Skip to main content

The Warburg effect modulates DHODH role in ferroptosis: a review

Abstract

Ferroptosis is an iron-dependent regulated cell death that suppresses tumor growth. It is activated by extensive peroxidation of membrane phospholipids caused by oxidative stress. GPX4, an antioxidant enzyme, reduces these peroxidized membrane phospholipids thereby inhibiting ferroptosis. This enzyme has two distinct subcellular localization; the cytosol and mitochondria. Dihydroorotate dehydrogenase (DHODH) complements mitochondrial GPX4 in reducing peroxidized membrane phospholipids. It is the rate-limiting enzyme in de novo pyrimidine nucleotide biosynthesis. Its role in ferroptosis inhibition suggests that DHODH inhibitors could have two complementary mechanisms of action against tumors; inhibiting de novo pyrimidine nucleotide biosynthesis and enhancing ferroptosis. However, the link between mitochondrial function and ferroptosis, and the involvement of DHODH in the ETC suggests that its role in ferroptosis could be modulated by the Warburg effect. Therefore, we reviewed relevant literature to get an insight into the possible effect of this metabolic reprogramming on the role of DHODH in ferroptosis. Furthermore, an emerging link between DHODH and cellular GSH pool has also been highlighted. These insights could contribute to the rational design of ferroptosis-based anticancer drugs.

Video Abstract

Background

Oncogenic mutations significantly contribute to tumorigenesis. An example is the mutation that affects Ras superfamily of small GTPases [1]. It was discovered that two chemical compounds; erastin and Ras-selective lethal compound 3 (RSL3) selectively induce cell death in Ras-mutant cancers. This cell death mechanism lacks the classical molecular and biochemical features of apoptosis. Rather, it was characterized by a high cellular level of oxidative stress markers and also susceptible to inhibition by iron chelation or blocking cellular iron uptake [2]. To capture its iron dependency, this novel cell death mechanism was termed “ferroptosis” [2]. RSL3 inhibits glutathione peroxidase 4 (GPX4), an antioxidant enzyme having two distinct subcellular localization; the cytosol and the mitochondria [3]. This enzyme protects membrane phospholipids from peroxidation [4]. Erastin is an inhibitor of cystine-glutamate antiporter (xCT), a transmembrane protein that imports cystine into the cell [5]. Cystine is then reduced to cysteine, the rate-limiting amino acid in the biosynthesis of reduced glutathione (GSH) [5]. GSH serves as a cofactor for GPX4.

Following its first description, ferroptosis has been implicated in the pathogenesis of cancer, neurological disorders, coronary heart diseases, liver, and kidney diseases [6]. However, most of the studies focused on the involvement of ferroptosis in cancer, and a recent finding is the contribution of dihydroorotate dehydrogenase (DHODH) to ferroptosis inhibition. It was reported that DHODH works along with mitochondrial GPX4 to inhibit ferroptosis [3]. DHODH is the rate-limiting enzyme in de novo pyrimidine nucleotide biosynthesis located in the mitochondria. It links de novo pyrimidine nucleotide biosynthesis to the electron transport chain (ETC) at complex III through Coenzyme Q (ubiquinone) pool [7]. This suggests that in addition to suppressing tumor growth by inhibiting pyrimidine biosynthesis, DHODH inhibitors could have a second mechanism of action that is, enhancement of ferroptosis.

In contrast to normal cells, which rely on mitochondrial oxidative phosphorylation to generate adenosine triphosphate (ATP), most cancer cells instead use aerobic glycolysis also known as the Warburg effect [8]. This metabolic reprogramming could be a biochemical adaptation that supports the biosynthetic requirements of rapidly proliferating cells [9]. Since nucleic acid synthesis is a key biosynthetic requirement of cancer cells, the metabolic reprogramming associated with the Warburg effect could enhance its efficiency. It is important to note that the Warburg effect affects the functioning of mitochondrial ETC which is linked to the de novo pyrimidine nucleotide biosynthesis by DHODH at complex III [7]. Given the role of the ETC in the generation of oxidative stress-inducing reactive oxygen species (ROS) and the involvement of DHODH in ETC function; we reviewed previous studies to understand the possible effect of the Warburg effect on the role of DHODH in ferroptosis.

The electron transport chain and ferroptosis

The ETC is a series of inner mitochondrial membrane protein complexes and biochemical mobile electron carriers which facilitate the movement of electrons extracted from fuel molecules to oxygen to form water [10]. Electron movement along the ETC occurs with a concurrent translocation of protons into the intermembrane space to generate a proton gradient. The proton gradient is dissipated when H+ re-enters the mitochondrial matrix through ATP synthase also known as complex V [11]. The other protein complexes are complex I, complex II, complex III, and complex IV. Electron leakage along the ETC produces O2. This primarily occurs at complex I or complex III [12].

It was reported that O2 produced by complex III of the ETC plays a pivotal role in the induction of ferroptosis by cysteine deprivation [13]. The study revealed that S3QEL, a molecule that suppresses O2 production by complex III and not S1QEL which suppresses O2 generation by complex I could protect mouse hepatoma Hepa 1–6 cells from lipid peroxidation and ferroptosis. In addition, S3QEL was able to inhibit ferroptosis in xCT-knockout mouse-derived embryonic fibroblasts [13]. The xCT knock out (KO) causes cysteine deficiency which inhibits glutathione biosynthesis. This study suggests that the functionality of complex III of the ETC may be required for pharmacological induction of ferroptosis. The ability of complex III-generated O2 to induce lipid peroxidation could be due to its bi-directional release both into the mitochondrial matrix and the intermembrane space (Fig. 1) as opposed to complex I-generated O2 which is only released into the mitochondrial matrix [12].

Fig. 1
figure 1

Contribution of ETC to ferroptosis induction. Electrons shuttled into the ETC come from NADH (represented as NAD-e), FADH2 (represented as FAD-e), and CoQ10H2 (represented as COQ-e). Complex III (CIII)-generated O2 has been shown to play a key role in the induction of ferroptosis activated by cysteine deprivation

It was earlier reported that the biochemical mechanism for the induction of ferroptosis by erastin is mitochondrial membrane potential hyperpolarization [14]. However, this study did not report the dynamics of O2 generation by the ETC under experimental conditions. It is conceivable that the membrane potential hyperpolarization observed during the study was linked to ferroptosis induction due to a positive correlation with O2 generation by the ETC. This view is supported by the failure of complex I and II inhibitors to sustainably suppress ferroptosis during the study, suggesting that provided electrons pass through complex III and IV, ferroptosis could be activated. The generation of O2 by complex III could be a more plausible hypothesis explaining the biochemical mechanism underlying ferroptosis induction by erastin. This is because of the well-established relationship between ferroptosis and oxidative stress.

