Logo
J Kermanshah Univ Med Sci

Image Credit:J Kermanshah Univ Med Sci

Therapeutic Effects of Taraxasterol on Triple-Negative Breast Cancer (MDA-MB-231 Cells) Through Modulation of the Novel AKT/mTOR/β-Catenin Signaling Pathway

Author(s):
Amir AbdolmalekiAmir Abdolmaleki1, Iraj RashidiIraj Rashidi2, Cyrus JaliliCyrus JaliliCyrus Jalili ORCID2, Ali GhanbariAli GhanbariAli Ghanbari ORCID2, Motahareh ZeinivandMotahareh Zeinivand3, Hadis NouriHadis Nouri4, Fuzieh Khani HematabadiFuzieh Khani HematabadiFuzieh Khani Hematabadi ORCID2,*
1Department of Operating Room, Nahavand Shool of Allied Medical Sciences, Hamadan University of Medical Sciences, Hamadan, Iran
2Department of Anatomical Sciences, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
3Department of Physiology, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
4Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran

Journal of Kermanshah University of Medical Sciences:Vol. 29, issue 2; e141551
Published online:Aug 15, 2023
Article type:Research Article
Received:Oct 10, 2023
Accepted:Jun 26, 2025
How to Cite:Abdolmaleki A, Rashidi I, Jalili C, Ghanbari A, Zeinivand M, et al. Therapeutic Effects of Taraxasterol on Triple-Negative Breast Cancer (MDA-MB-231 Cells) Through Modulation of the Novel AKT/mTOR/β-Catenin Signaling Pathway.J Kermanshah Univ Med Sci.2023;29(2):e141551.https://doi.org/10.5812/jkums-141551.

Abstract

Background:

Triple-negative breast cancer (TNBC) is a subtype of breast cancer (BC) with high resistance to chemotherapy. Taraxasterol is isolated from Taraxacum officinale (TO) with anti-cancer effects.

Objectives:

The present study aimed to investigate the anticancer effect of taraxasterol on TNBC cells.

Methods:

Taraxacum officinale extract was prepared and purified with concentrations ranging from 1 to 128 μM. MDA-MB-231 cells were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS) and treated with TO for 24 to 96 hours. Cell viability was assessed using the MTT assay, and apoptosis was evaluated by the diphenylamine method. Gene expression of HPRT, OPN, AKT1, mTOR, PTEN, and β-Catenin was measured by real-time PCR using the 2-ΔΔCt method on the StepOne system. Statistical analysis was performed using one-way ANOVA followed by Tukey’s post hoc test in SPSS, with significance set at P < 0.05.

Results:

After 24, 48, 72, and 96 h of treatment with taraxasterol, cell viability was decreased (P < 0.05). The IC50 values were respectively found 439.37 ± 6.8, 213.27 ± 5.78, 121 ± 7.98 and 27.86 ± 9.66 μM following 24, 48, 72, and 96 h of taraxasterol exposure. Diphenylamine assay showed that IC50 concentration of taraxasterol causes a significant increase in apoptosis after 24 h. Also, after 24 h of treatment, taroxasterol induced a significant decrease in gene expression of HPRT, OPN, AKT1, mTOR, and β-Catenin.

Conclusions:

Taraxasterol can reduce survival rate and induce apoptosis in TNBC cells through disruption of the AKT/mTOR/β-Catenin pathway.

1. Background

Breast cancer is the most common malignancy in females, approximately one-third of all women’s tumors (1). Pathologically, the BC is classified into four subtypes luminal A, luminal B, HER-2+, and triple-negative breast cancer (TNBC) (2). Among these, the TNBC is known for the lack of estrogen (ER), progesterone receptors (PR), and growth factor receptor-2 (HER-2). The TNBC accounts for 12 - 20% of all BC cases. The incidence rate of TNBC increases in premenopausal women with more prevalence in younger (3).

Triple-negative tumors are characterized by aggressive behaviors with primary metastasis to the central nervous system, bones, lungs, and liver. Triple-negative tumors contain poor response to therapies, poor prognosis, and low survival rates compared to other BC subtypes (4). Unlike ER and HER-2+ cancers, TNBCs are highly resistant to current therapies because they are insensitive to endocrine and molecularly targeted treatments. Currently, only systemic chemotherapy and surgery are considered the main management for TNBC cases (5).

