- Research
- Open access
- Published:
Essential oils from Amorpha fruticosa against hepatocellular carcinoma based on network pharmacology
BMC Complementary Medicine and Therapies volume 25, Article number: 29 (2025)
Abstract
Background
Amorpha fruticosa was used for treating burn, ambustion, carbuncle, and eczema in the traditional Chinese medicine. Although more and more attention has been paid to its biological activity recently, the antitumor activities of the essential oils (EOs) extracted from its leaves (AFLEO) and flowers (AFFEO), and their molecular mechanisms have never been reported up to now. The objective of present study was to examine the chemical compositions of AFLEO and AFFEO, then investigate the effects and pharmacological mechanism of EOs against hepatocellular carcinoma (HCC).
Methods
The chemical compositions of EOs were examined using gas chromatography-mass spectrometry (GC-MS). The inhibitory effect of the EOs on HCC was evaluated by MTT assay. The detected components of AFLEO and AFFEO were performed ADME screening to examine their drug-likeness. Then a PPI network, compound-target network, compound-target-pathway network, gene ontology, and KEGG enrichment for HCC were applied to identify the targets and pathways for AFLEO and AFFEO against HCC. Molecular docking of the main components and their targets was performed to predict the binding affinity. Western blotting was used to verify the results.
Results
30 components were identified from AFLEO, while 22 components from AFFEO. Both AFLEO and AFFEO inhibited the proliferation of HCC cells in a time and dose-dependent manner. 10 compounds of AFLEO and 9 compounds of AFFEO were screened out for further analysis. 28 hub targets of AFLEO and 40 hub targets of AFFEO were detected by PPI network. KEGG analysis revealed that pathways in cancer, chemical carcinogenesis - receptor activation and proteoglycans in cancer were related to the EOs against HCC. Molecular docking confirmed that the main component of the EOs has high affinity to the targets of HCC.
Conclusions
AFLEO and AFFEO may suppress HCC by acting on multiple targets and regulating multiple pathways.
Background
Hepatocellular carcinoma (HCC) is one of the most malignant and lethal cancers in the world. The most common cause of HCC is chronic viral infection, with about 80% of HCC cases attributed to chronic infection with hepatitis B and hepatitis C [1]. Currently, the clinically treatment strategies for HCC include surgery, chemotherapy, radiation therapy, molecular targeted therapy or their combination. Among them, chemotherapy is the standard treatment method for the advanced HCC [2]. However, due to the adverse reactions and drug resistance, the prognosis of HCC is still poor, and the five-year survival rate is only 18% [3]. In addition to the traditional malignant tumor treatments, exploring more effective and lower toxicity antitumor chemicals has become an important issue in the field of HCC diagnosis and treatment research.
Amorpha fruticosa L., which belongs to the genus Amorpha in Leguminosae family, contains flavonoids [4,5,6], stilbene glycosides [7], essential oil [8] and other chemical components. In the traditional Chinese medicine, Amorpha fruticosa was used for treating burn, ambustion, carbuncle, and eczema [9]. In recent years, more and more attention has been paid to its biological activity. Pharmacological research has demonstrated that the extracts of Amorpha fruticosa have potential medicinal values in anti-tumor [10,11,12], anti-inflammatory [13], liver protection [14] and antibacterial [5]. However, the research on the pharmacological activity of Amorpha fruticosa mainly focus on flavonoids. There is little literature on the compositions of the essential oils (EOs) extracted from the leaves (AFLEO) and flowers (AFFEO) of Amorpha fruticose [15], and the reported constituents are significantly different from what we detected possibly for the difference in geographical location. To the best of our knowledge, the antitumor activities of AFLEO and AFFEO and their molecular mechanisms have never been reported up to now.
In the present study, the fresh leaves and flowers of Amorpha fruticosa were collected first. Then the EOs of Amorpha fruticosa were prepared to identify their chemical compositions and antitumor activity. In addition, network pharmacology was used to predict potential processes and elucidate the molecular mechanisms for EOs of Amorpha fruticosa against HCC.
Materials and methods
Plant material
The fresh leaves and flowers of Amorpha fruticosa were purchased from Mingquan Amorpha fruticosa culture base, Henan Province of China in June 2021. The leaves and flowers were washed and air-dried at room temperature for 1 week.
