Pentagalloyl Glucose from Bouea macrophylla Suppresses the Epithelial–Mesenchymal Transition and Synergizes the Doxorubicin-Induced Anticancer and Anti-Migration Effects in Triple-Negative Breast Cancer
"> Figure 1
<p>The inhibitory effect of PGG and DOX on cell proliferation in TNBC cells, as assessed using the MTT assay. (<b>A</b>–<b>C</b>) Dose–response curves illustrating the cytotoxic effects of PGG on MCF-10A cells (<b>A</b>), DOX on MDA-MB231 cells (<b>B</b>), and PGG on MDA-MB231 cells (<b>C</b>) after 48 h of treatment. (<b>D</b>,<b>E</b>) Clonogenic survival analysis indicating the number of colonies formed by MDA-MB231 cells following treatment with varying concentrations of PGG (0, 2.5, 5, 10, 20, and 40 µM). Representative colony images are shown in (<b>D</b>), with quantification graphs in (<b>E</b>). The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> < 0.05 indicates a statistically significant difference compared to the control, while *** <span class="html-italic">p</span> < 0.001 denotes a highly significant difference from the control. PGG, pentagalloyl glucose; DOX, doxorubicin.</p> "> Figure 2
<p>PGG inhibits the migration and invasion capabilities of TNBC cells. (<b>A</b>) Representative microscopic images from wound healing assays performed on MDA-MB231 cells treated with varying concentrations of PGG, captured at 0 and 48 h. (<b>B</b>) Quantification of wound closure percentages, demonstrating the impact of PGG treatment. (<b>C</b>) Transwell chamber images showing cell migration following treatment with various concentrations of PGG. (<b>D</b>) Quantification of migrated cells: migrating cells were counted in five high-power fields and averaged. The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> < 0.05 indicates a statistically significant difference compared to the control. Scale bar = 100 µm (<b>A</b>,<b>C</b>). PGG, pentagalloyl glucose.</p> "> Figure 3
<p>Impact of combined treatment with 10, 20, and 40 µM PGG and varying concentrations of DOX on the viability of MDA-MB231 cells after 48 h, as assessed using the MTT assay (<b>A</b>). Fa-CI plot analysis depicting the interaction between DOX and PGG in MDA-MB231 cells. The dashed line at CI = 1 signifies an additive effect, while CI values less than, equal to, or greater than 1 indicate synergy, additivity, or antagonism, respectively (<b>B</b>). The effect (Fa) represents the degree of fractional inhibition associated with each combination index. PGG, pentagalloyl glucose; DOX, doxorubicin.</p> "> Figure 4
<p>The impact on apoptosis of PGG and DOX as monotherapies or in combination in TNBC cells. (<b>A</b>) Representative dot plots show the apoptotic response of MDA-MB231 cells to the indicated treatments. (<b>B</b>) Quantitative data represent the percentage of total cell death, as determined using flow cytometry. (<b>C</b>,<b>D</b>) The effects of PGG on mitochondrial membrane potential in MDA-MB231 cells were assessed using JC-1 staining: (<b>C</b>) representative images display JC-1 fluorescence across different treatment groups after 24 h, with FCCP as the positive control. Monomeric JC-1 exhibits green fluorescence, while aggregated JC-1 emits red fluorescence; (<b>D</b>) quantification of the red/green fluorescence ratio shown in a histogram. Data are presented as the mean ± SD of three independent experiments. (<b>E</b>) Western blot analysis of apoptotic markers, including Bax, Bcl-2, caspase-3, PARP, p-ERK, and t-ERK. The uncropped Western blot images are provided in <a href="#app1-pharmaceuticals-17-01729" class="html-app">Figure S1</a>. (<b>F</b>–<b>J</b>) The relative protein density values were quantified, with expression levels normalized to GAPDH as the loading control. The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> < 0.05 indicates a statistically significant difference compared to the control. Scale bar = 100 µm (<b>C</b>). PGG, pentagalloyl glucose; DOX, doxorubicin; PARP, poly ADP ribose polymerase; ERK, extracellular signal-regulated kinase.</p> "> Figure 4 Cont.