Over the thousands of years of human life, a large number of natural products of herbs have been used for the treatment of various diseases (6). In the 1950s, scientists systematically investigate natural organisms as a source of effective anticancer substances. Recently, it is proposed that the application of natural products is the most successful strategy for discovery of new anti-cancer drugs (7). Taraxasterol ((3β, 18α, 18α)-Urs-20(30)-en-3-ol)), as a pentacyclic triterpene, is isolated from Taraxacum officinale (TO) plant. Taraxacum officinale is used in traditional medicine for its milk-enhancing, choleretic, diuretic, and anti-inflammatory activities. Previous studies showed that TO extracts are effective in inhibiting proliferation and induction of apoptosis in cancer cells. According to the studies, taraxasterol is an important bioactive compound with strong anti-inflammatory and antioxidant activities (8).

The PI3K/AKT/mTOR signaling pathway plays an essential role in many cellular processes and is often altered in BC, leading to increased tumor growth. Small molecule inhibitors can effectively target the key elements in this pathway (9).

2. Objectives

Generally, development and evaluation of new agents against TNBC are difficult due to the high biological heterogeneity. Also, this type of tumor is considerably resistant to remedies which can potentially develop metastasis potential. Therefore, there is an urgent need for identification and development of new potential therapeutic drugs against TNBC with high efficiency and low side effects. Thus, the anticancer properties of taraxasterol on modulation of AKT/mTOR/β-Catenin signaling pathway (in TNBC cells) were investigated in the present laboratory-based study.

3. Methods

3.1. Taraxacum officinale Extraction and Taraxasterol Isolation

Taraxacum officinale was prepared by the Research Institute of Medicinal Plants (Tehran, Iran). The authenticity of the plant was confirmed by a herbalist. Then, the active substance of TO was extracted and purified under the supervision of the extraction company (Milad Herbal Medicine, Tehran, Iran). The desired extract was classified into different doses of 1, 2, 4, 8, 16, 32, 64, and 128 μM.

3.2. Cell Culture

The MDA-MB-231 cell line was obtained from the Pasteur Institute (Iran, Tehran). The cells were cultured (37°C, 5% CO2) in RPMI-1640 medium (Gibco, Germany) supplemented with 10% fetal bovine serum (FBS) (Gibco, Germany) and lack of antibiotics. All protocols of cell culture were considered carefully based on the previously published studies (10).

3.3. Laboratory Protocol

Following the preparation of various doses of TO, TNBC cells were cultured and exposed to TO. The procedures of exposure were applied based on the different times.

3.4. Cell Viability Assessment

2,5-diphenyl-2H-tetrazolium bromide (MTT) test was used to assess the cell viability. To this aim, 15 × 103 cells were cultured in each well of a 96-well plate and incubated overnight. Then, the cells were treated with various concentrations of taraxasterol (Sigma, Germany) for 24, 48, 72, and 96 h. The supernatant was discarded and MTT solution (Sigma, Germany) was exposed to each well (5 mg/mL in culture medium) for 3h. Following incubation, the supernatant was removed and formazan crystals were dissolved with 100 μL of DMSO. Optical density (OD) was measured at 570 nm with background subtraction at 690 nm. Data from three independent experiments were normalized using the following equation: Cell viability (%) = (OD sample value)/(OD cell control) × 100 (11).

3.5. Cell Apoptosis Investigation

The percentage of fragmented DNA following 24 h treatment with IC50 concentration of taraxasterol was calculated using diphenylamine assay. For the data report, the measurement of OD of samples was read by a spectrophotometer at 600 nm as described by Cohen and Duke (12).

3.6. Gene Expression Analysis

Gene expression assessment was performed by the real-time PCR method. Following the treatment of the cells with IC50 concentration for 24 h, the RNA was isolated from 1 × 106 cells of control and treatment cells using TRIZol reagent (Invitrogen, USA), and RNA purity and integrity were evaluated using a Nanodrop machine. Complementary DNA (cDNA) synthesis was carried out using a cDNA synthesis kit (Vivantis Technologies, Malaysia) and a real-time PCR process was performed by SYBR Premix Ex Taq Technology (TaKaRa Bio Inc., Japan). All laboratory processes were conducted according to the manufacturer’s instructions using the Applied Biosystems StepOne Real-Time PCR System. Changes were obtained based on the comparative method of Ct (2-ΔΔct) (13). GAPDH was used as an internal control and the primer sequences of various genes (HPRT, OPN, AKT1, mTOR, PTEN, β-Catenin, and GAPDH) were designed by GeneRunner software (v.3.05) according to Table 1.

