Skip to main content
Log in

Interference with MTHFD2 induces ferroptosis in ovarian cancer cells through ERK signaling to suppress tumor malignant progression

  • Research
  • Published:
Journal of Bioenergetics and Biomembranes Aims and scope Submit manuscript

Abstract

Ovarian cancer (OC) is a deadliest gynecological cancer with the highest mortality rate. Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2), a crucial tumor-promoting factor, is over-expressed in several malignancies including OC. The present study aimed to explore the role and mechanisms of MTHFD2 in OC malignant progression. Thus, cell proliferation, cycling, apoptosis, migration, and invasion were evaluated by CCK-8 assay, EdU assay, flow cytometry, wound healing, transwell assay and western blotting. Additionally, glycolysis was assessed by measuring the level of glucose and lactate production, as well as the expressions of GLUT1, HK2 and PKM2. Then the expression of ferroptosis-related proteins and ERK signaling was detected using western blotting. Ferroptosis was detected through the measurement of iron level, GSH, MDA and ROS activities. The results revealed that MTHFD2 was highly expressed in OC cells. Besides, interference with MTHFD2 induced ferroptosis, promoted ROS accumulation, destroyed mitochondrial function, reduced ATP content and inhibited glycolysis in OC cells. Subsequently, we further found that interference with MTHFD2 affected mitochondrial function and glycolysis in OC cells through ERK signaling. Moreover, interference with MTHFD2 affected ferroptosis to inhibit the malignant progression of OC cells. Collectively, our present study disclosed that interference with MTHFD2 induced ferroptosis in OC to inhibit tumor malignant progression through regulating ERK signaling.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9

Similar content being viewed by others

Data availability

The datasets used and/or analyzed in the current study are available from the corresponding author upon reasonable request.

References

  • Armstrong DK, Bundy B, Wenzel L, Huang HQ, Baergen R, Lele S, Copeland LJ, Walker JL, Burger RA (2006) Gynecologic oncology, intraperitoneal cisplatin and paclitaxel in ovarian cancer. N Engl J Med 354:34–43

    Article  CAS  PubMed  Google Scholar 

  • Basuli D, Tesfay L, Deng Z, Paul B, Yamamoto Y, Ning G, Xian W, McKeon F, Lynch M, Crum CP, Hegde P, Brewer M, Wang X, Miller LD, Dyment N, Torti FM (2017) Torti, Iron addiction: a novel therapeutic target in ovarian cancer. Oncogene 36:4089–4099

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Ben-Sahra I, Hoxhaj G, Ricoult SJH, Asara JM (2016) Manning, mTORC1 induces purine synthesis through control of the mitochondrial tetrahydrofolate cycle. Science 351:728–733

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  • Cookson BT, Brennan MA (2001) Pro-inflammatory programmed cell death. Trends Microbiol 9:113–114

    Article  CAS  PubMed  Google Scholar 

  • Cui X, Su H, Yang J, Wu X, Huo K, Jing X, Zhang S (2022) Up-regulation of MTHFD2 is associated with clinicopathological characteristics and poor survival in ovarian cancer, possibly by regulating MOB1A signaling. J Ovarian Res 15:23

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N, Cuny GD, Mitchison TJ, Moskowitz MA, Yuan J (2005) Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol 1:112–119

    Article  CAS  PubMed  Google Scholar 

  • Deng X, Liu X, Hu B, Liu J, Fu B, Zhang W (2023) Upregulation of MTHFD2 is associated with PD–L1 activation in bladder cancer via the PI3K/AKT pathway. Int J Mol Med 51.

  • Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, Morrison B, Stockwell BR (2012) Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149:1060–1072

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Ganapathy-Kanniappan S, Geschwind JF (2013) Tumor glycolysis as a target for cancer therapy: progress and prospects. Mol Cancer 12:152

    Article  PubMed  PubMed Central  Google Scholar 

  • Hurwitz LM, Pinsky PF, Trabert B (2021) General population screening for ovarian cancer. Lancet 397:2128–2130

    Article  PubMed  Google Scholar 

  • Jin Y, Chen L, Li L, Huang G, Huang H, Tang C (2022) SNAI2 promotes the development of ovarian cancer through regulating ferroptosis. Bioengineered 13:6451–6463