<p>The impact on apoptosis of PGG and DOX as monotherapies or in combination in TNBC cells. (<b>A</b>) Representative dot plots show the apoptotic response of MDA-MB231 cells to the indicated treatments. (<b>B</b>) Quantitative data represent the percentage of total cell death, as determined using flow cytometry. (<b>C</b>,<b>D</b>) The effects of PGG on mitochondrial membrane potential in MDA-MB231 cells were assessed using JC-1 staining: (<b>C</b>) representative images display JC-1 fluorescence across different treatment groups after 24 h, with FCCP as the positive control. Monomeric JC-1 exhibits green fluorescence, while aggregated JC-1 emits red fluorescence; (<b>D</b>) quantification of the red/green fluorescence ratio shown in a histogram. Data are presented as the mean ± SD of three independent experiments. (<b>E</b>) Western blot analysis of apoptotic markers, including Bax, Bcl-2, caspase-3, PARP, p-ERK, and t-ERK. The uncropped Western blot images are provided in <a href="#app1-pharmaceuticals-17-01729" class="html-app">Figure S1</a>. (<b>F</b>–<b>J</b>) The relative protein density values were quantified, with expression levels normalized to GAPDH as the loading control. The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> < 0.05 indicates a statistically significant difference compared to the control. Scale bar = 100 µm (<b>C</b>). PGG, pentagalloyl glucose; DOX, doxorubicin; PARP, poly ADP ribose polymerase; ERK, extracellular signal-regulated kinase.</p> "> Figure 5
<p>The impact of PGG combined with DOX on the migratory behavior of TNBC cells. (<b>A</b>) Representative microscopic images from wound healing assays and (<b>C</b>) quantification of wound closure percentages illustrating the effects of treatments with PGG (40 µM), DOX (0.75 µM), and their combination. (<b>B</b>) Transwell chamber images showing cell migration following treatment with PGG (40 µM), DOX (0.75 µM), or a combination of both. (<b>D</b>) Quantitative analysis of the number of migrating cells. The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> < 0.05 indicates a statistically significant difference compared to the control and DOX treatment alone. Scale bar = 100 µm (<b>A</b>,<b>B</b>). PGG, pentagalloyl glucose; DOX, doxorubicin.</p> "> Figure 6
<p>Reversal of EMT and suppression of EMT marker expression by PGG in TNBC cell lines. MDA-MB231 cells were treated for 48 h, as indicated, and the expression of EMT markers was analyzed using Western blot. (<b>A</b>) Representative Western blot images showing the levels of β-catenin, vimentin, E-cadherin, and GAPDH. The uncropped Western blot images are provided in <a href="#app1-pharmaceuticals-17-01729" class="html-app">Figure S2</a>. (<b>B</b>–<b>D</b>) Quantification of band intensities from the Western blot analysis. Relative protein levels were quantified and normalized to GAPDH as the loading control. The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> < 0.05 indicates a statistically significant difference compared to the control and DOX-only treatment. (<b>E</b>) Immunofluorescence staining of β-catenin (green), E-cadherin (green), and vimentin (red) in MDA-MB231 cells, with nuclei counterstained using DAPI (blue). Scale bar = 100 µm.</p> "> Figure 7