Table 1.The Primer Sequences of Genes
GenesSequences
HPRTF: 5´-TGGACAGGACTGAACGTCTTG-3´
R: 5´-CCAGCAGGTCAGCAAAGAATTTA-3´
OPNF: 5´-ACCCTTCCAAGTAAGTCCAACG-3´
R: 5´-GGTGAGAATCATCAGTGTCATCTAC-3´
AKT1F: 5´-AGCGACGTGGCTATTGTGAAG-3´
R: 5´-GTACTCCCCTCGTTTGTGCAG-3´
mTORF: 5´-AACTCCGAGAGATGAGTCAAGA-3´
R: 5´-AGTTGGTCATAGAAGCGAGTAGA-3´
PTENF: 5´-TGGATTCGACTTAGACTTGACCT-3´
R: 5´-TTTGGCGGTGTCATAATGTCTT-3´
β-cateninF: 5´-TACCTCCCAAGTCCTGTATGAG-3´
R: 5´-TGAGCAGCATCAAACTGTGTAG-3´
GAPDHF: 5´-TCCCTGAGCTGAACGGGAAG-3´
R: 5´-GGAGGAGTGGGTGTCGCTGT-3´

The Primer Sequences of Genes

3.7. Statistical Analysis

Following data extraction, the Kolmogorov-Smirnov test was the assessment of normal data distribution. One-way analysis of variance (one-way ANOVA) was used for statistical analysis and the Tukey post hoc test was conducted to determine the difference between the groups. Statistical Package for the Social Sciences 16 (SPSS Inc., Chicago, IL) was used for data analysis, and the results were expressed as Mean ± standard error. A P < 0.05 was also considered statistically significant (14).

4. Results

4.1. Taraxasterol and Triple-Negative Breast Cancer Cell Viability

Following 24, 48, 72, and 96 h of treatment with various concentrations of taraxasterol, the TNBC cell viability was decreased significantly (P < 0.05) in the concentration- and time-dependent manner. These time and concentration dependencies were 24h of taraxasterol exposure with concentrations of 32, 64, and 128 μM, 48 h of taraxasterol exposure with concentrations of 8, 16, 32, 64, and 128 μM, 72 h of taraxasterol exposure with concentrations of 4, 8, 16, 32, 64, and 128 μM and 96 h of taraxasterol exposure with concentrations of 2, 4, 8, 16, 32, 64 and 128 μM which caused low TNBC cells viability. Also, the IC50 of taraxasterol was detected at 439.37 ± 6.8, 213.27 ± 5.78, 121 ± 7.98 and 27.86 ± 9.66 μM respectively after 24, 48, 72, and 96 h of exposure. These findings approved that the viability of TNBC cells decreases dramatically following exposure to taraxasterol. Also, it was concluded that by the acceleration of the taraxasterol dose and the time of exposure, the Viability Index decreases significantly (Figure 1).

Effects of taraxasterol on viability of breast cancer (BC) cells. Data were presented as mean ± S.E.M. *P &lt; 0.05, **P &lt; 0.01, and ***P &lt; 0.01 compared to control.
Figure 1.

Effects of taraxasterol on viability of breast cancer (BC) cells. Data were presented as mean ± S.E.M. *P < 0.05, **P < 0.01, and ***P < 0.01 compared to control.

4.2. Taraxasterol and Triple-Negative Breast Cancer Cells Apoptosis

According to the data analysis, taraxasterol represented apoptotic features (44.02 ± 2.3% vs 1.01 ± 0.002% respectively in treatment vs control groups) significantly (P < 0.05) after 24 h at the IC50 concentration (439.37 µM) (Figure 2).

Effects of taraxasterol on apoptosis rate in breast cancer (BC) cells. Data were presented as mean ± S.E.M. ***P &lt; 0.01 compared to control.
Figure 2.

Effects of taraxasterol on apoptosis rate in breast cancer (BC) cells. Data were presented as mean ± S.E.M. ***P < 0.01 compared to control.

