Next Article in Journal
Structural Insights into Novel 15-Prostaglandin Dehydrogenase Inhibitors
Next Article in Special Issue
In Silico Identification of Possible Inhibitors for Protein Kinase B (PknB) of Mycobacterium tuberculosis
Previous Article in Journal
Controlled Drug Delivery Systems: Current Status and Future Directions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Network Pharmacology-Based Study to Uncover Potential Pharmacological Mechanisms of Korean Thistle (Cirsium japonicum var. maackii (Maxim.) Matsum.) Flower against Cancer

Department of Bio-Health Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Korea
*
Author to whom correspondence should be addressed.
Molecules 2021, 26(19), 5904; https://doi.org/10.3390/molecules26195904
Submission received: 4 September 2021 / Revised: 26 September 2021 / Accepted: 27 September 2021 / Published: 29 September 2021
(This article belongs to the Special Issue Virtual Screening in Chemical Biology)

Abstract

:
Cirsium japonicum var. maackii (Maxim.) Matsum. or Korean thistle flower is a herbal plant used to treat tumors in Korean folk remedies, but its essential bioactives and pharmacological mechanisms against cancer have remained unexplored. This study identified the main compounds(s) and mechanism(s) of the C. maackii flower against cancer via network pharmacology. The bioactives from the C. maackii flower were revealed by gas chromatography-mass spectrum (GC-MS), and SwissADME evaluated their physicochemical properties. Next, target(s) associated with the obtained bioactives or cancer-related targets were retrieved by public databases, and the Venn diagram selected the overlapping targets. The networks between overlapping targets and bioactives were visualized, constructed, and analyzed by RPackage. Finally, we implemented a molecular docking test (MDT) to explore key target(s) and compound(s) on AutoDockVina and LigPlot+. GC-MS detected a total of 34 bioactives and all were accepted by Lipinski’s rules and therefore classified as drug-like compounds (DLCs). A total of 597 bioactive-related targets and 4245 cancer-related targets were identified from public databases. The final 51 overlapping targets were selected between the bioactive targets network and cancer-related targets. With Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, a total of 20 signaling pathways were manifested, and a hub signaling pathway (PI3K-Akt signaling pathway), a key target (Akt1), and a key compound (Urs-12-en-24-oic acid, 3-oxo, methyl ester) were selected among the 20 signaling pathways via MDT. Overall, Urs-12-en-24-oic acid, 3-oxo, methyl ester from the C. maackii flower has potent anti-cancer efficacy by inactivating Akt1 on the PI3K-Akt signaling pathway.

Graphical Abstract

1. Introduction

The definition of cancer is that normal cells are damaged via an aberrant endogenous process such as an abnormality during DNA replication or instability of the DNA sequence, which transforms into malignancy [1]. The DNA damage responses represent chronic inflammation via the immune signaling pathway, which results in accelerating tumorigenesis [2]. The damaged normal cells undergo cellular senescence, triggering secretion in the inflammatory cytokines leading to cellular mechanical disruption [3,4]. Because inflammation is a leading factor in causing pathological symptoms such as unknown severe pain, fatigue, and comorbidity in cancer patients, anti-inflammation strategies are thus key therapeutics [5,6]. Current anti-inflammatory agents for cancer treatment NSAIDs, including Cox-2, due to fewer adverse effects and a lower mortality rate [7]. Most commonly, anti-cancer agents aim to inhibit DNA replication and induce cancer cell death; however, cancer chemotherapeutics also attack healthy cells resulting in serious side effects such as nausea, vomiting, hair loss, and fatigue [8,9,10]. On the other hand, traditional herbal plants with innovative bioactives and secondary metabolites play an essential role as effective anti-inflammatory, anti-oxidant, or anti-cancer agents [11]. For instance, plant extracts (Urtica membranaceae, Artemesia monosperma, and Origanum dayi Post) in combination with anti-cancer drugs showed enhanced potency against specific cancer cell lines (lung, breast, colon, and prostate cancer) without exposing normal lymphocytes and fibroblasts to cytotoxicity [12]. Herbal-derived bioactives possess fewer unwanted side effects than chemotherapy, and have led to new clinical drugs such as taxol from Taxus brevifolia L., vincristine from Catharanthus roseus G. Don, and Epipodophyllotoxin from Podophyllum peltatum L. [12,13].
C. maackii is a perennial herbal plant, belonging to the family of Compositae, and is widely distributed in the mountainous areas of Korea, Japan, and China [14]. Furthermore, Cirsium species have been reported to have diverse pharmacological activities such as anti-oxidant, anti-inflammation, anti-cancer, and hepatoprotection effects [15,16,17,18]. Specifically, a C. maackii extract at a concentration of 200 µg/mL showed 36.89% inhibition against a breast cancer cell line (MDA-MB-231) [19]. Another study demonstrated that HepG2 cells treated with a MeOH extract of C. maackii have potent antioxidant efficacy against severe oxidant conditions [20]. Anti-inflammatory agents assist in protection against cancer development, thereby preventing cytokine storms [21]. It implies that anti-inflammatory compounds are important agonists to protect normal cells adjacent to tumor cells because they can block the overflow of cytokines. Until now, C. maackii flower compounds were identified by HPLC and had only been reported for anti-Alzheimer efficacy by inhibiting BACE1 [22]. Generally, the identification of polar and mid-polar compounds from extracts is based on HPLC due to its good separation capability [23,24]. From a different perspective, we utilized GC-MS analysis to discover lipophilic bioactives, which mainly act as drug-like compounds and uptake efficiently into the cells. Lipophilicity is a significant physicochemical parameter that influences membrane permeability and affinity [25]. More importantly, GC-MS, along with the molecular docking test (MDT) and ADME (Absorption, Distribution, Metabolism, and Excretion) study, is an optimal analytical method to determine drug-like compounds [26]. At present, the bioactives and mechanisms of the C. maackii flower against cancer remain unknown. Hence, we aimed to uncover its potential bioactives with their fundamental mechanisms through network pharmacology.
Network pharmacology (NP) as a systemic method can analyze holistic bioactive–target–disease relationships [27]. It can decipher the unknown mechanism(s) with “multiple targets”, “multiple bioactives”, instead of “one target”, “one bioactive” [28]. This approach is a very effective method to recognize the mechanism of action for lead compounds discovered from herbal plants [29]. An existing drug may be re-modelled to bind on multiple targets with the concept of NP, thus it can be a guide for drug repurposing [30]. The NP application is a powerful tool to elucidate novel targets and bioactives from natural products, and is especially effective for anti-cancer research to investigate multi-target activity in biological pathways and interaction networks [31]. Here, we implemented NP to provide key bioactive(s), uppermost target(s), and potential mechanism(s) of the C. maackii flower against cancer. Mainly, the flower part of herbal plants are the most underexplored part compared to other parts such as leaves, roots, and stems. During the growing season of the C. maackii plant, the flowering part might be utilized as a source of essential bioactives or be taken as a functional food. In this study, we suggest that C. maackii flowers are valuable parts with potential anti-cancer compounds.
To prove their therapeutic value, we performed a GC-MS analysis, protein network investigation, and a molecular docking test (MDT). Its brief processes are discussed below.
Firstly, bioactives from the C. maackii flower were identified by GC-MS analysis and screened to find drug-likeness compounds via an in silico tool. Then, targets associated with bioactives or cancer were identified through public databases, and final overlapping targets were utilized to analyze protein–protein interaction (PPI) and the highest degree of a target. Thirdly, a signaling pathway–target protein–bioactive (S–T–B) relationship against cancer was identified by networking analysis. Lastly, we found the most potent bioactive and a hub target to alleviate cancer severity by exploring the molecular mechanism of the C. maackii flower based on MDT. The workflow diagram is displayed in Figure 1.

2. Results

2.1. Bioactives from C. maackii Flower

A total of 34 bioactives in the C. maackii flower were identified by GC-MS analysis (Figure 2), and the name of compounds, PubChem ID, retention time (mins), and peak area (%) are listed in Table 1. All 34 compounds were accepted by Lipinski’s rules (Molecular Weight ≤ 500 g/mol; Moriguchi octanol-water partition coefficient ≤ 4.15; Number of Nitrogen or Oxygen ≤ 10; Number of NH or OH ≤ 5), and all bioactives corresponded with the standard of “Abbott Bioavailability Score (>0.1)” through SwissADME. The TPSA (Topological Polar Surface Area) value of all bioactives was also accepted (Table 2).

2.2. Overlapping Targets between SEA and STP Associated with Bioactives

A total of 309 targets from SEA and 396 targets from STP connected to 34 bioactives were identified (Supplementary Table S1). The Venn diagram showed that 108 targets overlapped between the two public databases (Supplementary Table S1) (Figure 3A).

2.3. Overlapping Targets between Cancer-Associated Targets and the Final 51 Overlapping Targets

A total of 4245 targets related to cancer were selected via retrieval from TTD and OMIM databases (Supplementary Table S2). The Venn diagram results revealed 51 overlapping targets that were selected between 4245 targets associated with cancer and 108 overlapping targets (Figure 3B) (Supplementary Table S3).

2.4. Acquisition of a Hub Target from PPI Networks

From STRING analysis, 46 out of 51 overlapping targets were directly related to cancer occurrence and development, indicating 46 nodes and 145 edges (Figure 4). The five targets removed (PAM, EPHX1, PPARD, KDM5C, and BCHE) had no connectivity to the overlapping 51 targets. In PPI networks, the Akt1 target was the highest degree (29) and was considered a hub target (Table 3).

2.5. Identification of a Hub Signaling from Bubble Chart

The output of KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway enrichment analysis revealed that 20 signaling pathways were associated with 24 targets (False Discovery Rate < 0.05). The 20 signaling pathways were directly connected to cancer, suggesting that these 20 signaling pathways might be the noteworthy pathways of the C. maackii flower against cancer. The descriptions of 20 signaling pathways are displayed in Table 4. Additionally, a bubble chart suggested that the PI3K-Akt signaling pathway might be a hub signaling pathway of the C. maackii flower against cancer (Figure 5). Among the 20 signaling pathways, the Akt1 target was associated with 18 signaling pathways, representing the highest degree of value. Most importantly, Akt1 is directly related to the PI3K-Akt signaling pathway whereas both the PPAR signaling pathway and the Calcium signaling pathway are not correlated with Akt1.