<p>The effects of PGG and DOX, either alone or in combination, on STAT3 signaling proteins in TNBC cells. (<b>A</b>) Representative Western blot images showing the expression levels of phosphorylated STAT3 (p-STAT3), total STAT3 (t-STAT3), and GAPDH after treatment with PGG (40 µM), DOX (0.75 µM), or their combination for 48 h in MDA-MB231 cells. The uncropped Western blot images are provided in <a href="#app1-pharmaceuticals-17-01729" class="html-app">Figure S3</a>. (<b>B</b>) Bar graph depicting the fold change in protein expression. The relative protein densities were quantified and normalized to the GAPDH loading control. The results are presented as the mean ± SD from three independent experiments. * <span class="html-italic">p</span> < 0.05 indicates a statistically significant difference compared to the control and DOX treatment alone. PGG, pentagalloyl glucose; DOX, doxorubicin; STAT3, signal transducer and activator of transcription 3; p, phosphorylated; t, total; GAPDH, glyceraldehyde 3-phosphate dehydrogenase.</p> ">
Abstract
:1. Introduction
2. Results
2.1. PGG Inhibits Cell Proliferation and Survival of Triple-Negative Breast Cancer Cells
2.2. PGG Suppresses Invasion and Migration Potential and Alters the Expression Levels of EMT-Associated Proteins in Triple-Negative Breast Cancer Cells
2.3. PGG Enhances the Antitumor Effect of Doxorubicin in Triple-Negative Breast Cancer Cells
2.4. Combination Treatment of PGG and Doxorubicin Enhances Apoptosis in Triple-Negative Breast Cancer Cells
2.5. PGG and Doxorubicin Combination Enhances the Anti-Migration Effect of DOX in Triple-Negative Breast Cancer Cells
2.6. PGG-Mediated Reversal of the EMT Process Plays a Crucial Role in the Anti-Migration Effect Induced by Combination Treatment of PGG and DOX
2.7. Abrogation of STAT3 Is Integral to PGG-Mediated Inhibition of EMT, Invasion, and Migration of Triple-Negative Breast Cancer Cells
3. Discussion
4. Materials and Methods
4.1. Preparation of PGG (Penta-O-galloyl-β-D-glucose)
4.2. Cell Line and Cell Culture
4.3. Cell Viability Assay
4.4. Colony Formation Assay
4.5. Apoptosis Assay by Annexin V-FITC/PI Double Staining
4.6. Mitochondrial Membrane Potential
4.7. Wound Healing Assay
4.8. Transwell Migration Assay
4.9. Western Blot Analysis
4.10. Immunofluorescence
4.11. Statistical Analysis
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Yin, L.; Duan, J.J.; Bian, X.W.; Yu, S.C. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 2020, 22, 61. [Google Scholar] [CrossRef]
- Cao, L.; Niu, Y. Triple negative breast cancer: Special histological types and emerging therapeutic methods. Cancer Biol. Med. 2020, 17, 293–306. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.D.; Li, J.; Du, L.; Mahdi, F.; Le, T.P.; Chen, W.L.; Swanson, S.M.; Watabe, K.; Nagle, D.G. Biochemical and anti-triple negative metastatic breast tumor cell properties of Psammaplins. Mar. Drugs 2018, 16, 442. [Google Scholar] [CrossRef]
- Nedeljković, M.; Damjanović, A. Mechanisms of chemotherapy resistance in triple-negative breast cancer-How we can rise to the challenge. Cells 2019, 8, 957. [Google Scholar] [CrossRef] [PubMed]
- Nath, A.; Mitra, S.; Mistry, T.; Pal, R.; Nasare, V.D. Molecular targets and therapeutics in chemoresistance of triple-negative breast cancer. Med. Oncol. 2021, 39, 14. [Google Scholar] [CrossRef]