4.3. Taraxasterol and Gene Expression in Triple-Negative Breast Cancer Cells

Following 24 h of treatment with IC50 concentration (439.37 µM) of tarxasterol, a significant (P < 0.05) decrease was detected in the expression of HPRT (0.72 ± 0.2 in treatment group than 1 in control group), OPN (0.49 ± 0.1 in treatment group than 1 in control group), AKT1 (0.85 ± 0.1 in treatment group than 1 in control group), mTOR (0.73 ± 0.1 in treatment group than 1 in control group) and β-catenin (0.43 ± 0.11 in treatment group than 1 in control group) genes. It can be concluded that the gene expression of HPRT, OPN, AKT1, mTOR, and β-catenin was decreased respectively by 24%, 29%, 12%, 55%, and 53% following tarxasterol exposure. PTEN (1.08 ± 0.22 in the treatment group than 1 in the control group) gene expression was also increased significantly (P < 0.05) by 1.08 after tarxasterol exposure (Figure 3).

Effects of taraxasterol on gene expression of A, HPRT; B, OPN; C, AKT1; D, mTOR; E, β-Catenin; and F, PTEN in breast cancer (BC) cells. Data were presented mean ± S.E.M. * P &lt; 0.05 and *** P &lt; 0.001 compared to the control.
Figure 3.

Effects of taraxasterol on gene expression of A, HPRT; B, OPN; C, AKT1; D, mTOR; E, β-Catenin; and F, PTEN in breast cancer (BC) cells. Data were presented mean ± S.E.M. * P < 0.05 and *** P < 0.001 compared to the control.

5. Discussion

The TNBC is characterized by poor prognosis and resistance to conventional therapies. Phytochemicals such as taraxasterol, a bioactive compound from TO, have demonstrated anticancer properties. The current study confirms that taraxasterol induces apoptosis and reduces viability in MDA-MB-231 cells.

Taraxacum officinale antitumor activity was initially reported in 1981 (15). It exhibits cytotoxicity by promoting TNF-α and IL-1α production (16), key cytokines in apoptosis induction (17). Apoptosis is mediated via both extrinsic (death receptor) and intrinsic (mitochondrial) pathways. Taraxacum officinale also inhibits cancer cell invasiveness by attenuating phosphorylation of focal adhesion kinase (FAK) and Src, and reducing matrix metalloproteinases (MMP-2/9) activity (18).

Sox2, a pluripotency-associated transcription factor, is aberrantly expressed in multiple malignancies and facilitates proliferation and tumorigenesis, positioning it as a potential therapeutic target (19, 20).

RARβ2 encodes a nuclear receptor involved in retinoic acid-mediated transcriptional regulation, critical for morphogenesis and differentiation. Its expression is often suppressed in cancer, implicating its role as a tumor suppressor (15, 21).

TNF-related apoptosis-inducing ligand (TRAIL) is a selective inducer of apoptosis in cancer cells (22), although resistance mechanisms limit its efficacy. TIPRL inhibition sensitizes cells to TRAIL via MKK7-JNK pathway activation. Co-administration with TO enhances apoptosis through inhibition of the TIPRL-MKK7 interaction and JNK phosphorylation (15).

Taraxasterol markedly downregulated osteopontin (OPN), a multifunctional protein implicated in metastasis and poor outcomes in breast and other cancers (23). It also suppressed HPRT1 expression, a gene involved in nucleotide metabolism and highly expressed in TNBC. HPRT1 overexpression correlates with aggressive tumor phenotypes and regulates oncogenic pathways including Notch and ErbB, indicating its value as a biomarker and therapeutic target (24).

AKT1, commonly hyperactivated in BC (25), was downregulated following taraxasterol exposure, suggesting inhibition at both post-translational and transcriptional levels. mTOR expression was similarly reduced (26). PTEN expression, however, remained unchanged, aligning with prior findings where its modulation is context-dependent (27).

β-catenin, a key effector of Wnt signaling and regulator of cell adhesion and transcription, was also significantly reduced. Aberrant activation of β-catenin promotes oncogenesis by enhancing proliferation and invasion (28). Taraxasterol and similar plant-derived agents disrupt Wnt/β-catenin signaling and inhibit cancer stem cell function, thereby suppressing tumor progression (29).