2.6. S–T–B Network Analysis of C. maackii Flower against Cancer

The S–T–B network of the C. maackii flower is exhibited in Figure 6. There were 20 pathways, 24 targets, and 19 bioactives (63 nodes and 204 edges). The nodes represented a total of three elements: Signaling pathways-Target protein-Bioactive (S-T-B). The edges represented relationships of a total number of three elements. The S–T–B relationship suggested that the network might interact with therapeutic efficacy against cancer. Regarding the 20 signaling pathways, the highest degree among targets was “Akt1” with 18 degrees, and the highest degree among bioactives was “Urs-12-en-24-oic acid, 3-oxo-, methyl ester” with 36 degrees (Table 5).

2.7. MDT of 3 Targets and 10 Bioactives Connected to PI3K-Akt Signaling Pathway

The Akt1 protein was related to two bioactives (Urs-12-en-24-oic acid, 3-oxo-, methyl ester and α-Tocopherol), the VEGFA target to three bioactives (Allyl stearate, Isopropyl palmitate, and Methyl 3,6-anhydro hexopyranoside #), and the PRKCA target with seven bioactives (Allyl stearate, Isopropyl palmitate, Pentyl isobutyrate, 9-Heptadecanone, Palmitic acid, Stearic acid, and d-Lyxo-d-manno-nononic- 1,4-lactone). MDT was performed to verify the affinity of target protein(s) and biactive(s), which displayed the docking figure of a hub target—the uppermost bioactive (Figure 7). The Akt1 protein (PDB ID: 5KCV) connected to two compounds on the PI3K-Akt signaling pathway was subjected to MDT. It was observed that Urs-12-en-24-oic acid, 3-oxo-, methyl ester (−12.8 kcal/mol) docked on the Akt1 protein (PDB ID: 5KCV) manifested the highest binding energy, followed by α-Tocopherol (−5.8 kcal/mol). The docking detail results of two bioactives are shown in Table 6. The MDT score of three bioactives on the VEGFA protein (PDB ID: 3V2A) was analyzed in the “Homo Sapiens” mode. It was revealed that Methyl 3,6-anhydro hexopyranoside # (−5.2 kcal/mol) docked on the VEGFA protein (PDB ID: 3V2A) exhibited the highest binding energy followed by Allyl stearate (−5.1 kcal/mol) and Isopropyl palmitate (−5.1 kcal/mol). The docking detail results of three bioactives are shown in Table 7. The MDT score of seven bioactives on the PRKCA protein (PDB ID: 3IW4) was analyzed in the “Homo Sapiens” mode. It was exposed that d-Lyxo-d-manno-nononic- 1,4-lactone (−6.9 kcal/mol) docked on the PRKCA protein (PDB ID: 3IW4) demonstrated the highest binding energy, followed by Isopropyl palmitate (−6.3 kcal/mol), Stearic acid (−6.2 kcal/mol), Allyl stearate (−6.2 kcal/mol), 9-Heptadecanone (−5.4 kcal/mol), Pentyl isobutyrate (−5.0 kcal/mol), and Palmitic acid (−4.8 kcal/mol). The docking detail results of seven bioactives are shown in Table 8. Collectively, both VEGFA (PDB ID: 3V2A) and PRKCA proteins (PDB ID: 3IW4) showed that the affinity of each bioactive did not give a valid binding score (>−7.0 kcal/mol) [45].

2.8. Comparative Analysis of MDT against Positive Controls on a Hub Target

A key bioactive (Urs-12-en-24-oic acid, 3-oxo-, methyl ester) associated with the PI3K-Akt signaling pathway revealed the greatest binding score (−12.8 kcal/mol) on Akt1 (PDB ID: 5KCV). The 13 positive controls on Akt1 (PDB ID: 5KCV) are as follows. The MDT score of BAY1125976 (PubChem ID: 70817911) was the highest score with −9.1 kcal/mol, followed by Miransertib (PubChem ID: 53262401; −9.0 kcal/mol)), Akti-1/2 (PubChem ID: 135398501; −8.9 kcal/mol), Oridonin (PubChem ID: 5321010; −8.3 kcal/mol), MK-2206 dihydrochloride (PubChem ID: 46930998; −7.8 kcal/mol), AT7867 (PubChem ID: 11175137; −7.6 kcal/mol), A-674563 (PubChem ID: 11314340; −7.6 kcal/mol), Gsk-690693 (PubChem ID: 16725726; −7.4 kcal/mol), Ipatasertib (PubChem ID: 24788740; −7.3 kcal/mol), Capivasertib (Pubchem ID: 25227436; −7.1 kcal/mol), AT13148 (PubChem ID: 24905401; −6.9 kcal/mol), Afuresertib (PubChem ID: 46843057; −6.9 kcal/mol), and Uprosertib (PubChem ID: 51042438; −6.8 kcal/mol), respectively. In particular, the MDT score of Urs-12-en-24-oic acid, 3-oxo-, methyl ester on Akt1 (PDB ID: 5KCV) had ahigher affinity (−12.8 kcal/mol) compared to the 13 positive controls. Detailed information is listed in Table 9.

2.9. Toxicological Properties of a Selected Key Bioactive

Additionally, the toxicological properties of Urs-12-en-24-oic acid, 3-oxo-, methyl ester were predicted by the admetSAR online tool. Our result suggested that the bioactive did not disclose Ames toxicity, carcinogenic properties, acute oral toxicity, or rat acute toxicity properties (Table 10).

3. Discussion

The S–T–B network suggested that the therapeutic efficacy of the C. maackii flower against cancer was directly associated with 20 signaling pathways, 24 targets, and 19 bioactives. Through the network, we identified the most significant protein (Akt1) associated with the occurrence and development of cancer and a bioactive (Urs-12-en-24-oic acid, 3-oxo-, methyl ester) from the C. maackii flower. From a bubble chart, we identified a hub signaling pathway (PI3K-Akt signaling pathway) connected to the Akt1 target, indicating the lowest rich factor among 20 signaling pathways. A report demonstrated that the activated PI3K-Akt signaling pathway accelerates tumor cell proliferation, invasion, and metastasis, inhibiting apoptosis [46]. Furthermore, it was found that the Akt1 target was overexpressed in 15 out of 24 human hepatocellular carcinomas (63.3%) confirmed using PCR analysis through Northern blot [47]. Another research study suggested that the PI3K-Akt signaling pathway’s antagonists are potential anti-cancer candidates to regulate acute and chronic inflammatory responses [48].
In the S–T–B network, Urs-12-en-24-oic acid, 3-oxo-, methyl ester had the highest degree of value and was considered the uppermost bioactive of the C. maackii flower against cancer. Urs-12-en-24-oic acid, 3-oxo-, methyl ester is categorized into boswellic acids used widely as anti-inflammatory agents, including as an anti-cancer treatment. Additionally, the boswellic acids have potent anti-cancer efficacy against diverse malignant cancers [49,50]. The KEGG pathway enrichment analysis of 24 targets suggested that a total of 20 signaling pathways were involved in cancer occurrence and development. The relationships of the 20 signaling pathways with cancer are concisely discussed as follows. In the Peroxisome Proliferator-Activated Receptor (PPAR) signaling pathway, the activation of the PPAR signaling pathway functions as an anti-inflammatory agent, which can overwhelm the metabolic energy balance of cancer cells by inhibiting the fatty acid synthesis and accelerating fatty acid oxidation [51,52]. In the Mitogen-Activated Protein Kinase (MAPK) signaling pathway, MAPK inhibitors are efficient blockers to reduce pro-inflammatory cytokines and enhance the anti-cancer effect, particularly on human pancreatic cancer cells [53,54]. In the Rap1 (Ras-associated protein-1) signaling pathway, Rap1 promotes cytokine production during the inflammatory condition, which leads to tumor progression in human colorectal cancer [55,56]. Regarding the alcium signaling pathway, calcium is a significant second messenger to regulate inflammation; hence, blockers of the calcium channel can induce cancer cell death [57,58]. In the Cyclic AMP (cAMP) signaling pathway, an increased cAMP level has an anti-inflammatory effect, where the increased level can regulate DNA damage, DNA repair, and apoptosis of cancer cells [59,60]. Concerning the Hypoxia-Inducible Factor-1 (HIF-1) signaling pathway, HIF-1 is a central regulator to stimulate the production of inflammation. HIF-1 overexpression is related to increased tumor growth [61,62]. In the sphingolipid signaling pathway, sphingolipid is implicated in the inflammatory response, which has been involved in cancer cell proliferation [63,64]. In the Phospholipase D (PLD) signaling pathway, PLD inhibition can induce two functions, namely anti-inflammation and anti-cancer. Mainly, the blocking of PLD during chemotherapy can sensitize one to chemotherapeutics [65,66]. Regarding the Phosphoinositide 3-kinase-Akt (PI3K-Akt) signaling pathway, inhibition of the PI3K-Akt signaling pathway reduces the severity of inflammation in mice. This strategy is a promising mechanism for the treatment of cancers such as lung cancer, colorectal cancer, renal cancer, prostate cancer, triple-negative breast cancer, mucinous adenocarcinoma of the ovary, and skin cancer [67,68]. In the AMP-activated protein kinase (AMPK) signaling pathway, AMPK activation suppresses inflammatory responses and dampens cancer growth with cell metabolism and the cell cycle [69,70]. Regarding the Vascular Endothelial Growth Factor (VEGF) signaling pathway, VEGF is a core mediator in the formation of new blood vessels for cancer cells; thereby, cancer cells can survive, grow, and metastasize [71]. In the B cell receptor (BCR) signaling pathway, B cells are involved in the inflammatory T cell receptor, which is implicated in antibody production [72]. B cells or BCR-associated kinases may function with anti-cancer activity via B cells activation [73]. In the Fc epsilon RI signaling pathway, the expression of Fc epsilon on mast cells stimulates immunoglobulin E, leading to type 1 hypersensitivity-induced local inflammatory responses at the tumor sites [74]. In the insulin signaling pathway, insulin-resistant patients undergo excessive production of reactive oxygen species (ROS) that can harm DNA attributed to carcinogenesis [75]. In the estrogen signaling pathway, estrogen exposure to chronic inflammatory disease activity is a key risk in breast cancer progression [76]. With regards to the prolactin signaling pathway, prolactin functions as a cytokine immune system, especially in breast cancer, which has the strongest correlation with an increased expression level of prolactin and prolactin receptors [77,78]. In the thyroid signaling pathway, the optimal regulation of the cellular thyroid hormone is essential for an adequate role of immune cells during inflammation [79]. In terms of the adipocytokine signaling pathway, adipocytokine is an inflammatory mediator during immune-associated diseases, which accelerates cancer progression and metastasizes from organ to organ [80]. In the Relaxin signaling pathway, Relaxin alleviates the inflammatory severity and diminishes the amount of leucocytes and the expression level of cytokines [81]. In terms of the Advanced Glycation End-product (AGE)-Advanced Glycation End-product Receptor (RAGE) signaling pathway in diabetic complications, RAGE can stimulate inflammatory responses by binding with AGEs [82]. The inhibitor (papaverine) of RAGE is a promising target for anti-cancer activity, which blocks nuclear factor kappa B (NF- κB) [83].

