Inhibition of TFAM-Mediated Mitophagy by Oroxylin A Restored Sorafenib Sensitivity Under Hypoxia Conditions in HepG2 Cells
"> Figure 1
<p>TFAM contributed to hypoxia-induced resistance in HepG2 cells. (<b>A</b>) HepG2 cells were cultured in complete medium containing 50 μM CoCl<sub>2</sub> to establish a hypoxia model. HIF1α levels were assessed and analyzed by Western blot in the presence or absence of CoCl<sub>2</sub> solution. (<b>B</b>) After treatment with sorafenib for 24 h, the impact of sorafenib on HepG2 cell viability was evaluated using an MTT assay under hypoxic and normoxic conditions, respectively. (<b>C</b>) DCFH-DA flow cytometry was employed to measure ROS levels in the presence of sorafenib in hypoxic and normoxic environments. ROS levels under normoxic conditions as a control. (<b>D</b>) With the addition of 2 mmol/L NAC for 1 h under normal conditions, the influence of sorafenib on HepG2 cell viability was determined via the MTT assay. (<b>E</b>) The TCGA database was analyzed to identify differences in TFAM expression between liver cancer tissues and adjacent non-tumor tissues. (<b>F</b>) Patients with high TFAM expression in liver cancer exhibited worse prognosis. (<b>G</b>) RT-PCR showed that TFAM mRNA significantly increased in HepG2 cells under hypoxic conditions. GAPDH mRNA was used to normalize the mRNA level of each gene. (<b>H</b>) TFAM was knocked down by TFAMsiRNA, then HepG2 cells were treated with 10 μM sorafenib for 24 h under hypoxic conditions. DCFH-DA flow cytometry revealed that knockdown of TFAM enhanced the ROS levels in sorafenib-treated HepG2 cells in the resistance model. (<b>I</b>) The plate cloning assay showed that TFAM siRNA significantly decreased colonies of HepG2 cells treated with sorafenib for 24 h under hypoxic conditions. Data are expressed as mean ± SEM. * <span class="html-italic">p</span> < 0.05, ** <span class="html-italic">p</span> < 0.01, and *** <span class="html-italic">p</span> < 0.001 versus control.</p> "> Figure 1 Cont.
<p>TFAM contributed to hypoxia-induced resistance in HepG2 cells. (<b>A</b>) HepG2 cells were cultured in complete medium containing 50 μM CoCl<sub>2</sub> to establish a hypoxia model. HIF1α levels were assessed and analyzed by Western blot in the presence or absence of CoCl<sub>2</sub> solution. (<b>B</b>) After treatment with sorafenib for 24 h, the impact of sorafenib on HepG2 cell viability was evaluated using an MTT assay under hypoxic and normoxic conditions, respectively. (<b>C</b>) DCFH-DA flow cytometry was employed to measure ROS levels in the presence of sorafenib in hypoxic and normoxic environments. ROS levels under normoxic conditions as a control. (<b>D</b>) With the addition of 2 mmol/L NAC for 1 h under normal conditions, the influence of sorafenib on HepG2 cell viability was determined via the MTT assay. (<b>E</b>) The TCGA database was analyzed to identify differences in TFAM expression between liver cancer tissues and adjacent non-tumor tissues. (<b>F</b>) Patients with high TFAM expression in liver cancer exhibited worse prognosis. (<b>G</b>) RT-PCR showed that TFAM mRNA significantly increased in HepG2 cells under hypoxic conditions. GAPDH mRNA was used to normalize the mRNA level of each gene. (<b>H</b>) TFAM was knocked down by TFAMsiRNA, then HepG2 cells were treated with 10 μM sorafenib for 24 h under hypoxic conditions. DCFH-DA flow cytometry revealed that knockdown of TFAM enhanced the ROS levels in sorafenib-treated HepG2 cells in the resistance model. (<b>I</b>) The plate cloning assay showed that TFAM siRNA significantly decreased colonies of HepG2 cells treated with sorafenib for 24 h under hypoxic conditions. Data are expressed as mean ± SEM. * <span class="html-italic">p</span> < 0.05, ** <span class="html-italic">p</span> < 0.01, and *** <span class="html-italic">p</span> < 0.001 versus control.</p> "> Figure 2