- Cappetta, D.; De Angelis, A.; Sapio, L.; Prezioso, L.; Illiano, M.; Quaini, F.; Rossi, F.; Berrino, L.; Naviglio, S.; Urbanek, K. Oxidative stress and cellular response to doxorubicin: A common factor in the complex milieu of anthracycline cardiotoxicity. Oxid. Med. Cell. Longev. 2017, 2017, 1521020. [Google Scholar] [CrossRef]
- Ye, S.; Chen, S.; Yang, X.; Lei, X. Drug resistance in breast cancer is based on the mechanism of exocrine non-coding RNA. Discov. Onc. 2024, 15, 138. [Google Scholar] [CrossRef]
- Roche, J. The epithelial-to-mesenchymal transition in cancer. Cancers 2018, 10, 52. [Google Scholar] [CrossRef] [PubMed]
- Sistigu, A.; Di Modugno, F.; Manic, G.; Nisticò, P. Deciphering the loop of epithelial-mesenchymal transition, inflammatory cytokines and cancer immunoediting. Cytokine Growth Factor Rev. 2017, 36, 67–77. [Google Scholar] [CrossRef]
- Serrano-Gomez, S.J.; Maziveyi, M.; Alahari, S.K. Regulation of epithelial-mesenchymal transition through epigenetic and post-translational modifications. Mol. Cancer 2016, 15, 18. [Google Scholar] [CrossRef]
- Huang, Y.; Hong, W.; Wei, X. The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J. Hematol. Oncol. 2022, 15, 129. [Google Scholar] [CrossRef] [PubMed]
- Weadick, B.; Nayak, D.; Persaud, A.K.; Hung, S.W.; Raj, R.; Campbell, M.J.; Chen, W.; Li, J.; Williams, T.M.; Govindarajan, R. EMT-induced gemcitabine resistance in pancreatic cancer involves the functional loss of equilibrative nucleoside transporter 1. Mol. Cancer Ther. 2021, 20, 410–422. [Google Scholar] [CrossRef] [PubMed]
- Luo, M.; Wu, C.; Guo, E.; Peng, S.; Zhang, L.; Sun, W.; Liu, D.; Hu, G.; Hu, G. FOXO3a knockdown promotes radioresistance in nasopharyngeal carcinoma by inducing epithelial-mesenchymal transition and the Wnt/β-catenin signaling pathway. Cancer Lett. 2019, 455, 26–35. [Google Scholar] [CrossRef] [PubMed]
- Takaoka, Y.; Konno, M.; Koseki, J.; Colvin, H.; Asai, A.; Tamari, K.; Satoh, T.; Mori, M.; Doki, Y.; Ogawa, K.; et al. Mitochondrial pyruvate carrier 1 expression controls cancer epithelial-mesenchymal transition and radioresistance. Cancer Sci. 2019, 110, 1331–1339. [Google Scholar] [CrossRef]
- Mallini, P.; Lennard, T.; Kirby, J.; Meeson, A. Epithelial-to-mesenchymal transition: What is the impact on breast cancer stem cells and drug resistance. Cancer Treat. Rev. 2014, 40, 341–348. [Google Scholar] [CrossRef] [PubMed]
- Duran, G.E.; Wang, Y.C.; Moisan, F.; Francisco, E.B.; Sikic, B.I. Decreased levels of baseline and drug-induced tubulin polymerisation are hallmarks of resistance to taxanes in ovarian cancer cells and are associated with epithelial-to-mesenchymal transition. Br. J. Cancer 2017, 116, 1318–1328. [Google Scholar] [CrossRef]
- Garg, P.; Malhotra, J.; Kulkarni, P.; Horne, D.; Salgia, R.; Singhal, S.S. Emerging therapeutic strategies to overcome drug resistance in cancer cells. Cancers 2024, 16, 2478. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.J.; Fang, C.B.; Wang, S.S.; Chen, X.Q.; Li, Y.; Liu, Q.; Qi, Y.K.; Du, S.S. Design and synthesis of TH19P01-Camptothecin based hybrid peptides inducing effective anticancer responses on sortilin positive cancer cells. Bioorg. Med. Chem. 2024, 111, 117869. [Google Scholar] [CrossRef] [PubMed]