4. Materials and Methods

4.1. Plant Material Collection and Identification

The C. maackii flowers were collected from Mihogil of Bomunmyeon (Latitude: 36.666149, Longitude: 128.511759), Kyeongsang-bukdo, Republic of Korea, in October 2020, and the plant was identified by Dr. Dong Ha Cho, Plant biologist and Professor, Department of Bio-Health Convergence, College of Biomedical Science, Kangwon National University. A voucher number (CNB 015) has been deposited at the Kenaf Corporation in the Department of Bio-Health Convergence, and the material can only be used as research.

4.2. Plant Preparation, Extraction

The C. maackii flower was dried in a shady area at room temperature (20–22 °C) for 7 days, and dried leaves were powdered using an electric blender (Shinil, Cheonan, Korea). Approximately 50 g of C. maackii flower powder was soaked in 800 mL of 100% methanol (Daejung, Seohaean, Korea) for 5 days and repeated 3 times to collect extraction. The solvent extract was collected, filtered, and evaporated using a vacuum evaporator (RV8, IKA, Staufen, German). The evaporated sample was dried under a boiling water bath (HB10, IKA, Staufen, German) at 40 °C to obtain the extract.

4.3. GC-MS Analysis Condition

Agilent 7890A (Agilent, Santa Clara, CA, USA) was used to carry out the GC-MS analysis. GC was equipped with a DB-5 (30 m × 0.25 mm × 0.25 μm) capillary column (Agilent, Santa Clara, CA, USA). Initially, the instrument was maintained at a temperature of 100 °C for 2.1 min. The temperature rose to 300 °C at a rate of 25 °C/min and was maintained for 20 min. The injection port temperature and helium flow rate were sustained at 250 °C and 1.5 mL/min, respectively. The ionization voltage was 70 eV. The samples were injected in the split mode at 10:1. The MS scan range was set at 35–900 (m/z). The fragmentation patterns of mass spectra were compared with those stored in the W8N05ST Library MS database. The percentage of each compound was calculated from the relative peak area of each compound in the chromatogram. The concept of integration was used with the ChemStation integrator (Agilent, Santa Clara, CA, USA) algorithms (analyzed 19 May 2021) [84].

4.4. Bioactives Database Construction and Drug-Likeness Property

The bioactives from the C. maackii flower were identified by utilizing GC-MS analysis. Then, the GC-MS-detected bioactives were filtered in accordance with Lipinski’s rules through SwissADME (http://www.swissadme.ch/) (accessed on 3 June 2021) to confirm the “Drug-likeness” physicochemical properties. PubChem (https://pubchem.ncbi.nlm.nih.gov/) (accessed on 3 June 2021) was utilized to select the SMILES (Simplified Molecular Input Line Entry System) bioactives.

4.5. Target Targets Related to Selected Bioactives or Cancer

Targets connected to the bioactives were selected through both the Similarity Ensemble Approach (SEA) (http://sea.bkslab.org/) (accessed on 7 June 2021) [85] and Swiss Target Prediction (STP) (http://www.swisstargetprediction.ch/) (accessed on 9 June 2021) [86] with the “Homo Sapiens” setting, both of which are based on SMILES. The cancer-related targets in humans were obtained with keywords (cancer/tumor/neoplasia/carcinoma) from TTD (http://db.idrblab.net/ttd/) (accessed on 12 June 2021) and OMIM (https://www.omim.org/) (accessed on 13 June 2021). The overlapping targets between compounds of the C. maackii flower and cancer targets were illustrated by VENNY 2.1 (https://bioinfogp.cnb.csic.es/tools/venny/) (accessed on 14 June 2021).

4.6. Construction of PPI Networks and Bubble Chart

For the final overlapping targets, STRING (https://string-db.org/) (accessed on 16 June 2021) [87] was utilized to analyze the PPI network. Thereby, RPackage was used to identify the degree of value. Then, signaling pathways on STRING were visualized by RPackage, a hub signaling pathway (lowest rich factor) related to a hub target (highest degree of value from PPI).

4.7. Construction of a Size Map on S-T-B Network

Both the hub target (the highest degree of value among 20 signaling pathways) and the uppermost bioactive were identified via the S–T–B network. The S–T–B networks were utilized to construct a size map, based on the degree of values. In this size map, green rectangles (nodes) represented signaling pathways, pink triangles (nodes) represented targets, and orange circles (nodes) represented bioactives. The circle size represented the degree value, the size of pink triangles represented the number of connectivity with signaling pathways, and the size of orange circles represented the number of connections with targets. The merged networks were constructed using RPackage.

4.8. Preparation for MDT of Targets

Four targets of a hub signaling pathway, i.e., Akt1 (PDB ID: 5KCV), VEGFA (PDB ID: 3V2A), PRKCA (PDB ID: 3IW4), and PHLPP1 (PDB ID: N/A), were selected on STRING via RCSB PDB (https://www.rcsb.org/) (accessed on 19 June 2021). Specifically, the PHLPP1 protein structure was not determined; thereby, the PDB ID was not uploaded in the .pdb format. Thus, the final three proteins selected in the .pdb format were converted into the .pdbqt format via Autodock (http://autodock.scripps.edu/) (accessed on 22 June 2021) [88].

4.9. Preparation for MDT of Positive Standard Ligands

A total of 13 positive control compounds on Akt1 antagonists, i.e., MK-2206 dihydrochloride (PubChem ID: 46930998), Gsk-690693 (PubChem ID: 16725726), Ipatasertib (PubChem ID: 24788740), Capivasertib (PubChem ID: 25227436), AT7867 (PubChem ID: 11175137), A-674563 (PubChem ID: 11314340), Miransertib (PubChem ID: 53262401), BAY1125976 (PubChem ID: 70817911), Akti-1/2 (PubChem ID: 135398501), Uprosertib (PubChem ID: 51042438), Afuresertib (PubChem ID: 46843057), AT13148 (PubChem ID: 24905401), and Oridonin (PubChem ID: 5321010), were selected to verify the docking score.

4.10. Preparation for MDT of Ligand Molecules

The ligand molecules were converted from sdf in PubChem into the .pdb format using Pymol, and the ligand molecules were converted into the .pdbqt format through Autodock.

4.11. Ligand-Protein Docking

The ligand molecules were docked with targets utilizing autodock4 by setting up an energy range of 4 and exhaustiveness at 8 as the default to obtain 10 different positions of ligand molecules [89]. The center (the position of the middle coordinate point) in the target was X: −12.677, Y: 2.931, Z: −13.145 on AKT1 (PDB ID: 5KCV). The grid box size was set to 40 Å × 40 Å × 40 Å. The 2D binding interactions were used with LigPlot+ v.2.2 (https://www.ebi.ac.uk/thornton-srv/software/LigPlus/) (accessed on 23 June 2021) [90]. After docking, ligands of the lowest binding energy (highest affinity) were selected to visualize the ligand–protein interaction in Pymol.

4.12. Toxicological Properties Prediction by admetSAR

Toxicological properties of the key bioactive were established using the admetSAR web-service tool (http://lmmd.ecust.edu.cn/admetsar1/predict/) (accessed on 24 June 2021) [91] because toxicity is an essential factor in developing new drugs. Hence, Ames toxicity, carcinogenic properties, acute oral toxicity, and rat acute toxicity were predicted by admetSAR.

5. Conclusions

The bioactives and mechanisms of C. maackii flowers against cancer were firstly uncovered through network pharmacology. The findings suggested that 20 signaling pathways, 24 targets, and 19 bioactives are connected to cancer. Of these, the PI3K-Akt signaling pathway, Akt1, and Urs-12-en-24-oic acid, 3-oxo-, methyl ester were the hub signaling pathway, hub target, and key bioactive of C. maackii flowers against cancer, respectively. Furthermore, Urs-12-en-24-oic acid, 3-oxo-, methyl ester has the most potent efficacy on the Akt1 target protein than 13 other standard ligands. This study suggests that the mechanism of the C. maackii flower against cancer might strengthen anti-inflammatory responses by inactivating the PI3K-Akt signaling pathway, bound to Urs-12-en-24-oic acid, 3-oxo-, methyl ester on Akt1. From this viewpoint, we propose that C. maackii flowers can be utilized as functional or medicinal resources against cancer.

Supplementary Materials

The following are available online, Table S1: The 309, 396, and 108 targets from SEA, STP, and overlapping targets between SEA and STP, respectively. Table S2: The 4245 cancer-related targets from TTD and OMIM databases, Table S3: The final 51 targets against cancer.