A third study that reported the biochemical mechanism underlying the induction of ferroptosis by erastin also implicated O2 generation [15]. However, this study concluded that the ETC is not involved in erastin-induced ferroptosis but rather, the pyruvate dehydrogenase complex-catalyzed reaction. This is because treating HT1080 cells with Antimycin A or FCCP was found to have no significant effect on the cell’s sensitivity to erastin as opposed to the genetic inhibition of E1 or E3 subunits of pyruvate dehydrogenase complex. This study used changes in the E50 value of erastin as indicative of ferroptosis sensitivity. A possible drawback to the validity of the conclusion dissociating ETC from erastin-induced ferroptosis is the absence of data showing that the cells actually died as a result of ferroptosis. This data may be important because Antimycin and FCCP can interact with erastin to modify its pharmacological effect. In addition, although the decrease in oxygen consumption rate (OCR) observed as a result of Antimycin treatment is expected to correlate with decreased electron flow through the ETC resulting in decreased O2 production, a previous study has reported an increase in cellular O2 level in response to Antimycin treatment [16]. If this happens to be the case in this study, although a significant difference in OCR was observed between the Antimycin and FCCP treated group, there may be no significant difference in cellular O2 levels. Therefore, the use of a different pharmacological agent like S3QEL or a genetic approach to the inhibition of complex III of the ETC may tell a different story.

DHODH and ferroptosis

DHODH functions along with mitochondrial GPX4 to reduce peroxidized membrane phospholipids [17]. Metabolomics screening following the treatment of cancer cells with GPX4 inhibitors resulted in a significant depletion of N-carbamoyl-L-aspartate and the accumulation of uridine. N-carbamoyl-L-aspartate is a de novo pyrimidine nucleotide biosynthesis intermediate located upstream of DHODH while uridine is an intermediate downstream of DHODH. These changes suggest that GPX4 inhibition enhanced the conversion of N-carbamoyl-L-aspartate to uridine. Given that DHODH is the rate-limiting enzyme of this pathway, it is conceivable that the flow of metabolites can only increase if DHODH activity is upregulated. Supplementing the culture medium with dihydroorotate, a substrate for DHODH, suppressed ferroptosis activated by GPX4 inhibition while supplementing the medium with uridine, the end product of the pathway, enhanced ferroptosis induced by GPX4 inhibition. These observations suggested a complementary role between GPX4 and DHODH in ferroptosis inhibition, a hypothesis that was later confirmed during the study [17]. Further investigations revealed that the synergy is between mitochondrial GPX4 and DHODH. A mechanistic study showed that the lipophilic antioxidant, ubiquinol (CoQ10H2), produced during DHODH-catalyzed conversion of dihydroorotate to orotate is the anti-ferroptosis molecule. This mitochondria-localized ferroptosis defense mechanism has its cytosolic counterpart involving the conversion of ubiquinone (CoQ10) to CoQ10H2 by ferroptosis suppressor protein 1 (FSP-1) [18]. Whether other ubiquinol-generating reactions within the mitochondria inhibit ferroptosis is yet to be investigated. This biochemical mechanism linking DHODH with ferroptosis is represented in Fig. 2A.

Fig. 2
figure 2

A Ferroptosis inhibition by DHODH. DHODH-generated Ubiquinol (COQ-e-) reduces mitochondrial membrane phospholipids hydroperoxides to lipids alcohol, thereby inhibiting the onset of ferroptosis. B Ferroptosis induction by DHODH. DHODH-catalyzed reaction indirectly reduces the cellular level of GSH. This is because the nucleoside triphosphates (NTPs) it produces are required for RNR-catalyzed reaction which uses GSH as a cofactor. This can inhibit GPX4 via GSH depletion

A second link between DHODH and ferroptosis is found within the deoxyribonucleic acid (DNA) biosynthetic pathway. Nucleosides triphosphates (NTPs), which are the precursors for deoxynucleosides triphosphates (dNTPs) used for DNA synthesis, are derived from purines and pyrimidines [19]. Given that during DNA synthesis, purines pair with pyrimidines, insufficient pyrimidines could terminate the base-pairing process. The de novo pyrimidine biosynthetic pathway plays an important role in rapidly proliferating cells. This is due to the need to meet the cell’s high nucleic acid demand. Since DHODH catalyzes the rate-limiting step in de novo pyrimidine biosynthesis, its inhibition could decrease the cellular levels of pyrimidines. Low cellular levels of pyrimidines will slow down ribonucleotide reductase (RNR)-catalyzed reaction which converts NTPs to dNTPs. This assertion is supported by a significant decrease in dNTPs observed following DHODH inhibition [20]. A recent study reported that ferroptosis could be suppressed by inhibiting RNR [21]. This is because RNR uses GSH as a cofactor. When RNR is inhibited, the cellular GSH pool increases. This becomes available to GPX4 to use for reducing peroxidized membrane phospholipids. Therefore, DHODH depletion could enhance the efficiency of GPX4 via inhibiting RNR. However, DHODH depletion may not inhibit ferroptosis in cells with low GPX4 expression via this biochemical mechanism. This biochemical mechanism linking DHODH with ferroptosis is represented in Fig. 2B.

The role of DHODH and complex III in tumor growth

Tumor cells are characterized by rapid growth and division. As a result, most of the biosynthetic precursors are synthesized de novo [22]. To sustain growth in tumors, cell division must progress rapidly. This cannot occur when biosynthetic precursors required for the S phase of the cell cycle are insufficient. The S phase facilitates chromosome doubling that precedes cell division. This phase in tumors will require a large supply of pyrimidines that can only be made available when synthesized de novo. DHODH, the rate-limiting enzyme for de novo pyrimidine nucleotide biosynthesis, links this pathway to complex III of the ETC [23]. The reaction mechanism for DHODH involves the reduction of ubiquinone (CoQ10) to CoQ10H2. CoQ10 is a mobile electron carrier that accepts electrons at complexes I and II of the ETC and transfers them to complex III. However, DHODH-generated CoQ10H2 donates its electrons directly to complex III [23]. Therefore, inhibition of complex III will not only halt electron flow through the ETC but could also inhibit DHODH-catalyzed conversion of dihydroorotate to orotate, thereby shutting down de novo pyrimidine nucleotide biosynthesis. This could result in cell cycle arrest and the inhibition of tumor growth. Inhibition of complex III of mitochondrial ETC by antimycin A was able to inhibit proliferation and promote cellular differentiation in acute myeloid leukemia (AML) cells through a DHODH inhibition-mediated mechanism [24].

