Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2021 Jun 26;17(11):2703-2717.
doi: 10.7150/ijbs.59404. eCollection 2021.

Tagitinin C induces ferroptosis through PERK-Nrf2-HO-1 signaling pathway in colorectal cancer cells

Affiliations

Tagitinin C induces ferroptosis through PERK-Nrf2-HO-1 signaling pathway in colorectal cancer cells

Ruiran Wei et al. Int J Biol Sci. .

Abstract

Rationale: Colorectal cancer (CRC) is a common malignant tumor of the digestive system. However, the efficacy of surgery and chemotherapy is limited. Ferroptosis is an iron- and reactive oxygen species (ROS)-dependent form of regulated cell death (RCD) and plays a vital role in tumor suppression. Ferroptosis inducing agents have been studied extensively as a novel promising way to fight against therapy resistant cancers. The aim of this study is to investigate the mechanism of action of tagitinin C (TC), a natural product, as a novel ferroptosis inducer in tumor suppression. Methods: The response of CRC cells to tagitinin C was assessed by cell viability assay, clonogenic assay, transwell migration assay, cell cycle assay and apoptosis assay. Molecular approaches including Western blot, RNA sequencing, quantitative real-time PCR and immunofluorescence were employed as well. Results: Tagitinin C, a sesquiterpene lactone isolated from Tithonia diversifolia, inhibits the growth of colorectal cancer cells including HCT116 cells, and induced an oxidative cellular microenvironment resulting in ferroptosis of HCT116 cells. Tagitinin C-induced ferroptosis was accompanied with the attenuation of glutathione (GSH) levels and increased in lipid peroxidation. Mechanistically, tagitinin C induced endoplasmic reticulum (ER) stress and oxidative stress, thus activating nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2). As a downstream gene (effector) of Nrf2, heme oxygenase-1 (HO-1) expression increased significantly with the treatment of tagitinin C. Upregulated HO-1 led to the increase in the labile iron pool, which promoted lipid peroxidation, meanwhile tagitinin C showed synergistic anti-tumor effect together with erastin. Conclusion: In summary, we provided the evidence that tagitinin C induces ferroptosis in colorectal cancer cells and has synergistic effect together with erastin. Mechanistically, tagitinin C induces ferroptosis through ER stress-mediated activation of PERK-Nrf2-HO-1 signaling pathway. Tagitinin C, identified as a novel ferroptosis inducer, may be effective chemosensitizer that can expand the efficacy and range of chemotherapeutic agents.

Keywords: ER stress; Nrf2-HO-1 pathway; ROS; ferroptosis; tagitinin C.

PubMed Disclaimer

Conflict of interest statement

Competing Interests: The authors have declared that no competing interest exists.