Preparation of the essential oil
Air-dried Amorpha fruticosa leaves and flowers were crushed into powders. Approximately 150 g of each sample was submitted to hydrodistillation in a Clevenger-type apparatus at 100 ℃ for 5 h. The obtained EOs were extracted by ether and dried with anhydrous sodium sulfate. After filtration, pure oils were stored at 4℃ in dark.
Gas Chromatograph-Mass Spectrometry (GC-MS) analysis
The composition of EOs were identified by using an Agilent 7890 A gas chromatograph equipped with a HP-5MS fused silica capillary tubes column (30 m×250 μm×0.25 μm) connected to a 5975 C mass spectrometer (Agilent, Palo Alto, CA, U.S.A.). The carrier gas was Helium (purity > 99.999%) and the flow rate was 1 ml/min. The sample was diluted by hexane. The injection volume was 1 µl, and the split less mode was used. Temperature program was as follows: initial temperature of 40 ℃ for 5 min, increased 2.5 ℃/min up to 65 ℃ and held for 1 min. Then the temperature was raised to 200 ℃ at 8 °C/min and kept at that temperature for 2 min, finally increased to 250 ℃ at 10 °C/min, and maintained for 2 min. The injector and ion source temperature were 250 ℃ and 230 ℃. The ionization voltage was 70 eV, and the mass scan range 33–400 Da was used. Each component was identified according to its retention indices and mass spectra. Retention indices were calculated using a homologous series of C8-C20 n-alkanes injected under the same experimental conditions. Mass spectra were compared with the corresponding standard spectra in National Institute of Standards and Technology (NIST) library. The relative peak area of the components was calculated using the normalization method.
MTT assay
The Huh7 and HepG2 cells were cultured in DMEM medium supplemented with 10% FBS at 37℃ with a humid atmosphere containing 5% CO2. To perform MTT assay, Huh7 and HepG2 cells were seeded into a 96-well plate and for 48 h after treating with different concentrations of AFLEO and AFFEO (20, 50, 100, 200, 500 and 1000 µg/mL), respectively. Then, the cells were added 20 µl MTT and incubated for 4 h. After that, 100 µl of DMSO was added to dissolve the crystal, and the absorbance was measured at 490 nm by a microplate reader.
Identification of active ingredients and prediction of targets of AFLEO and AFFEO
SwissADME (http://www.swissadme.ch/) [16] was used to search for the active ingredients of AFLEO and AFFEO. An ingredient is considered to be active when it meets the conditions as follows: human gastrointestinal absorption (HIA) was “high”, blood-brain barrier (BBB) permeation was “yes”, and at least two models among five drug-likeness models (Lipinski, Ghose, Veber, Egan, and Muegge) were “yes”. Then SwissTargetPrediction database (http://www.swisstargetprediction.ch/) [17] was used to identify the protein targets of the active ingredients.
Protein targets related to HCC
To ensure comprehensive collection of genes associated with HCC, GeneCards (http://www.genecards.org/) [18] and DisGeNET (https://www.disgenet.org/) [19] were adopted to search for the HCC-related targets. Then the overlapping targets of AFLEO and HCC, as well as AFFEO and HCC, which were considered potential targets of AFLEO or AFFEO in the treatment of HCC, were identified by jvenn (https://jvenn.toulouse.inrae.fr/app/example.html) [20].
Construction of (protein–protein interaction) PPI Network
Direct and indirect interactions of the overlapping targets between AFLEO or AFFEO and HCC were identified by the Search Tool for the Retrieval of Interacting Genes (STRING) database (https://string-db.org/) [21]. The specie was “Homo sapiens” and the minimum required interaction score was 0.4. Then the PPI network was imported to Cytoscape 3.9.1 to evaluate its topological characteristics, including the degree, betweenness and closeness.
Gene Ontology (GO) and Kyoto Encyclopedia of genes and the genomes (KEGG) Enrichment analysis
GO and KEGG pathway enrichment analysis were performed using the Integrated Annotation and Discovery Visualization Database (DAVID) (https://david.ncifcrf.gov/) [22] and plotted by the online platform for data analysis and visualization (http://www.bioinformatics.com.cn/). GO functions and KEGG pathways with a p-value < 0.05 were considered as the main terms of clustering.
Network construction and analysis
The compound–target network and the compound–target–pathway network were constructed using Cytoscape 3.9.1 to analyze the complicated interconnection of compounds, targets and pathways.
