According to the docking studies, eight active constituents
reported greater binding energy with key EOC targets. Among those, more specifically,
two phytochemicals, β- Sitosterol and Stigmasterol, were prominent (Table 3).
Pharmaceutics 2022,14, 1970 12 of 17
Pharmaceutics 2022, 14, x FOR PEER REVIEW 14 of 20
1000 decent steps of energy minimization [46]. In order to avoid collisions and erroneous
compositions, non-standard residues were eliminated from the proteins. The docking
analysis was used to accurately predict the significant binding affinity between active
constituents and four target protein’s’ binding pockets. The eight active components of
C. fistula were docked with the four EOC potential targets (Figure 9). All compounds
and targets showed binding scores ranging from 5.8 to 9.2 kcal/mole. According to the
docking studies, eight active constituents reported greater binding energy with key EOC
targets. Among those, more specifically, two phytochemicals, β- Sitosterol and Stigmas-
terol, were prominent (Table 3).
Table 3. The binding affinities of prospective target genes with phytochemicals in the docking
analysis.
Sr. No Compound Binding Affinities (kcal/mol)
AKT1 CTNNB1 ESR1 CASP3
01 Rhein 7.5 6.9 6.8 6.2
02 Ellagic acid 7.9 7.2 6.6 6.4
03 Quercetin 7.7 6.5 7.7 6.1
04 Kaempferol 7.6 6.7 7.3 5.8
05 Gibberelin A3 8.2 6.7 6.6 6.7
06 Licoisoflavone 7.9 6.1 6.6 6.5
07 β- Sitosterol 8.8 7.0 6.7 6.8
08 Stigmasterol 9.2 6.7 7.0 6.8
(A)
Pharmaceutics 2022, 14, x FOR PEER REVIEW 15 of 20
(B)
(C)
(D)
Figure 9.
The binding site residues with the four proteins are shown in the docking complex of four
targets with their best binding components: (A) AKT1; (B) CTNNB1; (C) ESR1; (D) CASP3.
Pharmaceutics 2022,14, 1970 13 of 17
Table 3.
The binding affinities of prospective target genes with phytochemicals in the docking
analysis.
Sr. No Compound Binding Affinities (kcal/mol)
AKT1 CTNNB1 ESR1 CASP3
01 Rhein 7.5 6.9 6.8 6.2
02 Ellagic acid 7.9 7.2 6.6 6.4
03 Quercetin 7.7 6.5 7.7 6.1
04 Kaempferol 7.6 6.7 7.3 5.8
05 Gibberelin
A3 8.2 6.7 6.6 6.7
06
Licoisoflavone
7.9 6.1 6.6 6.5
07 β- Sitosterol 8.8 7.0 6.7 6.8
08 Stigmasterol 9.2 6.7 7.0 6.8
4. Discussion
The etiology and pathogenesis of EOC is very complex and not yet fully understood.
It is characterized by sneaking symptoms. That is why individuals are examined at a
later stage. Cyto-reduction surgeries (CRS) are used to treat this, followed by platinum-
based chemotherapy [
1
]. These types of medications incorporate adverse side effects on
the individual’s well-being [
2
]. Owing to the progress in the treatment of OC, targeted
medicines are available now. Nevertheless, targeted/focused medications are expensive
and the options are frequently restricted. Therefore, more effective, safe, and affordable
therapeutic medications are urgently needed for the OC.
Natural phytochemicals, especially plant-derived compounds, are widely employed
as alternative therapeutics, such as taxol for cancers [
47
]. The reason is that they are
inexpensive, more accessible, multi-targeting, and contain low toxicity. Herbal medicines
have been utilized in order to cure human illness since ancient times and are thought to be
the rich origin of drug discovery. C. fistula is a versatile plant that has shown the antioxidant,
anti-inflammatory, immunological, hepatoprotective, antipyretic, analgesic, and antitumor
properties [
30
]. More than 60 phytochemicals, including flavonoids, anthraquinones,
chromones, coumarins, alkaloids, phytosterols, long-chain hydrocarbons, phenolic, and
other phytochemicals have been found in C. fistula.
This study has provided a baseline for the screening of C. fistula’s bioactive compounds.
That was a unique therapeutic idea for future research of C. fistula’s processes for EOC
treatment. Anthraquinone, polyphenols, hormones, phytosterols, and flavonoids were the
most abundant bioactive chemicals discovered in C. fistula. These played a critical role in
the development of EOC. This is a strong indicator that a number of targets may work
together to provide a synergistic impact.
By PPI network topological analysis, the four key targets AKT1, CTNNB1, ESR1, and
CASP3 were evaluated for anti-EOC activity. This is a strong indicator that a number of
targets may work together to provide a synergistic impact. AKT1 promotes OC cell prolif-
eration, immigration, epithelial transformation to mesenchyme (EMT), gluconeogenesis,
and resistance to therapeutics and serves as a biomarker for OC therapy response [4851].
CTNNB1 is involved in the growth of OC due to its deregulated impact [
52
54
]. ESR1 is
implicated in amplification and penetration, and its hereditary characteristics are linked to
OC probability and advancement [
55
57
]. CASP3 activity and expression are linked to cell
death, immigration, amplification, susceptibility of OC cells to anticarcinogenic drugs, as
well as metastasis and prognostic outcomes in OC patients [5558].