Author Contributions

Conceptualization, methodology, formal analysis, investigation, visualization, data curation, writing—original draft, K.-K.O.; software, investigation, data curation, K.-K.O. and M.A.; validation, writing—review and editing, M.A.; supervision, project administration, D.-H.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data generated or analyzed during this study are included in this published article (and its Supplementary Information Files).

Acknowledgments

This research was acknowledged by the Department of Bio-Health Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Korea.

Conflicts of Interest

There is no conflict of interest.

Sample Availability

Samples of described compounds are available from the authors.

Abbreviations

ADMEAbsorption: Distribution, Metabolism, Excretion;
AGEAdvanced Glycation End-product;
AMPKAMP-activated protein kinase;
cAMPCyclic AMP;
C. maackiiCirsium japonicum var. maackii (Maxim.) Matsum.: C. maackii;
DLCsDrug-Like Compounds;
BCRB cell receptor;
GC-MSGas Chromatography Mass Spectrum;
HIF-1Hypoxia Inducible Factor-1;
KEGGKyoto Encyclopedia of Genes and Genomes;
MAPKMitogen Activated Protein Kinase;
MDTMolecular Docking Test;
NF- κBNuclear Factor Kappa B;
OMIMOnline Mendelian Inheritance in Man;
PI3K-AktPhosphoinositide 3-kinase-Akt;
PLDPhospholipase D;
PPARPeroxisome Proliferator Activated Receptor;
PPIProtein-protein interaction;
RAGEAdvanced Glycation End-product Receptor;
Rap1Ras-associated protein-1;
ROSReactive Oxygen Species;
SEASimilarity Ensemble Approach;
SMILESSimplified Molecular Input Line Entry System;
S-T-BSignaling pathway-Target protein-Bioactive;
STPSwissTargetPrediction;
t-BHPTert-butyl hydroperoxide;
TTDTherapeutic Target Database;
VEGFVascular Endothelial Growth Factor

