Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2021

Open Access 01-12-2021 | Breast Cancer | Research

Identification of a small molecule as inducer of ferroptosis and apoptosis through ubiquitination of GPX4 in triple negative breast cancer cells

Authors: Yahui Ding, Xiaoping Chen, Can Liu, Weizhi Ge, Qin Wang, Xin Hao, Mengmeng Wang, Yue Chen, Quan Zhang

Published in: Journal of Hematology & Oncology | Issue 1/2021

Login to get access

Abstract

Background

TNBC is the most aggressive breast cancer with higher recurrence and mortality rate than other types of breast cancer. There is an urgent need for identification of therapeutic agents with unique mode of action for overcoming current challenges in TNBC treatment.

Methods

Different inhibitors were used to study the cell death manner of DMOCPTL. RNA silencing was used to evaluate the functions of GPX4 in ferroptosis and apoptosis of TNBC cells and functions of EGR1 in apoptosis. Immunohistochemical assay of tissue microarray were used for investigating correlation of GPX4 and EGR1 with TNBC. Computer-aided docking and small molecule probe were used for study the binding of DMOCPTL with GPX4.

Results

DMOCPTL, a derivative of natural product parthenolide, exhibited about 15-fold improvement comparing to that of the parent compound PTL for TNBC cells. The cell death manner assay showed that the anti-TNBC effect of DMOCPTL mainly by inducing ferroptosis and apoptosis through ubiquitination of GPX4. The probe of DMOCPTL assay indicated that DMOCPTL induced GPX4 ubiquitination by directly binding to GPX4 protein. To the best of our knowledge, this is the first report of inducing ferroptosis through ubiquitination of GPX4. Moreover, the mechanism of GPX4 regulation of apoptosis is still obscure. Here, we firstly reveal that GPX4 regulated mitochondria-mediated apoptosis through regulation of EGR1 in TNBC cells. Compound 13, the prodrug of DMOCPTL, effectively inhibited the growth of breast tumor and prolonged the lifespan of mice in vivo, and no obvious toxicity was observed.

