Review
Emerging Mechanisms and Disease Relevance of Ferroptosis

https://doi.org/10.1016/j.tcb.2020.02.009Get rights and content

Highlights

  • There are three parallel pathways for defending against cell death by ferroptosis.

  • Iron availability is controlled by cellular iron import, storage, and efflux mechanisms.

  • Several tumor suppressor genes normally prevent tumor development by activating ferroptosis in developing tumor cells.

  • As cancers evolve to a more aggressive state, they become increasingly sensitive to ferroptosis.

  • The immune system uses ferroptosis as a means of eliminating cancer cells.

Cell death is an essential feature of development in multicellular organisms, a critical driver of degenerative diseases, and can be harnessed for treating some cancers. Understanding the mechanisms governing cell death is critical for addressing its role in disease. Similarly, metabolism is essential for normal energy and biomolecule production, and goes awry in many diseases. Metabolism and cell death are tightly linked in the phenomenon of ferroptosis, a form of regulated cell death driven by peroxidation of phospholipids. Glutathione peroxidase 4 (GPX4) uses glutathione to protect cells from ferroptosis by eliminating phospholipid peroxides. Recent data have revealed glutathione/GPX4-independent axes for suppressing ferroptosis, and insight into the regulation of iron and mitochondria in ferroptosis. Ferroptosis has recently been implicated in multiple diseases, and functions as a tumor suppression mechanism. Ferroptosis induction is a promising approach in treating several conditions, including neoplastic diseases. Here, we summarize these recent advances.

Section snippets

Ferroptosis Is a Recently Discovered Form of Cell Death That Is Controlled by Numerous Metabolic Pathways

Cells are the fundamental organizing unit of biological systems. The mechanisms governing the division, growth, proliferation, and death of cells are central to understanding the logic underlying the functioning of life on Earth, the possibility of life elsewhere in the universe, and the processes by which disease develops. Ferroptosis is a recently described form of cell death involving iron-dependent damage to membrane lipids; numerous metabolic pathways involving iron, lipids, and amino

Brief History of Ferroptosis

Cell death is critical for the normal development of multicellular organisms, and is aberrantly activated or suppressed in a large number of diseases [1]. Cell death was historically considered to be unregulated, until the 1950s, when the concept of ‘programmed cell death’ emerged, which led in subsequent decades to the discovery of apoptosis as a form of programmed cell death, along with expanding insight into its mechanisms of operation. For a time, apoptosis was synonymous with programmed

Ferroptosis Is Regulated by Numerous Pathways and Implicated in an Increasing Number of Diseases

Ferroptosis is a form of cell death first reported in 2012 [4], although many of the processes involved in ferroptosis had been observed in isolation decades earlier, but not integrated into a unified process [8]. The mechanisms governing ferroptosis that were elucidated in the first few years after it was discovered centered around cysteine and glutathione metabolism, and the ability of the phospholipid peroxidase GPX4 to prevent the accumulation of peroxidized lipids [9], which built on early

Regulation of Iron in Ferroptosis

Despite incorporating iron in the name, the role of and regulation of iron in ferroptosis only emerged recently. Initially, confusion surrounded as to whether the lipid peroxidation that drives ferroptosis was caused by the labile iron pool reacting with lipid peroxides to propagate these species in membranes, or whether iron-dependent enzymes might solely drive this peroxidation process. Studies from several laboratories revealed that iron-dependent lipoxygenases often initiate ferroptosis by

Lipid Peroxide Regulation

As noted earlier, the glutathione-dependent phospholipid peroxidase GPX4 was the first-discovered central inhibitor of ferroptosis [9]. GPX4 is a selenoprotein, implying that selenium availability impacts the sensitivity to ferroptosis. Indeed, delivery of selenium to cells or animals suppresses ferroptosis, including in a mouse model of intracerebral hemorrhage [26., 27., 28.].

A genetic screen for regulators of ferroptosis sensitivity revealed the surprising finding that the multidrug

Ferroptosis as a Tumor Suppression Mechanism

While illuminating the basic mechanisms by which ferroptosis operates is of value for understanding fundamental cell biology, determining the natural functions of ferroptosis will aid in understanding how and why this form of cell death emerged during evolution. Accumulating evidence indicates that ferroptotic cell death leads to tumor growth suppression. Nevertheless, it remains to be further defined whether ferroptosis acts as a critical barrier to cancer development. Inactivation of the p53

Ferroptosis Modulation as a Therapeutic Avenue

Although a definitive physiological function for ferroptosis has yet to be unambiguously demonstrated, the role of ferroptosis in human diseases has been established. A wealth of studies suggests that pharmacological modulation of this unique cell death modality, by either inhibiting it or stimulating it, may yield significant clinical benefit for certain diseases.

Concluding Remarks and Future Perspectives

The rapid expansion of our understanding of ferroptosis is due to the growing number of laboratories that are exploring the mechanisms and functions of this form of cell death. Given the recent advances, we know that there are at least three major pathways that control sensitivity of cells to ferroptosis: the glutathione–GPX4, NADPH–FSP1–CoQ10, and GCH1–BH4 pathways. Moreover, it appears clear that ferroptosis contributes to both degenerative disease pathology and tumor suppression, and that

Acknowledgments

Research by the authors is supported by the National Institutes of Health (NIH) R01CA204232 (to X.J.), a Geoffrey Beene Cancer Research fund (to X.J.), a Functional Genomic Initiative fund (to X.J), the National Cancer Institute R35CA209896, P01CA087497 (to B.R.S.), and R01CA085533 and RO1CA224272 (to W.G), and the National institute of Neurological Disorders and Stroke R61NS109407 (to B.R.S.).

References (100)

  • J.P. Friedmann Angeli et al.