Another study reported impaired tumor growth due to a dysfunctional complex III of the ETC. The biochemical mechanism underlying this growth inhibition involves the failure of complex III to re-oxidize CoQ10H2 back to CoQ10 rather than the failure of electrons to flow through the ETC [25]. During the study, ectopic expression of Ciona intestinalis alternative oxidase (AOX) which oxidizes CoQ10H2 to CoQ10 restored the lost tumor growth. In addition, a functional complex III and IV of the ETC alone sufficiently restored tumor growth of respiration-deficient cancer cells through the re-activation of de novo pyrimidine nucleotide biosynthesis [26]. These studies provide mechanistic insight into a previous observation where the acquisition of mitochondrial DNA (mtDNA) via horizontal transfer of whole mitochondria from host cells to mtDNA-deficient cancer cells restored lost tumorigenicity [27, 28]. The transferred host mitochondria could have facilitated the rate-limiting step in de novo pyrimidine nucleotide biosynthesis. Also, these findings are an addition to the accumulating evidence against complete mitochondrial damage as the biochemical basis for aerobic glycolysis in tumors. During aerobic glycolysis also known as the Warburg effect, some aspects of mitochondrial function could be inefficient; however, the ETC-linked DHODH-catalyzed rate-limiting step for de novo pyrimidine nucleotide biosynthesis should not be affected. This conclusion is further supported by the need for a functional complex III for hepatitis E virus replication [29]. In fact, disruption in certain aspects of mitochondrial function during aerobic glycolysis, in addition to conferring other tumorigenic advantages, appears to be aimed at optimizing de novo pyrimidine nucleotide biosynthesis. This optimization could be due to the high nucleic acid demand during the S-phase of the cell cycle [30]. For example, hypoxia can slow down the movement of electrons through the ETC due to insufficient oxygen molecules that can serve as terminal electron acceptors. A defect in complex I and II of the ETC associated with the Warburg effect [31, 32] could enable more efficient coupling of mitochondrial oxygen consumption to the conversion of dihydroorotate to orotate in an oxygen-deficient intracellular environment predominant in solid tumors.

The importance of de novo pyrimidine nucleotide biosynthesis for the proliferation of human Jurkat leukemic T cells is indirectly substantiated by a metabolic reprogramming activated by complex I inhibition. When the ETC is fully functional, glutamate–oxaloacetate transaminase 1 (GOT1) converts aspartate to oxaloacetate and α-ketoglutarate to glutamate concurrently. This reaction is part of the cytosolic portion of the malate-aspartate shuttle that transfers NADH produced during glycolysis from the cytosol into the mitochondria. The mitochondrial matrix portion of this shuttle comprises malate dehydrogenase 2 (MDH2) and glutamate–oxaloacetate transaminase 2 (GOT2). These enzymes synthesize aspartate which is transported to the cytosol for use by GOT1 and other enzymes. However, complex I inhibition inhibits the regeneration of NAD+ by the ETC. This leads to the inhibition of the mitochondrial matrix portion of the shuttle. Under this condition, GOT1-catalyzed reaction reverses, thereby producing aspartate from the oxaloacetate produced by ATP citrate lyase-catalyzed reaction in the cytosol. This citrate comes from the reductive decarboxylation of glutamine in the mitochondrial matrix [33]. Following complex I inhibition, these cells were found to rely on GOT1-dependent aspartate biosynthesis for proliferation [33].

A defect in complex I of the ETC rewires the malate-aspartate shuttle, shutting down GOT2-dependent aspartate biosynthesis while activating GOT1-dependent aspartate biosynthesis which is linked to the reductive decarboxylation of glutamine. This could be the reason why tumors which usually have a defective ETC due to the Warburg effect are heavily reliant on glutamine metabolism for proliferation [34, 35]. It is important to note that during the study, experimental data suggested that a significant fraction of the aspartate synthesized by GOT1 goes to purine and pyrimidine nucleotide biosynthesis [22]. Given that, GOT1-dependent aspartate biosynthesis could only support cell proliferation if DHODH and complex III are functional. The coupling of DHODH-catalyzed reaction to complex III of the ETC [36] suggests that reductive decarboxylation of glutamine which supplies the oxaloacetate used by GOT1 could result in the generation of ATP in the ETC through DHODH-mediated shuttling of CoQ10H2 to complex III [37].

High extracellular lactate characteristic of the Warburg effect induces reductive decarboxylation of glutamine in breast cancer cells [38] and tumor hypoxia which can also induce the Warburg effect making aspartate become a limiting metabolite for cell proliferation [39]. Aspartate was found to be limiting for cell proliferation because of its role in de novo purine and pyrimidine nucleotide biosynthesis. In pyrimidine biosynthesis, aspartate contributes three carbon atoms during the production of uridine monophosphate (UMP) and thymidine monophosphate (TMP) [39]. Under hypoxia and ETC dysfunction, 50 to 80% of orotate, dTMP, and UMP in solute carrier family 1 member 3 (SLC1A3)-expressing cells were derived from extracellularly imported aspartate. Thus suggests that hypoxia could reduce the intracellular concentration of pyrimidines synthesized de novo by more than 50% unless an adaptive metabolic rewiring occurs to supply aspartate. This study suggests a relationship between defective complex I, GOT1-facilitated aspartate biosynthesis, and nucleotide biosynthesis.

Coupling of de novo pyrimidine nucleotide biosynthesis to the ETC could be a strategy for meeting the high energy demand for pyrimidine biosynthesis when metabolic rewiring occurs that shuts down part of the energy-generating mitochondrial catabolism in favor of energy-demanding nucleotide biosynthesis. This is because DHODH-facilitated electron transfer to complex III and IV could result in pumping protons into the intermembrane space thereby generating some transmembrane electrochemical potential that could be used for ATP synthesis by complex V [40]. This notion is supported by the efficient induction of mitochondrial membrane potential depolarization by DHODH inhibitors from two distinct chemical series [41]. Findings from another study suggest that preceding UVB-Induced primary skin tumor formation, a metabolic rewiring occurs that shuts down glycolysis and the TCA cycle (thereby shutting down complex I and II) while upregulating the distal part of the ETC which includes a heightened DHODH activity [42]. In this study, DHODH was found to sustain ATP generation by the ETC.