- Fu, X.Y.; Yin, H.; Chen, X.T.; Yao, J.F.; Ma, Y.N.; Song, M.; Xu, H.; Yu, Q.Y.; Du, S.S.; Qi, Y.K.; et al. Three rounds of stability-guided optimization and systematical evaluation of oncolytic peptide LTX-315. J. Med. Chem. 2024, 67, 3885–3908. [Google Scholar] [CrossRef] [PubMed]
- Fu, C.; Yu, L.; Miao, Y.; Liu, X.; Yu, Z.; Wei, M. Peptide-drug conjugates (PDCs): A novel trend of research and development on targeted therapy, hype or hope? Acta Pharm. Sin. B 2023, 13, 498–516. [Google Scholar] [CrossRef]
- Wang, M.; Liu, J.; Xia, M.; Yin, L.; Zhang, L.; Liu, X.; Cheng, Y. Peptide-drug conjugates: A new paradigm for targeted cancer therapy. Eur. J. Med. Chem. 2024, 265, 116119. [Google Scholar] [CrossRef] [PubMed]
- Jakobušić Brala, C.; Karković Marković, A.; Kugić, A.; Torić, J.; Barbarić, M. Combination chemotherapy with selected polyphenols in preclinical and clinical studies—An update overview. Molecules 2023, 28, 3746. [Google Scholar] [CrossRef] [PubMed]
- Mitra, S.; Tareq, A.; Das, R.; Emran, T.; Nainu, F.; Chakraborty, A.; Ahmad, I.; Tallei, T.; Idris, A.; Simal-Gandara, J. Polyphenols: A first evidence in the synergism and bioactivities. Food Rev. Int. 2022, 39, 4419–4441. [Google Scholar] [CrossRef]
- Vladu, A.F.; Ficai, D.; Ene, A.G.; Ficai, A. Combination therapy using polyphenols: An efficient way to improve antitumoral activity and reduce resistance. Int. J. Mol. Sci. 2022, 23, 10244. [Google Scholar] [CrossRef]
- Wen, C.; Dechsupa, N.; Yu, Z.; Zhang, X.; Liang, S.; Lei, X.; Xu, T.; Gao, X.; Hu, Q.; Innuan, P.; et al. Pentagalloyl glucose: A review of anticancer properties, molecular targets, mechanisms of action, pharmacokinetics, and safety profile. Molecules 2023, 28, 4856. [Google Scholar] [CrossRef] [PubMed]
- Yang, H.; Yue, G.G.; Leung, P.C.; Wong, C.K.; Zhang, Y.J.; Lau, C.B. Anti-metastatic effects of 1,2,3,4,6-Penta-O-galloyl-β-D-glucose in colorectal cancer: Regulation of cathepsin B-mediated extracellular matrix dynamics and epithelial-to-mesenchymal transition. Pharmacol. Res. 2022, 184, 106457. [Google Scholar] [CrossRef] [PubMed]
- Fan, C.W.; Tang, J.; Jiang, J.C.; Zhou, M.M.; Li, M.S.; Wang, H.S. Pentagalloylglucose suppresses the growth and migration of human nasopharyngeal cancer cells via the GSK3β/β-catenin pathway in vitro and in vivo. Phytomedicine 2022, 102, 154192. [Google Scholar] [CrossRef]
- Mendonca, P.; Alghamdi, S.; Messeha, S.; Soliman, K.F.A. Pentagalloyl glucose inhibits TNF-α-activated CXCL1/GRO-α expression and induces apoptosis-related genes in triple-negative breast cancer cells. Sci. Rep. 2021, 11, 5649. [Google Scholar] [CrossRef] [PubMed]
- Kantapan, J.; Paksee, S.; Duangya, A.; Sangthong, P.; Roytrakul, S.; Krobthong, S.; Suttana, W.; Dechsupa, N. A radiosensitizer, gallotannin-rich extract from Bouea macrophylla seeds, inhibits radiation-induced epithelial-mesenchymal transition in breast cancer cells. BMC Complement. Med. Ther. 2021, 21, 189. [Google Scholar] [CrossRef]
- Kantapan, J.; Intachai, N.; Khamto, N.; Meepowpan, P.; Sangthong, P.; Wantanajittikul, K.; Dechsupa, N.; Chitapanarux, I. Pentagalloyl glucose and cisplatin combination treatment exhibits a synergistic anticancer effect in 2D and 3D models of head and neck carcinoma. Pharmaceuticals 2022, 15, 830. [Google Scholar] [CrossRef]