Molecular docking
The 3D structure of nerolidol was obtained through the PubChem (https://pubchem.ncbi.nlm.nih.gov/). The Protein Data Bank (PDB, https://www.rcsb.org/) was used to obtain the 3D structures of its target proteins. Autodock vina (https://autodock.scripps.edu/) was used to dock active ingredients with target proteins. The binding energy less than − 5.0 kcal/mol was considered as tight binding.
Western blotting
After treated with AFLEO for 48 h, HCC cells were lysed in ice-cold RIPA lysis buffer (Leagene, China) for 30 min and then centrifuged for 10 min (12, 000 × g, 4 ℃). Protein concentration was detected using the BCA Quantitation Kit (Thermo Fisher Scientific, USA). 20 µg protein was run on 10% SDS-PAGE gel and transferred to polyvinylidene fluoride membranes (PVDF) (0.45 μm pore size, Millipore, USA). The membranes were blocked with 5% non-fat powdered milk for 2 h at 25 ℃. Then the membranes were incubated overnight at 4 ℃ with primary antibodies, including anti-GAPDH (1:10000, Proteintech, China), anti-AKT1 (1:1000, Cell Signaling Technology, USA), anti-PPARG (1:1000, Wanleibio, China), and anti-MAPK3 (1:1000, Abcam, USA), and then incubated with horseradish peroxidase (HRP)-conjugated goat anti-rabbit IgG (1:10000, Sangon Biotech, China) secondary antibody for 2 h at 25 ℃. The luminescence reaction was conducted with an ECL kit (Geneview, USA). GAPDH was used as internal control.
Statistical analysis
All the experiments were conducted independently in triplicate (n = 3). The data are expressed as the mean ± SD. All analyses were performed using Graphpad 6.0.
Results
Chemical composition of the EOs
Both AFLEO and AFFEO were light yellow color with special aroma. The yield (v/w) of them were 0.67% and 0.43%, respectively. The chemical components of EOs were identified by GC-MS. The chromatogram spectrum can be seen in Fig. S1. The results are shown in Table 1. 30 components were identified from AFLEO, and 22 components were identified from AFFEO. AFLEO consisted primarily of alcohols (51.49%), along with olefins (32.77%), ketones (1.46%), alkanes (2.92%), benzene (1.88%) and naphthalene (1.12%). The major components of AFLEO were nerolidol (11.7%), farnesol (6.73%) and phytol (6.63%). Similar to AFLEO, AFFEO was also abundant in alcohols (46.15%), olefins (28.75%), alkanes (9.73%), ketones (5.77%), benzene (3.42%) and naphthalene (1.89%). The major components of AFFEO were nerolidol (11.07%), farnesol (8.21%) and viridiforol (8.0%).
Antitumor activity
Antitumor activities of AFLEO and AFFEO were determined by MTT assay. As seen in Fig. 1, both AFLEO and AFFEO inhibited the proliferation of Huh7 and HepG2 cells in a time and dose-dependent manner.
Prediction of EOs targets and HCC targets
10 active ingredients of AFLEO and 9 of AFFEO were screened out according to the parameters of HIA, BBB permeation, and drug-likeness (Table S1 & S2). SwissTargetPrediction was used to obtain the potential targets of each ingredient. After removing the duplicate targets, 240 targets of AFLEO and 298 targets of AFFEO were obtained. Then a total of 8685 targets related to HCC were found by GeneCards and DisGeNET database. Venn diagrams were created to obtain the overlapping targets. As seen in Fig. 2, there were 176 AFLEO targets and 209 AFFEO targets that overlapped with HCC targets and were considered potential targets for AFLEO or AFFEO in treating HCC.
Construction and analysis of PPI Network
A PPI network was obtained after importing the overlapping targets into the STRING database. As shown in Fig. 3, for AFLEO, with closeness ≥ 0.002553852, betweenness ≥ 232.1169591 and degree ≥ 16.24561404, the PPI network with 28 nodes and 214 edges was obtained. The top 8 targets included AKT1, PPARG, EGFR, PTGS2, ESR1, HIF1A, MAPK3 and ERBB2. For AFFEO, with closeness ≥ 0.002136257, betweenness ≥ 275.1764706 and degree ≥ 17.5, the PPI network with 40 nodes and 348 edges was obtained. The top 8 targets included TNF, PPARG, EGFR, PTGS2, ESR1, HIF1A, MMP9 and GSK3B. Besides, the associations between these top 8 hub targets and HCC were assessed according to gifts in Genecards database. As shown in table S3, these hub targets were closely related to HCC.