<p>Silencing TFAM enhanced the sensitivity of HepG2 cells to sorafenib by inhibiting mitophagy. The changes in mitochondria were observed after sorafenib intervention for 24 h in HepG2 cells, with or without TFAM knockdown. (<b>A</b>) Upon TFAM knockdown, the mitochondrial morphology was detected by TEM in sorafenib-treated HepG2 cells under hypoxia conditions. The black arrows indicated normal mitochondria, and the red arrows indicated the damaged mitochondria. The left scale bar: 2.0 μm; the right scale bar: 500 nm. (<b>B</b>) Mitophagy-related proteins (parkin and pink1) were analyzed by Western blot. (<b>C</b>) Flow cytometry measured the effect of TFAM knockdown on MMP levels in hypoxia. HepG2 cells were treated with 5 μM CCCP for 24 h. The experiment was repeated three times. (<b>D</b>) Plate cloning assays showed that combination of 5 μM CCCP and TFAM knockdown under hypoxia conditions significantly promoted the inhibitory effect on cell proliferation. Data are expressed as mean ± SEM, where * <span class="html-italic">p</span> < 0.05, ** <span class="html-italic">p</span> < 0.01, and *** <span class="html-italic">p</span> < 0.001 denote statistical significance.</p> "> Figure 3
<p>OA enhanced the sensitivity of HepG2 cells to sorafenib by inhibiting mitophagy. HepG2 cells were treated with different concentrations of OA or 10 μM sorafenib for 24 h under hypoxia conditions. (<b>A</b>) The MTT assay determined the effect of OA on the viability of HepG2 cells. (<b>B</b>) The inhibitory effects of OA (10 μM) and sorafenib (10 μM), either individually or in combination, on HepG2 cells were evaluated under hypoxia conditions. (<b>C</b>) The DCFH-DA assay determined the effect of OA (10 μM) and sorafenib (10 μM), either individually or in combination, on ROS levels. (<b>D</b>) Flow cytometry detected the effect of OA (10 μM) and sorafenib (10 μM), either individually or in combination, on MMP levels. (<b>E</b>) The mitochondrial count was evaluated using Mito-Tracker Red CMXRos after individual or combined treatments. Scale bar: 10 μm. (<b>F</b>) The levels of mitophagy-related proteins were determined by Western blot after individual or combined treatments. (<b>G</b>) HepG2 cells were transiently transfected with the Cox8-EGFP-mCherry plasmid and subsequently treated with OA, either alone or in combination with sorafenib for 24 h. The Cox8-EGFP-mCherry dual fluorescence reporter system was analyzed using confocal microscopy. Both OA treatment alone and the combined treatment with sorafenib markedly enhanced the green fluorescence intensity (EGFP). Scale bar: 2.5 μm. (<b>H</b>) Laser confocal microscopy assessed the colocalization of mitochondria and lysosomes. Scale bar: 10 μm. Data are expressed as mean ± SEM, where * <span class="html-italic">p</span> < 0.05, ** <span class="html-italic">p</span> < 0.01, and *** <span class="html-italic">p</span> < 0.001 denote statistical significance.</p> "> Figure 3 Cont.