Other defects in mitochondrial function that could selectively shut down complex I and II while preserving DHODH-catalyzed reaction and complex III include a tumor-associated defect in three enzymes of the TCA cycle [43]. These enzymes are succinate dehydrogenase (SDH), fumarate hydratase (FH), and isocitrate dehydrogenase (IDH). Defects in these enzymes affect cellular ROS status in a way that induces oxidative stress and stabilizes HIF1-α. While a deficiency in SDH increases ROS levels that result in HIF-1α stabilization, IDH1 and IDH2 mutation lowers the level of GSH and NADPH thereby reducing the efficiency of GSH and NADPH-dependent antioxidant enzymes. However, to manifest the metabolic phenotype of SDH mutant tumors, additional inhibition of complex I is required [44]. The absence of complex I inhibition has been postulated to be the differentiating factor between neurodegeneration-linked SDH deficiency and tumor-linked SDH deficiency [44]. A functional complex I could support glutaminolysis and the malate-aspartate shuttle, two pathways that could transfer electrons to complex I thereby reducing the efficient coupling of DHODH-catalyzed reaction to the utilization of the limited oxygen available in a tumor hypoxic environment. The mutation in FH inhibits complex I through the succination of its Fe-S proteins and complex II through a feedback inhibition activated by fumarate [45]. Role of DHODH and complex III in tumor growth is schematically represented in Fig. 3.

Fig. 3
figure 3

Role of DHODH and complex III in tumor growth. DHODH-catalyzed reaction is the rate-limiting step in de novo pyrimidine nucleotide biosynthesis. It catalyzes the conversion of dihydroorotate to orotate used for nucleic acid biosynthesis. This reaction needs the activity of complex III of the ETC and it involves the conversion of ubiquinol (COQ-e-) to ubiquinone (COQ)

Warburg effect and its modulation of the role of DHODH in ferroptosis

Although DHODH complements mitochondrial GPX4 to inhibit ferroptosis, the prevalence of Warburg effect in tumors, indispensability of DHODH-catalyzed reaction for tumor growth and the role of ETC in O2 generation and ferroptosis induction suggest that inhibiting DHODH in some tumors could result in ferroptosis inhibition. This could result in drug resistance. Solid tumors are characterized by oxidative stress [46]. As ROS inhibit complex I and II of the ETC and the TCA cycle, it stabilizes HIF1-α which upregulates glycolysis as an adaptive response for ATP generation [47]. HIF1-α is an important component of HIF-1, a transcription factor that targets key glycolytic enzymes [47]. Active HIF-1 complex is a heterodimer made up of an oxygen-sensitive HIF-1α subunit and a constitutively expressed HIF-1β subunit. The HIF-1α subunit has an oxygen-dependent degradation domain (ODD) which contains two regulatory prolyl residues; P402 and P564. Under normal cellular oxygen concentration, these amino acid residues become hydroxylated by HIF prolyl-hydroxylases (PHDs). HIF-1 prolyl-hydroxylation facilitates the proteasomal degradation of this transcription factor [48]. However, even in the presence of oxygen, studies have shown that HIF-1α can be stabilized by ROS produced by the ETC.

In a study to establish the molecular mechanism underlying the activation of HIF-1 by angiotensin-II in vascular smooth muscle cells, it was discovered that ROS produced by the ETC play a key role [49]. The use of mitochondrial ETC complex III inhibitors, stigmatellin, and myxothiazol were found to inhibit the accumulation of HIF-1 induced by angiotensin-II. A second approach used in the study involved suppressing ROS production through siRNA-mediated inhibition of Rieske Fe-S protein, an essential component of complex III. This equally resulted in the inhibition of HIF-1α accumulation. The use of SkQ1, an antioxidant targeting mitochondrial ETC also produced the same result. This suggests that O2 generated by the ETC plays an important role in the stabilization of HIF-1α. Genetic and pharmacological inhibition of NADPH oxidase (NOX), a mitochondrial enzyme that produces O2 failed to sustain HIF-1α accumulation under non-hypoxic conditions [49]. This study concluded that ROS generated by the mitochondria is responsible for blocking HIF-1α hydroxylation and its subsequent proteasomal degradation during Angiotensin II treatment in vascular smooth muscle cells. Stabilization of HIF-1 by oxidative stress induces the Warburg effect.

Another study using hepatoma cells also supports the involvement of oxidative stress in inducing the Warburg effect even in the presence of oxygen [50]. During this study, it was discovered that the ability of hypoxia to upregulate glycolysis is dependent on ROS. This is because, in the presence of ROS scavengers, hypoxia failed to upregulate glycolysis. Hypoxia could increase both ROS level and HIF1-α activity. However, when ROS level was reduced using the antioxidant α-Lipoic Acid, the expression of HIF1-α was downregulated. The study further revealed that increasing endogenous ROS generation by xanthine oxidase gene transfection or decreasing it using antioxidants or MnSOD transfection could modulate cellular glycolytic activity independent of hypoxia status. Another group used mitoubiquinone, a mitochondria-targeted antioxidant to confirm the involvement of mitochondrial ROS in the stabilization of HIF-1α [51]. Furthermore, data from a study designed to assess the limiting metabolites required for cell proliferation under hypoxia suggests that tumor hypoxia is sufficient to inhibit the ETC [39]. It should be noted that if this inhibition affects complex III, tumor growth could be suppressed.

PTEN-induced kinase 1 (PINK1) inhibits the Warburg effect by reducing mitochondrial ROS generation. It also suppresses the growth of glioblastoma [52] suggesting that the Warburg effect, which is characterized by defective ETC, supports tumor growth. PINK1 suppresses ROS and tumor growth through FOXO3a, a transcription factor that regulates SOD2. ROS-mediated stabilization of HIF1-α was observed in PINK1 knockout (KO) cells. This resulted in the upregulation of genes that shifted glucose metabolism to aerobic glycolysis [52]. However, although the M2 isoform of pyruvate kinase (M2-PK) required for glycolysis was also upregulated, it remained in the inactive dimeric form due to ROS-mediated oxidation of its cysteine residues. The inactive dimeric form of M2-PK caused the accumulation of phospho-metabolites above phosphoenolpyruvate (PEP) in the glycolytic pathway. These metabolites were channeled to biosynthetic reactions including nucleic acid biosynthesis [53].