The analysis of GO functions and KEGG pathway enrichment
GO functional analysis of the 175 targets of AFLEO and 208 targets of AFFEO against HCC were conducted to identify the functional role of them. As shown in Fig. 4A and B, the top 10 terms of BP, CC, and MF were ranked according to the p-value. The BP analysis revealed that the targets of AFLEO and AFFEO were mainly related to the protein phosphorylation, inflammatory response, and intracellular receptor signaling pathway. The CC analysis of AFLEO and AFFEO showed that the genes were highly enriched in cytoplasm, cytosol and plasma membrane. According to the MF, targets of AFLEO and AFFEO were mainly related to protein serine/threonine/tyrosine kinase activity, ligand-activated sequence-specific DNA binding, protein kinase activity and enzyme binding.
The top 20 pathways of the KEGG enrichment analysis ranked according to the p-value were shown in Fig. 4C and D. The KEGG pathways of AFLEO and AFFEO against HCC were mainly related to pathways in cancer, Chemical carcinogenesis - receptor activation, inflammatory mediator regulation of TRP channels, proteoglycans in cancer and PD-L1 expression and PD-1 checkpoint pathway in cancer.
Construction of component-target network
The compound-target networks were constructed by the 29 key targets of AFLEO and 40 key targets of AFFEO obtained by topological analysis. As seen Fig. 5A and B, the network of AFLEO consisted of 39 nodes and 133 edges. While the network of AFFEO consisted of 51 nodes and 153 edges.
The results of the compound-target-pathway network were demonstrated in Fig. 5C and D. The network of AFLEO consisted of 49 nodes and 203 edges. While the network of AFFEO consisted of 61 nodes and 229 edges. The results demonstrated that multiple components of AFLEO and AFFEO had multiple targets to treat HCC through various pathways.
Molecular docking analysis
As nerolidol is the most abundant component of both AFLEO and AFFEO, the overlapping targets of it and key protein targets of AFLEO and AFFEO were selected to do molecular docking. The binding energy of nerolidol and the protein target was shown in Table 2. Some docking results were visualized in Fig. 6.
Validation of target proteins via Western blotting
To verify the target proteins obtained via the above network, western blotting was performed. As shown in Fig. 7, the expression of AKT1 and MAPK3 decreased, while the expression of PPARG increased in HCC cells. Therefore, western blotting confirmed the reliability of the network pharmacology results.
Discussion
EOs are mixtures extracted from plants. Many kinds of EOs have the effects of antibacterial, antioxidant, antiviral, anti-inflammatory and antitumor. The bioactivity of EOs is related to their compositions [23]. Many compounds of AFLEO and AFFEO have important known biological activities, such as nerolidol, α-bisabolol, β-eudesmol and caryophyllene oxide. Nerolidol and α-bisabolol have antibacterial, antitumor, anti-inflammatory and antioxidant activities [24,25,26]. β-Eudesmol and caryophyllene oxide have anticancer and anti-inflammatory effects [27,28,29,30]. Therefore, considering the active substances identified in AFLEO and AFFEO, the EOs should have antitumor effects. To confirm the antitumor activity of AFLEO and AFFEO, MTT assays were performed. The decreased cell viability of Huh7 and HepG2 proved that AFLEO and AFFEO inhibit the proliferation of HCC cells.
Except the diverse chemical compositions of EOs, the mechanism of their pharmacological action is also complex as they can influence biological processes at cellular levels by interacting with multiple biological targets [23]. The network pharmacology analysis combined with molecular docking method were applied to determine the mechanisms of AFLEO and AFFEO against HCC. As shown in PPI network, AFLEO had 28 hub targets, such as AKT1, PPARG, EGFR, PTGS2, ESR1, HIF1A, MAPK3 and ERBB2, whereas AFFEO has 40 hub targets, such as TNF, PPARG, EGFR, PTGS2, ESR1, HIF1A, MMP9 and GSK3B. Previous studies have revealed that those targets are closely related to cell proliferation, differentiation, apoptosis [31,32,33,34,35], metabolism [36, 37], and immune microenvironment, which are involved in the development of HCC [38,39,40,41].