<p>OA enhanced the sensitivity of HepG2 cells to sorafenib by inhibiting mitophagy. HepG2 cells were treated with different concentrations of OA or 10 μM sorafenib for 24 h under hypoxia conditions. (<b>A</b>) The MTT assay determined the effect of OA on the viability of HepG2 cells. (<b>B</b>) The inhibitory effects of OA (10 μM) and sorafenib (10 μM), either individually or in combination, on HepG2 cells were evaluated under hypoxia conditions. (<b>C</b>) The DCFH-DA assay determined the effect of OA (10 μM) and sorafenib (10 μM), either individually or in combination, on ROS levels. (<b>D</b>) Flow cytometry detected the effect of OA (10 μM) and sorafenib (10 μM), either individually or in combination, on MMP levels. (<b>E</b>) The mitochondrial count was evaluated using Mito-Tracker Red CMXRos after individual or combined treatments. Scale bar: 10 μm. (<b>F</b>) The levels of mitophagy-related proteins were determined by Western blot after individual or combined treatments. (<b>G</b>) HepG2 cells were transiently transfected with the Cox8-EGFP-mCherry plasmid and subsequently treated with OA, either alone or in combination with sorafenib for 24 h. The Cox8-EGFP-mCherry dual fluorescence reporter system was analyzed using confocal microscopy. Both OA treatment alone and the combined treatment with sorafenib markedly enhanced the green fluorescence intensity (EGFP). Scale bar: 2.5 μm. (<b>H</b>) Laser confocal microscopy assessed the colocalization of mitochondria and lysosomes. Scale bar: 10 μm. Data are expressed as mean ± SEM, where * <span class="html-italic">p</span> < 0.05, ** <span class="html-italic">p</span> < 0.01, and *** <span class="html-italic">p</span> < 0.001 denote statistical significance.</p> "> Figure 4
<p>OA targeted TFAM to inhibit mitophagy in HepG2 cells. HepG2 cells were treated with OA and 10 μM sorafenib, either individually or in combination, for 24 h under hypoxic conditions. (<b>A</b>) Molecular docking results showed a binding pocket between OA and TFAM in the 3D structure. OA formed hydrogen bonds with TYR211 and LYS145 of TFAM with hydrogen bond lengths of 2.9 and 4.0, respectively. The compound formed hydrophobic interactions with ARG157 and LYS156 of TFAM and π–cation interactions with LYS154 and LYS146 of the protein. (<b>B</b>) Thermal shift assays showed that treatment with OA decreased the degradation rate of TFAM. The thermal melting curve displayed a significant rightward shift following the administration of OA. (<b>C</b>) The effects of OA on TFAM expression were determined and analyzed by Western blot in hypoxia-induced resistance. (<b>D</b>) OA reduced the expression of mitophagy-related proteins in a concentration-dependent manner, which was detected by Western blot. (<b>E</b>) Western blot revealed that either knocking down TFAM or using OA reduced mitophagy-related proteins in HepG2 cells under hypoxia conditions. (<b>F</b>) Western blot analysis indicated that overexpression of TFAM could reverse the OA-mediated inhibition of mitophagy in hypoxia-induced resistant HepG2 cells. (<b>G</b>,<b>I</b>) Laser confocal microscopy analysis revealed that TFAM overexpression could counteract the OA-mediated suppression of mitophagy under hypoxia conditions. Scale bar: 10 μm. (<b>H</b>,<b>J</b>) Flow cytometry analysis showed that overexpression of TFAM could reverse OA-induced downregulation of MMP level in HepG2 cells. Data are expressed as mean ± SEM, where * <span class="html-italic">p</span> < 0.05, ** <span class="html-italic">p</span> < 0.01, and *** <span class="html-italic">p</span> < 0.001 denote statistical significance.</p> "> Figure 5