Gene enrichment analysis reported that anti-EOC target genes of C. fistula were pri-
marily involved in different molecular processes such as protein serine activity, protein
serine/threonine kinase activity, transmembrane receptor protein tyrosine kinase activity,
Pharmaceutics 2022,14, 1970 14 of 17
and transmembrane receptor protein kinase activity. The pathway enrichment analysis
evaluate that genes were focused to EOC-associated signaling pathways: the ‘PI3K-Akt
signaling pathway, proteoglycans in cancer, the TNF signaling pathway, and the IL-17
signaling pathway. The PI3K-Akt signaling pathway might boost tumor progression,
amplification, and uncontrolled mitosis OC cells [
56
,
57
,
59
]. Proteoglycans in cancer mod-
ulate adhesion and migration are also associated to mutagenic and angiogenic growth
factors [
60
]. TNF signaling pathways promote unregulated inflammation, which can be sup-
pressed with medication, and their expression profiles are linked to OC cancer [
61
,
62
]. The
pro-inflammatory cytokine interleukin-17 (IL-17) has been linked to low tumor grade [
63
].
Through the network analysis, the four main targets AKT1, CTNNB1, ESR1, and
CASP3 were evaluated for their binding energies to eight active components of C. fistula. It
exemplifies the expression of multiple targets, components, and pathways. In molecular
docking, more negative the binding energy, the greater the expected affinity for binding
of the ligand to the target [
64
]. The
β
-Sitosterol showed lower binding affinities to AKT1
and CTNNB1. Stigmasterol interacted to AKT1 and ESR1 with lower binding affinities.
This revealed that C. fistula has anti-EOC properties, inhibiting EOC main targeting genes.
Network pharmacology integrated to molecular docking approach appeared to be effective
for the identification of biologically active phytochemicals, associated potential genes, and
linked signaling pathways to treat EOC. It provided a systematic framework for subsequent
investigation of phytochemicals against a wide range of disorders.
5. Conclusions
This research set the foundation to establish the efficacy of multicomponent, multi-
target chemical formulae and identification of genes that target to treat EOC. Network
pharmacology and molecular docking approaches were incorporated in order to reveal
the underlying mechanisms of C. fistula to treat EOC. Furthermore, our data suggest that
the AKT1, CTNNB1, ESR1, and CASP3 targets are intriguingly beneficial to slow down
the prevalence of EOC, potentially resulting in therapeutic benefits in EOC. This is the
first time four genes have been reported against the EOC from C. fistula. However, more
pharmacological and clinical studies are required to confirm our findings. This method
lays the framework for future research into C. fistula for EOC-protective mechanisms and
the use of network pharmacology in drug development.
Supplementary Materials:
The following supporting information can be downloaded at: https:
//www.mdpi.com/article/10.3390/pharmaceutics14091970/s1. Figure S1: PPI network constructed
by STRING. It represents the interaction of potential genes of C. fistula against EOC. Table S1:
Phytochemical library of C. fistula. Table S2: The unique genes and variants of EOC fetched from
DisGeNET & Genecards databases. Table S3: Potential targets of C. fistula for EOC.
Author Contributions:
Conceptualization, U.A.A. and I.R.; methodology, H.N.; software, U.A.A.;
validation, F.A., H.N. and A.K.M.H.K.; formal analysis, A.K. and F.A.; investigation, A.K.; resources,
R.M.A.; data curation, A.K.; writing—original draft preparation, A.K. and R.M.A.; writing—review
and editing, I.R. and M.S.R.R.; visualization, R.M.A.; supervision, I.R.; project administration, I.R. All
authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: Not applicable.
Acknowledgments:
We are thankful to Department of Bioinformatics and Biotechnology, Govern-
ment College University, Faisalabad, Pakistan for providing computational facilities for successful
completion of this project. Another acknowledgement is due to Aamer Shaheen, Department of
English Literature, Government College University Faisalabad, Pakistan. He helped us in correcting
the English language and grammatical errors. Moreover, Najma Hameed and Hira Saleem deserve
special thanks for their assistance during formal analysis and docking.
Pharmaceutics 2022,14, 1970 15 of 17
Conflicts of Interest:
The funders had no role in the design of the study; in the collection, analyses,
or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.
References
1.
DeSantis, C.E.; Miller, K.D.; Dale, W.; Mohile, S.G.; Cohen, H.J.; Leach, C.R.; Sauer, A.G.; Jemal, A.; Siegel, R.L. Cancer statistics
for adults aged 85 years and older, 2019. CA Cancer J. Clin. 2019,69, 452–467. [CrossRef] [PubMed]
2.
Oronsky, B.; Ray, C.M.; Spira, A.I.; Trepel, J.B.; Carter, C.A.; Cottrill, H.M. A brief review of the management of platinum-resistant–
platinum-refractory ovarian cancer. Med. Oncol. 2017,34, 103. [CrossRef] [PubMed]
3.
Kurman, R.J.; Shih, I.M. The origin and pathogenesis of epithelial ovarian cancer-a proposed unifying theory. Am. J. Surg. Pathol.
2010,34, 433–443. [CrossRef]
4.