GO enrichment analysis showed that the targets of both AFLEO and AFFEO were closely related to 3 biological processes, including protein phosphorylation, inflammatory response, and signal transduction. KEGG pathway enrichment analysis showed that the targets of both AFLEO and AFFEO against to HCC were mainly enriched in pathways in cancer, Chemical carcinogenesis - receptor activation, inflammatory mediator regulation of TRP channels, proteoglycans in cancer, EGFR tyrosine kinase inhibitor resistance, and in cancer. It is well known that phosphorylation is one important post translational modifications, which could regulate cell growth, differentiation, apoptosis and cell signaling. Dysregulation of protein phosphorylation may cause many serious diseases, especially cancer [42]. Chronic inflammation promotes tumor development, including HCC. Inflammatory tumor microenvironment may lead to the proliferation, angiogenesis and metastasis of malignant cells [43]. The activity of signaling pathways is always altered in tumor cells, which will influence growth, proliferation, apoptosis, invasiveness, and metastasis of the cells. From the KEGG results, several signaling pathways closely related to HCC were enriched, such as VEGF signaling pathway and Ras signaling pathway. The result of KEGG showed that inflammatory mediator regulation of TRP channels may be an important pathway for both AFLEO and AFFEO in treating HCC. The association between TRP channels and cardiovascular diseases has been widely studied. However, more and more research found that TRP channels also play crucial roles in tumorigenesis and progression. Dysregulated TRP channel expression promotes cell proliferation, promote migration and invasion [44], protect cells from apoptosis through hypoxia [45], elevated oxidative stress and abnormal redox [46]. TRP channel also affects cancer progression by influence the immunocytes and tumor microenvironment [47]. We also noticed that the targets were also enriched into PD-L1 expression and PD-1 checkpoint pathway in cancer, which is related to T cell immunity. As the key targets, AKT1 is involved in VEGF signaling pathway and HIF-1 signaling pathway, MAPK3 is involved in Ras signaling pathway, while PPARG is involved in insulin resistance pathway. The result of GO and KEGG analysis indicated that AFLEO and AFFEO not only act directly on the tumor cells, but also affect the tumor microenvironment.
The binding ability between nerolidol and the key targets of AFLEO and AFFEO was detected by the molecular docking method. The binding energy between the receptor and ligand was lower than − 5 kcal/mol, which indicated that nerolidol and its targets may play a key role in the treatment of HCC. Among these targets, NR1I2, CYP2C9, CYP3A4, PTGS2, PGR, and JAK2 have higher binding energy. NR1I2, CYP2C9 and CYP3A4 are important regulators of hepatocellular carcinoma cellular resistance to antitumor drugs [48,49,50]. PTGS2 and JAK2 are related to proliferation, invasion, metastasis, and apoptosis of hepatocellular carcinoma cells [33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51]. PGR is associated with abnormal epigenetic silencing mechanisms in HCC cells [52]. However, the results of network pharmacology and molecular docking were only preliminary, and current results did not fully indicate whether AFLEO and AFFEO could be applied to HCC treatment. Further in vivo and in vitro experiments are needed to verify the active ingredients, targets and pathways of AFLEO and AFFEO against HCC.
Conclusions
The components of AFLEO and AFFEO were analyzed by GC-MS. A total of 30 and 22 compounds were identified in AFLEO and AFFEO, respectively. Nerolidol was the most abundant component of both essential oils. Both AFLEO and AFFEO can inhibit the proliferation of HCC. AFLEO and AFFEO may exert their antitumor effect by regulating multiple targets, such as PPARG, EGFR, PTGS2, ESR1 and HIF1A. Moreover, their therapeutic effects may relate to multiple pathways. Therefore, AFLEO and AFFEO have the potential to be studied and developed as candidate drugs for HCC treatment.
Data availability
All the data are included in the article and supplementary material, further inquiries can be directed to the corresponding authors.
Abbreviations
- AFLEO:
-
Essential oil extracted from Amorpha fruticosa leaves
- AFFEO:
-
Essential oil extracted from Amorpha fruticosa flowers
- EOs:
-
Essential oils
- GC-MS:
-
Gas chromatography-mass spectrometry
- HCC:
-
Hepatocellular carcinoma
- NIST:
-
National Institute of Standards and Technology
- HIA:
-
Human gastrointestinal absorption
- BBB:
-
Blood-brain barrier
- PPI:
-
Protein–protein interaction
- STRING:
-
Search Tool for the Retrieval of Interacting Genes
- GO:
-
Gene Ontology
- KEGG:
-
Kyoto Encyclopedia of Genes and the Genomes
- DAVID:
-
Integrated Annotation and Discovery Visualization Database
References
de Martel C, Maucort-Boulch D, Plummer M, Franceschi S. World-wide relative contribution of hepatitis B and C viruses in hepatocellular carcinoma. Hepatology. 2015;62(4):1190–200.