References

  1. Bertram, J.S. The molecular biology of cancer. Mol. Asp. Med. 2000, 21, 167–223. [Google Scholar] [CrossRef]
  2. Nakad, R.; Schumacher, B. DNA Damage Response and Immune Defense: Links and Mechanisms. Front. Genet. 2016, 7, 147. [Google Scholar] [CrossRef] [Green Version]
  3. Welsh, C.; Day, R.; McGurk, C.; Masters, J.R.; Wood, R.D.; Köberle, B. Reduced levels of XPA, ERCC1 and XPF DNA repair proteins in testis tumor cell lines. Int. J. Cancer 2004, 110, 352–361. [Google Scholar] [CrossRef] [PubMed]
  4. Munn, L.L. Cancer and inflammation. Wiley Interdiscip. Rev. Syst. Biol. Med. 2017, 9, e1370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lacourt, T.E.; Vichaya, E.G.; Chiu, G.S.; Dantzer, R.; Heijnen, C.J. The high costs of low-grade inflammation: Persistent fatigue as a consequence of reduced cellular-energy availability and non-adaptive energy expenditure. Front. Behav. Neurosci. 2018, 12, 78. [Google Scholar] [CrossRef] [Green Version]
  6. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
  7. Rayburn, E.R.; Ezell, S.J.; Zhang, R. Anti-inflammatory agents for cancer therapy. Mol. Cell. Pharmacol. 2009, 1, 29–43. [Google Scholar] [CrossRef]
  8. Subramaniam, M.; Liew, S.K.; In, L.L.; Awang, K.; Ahmed, N.; Nagoor, N.H. Inactivation of nuclear factor κB by MIP-based drug combinations augments cell death of breast cancer cells. Drug Des. Dev. Ther. 2018, 12, 1053–1063. [Google Scholar] [CrossRef] [Green Version]
  9. Shewach, D.S.; Kuchta, R.D. Introduction to Cancer Chemotherapeutics. Chem. Rev. 2009, 109, 2859–2861. [Google Scholar] [CrossRef] [Green Version]
  10. Kayl, A.E.; Meyers, C.A. Side-effects of chemotherapy and quality of life in ovarian and breast cancer patients. Curr. Opin. Obstet. Gynecol. 2006, 18, 24–28. [Google Scholar] [CrossRef]
  11. Aravindaram, K.; Yang, N.-S. Anti-Inflammatory Plant Natural Products for Cancer Therapy. Planta Medica 2010, 76, 1103–1117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Greenwell, M.; Rahman, P. Medicinal Plants: Their Use in Anticancer Treatment. Int. J. Pharm. Sci. Res. 2015, 6, 4103–4112. [Google Scholar] [CrossRef] [PubMed]
  13. Desai, A.G.; Qazi, G.N.; Ganju, R.K.; El-Tamer, M.; Singh, J.; Saxena, A.K.; Bedi, Y.S.; Taneja, S.C.; Bhat, H.K. Medicinal Plants and Cancer Chemoprevention. Curr. Drug Metab. 2008, 9, 581–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Wagle, A.; Seong, S.H.; Shrestha, S.; Jung, H.A.; Choi, J.S. Korean Thistle (Cirsium japonicum var. maackii (Maxim.) Matsum.): A Potential Dietary Supplement against Diabetes and Alzheimer’s Disease. Molecules 2019, 24, 649. [Google Scholar] [CrossRef] [Green Version]
  15. Nazaruk, J.; Wajs-Bonikowska, A.; Bonikowski, R. Components and antioxidant activity of fruits of Cirsium palustre and C. rivulare. Chem. Nat. Compd. 2012, 48, 8–10. [Google Scholar] [CrossRef]
  16. Kim, E.-Y.; Jho, H.-K.; Kim, N.-I.; Rhyu, M.R. Cirsium japonicum elicits endothelium-dependent relaxation via histamine H1-receptor in rat thoracic aorta. J. Ethnopharmacol. 2008, 116, 223–227. [Google Scholar] [CrossRef]
  17. Liu, S.; Zhang, J.; Li, D.; Liu, W.; Luo, X.; Zhang, R.; Li, L.; Zhao, J. Anticancer activity and quantitative analysis of flavone of Cirsium japonicum DC. Nat. Prod. Res. 2007, 21, 915–922. [Google Scholar] [CrossRef]
  18. Ma, Q.; Guo, Y.; Luo, B.; Liu, W.; Wei, R.; Yang, C.; Ding, C.; Xu, X.; He, M. Hepatoprotective phenylethanoid glycosides from Cirsium setosum. Nat. Prod. Res. 2015, 30, 1824–1829. [Google Scholar] [CrossRef]
  19. Park, J.Y.; Kim, H.Y.; Shibamoto, T.; Jang, T.S.; Lee, S.C.; Shim, J.S.; Hahm, D.-H.; Lee, H.-J.; Lee, S.; Kang, K.S. Beneficial effects of a medicinal herb, Cirsium japonicum var. maackii, extract and its major component, cirsimaritin on breast cancer metastasis in MDA-MB-231 breast cancer cells. Bioorg. Med. Chem. Lett. 2017, 27, 3968–3973. [Google Scholar] [CrossRef] [PubMed]
  20. Jung, H.A.; Abdul, Q.A.; Byun, J.S.; Joung, E.-J.; Gwon, W.-G.; Lee, M.-S.; Kim, H.-R.; Choi, J.S. Protective effects of flavonoids isolated from Korean milk thistle Cirsium japonicum var. maackii (Maxim.) Matsum on tert-butyl hydroperoxide-induced hepatotoxicity in HepG2 cells. J. Ethnopharmacol. 2017, 209, 62–72. [Google Scholar] [CrossRef]
  21. Gartung, A.; Yang, J.; Sukhatme, V.P.; Bielenberg, D.R.; Fernandes, D.; Chang, J.; Schmidt, B.A.; Hwang, S.H.; Zurakowski, D.; Huang, S.; et al. Suppression of chemotherapy-induced cytokine/lipid mediator surge and ovarian cancer by a dual COX-2/sEH inhibitor. Proc. Natl. Acad. Sci. USA 2019, 116, 1698–1703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Bhatarrai, G.; Seong, S.H.; Jung, H.A.; Choi, J.S. Isolation and Quantitative Analysis of BACE1 Inhibitory Compounds from Cirsium maackii Flower. Nat. Prod. Sci. 2019, 25, 326–333. [Google Scholar] [CrossRef] [Green Version]
  23. Aral, H.; Aral, T.; Çelik, K.S.; Ziyadanoğulları, B.; Ziyadanoğulları, R. HPLC Separation of Different Groups of Small Polar Compounds on a Novel Amide-Embedded Stationary Phase. Chromatographia 2014, 77, 771–781. [Google Scholar] [CrossRef]
  24. De La Torre, M.P.D.; Priego-Capote, F.; De Castro, M.D.L. Tentative identification of polar and mid-polar compounds in extracts from wine lees by liquid chromatography-tandem mass spectrometry in high-resolution mode. J. Mass Spectrom. 2015, 50, 826–837. [Google Scholar] [CrossRef] [PubMed]
  25. Constantinescu, T.; Lungu, C.N.; Lung, I. Lipophilicity as a Central Component of Drug-Like Properties of Chalchones and Flavonoid Derivatives. Molecules 2019, 24, 1505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Sympli, H.D. Estimation of drug-likeness properties of GC–MS separated bioactive compounds in rare medicinal Pleione maculata using molecular docking technique and SwissADME in silico tools. Netw. Model. Anal. Heal. Informatics Bioinform. 2021, 10, 1–36. [Google Scholar] [CrossRef]
  27. Oh, K.K.; Adnan, M.; Cho, D.H. Network pharmacology of bioactives from Sorghum bicolor with targets related to diabetes mellitus. PLoS ONE 2020, 15, e0240873. [Google Scholar] [CrossRef]
  28. Hopkins, A.L. Network pharmacology: The next paradigm in drug discovery. Nat. Chem. Biol. 2008, 4, 682–690. [Google Scholar] [CrossRef]
  29. Wang, Y.; Hu, B.; Feng, S.; Wang, J.; Zhang, F. Target recognition and network pharmacology for revealing anti-diabetes mechanisms of natural product. J. Comput. Sci. 2020, 45, 101186. [Google Scholar] [CrossRef]
  30. Ye, H.; Wei, J.; Tang, K.; Feuers, R.; Hong, H. Drug Repositioning Through Network Pharmacology. Curr. Top. Med. Chem. 2016, 16, 3646–3656. [Google Scholar] [CrossRef]
  31. 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. [Google Scholar] [CrossRef]
  32. Nisa, S.; Khan, N.; Shah, W.; Sabir, M.; Khan, W.; Bibi, Y.; Jahangir, M.; Haq, I.U.; Alam, S.; Qayyum, A. Identification and Bioactivities of Two Endophytic Fungi Fusarium fujikuroi and Aspergillus tubingensis from Foliar Parts of Debregeasia salicifolia. Arab. J. Sci. Eng. 2020, 45, 4477–4487. [Google Scholar] [CrossRef]
  33. Bassole, I.H.N.; Nebie, R.; Savadogo, A.; Ouattara, C.T.; Barro, N.; Traore, S.A. Composition and antimicrobial activities of the leaf and flower essential oils of Lippia chevalieri and Ocimum canum from Burkina Faso. Afr. J. Biotechnol. 2005, 4, 1156–1160. [Google Scholar] [CrossRef]
  34. Lee, J.-Y.; Lee, H.-J.; Jeong, J.-A.; Jung, J.-W. Palmitic acid inhibits inflammatory responses in lipopolysaccharide-stimulated mouse peritoneal macrophages. Orient. Pharm. Exp. Med. 2010, 10, 37–43. [Google Scholar] [CrossRef]
  35. Sebastianes, F.L.S.; Cabedo, N.; El Aouad, N.; Valente, A.M.M.P.; Lacava, P.T.; Azevedo, J.L.; Pizzirani-Kleiner, A.A.; Cortes, D. 3-Hydroxypropionic Acid as an Antibacterial Agent from Endophytic Fungi Diaporthe phaseolorum. Curr. Microbiol. 2012, 65, 622–632. [Google Scholar] [CrossRef] [PubMed]
  36. Cardoso, V.M.; Solano, A.G.R.; Prado, M.A.F.; Nunan, E.D.A. Investigation of fatty acid esters to replace isopropyl myristate in the sterility test for ophthalmic ointments. J. Pharm. Biomed. Anal. 2006, 42, 630–634. [Google Scholar] [CrossRef]
  37. Ivanova, E.P.; Nguyen, S.H.; Guo, Y.; Baulin, V.; Webb, H.; Truong, V.K.; Wandiyanto, J.V.; Garvey, C.J.; Mahon, P.; Mainwaring, D.E.; et al. Bactericidal activity of self-assembled palmitic and stearic fatty acid crystals on highly ordered pyrolytic graphite. Acta Biomater. 2017, 59, 148–157. [Google Scholar] [CrossRef]
  38. Maruthi, R.; Chandan, R.; Barath, M.; Datta, G.N.; D’Silva, M.; Kumari, M.K.; Ahmad, F.; Geetha, R. Analytical method development and validation of teneligliptin by rp-uflc. Res. J. Pharm. Technol. 2020, 13, 4035. [Google Scholar] [CrossRef]
  39. PCIDB. Available online: https://www.genome.jp/db/pcidb (accessed on 19 March 2021).