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Khamseekaew J, Kumfu S, Wongjaikam S, Kerdphoo S, Jaiwongkam T, Srichairatanakool S, Fucharoen S, Chattipakorn SC, Chattipakorn N (2017) Effects of iron overload, an iron chelator and a T-Type calcium channel blocker on cardiac mitochondrial biogenesis and mitochondrial dynamics in thalassemic mice. Eur J Pharmacol 799:118–127

    Article  CAS  PubMed  Google Scholar 

  • Koppula P, Zhuang L, Gan B (2021) Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell 12:599–620

    Article  CAS  PubMed  Google Scholar 

  • Koufaris C, Gallage S, Yang T, Lau CH, Valbuena GN, Keun HC (2016) Suppression of MTHFD2 in MCF-7 breast Cancer cells increases glycolysis, dependency on exogenous Glycine, and sensitivity to Folate Depletion. J Proteome Res 15:2618–2625

    Article  CAS  PubMed  Google Scholar 

  • Kumfu S, Chattipakorn S, Fucharoen S, Chattipakorn N (2012) Mitochondrial calcium uniporter blocker prevents cardiac mitochondrial dysfunction induced by iron overload in thalassemic mice. Biometals 25:1167–1175

    Article  CAS  PubMed  Google Scholar 

  • Li Q, Yang F, Shi X, Bian S, Shen F, Wu Y, Zhu C, Fu F, Wang J, Zhou J, Chen Y (2021) MTHFD2 promotes ovarian cancer growth and metastasis via activation of the STAT3 signaling pathway. FEBS Open Bio 11:2845–2857

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Liu Y, Liu X, Wang H, Ding P, Wang C (2022a) Agrimonolide inhibits cancer progression and induces ferroptosis and apoptosis by targeting SCD1 in ovarian cancer cells. Phytomedicine 101:154102

    Article  CAS  PubMed  Google Scholar 

  • Liu Z, Xu Y, Liu X, Wang B (2022b) PCDH7 knockdown potentiates colon cancer cells to chemotherapy via inducing ferroptosis and changes in autophagy through restraining MEK1/2/ERK/c-Fos axis. Biochem Cell Biol 100:445–457

    Article  CAS  PubMed  Google Scholar 

  • Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, Koppula P, Wu S, Zhuang L, Fang B, Poyurovsky MV, Olszewski K, Gan B (2021) DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 593:586–590

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  • Matz M, Coleman MP, Carreira H, Salmeron D, Chirlaque MD, Allemani C, Group CW (2017) Worldwide comparison of ovarian cancer survival: histological group and stage at diagnosis (CONCORD-2). Gynecol Oncol 144:396–404

    Article  PubMed  Google Scholar 

  • Moore K, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, Lisyanskaya A, Floquet A, Leary A, Sonke GS, Gourley C, Banerjee S, Oza A, Gonzalez-Martin A, Aghajanian C, Bradley W, Mathews C, Liu J, Lowe ES, Bloomfield R, DiSilvestro P (2018) Maintenance Olaparib in patients with newly diagnosed Advanced Ovarian Cancer. N Engl J Med 379:2495–2505

    Article  CAS  PubMed  Google Scholar 

  • Morscher RJ, Ducker GS, Li SH, Mayer JA, Gitai Z, Sperl W, Rabinowitz JD (2018) Mitochondrial translation requires folate-dependent tRNA methylation. Nature 554:128–132

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  • Nilsson R, Jain M, Madhusudhan N, Sheppard NG, Strittmatter L, Kampf C, Huang J, Asplund A, Mootha VK (2014) Metabolic enzyme expression highlights a key role for MTHFD2 and the mitochondrial folate pathway in cancer. Nat Commun 5:3128

    Article  ADS  PubMed  Google Scholar 

  • Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, Kristensen G, Carey MS, Beale P, Cervantes A, Kurzeder C, du Bois A, Sehouli J, Kimmig R, Stahle A, Collinson F, Essapen S, Gourley C, Lortholary A, Selle F, Mirza MR, Leminen A, Plante M, Stark D, Qian W, Parmar MK, Oza AM, Investigators I (2011) A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med 365:2484–2496

    Article  CAS  PubMed  Google Scholar 

  • Schweichel JU, Merker HJ (1973) The morphology of various types of cell death in prenatal tissues. Teratology 7:253–266

    Article  CAS  PubMed  Google Scholar 

  • Siegel RL, Miller KD, Fuchs HE, Jemal A (2021) Cancer Stat 2021 CA Cancer J Clin 71:7–33

    Google Scholar 

  • Song Q, Peng S, Che F, Zhu X (2022) Artesunate induces ferroptosis via modulation of p38 and ERK signaling pathway in glioblastoma cells. J Pharmacol Sci 148:300–306

    Article  CAS  PubMed  Google Scholar 

  • Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascon S, Hatzios SK, Kagan VE, Noel K, Jiang X, Linkermann A, Murphy ME, Overholtzer M, Oyagi A, Pagnussat GC, Park J, Ran Q, Rosenfeld CS, Salnikow K, Tang D, Torti FM, Torti SV, Toyokuni S, Woerpel KA, Zhang DD (2017) Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell 171: 273–285

  • Tan Z, Huang H, Sun W, Li Y, Jia Y (2022) Current progress of ferroptosis study in ovarian cancer. Front Mol Biosci 9:966007

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Torre LA, Trabert B, DeSantis CE, Miller KD, Samimi G, Runowicz CD, Gaudet MM, Jemal A, Siegel RL (2018) Ovarian cancer statistics, 2018. CA Cancer J Clin 68:284–296

    Article  PubMed  PubMed Central  Google Scholar 

  • Wan X, Wang C, Huang Z, Zhou D, Xiang S, Qi Q, Chen X, Arbely E, Liu CY, Du P, Yu W (2020) Cisplatin inhibits SIRT3-deacetylation MTHFD2 to disturb cellular redox balance in colorectal cancer cell. Cell Death Dis 11:649

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Wang W, Green M, Choi JE, Gijon M, Kennedy PD, Johnson JK, Liao P, Lang X, Kryczek I, Sell A, Xia H, Zhou J, Li G, Li J, Li W, Wei S, Vatan L, Zhang H, Szeliga W, Gu W, Liu R, Lawrence TS, Lamb C, Tanno Y, Cieslik M, Stone E, Georgiou G, Chan TA, Chinnaiyan A, Zou W (2019) CD8(+) T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 569:270–274

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  • Wu S, Cai W, Shi Z, Ming X, Yang X, Zhou Y, Chen X, Yang M (2022) Knockdown of MTHFD2 inhibits proliferation and migration of nasopharyngeal carcinoma cells through the ERK signaling pathway. Biochem Biophys Res Commun 614:47–55

    Article  CAS  PubMed  Google Scholar 

  • Yue L, Pei Y, Zhong L, Yang H, Wang Y, Zhang W, Chen N, Zhu Q, Gao J, Zhi M, Wen B, Zhang S, Xiang J, Wei Q, Liang H, Cao S, Lou H, Chen Z, Han J (2020) Mthfd2 modulates mitochondrial function and DNA repair to maintain the pluripotency of mouse stem cells. Stem Cell Rep 15:529–545

    Article  CAS  Google Scholar 

  • Zhang C, Liu X, Jin S, Chen Y, Guo R (2022) Ferroptosis in cancer therapy: a novel approach to reversing drug resistance. Mol Cancer 21:47

    Article  PubMed  PubMed Central  Google Scholar 

  • Zhang H, Zhu S, Zhou H, Li R, Xia X, Xiong H (2023) Identification of MTHFD2 as a prognostic biomarker and ferroptosis regulator in triple-negative breast cancer. Front Oncol 13:1098357

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhu J, Xiong Y, Zhang Y, Wen J, Cai N, Cheng K, Liang H, Zhang W (2020) The Molecular Mechanisms of Regulating Oxidative Stress-Induced Ferroptosis and Therapeutic Strategy in Tumors, Oxid Med Cell Longev 2020:8810785

Download references

Acknowledgements

Not applicable.

Funding

Development and Application of Appropriate Medical and Health Technologies in Guangxi (S2022076); The excellent Medical Talents Training Program of The First Affiliated Hospital of Guangxi Medical University (2018-12-9).

Author information

Authors and Affiliations

Authors

Contributions

XM and KL conceived and designed the study, acquired and interpreted the data. XM was a major contributor in writing the manuscript. YS and QL collected and analyzed the data. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Xiaoliang Mo.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mo, X., Liu, Q., Liang, K. et al. Interference with MTHFD2 induces ferroptosis in ovarian cancer cells through ERK signaling to suppress tumor malignant progression. J Bioenerg Biomembr (2024). https://doi.org/10.1007/s10863-024-10014-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s10863-024-10014-1

Keywords

Navigation