Cytotoxicity analysis revealed that taraxasterol reduced cell viability in a time-dependent manner, with IC50 values declining from 439.37 ± 6.8 μM at 24 h to 27.86 ± 9.66 μM at 96 h (P < 0.05), indicating cumulative toxicity. Apoptosis induction was confirmed via diphenylamine assay. Gene expression profiling showed significant downregulation of HPRT1, OPN, AKT1, mTOR, and β-catenin at 24 h.

The AKT/mTOR/β-catenin signaling axis exhibits extensive crosstalk with Wnt and Notch pathways (15, 18, 29). Wnt signaling enhances β-catenin stabilization and nuclear translocation, while AKT/mTOR potentiates its activity. Notch signaling further modulates β-catenin via NICD interactions, reinforcing proliferative and survival signaling. Therefore, simultaneous targeting of these interconnected pathways may offer an effective strategy for TNBC therapy.

5.1. Conclusions

Taraxasterol as a pentacyclic triterpenoid available in the TO herb can potentially exert anticancer features on TNBC cells. The process of anti-proliferation of taraxasterol occurred following two stages of reduced survival rate and increased apoptosis levels through disruption of AKT/mTOR/β-Catenin signaling pathway. Since the administration of taraxasterol for cancer inhibition was found in a dose and time-associated manner, it is recommended to design an animal study for a deep assessment of the treatment.

5.2. Limitations

Although the plant authenticity was confirmed and extraction supervised, potential variability in the purity and composition of taraxasterol extract may affect reproducibility and consistency of results, especially since detailed characterization (e.g., purity percentage, batch-to-batch variation) is not described. The study relies solely on the MDA-MB-231 TNBC cell line, which may limit the generalizability of findings across other TNBC subtypes or BC models with different genetic backgrounds. Apoptosis was assessed only by diphenylamine assay measuring DNA fragmentation at 24 hours, without complementary methods (e.g., flow cytometry, caspase activity) to confirm apoptotic pathways and kinetics.