- Ryu, H.G.; Jeong, S.J.; Kwon, H.Y.; Lee, H.J.; Lee, E.O.; Lee, M.H.; Choi, S.H.; Ahn, K.S.; Kim, S.H. Penta-O-galloyl-β-D-glucose attenuates cisplatin-induced nephrotoxicity via reactive oxygen species reduction in renal epithelial cells and enhances antitumor activity in Caki-2 renal cancer cells. Toxicol. In Vitro 2012, 26, 206–214. [Google Scholar] [CrossRef]
- Wang, K.; Zhang, C.; Bao, J.; Jia, X.; Liang, Y.; Wang, X.; Chen, M.; Su, H.; Li, P.; Wan, J.B.; et al. Synergistic chemopreventive effects of curcumin and berberine on human breast cancer cells through induction of apoptosis and autophagic cell death. Sci. Rep. 2016, 6, 26064. [Google Scholar] [CrossRef] [PubMed]
- To, S.Q.; Dmello, R.S.; Richards, A.K.; Ernst, M.; Chand, A.L. STAT3 signaling in breast cancer: Multicellular actions and therapeutic potential. Cancers 2022, 14, 429. [Google Scholar] [CrossRef]
- Ma, J.H.; Qin, L.; Li, X. Role of STAT3 signaling pathway in breast cancer. Cell Commun. Signal. 2020, 18, 33. [Google Scholar] [CrossRef] [PubMed]
- Gu, Y.; Mohammad, I.S.; Liu, Z. Overview of the STAT-3 signaling pathway in cancer and the development of specific inhibitors. Oncol. Lett. 2020, 19, 2585–2594. [Google Scholar] [CrossRef]
- Zhang, G.; Hou, S.; Li, S.; Wang, Y.; Cui, W. Role of STAT3 in cancer cell epithelial-mesenchymal transition (Review). Int. J. Oncol. 2024, 64, 48. [Google Scholar] [CrossRef] [PubMed]
- Rizeq, B.; Gupta, I.; Ilesanmi, J.; AlSafran, M.; Rahman, M.M.; Ouhtit, A. The power of phytochemicals combination in cancer chemoprevention. J. Cancer 2020, 11, 4521–4533. [Google Scholar] [CrossRef] [PubMed]
- Rahman, M.A.; Hannan, M.A.; Dash, R.; Rahman, M.H.; Islam, R.; Uddin, M.J.; Sohag, A.A.M.; Rahman, M.H.; Rhim, H. Phytochemicals as a complement to cancer chemotherapy: Pharmacological modulation of the autophagy-apoptosis pathway. Front. Pharmacol. 2021, 12, 639628. [Google Scholar] [CrossRef] [PubMed]
- Chai, Y.; Lee, H.J.; Shaik, A.A.; Nkhata, K.; Xing, C.; Zhang, J.; Jeong, S.J.; Kim, S.H.; Lu, J. Penta-O-galloyl-β-D-glucose induces G1 arrest and DNA replicative S-phase arrest independently of P21 cyclin-dependent kinase inhibitor 1A, P27 cyclin-dependent kinase inhibitor 1B and P53 in human breast cancer cells and is orally active against triple-negative xenograft growth. Breast Cancer Res. 2010, 12, R67. [Google Scholar] [CrossRef] [PubMed]
- Chen, R.H.; Yang, L.J.; Hamdoun, S.; Chung, S.K.; Lam, C.W.; Zhang, K.X.; Guo, X.; Xia, C.; Law, B.Y.K.; Wong, V.K.W. 1,2,3,4,6-Pentagalloyl glucose, a RBD-ACE2 binding inhibitor to prevent SARS-CoV-2 infection. Front. Pharmacol. 2021, 12, 634176. [Google Scholar] [CrossRef] [PubMed]
- Ding, X.Q.; Zhao, S.; Wang, J.Y.; Zheng, H.C.; Ma, C.M. Inhibitory effects and molecular mechanisms of pentagalloyl glucose in combination with 5-FU on aggressive phenotypes of HepG2 cells. Nat. Prod. Res. 2021, 35, 815–818. [Google Scholar] [CrossRef] [PubMed]
- Sangweni, N.F.; Gabuza, K.; Huisamen, B.; Mabasa, L.; van Vuuren, D.; Johnson, R. Molecular insights into the pathophysiology of doxorubicin-induced cardiotoxicity: A graphical representation. Arch. Toxicol. 2022, 96, 1541–1550. [Google Scholar] [CrossRef] [PubMed]