Lau WY, Ho SK, Yu SC, Lai EC, Liew CT, Leung TW. Salvage surgery following downstaging of unresectable hepatocellular carcinoma. Ann Surg. 2004;240(2):299–305.
Altekruse SF, McGlynn KA, Dickie LA, Kleiner DE. Hepatocellular carcinoma confirmation, treatment, and survival in surveillance, epidemiology, and end results registries, 1992–2008. Hepatology. 2012;55(2):476–82.
Somleva T, Ognyanov I. New rotenoids in Amorpha fruticosa fruits. Planta Med. 1985;51(3):219–21.
Masayoshi O, Toshiyuki T, Munekazu I. A prenylated flavanone from roots of Amorpha fruticosa. Phytochemistry. 1998;48(5):907–9.
Kim YS, Ryu YB, Curtis-Long MJ, Yuk HJ, Cho JK, Kim JY, Kim KD, Lee WS, Park KH. Flavanones and rotenoids from the roots of Amorpha fruticosa L. that inhibit bacterial neuraminidase. Food Chem Toxicol. 2011;49(8):1849–56.
Kemal M, Wahba Khalil SK, Rao NG, Woolsey NF. Isolation and identification of a cannabinoid-like compound from Amorpha species. J Nat Prod. 1979;42(5):463–8.
Stoyanova A, Georgiev E, Lis A, Majda T, Gora J. Essential oil from stored fruits of Amorpha fruticosa L. J Essent Oil Bearing Plants. 2003;6(3):195–7.
Das PK, Goswami S, Chinniah A, Panda N, Banerjee S, Sahu NP, Achari B. Woodfordia fruticosa: traditional uses and recent findings. J Ethnopharmacol. 2007;110(2):189–99.
Li L, Wang HK, Chang JJ, et al. Antitumor agents, 138. Rotenoids and isoflavones as cytotoxic constitutents from Amorpha fruticosa. J Nat Prod. 1993;56(5):690–8.
Dat NT, Lee JH, Lee K, Hong YS, Kim YH, Lee JJ. Phenolic constituents of Amorpha fruticosa that inhibit NF-kappaB activation and related gene expression. J Nat Prod. 2008;71(10):1696–700.
Choi Y, Lee JH. The combination of tephrosin with 2-deoxy-D-glucose enhances the cytotoxicity via accelerating ATP depletion and blunting autophagy in human cancer cells. Cancer Biol Ther. 2011;12(11):989–96.
Fuhr L, Rousseau M, Plauth A, Schroeder FC, Sauer S. Amorfrutins are natural PPARγ agonists with potent anti-inflammatory properties. J Nat Prod. 2015;78(5):1160–4.
Kloutek E, Popov A, Drenska D, Uzunov P. Eksperimentalni prouchvaniia za khepatoprotektivnata aktivnost na izolirani flavonoidi ot Amorpha fructiosa [Experimental research on the hepatoprotective activity of flavonoids isolated from Amorpha Fructiosa]. Eksp Med Morfol. 1985;24(2):50–4. Bulgarian.
Anna Lis &Józef Góra. Essential oil of Amorpha fruticosa L. J Essent Oil Res. 2001;13(5):340–2.
Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017;7:42717.
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(W1):W357–64.
Stelzer G, Rosen N, Plaschkes I, Zimmerman S, Twik M, Fishilevich S et al. The GeneCards Suite: From Gene Data Mining to Disease Genome Sequence Analyses. Curr Protoc Bioinformatics. 2016;54:1.30.1–1.30.33.
Piñero J, Saüch J, Sanz F, Furlong LI. The DisGeNET cytoscape app: exploring and visualizing disease genomics data. Comput Struct Biotechnol J. 2021;19:2960–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.csbj.2021.05.015.
Philippe, Bardou. Jérôme Mariette, Frédéric Escudié, Christophe Djemiel, Christophe Klopp. Jvenn: an interactive Venn diagram viewer. BMC Bioinformatics. 2014;15:293.
Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, et al. The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017;45(D1):D362–8.
Sherman BT, Hao M, Qiu J, Jiao X, Baseler MW, Lane HC, et al. DAVID: a web server for functional enrichment analysis and functional annotation of gene lists (2021 update). Nucleic Acids Res. 2022;50(W1):W216–21.
de Sousa DP, Damasceno ROS, Amorati R, Elshabrawy HA, de Castro RD, Bezerra DP, et al. Essential oils: Chemistry and pharmacological activities. Biomolecules. 2023;13(7):1144. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biom13071144.
Alonso L, Fernandes KS, Mendanha SA, Gonçalves PJ, Gomes RS, Dorta ML, et al. In vitro antileishmanial and cytotoxic activities of nerolidol are associated with changes in plasma membrane dynamics. Biochim Biophys Acta Biomembr. 2019;1861(6):1049–56.
de Moura DF, Rocha TA, de Melo Barros D, da Silva MM, Dos Santos Santana M, Neta BM, et al. Evaluation of the antioxidant, antibacterial, and antibiofilm activity of the sesquiterpene nerolidol. Arch Microbiol. 2021;203(7):4303–11.
Ramazani E, Akaberi M, Emami SA, Tayarani-Najaran Z. Pharmacological and biological effects of alpha-bisabolol: an updated review of the molecular mechanisms. Life Sci. 2022;304:120728.
Nam SY, Kim HY, Kim HM, Jeong HJ. Βeta-eudesmol reduces stem cell factor-induced mast cell migration. Int Immunopharmacol. 2017;48:1–7.
Bomfim DS, Ferraz RP, Carvalho NC, Soares MB, Pinheiro ML, Costa EV, Bezerra DP. Eudesmol isomers induce caspase-mediated apoptosis in human hepatocellular carcinoma HepG2 cells. Basic Clin Pharmacol Toxicol. 2013;113(5):300–6.
Chavan MJ, Wakte PS, Shinde DB. Analgesic and anti-inflammatory activity of Caryophyllene oxide from Annona squamosa L. bark. Phytomedicine. 2010;17(2):149–51.
Delgado C, Mendez-Callejas G, Celis C. Caryophyllene Oxide, the active compound isolated from leaves of Hymenaea courbaril L. (Fabaceae) with Antiproliferative and apoptotic effects on PC-3 androgen-independent prostate Cancer Cell line. Molecules. 2021;26(20):6142.
Chen J, Liang J, Liu S, Song S, Guo W, Shen F. Differential regulation of AKT1 contributes to survival and proliferation in hepatocellular carcinoma cells by mediating Notch1 expression. Oncol Lett. 2018;15(5):6857–64.
Bang J, Jun M, Lee S, Moon H, Ro SW. Targeting EGFR/PI3K/AKT/mTOR signaling in Hepatocellular Carcinoma. Pharmaceutics. 2023;15(8):2130.
Chen Y, Chen HN, Wang K, Zhang L, Huang Z, Liu J, et al. Ketoconazole exacerbates mitophagy to induce apoptosis by downregulating cyclooxygenase-2 in hepatocellular carcinoma. J Hepatol. 2019;70(1):66–77.
Hu X, Pan H, Zhou S, Pang Q, Wang Y, Zhu C, et al. HS1BP3, transcriptionally regulated by ESR1, promotes hepatocellular carcinoma progression. Biochem Biophys Res Commun. 2022;623:111–9.
Li Y, Zhou Y, Ma L, Liu D, Dai Z, Shen J. Mir-3677-3p promotes hepatocellular carcinoma progression via inhibiting GSK3β. Acta Biochim Biophys Sin (Shanghai). 2020;52(12):1404–12.
Zhao Y, Tan H, Zhang X, Zhu J. Roles of peroxisome proliferator-activated receptors in hepatocellular carcinoma. J Cell Mol Med. 2024;28(5):e18042.
Zhang MS, Cui JD, Lee D, Yuen VW, Chiu DK, Goh CC, et al. Hypoxia-induced macropinocytosis represents a metabolic route for liver cancer. Nat Commun. 2022;13(1):954.
Sun H, Qian X, Yang W, Zhou W, Zhou C, Liu S, et al. Novel prognostic signature based on HRAS, MAPK3 and TFRC identified to be associated with ferroptosis and the immune microenvironment in hepatocellular carcinoma. Am J Transl Res. 2022;14(10):6924–40.