<p>OA suppressed mitophagy by downregulating TFAM to reduce p53 acetylation under hypoxia conditions. (<b>A</b>) Western blot showed that knocking down TFAM could reduce p53 expression under hypoxia conditions. (<b>B</b>) RT-PCR analysis was used to determine the levels of p53 mRNA in HepG2 cells after TFAM knockdown. (<b>C</b>) Western blot assessed the effects of combining CHX or MG132 with OA on p53 protein. (<b>D</b>) After treatment with CHX in the presence or absence of OA, Western blot analysis evaluated the expression of p53 protein at the indicated time. (<b>E</b>) Co-IP detected the ubiquitination levels of p53 following treatment with either TFAM knockdown or 10 μM OA for 24 h. (<b>F</b>) Western blot investigated the effects of TFAM knockdown on the expression of acetylated p53 protein and its downstream target proteins. (<b>G</b>) After treatment with OA for 24 h, Western blot assessed the impact of OA on acetylated p53 and its downstream target protein. (<b>H</b>) Western blot showed that overexpressing TFAM could reverse the effects of OA on p53 acetylation and its downstream target genes. Data are expressed as mean ± SEM, where * <span class="html-italic">p</span> < 0.05, ** <span class="html-italic">p</span> < 0.01, and *** <span class="html-italic">p</span> < 0.001 denote statistical significance.</p> "> Figure 6
<p>OA enhanced the therapeutic effect of sorafenib on xenograft tumor in vivo. (<b>A</b>) Changes in body weight of mice in each group. (<b>B</b>) Quantification of tumor weight in each group. (<b>C</b>) Volume changes of tumors in each group. (<b>D</b>) Immunohistochemical analysis of TFAM expression in tumor tissues of mice in each group. (<b>E</b>) Western blot analysis of the expression of mitophagy-related proteins in tumor tissues. Data are expressed as mean ± SEM, where * <span class="html-italic">p</span> < 0.05, ** <span class="html-italic">p</span> < 0.01, and *** <span class="html-italic">p</span> < 0.001 denote statistical significance.</p> ">
Abstract
:1. Introduction
2. Results
2.1. TFAM Contributed to Hypoxia-Induced Resistance in HepG2 Cells
2.2. Silencing TFAM Enhanced the Sensitivity of HepG2 Cells to Sorafenib by Inhibiting Mitophagy
2.3. OA Enhanced the Sensitivity of HepG2 Cells to Sorafenib by Inhibiting Mitophagy
2.4. OA Targeted TFAM to Inhibit Mitophagy in HepG2 Cells
2.5. OA Suppressed Mitophagy by Downregulating TFAM to Reduce p53 Acetylation Under Hypoxia Conditions
2.6. OA Enhanced the Therapeutic Effect of Sorafenib on Xenograft Tumor In Vivo
3. Discussion
4. Materials and Methods
4.1. Cell Culture and Drug Treatment
4.2. Real-Time PCR Analysis
4.3. Western Blot Analysis
4.4. MTT Assay
4.5. Transmission Electron Microscopy (TEM)
4.6. Cellular Thermal Shift Assay (CETSA)
4.7. Immunohistochemistry