NADH:ubiquinone oxidoreductase subunit A10 (NDUFA10) is a key component of complex I of the ETC. PINK1 loss of function mutation affects complex 1 activity through the failure of serine 250 of NDUFA10 to become phosphorylated [54]. This is because a phosphorylated NDUFA10 is required for the reduction of ubiquinone by complex I [54]. This suggests that in addition to shifting glucose metabolism towards aerobic glycolysis through an oxidative stress-mediated stabilization of HIF-1, loss of PINK1 activity could affect the ETC by direct inhibition of complex I. The question now arises: between oxidative stress and the inhibition of complex I activity, which of the two plays the most important role in the induction of aerobic glycolysis? A study that assessed the effect of complex I deficiency and a defect in complex I assembly on the Warburg effect suggest that in the absence of ROS, a deficiency in complex I activity may not be sufficient to induce aerobic glycolysis [31]. Complex I is made up of the matrix arm and the membrane arm. The matrix arm is composed of two modules, the N (NADH binding) and Q (Quinone binding) module while the membrane arm comprises the P (Proton pumping) module. While the P module is encoded by mitochondrial DNA, the N and Q modules are encoded by nuclear DNA. These modules are built independently and later assembled together [55]. This study revealed that a defect in complex I assembly results in excessive ROS generation which resulted in the inhibition of pyruvate dehydrogenase in an AMPK-dependent manner.

Another mechanism through which mitochondrial ROS activates the Warburg effect involves the upregulation of mitochondrial uncoupling proteins (UCPs). Induction of oxidative stress in mouse skin tissues and embryonic fibroblast primary cell culture through SOD2 KO induced the Warburg effect. It was discovered that the oxidative stress-induced the uncoupling of the ETC to ATP synthesis which upregulated glycolysis to compensate for the reduction in ATP generated by the mitochondria [56]. A mechanistic study revealed that SOD2 deficiency caused the nuclear receptor; peroxisome proliferators activated receptors α (PPARα) to upregulate the expression of UCPs. This study also reported an increase in HIF-1α in response to oxidative stress and it was concluded that antioxidant-deficient mitochondria may serve as early cellular signaling responsible for the activation of the Warburg Effect in cancers.

An observation that contributed to the biochemical description of ferroptosis involves the ability of erastin and RSL3 to selectively inhibit the growth of Ras-mutant cancers by inducing a non-apoptotic iron-dependent regulated cell death. It is therefore conceivable that this mutation drives a metabolic reprogramming that makes tumors selectively susceptible to ferroptosis. RSL3 inhibits GPX4, the key antioxidant enzyme that prevents the onset of ferroptosis while erastin is a voltage-dependent anion channels 2 and 3 (VDAC2/3) and cystine-glutamate antiporter (xCT) inhibitor [2]. Mutation in the RAS family of small GTPases (HRAS, NRAS, and KRAS) affects nearly 30% of tumors [1]. Among these, KRAS-driven cancers are characterized by upregulated glycolysis, glutaminolysis, and nucleotide biosynthesis [57]. Upregulation of nucleotide biosynthesis and glutaminolysis suggests the Warburg effect and an active de novo pyrimidine nucleotide biosynthesis which requires glutamine metabolism to provide aspartate [34, 35]. This assertion is supported by the dependence of mutant K-RAS-driven cancers on DHODH [41]. The finding that xCT inhibitors only induce ferroptosis in a fraction of tumor cells sufficiently expressing solute carrier family 7 member 11 (SLC7A11) suggests that in addition to their effect on the functionality of the antioxidant enzyme system, there could be other biochemical mechanisms for ferroptosis induction [58]. It could also imply that to successfully induce ferroptosis, the metabolic phenotype of the tumor must closely resemble that of RAS-mutant cancers where ferroptosis was first induced through SLC7A11 inhibition. Accumulating evidence suggests that one of the metabolic pathways significantly contributing to the sensitivity of RAS-mutant cancers to ferroptosis inducers is de novo pyrimidine nucleotide biosynthesis which under Warburg effect condition, facilitates the shuttling of electrons from complex III to IV.

Oxidation of CoQ10H2 generated by DHODH to CoQ10 by the ETC could result in complex III-mediated generation of O2 even when the functionality of complex I and II are defective due to the Warburg effect. Generation of O2 by complex III could be required for erastin-induced ferroptosis [13] and possibly, ferroptosis induction in general. Therefore under this condition, if DHODH is inhibited, ferroptosis could equally be inhibited (Fig. 4A and B). The indispensability of a functional DHODH and complex III for tumor growth and the inhibition of complex I and II of the ETC in most tumors by the Warburg effect suggests that under the Warburg effect condition, DHODH-catalyzed reaction could play a significant role in ETC function, O2 generation, and ferroptosis induction. It is therefore conceivable that in addition to DNA synthesis which consumes cellular GSH, the Warburg effect could affect the anti-ferroptosis role of DHODH.

Fig. 4
figure 4

Warburg effect and its modulation of the anti-ferroptosis function of DHODH. A In the absence of functional complexes I and II, DHODH sustains the ETC and O2 generation by shuttling electrons to complex III. Complex III-generated O2 induces oxidative stress that results in ferroptosis. B When complexes I and II of the ETC are inhibited by the Warburg effect, DHODH inhibition could cause a complete shutdown of the ETC resulting in the failure of complex III to produce O2. Differences between A and B has been highlighted in yellow

Conclusion and future perspective

DHODH is a versatile drug target involved in the clinical management of many diseases. It is also an attractive drug target in cancer management. As a result of the importance of de novo pyrimidine nucleotide biosynthesis for tumor growth, an attempt has been made to suppress tumor progression using DHODH inhibitors. One such drug, however, showed unsatisfactory efficacy in phase I and II clinical trials [59, 60]. The role of DHODH in ferroptosis inhibition suggests that its inhibitors could have two complementary mechanisms of action for the suppression of tumor growth that is; inhibition of nucleotide biosynthesis and induction of ferroptosis. However, reports that revealed the indispensability of DHODH-catalyzed reaction for tumor growth, the prevalence of the Warburg effect in solid tumors, and the importance of complex III-generated O2 for ferroptosis induction suggests that under Warburg effect condition, DHODH could be responsible for the generation of O2 by complex III hence making its inhibition to result in suppressing O2 generation and ferroptosis. Inhibiting DHODH could also increase cellular GSH levels thereby making GPX4 to be more efficient in inhibiting ferroptosis. There is therefore a need to investigate the relationship between DHODH inhibitors and how they modulate ferroptosis in solid tumors and how that affects their efficacy. The conclusion and future perspective is schematically represented by Fig. 5.