Desai, A.; Xu, J.; Aysola, K.; Qin, Y.; Okoli, C.; Hariprasad, R.; Chinemerem, U.; Gates, C.; Reddy, A.; Danner, O.; et al. Epithelial
ovarian cancer: An overview. World J. Transl. Med. 2014,3, 1–8. [CrossRef] [PubMed]
5.
Casagrande, J.; Louie, E.; Pike, M.; Roy, S.; Ross, R.J.L.K.; Henderson, B.E. Incessant ovulation and ovarian cancer. Lancet
1979
,28,
170–173. [CrossRef]
6. Fathalla, M.J.L. Incessant ovulation—A factor in ovarian neoplasia. Lancet 1971,2, 163. [CrossRef]
7.
Cramer, D.W.; Wrjjotnci, W. Determinants of ovarian cancer risk. II. Inferences regarding pathogenesis. J. Natl. Cancer Inst.
1983
,
71, 717–721.
8.
Cramer, D.; Barbieri, R.; Fraer, A.; Harlow, B. Determinants of early follicular phase gonadotrophin and estradiol concentrations
in women of late reproductive age. Hum. Reprod. 2002,17, 221–227. [CrossRef]
9.
Rahmani, A.H.; Smjajpcr, A. Nigella sativa and its active constituents thymoquinone shows pivotal role in the diseases prevention
and treatment. Asian J. Pharm. Clin. Res. 2015,8, 48–53.
10.
Rahmani, A.H.; Aly, S.M.; Ali, H.; Babiker, A.Y.; Srikar, S.; Khan, A.A. Therapeutic effects of date fruits (Phoenix dactylifera) in the
prevention of diseases via modulation of anti-inflammatory, anti-oxidant and anti-tumour activity. Int. J. Clin. Exp. Med.
2014
,7,
483–491.
11.
Rahmani, A.; Pathophysiology, P. Active ingredients of ginger as potential candidates in the prevention and treatment of diseases
via modulation of biological activities. Int. J. Phys. Pathophysiol. Pharm. 2014,6, 125–136.
12. Hopkins, A. Network pharmacology. Nat. Biotechnol. 2007,25, 1110–1111. [CrossRef] [PubMed]
13.
Zhang, B.; Wang, X.; Li, S. An integrative platform of TCM network pharmacology and its application on a herbal formula,
Qing-Luo-Yin. Altern. Med. 2013,2013, 456747. [CrossRef] [PubMed]
14. Berger, S.; Iyengar, R. Network analyses in systems pharmacology. Bioinformatics 2009,25, 2466–2472. [CrossRef] [PubMed]
15.
Kibble, M.; Saarinen, N.; Tang, J.; Wennerberg, K.; Mäkelä, S.; Aittokallio, T. Network pharmacology applications to map the
unexplored target space and therapeutic potential of natural products. Nat. Prod. Rep.
2015
,32, 1249–1266. [CrossRef] [PubMed]
16.
Rajesh, Y.; Jain, C.G. Effect of Petroleum ether extract of Cassia fistula Seeds on Uterine Histoarchitecture of Ovariectomized
Female Rats. Indian J. Fundam. Appl. Life Sci. 2009,1, 438–444.
17.
Yadava, R.; Verma, V. A new biologically active flavone glycoside from the seeds of Cassia fistula (Linn.). J. Asian Nat. Prod. Res.
2003,5, 57–61. [CrossRef]
18.
Bhakta, T.; Mukherjee, P.; Saha, K.; Pal, M.; Saha, B. Hypoglycemic activity of Cassia fistula Linn.(Leguminosae) leaf (Methanol
extract) in alloxan-induced diabetic rats. Indian J. Pharm. Sci. 1997,9, 35–38.
19.
Sangita, D.; Sarkar, P.; Sengupta, A.; Chattopadhyay, A. Clinical study of Aragvadha (Cassia fistula Linn) on Vicharchika (Eczema).
Ayu 2008,39, 9–15.
20.
Karthikeyan, S.; Gobianand, K. Antiulcer activity of ethanol leaf extract of Cassia fistula.Pharm. Biol.
2010
,48, 869–877. [CrossRef]
21.
Silawat, N.; Jarald, E.E.; Jain, N.; Yadav, A.; Deshmukh, P. The mechanism of hypoglycemic and antidiabetic action of hydroalcholic
extract of Cassia fistula Linn. in rats. Biol. Chem. 2009,1, 82–92.
22.
Singh, K.; Bharadwaj, U. Hypoglycaemic activity of Albizzia stipulata, Albizzia moluccana and Cassia fistula leguminous seed
diets on normal young rats. Indian J. Pharm. Sci. 1975,7, 47–49.
23.
Manonmani, G.; Bhavapriya, V.; Kalpana, S.; Govindasamy, S.; Apparanantham, T. Antioxidant activity of Cassia fistula (Linn.)
flowers in alloxan induced diabetic rats. J. Ethnopharmacol. 2005,97, 39–42. [CrossRef] [PubMed]
24.
Duraipandiyan, V.; Ignacimuthu, S. Antibacterial and antifungal activity of Cassia fistula L.: An ethnomedicinal plant. J.
Ethnopharmacol. 2007,112, 590–594. [CrossRef]
25.
Bhatnagar, M.; Vimal, S.; Vyas, Y.; Sharma, D.; Sharma, K. Antioxidant activity of fruit pulp powder of Cassia fistula.Pharmacogn.