Conclusions

These findings firstly revealed novel manner to induce ferroptosis through ubiquitination of GPX4 and provided mechanism for GPX4 inducing mitochondria-mediated apoptosis through up-regulation of EGR1 in TNBC cells. Moreover, compound 13 deserves further studies as a lead compound with novel mode of action for ultimate discovery of effective anti-TNBC drug.
Appendix
Available only for authorised users
Literature
1.
go back to reference Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149:1060–72.CrossRef Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149:1060–72.CrossRef
2.
go back to reference Stockwell BR, Friedmann Angeli JP, Bayir H, et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 2017;171:273–85.CrossRef Stockwell BR, Friedmann Angeli JP, Bayir H, et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 2017;171:273–85.CrossRef
3.
go back to reference Yu H, Guo P, Xie X, et al. Ferroptosis, a new form of cell death, and its relationships with tumourous diseases. J Cell Mol Med. 2017;21:648–57.CrossRef Yu H, Guo P, Xie X, et al. Ferroptosis, a new form of cell death, and its relationships with tumourous diseases. J Cell Mol Med. 2017;21:648–57.CrossRef
4.
go back to reference Xu T, Ding W, Ji X, et al. Molecular mechanisms of ferroptosis and its role in cancer therapy. J Cell Mol Med. 2019;23:4900–12.CrossRef Xu T, Ding W, Ji X, et al. Molecular mechanisms of ferroptosis and its role in cancer therapy. J Cell Mol Med. 2019;23:4900–12.CrossRef
5.
go back to reference Mou Y, Wang J, Wu J, et al. Ferroptosis, a new form of cell death: opportunities and challenges in cancer. J Hematol Oncol. 2019;12:34.CrossRef Mou Y, Wang J, Wu J, et al. Ferroptosis, a new form of cell death: opportunities and challenges in cancer. J Hematol Oncol. 2019;12:34.CrossRef
6.
go back to reference Xie Y, Hou W, Song X, et al. Ferroptosis: process and function. Cell Death Differ. 2016;23:369–79.CrossRef Xie Y, Hou W, Song X, et al. Ferroptosis: process and function. Cell Death Differ. 2016;23:369–79.CrossRef
7.
go back to reference Shen Z, Song J, Yung BC, et al. Emerging strategies of cancer therapy based on ferroptosis. Adv Mater. 2018;30:e1704007.CrossRef Shen Z, Song J, Yung BC, et al. Emerging strategies of cancer therapy based on ferroptosis. Adv Mater. 2018;30:e1704007.CrossRef
8.
go back to reference Hassannia B, Vandenabeele P, Vanden BT. Targeting ferroptosis to iron out cancer. Cancer Cell. 2019;35:830–49.CrossRef Hassannia B, Vandenabeele P, Vanden BT. Targeting ferroptosis to iron out cancer. Cancer Cell. 2019;35:830–49.CrossRef
9.
go back to reference Liang C, Zhang X, Yang M, et al. Recent progress in ferroptosis inducers for cancer therapy. Adv Mater. 2019;31:e1904197.CrossRef Liang C, Zhang X, Yang M, et al. Recent progress in ferroptosis inducers for cancer therapy. Adv Mater. 2019;31:e1904197.CrossRef
10.
go back to reference Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat Rev Cancer. 2019;19:405–14.CrossRef Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat Rev Cancer. 2019;19:405–14.CrossRef
11.
go back to reference Lu B, Chen XB, Ying MD, et al. The role of ferroptosis in cancer development and treatment response. Front Pharmacol. 2017;8:992.CrossRef Lu B, Chen XB, Ying MD, et al. The role of ferroptosis in cancer development and treatment response. Front Pharmacol. 2017;8:992.CrossRef
12.
go back to reference Murphy MP. Metabolic control of ferroptosis in cancer. Nat Cell Biol. 2018;20:1104–5.CrossRef Murphy MP. Metabolic control of ferroptosis in cancer. Nat Cell Biol. 2018;20:1104–5.CrossRef
13.
go back to reference Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.CrossRef Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.CrossRef
14.
go back to reference Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7–30.CrossRef Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7–30.CrossRef
15.
go back to reference Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med. 2010;363:1938–48.CrossRef Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med. 2010;363:1938–48.CrossRef
16.