Acknowledgments

Footnotes

References

  • 1.
    Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209-49. [PubMed ID: 33538338]. https://doi.org/10.3322/caac.21660.
  • 2.
    Wu J, Hicks C. Breast Cancer Type Classification Using Machine Learning. J Pers Med. 2021;11(2). [PubMed ID: 33498339]. [PubMed Central ID: PMC7909418]. https://doi.org/10.3390/jpm11020061.
  • 3.
    Manjunath M, Choudhary B. Triple-negative breast cancer: A run-through of features, classification and current therapies. Oncol Lett. 2021;22(1):512. [PubMed ID: 33986872]. [PubMed Central ID: PMC8114477]. https://doi.org/10.3892/ol.2021.12773.
  • 4.
    Dass SA, Tan KL, Selva Rajan R, Mokhtar NF, Mohd Adzmi ER, Wan Abdul Rahman WF, et al. Triple Negative Breast Cancer: A Review of Present and Future Diagnostic Modalities. Medicina (Kaunas). 2021;57(1). [PubMed ID: 33445543]. [PubMed Central ID: PMC7826673]. https://doi.org/10.3390/medicina57010062.
  • 5.
    Lebert JM, Lester R, Powell E, Seal M, McCarthy J. Advances in the systemic treatment of triple-negative breast cancer. Curr Oncol. 2018;25(Suppl 1):S142-50. [PubMed ID: 29910657]. [PubMed Central ID: PMC6001760]. https://doi.org/10.3747/co.25.3954.
  • 6.
    Mandlik DS, Mandlik SK. Herbal and Natural Dietary Products: Upcoming Therapeutic Approach for Prevention and Treatment of Hepatocellular Carcinoma. Nutr Cancer. 2021;73(11-12):2130-54. [PubMed ID: 33073617]. https://doi.org/10.1080/01635581.2020.1834591.
  • 7.
    Tulp M, Bohlin L. Functional versus chemical diversity: is biodiversity important for drug discovery? Trends Pharmacol Sci. 2002;23(5):225-31. [PubMed ID: 12008000]. https://doi.org/10.1016/s0165-6147(02)02007-2.
  • 8.
    Jiao F, Tan Z, Yu Z, Zhou B, Meng L, Shi X. The phytochemical and pharmacological profile of taraxasterol. Front Pharmacol. 2022;13:927365. [PubMed ID: 35991893]. [PubMed Central ID: PMC9386448]. https://doi.org/10.3389/fphar.2022.927365.
  • 9.
    Li H, Prever L, Hirsch E, Gulluni F. Targeting PI3K/AKT/mTOR Signaling Pathway in Breast Cancer. Cancers (Basel). 2021;13(14). [PubMed ID: 34298731]. [PubMed Central ID: PMC8304822]. https://doi.org/10.3390/cancers13143517.
  • 10.
    Abdolmaleki A, Kondori BJ, Raei M, Ghaleh HEG. Cell therapy procedure using anti-inflammatory macrophage M2 can potentially reduce Clinical Score in animals with Experimental Autoimmune Encephalomyelitis: A preclinical systematic review and meta-analysis study. Fundam Clin Pharmacol. 2023;37(2):215-25. [PubMed ID: 36300567]. https://doi.org/10.1111/fcp.12844.
  • 11.
    Ghanbari A, Jalili C, Abdolmaleki A, Shokri V. Effects of cisplatin and acacetin on total antioxidant status, apoptosis and expression of OCTN3 in mouse testis. Biotech Histochem. 2022;97(3):185-91. [PubMed ID: 33998937]. https://doi.org/10.1080/10520295.2021.1925347.
  • 12.
    Salahshoor MR, Abdolmaleki A, Jalili C, Ziapoor A, Roshankhah S. Improvement of Petroselinum crispum on morphine toxicity in prefrontal cortex in rats. International Journal of Applied and Basic Medical Research. 2020;10(2):110-6. https://doi.org/10.4103/ijabmr.IJABMR_126_19.
  • 13.
    Abdolmaleki A, Jalili C, Mansouri K, Bakhtiari M. New rat to mouse xenograft transplantation of endometrium as a model of human endometriosis. Animal Model Exp Med. 2021;4(3):268-77. [PubMed ID: 34557653]. [PubMed Central ID: PMC8446700]. https://doi.org/10.1002/ame2.12181.
  • 14.
    Salahshoor MR, Abdolmaleki A, Shabanizadeh A, Jalali A, Roshankhah S. Ipomoea aquatica extract reduces hepatotoxicity by antioxidative properties following dichlorvos administration in rats. Chin J Physiol. 2020;63(2):77-84. [PubMed ID: 32341233]. https://doi.org/10.4103/CJP.CJP_89_19.
  • 15.
    Jalili C, Taghadosi M, Pazhouhi M, Bahrehmand F, Miraghaee SS, Pourmand D, et al. An overview of therapeutic potentials of Taraxacum officinale (dandelion): A traditionally valuable herb with a reach historical background. WCRJ World Cancer Res. J. 2020;7. e1679.
  • 16.
    Aras Ş, Aksu KiliÇLe P, ElİŞ Yildiz S, Uluman E, Bakir B, KaradaĞ Sari E, et al. Determination of the effects of Chia (Salvia hispanica L.) oil and Dandelion (Taraxacum Officinale) extract on Tumor Necrosis Factor-α (TNF-α) and Interleukin 6 (IL-6) release in liver tissue of diabetic rats. Turkish Journal of Veterinary Research. 2022;6(2):43-52. https://doi.org/10.47748/tjvr.1062162.