Figures

Figure 1
Figure 1
Tagitinin C suppresses cell growth and induces cell death. (A) The chemical structure of tagitinin C. (B-D) Cell viability of SW480, DLD1, and HCT116 cells were measured by MTT assay after treatment with indicate concentration of tagitinin C (5, 10, 20 µM) at 12, 24, 48, 72 h. (E) Cell morphology of SW480, DLD1, and HCT116 after treatment with concentration of tagitinin C (20 µM) at 0, 12, 24, 48, and 72 h (magnification, ×10). Data were presented as Mean ± SD. Scale bar indicates 40 µm. Statistical analysis was carried out between tagitinin C-treated group and DMSO group: * p ≤ 0.05, **p ≤ 0.01, *** p ≤ 0.001.
Figure 2
Figure 2
Tagitinin C inhibits the colony formation, cell migration abilities and induces G2/M cell cycle arrest in HCT116 cells. (A-B) Tagitinin C inhibited the colony formation ability of HCT116 cells in a dose-dependent manner. (A) Representative images of cell colonies after 14 days treatment tagitinin C (1, 1.5, 2 µM). (B) Bar graph shows the quantification of colonies numbers by measuring absorbance at 540 nm. (C-D) Tagitinin C inhibited the migration ability of HCT116 cells. (C) Transwell migration assay was performed in HCT116 cells treated with tagitinin C (5, 10, 20 µM) and representative images were shown. (D) Quantitative analysis for number of migrated HCT116 cells. (E-F) Tagitinin C induced G2/M cell cycle arrest in HCT116 cells. Representative images (E) and quantifications of populations of HCT116 cells in the cell cycle (F) after cells were treated with increasing concentration of tagitinin C (5, 10, 20 µM) for 24 h. Data were presented as Mean ± SD. Scale bar indicates 100 µm. Statistical analysis was carried out between tagitinin C-treated group and DMSO group: * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001.
Figure 3
Figure 3
Tagitinin C induces ferroptosis in HCT116 cells. (A-B) Flow cytometric analysis of apoptosis in the HCT116 cells treated with tagitinin C (20 µM) after 0, 12, 24, 48 h. Image (A) and quantitative analysis for apoptotic cells (B) were presented. (C) Cell viability was measured using CCK-8 assay in control cells and cells treated with tagitinin C (20 µM) with or without Fer-1 (1 µM), DFO (5 µM), Z-VAD-FMK (20 µM), 3-MA (2 mM), and Nec-1 (50 µM) at 12 h. (D) Fer-1 (1 µM) and DFO (5 µM) rescued tagitinin C-induced cell death at 12 h. (E) Fer-1 and DFO blocked tagitinin C-induced lipid peroxidation, quantified using C11-BODIPY lipid probe using flow cytometry in HCT116 cells. (F) Quantification of cellular MDA levels using the TBA method. (G) Quantification of cellular LIP levels using the calcein-AM (C-AM) method. The mean fluorescence intensity (MFI) of C-AM is subtracted from the MFI of C-AM treated with DFO. Data were presented as Mean ± SD. Statistical analysis was carried out between tagitinin C-treated group and DMSO group: * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001.
Figure 4
Figure 4
Tagitinin C induces ROS generation and contributes to mitochondrial dysfunction. (A-C) DCFH-DA probe was used to detected ROS levels in HCT116 cells by Incucyte S3 (A), flow cytometry (B) and fluorescence microscope (C) (magnification, ×20) respectively. (D-E) Tagitinin C (20 µM) increased the level of lipid peroxidation (D), MDA (E) and LIP (F), which could be blocked by NAC (200 µM). (G) Cell viability was measured using CCK-8 assay in control cells and cells treated with tagitinin C with or without NAC (200 µM) at 12 h. (H) Quantification of the reduced cellular GSH levels using the MCB method. (I) Tagitinin C (20 µM) decreased the level of GSH, which could be blocked by NAC (200 µM). (J) Tagitinin C (20 µM) decreased the TMRE fluorescence in HCT116 cells, which could be blocked by NAC (200 µM), Fer-1 (1µM) and DFO (5 µM). Data were presented as Mean ± SD. Scale bar indicates 100 µm. Statistical analysis was carried out between tagitinin C-treated group and DMSO group: * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001.
Figure 5
Figure 5
Tagitinin C activates Nrf2-HO-1 signaling pathway. (A) Heat map showed differentially up-regulated genes in response to tagitinin C treatment measured using RNA-seq analysis. Low expression is depicted in white, and high expression is depicted in black. (B-C) Nrf2 and HO-1 mRNA was measured at indicated concentrations or time after treatment of tagitinin C. Data were presented as Mean ± SD. Statistical analysis was carried out between tagitinin C-treated group and DMSO group: * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001.
Figure 6
Figure 6
Tagitinin C induces ER stress, which mediates Nrf2-HO-1 activation. (A) HCT116 cells stimulated ER stress. HCT116 cells were treated with 20 µM tagitinin C for the indicated time. ER stress-related proteins were determination by Western blot analysis. (B) Cell viability was measured using CCK-8 assay in control cells and cells treated with tagitinin C (20 µM) with/without 4-PBA (20 µM). (C) HCT116 cells were treated with tagitinin C (20 µM) with 4-PBA (20 µM) for 6 h, the cells were collected and used for Nrf2 and HO-1 mRNA determination. (D) HCT116 cells were treated with 20 µM tagitinin C for the indicated time intervals. Cells were collected for PERK, eIF2a, Nrf2, and HO-1 determination by Western blot analysis. (E) Cell viability was measured using CCK-8 assay in control cells and cells treated with tagitinin C (20 µM) with/without GSK2606414 (5 µM). (F) HCT116 cells were treated with 20 µM tagitinin C and/or GSK2606414 for 8 h, the cells were collected and used for Western blot analysis. (G) HCT116 cells were treated with tagitinin C (20 µM) with GSK2606414 for 6 h, the cells were collected and used for Nrf2 and HO-1 mRNA determination. (H) HCT116 cells were grown on coverslips and treated with tagitinin C. Tagitinin C-induced Nrf2 nuclear translocation was observed under confocal microscope by IF. Cells were stained with Nrf2 (red) and DAPI (blue) (magnification, ×100). Data were presented as Mean ± SD. Scale bar indicates 100 µm. Statistical analysis was carried out between tagitinin C-treated group and DMSO group: * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001.
Figure 7
Figure 7
Combination of erastin and tagitinin C synergistically induces cell death. (A) Cell viability was measured using MTT assay in HCT116 cells treated with tagitinin C with or without erastin at 12, 24, 36 and 48 h. (B) The HCT116 cell morphology after treatment with concentration of tagitinin C (10 µM) and/or erastin (20 µM) at 0, 12, 24, 36, 48 h (magnification, ×10). (C) The contents of the cellular ROS. (D-F) The contents of the cellular lipid peroxidation (D), MDA (E), LIP (F) and GSH (G) in HCT116 cells at 12 h under tagitinin C (10 μM) and/or erastin (20 μM) were determined. (H) HCT116 cells were treated with tagitinin C (10 µM) and/or erastin (20 μM) for 12 h, the cells were collected and used for Western blot analysis. (I) HCT116 cells were treated with tagitinin C (10 µM) and/or erastin (20 µM) for 6 h, the cells were collected and mRNA level of Nrf2 and HO-1 were determination. Data were presented as Mean ± SD. Scale bar indicates 40 µm. Statistical analysis was carried out between tagitinin C-treated group and DMSO group: * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001.
Figure 8
Figure 8
A proposed model showing that tagitinin C induces ferroptosis in HCT116 cells.

References

    1. Brody H, Grayson M, Miller K, Bender E, Haines N. Colorectal cancer. Nature. 2015;521:S1. - PubMed
    1. Weitz J, Koch M, Debus J, Höhler T, Galle PR, Büchler MW. Colorectal cancer. Lancet. 2005;365:153–165. - PubMed
    1. Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, Diaz LA Jr. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nature Reviews Gastroenterology & Hepatology. 2019;16:361–375. - PMC - PubMed
    1. Kopetz S. New therapies and insights into the changing landscape of colorectal cancer. Nature Reviews Gastroenterology & Hepatology. 2019;16:79–80. - PubMed
    1. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE. et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 2012;149:1060–1072. - PMC - PubMed

Publication types

MeSH terms