Fig. 5
figure 5

Schematic representation of conclusion and future perspective. As a result of their potential effect on RNR-catalyzed reaction, a reaction that uses GSH to convert NTPs to dNTPs for DNA synthesis, DHODH inhibitors could increase the intracellular level of GSH. Furthermore, under Warburg effect condition, they could inhibit the ETC and O2 generation. How this affect the onset of ferroptosis and drug efficacy needs to be investigated

Availability of data and materials

Not applicable.

Abbreviations

GPX4:

Glutathione peroxidase 4

DHODH:

Dihydroorotate dehydrogenase

ETC:

Electron transport chain

GSH:

Reduced glutathione

RSL3:

Ras-selective lethal compound 3

xCT:

Cystine-glutamate antiporter

ATP:

Adenosine triphosphate

ROS:

Reactive oxygen species

S3QEL:

Selective inhibitor of superoxide production from the outer Q-binding site of mitochondrial respiratory complex site III

S1QEL:

Selective inhibitor of superoxide production from the outer Q-binding site of mitochondrial respiratory complex site I

KO:

Knock out

FCCP:

Carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone

E1:

Alpha 1

E2:

Alpha 2

OCR:

Oxygen consumption rate

NADH:

Reduced nicotinamide adenine dinucleotide

NAD+ :

Oxidized nicotinamide adenine dinucleotide

FADH2 :

Reduced flavin adenine dinucleotide

CoQ10:

Ubiquinone

CoQ10H2 :

Ubiquinol

FSP1:

Ferroptosis suppressor protein 1

DNA:

Deoxyribonucleic acid

NTPs:

Nucleosides triphosphates

dNTPs:

Deoxynucleosides triphosphates

RNR:

Ribonucleotide reductase

GSSG:

Oxidized glutathione

AML:

Acute myeloid leukemia

AOX:

Alternative oxidase

mtDNA:

Mitochondrial DNA

GOT1:

Glutamate–oxaloacetate transaminase 1

MDH2:

Malate dehydrogenase 2

GOT2:

Glutamate–oxaloacetate transaminase 2

UMP:

Uridine monophosphate

TMP:

Thymidine monophosphate

dTMP:

Deoxythymidine monophosphate

SLC1A3:

Solute carrier family 1 member 3

UVB:

Ultraviolet B

SDH:

Succinate dehydrogenase

FH:

Fumarate hydratase

IDH:

Isocitrate dehydrogenase

HIF1-α:

Hypoxia inducible factor 1 alpha

ODD:

Oxygen-dependent degradation domain

PHDs:

Prolyl-hydroxylases

NOX:

NADPH oxidase

MnSOD:

Manganese superoxide dismutase

PINK1:

PTEN-induced kinase 1

FOXO3a:

Forkhead box O3a

SOD2:

Superoxide dismutase 2

M2-PK:

M2 isoform of pyruvate kinase

PEP:

Phosphoenolpyruvate

NDUFA10:

NADH:ubiquinone oxidoreductase subunit A10

AMPK:

Adenosine monophosphate kinase

UCPs:

Uncoupling proteins

PPAR-α:

Peroxisome proliferators activated receptors α

VDAC2/3:

Voltage-dependent anion channels 2 and 3

SLC7A11:

Solute carrier family 7 member 11

References

  1. Vigil D, Cherfils J, Rossman KL, Der CJ. Ras superfamily GEFs and GAPs: validated and tractable targets for cancer therapy? Nat Rev Cancer. 2010;10(12):842–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. 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(5):1060–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 2021;593(7860):586–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Yang Wan S, SriRamaratnam R, Welsch Matthew E, Shimada K, Skouta R, Viswanathan Vasanthi S, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156(1):317–31.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Shibata Y, Yasui H, Higashikawa K, Miyamoto N, Kuge Y. Erastin, a ferroptosis-inducing agent, sensitized cancer cells to X-ray irradiation via glutathione starvation in vitro and in vivo. PloS one. 2019;14(12):e0225931.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Han C, Liu Y, Dai R, Ismail N, Su W, Li B. Ferroptosis and its potential role in human diseases. Front Pharmacol. 2020;11:239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Boukalova S, Hubackova S, Milosevic M, Ezrova Z, Neuzil J, Rohlena J. Dihydroorotate dehydrogenase in oxidative phosphorylation and cancer. Biochim Biophys Acta Mol Basis Dis. 2020;1866(6): 165759.

    Article  CAS  PubMed  Google Scholar 

  8. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324(5930):1029–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Liberti MV, Locasale JW. The Warburg effect: how does it benefit cancer cells? Trends Biochem Sci. 2016;41(3):211–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Nolfi-Donegan D, Braganza A, Shiva S. Mitochondrial electron transport chain: oxidative phosphorylation, oxidant production, and methods of measurement. Redox Biol. 2020;37:101674.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Greene J, Segaran A, Lord S. Targeting OXPHOS and the electronic transport chain in cancer; molecular and therapeutic implications. Semin Cancer Biol. 2022. https://doi.org/10.1016/j.semcancer.2022.02.002.

    Article  PubMed  Google Scholar 

  12. Muller FL, Liu Y, Van Remmen H. Complex III releases superoxide to both sides of the inner mitochondrial membrane. J Biol Chem. 2004;279(47):49064–73.

    Article  CAS  PubMed  Google Scholar 

  13. Homma T, Kobayashi S, Sato H, Fujii J. Superoxide produced by mitochondrial complex III plays a pivotal role in the execution of ferroptosis induced by cysteine starvation. Arch Biochem Biophys. 2021;700: 108775.

    Article  CAS  PubMed  Google Scholar 

  14. Gao MH, Yi JM, Zhu JJ, Minikes AM, Monian P, Thompson CB, et al. Role of mitochondria in ferroptosis. Molecular Cell. 2019;73(2):354.

    Article  CAS  PubMed  Google Scholar 

  15. Vučković A-M, Venerando R, Tibaldi E, Bosello Travain V, Roveri A, Bordin L, et al. Aerobic pyruvate metabolism sensitizes cells to ferroptosis primed by GSH depletion. Free Radical Biol Med. 2021;167:45–53.

    Article  Google Scholar 

  16. Wang L, Duan Q, Wang T, Ahmed M, Zhang N, Li Y, et al. Mitochondrial respiratory chain inhibitors involved in ROS production induced by acute high concentrations of iodide and the effects of SOD as a protective factor. Oxid Med Cell Longev. 2015;2015: 217670.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Mao C, Liu XG, Zhang YL, Lei G, Yan YL, Lee H, et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 2021;593(7860):586.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575(7784):693.

    Article  CAS  PubMed  Google Scholar 

  19. Hollenstein M, Smith CC, Räz M. Nucleoside triphosphates–from synthesis to biochemical characterization. J Vis Exp. 2014;86:51385.

    Google Scholar 

  20. Desler C, Durhuus JA, Hansen TL-L, Anugula S, Zelander NT, Bøggild S, et al. Partial inhibition of mitochondrial-linked pyrimidine synthesis increases tumorigenic potential and lysosome accumulation. Mitochondrion. 2022;64:73–81.

    Article  CAS  PubMed  Google Scholar 

  21. Tarangelo A, Rodencal J, Kim JT, Magtanong L, Long JZ, Dixon SJ. Nucleotide biosynthesis links glutathione metabolism to ferroptosis sensitivity. Life Sci Alliance. 2022. https://doi.org/10.26508/lsa.202101157.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Robinson AD, Eich ML, Varambally S. Dysregulation of de novo nucleotide biosynthetic pathway enzymes in cancer and targeting opportunities. Cancer Lett. 2020;470:134–40.

    Article  CAS  PubMed  Google Scholar 

  23. Khutornenko AA, Roudko VV, Chernyak BV, Vartapetian AB, Chumakov PM, Evstafieva AG. Pyrimidine biosynthesis links mitochondrial respiration to the p53 pathway. Proc Natl Acad Sci USA. 2010;107(29):12828–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhang YP, Luo T, Ding XY, Chang YT, Liu CX, Zhang YQ, et al. Inhibition of mitochondrial complex III induces differentiation in acute myeloid leukemia. Biochem Biophys Res Commun. 2021;547:162–8.

    Article  CAS  PubMed  Google Scholar 

  25. Martinez-Reyes I, Cardona LR, Kong H, Vasan K, McElroy GS, Werner M, et al. Mitochondrial ubiquinol oxidation is necessary for tumour growth. Nature. 2020;585(7824):288.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bajzikova M, Kovarova J, Coelho AR, Boukalova S, Oh S, Rohlenova K, et al. Reactivation of dihydroorotate dehydrogenase-driven pyrimidine biosynthesis restores tumor growth of respiration-deficient cancer cells. Cell Metab. 2019;29(2):399-416.e10.

    Article  CAS  PubMed  Google Scholar 

  27. Dong LF, Kovarova J, Bajzikova M, Bezawork-Geleta A, Svec D, Endaya B, et al. Horizontal transfer of whole mitochondria restores tumorigenic potential in mitochondria! DNA-deficient cancer cells. Elife. 2017;6:e22187.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Tan AS, Baty JW, Dong LF, Bezawork-Geleta A, Endaya B, Goodwin J, et al. Mitochondrial genome acquisition restores respiratory function and tumorigenic potential of cancer cells without mitochondrial DNA. Cell Metab. 2015;21(1):81–94.

    Article  CAS  PubMed  Google Scholar 

  29. Qu CB, Zhang SS, Wang WS, Li M, Wang YJ, van der Heijde-Mulder M, et al. Mitochondrial electron transport chain complex III sustains hepatitis E virus replication and represents an antiviral target. FASEB J. 2019;33(1):1008–19.

    Article  CAS  PubMed  Google Scholar 

  30. Laskey RA, Fairman MP, Blow JJ. S phase of the cell cycle. Science. 1989;246(4930):609–14.

    Article  CAS  PubMed  Google Scholar 

  31. Desquiret-Dumas V, Leman G, Wetterwald C, Chupin S, Lebert A, Khiati S, et al. Warburg-like effect is a hallmark of complex I assembly defects. BBA-Mol Basis Dis. 2019;1865(9):2475–89.

    Article  CAS  Google Scholar 

  32. Holley AK, Dhar SK, St Clair DK. Curbing cancer’s sweet tooth: is there a role for MnSOD in regulation of the Warburg effect? Mitochondrion. 2013;13(3):170–88.

    Article  CAS  PubMed  Google Scholar 

  33. Birsoy K, Wang T, Chen WW, Freinkman E, Abu-Remaileh M, Sabatini DM. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell. 2015;162(3):540–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yoo HC, Yu YC, Sung Y, Han JM. Glutamine reliance in cell metabolism. Exp Mol Med. 2020;52(9):1496–516.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Yang LF, Venneti S, Nagrath D. Glutaminolysis: a Hallmark of Cancer Metabolism. In: Yarmush ML, editor. Annual review of biomedical engineering, vol 19, 192017: 163–194.

  36. Khutornenko AA, Dalina AA, Chernyak BV, Chumakov PM, Evstafieva AG. The role of dihydroorotate dehydrogenase in apoptosis induction in response to inhibition of the mitochondrial respiratory chain complex III. Acta Naturae. 2014;6(1):69–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Desbats MA, Lunardi G, Doimo M, Trevisson E, Salviati L. Genetic bases and clinical manifestations of coenzyme Q10 (CoQ10) deficiency. J Inherit Metab Dis. 2015;38(1):145–56.

    Article  CAS  PubMed  Google Scholar 

  38. Brodsky AN, Odenwelder DC, Harcum SW. High extracellular lactate causes reductive carboxylation in breast tissue cell lines grown under normoxic conditions. Plos One. 2019;14(6):e0213419.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Garcia-Bermudez J, Baudrier L, La K, Zhu XG, Fidelin J, Sviderskiy VO, et al. Aspartate is a limiting metabolite for cancer cell proliferation under hypoxia and in tumours. Nat Cell Biol. 2018;20(10):1228.

    Article  CAS  PubMed  Google Scholar 

  40. Stuchebrukhov AA. Redox-driven proton pumps of the respiratory chain. Biophys J. 2018;115(5):830–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Koundinya M, Sudhalter J, Courjaud A, Lionne B, Touyer G, Bonnet L, et al. Dependence on the pyrimidine biosynthetic enzyme DHODH is a synthetic lethal vulnerability in mutant KRAS-driven cancers. Cell Chem Biol. 2018;25(6):705.