Yu C, Zhang X, Wang M, Xu G, Zhao S, Feng Y, et al. Afatinib combined with anti-PD1 enhances immunotherapy of hepatocellular carcinoma via ERBB2/STAT3/PD-L1 signaling. Front Oncol. 2023;13:1198118. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fonc.2023.1198118.
Shrestha R, Bridle KR, Crawford DHG, Jayachandran A. TNF–α–mediated epithelial–to–mesenchymal transition regulates expression of immune checkpoint molecules in hepatocellular carcinoma. Mol Med Rep. 2020;21(4):1849–60.
Liu G, Yin L, Ouyang X, Zeng K, Xiao Y, Li Y. M2 macrophages promote HCC Cells Invasion and Migration via miR-149-5p/MMP9 signaling. J Cancer. 2020;11(5):1277–87.
Singh V, Ram M, Kumar R, Prasad R, Roy BK, Singh KK. Phosphorylation: implications in Cancer. Protein J. 2017;36(1):1–6.
Lin Y, Jiang M, Chen W, Zhao T, Wei Y. Cancer and ER stress: mutual crosstalk between autophagy, oxidative stress and inflammatory response. Biomed Pharmacother. 2019;118:109249.
Elzamzamy OM, Penner R, Hazlehurst LA. The role of TRPC1 in modulating Cancer Progression. Cells. 2020;9(2):388.
Wang Y, Qi YX, Qi Z, Tsang SY. TRPC3 regulates the proliferation and apoptosis resistance of Triple negative breast Cancer cells through the TRPC3/RASA4/MAPK pathway. Cancers (Basel). 2019;11(4):558.
Chay DB, Han GH, Cho H, Kim S, Kim JH. High expression of TRPV-1 is associated with poor prognosis in cervical cancer. Gynecol Oncol. 2019;154:270.
Parenti A, De Logu F, Geppetti P, Benemei S. What is the evidence for the role of TRP channels in inflammatory and immune cells? Br J Pharmacol. 2016;173(6):953–69.
Wang H, Chu F, Zhang XF, Zhang P, Li LX, Zhuang YL, et al. TPX2 enhances the transcription factor activation of PXR and enhances the resistance of hepatocellular carcinoma cells to antitumor drugs. Cell Death Dis. 2023;14(1):64.
Wang Y, Su K, Wang C, Deng T, Liu X, Sun S, et al. Chemotherapy-induced acetylation of ACLY by NAT10 promotes its nuclear accumulation and acetyl-CoA production to drive chemoresistance in hepatocellular carcinoma. Cell Death Dis. 2024;15(7):545.
Özkan A, Stolley DL, Cressman ENK, McMillin M, DeMorrow S, Yankeelov TE, et al. Tumor Microenvironment alters Chemoresistance of Hepatocellular Carcinoma through CYP3A4 metabolic activity. Front Oncol. 2021;11:662135.
Wu L, Li J, Liu T, Li S, Feng J, Yu Q, et al. Quercetin shows anti-tumor effect in hepatocellular carcinoma LM3 cells by abrogating JAK2/STAT3 signaling pathway. Cancer Med. 2019;8(10):4806–20.
Kondo Y, Shen L, Suzuki S, Kurokawa T, Masuko K, Tanaka Y, et al. Alterations of DNA methylation and histone modifications contribute to gene silencing in hepatocellular carcinomas. Hepatol Res. 2007;37(11):974–83.
Funding
This work was supported by the Key Scientific and Technological Project of Henan Province under Grant (No. 242102310387); Project of Basic Research Fund of Henan Institute of Medical and Pharmacological Sciences under Grant (No. 2024BP0102/2024BP0206/2024BP0203).
Author information
Authors and Affiliations
Contributions
Yixian Liu, Huiping Chen and Jintao Zhang contributed to the study conception and design. Material preparation, data collection and analysis were performed by Xiaojun Zhang, Jiacong Hao and Ying Zhao, Huiping Chen and Min Zou reviewed and checked all the experimental data. The first draft of the manuscript was written by Yixian Liu, and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Liu, Y., Zhang, X., Hao, J. et al. Essential oils from Amorpha fruticosa against hepatocellular carcinoma based on network pharmacology. BMC Complement Med Ther 25, 29 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12906-025-04766-5
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12906-025-04766-5