  • 17.
    Schuerwegh AJ, Dombrecht EJ, Stevens WJ, Van Offel JF, Bridts CH, De Clerck LS. Influence of pro-inflammatory (IL-1α, IL-6, TNF-α, IFN-γ) and anti-inflammatory (IL-4) cytokines on chondrocyte function. Osteoarthritis and Cartilage. 2003;11(9):681-7. https://doi.org/10.1016/s1063-4584(03)00156-0.
  • 18.
    Lee CW, Lin CC, Lin WN, Liang KC, Luo SF, Wu CB, et al. TNF-alpha induces MMP-9 expression via activation of Src/EGFR, PDGFR/PI3K/Akt cascade and promotion of NF-kappaB/p300 binding in human tracheal smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2007;292(3):L799-812. [PubMed ID: 17158602]. https://doi.org/10.1152/ajplung.00311.2006.
  • 19.
    Mamun MA, Mannoor K, Cao J, Qadri F, Song X. SOX2 in cancer stemness: tumor malignancy and therapeutic potentials. J Mol Cell Biol. 2020;12(2):85-98. [PubMed ID: 30517668]. [PubMed Central ID: PMC7109607]. https://doi.org/10.1093/jmcb/mjy080.
  • 20.
    Zhang S, Xiong X, Sun Y. Functional characterization of SOX2 as an anticancer target. Signal Transduct Target Ther. 2020;5(1):135. [PubMed ID: 32728033]. [PubMed Central ID: PMC7391717]. https://doi.org/10.1038/s41392-020-00242-3.
  • 21.
    Bahmani L, Baghi M, Peymani M, Javeri A, Ghaedi K. MiR-141-3p and miR-200a-3p are involved in Th17 cell differentiation by negatively regulating RARB expression. Hum Cell. 2021;34(5):1375-87. [PubMed ID: 34086186]. https://doi.org/10.1007/s13577-021-00558-4.
  • 22.
    Fulga A, Todiras M, Gudumac V, Tagadiuc O. Taraxacum officinale Leaves and Roots Suppress Glioma Cell Viability. Journal of Biosciences and Medicines. 2022;10(3):175-89. https://doi.org/10.4236/jbm.2022.103017.
  • 23.
    Zahed Panah M, Nikbakht M, Sajjadi SM, Rostami S, Norooznezhad AH, Kamranzadeh Fumani H, et al. Anti-Apoptotic Effects of Osteopontin via the Up-Regulation of AKT/mTOR/beta-Catenin Loop in Acute Myeloid Leukemia Cells. Int J Hematol Oncol Stem Cell Res. 2017;11(2):148-57. [PubMed ID: 28875010]. [PubMed Central ID: PMC5574411].
  • 24.
    J. Sedano M, I. Ramos E, Choudhari R, L. Harrison A, Subramani R, Lakshmanaswamy R, et al. Hypoxanthine Phosphoribosyl Transferase 1 Is Upregulated, Predicts Clinical Outcome and Controls Gene Expression in Breast Cancer. Cancers (Basel). 2020;12(6). [PubMed ID: 32532008]. [PubMed Central ID: PMC7352670]. https://doi.org/10.3390/cancers12061522.
  • 25.
    George B, Gui B, Raguraman R, Paul AM, Nakshatri H, Pillai MR, et al. AKT1 Transcriptomic Landscape in Breast Cancer Cells. Cells. 2022;11(15). [PubMed ID: 35892586]. [PubMed Central ID: PMC9332453]. https://doi.org/10.3390/cells11152290.
  • 26.
    Hare SH, Harvey AJ. mTOR function and therapeutic targeting in breast cancer. Am J Cancer Res. 2017;7(3):383-404. [PubMed ID: 28400999]. [PubMed Central ID: PMC5385631].
  • 27.
    Chen J, Sun J, Wang Q, Du Y, Cheng J, Yi J, et al. Systemic Deficiency of PTEN Accelerates Breast Cancer Growth and Metastasis. Front Oncol. 2022;12:825484. [PubMed ID: 35372075]. [PubMed Central ID: PMC8971716]. https://doi.org/10.3389/fonc.2022.825484.
  • 28.
    Wang Z, Zhang H, Hou J, Niu J, Ma Z, Zhao H, et al. Clinical implications of beta-catenin protein expression in breast cancer. Int J Clin Exp Pathol. 2015;8(11):14989-94. [PubMed ID: 26823833]. [PubMed Central ID: PMC4713619].
  • 29.
    Arzi L, Mollaei H, Hoshyar R. Countering Triple Negative Breast Cancer via Impeding Wnt/beta-Catenin Signaling, a Phytotherapeutic Approach. Plants (Basel). 2022;11(17). [PubMed ID: 36079579]. [PubMed Central ID: PMC9460573]. https://doi.org/10.3390/plants11172191.

Crossmark
Crossmark
Checking
Share on
Cited by
Metrics

Purchasing Reprints

  • Copyright Clearance Center (CCC) handles bulk orders for article reprints for Brieflands. To place an order for reprints, please click here (   https://www.copyright.com/landing/reprintsinquiryform/ ). Clicking this link will bring you to a CCC request form where you can provide the details of your order. Once complete, please click the ‘Submit Request’ button and CCC’s Reprints Services team will generate a quote for your review.
Search Relations

Author(s):

Related Articles