J. 2010,2, 219–228. [CrossRef]
26.
Abo, K.; Lasaki, S.; Adeyemi, A. Laxative and antimicrobial properties of Cassia species growing in Ibadan. Niger. J. Nat. Prod.
Med. 1999,3, 47–50. [CrossRef]
27.
Dutta, A.; Bratati, D. Seasonal variation in the content of sennosides and rhein in leaves and pods of Cassia fistula.Pharm. Sci.
1998,60, 388.
28.
Aminah, N.; Tun, K.; Kristanti, A.; Aung, H.; Takaya, Y.; Choudhary, M. Chemical constituents and their biological activities from
Taunggyi (Shan state) medicinal plants. Heliyon 2021,7, e06173. [CrossRef]
29. Ali, M. Cassia fistula Linn: A review of phytochemical and pharmacological studies. Int. J. Pharm. Sci. Res. 2014,5, 2125–2130.
Pharmaceutics 2022,14, 1970 16 of 17
30.
Mwangi, R.W.; Macharia, J.M.; Wagara, I.N.; Bence, R.L. The medicinal properties of Cassia fistula L.: A review. Biomed.
Pharmacother. 2021,144, 112240. [CrossRef]
31.
Wijaya, S.H.; Tanaka, Y.; Amin, A.U.; Morita, A.H.; Afendi, F.M.; Batubara, I.; Ono, N.; Darusman, L.K.; Kanaya, S. Utilization of
KNApSAcK family databases for developing herbal medicine systems. J. Comput. Aided Chem. 2016,17, 1–7. [CrossRef]
32.
Li, J.; Huang, Y.; Zhao, S.; Guo, Q.; Zhou, J.; Han, W.; Xu, Y. Based on network pharmacology to explore the molecular mechanisms
of Astragalus membranaceus for treating T2 diabetes mellitus. Ann. Transl. Med. 2019,7, 633. [CrossRef] [PubMed]
33.
Apweiler, R.; Bairoch, A.; Wu, C.H.; Barker, W.C.; Boeckmann, B.; Ferro, S.; Gasteiger, E.; Huang, H.; Lopez, R.; Magrane, M.; et al.
UniProt: The universal protein knowledgebase. Nucleic Acids Res. 2018,46, 2699. [CrossRef]
34.
Piñero, J.; Ramírez-Anguita, J.M.; Saüch-Pitarch, J.; Ronzano, F.; Centeno, E.; Sanz, F.; Furlong, L.I. The DisGeNET knowledge
platform for disease genomics: 2019 update. Nucleic Acids Res. 2020,48, D845–D855. [CrossRef] [PubMed]
35.
Fishilevich, S.; Nudel, R.; Rappaport, N.; Hadar, R.; Plaschkes, I.; Stein, I.T.; Rosen, N.; Kohn, A.; Twik, M.; Safran, M.; et al.
GeneHancer: Genome-wide integration of enhancers and target genes in GeneCards. Database 2017,2017, bax028. [CrossRef]
36.
Bardou, P.; Mariette, J.; Escudié, F.; Djemiel, C.; Klopp, C. Jvenn: An interactive Venn diagram viewer. BMC Bioinform.
2014
,15,
1–7. [CrossRef]
37.
Shannon, P.; Markiel, A.; Ozier, O.; Baliga, N.S.; Wang, J.T.; Ramage, D.; Amin, N.; Schwikowski, B.; Ideker, T. Cytoscape: A
software environment for integrated models of biomolecular interaction networks. Genome Res. 2003,13, 2498–2504. [CrossRef]
38.
De Jong, H.; Geiselmann, J.; Hernandez, C.; Page, M. Genetic Network Analyzer: Qualitative simulation of genetic regulatory
networks. Bioinformatics 2003,19, 336–344. [CrossRef]
39.
Li, H.; Hung, A.; Yang, A.W.H. Herb-target virtual screening and network pharmacology for prediction of molecular mechanism
of Danggui Beimu Kushen Wan for prostate cancer. Sci. Rep. 2021,11, 6656. [CrossRef]
40.
Szklarczyk, D.; Gable, A.L.; Lyon, D.; Junge, A.; Wyder, S.; Huerta-Cepas, J.; Simonovic, M.; Doncheva, N.T.; Morris, J.H.;
Bork, P.; et al. STRING v11: Protein–protein association networks with increased coverage, supporting functional discovery in
genome-wide experimental datasets. Nucleic Acids Res. 2019,47, D607–D613. [CrossRef]
41.
Lu, X.; Zheng, Y.; Wen, F.; Huang, W.; Chen, X.; Ruan, S.; Gu, S.; Hu, Y.; Teng, Y.; Shu, P. Study of the active ingredients and
mechanism of Sparganii rhizoma in gastric cancer based on HPLC-Q-TOF–MS/MS and network pharmacology. Sci. Rep.
2021
,
11, 1905. [CrossRef] [PubMed]
42.
Kanehisa, M.; Sato, Y.; Furumichi, M.; Morishima, K.; Tanabe, M. New approach for understanding genome variations in KEGG.
Nucleic Acids Res. 2019,47, D590–D595. [CrossRef] [PubMed]
43.
Kim, S.; Chen, J.; Cheng, T.; Gindulyte, A.; He, J.; He, S.; Li, Q.; Shoemaker, B.A.; Thiessen, P.A.; Yu, B.; et al. PubChem 2019
update: Improved access to chemical data. Nucleic Acids Res. 2019,47, D1102–D1109. [CrossRef] [PubMed]
44.
Bittrich, S.; Rose, Y.; Segura, J.; Lowe, R.; Westbrook, J.D.; Duarte, J.M.; Burley, S.K. RCSB Protein Data Bank: Improved annotation,
search and visualization of membrane protein structures archived in the PDB. Bioinformatics 2022,38, 1452–1454. [CrossRef]
45.
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. 2021,61, 3891–3898. [CrossRef]
46.
Mirzaei, H.; Zarbafian, S.; Villar, E.; Mottarella, S.; Beglov, D.; Vajda, S.; Paschalidis, I.C.; Vakili, P.; Kozakov, D. Energy
minimization on manifolds for docking flexible molecules. J. Chem. Theory Comput. 2015,11, 1063–1076. [CrossRef]
47.
Nobili, S.; Lippi, D.; Witort, E.; Donnini, M.; Bausi, L.; Mini, E.; Capaccioli, S. Natural compounds for cancer treatment and
prevention. Pharmacol. Res. 2009,59, 365–378. [CrossRef]
48.
Chen, L.; Cheng, X.; Tu, W.; Qi, Z.; Li, H.; Liu, F.; Yang, F.; Zhang, Z.; Wang, Z. Apatinib inhibits glycolysis by suppressing the
VEGFR2/AKT1/SOX5/GLUT4 signaling pathway in ovarian cancer cells. Cell Oncol. 2019,42, 679–690. [CrossRef]
49.
Etemadmoghadam, D.; Bowtell, D. AKT1 gene amplification as a biomarker of treatment response in ovarian cancer: Mounting
evidence of a therapeutic target. Gynecol. Oncol. 2014,135, 409–410. [CrossRef]
50.
Hua, G.; He, C.; Lv, X.; Fan, L.; Wang, C.; Remmenga, S.W.; Rodabaugh, K.J.; Yang, L.; Lele, S.M.; Yang, P.; et al. The four and a
half LIM domains 2 (FHL2) regulates ovarian granulosa cell tumor progression via controlling AKT1 transcription. Cell Death Dis.
2016,7, e2297. [CrossRef]
51.
Chen, Q.; He, Q.; Zhuang, L.; Wang, K.; Yin, C.; He, L. IP10-CDR3 Reduces the Viability and Induces the Apoptosis of Ovarian
Cancer Cells by Down-Regulating the Expression of Bcl-2 and Caspase 3. Onco Targets Ther.
2019
,12, 9697. [CrossRef] [PubMed]
52.
Gregoraszczuk, E.; Rak-Mardyla, A.; Rys, J.; Jakubowicz, J.; Urba´nski, K. Effect of chemotherapeutic drugs on caspase-3 activity,
as a key biomarker for apoptosis in ovarian tumor cell cultured as monolayer. A pilot study. Iran. J. Pharm. Res.
2015
,14,
1153–1161. [PubMed]
53.
Kleinberg, L.; Dong, H.P.; Holth, A.; Risberg, B.; Trope, C.G.; Nesland, J.M.; Flørenes, V.A.; Davidson, B. Cleaved caspase-3 and
nuclear factor-
κ
B p65 are prognostic factors in metastatic serous ovarian carcinoma. Hum. Pathol.
2009
,40, 795–806. [CrossRef]
[PubMed]
54.
Yang, X.; Zheng, F.; Xing, H.; Gao, Q.; Wei, W.; Lu, Y.; Wang, S.; Zhou, J.; Hu, W.; Ma, D. Resistance to chemotherapy-induced
apoptosis via decreased caspase-3 activity and overexpression of antiapoptotic proteins in ovarian cancer. J. Cancer Res. Clin.
Oncol. 2004,130, 423–428. [CrossRef]
55.
Zhao, X.; Wang, D.; Zhao, Z.; Xiao, Y.; Sengupta, S.; Xiao, Y.; Zhang, R.; Lauber, K.; Wesselborg, S.; Feng, L.; et al. Caspase-3-
dependent activation of calcium-independent phospholipase A2 enhances cell migration in non-apoptotic ovarian cancer cells. J.
Biol. Chem. 2006,281, 29357–29368. [CrossRef]
Pharmaceutics 2022,14, 1970 17 of 17
56.
Zhou, C.; Qiu, L.; Sun, Y.; Healey, S.; Wanebo, H.; Kouttab, N.; Di, W.; Yan, B.; Wan, Y. Inhibition of EGFR/PI3K/AKT cell
survival pathway promotes TSA’s effect on cell death and migration in human ovarian cancer cells. Int. J. Oncol.
2006
,29, 269–278.
[CrossRef]
57.
Sheppard, K.E.; Cullinane, C.; Hannan, K.M.; Wall, M.; Chan, J.; Barber, F.; Foo, J.; Cameron, D.; Neilsen, A.; Ng, P.; et al.
Synergistic inhibition of ovarian cancer cell growth by combining selective PI3K/mTOR and RAS/ERK pathway inhibitors. Eur.
J. Cancer 2013,49, 3936–3944. [CrossRef]
58.
Zhang, X.; Yang, X.; Chen, M.; Zheng, S.; Li, J.; Lin, S.; Wang, X. ST3Gal3 confers paclitaxel—mediated chemoresistance in ovarian
cancer cells by attenuating caspase-8/3 signaling. Mol. Med. Rep. 2019,20, 4499–4506. [CrossRef]
59.
Ediriweera, M.K.; Tennekoon, K.H.; Samarakoon, S.R. Role of the PI3K/AKT/mTOR signaling pathway in ovarian cancer:
Biological and therapeutic significance. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2019.
60.
Sanderson, R.D. Heparan sulfate proteoglycans in invasion and metastasis. In Seminars in Cell & Developmental Biology; Elsevier:
Amsterdam, The Netherlands, 2001.
61.
Gupta, M.; Babic, A.; Beck, A.H.; Terry, K. TNF-
α
expression, risk factors, and inflammatory exposures in ovarian cancer: Evidence
for an inflammatory pathway of ovarian carcinogenesis? Hum. Pathol. 2016,54, 82–91. [CrossRef]
62.
Thaklaewphan, P.; Ruttanapattanakul, J.; Monkaew, S.; Buatoom, M.; Sookkhee, S.; Nimlamool, W.; Potikanond, S. Kaempferia
parviflora extract inhibits TNF-
α
-induced release of MCP-1 in ovarian cancer cells through the suppression of NF-
κ
B signaling.
Biomed. Pharmacother. 2021,141, 111911. [CrossRef]
63.
Girondel, C.; Lévesque, K.; Langlois, M.-J.; Pasquin, S.; Saba-El-Leil, M.K.; Rivard, N.; Friesel, R.; Servant, M.J.; Gauchat, J.-F.;
Lesage, S.; et al. Loss of interleukin-17 receptor D promotes chronic inflammation-associated tumorigenesis. Oncogene
2021
,40,
452–464. [CrossRef] [PubMed]
64.
Trott, O.; Olson, A. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient
optimization, and multithreading. J. Comput. Chem. 2010,31, 455–461. [CrossRef] [PubMed]
... The result obtained indicated that SE induce apoptosis through EGEF protein inhibition. Compared with a previous study (Kanwal et al., 2022), this study revealed that EGFR protein expression is one potentially target for the anticancer effects of Cassia fistula. Figure 6. Figure 6. Figure 6. Figure 6. ...
Article
Full-text available
This study aimed to determine the anticancer effects of extracts prepared from various parts of Cassia fistula L. (CF), i.e., flower extract (FE), fruit pulp extract (FPE) and seed extract (SE), on MCF-7 breast cancer cells. The anticancer effects of the extracts were assessed for cell toxicity, cell proliferation, cell migration, cell apoptosis, and production of reactive oxygen species (ROS). Effective cancer treatments have focused on inhibiting epidermal growth factor receptor (EGFR) signalling. Thus, the expression of EGFR protein after extract-treated cells was also determined. Following a 72 incubation, high potential cytotoxicity on MCF-7 cells was observed after SE treatment, followed by FE and FPE treatment. FE, FPE, and SE significantly inhibited cell growth at concentrations of 500, 1,000, and 250 µg/mL, respectively. Also, FE, FPE, and SE markedly suppressed migration of MCF-7 cells at concentrations of 500, 500, and 100 µg/mL, respectively. These results can be concluded that SE had the highest potential anticancer effect on MCF-7 cells when compared with FE and FPE. Thus, SE might be a potential source of preventative and therapeutic agents against breast cancer. Since most anticancer drugs cause ROS production in cancer cells and it is known that ROS induce cell death; therefore, cell apoptosis and ROS formation induced by SE were further studied. The results showed SE induced MCF-7 cell apoptosis in a concentration-dependent way. SE caused a significant increase in ROS formation when compared with the control group. Western blot analysis showed low levels of EGFR protein expression after SE-treated cells at 1,000 mg/mL. Therefore, besides ROS formation, it may be concluded that the downregulation of EGFR protein expression is potentially one of the fundamental mechanisms driving the anticancer effects of SE.
Article
Full-text available
Motivation Membrane proteins are encoded by approximately one fifth of human genes but account for more than half of all US FDA approved drug targets. Thanks to new technological advances, the number of membrane proteins archived in the PDB is growing rapidly. However, automatic identification of membrane proteins or inference of membrane location is not a trivial task. Results We present recent improvements to the RCSB Protein Data Bank web portal (RCSB PDB, rcsb.org) that provide a wealth of new membrane protein annotations integrated from 4 external resources: OPM, PDBTM, MemProtMD, and mpstruc. We have substantially enhanced the presentation of data on membrane proteins. The number of membrane proteins with annotations available on rcsb.org was increased by ∼80%. Users can search for these annotations, explore corresponding tree hierarchies, display membrane segments at the 1D amino acid sequence level, and visualize the predicted location of the membrane layer in 3D. Availability Annotations, search, tree data, and visualization are available at our rcsb.org web portal. Membrane visualization is supported by the open-source Mol* viewer (molstar.org and github.com/molstar/molstar). Supplementary information Supplementary data are available at Bioinformatics online.
Article
Full-text available
Medicinal plant species contain vast and unexploited riches of chemical substances with high medical potential making these plant species valuable as biomedicine sources. Cassia fistula L is an important medicinal plant used in many traditional medicinal systems including Ayurveda and Chinese Traditional Medicine. It is a deciduous medium sized tree with elongated and rod-shaped fruits having pulp and have bright yellow flowers, earning the name 'Yellow Shower'. The present review provides a version of updated information on its botanical description and pharmacological properties including antioxidant, antimicrobial, anti-inflammatory, antidiabetic, anti-tumor, hepatoprotective among other activities. Pharmacological reviews on medicinal plants will provide valuable information; thus, Cassia fistula L can provide important discoveries of valuable bioactive natural products facilitating in developing novel pharmaceuticals products.
Article
Full-text available
Ayurveda is an Indian traditional system of medicine used for various disease conditions. Plants are an important source of treatments and are extensively used for the preparation of different formulations. Herbal drugs are separately or in combination used for the treatment of various ailments in the traditional system of medicines. Nalpamaram is an important group of four trees used in Ayurveda which comprises trees like Ficus racemosa, Ficus microcarpa, Ficus benghalensis, and Ficus religiosa belongs to the family Moraceae. Trees have enormous medicinal values and they are lactiferous in nature. These trees are separately or in combination used for the treatment of various ailments in the traditional system of medicines. Different plant parts like bark, leaves, and fruits of ficus species are used as astringent, hemostatic, antiseptic, anti-inflammatory, anti-oxidant, and anti-cancer, and it useful in pitta and Kapha. They are also effective in the treatment of diarrhoea, dysentery, ulcers, vaginal disorders, leucorrhoea, menorrhea, and deficient lactation conditions. The barks of these trees are an important ingredient of many Ayurvedic formulations like Nalpamaradi Choorna, Nalpamaradi Tailam, Saribadyasavam, and Chandanasavam. The bark of tree is boiled with water; its decoction is used to cleanse the body of pregnant women towards reduce inflammation. This review article compiled the ethnobotanical and phytopharmacological properties of four ficus species trees of Nalpamaram.
Article
Full-text available
Prostate cancer (PCa) is a cancer that occurs in the prostate with high morbidity and mortality. Danggui Beimu Kushen Wan (DBKW) is a classic formula for patients with difficult urination including PCa. This study aimed to investigate the molecular mechanisms of DBKW for PCa. We obtained DBKW compounds from our previous reviews. We identified potential targets for PCa from literature search, currently approved drugs and Open Targets database and filtered them by protein–protein interaction network analysis. We selected 26 targets to predict three cancer-related pathways. A total of 621 compounds were screened via molecular docking using PyRx and AutoDock Vina against 21 targets for PCa, producing 13041 docking results. The binding patterns and positions showed that a relatively small number of tight-binding compounds from DBKW were predicted to interact strongly and selectively with three targets. The top five high-binding-affinity compounds were selected to generate a network, indicating that compounds from all three herbs had high binding affinity against the 21 targets and may have potential biological activities with the targets. DBKW contains multi-targeting agents that could act on more than one pathway of PCa simultaneously. Further studies could focus on validating the computational results via experimental studies.
Article
Full-text available
Medicinal plants are essential aspects of readily available primary healthcare remedies. Phytochemical constituents of medicinal plants cover a broad variety of chemical fields to explore medicines. This review highlights selected empirical data on traditional uses, phytochemistry, and pharmacological properties of Taunggyi medicinal plants, Andrographis paniculata, Physalis peruviana, and Cassia fistula. Historically, these plants have been used for many infections and diseases in Taunggyi. More than 361 chemical compounds have been isolated and identified from the selected plants. Some of the chemical constituents have substantial pharmacological properties. It is clear that these herbs have significant potential for useful natural supplements in many contemporary diseases. Thus, the aim of this review compiles an ethnobotanical survey and documentation of medicinal plants in Taunggyi (Myanmar). This review will also inspire Myanmar researcher's to further investigate the potential of these plants in their future work into new compound and new drugs.
Article
Full-text available
Sparganii rhizoma (SL) has potential therapeutic effects on gastric cancer (GC), but its main active ingredients and possible anticancer mechanism are still unclear. In this study, we used HPLC-Q-TOF–MS/MS to comprehensively analyse the chemical components of the aqueous extract of SL. On this basis, a network pharmacology method incorporating target prediction, gene function annotation, and molecular docking was performed to analyse the identified compounds, thereby determining the main active ingredients and hub genes of SL in the treatment of GC. Finally, the mRNA and protein expression levels of the hub genes of GC patients were further analysed by the Oncomine, GEPIA, and HPA databases. A total of 41 compounds were identified from the aqueous extract of SL. Through network analysis, we identified seven main active ingredients and ten hub genes: acacetin, sanleng acid, ferulic acid, methyl 3,6-dihydroxy-2-[(2-hydroxyphenyl) ethynyl]benzoate, caffeic acid, adenine nucleoside, azelaic acid and PIK3R1, PIK3CA, SRC, MAPK1, AKT1, HSP90AA1, HRAS, STAT3, FYN, and RHOA. The results indicated that SL might play a role in GC treatment by controlling the PI3K-Akt and other signalling pathways to regulate biological processes such as proliferation, apoptosis, migration, and angiogenesis in tumour cells. In conclusion, this study used HPLC-Q-TOF–MS/MS combined with a network pharmacology approach to provide an essential reference for identifying the chemical components of SL and its mechanism of action in the treatment of GC.