  40. Pédeboscq, S.; Rey, C.; Petit, M.; Harpey, C.; De Giorgi, F.; Ichas, F.; Lartigue, L. Non-Antioxidant Properties of α-Tocopherol Reduce the Anticancer Activity of Several Protein Kinase Inhibitors In Vitro. PLoS ONE 2012, 7, e36811. [Google Scholar] [CrossRef] [PubMed]
  41. Oh, K.K.; Adnan, M.; Ju, I.; Cho, D.H. A network pharmacology study on main chemical compounds from Hibiscus cannabinus L. leaves. RSC Adv. 2021, 11, 11062–11082. [Google Scholar] [CrossRef]
  42. Zhong, R.-F.; Xu, G.-B.; Wang, Z.; Wang, A.-M.; Guan, H.-Y.; Li, J.; He, X.; Liu, J.-H.; Zhou, M.; Li, Y.-J.; et al. Identification of anti-inflammatory constituents from Kalimeris indica with UHPLC-ESI-Q-TOF-MS/MS and GC–MS. J. Ethnopharmacol. 2015, 165, 39–45. [Google Scholar] [CrossRef]
  43. Wonkam, A.K.N.; Ngansop, C.A.N.; Wouamba, S.C.N.; Jouda, J.B.; Happi, G.M.; Boyom, F.F.; Sewald, N.; Lenta, B.N. Rothmanniamide and other constituents from the leaves of Rothmannia hispida (K.Schum.) fagerl. (Rubiaceae) and their chemophenetic significance. Biochem. Syst. Ecol. 2020, 93, 104137. [Google Scholar] [CrossRef]
  44. Kühnl, A.; Musiol, A.; Heitzig, N.; Johnson, D.E.; Ehrhardt, C.; Grewal, T.; Gerke, V.; Ludwig, S.; Rescher, U. Late Endosomal/Lysosomal Cholesterol Accumulation Is a Host Cell-Protective Mechanism Inhibiting Endosomal Escape of Influenza A Virus. mBio 2018, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Mohan, R.; Wilson, M.; Gorham, R.; Harrison, R.E.S.; Morikis, V.A.; Kieslich, C.; Orr, A.A.; Coley, A.V.; Tamamis, P.; Morikis, D. Virtual Screening of Chemical Compounds for Discovery of Complement C3 Ligands. ACS Omega 2018, 3, 6427–6438. [Google Scholar] [CrossRef]
  46. Liu, R.; Chen, Y.; Liu, G.; Li, C.; Song, Y.; Cao, Z.; Li, W.; Hu, J.; Lu, C.; Liu, Y. PI3K/AKT pathway as a key link modulates the multidrug resistance of cancers. Cell Death Dis. 2020, 11, 1–12. [Google Scholar] [CrossRef] [PubMed]
  47. Liu, L.-X.; Liu, Z.-H.; Jiang, H.-C.; Qi, S.-Y.; Zhang, W.-H.; Zhu, A.-L.; Wang, X.-Q.; Wu, M. Overexpression of Akt-1 gene in human hepatocellular carcinoma. Chin. J. Cancer Res. 2002, 14, 161–164. [Google Scholar] [CrossRef]
  48. Di Lorenzo, A.; Fernandez-Hernando, C.; Cirino, G.; Sessa, W.C. Akt1 is critical for acute inflammation and histamine-mediated vascular leakage. Proc. Natl. Acad. Sci. USA 2009, 106, 14552–14557. [Google Scholar] [CrossRef] [Green Version]
  49. Chenniappan, J.; Sankaranarayanan, A.; Arjunan, S. Evaluation of Antimicrobial Activity of Cissus quadrangularis L. stem extracts against Avian Pathogens and Determination of its Bioactive Constituents using GC-MS. J. Sci. Res. 2020, 64, 90–96. [Google Scholar] [CrossRef]
  50. Sharma, T.; Jana, S. Boswellic acids as natural anticancer medicine: Precious gift to humankind. J. Herb. Med. 2020, 20, 100313. [Google Scholar] [CrossRef]
  51. Fuentes, E.; Guzmán-Jofre, L.; Moore-Carrasco, R.; Palomo, I. Role of PPARs in inflammatory processes associated with metabolic syndrome (Review). Mol. Med. Rep. 2013, 8, 1611–1616. [Google Scholar] [CrossRef] [Green Version]
  52. Grabacka, M.; Reiss, K. Anticancer Properties of PPARα-Effects on Cellular Metabolism and Inflammation. PPAR Res. 2008, 2008, 930705. [Google Scholar] [CrossRef] [Green Version]
  53. Kaminska, B. MAPK signalling pathways as molecular targets for anti-inflammatory therapy—From molecular mechanisms to therapeutic benefits. In Proceedings of the Biochimica et Biophysica Acta—Proteins and Proteomics, Warsaw, Poland, 25–29 June 2005; pp. 253–262. [Google Scholar]
  54. Qian, C.; Qi, Y.; Zhong, S.; Zeng, J.; Chen, X.; Yao, J. Mitogen-activated protein kinase inhibition enhances the antitumor effects of sporamin in human pancreatic cancer cells. Oncol. Lett. 2018, 16, 1237–1242. [Google Scholar] [CrossRef] [Green Version]
  55. Cai, Y.; Sukhova, G.K.; Wong, H.K.; Xu, A.; Tergaonkar, V.; Vanhoutte, P.M.; Tang, E.H.C. Rap1 induces cytokine production in pro-inflammatory macrophages through NFκB signaling and is highly expressed in human atherosclerotic lesions. Cell Cycle 2015, 14, 3580–3592. [Google Scholar] [CrossRef] [Green Version]
  56. Gao, W.; Ye, G.; Liu, L.; Wei, L. The downregulation of Rap1 GTPase-activating protein is associated with a poor prognosis in colorectal cancer and may impact on tumor progression. Oncol. Lett. 2018, 15, 7661–7668. [Google Scholar] [CrossRef]
  57. Dalal, P.J.; Muller, W.A.; Sullivan, D.P. Endothelial Cell Calcium Signaling during Barrier Function and Inflammation. Am. J. Pathol. 2020, 190, 535–542. [Google Scholar] [CrossRef] [Green Version]
  58. Bong, A.H.; Monteith, G.R. Calcium signaling and the therapeutic targeting of cancer cells. Biochim. Biophys. Acta (BBA)—Mol. Cell Res. 2018, 1865, 1786–1794. [Google Scholar] [CrossRef]
  59. Erdogan, S.; Aslantas, O.; Celik, S.; Atik, E. The effects of increased cAMP content on inflammation, oxidative stress and PDE4 transcripts during Brucella melitensis infection. Res. Vet. Sci. 2008, 84, 18–25. [Google Scholar] [CrossRef]
  60. Zou, T.; Liu, J.; She, L.; Chen, J.; Zhu, T.; Yin, J.; Li, X.; Li, X.; Zhou, H.; Liu, Z. A perspective profile of ADCY1 in cAMP signaling with drug-resistance in lung cancer. J. Cancer 2019, 10, 6848–6857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Gaspar, J.M.; Velloso, L.A. Hypoxia inducible factor as a central regulator of metabolism—Implications for the development of obesity. Front. Neurosci. 2018, 12, 813. [Google Scholar] [CrossRef] [PubMed]
  62. Semenza, G.L. Evaluation of HIF-1 inhibitors as anticancer agents. Drug Discov. Today 2007, 12, 853–859. [Google Scholar] [CrossRef] [PubMed]
  63. Nixon, G.F. Sphingolipids in inflammation: Pathological implications and potential therapeutic targets. Br. J. Pharmacol. 2009, 158, 982–993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Ponnusamy, S.; Meyers-Needham, M.; Senkal, C.E.; Saddoughi, S.A.; Sentelle, D.; Selvam, S.P.; Salas, A.; Ogretmen, B. Sphingolipids and cancer: Ceramide and sphingosine-1-phosphate in the regulation of cell death and drug resistance. Futur. Oncol. 2010, 6, 1603–1624. [Google Scholar] [CrossRef] [Green Version]
  65. Kang, D.W.; Choi, K.-Y.; Min, D.S. Functional Regulation of Phospholipase D Expression in Cancer and Inflammation. J. Biol. Chem. 2014, 289, 22575–22582. [Google Scholar] [CrossRef] [Green Version]
  66. Cho, J.H.; Han, J.-S. Phospholipase D and Its Essential Role in Cancer. Mol. Cells 2017, 40, 805–813. [Google Scholar] [CrossRef]
  67. Hawkins, P.; Stephens, L. PI3K signalling in inflammation. Biochim. Biophys. Acta (BBA)—Mol. Cell Biol. Lipids 2015, 1851, 882–897. [Google Scholar] [CrossRef] [Green Version]
  68. Yang, J.; Nie, J.; Ma, X.; Wei, Y.; Peng, Y.; Wei, X. Targeting PI3K in cancer: Mechanisms and advances in clinical trials. Mol. Cancer 2019, 18, 1–28. [Google Scholar] [CrossRef] [Green Version]
  69. Chen, X.; Li, X.; Zhang, W.; He, J.; Xu, B.; Lei, B.; Wang, Z.; Cates, C.; Rousselle, T.; Li, J. Activation of AMPK inhibits inflammatory response during hypoxia and reoxygenation through modulating JNK-mediated NF-κB pathway. Metabolism 2018, 83, 256–270. [Google Scholar] [CrossRef] [PubMed]
  70. Chuang, H.-C.; Chou, C.-C.; Kulp, S.; Chen, C.-S. AMPK as a Potential Anticancer Target—Friend or Foe? Curr. Pharm. Des. 2014, 20, 2607–2618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Carmeliet, P. VEGF as a Key Mediator of Angiogenesis in Cancer. Oncology 2005, 69, 4–10. [Google Scholar] [CrossRef] [PubMed]
  72. Zeng, Q.-H.; Wei, Y.; Lao, X.-M.; Chen, D.-P.; Huang, C.-X.; Lin, Q.-Y.; He, M.; Liao, Y.; Zheng, L.; Li, B.; et al. B cells polarize pathogenic inflammatory T helper subsets through ICOSL-dependent glycolysis. Sci. Adv. 2020, 6, eabb6296. [Google Scholar] [CrossRef] [PubMed]
  73. Burger, J.A.; Wiestner, A. Targeting B cell receptor signalling in cancer: Preclinical and clinical advances. Nat. Rev. Cancer 2018, 18, 148–167. [Google Scholar] [CrossRef]
  74. Luiten, R.M.; Warnaar, S.O.; Schuurman, J.; Pasmans, S.G.; Latour, S.; Daëron, M.; Fleuren, G.J.; Litvinov, S.V. Chimeric immunoglobulin E reactive with tumor-associated antigen activates human FcεRI bearing cells. Hum. Antibodies 1997, 8, 169–180. [Google Scholar] [CrossRef]
  75. Arcidiacono, B.; Iiritano, S.; Nocera, A.; Possidente, K.; Nevolo, M.T.; Ventura, V.; Foti, D.; Chiefari, E.; Brunetti, A. Insulin Resistance and Cancer Risk: An Overview of the Pathogenetic Mechanisms. Exp. Diabetes Res. 2012, 2012, 1–12. [Google Scholar] [CrossRef] [Green Version]
  76. Monteiro, R.; Teixeira, D.; Calhau, C. Estrogen Signaling in Metabolic Inflammation. Mediat. Inflamm. 2014, 2014, 615917. [Google Scholar] [CrossRef] [Green Version]
  77. Orbach, H.; Shoenfeld, Y. Hyperprolactinemia and autoimmune diseases. Autoimmun. Rev. 2007, 6, 537–542. [Google Scholar] [CrossRef] [PubMed]
  78. Sethi, B.K.; Chanukya, G.; Nagesh, V.S. Prolactin and cancer: Has the orphan finally found a home? Indian J. Endocrinol. Metab. 2012, 16, S195–S198. [Google Scholar] [CrossRef] [PubMed]