    Article  CAS  PubMed  Google Scholar 

  42. Hosseini M, Dousset L, Mahfouf W, Serrano-Sanchez M, Redonnet-Vernhet I, Mesli S, et al. Energy metabolism rewiring precedes UVB-induced primary skin tumor formation. Cell Rep. 2018;23(12):3621–34.

    Article  CAS  PubMed  Google Scholar 

  43. Cardaci S, Ciriolo MR. TCA cycle defects and cancer: when metabolism tunes redox state. Int J Cell Biol. 2012;2012: 161837.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Lorendeau D, Rinaldi G, Boon R, Spincemaille P, Metzger K, Jager C, et al. Dual loss of succinate dehydrogenase (SDH) and complex I activity is necessary to recapitulate the metabolic phenotype of SDH mutant tumors. Metab Eng. 2017;43:187–97.

    Article  CAS  PubMed  Google Scholar 

  45. Tyrakis PA, Yurkovich ME, Sciacovelli M, Papachristou EK, Bridges HR, Gaude E, et al. Fumarate hydratase loss causes combined respiratory chain defects. Cell Rep. 2017;21(4):1036–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. McCarty MF, Barroso-Aranda J, Contreras F. Oxidative stress therapy for solid tumors–a proposal. Med Hypotheses. 2010;74(6):1052–4.

    Article  CAS  PubMed  Google Scholar 

  47. Jiang Y, Wu GH, He GD, Zhuang QL, Xi QL, Zhang B, et al. The effect of silencing HIF-1α gene in BxPC-3 cell line on glycolysis-related gene expression, cell growth, invasion, and apoptosis. Nutr Cancer. 2015;67(8):1314–23.

    Article  PubMed  Google Scholar 

  48. He WG, Batty-Stuart S, Lee JE, Ohh M. HIF-1 alpha hydroxyprolines modulate oxygen-dependent protein stability via single VHL interface with comparable effect on ubiquitination rate. J Mol Biol. 2021;433(22):17.

    Article  Google Scholar 

  49. Patten DA, Lafleur VN, Robitaille GA, Chan DA, Giaccia AJ, Richard DE. Hypoxia-inducible factor-1 activation in nonhypoxic conditions: the essential role of mitochondrial-derived reactive oxygen species. Mol Biol Cell. 2010;21(18):3247–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Shi DY, Xie FZ, Zhai C, Stern JS, Liu Y, Liu SL. The role of cellular oxidative stress in regulating glycolysis energy metabolism in hepatoma cells. Mol Cancer. 2009;8(1):1–5.

    Article  Google Scholar 

  51. Sanjuan-Pla A, Cervera AM, Apostolova N, Garcia-Bou R, Victor VM, Murphy MP, et al. A targeted antioxidant reveals the importance of mitochondrial reactive oxygen species in the hypoxic signaling of HIF-1 alpha. FEBS Lett. 2005;579(12):2669–74.

    Article  CAS  PubMed  Google Scholar 

  52. Agnihotri S, Golbourn B, Huang X, Remke M, Younger S, Cairns RA, et al. PINK1 is a negative regulator of growth and the Warburg effect in glioblastoma. Can Res. 2016;76(16):4708–19.

    Article  CAS  Google Scholar 

  53. Zahra K, Dey T, Ashish Mishra SP, Pandey U. Pyruvate kinase M2 and cancer: the role of PKM2 in promoting tumorigenesis. Front Oncol. 2020;10:159.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Morais VA, Haddad D, Craessaerts K, De Bock PJ, Swerts J, Vilain S, et al. PINK1 loss-of-function mutations affect mitochondrial complex I activity via NdufA10 ubiquinone uncoupling. Science. 2014;344(6180):203–7.

    Article  CAS  PubMed  Google Scholar 

  55. Sánchez-Caballero L, Guerrero-Castillo S, Nijtmans L. Unraveling the complexity of mitochondrial complex I assembly: a dynamic process. Biochimica et Biophysica Acta (BBA) Bioenergetics. 2016;1857(7):980–90.

    Article  PubMed  Google Scholar 

  56. Xu Y, Miriyala S, Fang F, Bakthavatchalu V, Noel T, Schell DM, et al. Manganese superoxide dismutase deficiency triggers mitochondrial uncoupling and the Warburg effect. Oncogene. 2015;34(32):4229–37.

    Article  PubMed  Google Scholar 

  57. Kawada K, Toda K, Sakai Y. Targeting metabolic reprogramming in KRAS-driven cancers. Int J Clin Oncol. 2017;22(4):651–9.

    Article  CAS  PubMed  Google Scholar 

  58. Zheng J, Sato M, Mishima E, Sato H, Proneth B, Conrad M. Sorafenib fails to trigger ferroptosis across a wide range of cancer cell lines. Cell Death Dis. 2021;12(7):698.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Arteaga CL, Brown TD, Kuhn JG, Shen HS, O’Rourke TJ, Beougher K, et al. Phase I clinical and pharmacokinetic trial of Brequinar sodium (DuP 785; NSC 368390). Cancer Res. 1989;49(16):4648–53.

    CAS  PubMed  Google Scholar 

  60. Peters GJ. Re-evaluation of Brequinar sodium, a dihydroorotate dehydrogenase inhibitor. Nucleosides Nucleotides Nucleic Acids. 2018;37(12):666–78.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable

Funding

 This work was supported by a grant from the National Natural Science Foundation of China [No. 82172804;82003232], China International Medical Foundation [No. NTC03932266], Key Program of Jiangsu Provincial Department of Science and Technology [No. BE2019756], Key Program of Jiangsu Commission of Health [No.K2019028], The sixth "333 Talents" Outstanding Young Talents Project of Jiangsu Province and Nasopharyngeal Carcinoma Cohort Program of Nanjing Medical University [No.NMUC2021011A].

Author information

Authors and Affiliations

Authors

Contributions

The Idea was conceived by AA, AA and AX performed the literature search and drafted the manuscript; LW and HX critically revised the article. All authors read and approved the final manuscript.

Corresponding author

Correspondence to He Xia.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

No competing interest.

Additional information

Publisher's Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Amos, A., Amos, A., Wu, L. et al. The Warburg effect modulates DHODH role in ferroptosis: a review. Cell Commun Signal 21, 100 (2023). https://doi.org/10.1186/s12964-022-01025-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-022-01025-9

Keywords