Article
Full-text available
Interleukin-17 receptor D (IL-17RD), also known as similar expression to Fgf genes (SEF), is proposed to act as a signaling hub that negatively regulates mitogenic signaling pathways, like the ERK1/2 MAP kinase pathway, and innate immune signaling. The expression of IL-17RD is downregulated in certain solid tumors, which has led to the hypothesis that it may exert tumor suppressor functions. However, the role of IL-17RD in tumor biology remains to be studied in vivo. Here, we show that genetic disruption of Il17rd leads to the increased formation of spontaneous tumors in multiple tissues of aging mice. Loss of IL-17RD also promotes tumor development in a model of colitis-associated colorectal cancer, associated with an exacerbated inflammatory response. Colon tumors from IL-17RD-deficient mice are characterized by a strong enrichment in inflammation-related gene signatures, elevated expression of pro-inflammatory tumorigenic cytokines, such as IL-17A and IL-6, and increased STAT3 tyrosine phosphorylation. We further show that RNAi depletion of IL-17RD enhances Toll-like receptor and IL-17A signaling in colon adenocarcinoma cells. No change in the proliferation of normal or tumor intestinal epithelial cells was observed upon genetic inactivation of IL-17RD. Our findings establish IL-17RD as a tumor suppressor in mice and suggest that the protein exerts its function mainly by limiting the extent and duration of inflammation.
Article
AutoDock Vina is arguably one of the fastest and most widely used open-source programs for molecular docking. However, compared to other programs in the AutoDock Suite, it lacks support for modeling specific features such as macrocycles or explicit water molecules. Here, we describe the implementation of this functionality in AutoDock Vina 1.2.0. Additionally, AutoDock Vina 1.2.0 supports the AutoDock4.2 scoring function, simultaneous docking of multiple ligands, and a batch mode for docking a large number of ligands. Furthermore, we implemented Python bindings to facilitate scripting and the development of docking workflows. This work is an effort toward the unification of the features of the AutoDock4 and AutoDock Vina programs. The source code is available at https://github.com/ccsb-scripps/AutoDock-Vina.
Article
Ovarian clear cell carcinoma (OCCC) is an uncommon subtype of epithelial cell ovarian cancers (EOCs) that has poor response to conventional platinum-based therapy. Therefore, finding new potential therapeutic agents is required. Since inflammatory cytokine, tumor necrosis factor alpha (TNF-α), is strongly expressed in EOCs and associated with the level of tumor grade, disruption of this inflammation pathway may provide another potential target for OCCC treatment. We previously reported that Kaempferia parviflora (KP) extract decreased cell proliferation and induced apoptosis. However, the effects of KP on OCCC, especially the aspects related to inflammatory cytokines, have not been elucidated. Our current study demonstrated the effects of KP extract on cytokine production in TNF-α-induced OCCC TOV-21G cell line. This study showed that KP extract inhibited interleukin 6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1) production at both transcription and translation levels via the suppression of nuclear factor-kappa B (NF-κB) signal transduction. In contrast, KP extract increased the expression of inhibitor kappa B (IκB) protein which may delay NF-κB translocation into the nucleus upon TNF-α activation. Moreover, the suppression of cytokines released from KP treated-TOV-21G reduced the migration of monocyte cell (THP-1). KP extract also exhibited the inhibition of IL-6 and MCP-1 production from THP-1 activated by lipopolysaccharides (LPS). Cells treated with KP extract exhibited a decrease in extracellular signal-regulated kinases (ERK1/2) and protein kinase B (AKT) phosphorylation and induced myeloid leukemia cell differentiation protein Mcl-1 (MCL-1) expression. Suppression of inflammatory cytokine and chemokine production and inhibition of tumor-associated macrophage (TAM) migration support the possibility of using KP for OCCC treatment.
Article
Background: Astragalus membranaceus refers to a type of traditional Chinese medicine (TCM) used to treat type 2 diabetes mellitus (T2DM), whereas its molecular mechanism remains unclear. In the presented study, network pharmacology was performed to analyze the molecular mechanism of astragalus membranaceus against T2DM. Methods: First, we found common targets of astragalus membranaceus and disease, protein-protein interaction (PPI) network was built by String, and then key targets were screened from these common targets by topological analysis. Subsequently, common targets were introduced into DAVID to achieve the results of gene ontology (GO) and KEGG enrichment analysis. The therapeutic effect of astragalus was observed, and several key targets were verified by an animal experiment. Results: First, 13 key targets (EGFR, KDR, SRC, ERBB2, FYN, ESR1, AR, HSP90AA1, PTGS2, ABCG2, AB1, MMP2, and CYP1) were found by topological analysis. Then, the results of GO and KEGG suggested that the anti-diabetes effect of astragalus membranaceus was strongly associated with the activation of receptor protein tyrosine kinase (RPTK). The results of animal experiments revealed that astragalus could enhance the morphology of rat pancreas and up-regulate the expression of tyrosine receptor. Conclusions: In brief, 13 key targets were found in this study, and astragalus membranaceus was found up-regulating insulin signaling pathways by improving the activity of casein kinase, regulating lipid metabolism, and enhancing insulin resistance to treat T2DM. The present study lays a basis for subsequent experimental research and broadens the clinical application of astragalus membranaceus.