  79. van der Spek, A.H.; Fliers, E.; Boelen, A. Thyroid hormone in inflammation. Endocr. Abstr. 2019, 65. [Google Scholar] [CrossRef]
  80. Liu, C.; Zhao, Q.; Yu, X. Bone Marrow Adipocytes, Adipocytokines, and Breast Cancer Cells: Novel Implications in Bone Metastasis of Breast Cancer. Front. Oncol. 2020, 10, 561595. [Google Scholar] [CrossRef] [PubMed]
  81. Thanasupawat, T.; Glogowska, A.; Nivedita-Krishnan, S.; Wilson, B.; Klonisch, T.; Hombach-Klonisch, S. Emerging roles for the relaxin/RXFP1 system in cancer therapy. Mol. Cell. Endocrinol. 2019, 487, 85–93. [Google Scholar] [CrossRef] [PubMed]
  82. Logan, S.M.; Storey, K.B. Pro-inflammatory AGE-RAGE signaling is activated during arousal from hibernation in ground squirrel adipose. PeerJ 2018, 6, e4911. [Google Scholar] [CrossRef] [PubMed]
  83. El-Far, A.; Munesue, S.; Harashima, A.; Sato, A.; Shindo, M.; Nakajima, S.; Inada, M.; Tanaka, M.; Takeuchi, A.; Tsuchiya, H.; et al. In vitro anticancer effects of a RAGE inhibitor discovered using a structure-based drug design system. Oncol. Lett. 2018, 15, 4627–4634. [Google Scholar] [CrossRef]
  84. Oh, K.K.; Adnan, M.; Cho, D.H. Active ingredients and mechanisms of Phellinus linteus (grown on Rosa multiflora) for alleviation of Type 2 diabetes mellitus through network pharmacology. Gene 2021, 768, 145320. [Google Scholar] [CrossRef]
  85. Keiser, M.; Roth, B.L.; Armbruster, B.N.; Ernsberger, P.; Irwin, J.; Shoichet, B.K. Relating protein pharmacology by ligand chemistry. Nat. Biotechnol. 2007, 25, 197–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Daina, A.; Michielin, O.; Zoete, V. SwissTargetPrediction: Updated data and new features for efficient prediction of protein targets of small molecules. Nucleic Acids Res. 2019, 47, W357–W364. [Google Scholar] [CrossRef] [Green Version]
  87. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Morris, G.M.; Huey, R.; Lindstrom, W.; Sanner, M.F.; Belew, R.K.; Goodsell, D.S.; Olson, A.J. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J. Comput. Chem. 2009, 30, 2785–2791. [Google Scholar] [CrossRef] [Green Version]
  89. Khanal, P.; Patil, B.M.; Chand, J.; Naaz, Y. Anthraquinone Derivatives as an Immune Booster and their Therapeutic Option Against COVID-19. Nat. Prod. Bioprospect. 2020, 10, 325–335. [Google Scholar] [CrossRef]
  90. Laskowski, R.A.; Swindells, M.B. LigPlot+: Multiple ligand-protein interaction diagrams for drug discovery. ACS Pub. 2011, 51, 2778–2786. [Google Scholar] [CrossRef]
  91. Yang, H.; Lou, C.; Sun, L.; Li, J.; Cai, Y.; Wang, Z.; Li, W.; Liu, G.; Tang, Y. admetSAR 2.0: Web-service for prediction and optimization of chemical ADMET properties. Bioinformatics 2018, 35, 1067–1069. [Google Scholar] [CrossRef]
Figure 1. Workflow diagram of network pharmacology analysis of the C. maackii flower against cancer.
Figure 1. Workflow diagram of network pharmacology analysis of the C. maackii flower against cancer.
Molecules 26 05904 g001
Figure 2. GC-MS peak of the C. maackii flower MeOH extract and an indication of the uppermost bioactive.
Figure 2. GC-MS peak of the C. maackii flower MeOH extract and an indication of the uppermost bioactive.
Molecules 26 05904 g002
Figure 3. (A) Overlapping targets (108 targets) between SEA (309 targets) and STP (396 targets). (B) Overlapping targets (51 targets) between 108 overlapping targets from two databases (SEA and STP) and cancer associated with targets (4245 targets).
Figure 3. (A) Overlapping targets (108 targets) between SEA (309 targets) and STP (396 targets). (B) Overlapping targets (51 targets) between 108 overlapping targets from two databases (SEA and STP) and cancer associated with targets (4245 targets).
Molecules 26 05904 g003aMolecules 26 05904 g003b
Figure 4. PPI networks (46 nodes, 145 edges). The size of circles represents the degree of values.
Figure 4. PPI networks (46 nodes, 145 edges). The size of circles represents the degree of values.
Molecules 26 05904 g004
Figure 5. Bubble chart of 20 signaling pathways connected to cancer.
Figure 5. Bubble chart of 20 signaling pathways connected to cancer.
Molecules 26 05904 g005
Figure 6. S–T–B networks (63 nodes, 204 edges). Green rectangle: Signaling pathway; pink triangle: Target protein; orange circle: Bioactive.
Figure 6. S–T–B networks (63 nodes, 204 edges). Green rectangle: Signaling pathway; pink triangle: Target protein; orange circle: Bioactive.
Molecules 26 05904 g006
Figure 7. MDT of Urs-12-en-24-oic acid, methyl ester on Akt1 (PDB ID: 5KCV).
Figure 7. MDT of Urs-12-en-24-oic acid, methyl ester on Akt1 (PDB ID: 5KCV).
Molecules 26 05904 g007
Table 1. A list of the 34 bioactives identified from C. maackii through GC-MS and profiling of bioactivities.
Table 1. A list of the 34 bioactives identified from C. maackii through GC-MS and profiling of bioactivities.
No.CompoundsPubchem IDRT (mins)Area (%)Pharmacological Activities (References)
1Glyceraldehyde7513.510 0.91No reported
26-Methyluracil122834.279 0.88No reported
33-Hydroxy-2,3-dihydromaltol1198384.789 5.19No reported
4Pyranopyridine5340335.414 0.69No reported
5Formicin693656.096, 6.2120.58No reported
64-Nitro-2-picoline N-oxide952916.721 0.6No reported
72-Vinyl-9-[.beta.-d-ribofuranosyl]hypoxanthine1354930116.933 1.35Antibacterial and antifungal activity [32]
82-Chloromethyl-3,3-dichloropropene434927.058 0.51No reported
9d-Lyxo-d-manno-nononic-1,4-lactone5355567.183 0.43No reported
10Pentyl isobutyrate755547.327 0.26Antimicrobial activity [33]
112-Methyl-3-nitrosooxazolidine383577.673 0.23No reported
12N-Nitrosomethylethanolamine336467.808 2.71No reported
131,4,7,10-Tetraoxacyclododecan-2-one5336468.145 0.15No reported
144-methylpyrimidin-2-ol4070918.231 0.58No reported
15Diethyl malonate77618.750 0.46No reported
16Palmitic acid9858.943, 9.443, 9.760, 10.1064.03Anti-inflammation [34]
17Methyl 3,6-anhydrohexopyranoside #916913849.154 0.65No reported
183-Hydroxypropionic acid681529.625 1.17Antibacterial activity [35]
192,2,4-Trichloro-1,3-cyclopentenedione1507579.846 0.65No reported
20Isopropyl palmitate890710.673 0.54Antibacterial activity [36]
21Stearic acid528110.933 0.72Antibacterial activity [37]
22Pregn-5,7-diene-3-ol-20-one2111740311.068 0.25No reported
239-Heptadecanone1088711.173 0.95Antibacterial activity [38]
24Allyl stearate8050012.087 1.05No reported
25Squalene63807212.250 0.54Anti-oxidant [39]
26Prexanthoperol62874213.952 13.65No reported
27α-Tocopherol1498514.664 0.94Anticancer [40]
28β-Stigmasterol643274516.289 1.68Anti-inflammation [39]
294,4-Dimethylcholest-7-en-3-ol546007617.077 1.06No reported
30Aristol-9-en-8-one #643265117.837 1.11No reported [39]
31α-Amyrin7317018.587 2.53Anti-obesity [41]
32Urs-12-en-24-oic acid, 3-oxo-, methyl ester61282219.125 9.04Potential Anti-inflammation [42]
33Lupenyl acetate32307420.019 24.42Antibacterial activity [43]
34Cholesterol599721.693, 21.95215.19Protective cell against pathogens [44]
PCIDB: PhytoChemical Interactions DB.
Table 2. Physicochemical properties of bioactives for good oral bioavailability and cell membrane permeability.
Table 2. Physicochemical properties of bioactives for good oral bioavailability and cell membrane permeability.
CompoundsLipinski RulesLipinski’s ViolationsBioavailability ScoreTPSA (Å2)
MWHBAHBDMLog P
No.<500<10≤5≤4.15≤1>0.1<140
1Glyceraldehyde90.0832−1.6600.5557.53
26-Methyluracil126.1122−0.3900.5565.72
33-Hydroxy-2,3-dihydromaltol144.1342−1.7700.8566.76
4Pyranopyridine133.15200.7300.5522.12
5Formicin89.0922−0.8500.5549.33
64-Nitro-2-picoline N-oxide154.1230−0.1500.5571.28
72-Vinyl-9-[.beta.-d-ribofuranosyl]hypoxanthine294.2674−1.7700.55133.49
82-Chloromethyl-3,3-dichloropropene159.44002.8100.550.00
9d-Lyxo-d-manno-nononic-1,4-lactone268.2297−3.8510.55167.91
10Pentyl isobutyrate158.24202.2800.5526.30
112-Methyl-3-nitrosooxazolidine116.1230−0.4200.5541.90
12N-Nitrosomethylethanolamine104.1131−0.8900.5552.90
131,4,7,10-Tetraoxacyclododecan-2-one190.1950−0.9900.5553.99
144-methylpyrimidin-2-ol110.1121−0.2700.5545.75
15Diethyl malonate160.17400.600.5552.60
16Palmitic acid256.42214.1910.8537.30
17Methyl 3,6-anhydrohexopyranoside #176.1752−1.5900.5568.15
183-Hydroxypropionic acid90.0832−0.8500.8557.53
192,2,4-Trichloro-1,3-cyclopentenedione199.42200.5500.5534.14
20Isopropyl palmitate298.5204.9110.5526.30
21Stearic acid284.48214.6710.8537.30
22Pregn-5,7-diene-3-ol-20-one314.46213.9500.5537.30
239-Heptadecanone254.45104.5510.5517.07
24Allyl stearate324.54205.2510.5526.30
25Squalene410.72007.9310.550.00
26Prexanthoperol314.42312.9400.5554.37
27Vitamin E430.71216.1410.5529.46
28β-Stigmasterol412.69116.6210.5520.23
294,4-Dimethylcholest-7-en-3-ol414.71116.7310.5520.23
30Aristol-9-en-8-one #218.33103.5600.5517.07
31α-Amyrin426.72116.9210.5520.23
32Urs-12-en-24-oic acid, 3-oxo-, methyl ester468.71305.9210.5543.37