- Gogvadze, V.; Orrenius, S.; Zhivotovsky, B. Multiple pathways of cytochrome c release from mitochondria in apoptosis. Biochim. Biophys. Acta (BBA) Bioenerg. 2006, 1757, 639–647. [Google Scholar] [CrossRef]
- Deschênes-Simard, X.; Malleshaiah, M.; Ferbeyre, G. Extracellular signal-regulated kinases: One pathway, multiple fates. Cancers 2023, 16, 95. [Google Scholar] [CrossRef] [PubMed]
- Taylor, C.A.; Zheng, Q.; Liu, Z.; Thompson, J.E. Role of p38 and JNK MAPK signaling pathways and tumor suppressor p53 on induction of apoptosis in response to Ad-eIF5A1 in A549 lung cancer cells. Mol. Cancer 2013, 12, 35. [Google Scholar] [CrossRef] [PubMed]
- Karagiannis, G.S.; Condeelis, J.S.; Oktay, M.H. Chemotherapy-induced metastasis: Mechanisms and translational opportunities. Clin. Exp. Metastasis 2018, 35, 269–284. [Google Scholar] [CrossRef] [PubMed]
- Yang, Q.; Huang, J.; Wu, Q.; Cai, Y.; Zhu, L.; Lu, X.; Chen, S.; Chen, C.; Wang, Z. Acquisition of epithelial-mesenchymal transition is associated with Skp2 expression in paclitaxel-resistant breast cancer cells. Br. J. Cancer 2014, 110, 1958–1967. [Google Scholar] [CrossRef] [PubMed]
- Mirzaei, S.; Abadi, A.J.; Gholami, M.H.; Hashemi, F.; Zabolian, A.; Hushmandi, K.; Zarrabi, A.; Entezari, M.; Aref, A.R.; Khan, H.; et al. The involvement of epithelial-to-mesenchymal transition in doxorubicin resistance: Possible molecular targets. Eur. J. Pharmacol. 2021, 908, 174344. [Google Scholar] [CrossRef] [PubMed]
- Kantapan, J.; Dechsupa, N.; Tippanya, D.; Nobnop, W.; Chitapanarux, I. Gallotannin from Bouea macrophylla seed extract suppresses cancer stem-like cells and radiosensitizes head and neck cancer. Int. J. Mol. Sci. 2021, 22, 9253. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kantapan, J.; Innuan, P.; Kongkarnka, S.; Sangthong, P.; Dechsupa, N. Pentagalloyl Glucose from Bouea macrophylla Suppresses the Epithelial–Mesenchymal Transition and Synergizes the Doxorubicin-Induced Anticancer and Anti-Migration Effects in Triple-Negative Breast Cancer. Pharmaceuticals 2024, 17, 1729. https://doi.org/10.3390/ph17121729
Kantapan J, Innuan P, Kongkarnka S, Sangthong P, Dechsupa N. Pentagalloyl Glucose from Bouea macrophylla Suppresses the Epithelial–Mesenchymal Transition and Synergizes the Doxorubicin-Induced Anticancer and Anti-Migration Effects in Triple-Negative Breast Cancer. Pharmaceuticals. 2024; 17(12):1729. https://doi.org/10.3390/ph17121729
Chicago/Turabian StyleKantapan, Jiraporn, Phattarawadee Innuan, Sarawut Kongkarnka, Padchanee Sangthong, and Nathupakorn Dechsupa. 2024. "Pentagalloyl Glucose from Bouea macrophylla Suppresses the Epithelial–Mesenchymal Transition and Synergizes the Doxorubicin-Induced Anticancer and Anti-Migration Effects in Triple-Negative Breast Cancer" Pharmaceuticals 17, no. 12: 1729. https://doi.org/10.3390/ph17121729
APA StyleKantapan, J., Innuan, P., Kongkarnka, S., Sangthong, P., & Dechsupa, N. (2024). Pentagalloyl Glucose from Bouea macrophylla Suppresses the Epithelial–Mesenchymal Transition and Synergizes the Doxorubicin-Induced Anticancer and Anti-Migration Effects in Triple-Negative Breast Cancer. Pharmaceuticals, 17(12), 1729. https://doi.org/10.3390/ph17121729