    Selenium and GPX4, a vital symbiosis

    Free Radic. Biol. Med.

    (2018)
  • E.R. Kastenhuber et al.

    Putting p53 in context

    Cell

    (2017)
  • M. Gao

    Glutaminolysis and transferrin regulate ferroptosis

    Mol. Cell

    (2015)
  • S.J. Wang

    Acetylation is crucial for p53-mediated ferroptosis and tumor suppression

    Cell Rep.

    (2016)
  • T. Ishimoto

    CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc(-) and thereby promotes tumor growth

    Cancer Cell

    (2011)
  • X.X. Liu

    MicroRNA-26b is underexpressed in human breast cancer and induces cell apoptosis by targeting SLC7A11

    FEBS Lett.

    (2011)
  • W. Guo

    Disruption of xCT inhibits cell growth via the ROS/autophagy pathway in hepatocellular carcinoma

    Cancer Lett.

    (2011)
  • I.I.C. Chio et al.

    ROS in cancer: the burning question

    Trends Mol. Med.

    (2017)
  • J.D. Hayes et al.

    NRF2 and KEAP1 mutations: permanent activation of an adaptive response in cancer

    Trends Biochem. Sci.

    (2009)
  • J.P. Kruse et al.

    Modes of p53 regulation

    Cell

    (2009)
  • J.L. Moran

    A mouse mutation in the 12R-lipoxygenase, Alox12b, disrupts formation of the epidermal permeability barrier

    J Invest Dermatol

    (2007)
  • Z. Yu

    Mutations associated with a congenital form of ichthyosis (NCIE) inactivate the epidermal lipoxygenases 12R-LOX and eLOX3

    Biochim. Biophys. Acta

    (2005)
  • P. Krieg

    Aloxe3 knockout mice reveal a function of epidermal lipoxygenase-3 as hepoxilin synthase and its pivotal role in barrier formation

    J Invest Dermatol

    (2013)
  • E.J. Park

    Whole cigarette smoke condensates induce ferroptosis in human bronchial epithelial cells

    Toxicol. Lett.

    (2019)
  • L.J. Yant

    The selenoprotein GPX4 is essential for mouse development and protects from radiation and oxidative damage insults

    Free Radic. Biol. Med.

    (2003)
  • E.A. McCullagh et al.

    Behavioral characterization of system xc- mutant mice

    Behav. Brain Res.

    (2014)
  • H. Sato

    Redox imbalance in cystine/glutamate transporter-deficient mice

    J. Biol. Chem.

    (2005)
  • B.R. Stockwell et al.

    A physiological function for ferroptosis in tumor suppression by the immune system

    Cell Metab.

    (2019)
  • L. Galluzzi

    Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018

    Cell Death Differ.

    (2018)
  • N. Yagoda

    RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels

    Nature

    (2007)
  • S.J. Dixon

    Pharmacological inhibition of cystine–glutamate exchange induces endoplasmic reticulum stress and ferroptosis

    eLife

    (2014)
  • H. Eagle

    Nutrition needs of mammalian cells in tissue culture

    Science

    (1955)
  • H. Eagle

    The specific amino acid requirements of a human carcinoma cell (Stain HeLa) in tissue culture

    J. Exp. Med.

    (1955)
  • W.Z. Potter

    Acetaminophen-induced hepatic necrosis. 3. Cytochrome P-450-mediated covalent binding in vitro

    J. Pharmacol. Exp. Ther.

    (1973)
  • J.R. Mitchell

    Acetaminophen-induced hepatic necrosis. IV. Protective role of glutathione

    J. Pharmacol. Exp. Ther.

    (1973)
  • J.R. Mitchell

    Acetaminophen-induced hepatic necrosis. I. Role of drug metabolism

    J. Pharmacol. Exp. Ther.

    (1973)
  • D.J. Jollow

    Acetaminophen-induced hepatic necrosis. II. Role of covalent binding in vivo

    J. Pharmacol. Exp. Ther.

    (1973)
  • R. Shah

    Resolving the role of lipoxygenases in the initiation and execution of ferroptosis

    ACS Cent. Sci.

    (2018)
  • S.E. Wenzel

    PEBP1 wardens ferroptosis by enabling lipoxygenase generation of lipid death signals

    Cell

    (2017)
  • Y. Zou

    Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis

    Nat. Chem. Biol.

    (2020)
  • W. Hou

    Autophagy promotes ferroptosis by degradation of ferritin

    Autophagy

    (2016)
  • M. Gao

    Ferroptosis is an autophagic cell death process

    Cell Res.

    (2016)
  • C.W. Brown

    Prominin2 drives ferroptosis resistance by stimulating iron export

    Dev. Cell

    (2019)
  • I. Alim

    Selenium drives a transcriptional adaptive program to block ferroptosis and treat stroke

    Cell

    (2019)
  • I. Ingold

    Selenium utilization by GPX4 is required to prevent hydroperoxide-induced ferroptosis

    Cell

    (2018)
  • J.Y. Cao

    A genome-wide haploid genetic screen identifies regulators of glutathione abundance and ferroptosis sensitivity

    Cell Rep.

    (2019)
  • K. Shimada

    Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis

    Nat. Chem. Biol.

    (2016)
  • S. Doll

    FSP1 is a glutathione-independent ferroptosis suppressor

    Nature

    (2019)
  • K. Bersuker

    The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis

    Nature

    (2019)
  • V.A.N. Kraft

    GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling

    ACS Cent. Sci.

    (2020)
  • Cited by (661)

    • Coenzyme Q10 in atherosclerosis

      2024, European Journal of Pharmacology
    • GPX4, ferroptosis, and diseases

      2024, Biomedicine and Pharmacotherapy
    View all citing articles on Scopus
    @

    Twitter: @bstockwell

    View full text