33Lupenyl acetate144.88207.0810.5526.30
34Cholesterol386.65116.3410.5520.23
MW, Molecular Weight (g/mol); HBA, Hydrogen Bond Acceptor; HBD, Hydrogen Bond Donor; MLog P, Lipophilicity; Bioavailability Score, the ability of a drug or other substance to be absorbed and used by the body.
Table 3. The degree value of PPI networks.
Table 3. The degree value of PPI networks.
No.TargetDegreeNo.TargetDegree
1AKT12924MGLL5
2VEGFA2125S1PR15
3ESR11626SHH5
4AR1327VDR5
5CYP19A11128TOP2A5
6PPARG1129ADORA2B4
7IL1B1030PTGER24
8CNR1931S1PR34
9TRPV1932ADORA2A3
10PPARA933CYP24A13
11PTPN1934FOLH13
12HMGCR835PTPN63
13ESR2736CA22
14PRKCA737FABP32
15ABCB1638NR1H32
16CDC25B639PHLPP12
17PTGER4640PTPRF2
18SHBG641PRKCE2
19CYP17A1642PTGFR2
20SRD5A2643CA11
21ADORA3544FNTB1
22AKR1B1545NR1H41
23CDC25A546IMPDH21
Table 4. The degree value of S–T–B networks.
Table 4. The degree value of S–T–B networks.
No.TargetDegreeBioactiveDegreeNo.TargetDegreeBioactiveDegree
1AKT118Urs-12-en-24-oic acid,3-oxo-, methyl ester3613S1PR11Cholesterol14
2PRKCA12Palmitic acid2714S1PR31Pregn-5,7-diene-3-ol-20-one10
3VEGFA7Stearic acid2615PTPN61Methyl 3,6-anhydro hexopyranoside #7
4PPARA3α-Tocopherol2616CNR11Aristol-9-en-8-one #7
5ESR13Isopropyl palmitate2617HMGCR1Squalene3
6PPARG2Allyl stearate2518PTPN11Lupenyl acetate3
7ESR22α-Amyrin2119PTPRF12-Vinyl-9- [β-d-ribofuranosyl] hypoxanthine1
8PRKCE29-Heptadecanone2120CDC25B1
9IL1B2β-Stigmasterol2021PTGFR1
10NR1H314,4-Dimethyl-cholest-7-en-3-ol2022ADORA2B1
11FABP31Pentyl isobutyrate1723PTGER21
12CYP17A11d-Lyxo-d-manno-nononic-1,4-lactone1524PHLPP11
Table 5. Targets in 20 signaling pathways’ enrichment related to cancer.
Table 5. Targets in 20 signaling pathways’ enrichment related to cancer.
KEGG ID & DescriptionTargetsFalse Discovery Rate
hsa03320: PPAR signaling pathwayPPARA, PPARD, PPARG, NR1H3, FABP30.0000695
hsa04917: Prolactin signaling pathwayAKT1, ESR1, ESR2, CYP17A10.00063
hsa04933: AGE-RAGE signaling pathway in diabetic complicationsAKT1, VEGFA, PRKCA, PRKCE, IL1B0.00018
hsa04370: VEGF signaling pathwayAKT1, VEGFA, PRKCA0.0068
hsa04071: Sphingolipid signaling pathwayAKT1, VEGFA, PRKCE, S1PR1, S1PR30.00027
hsa04066: HIF-1 signaling pathwayAKT1, VEGFA, PRKCA0.016
hsa04664: Fc epsilon RI signaling pathwayAKT1, PRKCA0.0492
hsa04920: Adipocytokine signaling pathwayAKT1, PPARA0.0492
hsa04662: B cell receptor signaling pathwayAKT1, PTPN60.0492
hsa04919: Thyroid signaling pathwayAKT1, ESR1, PRKCA0.0236
hsa04015: Rap1 signaling pathwayAKT1, VEGFA, PRKCA, VEGFA, ADORA2B0.003
hsa04152: AMPK signaling pathwayAKT1, PPARG, HMGCR0.025
hsa04926: Relaxin signaling pathwayAKT1, VEGFA, PRKCA0.0278
hsa04915: Estrogen signaling pathwayAKT1, ESR1, ESR20.0279
hsa04910: Insulin signaling pathwayAKT1, PTPN1, PTPRF0.0279
hsa04072: Phospholipase D signaling pathwayAKT1, PRKCA0.0324
hsa04010: MAPK signaling pathwayAKT1, VEGFA, PRKCA, IL1B0.0103
hsa04020: Calcium signaling pathwayPRKCA, ADORA2B0.0457
hsa04024: cAMP signaling pathwayAKT1, PPARA, PTGER20.0492
hsa04151: PI3K-Akt signaling pathwayAKT1, VEGFA, PRKCA, PHLPP10.0487
Table 6. Binding energy of potential bioactives on Akt1 (PDB ID: 5KCV).
Table 6. Binding energy of potential bioactives on Akt1 (PDB ID: 5KCV).
Hydrogen Bond InteractionsHydrophobic Interactions
ProteinLigandPubChem IDBinding Energy (kcal/mol)Amino Acid ResidueR Group(s) Involved in Hydrogen Bonding Distance (Å)Amino Acid Residue
5KCVUrs-12-en-24-oic acid, 3-oxo-, methyl ester612822−12.8N/AN/AN/AGln59, Lys268, Asn53
Trp80, Leu202, Leu78
α-Tocopherol14985−5.8Leu78R-OH2.71Ala58, Val270, Lys268
Val201, Leu202, Gln203
Trp80, Asn53
Table 7. Binding energy of potential bioactives on VEGFA (PDB ID: 3V2A).
Table 7. Binding energy of potential bioactives on VEGFA (PDB ID: 3V2A).
Hydrogen Bond InteractionsHydrophobic Interactions
ProteinLigandPubChem IDBinding Energy (kcal/mol)Amino Acid ResidueR Group(s) Involved in Hydrogen Bonding Distance (Å)Amino Acid Residue
3V2AAllyl stearate80500−5.1Phe47RCOOR′3.18Ile46, Lys48, Lys286
Asp276, Arg275, Pro40
Phe36
Isopropyl palmitate8907−5.1N/AN/AN/AAsp276, Asp34, Ile46
Phe47, Lys48, Lys286
Phe36, Pro40
Methyl 3,6-anhydro hexopyranoside #91691384−5.2Ser310, Pro85R-O-R′, R-OH2.98, 3.23, 2.82Lys84, Gln87, Gly312
Asp257, Gly255, Ile256
Glu44
Table 8. Binding energy of potential bioactives on PRKCA (PDB ID: 3IW4).
Table 8. Binding energy of potential bioactives on PRKCA (PDB ID: 3IW4).
Hydrogen Bond Interactions Hydrophobic Interactions
ProteinLigandPubChem IDBinding Energy (kcal/mol)Amino Acid ResidueR Group(s) Involved in Hydrogen Bonding Distance (Å)Amino Acid Residue
3IW4Allyl stearate80500−6.2Lys 396RCOOR′3.21Asp395, Leu393, Pro397
Arg608, Pro398, Val664
Pro666, Ser473, Glu474
His665, Lys478, Ile667
Gln402, Asn660
Isopropyl palmitate8907−6.3Asn660RCOOR′2.92Asp395, Leu393, Pro397
Lys396, Gln402,Pro398
Arg608, Ile667, Pro666
Val664, His665, Lys478
N/AN/AN/APro502, Gln650, Ile645
Pentyl isobutyrate75554−5.0 Gly540, Asp542, Asp503
Asp539, Gln642, Leu546
Glu543
9-Heptadecanone10887−5.4Lys478RCOR′2.80 Asn607, Gln548, Pro398
Arg608, Ile667, Tyr419
Asn421, Ser473, Pro666
Glu418, Asp472, His665
Val664, Gln402, Glu552
Palmitic acid985−4.8Asn660, Lys396RCOR′, R-OH2.93, 2.95Val664, Pro398, Glu552
Gln662, Asp395, Leu393
Leu394, Gln402, Pro397
Stearic acid5281−6.2Lys396, Leu393R-OH3.04, 2.95Leu394, Gln402, Pro398
Lys478, His665, Ile667
Pro666, Val664, Arg608
Pro397, Asn660
d-Lyxo-d-manno-nononic- 1,4-lactone535556−6.9Leu393, Lys396, Pro397RCOOR′, R-OH3.30, 2.73, 3.31Leu394, Asp395, Gln662
Gln402 3.23, 3.19, 2.66Asn660, Pro398
3.08, 3.03, 3.15
3.26, 2.88
Table 9. Binding energy of the positive controls on Akt1 (PDB ID: 5KCV).
Table 9. Binding energy of the positive controls on Akt1 (PDB ID: 5KCV).
Hydrogen Bond Interactions Hydrophobic Interactions
ProteinLigandPubChem IDBinding Energy (kcal/mol)Amino Acid ResidueR Group(s) Involved in Hydrogen Boding Distance (Å)Amino Acid Residue
5KCVMK-2206 dihydrochloride46930998−7.8N/AN/AN/ALeu78, Leu202, Gln203
Lys268, Trp80, Ala58
Gln59
Gsk-69069316725726−7.4Tyr18, Asp274RNR′,RNHH3.18, 3.06Glu85, Phe161, Val83
Cys296, Gly294, Phe293
Lys276, Leu316, Pro313
Arg273, Leu295, Ile84
Ipatasertib24788740−7.3N/AN/AN/ALys268, Asn53, Gln79
Ala58, Phe225, Leu223
Leu202, Leu78, Trp80
Capivasertib25227436−7.1Val83ROH2.95Ile84, Arg273, Asn279
Asp274, Thr291, Glu278
Phe293, Gly294, Cys296
Glu85, Glu17
AT786711175137−7.6Ser56RNH3.04Lys268, Trp80, Asn53
Leu78, Cys60, Gln79
Ala58, Cys77, Val201
Leu202, Gln203
A-67456311314340−7.6Gln79, Gln203RNHH, RNH3.16, 3.20Ser56, Trp80, Leu202
Lys268, Asn53, Ala58
Leu78
Miransertib53262401−9.0Gln203, Leu78RNHH3.19, 3.22Lys268, Trp80, Leu202
Phe225, Ser216, Val201
Gln59, Gln79, Ala58
BAY112597670817911−9.1Asp274RNHH3.29Gly294, Thr291, Glu278
Asn279, Tyr229, Leu156
Glu234, Phe293, Lys158
Cys296
Akti-1/2135398501−8.9Cys296, Arg15RCOR′, RNR′3.03, 3.15, 3.19Glu278, Gly294, Leu295
Val83, Gln85, Glu17
Tyr18, Ile84, Phe293
Asn279
Uprosertib51042438−6.8Cys296, Leu295, Glu278RCOR′, RCOR′, RNHH2.93, 2.86, 2.93Tyr18, Arg273, Asp274
Lys276, Thr291, Phe293
Asn279, Gly294, Ile84
Thr82
Afuresertib46843057−6.9Gly394, Gly395RNHH3.10, 2.95Ala50, Arg328, Lys389
Pro388, Ala329, Asp325
Gly327, Tyr326, Phe55
Ile36, Leu52, Pro51
AT1314824905401−6.9Glu341, Tyr315, His354ROR′, ROR′, RNR′3.01, 2.70, 3.14Phe236, Glu278, Leu347
Pro313, Tyr350, Glu314
Oridonin5321010−8.3Asp325, Ala50, Arg328RCOR′, ROH, ROH, ROH2.67, 2.71, 2.80, 3.24Phe55, Ile36, Leu52
Gly394, Ala329, Gly327
Tyr326
Table 10. Toxicological properties of the uppermost bioactive on AKT1 (PDB ID: 5KCV) in MDT.
Table 10. Toxicological properties of the uppermost bioactive on AKT1 (PDB ID: 5KCV) in MDT.
ParametersCompound Name
Urs-12-en-24-oic acid, 3-oxo-, Methyl Ester
Ames toxicityNAT
CarcinogensNC
Acute oral toxicity
Rat acute toxicity2.1675
AT: Ames toxic; NAT: Non-Ames toxic; NC: Non-carcinogenic; Category-II means (50 mg/kg > LD50 < 500 mg/kg); Category-III means (500 mg/kg > LD50 < 5000 mg/kg).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Oh, K.-K.; Adnan, M.; Cho, D.-H. Network Pharmacology-Based Study to Uncover Potential Pharmacological Mechanisms of Korean Thistle (Cirsium japonicum var. maackii (Maxim.) Matsum.) Flower against Cancer. Molecules 2021, 26, 5904. https://doi.org/10.3390/molecules26195904

AMA Style

Oh K-K, Adnan M, Cho D-H. Network Pharmacology-Based Study to Uncover Potential Pharmacological Mechanisms of Korean Thistle (Cirsium japonicum var. maackii (Maxim.) Matsum.) Flower against Cancer. Molecules. 2021; 26(19):5904. https://doi.org/10.3390/molecules26195904

Chicago/Turabian Style

Oh, Ki-Kwang, Md. Adnan, and Dong-Ha Cho. 2021. "Network Pharmacology-Based Study to Uncover Potential Pharmacological Mechanisms of Korean Thistle (Cirsium japonicum var. maackii (Maxim.) Matsum.) Flower against Cancer" Molecules 26, no. 19: 5904. https://doi.org/10.3390/molecules26195904

Article Metrics

Back to TopTop