go back to reference Gluz O, Liedtke C, Gottschalk N, et al. Triple-negative breast cancer–current status and future directions. Ann Oncol. 2009;20:1913–27.CrossRef Gluz O, Liedtke C, Gottschalk N, et al. Triple-negative breast cancer–current status and future directions. Ann Oncol. 2009;20:1913–27.CrossRef
17.
go back to reference Penault-Llorca F, Viale G. Pathological and molecular diagnosis of triple-negative breast cancer: a clinical perspective. Ann Oncol. 2012;23(Suppl 6):vi19-22.CrossRef Penault-Llorca F, Viale G. Pathological and molecular diagnosis of triple-negative breast cancer: a clinical perspective. Ann Oncol. 2012;23(Suppl 6):vi19-22.CrossRef
18.
go back to reference Denkert C, Liedtke C, Tutt A, et al. Molecular alterations in triple-negative breast cancer-the road to new treatment strategies. Lancet. 2017;389:2430–42.CrossRef Denkert C, Liedtke C, Tutt A, et al. Molecular alterations in triple-negative breast cancer-the road to new treatment strategies. Lancet. 2017;389:2430–42.CrossRef
19.
go back to reference Yu M, Gai C, Li Z, et al. Targeted exosome-encapsulated erastin induced ferroptosis in triple negative breast cancer cells. Cancer Sci. 2019;110:3173–82.CrossRef Yu M, Gai C, Li Z, et al. Targeted exosome-encapsulated erastin induced ferroptosis in triple negative breast cancer cells. Cancer Sci. 2019;110:3173–82.CrossRef
20.
go back to reference Yu H, Yang C, Jian L, et al. Sulfasalazineinduced ferroptosis in breast cancer cells is reduced by the inhibitory effect of estrogen receptor on the transferrin receptor. Oncol Rep. 2019;42:826–38.PubMed Yu H, Yang C, Jian L, et al. Sulfasalazineinduced ferroptosis in breast cancer cells is reduced by the inhibitory effect of estrogen receptor on the transferrin receptor. Oncol Rep. 2019;42:826–38.PubMed
21.
go back to reference Ma S, Henson ES, Chen Y, et al. Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 2016;7:e2307.CrossRef Ma S, Henson ES, Chen Y, et al. Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 2016;7:e2307.CrossRef
22.
go back to reference Hasegawa M, Takahashi H, Rajabi H, et al. Functional interactions of the cystine/glutamate antiporter, CD44v and MUC1-C oncoprotein in triple-negative breast cancer cells. Oncotarget. 2016;7:11756–69.CrossRef Hasegawa M, Takahashi H, Rajabi H, et al. Functional interactions of the cystine/glutamate antiporter, CD44v and MUC1-C oncoprotein in triple-negative breast cancer cells. Oncotarget. 2016;7:11756–69.CrossRef
23.
go back to reference Chen MS, Wang SF, Hsu CY, et al. CHAC1 degradation of glutathione enhances cystine-starvation-induced necroptosis and ferroptosis in human triple negative breast cancer cells via the GCN2-eIF2alpha-ATF4 pathway. Oncotarget. 2017;8:114588–602.CrossRef Chen MS, Wang SF, Hsu CY, et al. CHAC1 degradation of glutathione enhances cystine-starvation-induced necroptosis and ferroptosis in human triple negative breast cancer cells via the GCN2-eIF2alpha-ATF4 pathway. Oncotarget. 2017;8:114588–602.CrossRef
24.
go back to reference Ghantous A, Sinjab A, Herceg Z, et al. Parthenolide: from plant shoots to cancer roots. Drug Discov Today. 2013;18:894–905.CrossRef Ghantous A, Sinjab A, Herceg Z, et al. Parthenolide: from plant shoots to cancer roots. Drug Discov Today. 2013;18:894–905.CrossRef
25.
go back to reference Guzman ML, Rossi RM, Neelakantan S, et al. An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells. Blood. 2007;110:4427–35.CrossRef Guzman ML, Rossi RM, Neelakantan S, et al. An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells. Blood. 2007;110:4427–35.CrossRef
26.
go back to reference Lesiak K, Koprowska K, Zalesna I, et al. Parthenolide, a sesquiterpene lactone from the medical herb feverfew, shows anticancer activity against human melanoma cells in vitro. Melanoma Res. 2010;20:21–34.CrossRef Lesiak K, Koprowska K, Zalesna I, et al. Parthenolide, a sesquiterpene lactone from the medical herb feverfew, shows anticancer activity against human melanoma cells in vitro. Melanoma Res. 2010;20:21–34.CrossRef
27.
go back to reference Nasim S, Crooks PA. Antileukemic activity of aminoparthenolide analogs. Bioorg Med Chem Lett. 2008;18:3870–3.CrossRef Nasim S, Crooks PA. Antileukemic activity of aminoparthenolide analogs. Bioorg Med Chem Lett. 2008;18:3870–3.CrossRef
28.