4.8. Colocalization of Mitochondria and Lysosomes
4.9. Co-Immunoprecipitation (Co-IP)
4.10. Cox8-EGFP-mCherry to Monitor Mitophagy
4.11. TCGA Database Analysis
4.12. Plate Colony Formation Assay
4.13. The Molecular Docking
4.14. Animal Experiments
4.15. Measurement of Intracellular Reactive Oxygen Species (ROS) Levels
4.16. Mitochondrial Membrane Potential (MMP) Measurement
4.17. Statistical Analysis
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
References
- Vasan, N.; Baselga, J.; Hyman, D.M. A view on drug resistance in cancer. Nature 2019, 575, 299–309. [Google Scholar] [CrossRef] [PubMed]
- Bukowski, K.; Kciuk, M.; Kontek, R. Mechanisms of Multidrug Resistance in Cancer Chemotherapy. Int. J. Mol. Sci. 2020, 21, 3233. [Google Scholar] [CrossRef] [PubMed]
- Marine, J.C.; Dawson, S.J.; Dawson, M.A. Non-genetic mechanisms of therapeutic resistance in cancer. Nat. Rev. Cancer 2020, 20, 743–756. [Google Scholar] [CrossRef]
- Sweeney, P.L.; Suri, Y.; Basu, A.; Koshkin, V.S.; Desai, A. Mechanisms of tyrosine kinase inhibitor resistance in renal cell carcinoma. Cancer Drug Resist. 2023, 6, 858–873. [Google Scholar] [CrossRef]
- Man, K.F.; Ma, S. Mechanisms of resistance to tyrosine kinase inhibitors in liver cancer stem cells and potential therapeutic approaches. Essays Biochem. 2022, 66, 371–386. [Google Scholar]
- Nagasaki, J.; Ishino, T.; Togashi, Y. Mechanisms of resistance to immune checkpoint inhibitors. Cancer Sci. 2022, 113, 3303–3312. [Google Scholar] [CrossRef] [PubMed]
- Kim, D.W.; Talati, C.; Kim, R. Hepatocellular carcinoma (HCC): Beyond sorafenib-chemotherapy. J. Gastrointest. Oncol. 2017, 8, 256–265. [Google Scholar] [CrossRef] [PubMed]
- Cheng, Z.; Wei-Qi, J.; Jin, D. New insights on sorafenib resistance in liver cancer with correlation of individualized therapy. Biochim. Biophys. Acta Rev. Cancer 2020, 1874, 188382. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.T.; Xiang, D.; Zhao, X.Y.; Chu, X.Y. Upregulation of lncRNA NIFK-AS1 in hepatocellular carcinoma by m6A methylation promotes disease progression and sorafenib resistance. Hum. Cell 2021, 34, 1800–1811. [Google Scholar] [CrossRef]
- Chen, S.; Du, Y.; Guan, X.Y.; Yan, Q. The current status of tumor microenvironment and cancer stem cells in sorafenib resistance of hepatocellular carcinoma. Front. Oncol. 2023, 13, 1204513. [Google Scholar] [CrossRef]
- Chen, Y.; Hao, X.; Sun, R.; Wei, H.; Tian, Z. Natural Killer Cell-Derived Interferon-Gamma Promotes Hepatocellular Carcinoma Through the Epithelial Cell Adhesion Molecule-Epithelial-to-Mesenchymal Transition Axis in Hepatitis B Virus Transgenic Mice. Hepatology 2019, 69, 1735–1750. [Google Scholar] [CrossRef]
- Ul-Islam, S.; Ahmed, M.B.; Shehzad, A.; Ul-Islam, M.; Lee, Y.S. Failure of Chemotherapy in Hepatocellular Carcinoma Due to Impaired and Dysregulated Primary Liver Drug Metabolizing Enzymes and Drug Transport Proteins: What to Do? Curr. Drug Metab. 2018, 19, 819–829. [Google Scholar] [CrossRef]
- Ladd, A.D.; Duarte, S.; Sahin, I.; Zarrinpar, A. Mechanisms of drug resistance in HCC. Hepatology 2024, 79, 926–940. [Google Scholar] [CrossRef]
- Niu, L.; Liu, L.; Yang, S.; Ren, J.; Lai, P.B.S.; Chen, G.G. New insights into sorafenib resistance in hepatocellular carcinoma: Responsible mechanisms and promising strategies. Biochim. Biophys. Acta Rev. Cancer 2017, 1868, 564–570. [Google Scholar] [CrossRef] [PubMed]
- Harada, H. Hypoxia-inducible factor 1-mediated characteristic features of cancer cells for tumor radioresistance. J. Radiat. Res. 2016, 57 (Suppl. S1), i99–i105. [Google Scholar] [CrossRef] [PubMed]
- Wang, Q.; Liu, C. Mitophagy plays a “double-edged sword” role in the radiosensitivity of cancer cells. J. Cancer Res. Clin. Oncol. 2024, 150, 14. [Google Scholar] [CrossRef] [PubMed]
- Uoselis, L.; Nguyen, T.N.; Lazarou, M. Mitochondrial degradation: Mitophagy and beyond. Mol. Cell 2023, 83, 3404–3420. [Google Scholar] [CrossRef]
- Glytsou, C.; Chen, X.; Zacharioudakis, E.; Al-Santli, W.; Zhou, H.; Nadorp, B.; Lee, S.; Lasry, A.; Sun, Z.; Papaioannou, D.; et al. Mitophagy Promotes Resistance to BH3 Mimetics in Acute Myeloid Leukemia. Cancer Discov. 2023, 13, 1656–1677. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Pang, C.; Zhang, C.; Wang, Y.; Wang, P.; Chen, Y.; Wang, J.; Hu, Y.; Liu, C.; Liang, H.; et al. HILPDA-mediated lipidomic remodelling promotes radiotherapy resistance in nasopharyngeal carcinoma by accelerating mitophagy. Cell Mol. Life Sci. 2023, 80, 242. [Google Scholar] [CrossRef]
- Wu, H.; Wang, T.; Liu, Y.; Li, X.; Xu, S.; Wu, C.; Zou, H.; Cao, M.; Jin, G.; Lang, J.; et al. Mitophagy promotes sorafenib resistance through hypoxia-inducible ATAD3A dependent Axis. J. Exp. Clin. Cancer Res. 2020, 39, 274. [Google Scholar] [CrossRef]
- Ponneri Babuharisankar, A.; Kuo, C.L.; Chou, H.Y.; Tangeda, V.; Fan, C.C.; Chen, C.H.; Kao, Y.H.; Lee, A.Y. Mitochondrial Lon-induced mitophagy benefits hypoxic resistance via Ca2+-dependent FUNDC1 phosphorylation at the ER-mitochondria interface. Cell Death Dis. 2023, 14, 199. [Google Scholar] [CrossRef]
- Song, C.; Pan, S.; Zhang, J.; Li, N.; Geng, Q. Mitophagy: A novel perspective for insighting into cancer and cancer treatment. Cell Prolif. 2022, 55, e13327. [Google Scholar] [CrossRef]
- Campbell, C.T.; Kolesar, J.E.; Kaufman, B.A. Mitochondrial transcription factor A regulates mitochondrial transcription initiation, DNA packaging, and genome copy number. Biochim. Biophys. Acta 2012, 1819, 921–929. [Google Scholar] [CrossRef]
- Wang, W.; Jiang, C.F.; Yin, H.S.; Gao, S.; Yu, B.P. Targeting mitochondrial transcription factor A sensitizes pancreatic cancer cell to gemcitabine. Hepatobiliary Pancreat. Dis. Int. 2023, 22, 519–527. [Google Scholar] [CrossRef] [PubMed]
- Baixauli, F.; Acín-Pérez, R.; Villarroya-Beltrí, C.; Mazzeo, C.; Nuñez-Andrade, N.; Gabandé-Rodriguez, E.; Ledesma, M.D.; Blázquez, A.; Martin, M.A.; Falcón-Pérez, J.M.; et al. Mitochondrial Respiration Controls Lysosomal Function during Inflammatory T Cell Responses. Cell Metab. 2015, 22, 485–498. [Google Scholar] [CrossRef] [PubMed]
- Hafner, A.; Bulyk, M.L.; Jambhekar, A.; Lahav, G. The multiple mechanisms that regulate p53 activity and cell fate. Nat. Rev. Mol. Cell Biol. 2019, 20, 199–210. [Google Scholar] [CrossRef] [PubMed]
- Brooks, C.L.; Gu, W. Ubiquitination, phosphorylation and acetylation: The molecular basis for p53 regulation. Curr. Opin. Cell Biol. 2003, 15, 164–171. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Chen, Y.; Chen, Q.; Zhang, X.; Wang, H.; Wang, Z.; Wang, J.; Tian, C. The role of acetylation sites in the regulation of p53 activity. Mol. Biol. Rep. 2020, 47, 381–391. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Luo, J.; Brooks, C.L.; Gu, W. Acetylation of p53 inhibits its ubiquitination by Mdm2. J. Biol. Chem. 2002, 277, 50607–50611. [Google Scholar] [CrossRef] [PubMed]
- Feng, L.; Lin, T.; Uranishi, H.; Gu, W.; Xu, Y. Functional analysis of the roles of posttranslational modifications at the p53 C terminus in regulating p53 stability and activity. Mol. Cell Biol. 2005, 25, 5389–5395. [Google Scholar] [CrossRef]
- Sajeev, A.; Hegde, M.; Girisa, S.; Devanarayanan, T.N.; Alqahtani, M.S.; Abbas, M.; Sil, S.K.; Sethi, G.; Chen, J.T.; Kunnumakkara, A.B. OA: A Promising Flavonoid for Prevention and Treatment of Chronic Diseases. Biomolecules 2022, 12, 1185. [Google Scholar] [CrossRef]
- Li, X.; Miao, H.; Zhang, Y.; Li, W.; Li, Z.; Zhou, Y.; Zhao, L.; Guo, Q. Bone marrow microenvironment confers imatinib resistance to chronic myelogenous leukemia and OA reverses the resistance by suppressing Stat3 pathway. Arch. Toxicol. 2015, 89, 121–136. [Google Scholar] [CrossRef]
- Ding, Y.; Zhou, Y.; Li, Z.; Zhang, H.; Yang, Y.; Qin, H.; Xu, Q.; Zhao, L. Oroxylin A reversed Fibronectin-induced glioma insensitivity to Temozolomide by suppressing IP3R1/AKT/β-catenin pathway. Life Sci. 2020, 260, 118411. [Google Scholar] [CrossRef]
- Zhu, J.; Ao, H.; Liu, M.; Cao, K.; Ma, J. UBE2T promotes autophagy via the p53/AMPK/mTOR signaling pathway in lung adenocarcinoma. J. Transl. Med. 2021, 19, 374. [Google Scholar] [CrossRef]
- Pietrocola, F.; Izzo, V.; Niso-Santano, M.; Vacchelli, E.; Galluzzi, L.; Maiuri, M.C.; Kroemer, G. Regulation of autophagy by stress-responsive transcription factors. Semin. Cancer Biol. 2013, 23, 310–322. [Google Scholar] [CrossRef] [PubMed]
- Donato, M.T.; Tolosa, L.; Gómez-Lechón, M.J. Culture and Functional Characterization of Human Hepatoma HepG2 Cells. Methods Mol. Biol. 2015, 1250, 77–93. [Google Scholar]
- Štancl, P.; Gršković, P.; Držaić, S.; Vičić, A.; Karlić, R.; Korać, P. RNA-Sequencing Identification of Genes Supporting HepG2 as a Model Cell Line for Hepatocellular Carcinoma or Hepatocytes. Genes 2024, 15, 1460. [Google Scholar] [CrossRef] [PubMed]
- Wei, J.; Fang, D. Endoplasmic Reticulum Stress Signaling and the Pathogenesis of Hepatocarcinoma. Int. J. Mol. Sci. 2021, 22, 1799. [Google Scholar] [CrossRef]
- Xiong, X.X.; Qiu, X.Y.; Hu, D.X.; Chen, X.Q. Advances in Hypoxia-Mediated Mechanisms in Hepatocellular Carcinoma. Mol. Pharmacol. 2017, 92, 246–255. [Google Scholar] [CrossRef]
- Gutsaeva, D.R.; Carraway, M.S.; Suliman, H.B.; Demchenko, I.T.; Shitara, H.; Yonekawa, H.; Piantadosi, C.A. Transient hypoxia stimulates mitochondrial biogenesis in brain subcortex by a neuronal nitric oxide synthase-dependent mechanism. J. Neurosci. 2008, 28, 2015–2024. [Google Scholar] [CrossRef]
- Zhu, L.; Wang, Q.; Zhang, L.; Fang, Z.; Zhao, F.; Lv, Z.; Gu, Z.; Zhang, J.; Wang, J.; Zen, K.; et al. Hypoxia induces PGC-1α expression and mitochondrial biogenesis in the myocardium of TOF patients. Cell Res. 2010, 20, 676–687. [Google Scholar] [CrossRef]
- Glick, D.; Barth, S.; Macleod, K.F. Autophagy: Cellular and molecular mechanisms. J. Pathol. 2010, 221, 3–12. [Google Scholar] [CrossRef] [PubMed]
- Mizushima, N. Autophagy: Process and function. Genes Dev. 2007, 21, 2861–2873. [Google Scholar] [CrossRef] [PubMed]
- Tasdemir, E.; Chiara Maiuri, M.; Morselli, E.; Criollo, A.; D’Amelio, M.; Djavaheri-Mergny, M.; Cecconi, F.; Tavernarakis, N.; Kroemer, G. A dual role of p53 in the control of autophagy. Autophagy 2008, 4, 810–814. [Google Scholar] [CrossRef]
- Hsieh, Y.T.; Tu, H.F.; Yang, M.H.; Chen, Y.F.; Lan, X.Y.; Huang, C.L.; Chen, H.M.; Li, W.C. Mitochondrial genome and its regulator TFAM modulates head and neck tumourigenesis through intracellular metabolic reprogramming and activation of oncogenic effectors. Cell Death Dis. 2021, 12, 961. [Google Scholar] [CrossRef] [PubMed]
- Fei, P.; Wang, W.; Kim, S.H.; Wang, S.; Burns, T.F.; Sax, J.K.; Buzzai, M.; Dicker, D.T.; McKenna, W.G.; Bernhard, E.J.; et al. Bnip3L is induced by p53 under hypoxia, and its knockdown promotes tumor growth. Cancer Cell 2004, 6, 597–609. [Google Scholar] [CrossRef] [PubMed]
Gene (Human) | Forward Sequence | Reverse Sequence |
---|---|---|
p53 | GATCAGCAGAGCATTGTTCACATTG | GGGTCGTCGCCTCCAGTTG |
TFAM | CCGAGGTGGTTTTCATCTGT | TATATACCTGCCACTCCGCC |
GADPH | ATTCCACCCATGGCAAATTCC | GACTCCACGACGTACTCAGC |
Antibody | Catalog Number | Company |
---|---|---|
Anti-TFAM | AF0531 | Affinity Biosciences (Liyang, China) |
Anti-p53 | 2524T | CST (Danvers, MA, USA) |
Anti-HIF 1α | AF02369 | AiFang biological (Changsha, China) |
Anti-Acetyl p53 | Ab179484 | Abcam (Cambridge, UK) |
Anti-PINK1 | Ab300623 | Abcam (Cambridge, UK) |
Anti-PARKIN | 14060-1-AP | Proteintech (Wuhan, China) |
Anti-LC3B | AF11004 | AiFang biological (Changsha, China) |
Anti-FAS | AF301026 | AiFang biological (Changsha, China) |
Anti-PUMA | AF300458 | AiFang biological (Changsha, China) |
Anti-β-actin | 20536-1-AP | Proteintech (Wuhan, China) |
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
Ji, S.; Xu, X.; Li, Y.; Sun, S.; Fu, Q.; Qiu, Y.; Wang, S.; Xia, S.; Wang, F.; Zhang, F.; et al. Inhibition of TFAM-Mediated Mitophagy by Oroxylin A Restored Sorafenib Sensitivity Under Hypoxia Conditions in HepG2 Cells. Pharmaceuticals 2024, 17, 1727. https://doi.org/10.3390/ph17121727
Ji S, Xu X, Li Y, Sun S, Fu Q, Qiu Y, Wang S, Xia S, Wang F, Zhang F, et al. Inhibition of TFAM-Mediated Mitophagy by Oroxylin A Restored Sorafenib Sensitivity Under Hypoxia Conditions in HepG2 Cells. Pharmaceuticals. 2024; 17(12):1727. https://doi.org/10.3390/ph17121727
Chicago/Turabian StyleJi, Shufan, Xuefen Xu, Yujia Li, Sumin Sun, Qiuyu Fu, Yangling Qiu, Shuqi Wang, Siwei Xia, Feixia Wang, Feng Zhang, and et al. 2024. "Inhibition of TFAM-Mediated Mitophagy by Oroxylin A Restored Sorafenib Sensitivity Under Hypoxia Conditions in HepG2 Cells" Pharmaceuticals 17, no. 12: 1727. https://doi.org/10.3390/ph17121727
APA StyleJi, S., Xu, X., Li, Y., Sun, S., Fu, Q., Qiu, Y., Wang, S., Xia, S., Wang, F., Zhang, F., Xuan, J., & Zheng, S. (2024). Inhibition of TFAM-Mediated Mitophagy by Oroxylin A Restored Sorafenib Sensitivity Under Hypoxia Conditions in HepG2 Cells. Pharmaceuticals, 17(12), 1727. https://doi.org/10.3390/ph17121727