go back to reference Zhang Q, Lu Y, Ding Y, et al. Guaianolide sesquiterpene lactones, a source to discover agents that selectively inhibit acute myelogenous leukemia stem and progenitor cells. J Med Chem. 2012;55:8757–69.CrossRef Zhang Q, Lu Y, Ding Y, et al. Guaianolide sesquiterpene lactones, a source to discover agents that selectively inhibit acute myelogenous leukemia stem and progenitor cells. J Med Chem. 2012;55:8757–69.CrossRef
29.
go back to reference Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156:317–31.CrossRef Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156:317–31.CrossRef
30.
go back to reference Araujo TG, Vecchi L, Lima P, et al. Parthenolide and its analogues: a new potential strategy for the treatment of triple-negative breast tumors. Curr Med Chem. 2020;27:6628–42.CrossRef Araujo TG, Vecchi L, Lima P, et al. Parthenolide and its analogues: a new potential strategy for the treatment of triple-negative breast tumors. Curr Med Chem. 2020;27:6628–42.CrossRef
31.
go back to reference Ge W, Hao X, Han F, et al. Synthesis and structure-activity relationship studies of parthenolide derivatives as potential anti-triple negative breast cancer agents. Eur J Med Chem. 2019;166:445–69.CrossRef Ge W, Hao X, Han F, et al. Synthesis and structure-activity relationship studies of parthenolide derivatives as potential anti-triple negative breast cancer agents. Eur J Med Chem. 2019;166:445–69.CrossRef
32.
go back to reference Zhao H, Ji B, Chen J, et al. Gpx 4 is involved in the proliferation, migration and apoptosis of glioma cells. Pathol Res Pract. 2017;213:626–33.CrossRef Zhao H, Ji B, Chen J, et al. Gpx 4 is involved in the proliferation, migration and apoptosis of glioma cells. Pathol Res Pract. 2017;213:626–33.CrossRef
33.
go back to reference Imai H, Nakagawa Y. Biological significance of phospholipid hydroperoxide glutathione peroxidase (PHGPx, GPx4) in mammalian cells. Free Radic Biol Med. 2003;34:145–69.CrossRef Imai H, Nakagawa Y. Biological significance of phospholipid hydroperoxide glutathione peroxidase (PHGPx, GPx4) in mammalian cells. Free Radic Biol Med. 2003;34:145–69.CrossRef
34.
go back to reference Sakamoto K, Sogabe S, Kamada Y, et al. Discovery of GPX4 inhibitory peptides from random peptide T7 phage display and subsequent structural analysis. Biochem Biophys Res Commun. 2017;482:195–201.CrossRef Sakamoto K, Sogabe S, Kamada Y, et al. Discovery of GPX4 inhibitory peptides from random peptide T7 phage display and subsequent structural analysis. Biochem Biophys Res Commun. 2017;482:195–201.CrossRef
35.
go back to reference Myung DS, Park YL, Kim N, et al. Expression of early growth response-1 in colorectal cancer and its relation to tumor cell proliferation and apoptosis. Oncol Rep. 2014;31:788–94.CrossRef Myung DS, Park YL, Kim N, et al. Expression of early growth response-1 in colorectal cancer and its relation to tumor cell proliferation and apoptosis. Oncol Rep. 2014;31:788–94.CrossRef
36.
go back to reference Vagia E, Mahalingam D, Cristofanilli M. The landscape of targeted therapies in TNBC. Cancers (Basel). 2020;12:916.CrossRef Vagia E, Mahalingam D, Cristofanilli M. The landscape of targeted therapies in TNBC. Cancers (Basel). 2020;12:916.CrossRef
37.
go back to reference Gaschler MM, Andia AA, Liu H, et al. FINO2 initiates ferroptosis through GPX4 inactivation and iron oxidation. Nat Chem Biol. 2018;14:507–15.CrossRef Gaschler MM, Andia AA, Liu H, et al. FINO2 initiates ferroptosis through GPX4 inactivation and iron oxidation. Nat Chem Biol. 2018;14:507–15.CrossRef
38.
go back to reference Shimada K, Skouta R, Kaplan A, et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. 2016;12:497–503.CrossRef Shimada K, Skouta R, Kaplan A, et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. 2016;12:497–503.CrossRef
Metadata
Title
Identification of a small molecule as inducer of ferroptosis and apoptosis through ubiquitination of GPX4 in triple negative breast cancer cells
Authors
Yahui Ding
Xiaoping Chen
Can Liu
Weizhi Ge
Qin Wang
Xin Hao
Mengmeng Wang
Yue Chen
Quan Zhang
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2021
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-020-01016-8

Other articles of this Issue 1/2021

Journal of Hematology & Oncology 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine