Next Article in Journal
Serum TLR2 and TLR9 in Prostate Cancer Patients in Relation to EBV Status
Previous Article in Journal
The Beneficial Effects of Prenatal Biotin Supplementation in a Rat Model of Intrauterine Caloric Restriction to Prevent Cardiometabolic Risk in Adult Female Offspring
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tricarboxylic Acid Cycle Relationships with Non-Metabolic Processes: A Short Story with DNA Repair and Its Consequences on Cancer Therapy Resistance

by
Enol Álvarez-González
1,2,3 and
Luisa María Sierra
1,2,3,*
1
Departamento de Biología Funcional, Área de Genética, University of Oviedo, C/Julián Clavería s/n, 33006 Oviedo, Spain
2
Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
3
Instituto de Investigación Sanitaria del Principado de Asturias, Avda. HUCA s/n, 33011 Oviedo, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(16), 9054; https://doi.org/10.3390/ijms25169054 (registering DOI)
Submission received: 18 July 2024 / Revised: 8 August 2024 / Accepted: 20 August 2024 / Published: 21 August 2024
(This article belongs to the Special Issue DNA Damage, Oxidative Stress and Metabolism in Cancer)

Abstract

:
Metabolic changes involving the tricarboxylic acid (TCA) cycle have been linked to different non-metabolic cell processes. Among them, apart from cancer and immunity, emerges the DNA damage response (DDR) and specifically DNA damage repair. The oncometabolites succinate, fumarate and 2-hydroxyglutarate (2HG) increase reactive oxygen species levels and create pseudohypoxia conditions that induce DNA damage and/or inhibit DNA repair. Additionally, by influencing DDR modulation, they establish direct relationships with DNA repair on at least four different pathways. The AlkB pathway deals with the removal of N-alkylation DNA and RNA damage that is inhibited by fumarate and 2HG. The MGMT pathway acts in the removal of O-alkylation DNA damage, and it is inhibited by the silencing of the MGMT gene promoter by 2HG and succinate. The other two pathways deal with the repair of double-strand breaks (DSBs) but with opposite effects: the FH pathway, which uses fumarate to help with the repair of this damage, and the chromatin remodeling pathway, in which oncometabolites inhibit its repair by impairing the homologous recombination repair (HRR) system. Since oncometabolites inhibit DNA repair, their removal from tumor cells will not always generate a positive response in cancer therapy. In fact, their presence contributes to longer survival and/or sensitization against tumor therapy in some cancer patients.

1. Introduction

Basic cellular metabolic processes are not identical in all cells since at least cancer cells present a distinct metabolic phenotype known as “aerobic glycolysis” [1,2]. In the last decades, research has shown the relevance of the tricarboxylic acid (TCA) cycle. Metabolic changes involving this central hub for energy metabolism, macromolecule synthesis and redox balance [3,4,5] are linked to different non-metabolic cell processes. Some of these changes are related to variations in the levels of some TCA cycle metabolites in normal cellular function [5]. However, most of them are the consequence of mutations in genes related to the TCA cycle [5,6,7,8]. In fact, the discovery of these mutations, as well as their effects, represents a turning point in the fields of cancer genetics and metabolism [7,9,10,11,12]. Moreover, they show an important connection between the TCA cycle, immunity and cancer [13,14,15,16,17,18,19].
Furthermore, in recent years, an additional link has emerged between the TCA cycle and epigenetic processes [20,21,22,23,24]. Epigenetics impacts aging [25], innate immune memory [18] and, most importantly, DNA damage repair [8,11,24,26,27,28,29,30,31,32,33]. The interaction between the TCA cycle and DNA repair might be involved in cancer therapy resistance, and this is the main subject of this review.

2. TCA Cycle

The TCA cycle is a metabolic process taking place in mitochondria and linking glucose oxidation with the respiratory chain. This is a specific aerobic biodegradation process that starts with the catabolism of acetyl-coenzyme A (acetyl-CoA) to produce the reduced coenzymes NADH and FADH2, as well as CO2 [34] (Figure 1).
This cycle provides cells with energy and intermediates for biosynthetic pathways [12], and it is defined as the central biochemical process in eukaryotic life [18].
Since its discovery by Krebs and Johnson in 1937 [35], many reviews have been published about the TCA cycle, several of them in the last two decades. Among them, there are nice and thorough works about energy generation and/or metabolism remodeling [36,37] and even one modifying Krebs’ original concept [38]. Several reviews were also published about the TCA cycle relationships with human inborn errors of metabolism [39], neurometabolic disorders [40], immunity and inflammation [18,41,42,43], viral infections [44] and the control of mammalian stem cell fate [45].
Most of the relationships between the TCA cycle and these non-metabolic cell processes arise from the fact that some of the cycle metabolites have additional functions outside of the TCA cycle. Among them, there are direct metabolites such as citrate, α-KG, succinate and fumarate, but also new and indirectly generated metabolites, like itaconate (Figure 2A) and 2-hydroxyglutarate, 2HG (present as two enantiomers [39]) (Figure 2B) [10,15,16,17,18,19,42,43,46,47,48]. In fact, there is quite a lot of information on the additional roles of citrate [12,13,16,17,18,42,43,49,50,51,52,53], itaconate [15,16,17,18,43,49,54,55,56], α-KG [12,57,58,59,60], 2HG [16,39,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79], succinate [13,15,16,18,42,43,63,80,81,82,83] and fumarate [7,43,63,84,85,86,87] in the mentioned relationships.
There is also a relevant connection between the TCA cycle and cell proliferation and cancer [40,88,89,90,91,92,93]. It constitutes the foundation for the newly described relationship we would like to highlight: the relationship established by 2HG, succinate and fumarate with DNA repair. This interaction is established either with the epigenetic control of chromatin structure [4,11,23,25] and, therefore, indirectly with DNA damage repair [8,11,24,28,29] or directly with some DNA repair proteins [8,26,27,30,31,32,33]. This new connection constitutes one of the most important side effects of the TCA cycle metabolites, since it might provide new potential approaches and new therapeutic targets [7,12,22] in cancer treatments. Specifically, the effects of metabolites on DNA damage repair, mostly inhibitory, might be linked to the suppression of drug resistance. Consequently, this effect of metabolites is especially noteworthy due to its potential implications for overcoming the resistance commonly encountered in cancer therapies.

3. TCA Cycle Metabolites, Cell Proliferation and Cancer

α-KG is one of the most important TCA cycle metabolites. In addition to its role in linking this cycle with non-metabolic processes, it is the fundamental key in the relationship between cell proliferation and cancer and, therefore, with the new connection with DNA repair. This metabolite is a co-substrate for the α-KG/Fe2+-dependent dioxygenases (α-KGDD), a family of enzymes able to catalyze the hydroxylation of different substrates, including nucleic acids, proteins, and fatty acids, using Fe2+ as a cofactor [94,95]. Some of the relevant enzymes in this superfamily are (i) prolyl hydrolase domain-containing proteins (PHDs) [96], of the hypoxia-sensing system; (ii) Ten-Eleven Translocation (TET) DNA demethylases [97]; (iii) Jumonji domain-containing (JmjCs) histone-lysine demethylases [98]; and (iv) AlkB homolog proteins [99] that remove nitrogen alkylation damage from the DNA [100,101].
α-KG plays a crucial role in the hypoxia-sensing system, a mechanism that has evolved in multicellular organisms to adapt to low oxygen levels [102,103,104]. The system is based on the existence of two types of proteins. The first one is that formed by oxygen-dependent hypoxia-inducible factors (HIF-α), including the most relevant HIF-1α, which can detect and activate transcriptional responses when oxygen levels are low [57,104,105]. The second type is constituted by PHD proteins, present in normal oxygen conditions, that hydroxylate different subunits of HIF-α factors, leading to their degradation in the proteasome [106,107].
The TET 5-methylcytosine (5-mC) dioxygenases catalyze the oxidation of 5-mC to 5-hydroxymethylcytosine (5-hmC) [108,109] and then to 5-formylcytosine (5-fC) and 5-carboxylcytosine (5-cC) [110]. With these reactions, they demethylate DNA in a replication-independent process [110]. They were proposed to work in regulating DNA methylation fidelity [111]. Moreover, since their inactivation contributes to both DNA [111] and RNA hypermethylation [112], they also work on the regulation of gene expression [110,112].
The JmjC histone-lysine demethylases (KDMs) form one of the two groups of histone-lysine demethylases, which are subdivided into seven families [113,114]. Described by Tsukada et al. [98] as enzymes that depend on Fe2+ and α-KG for their activity, they may be modulated by metabolites [115].
The AlkB homolog proteins that are present in different organisms, including humans, remove alkyl groups from nitrogen atoms present on the nitrogen bases of the DNA and also of the RNA [99,100,101,116,117,118].
The role of α-KG is equivalent in all these proteins. Low levels of this metabolite would inactivate them, maintaining (i) stable levels of HIF-α factors, even in oxygen normal levels [106]; (ii) high levels of DNA and histone methylation [21,22,23], which is key to keeping a closed chromatin structure and inhibition of gene expression [110,119]; and (iii) high levels of DNA damage that could be the origin of mutations [120,121]. All these altered processes generate cellular conditions that might be the starting point of cell proliferation.
The same effects caused by low levels of α-KG may be achieved by the aberrant accumulation of some TCA cycle metabolites due to mutated enzymes. Since those metabolites may present pro-oncogenic activity that contributes to tumor development and progression, they are called oncometabolites. This is the case for 2HG [10], succinate [122] and fumarate [84].
2HG is present in two enantiomers: D- and L-2HG, also known as (R)- and (S)-, respectively, with IUPAC nomenclature [39,62,65]. When erroneously accumulated, they contribute to the development of different types of tumors. D-2HG accumulates in certain brain tumors, such as gliomas and glioblastomas, with gain-of-function mutations in the IDH1 and IDH2 genes encoding the IDH1 and IDH2 enzymes, respectively [10,62,65,123,124,125]. L-2HG accumulates in brain tumors [63] and also in kidney tumors (like renal cell carcinoma, RCC) as the consequence of a mutated L-2HGDH enzyme [61,63,64] (Figure 2B).
Succinate accumulation, due to loss-of-function mutations in genes encoding the subunits of SDH [90,126,127,128,129,130], leads to the development of various types of tumors, such as paragangliomas and pheochromocytomas [9,29,126,130,131,132,133,134], gastrointestinal stromal tumors (GISTs) [127,129,132,135,136,137,138,139], renal carcinomas [131,132,140,141], pituitary tumors [131,142,143] and others [127,131,132,134,144]. All this information has led to the classification of SDH as a tumor suppressor [131,135,141,145].
The accumulation of fumarate [7,84] due to loss-of-function mutations at the FH encoding gene [84,146,147] causes the development of hereditary leiomyomatosis and renal cell cancer (HLRCC) [84,146,148,149,150,151,152]. Additionally, it is also the origin of renal cancer (RCC) [63,153,154], leiomyoma with bizarre nuclei [155], malignant pheochromocytoma and paragangliomas [133,156,157], and even breast and bladder carcinomas [126,149], as well as neuroblastomas and gliomas [158]. Moreover, the reduction of FH activity may induce disease progression in chronic myeloid leukemia [159]. Because of its role in these carcinogenesis processes, FH was classified as a tumor suppressor [84,86,158,160].
Succinate, fumarate and 2HG work as oncometabolites because they share a chemical structure similar to that of α-KG (see Figure 1) that enables them to act as competitive inhibitors of αKGDD enzymes [22,61,161,162].
The link between 2HG and the hypoxia-response pathway is unclear due to the conflicting roles of its two enantiomers [62,63,64,65]. However, the accumulation of succinate and fumarate clearly stabilizes the hypoxia-inducible factor HIF1-α in normoxic conditions [131,145,146,158,163,164,165,166], causing pseudohypoxia. Succinate, fumarate and 2HG also inhibit, although in different degrees [63], TET DNA (L-2HG appears to be a more potent inhibitor of TET enzymes than D-2HG [65]) and JmjC KDM histone demethylases. This inhibition results in increased genomic DNA methylation and chromatin condensation, which involves the oncometabolites in the epigenetic regulation of gene expression [22,61,63,135,136,147,158,162,165,167,168,169,170]. Their accumulation in epithelial cells causes epithelial-to-mesenchymal transition (EMT), giving these cells increased invasive and migratory abilities [63,85,168,171,172,173]. They are also involved in the development and progression of renal cell cancer [63]. Finally, at least 2HG and fumarate inhibit AlkB homolog proteins [116,117,118].
Moreover, these oncometabolites present other effects that might also be important for cell proliferation, such as the impairment of glutathione (GSH) production [77], increasing reactive oxygen species (ROS) production [77,78,127] or influencing tumor immune responses by affecting dendritic cells or macrophages in tumor micro-environments [174,175]. D-2HG has been found to inhibit the activity of cytochrome c oxidase in the mitochondrial electron transport chain [65]. Furthermore, fumarate integrates immune and metabolic circuits to generate monocyte epigenetic reprogramming in the induction of trained immunity through the inhibition of KDM5 histone demethylases [81,176].

4. TCA Cycle, Oncometabolites and DNA Repair

As already mentioned, one of the most relevant relationships between the TCA cycle and a non-metabolic process is that established with the DNA damage response (DDR) and specifically with DNA repair mechanisms [24,26,27,28,29,30,31,32,33]. DDR is the set of mechanisms that detects DNA damage, signals its presence and promotes its repair [177,178,179,180,181,182].
The different types of lesions or adducts that modify nitrogen bases and/or the deoxyribose-phosphate backbone, commonly known as DNA damage, can be detected in all kinds of organisms [120,121]. They can be generated either as the result of endogenous cellular metabolism or as the consequence of exposure to external genotoxic agents [120,121]. The types of DNA damage include (i) monoadducts—lesions that affect one nitrogen base; (ii) cross-links—intra- and inter-strand ones—which are lesions affecting two nitrogen bases in the same strand or in opposite ones, respectively; and (iii) single- and double-strand breaks—breaks in the sugar-phosphate bonds—in one (SSB) or both strands (at the same position, DSB) of the DNA molecule [120,121].
If not removed from the DNA, all these types of DNA damage may be the source of genetic instability and mutations [120,121,178,179,180,182]. To prevent this possibility, all organisms have DNA damage response mechanisms to remove the damage or to process it to avoid its immediate danger [8,120,179]. These DDR mechanisms depend on the extent of the damage [183]. They start with (1) signaling or detection of the damage; (2) the cell response to provide time for its removal/processing, which in eukaryotes includes the control of cell cycle progression; (3) the actual removal and/or processing of the damage by DNA repair systems; and (4) cell apoptosis or cell senescence that occurs when the DNA damage cannot be removed in time [120,169,177,178,179,180,181,182,184].
Among the types of DNA damage, the most toxic or lethal ones, when not removed, are the DSBs [169,177,185,186,187], which can lead to chromosomal mutations and/or rearrangements [120,169,179]. Perhaps because of their danger, there are several mechanisms that can remove or process DSBs in a fast way [187], like homologous recombination repair (HRR), non-homologous end joining (NHEJ), alternative end joining (Alt-EJ) and single-strand annealing (SSA) [120,188,189,190,191,192,193]. The HRR system is an error-free DNA repair mechanism, whereas the others may introduce errors in the DNA when removing the DSBs [120,189,191,192]. Which mechanism should act to remove a specific DSB is not completely known. It seems to depend on several factors, such as the cell cycle stage, the 53BP1 protein and also the nature of the break ends [184,188,191,192,193]. DSBs induced by external agents might present base lesions at the break sites that can play a role in the choice of the repair mechanism [187].
It is in this context of DNA damage and DDR, or specifically DNA repair that the TCA cycle presents new and important non-metabolic effects. First, because oncometabolites induce increased levels of ROS [63,67,77,78,127,194], and ROS are a source of DNA damage, both endogenous and exogenous [177,195]. Second, because by inhibiting PHD proteins, oncometabolites induce pseudohypoxia conditions [131,145,146,158,163,164,165,166]. Hypoxia contributes to ROS induction [196] and also to a decreased expression of DNA repair genes in some tumor tissues [196,197], inhibiting homology-directed repair, mismatch repair and base excision repair pathways [196,197,198].
Third, and more importantly, oncometabolites influence DDR modulation through their effects on at least four different pathways. The first one, which we call the AlkB pathway, deals with the removal of alkylation damage from nitrogen atoms of nitrogen bases, and it is inhibited by at least fumarate and 2HG [26,27,199]. The second one, the MGMT pathway, removes alkylation damage from the oxygen atom at position 6 of guanine (O6-G) by direct DNA damage reversal [116,200], and it is inhibited in IDH and SDH mutated cell tumors [201,202]. The last two pathways deal with the repair of DSBs, although with opposite effects: (i) the FH pathway generates fumarate in the nucleus to help with the repair of this damage [30,31,32,33,160]; and (ii) the chromatin remodeling pathway activates the HRR system to remove DSBs, and it is inhibited by the three oncometabolites [24,28,29].

4.1. The AlkB Pathway

The first pathway in the relationship between the TCA cycle, oncometabolites and DNA repair is established because some repair proteins, such as AlkB and its homologs in different organisms, are α-KG–dependent dioxygenases [99] that may be inhibited by oncometabolites [26,27,199] (Figure 3).
The Escherichia coli inducible protein AlkB was demonstrated to work as a DNA repair enzyme on induced alkylation damage [100,101,203].
Firstly, N1-methyladenine (N1-metA) and N3-methylcytosine (N3-metC) replication-blocking lesions, generated in single-stranded and double-stranded DNA, were the only described substrates of this enzyme [100,101]. Later, more lesions were found to be repaired by this enzyme: (i) all the four possible exocyclic etheno adducts of the DNA nucleobases; (ii) other DNA monoalkyl lesions, like N6-methyladenine (N6-metA), N1-methylguanine (N1-metG), N2-methylguanine (N2-metG), N4-methylcytosine (N4-metC) and N3-methylthymine (N3-metT); and (iii) even monoalkyl RNA lesions, N1-metA, N3-metC and N1-metG [116,117,118]. The enzyme can work on single- and double-stranded substrates in DNA and RNA [117,118].
Although all these lesions are methylated nucleobases, AlkB seems to be capable of also removing higher-order alkyl adducts [117]. Among the DNA adduct substrates of AlkB activity, special attention has been given to exocyclic N6-metA, N2-metG and N4-metC since they are not deleterious, do not generate mutations and might be post-replicative markers [117,203]. However, they are removed from the DNA as other mutagenic or replication-blocking adducts. This situation suggests a potential additional biological function for AlkB protein besides DNA repair, like regulating (i) the discrimination between DNA strands, (ii) the replication start or (iii) even the gene expression through the control of these post-replicative markers [117,203].
These lesions are removed by oxidative demethylation, generating oxy-ferryl intermediates that decompose and are released as aldehydes [100,116].
The AlkB homologs in mammals show a narrower range of substrates that include etheno adducts, N1-metA, N3-metC and N3-metT DNA mono-alkyl lesions, as well as mono-alkyl RNA lesions [117,118].
The connection between AlkB homologs and oncometabolites was discovered when some IDH-mutant glioma patients responded to a combination of alkylating agent chemotherapy, and this outcome was linked to the inhibition of the AlkB human homologs, ALKBH repair proteins, by the oncometabolite 2HG [26,62].
Like the JmjC KDMs and TET proteins, AlkB belongs to the Fe2+/α-KG-dependent dioxygenases [99], and the family includes nine distinct proteins in human cells (AlkB homolog ALKBH1 to ALKBH8 and FTO) [117,118]. These enzymes were inhibited in vitro not only by D-2HG [26,27,62] but also by L-2HG [27], and the repair of alkylation DNA damage was impaired in IDH-mutant glioma cells [26,62].
Later, fumarate was described to also inhibit these enzymes in vitro and in vivo [199]. The possible effect of succinate on the AlkB homologs has not yet been reported, although it is expected to be found.
In this pathway, the accumulation of oncometabolites leads to the inhibition of DNA damage repair, which might be a desirable situation in chemotherapy treatments.

4.2. The MGMT Pathway

In eukaryotes, the MGMT gene encodes the O6-methylguanine DNA methyltransferase, or MGMT, protein present in all kinds of organisms [204]. In prokaryotes, proteins with the same function are encoded by the ada and ogt genes [205,206]. The function of MGMT proteins, also known as alkyltransferases, and even as suicide proteins, is the direct removal of alkyl groups from oxygen atoms in DNA, specifically from O6-guanine (O6-G), in a process that inactivates them [116,200,204,207]. Alkylation of O6-G is not a very common type of induced DNA damage. Only alkylating agents following an SN1 unimolecular substitution reaction can generate it, and at low frequencies [116,120,200,204,208]. However, it is a miscoding pre-mutagenic and pre-carcinogenic DNA lesion [116,200]. It is induced by many of the alkylating agents used in cancer chemotherapy, like temozolomide (TMZ) [200,204,207,209], and its removal from the treated cells by MGMT normally results in therapy failure. In fact, low levels of, or not detectable, MGMT activity are linked to better therapy response and longer survival [201,209,210,211].
In most cases, the lack of MGMT activity is directly associated with the methylation of CpG islands on the gene promoter [210]. Consequently, the methylation status of this gene promoter is used as a prognostic and predictive biomarker in brain tumors like glioblastoma, glioma, astrocytoma or oligodendroglioma [210,212,213,214,215]. Moreover, the relevance of MGMT activity was detected for other tumors, like gastric cancer [216], GISTs [202,217] and lung cancer [218], but not for breast cancer [219].
The influence of oncometabolites on this repair process was discovered because many of the brain tumor patients with a methylated MGMT promoter in tumor cells were IDH mutants [220,221,222], and they showed the best response to chemotherapy with alkylating agents [223,224,225,226,227]. Furthermore, some malignant SDHB-mutated pheochromocytoma and paraganglioma were associated with hypermethylation of the MGMT promoter and responded to TMZ [228], as well as some SDH-deficient GIST tumors [229].
The MGMT promoter methylation seems to be linked to a hyper-methylator phenotype, also known as CpG island methylator phenotype (CIMP) that can be established through the inhibition of TET and KDM proteins by mutations at IDH [230,231,232,233] or SDH genes [202,217,228] (Figure 3).
It is clear, then, that the accumulation of at least 2HG and succinate inhibits MGMT repair through the induction of a hyper-methylator phenotype, and this is a welcome situation in the response to chemotherapy.

4.3. The FH Pathway

FH is an enzyme present in all kinds of organisms, prokaryotic and eukaryotic [32,33,160,234], and in these last ones, equally distributed between cytosol and mitochondria [160].
The third pathway linking the TCA cycle and DNA repair was found when studying the role of the yeast cytosolic FH enzyme [30] (Figure 4). As a member of the DDR, this enzyme is translocated to the nucleus after the induction of DNA damage, and it helps with the recognition of DSB sites in the chromatin and with cell recovery in a function that seems to be HIF-independent [30]. In fact, FH levels increase after the induction of DNA damage and when FH is not working, the cells are sensitive to DSBs because their repair is impaired, both in yeast and human cells [30].
Since its discovery, more information has been gathered on this pathway. Cytosolic yeast FH, affected by post-translational modifications, is not active when there is no damage in the DNA [160]. Phosphorylation of its Ser 46 by PAK4 kinase retains this enzyme in the cytosol of human lung cells [235]. When DNA damage is induced, FH levels increase, and the enzyme is activated by removing these modifications or by synthesizing new molecules without modifications [160], and it moves to the nucleus, specifically to DSB sites [31,32,33,160]. At least in human cells, DNA-PK phosphorylates the Thr236 position of the translocated FH molecules, leading to their interaction with the H2A.Z histone variant [31]. This histone is present in nucleosomes at DSBs and contributes to shifting the chromatin to an open conformation [236]. FH then synthesizes fumarate from malate [31,158,160]. This locally generated fumarate inhibits KDM2B histone demethylase, enhancing the dimethylation of histone H3 lysine 36 (H3K36) [31,158,160], signaling the position of DNA damage and opening the chromatin to the necessary repair proteins [237]. In fact, in human cells, all these steps lead to the accumulation of Ku70-containing DNA-PK at DSB regions for the NHEJ system to work and help with cell survival [31,32]. These findings reveal a feedback mechanism that underlies DNA-PK regulation by chromatin-associated fumarase and an instrumental function of fumarase in regulating histone H3 methylation and DNA repair [31].
Contrary to what happens in human cells, in yeast, the repair process activated by this FH pathway is the HRR, with a relevant role of this protein in the DSB resection through its interaction with the Sae2 endonuclease [32,33]. Moreover, during DNA replication stress, the fumarate in the nucleus enhances the survival of yeast lacking Htz1p histone (H2A.Z in mammals) by increasing the methylation of histone H3 lysine 4 (H3K4) through inhibition of the corresponding histone demethylase (Jhd2p) [238]. Finally, fumarase seems to indirectly influence DDR by binding to the desulfurase Nfs1p in the mitochondria, protecting it and allowing the formation of Fe–S clusters, which are essential cofactors for DNA repair enzymes [239].
The role of FH in the response to DSBs is not restricted to eukaryotic cells since a fumarase protein involved in DNA damage response and induced by the presence of DNA damage was also found in Bacillus subtilis [240].
This pathway, with the accumulation of fumarate at specific DNA sites, contributes to the repair of DSBs.

4.4. The Chromatin Remodeling Pathway

To facilitate the access of repair proteins to the damage sites for an accurate repair of DNA, the cells need to present an open chromatin structure, which may be achieved through the work of histone modifiers [186,237,241], DNA methylation control [110] and chromatin remodelers that are recruited at the DNA lesion site [186,237,241,242].
Histone modifications play a fundamental role in chromatin structure, dynamics and functions and, among them, methylation of lysine residues in H3 and H4 and their corresponding demethylations are essential to chromatin regulation [98,114,170,243]. The JmjCs histone-lysine KDM4 (A–D) demethylase family contains the most relevant histone modifiers in the context of the epigenetic regulation of chromatin structure [98,113,169,170]. They remove methyl residues from the trimethylated histone H3 lysine 9 (H3K9me3) [113,114,169], which is necessary for maintaining an open chromatin structure [170]. H3K9me3 represents a barrier to DSB repair [169], and the enzyme removing it is then part of the DDR [169,244]. In fact, the C-terminal region of KDM4D mediates its rapid recruitment to DNA damage sites, where it is required for the efficient phosphorylation of some substrates of the DDR sensor ATM protein [244]. This recruitment depends on the poly (ADP-ribose) polymerase 1 (PARP1), which ADP-ribosylates KDM4D [244].
After the discovery of the effect of oncometabolites in chromatin structure through the inhibition of KDM demethylases [22,65,136,147,158,162,165,169,170], one question started to arise: were the oncometabolites modulating DNA damage induction and/or DNA damage cellular responses in ways not related to FH, AlkB or MGMT? [183,242,245,246]. The answer to this question constitutes the chromatin remodeling pathway in the relationship between the TCA cycle and DNA repair and was quite recently completely deciphered [24,28,29] (Figure 5).
The first insights into this pathway were provided by the reports about the following effects: (i) of hypoxia on inducing DNA damage and inhibiting DNA repair [196,197,198]; (ii) of D-2HG on inducing an impaired HRR system that rendered cells sensitive to DNA damaging agents and PARP1 inhibitors [28,247]; and (iii) of succinate and fumarate on inducing this same HRR problem, with the same consequences [29,248].
These findings uncover an unexpected connection between oncometabolites, altered DNA repair and genetic instability [28]. They were followed by results showing that the repair activity of DSBs induced by ionizing radiation was markedly reduced in IDH mutant cell lines [28] and in FH and SDH mutant cells [29]. Moreover, IDH mutant cells and those with increased levels of fumarate and succinate show an increased persistence of unrepaired DSBs, even in untreated cells, as shown with the neutral comet assay [28,158]. However, it is not clear whether 2HG, succinate or fumarate induce DNA damage in vitro [28,29,245,246] since exogenous exposures to any of them increase the levels of DSBs [24,246].
In normal cellular conditions, the induction of DSBs generates a very rapid, high and short-timed increase of H3K9me3, brought down by KDM4 demethylases [28,29]. This is the most important signal for activating the HRR system through its interaction with TIP60 acetyltransferase [249]. However, if the levels of H3K9me3 were increased because exposure to oncometabolites inhibits KDM4 demethylases, this signal is masked [24]. The lack of this signal impairs the activation of TIP60 [249]. Consequently, the recruitment of ATM protein to sites of DSBs is inhibited, which, in turn, attenuates the foci of the RAD51 and BRCA2 proteins. The impairment of these proteins, all of which are essential for normal HRR activity, inhibits this system, with a defect at the end-resection step [24].
In this chromatin remodeling pathway, the accumulation of oncometabolites inhibits the repair of DSBs. This lack of repair might be a desirable situation in radiotherapy treatments.
In addition to directly impairing the HRR system, this chromatin remodeling pathway might be linked to the inhibition of other DNA repair systems, like the nucleotide excision repair (NER), the base excision repair (BER) or the mismatch repair (MMS) systems through maintaining a closed chromatin structure. In that case, the inhibition of TET DNA demethylases [167], together with that of KDM histone demethylases, would be the key to the oncometabolite control process, although some control through maintaining hypoxia levels should not be excluded [196,197,198].
Obviously, chromatin remodeling might be achieved through other processes, some of which are also related to the TCA cycle, like the histone acetylation/deacetylation alternation, which is also used to devise chemotherapy treatments with the use of histone deacetylation inhibitors [250,251,252]; however, since the link with the TCA cycle is the acetyl-CoA metabolite [253,254,255,256] and not the oncometabolites, no more information about this process will be provided in this review.

5. Oncometabolites, DNA Repair and Therapy Treatment Resistance

The activities of many chemotherapy drugs and of radiotherapy depend on the generation of DNA damage that would induce apoptosis and cell death in the treated tumor cells [257,258]. It is evident, then, that functional DNA repair systems that remove the DNA damage before it kills the cells would help with tumor survival and, therefore, contribute to therapy resistance [179,224,258,259,260].
In fact, DNA repair systems have been linked to drug resistance since a long time ago [261,262,263], especially when dealing with relevant chemotherapy drugs (because they are the best available treatment for some specific tumor types), like, for instance, cisplatin or TMZ [224,257,258,264,265]. The role of DNA repair in drug resistance is so relevant from a negative point of view that there are proteins from the different repair systems that are used as biomarkers, prognostic and/or predictive, in tumor therapy [201,210,212,213,214,215]. Moreover, some processes in the repair systems are used to develop selective agents targeting specific DNA repair pathways [181,257].
As previously discussed, increased levels of oncometabolites are the origin of tumor development; therefore, a good approach to treating these tumors was to reduce such high oncometabolite levels using inhibitors of the mutant enzymes that generate them [5,63,266,267,268,269,270,271,272,273]. This type of treatment is only possible for IDH mutant tumors since IDH enzymes are the only ones with gain-of-function mutations [7,10,62]. However, this limitation is not a problem, considering the many tumor types due to increased 2HG levels [67]. Although the use of inhibitors provides a good response at the beginning of the treatment, after some time, some patients develop chemotherapy resistance [268,269,271,274,275,276]. The resistance is linked to (i) new additional mutations in IDH genes [273,277]; (ii) the already mentioned hyper-methylator phenotype of oncometabolites, which may contribute to resistance depending on the silenced genes [268,274,278]; (iii) the inhibition of ferroptosis [279,280]; or (iv) the angiogenesis-promoting phenotype of tumor-associated macrophages (TAMs), which is supposed to be activated by hypoxic tumor cell-derived oncometabolites, as demonstrated in the case of succinate [281].
In addition, high levels of oncometabolites seem to be directly related to resistance to different therapy drugs by several very specific mechanisms, as already reviewed [274]. In particular, high levels of oncometabolites may directly promote cancer treatment resistance (i) by upregulating the nuclear factor NRF2 pathway, which inhibits apoptosis and increases both antioxidant responses and the expression of efflux proteins, like in the case of fumarate through the KEAP protein [282] or succinate through hyper-succinylation [274]; (ii) by the reduction of SDH activity, which increases succinate levels and stabilizes HIF-1α protein after treatment with transforming growth factor β (TGF-β) in osteosarcoma cells [283]; (iii) by inhibiting the anti-tumor immune response, like in the case of 2HG [284]; or (iv) by promoting angiogenesis, like fumarate and 2HG [282]. Additionally, resistance to the topoisomerase II inhibitor etoposide was linked to the presence of mutated IDH genes but not to the accumulation of 2HG metabolite [285].
The connections between TCA cycle oncometabolites and DNA repair might be the reason for some of the therapy resistance cases. When they are present, like in the case of the locally generated fumarate, they help with the repair of induced DNA damage [30,31,32,33,286]. When the oncometabolites are removed from the cells, the repair systems are no longer inhibited [24,26,27,28,29,287,288] (Figure 6). However, the oncometabolite effects on therapy responses were not always identified in the detected responses. Some of the resistance responses were considered cases that do not respond to chemotherapy with TCA cycle enzyme inhibitors [266,289]. Other treatment responses were viewed as cases of longer survival in the presence of oncometabolites [220,224,288,289,290,291] or as a sensitization effect of oncometabolites [224,272,273,291,292,293]. None of these responses were ever related to the oncometabolite effects on DNA repair.
One of the pathways identified so far between the TCA cycle and DNA repair is that established with the MGMT protein: the oncometabolites create a hyper-methylator phenotype in tumor cells that inhibits MGMT gene expression [202,217,228,229,230,231,232]. In tumors treated with alkylating agents, the presence of oncometabolites inhibits the synthesis of this protein [220,221,222], and it re-sensitizes [223,288,294] the cells that were resistant to therapy due to MGMT activity [201,209,210,211,295].
Increased levels of oncometabolites also contribute to sensitization against alkylating agents via inhibition of the human ALKBH1 to ALKBH8 and FTO proteins, homologs of the AlkB protein. Since these proteins repair alkylation DNA and RNA damage, they protect against this kind of damage [288,296]. In fact, these proteins are described to be responsible for chemotherapy resistance in different tumors, not only to alkylating agents [297] but also to cisplatin [298]. Their inhibition by 2HG and fumarate sensitizes the tumors against these chemicals [26,62,288,295]. However, the depletion of FTO and ALKBH5 in epithelial ovarian tumors renders them resistant to PARP inhibitor treatment [299].
The cases of longer survival in the presence of oncometabolites and many of the sensitization ones were detected in patients with tumors identified as IDH mutant [224,272,288,289,290,291,292,293,300] (Figure 6). Nevertheless, these types of cases might also be expected in SDH and FH mutant tumors if they could be compared with equivalent non-mutant ones. In addition to their effect on AlkB genes, these cases are due to the role of oncometabolites in the inhibition of KDM proteins, which triggers the impairment of the HRR system [24]. When the chemotherapy-induced DNA damage cannot be repaired because of the oncometabolite levels, tumor cells die (by whatever pathway), and patients present with longer survival [8]. In this case, effects would be expected when cancer therapy is performed with agents inducing DSBs, such as ionizing radiation, radiation mimetic chemicals or topoisomerase II poisons, like etoposide, for instance.
It should be noted that to prevent tumor resistance to therapy, a novel strategy that combines metabolic/epigenetic alterations with immunotherapy is being used to obtain better antitumor responses [284,301,302,303].There is another effect of oncometabolites, specifically of fumarate, that might be important in drug resistance. As discussed in the FH pathway, the combination of FH activity with the NHEJ system in the nucleus contributes to the repair and/or processing of DSBs in DNA and, consequently, also to the maintenance of genomic stability and cell survival [31,32]. The role in this pathway gives FH a second activity as a tumor suppressor [32]. However, the result of this activity is opposed to the FH effects on the AlkB and chromatin remodeling pathways. In the FH pathway, a functional FH contributes to DNA damage repair through the generation of fumarate. In the AlkB and chromatin remodeling pathways, a mutated, non-functional FH inhibits DNA damage repair processes, also through the generation of fumarate. Why does fumarate induced at DSB sites not inhibit DNA repair? And why does fumarate induced in the mitochondria not help with DNA repair? The levels and localization of fumarate increase, as well as the time in the cell cycle when fumarate accumulates in both cases, which might explain these differences [32]. Furthermore, although in cultured cells, sensitization to chemotherapy with cisplatin was linked to increased levels of fumarate and malate generated outside the TCA cycle [274], we have not found any case of drug resistance or sensitization linked to this FH pathway in tumor patients, perhaps because it would not be easy to detect.

6. Conclusions

The central hub for energy metabolism—the TCA cycle—establishes relationships with non-metabolic processes through some of its metabolites, both direct and indirect. In addition to the clear and well-studied connections between cancer and oncometabolites and between immunity and signaling metabolites, a new relationship has emerged between oncometabolites and DNA repair that already shows its relevance in the response to DNA damage.
Relevance because, on one hand, the localized accumulation of fumarate aids in the repair, or at least with the processing, of DNA damage as lethal as DSBs. And this help occurs in normal cells exposed to DSB-inducing agents.
On the other hand, relevance because the accumulation of oncometabolites in tumor cells renders them sensitive to DSBs and also to alkylation DNA damage through the inhibition of HRR, AlkB and MGMT repair systems, respectively. These effects of oncometabolites on DNA repair might be the primary reason for their name and the basis for their effect on cancer. However, when considering tumor cells, this lack of repair could be a desirable cell trait in cases of cancer therapy, both chemo- and radiotherapy. At this moment, with the exception of MGMT inhibition, this trait is not clearly exploited because many therapy treatments are based on the use of oncometabolite inhibitors. However, if you remove the oncometabolites, you also eliminate the DNA repair inhibition. Maybe this is the reason why many cases of DNA repair restoration, through the elimination of oncometabolites, are still considered only a failure of chemotherapy with TCA cycle enzyme inhibitors.
Moreover, all the information presented until now opens the possibility of further effects of oncometabolites on other DNA repair systems, although not necessarily in a direct way. Since there are not more dioxygenases among the known repair proteins, other oncometabolite effects might be expected from their role in chromatin remodeling, as indicated. Moreover, why not expect that oncometabolites might influence the ataxia-telangiectasia-related ATR protein, which is a sensor of different types of chemically induced DNA damage?

Author Contributions

Conceptualization and design, L.M.S.; writing and discussion, L.M.S. and E.Á.-G.; work with references, E.Á.-G. and L.M.S.; work with figures, E.Á.-G. and L.M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Gobierno del Principado de Asturias (Oviedo, Spain) through Plan de Ciencia, Tecnología e Innovación (PCTI) co-financed by FEDER funds (Ref. SV-PA-21-AYUD/2021/51399) and by the Ministerio de Ciencia e Innovación (MCI) of Spain under the Project MCI-20-PID2019-104334RB-100.

Informed Consent Statement

Not applicable.

Acknowledgments

E.A.G. acknowledges the Instituto de Investigación Sanitaria del Principado de Asturias (ISPA) (Spain) for funding his predoctoral fellowship.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Nowicki, S.; Gottlieb, E. Oncometabolites: Tailoring our genes. FEBS J. 2015, 282, 2796–2805. [Google Scholar] [PubMed]
  2. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [PubMed]
  3. Arnold, P.K.; Finley, L.W.S. Regulation and function of the mammalian tricarboxylic acid cycle. JBC 2023, 299, 102838. [Google Scholar]
  4. Ciccarone, F.; Vegliante, R.; Di Leo, L.; Ciriolo, M.R. The TCA cycle as a bridge between oncometabolism and DNA transactions in cancer. Semin. Cancer Biol. 2017, 47, 50–56. [Google Scholar] [PubMed]
  5. Anderson, N.M.; Mucka, P.; Kern, J.G.; Feng, H. The emerging role and targetability of the TCA cycle in cancer metabolism. Protein Cell 2018, 9, 216–237. [Google Scholar]
  6. Sciacovelli, M.; Frezza, C. Oncometabolites: Unconventional triggers of oncogenic signalling cascades. Free Radic. Biol. Med. 2016, 100, 175–181. [Google Scholar]
  7. Collins, R.R.J.; Patel, K.; Putnam, W.C.; Kapur, P.; Rakheja, D. Oncometabolites: A new paradigm for oncology, metabolism, and the clinical laboratory. Clin. Chem. 2017, 63, 1812–1820. [Google Scholar]
  8. Gueble, S.E.; Bindra, R.S. Oncometabolites as Regulators of DNA Damage Response and Repair. Semin. Radiat. Oncol. 2022, 32, 82–94. [Google Scholar]
  9. Bardella, C.; Pollard, P.J.; Tomlinson, I. SDH mutations in cancer. Biochim. Biophys. Acta 2011, 1807, 1432–1443. [Google Scholar]
  10. Dang, L.; White, D.W.; Gross, S.; Bennett, B.D.; Bittinger, M.A.; Driggers, E.M.; Fantin, V.R.; Jang, H.G.; Jin, S.; Keenan, M.C.; et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 2009, 462, 739–744. [Google Scholar]
  11. Savani, M.R.; Abdullah, K.G.; McBrayer, S.K. Amplifying the Noise: Oncometabolites Mask an Epigenetic Signal of DNA Damage. Mol. Cell 2020, 79, 368–370. [Google Scholar] [PubMed]
  12. Eniafe, J.; Jiang, S. The functional roles of TCA cycle metabolites in cancer. Oncogene 2021, 40, 3351–3363. [Google Scholar]
  13. Mills, E.; O’Neill, L.A. Succinate: A metabolic signal in inflammation. Trends Cell Biol. 2014, 24, 313–320. [Google Scholar]
  14. Mills, E.L.; Kelly, B.; O’Neill, L.A.J. Mitochondria are the powerhouses of immunity. Nat. Immunol. 2017, 18, 488–498. [Google Scholar] [PubMed]
  15. Murphy, M.P.; O’Neill, L.A.J. Krebs Cycle Reimagined: The Emerging Roles of Succinate and Itaconate as Signal Transducers. Cell 2018, 174, 780–784. [Google Scholar]
  16. Ryan, D.G.; Murphy, M.P.; Frezza, C.; Prag, H.A.; Chouchani, E.T.; O’Neill, L.A.; Mills, E.L. Coupling Krebs cycle metabolites to signalling in immunity and cancer. Nat. Metab. 2019, 1, 16–33. [Google Scholar]
  17. Viola, A.; Munari, F.; Sánchez-Rodríguez, R.; Scolaro, T.; Castegna, A. The Metabolic Signature of Macrophage Responses. Front. Immunol. 2019, 10, 1462. [Google Scholar]
  18. Ryan, D.G.; O’Neill, L.A.J. Krebs Cycle Reborn in Macrophage Immunometabolism. Annu. Rev. Immunol. 2020, 38, 289–313. [Google Scholar]
  19. Scagliola, A.; Mainini, F.; Cardaci, S. The tricarboxylic acid cycle at the crossroad between cancer and immunity. Antioxid. Redox Signal 2020, 32, 834–852. [Google Scholar]
  20. Cervera, A.M.; Bayley, J.P.; Devilee, P.; McCreath, K.J. Inhibition of succinate dehydrogenase dysregulates histone modification in mammalian cells. Mol. Cancer 2009, 8, 89. [Google Scholar]
  21. Letouzé, E.; Martinelli, C.; Loriot, C.; Burnichon, N.; Abermil, N.; Ottolenghi, C.; Janin, M.; Menara, M.; Nguyen, A.T.; Benit, P.; et al. SDH Mutations Establish a Hypermethylator Phenotype in Paraganglioma. Cancer Cell 2013, 23, 739–752. [Google Scholar] [PubMed]
  22. Xiao, M.; Yang, H.; Xu, W.; Ma, S.; Lin, H.; Zhu, H.; Liu, L.; Liu, Y.; Yang, C.; Xu, Y.; et al. Inhibition of α-KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes. Dev. 2012, 26, 1326–1338. [Google Scholar]
  23. Martínez-Reyes, I.; Chandel, N.S. Mitochondrial TCA cycle metabolites control physiology and disease. Nat. Commun. 2020, 11, 102. [Google Scholar] [PubMed]
  24. Sulkowski, P.L.; Oeck, S.; Dow, J.; Economos, N.G.; Mirfakhraie, L.; Liu, Y.; Noronha, K.; Bao, X.; Li, J.; Shuch, B.M.; et al. Oncometabolites suppress DNA repair by disrupting local chromatin signalling. Nature 2020, 582, 586–591. [Google Scholar]
  25. Salminen, A.; Kauppinen, A.; Hiltunen, M.; Kaarniranta, K. Krebs cycle intermediates regulate DNA and histone methylation: Epigenetic impact on the aging process. Ageing Res. Rev. 2014, 16, 45–65. [Google Scholar]
  26. Wang, P.; Wu, J.; Ma, S.; Zhang, L.; Yao, J.; Hoadley, K.A.; Wilkerson, M.D.; Perou, C.M.; Guan, K.L.; Ye, D.; et al. Oncometabolite D-2-Hydroxyglutarate Inhibits ALKBH DNA Repair Enzymes and Sensitizes IDH Mutant Cells to Alkylating Agents. Cell Rep. 2015, 13, 2353–2361. [Google Scholar]
  27. Chen, F.; Bian, K.; Tang, Q.; Fedeles, B.I.; Singh, V.; Humulock, Z.T.; Essigmann, J.M.; Li, D. Oncometabolites d- and l-2-Hydroxyglutarate Inhibit the AlkB Family DNA Repair Enzymes under Physiological Conditions. Chem. Res. Toxicol. 2017, 30, 1102–1110. [Google Scholar]
  28. Sulkowski, P.L.; Corso, C.D.; Robinson, N.D.; Scanlon, S.E.; Purshouse, K.R.; Bai, H.; Liu, Y.; Sundaram, R.K.; Hegan, D.C.; Fons, N.R.; et al. 2-Hydroxyglutarate produced by neomorphic IDH mutations suppresses homologous recombination and induces PARP inhibitor sensitivity. Sci. Transl. Med. 2017, 9, eaal2463. [Google Scholar] [PubMed]
  29. Sulkowski, P.L.; Sundaram, R.K.; Oeck, S.; Corso, C.D.; Liu, Y.; Noorbakhsh, S.; Niger, M.; Boeke, M.; Ueno, D.; Kalathil, A.N. Krebs-cycle-deficient hereditary cancer syndromes are defined by defects in homologous-recombination DNA repair. Nat. Genet. 2018, 50, 1086–1092. [Google Scholar]
  30. Yogev, O.; Yogev, O.; Singer, E.; Shaulian, E.; Goldberg, M.; Fox, T.D.; Pines, O. Fumarase: A mitochondrial metabolic enzyme and a cytosolic/nuclear component of the DNA damage response. PLoS Biol. 2010, 8, e1000328. [Google Scholar]
  31. Jiang, Y.; Qian, X.; Shen, J.; Wang, Y.; Li, X.; Liu, R.; Xia, Y.; Chen, Q.; Peng, G.; Lin, S.Y.; et al. Local generation of fumarate promotes DNA repair through inhibition of histone H3 demethylation. Nat. Cell Biol. 2015, 17, 1158–1168. [Google Scholar]
  32. Leshets, M.; Silas, Y.B.H.; Lehming, N.; Pines, O. Fumarase: From the TCA Cycle to DNA Damage Response and Tumor Suppression. Front. Mol. Biosci. 2018, 5, 68. [Google Scholar]
  33. Leshets, M.; Ramamurthy, D.; Lisby, M.; Lehming, N.; Pines, O. Fumarase is involved in DNA double-strand break resection through a functional interaction with Sae2. Curr. Genet. 2018, 64, 697–712. [Google Scholar] [PubMed]
  34. Dashty, M. A quick look at biochemistry: Carbohydrate metabolism. Clin. Biochem. 2013, 46, 1339–1352. [Google Scholar]
  35. Kornberg, H. Krebs and his trinity of cycles. Nat. Rev. Mol. Cell Biol. 2000, 1, 225–228. [Google Scholar] [PubMed]
  36. Akram, M. Citric Acid Cycle and Role of its Intermediates in Metabolism. Cell Biochem. Biophys. 2014, 68, 475–478. [Google Scholar]
  37. Bénit, P.; Letouzé, E.; Rak, M.; Aubry, L.; Burnichon, N.; Favier, J.; Gimenez-Roqueplo, A.P.; Rustin, P. Unsuspected task for an old team: Succinate, fumarate and other Krebs cycle acids in metabolic remodeling. Biochim. Biophys. Acta 2014, 1837, 1330–1337. [Google Scholar]
  38. Roosterman, D.; Cottrell, G.S. Rethinking the citric acid cycle: Connecting pyruvate carboxylase and citrate synthase to the flow of energy and material. Int. J. Mol. Sci. 2021, 22, 604. [Google Scholar] [CrossRef]
  39. Kranendijk, M.; Struys, E.A.; Salomons, G.S.; Van der Knaap, M.S.; Jakobs, C. Progress in understanding 2-hydroxyglutaric acidurias. J. Inherit. Metab. Dis. 2012, 35, 571–587. [Google Scholar]
  40. Kang, W.; Suzuki, M.; Saito, T.; Miyado, K. Emerging role of TCA cycle-related enzymes in human diseases. Int. J. Mol. Sci. 2021, 22, 13057. [Google Scholar] [CrossRef]
  41. Weinberg, S.E.; Sena, L.A.; Chandel, N.S. Mitochondria in the regulation of innate and adaptive immunity. Immunity 2015, 42, 406–417. [Google Scholar] [PubMed]
  42. Ryan, D.G.; O’Neill, L.A.J. Krebs cycle rewired for macrophage and dendritic cell effector functions. FEBS Lett. 2017, 591, 2992–3006. [Google Scholar] [PubMed]
  43. Diskin, C.; Ryan, T.A.J.; O’Neill, L.A.J. Modification of Proteins by Metabolites in Immunity. Immunity 2021, 54, 19–31. [Google Scholar]
  44. Sánchez-García, F.J.; Pérez-Hernández, C.A.; Rodríguez-Murillo, M.; Moreno-Altamirano, M.M.B. The Role of Tricarboxylic Acid Cycle Metabolites in Viral Infections. Front. Cell. Infect. Microbiol. 2021, 11, 725043. [Google Scholar]
  45. Chakrabarty, R.P.; Chandel, N.S. Mitochondria as Signaling Organelles Control Mammalian Stem Cell Fate. Cell Stem Cell 2021, 28, 394–408. [Google Scholar]
  46. Merkley, E.D.; Metz, T.O.; Smith, R.D.; Baynes, J.W.; Frizzell, N. The succinated proteome. Mass. Spectrom. Rev. 2014, 33, 98–109. [Google Scholar]
  47. De Castro Fonseca, M.; Aguiar, C.J.; Da Rocha Franco, J.A.; Gingold, R.N.; Leite, M.F. GPR91: Expanding the frontiers of Krebs cycle intermediates. Cell Commun. Signal 2016, 14, 3. [Google Scholar] [PubMed]
  48. Liu, S.; He, L.; Yao, K. The Antioxidative Function of Alpha-Ketoglutarate and Its Applications. BioMed Res. Int. 2018, 2018, 3408467. [Google Scholar]
  49. Williams, N.C.; O’Neill, L.A.J. A role for the krebs cycle intermediate citrate in metabolic reprogramming in innate immunity and inflammation. Front. Immunol. 2018, 9, 141. [Google Scholar]
  50. Choudhary, C.; Weinert, B.T.; Nishida, Y.; Verdin, E.; Mann, M. The growing landscape of lysine acetylation links metabolism and cell signalling. Nat. Rev. Mol. Cell Biol. 2014, 15, 536–550. [Google Scholar]
  51. Ren, J.G.; Seth, P.; Ye, H.; Guo, K.; Hanai, J.I.; Husain, Z.; Sukhatme, V.P. Citrate Suppresses Tumor Growth in Multiple Models through Inhibition of Glycolysis, the Tricarboxylic Acid Cycle and the IGF-1R Pathway. Sci. Rep. 2017, 7, 4537. [Google Scholar]
  52. Philippe, I.; Hubert, L. The reduced concentration of citrate in cancer cells: An indicator of cancer aggressiveness and a possible therapeutic target. Drug Resis Updat. 2016, 29, 47–53. [Google Scholar]
  53. Li, Y.; Li, Y.C.; Liu, X.T.; Zhang, L.; Chen, Y.H.; Zhao, Q.; Gao, W.; Liu, B.; Yang, H.; Li, P. Blockage of citrate export prevents TCA cycle fragmentation via Irg1 inactivation. Cell Rep. 2022, 38, 110391. [Google Scholar]
  54. Lampropoulou, V.; Sergushichev, A.; Bambouskova, M.; Nair, S.; Vincent, E.E.; Loginicheva, E.; Cervantes-Barragan, L.; Ma, X.; Huang, S.C.C.; Griss, T.; et al. Itaconate Links Inhibition of Succinate Dehydrogenase with Macrophage Metabolic Remodeling and Regulation of Inflammation. Cell Metabol. 2016, 24, 158–166. [Google Scholar]
  55. Mills, E.L.; Ryan, D.G.; Prag, H.A.; Dikovskaya, D.; Menon, D.; Zaslona, Z.; Jedrychowski, M.P.; Costa, A.S.; Higgins, M.; Hams, E.; et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature 2018, 556, 113–117. [Google Scholar]
  56. Ni, L.; Lin, Z.; Hu, S.; Shi, Y.; Jiang, Z.; Zhao, J.; Zhou, Y.; Wu, Y.; Tian, N.; Sun, L.; et al. Itaconate attenuates osteoarthritis by inhibiting STING/NF-κB axis in chondrocytes and promoting M2 polarization in macrophages. Biochem. Pharmacol. 2022, 198, 114935. [Google Scholar]
  57. Abla, H.; Sollazzo, M.; Gasparre, G.; Iommarini, L.; Porcelli, A.M. The multifaceted contribution of α-ketoglutarate to tumor progression: An opportunity to exploit? Semin. Cell Dev. Biol. 2020, 98, 26–33. [Google Scholar] [PubMed]
  58. Xiao, D.; Zeng, L.; Yao, K.; Kong, X.; Wu, G.; Yin, Y. The glutamine-alpha-ketoglutarate (AKG) metabolism and its nutritional implications. Amino Acids 2016, 48, 2067–2080. [Google Scholar] [PubMed]
  59. Liu, P.S.; Wang, H.; Li, X.; Chao, T.; Teav, T.; Christen, S.; Di Conza, G.; Cheng, W.C.; Chou, C.H.; Vavakova, M.; et al. A-Ketoglutarate Orchestrates Macrophage Activation Through Metabolic and Epigenetic Reprogramming. Nat. Immunol. 2017, 18, 985–994. [Google Scholar]
  60. Morris, J.P.; Yashinskie, J.J.; Koche, R.; Chandwani, R.; Tian, S.; Chen, C.C.; Baslan, T.; Marinkovic, Z.S.; Sánchez-Rivera, F.J.; Leach, S.D.; et al. α-Ketoglutarate links p53 to cell fate during tumour suppression. Nature 2019, 573, 595–599. [Google Scholar] [PubMed]
  61. Shim, E.H.; Livi, C.B.; Rakheja, D.; Tan, J.; Benson, D.; Parekh, V.; Kho, E.Y.; Ghosh, A.P.; Kirkman, R.; Velu, S.; et al. L-2-hydroxyglutarate: An epigenetic modifier and putative oncometabolite in renal cancer. Cancer Discov. 2014, 4, 1290–1298. [Google Scholar]
  62. Gagné, L.M.; Boulay, K.; Topisirovic, I.; Huot, M.É.; Mallette, F.A. Oncogenic Activities of IDH1/2 Mutations: From Epigenetics to Cellular Signaling. Trends Cell Biol. 2017, 27, 738–752. [Google Scholar]
  63. Yong, C.; Stewart, G.D.; Frezza, C. Oncometabolites in renal cancer. Nat. Rev. Nephrol. 2020, 16, 156–172. [Google Scholar]
  64. Du, X.; Hu, H. The Roles of 2-Hydroxyglutarate. Front. Cell Dev. Biol. 2021, 9, 651317. [Google Scholar]
  65. Wahl, D.R.; Venneti, S. 2-Hydoxyglutarate: D/Riving Pathology in gLiomaS. Brain Pathol. 2015, 25, 760–768. [Google Scholar]
  66. Struys, E.A.; Verhoeven, N.M.; Ten Brink, H.J.; Wickenhagen, W.V.; Gibson, K.M.; Jakobs, C. Kinetic characterization of human hydroxyacid-oxoacid transhydrogenase: Relevance to D-2-hydroxyglutaric and gamma-hydroxybutyric acidurias. J. Inherit. Metab. Dis. 2005, 28, 921–930. [Google Scholar] [PubMed]
  67. Ježek, P. 2-Hydroxyglutarate in Cancer Cells. Antioxid. Redox Signal 2020, 33, 903–926. [Google Scholar]
  68. Achouri, Y.; Noël, G.; Vertommen, D.; Rider, M.H.; Veiga-Da-Cunha, M.; Van Schaftingen, E. Identification of a dehydrogenase acting on D-2-hydroxyglutarate. Biochem. J. 2004, 381, 35–42. [Google Scholar] [PubMed]
  69. Toplak, M.; Brunner, J.; Schmidt, J.; Macheroux, P. Biochemical characterization of human D-2-hydroxyglutarate dehydrogenase and two disease related variants reveals the molecular cause of D-2-hydroxyglutaric aciduria. Biochim. Biophys. Acta Proteins Proteom. 2019, 1867, 140255. [Google Scholar]
  70. Rzem, R.; Vincent, M.F.; Van Schaftingen, E.; Veiga-da-Cunha, M. L-2-hydroxyglutaric aciduria, a defect of metabolite repair. J. Inherit. Metab. Dis. 2007, 30, 681–689. [Google Scholar]
  71. Rzem, R.; Veiga-da-Cunha, M.; Noël, G.; Goffette, S.; Nassogne, M.C.; Tabarki, B.; Schöller, C.; Marquardt, T.; Vikkula, M.; Van Schaftingen, E. A gene encoding a putative FAD-dependent L-2-hydroxyglutarate dehydrogenase is mutated in L-2-hydroxyglutaric aciduria. Proc. Natl. Acad. Sci. USA 2004, 101, 16849–16854. [Google Scholar]
  72. Topçu, M.; Jobard, F.; Halliez, S.; Coskun, T.; Yalçinkayal, C.; Gerceker, F.O.; Wanders, R.J.; Prud’homme, J.F.; Lathrop, M.; Özguc, M.; et al. L-2-Hydroxyglutaric aciduria: Identification of a mutant gene C14orf160, localized on chromosome 14q22.1. Hum. Mol. Genet. 2004, 13, 2803–2811. [Google Scholar]
  73. Steenweg, M.E.; Jakobs, C.; Errami, A.; van Dooren, S.J.; Adeva Bartolomé, M.T.; Aerssens, P.; Augoustides-Savvapoulou, P.; Baric, I.; Baumann, M.; Bonafé, L.; et al. An overview of L-2-hydroxyglutarate dehydrogenase gene (L2HGDH) variants: A genotype-phenotype study. Hum. Mutat. 2010, 31, 380–390. [Google Scholar] [PubMed]
  74. Rzem, R.; Achouri, Y.; Marbaix, E.; Schakman, O.; Wiame, E.; Marie, S.; Gailly, P.; Vincent, M.F.; Veiga-da-Cunha, M.; Van Schaftingen, E. A mouse model of L-2-hydroxyglutaric aciduria, a disorder of metabolite repair. PLoS ONE 2015, 10, e0119540. [Google Scholar]
  75. Intlekofer, A.M.; DeMatteo, R.G.; Venneti, S.; Finley, L.W.; Lu, C.; Judkins, A.R.; Rustenburg, A.S.; Grinaway, P.B.; Chodera, J.D.; Cross, J.R.; et al. Hypoxia Induces Production of L-2-Hydroxyglutarate. Cell Metab. 2015, 22, 304–311. [Google Scholar] [PubMed]
  76. Oldham, W.M.; Clish, C.B.; Yang, Y.; Loscalzo, J. Hypoxia-Mediated Increases in L-2-hydroxyglutarate Coordinate the Metabolic Response to Reductive Stress. Cell Metab. 2015, 22, 291–303. [Google Scholar]
  77. Shi, J.; Zuo, H.; Ni, L.; Xia, L.; Zhao, L.; Gong, M.; Nie, D.; Gong, P.; Cui, D.; Shi, W.; et al. An IDH1 mutation inhibits growth of glioma cells via GSH depletion and ROS generation. Neurol. Sci. 2014, 35, 839–845. [Google Scholar]
  78. Böttcher, M.; Renner, K.; Berger, R.; Mentz, K.; Thomas, S.; Cardenas-Conejo, Z.E.; Dettmer, K.; Oefner, P.J.; Mackensen, A.; Kreutz, M.; et al. D-2-hydroxyglutarate interferes with HIF-1α stability skewing T-cell metabolism towards oxidative phosphorylation and impairing Th17 polarization. Oncoimmunology 2018, 7, e1445454. [Google Scholar]
  79. Xu, T.; Stewart, K.M.; Wang, X.; Liu, K.; Xie, M.; Ryu, J.K.; Li, K.; Ma, T.; Wang, H.; Ni, L.; et al. Metabolic control of TH17 and induced Treg cell balance by an epigenetic mechanism. Nature 2017, 548, 228–233. [Google Scholar]
  80. Tannahill, G.M.; Curtis, A.M.; Adamik, J.; Palsson-McDermott, E.M.; McGettrick, A.F.; Goel, G.; Frezza, C.; Bernard, N.J.; Kelly, B.; Foley, N.H.; et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature 2013, 496, 238–242. [Google Scholar]
  81. Smestad, J.; Erber, L.; Chen, Y.; Maher, L.J. 3rd. Chromatin Succinylation Correlates with Active Gene Expression and Is Perturbed by Defective TCA Cycle Metabolism. iScience 2018, 2, 63–75. [Google Scholar]
  82. Toma, I.; Kang, J.J.; Sipos, A.; Vargas, S.; Bansal, E.; Hanner, F.; Meer, E.; Peti-Peterdi, J. Succinate receptor GPR91 provides a direct link between high glucose levels and rennin release in murine and rabbit kidney. J. Clin. Investig. 2008, 118, 2526–2534. [Google Scholar]
  83. Mu, X.; Zhao, T.; Xu, C.; Shi, W.; Geng, B.; Shen, J.; Zhang, C.; Pan, J.; Yang, J.; Hu, S.; et al. Oncometabolite succinate promotes angiogenesis by upregulating VEGF expression through GPR91-mediated STAT3 and ERK activation. Oncotarget 2017, 8, 13174–13185. [Google Scholar]
  84. Yang, M.; Soga, T.; Pollard, P.J.; Adam, J. The emerging role of fumarate as an oncometabolite. Front. Oncol. 2012, 2, 85. [Google Scholar]
  85. Zyla, R.E.; Hodgson, A. Gene of the month: FH. J. Clin. Pathol. 2021, 74, 615–619. [Google Scholar]
  86. Sullivan, L.B.; Martinez-Garcia, E.; Nguyen, H.; Mullen, A.R.; Dufour, E.; Sudarshan, S.; Licht, J.D.; Deberardinis, R.J.; Chandel, N.S. The Proto-oncometabolite Fumarate Binds Glutathione to Amplify ROS-dependent signaling. Mol. Cell 2013, 51, 236–248. [Google Scholar]
  87. Tyrakis, P.A.; Yurkovich, M.E.; Sciacovelli, M.; Papachristou, E.K.; Bridges, H.R.; Gaude, E.; Schreiner, A.; D’Santos, C.; Hirst, J.; Hernandez-Fernaud, J.; et al. Fumarate Hydratase Loss Causes Combined Respiratory Chain Defects. Cell Rep. 2017, 21, 1036–1047. [Google Scholar]
  88. Lunt, S.Y.; Vander Heiden, M.G. Aerobic glycolysis: Meeting the metabolic requirements of cell proliferation. Annu. Rev. Cell Dev. Biol. 2011, 27, 441–464. [Google Scholar]
  89. Altman, B.J.; Stine, Z.E.; Dang, C.V. From Krebs to clinic: Glutamine metabolism to cancer therapy. Nat. Rev. Cancer 2016, 16, 619–634. [Google Scholar]
  90. Sajnani, K.; Islam, F.; Smith, R.A.; Gopalan, V.; Lam, A.K. Genetic alterations in Krebs cycle and its impact on cancer pathogenesis. Biochimie 2017, 135, 164–172. [Google Scholar]
  91. Farhadi, P.; Yarani, R.; Dokaneheifard, S.; Mansouri, K. The emerging role of targeting cancer metabolism for cancer therapy. Tumor Biol. 2020, 42, 1010428320965284. [Google Scholar]
  92. Gasmi, A.; Peana, M.; Arshad, M.; Butnariu, M.; Menzel, A.; Bjørklund, G. Krebs cycle: Activators, inhibitors and their roles in the modulation of carcinogenesis. Arch. Toxicol. 2021, 95, 1161–1178. [Google Scholar]
  93. Liu, Y.; Yang, C. Oncometabolites in cancer: Current understanding and challenges. Cancer Res. 2021, 81, 2820–2823. [Google Scholar] [PubMed]
  94. McDonough, M.A.; Loenarz, C.; Chowdhury, R.; Clifton, I.J.; Schofield, C.J. Structural studies on human 2-oxoglutarate dependent oxygenases. Curr. Opin. Struct. Biol. 2010, 20, 659–672. [Google Scholar] [PubMed]
  95. Loenarz, C.; Schofield, C.J. Physiological and biochemical aspects of hydroxylations and demethylations catalyzed by human 2-oxoglutarate oxygenases. Trends Biochem. Sci. 2011, 36, 7–18. [Google Scholar]
  96. Jokilehto, T.; Jaakkola, P.M. The role of HIF prolyl hydroxylases in tumour growth. J. Cell. Mol. Med. 2010, 14, 758–770. [Google Scholar] [PubMed]
  97. Iyer, L.M.; Tahiliani, M.; Rao, A.; Aravind, L. Prediction of novel families of enzymes involved in oxidative and other complex modifications of bases in nucleic acids. Cell Cycle 2009, 8, 1698–1710. [Google Scholar] [PubMed]
  98. Tsukada, Y.I.; Fang, J.; Erdjument-Bromage, H.; Warren, M.E.; Borchers, C.H.; Tempst, P.; Zhang, Y. Histone demethylation by a family of JmjC domain-containing proteins. Nature 2006, 439, 811–816. [Google Scholar] [PubMed]
  99. Aravind, L.; Koonin, E.V. The DNA-repair protein AlkB, EGL-9, and leprecan define new families of 2-oxoglutarate- and iron-dependent dioxygenases. Genome Biol. 2001, 2, research0007. [Google Scholar]
  100. Trewick, S.C.; Henshaw, T.F.; Hausinger, R.P.; Lindahl, T.; Sedgwick, B. Oxidative demethylation by Escherichia coli AlkB directly reverts DNA base damage. Nature 2002, 419, 174–178. [Google Scholar]
  101. Falnes, P.Ø.; Johansen, R.F.; Seeberg, E. AlkB-mediated oxidative demethylation reverses DNA damage in Escherichia coli. Nature 2000, 419, 178–182. [Google Scholar]
  102. Brahimi-Horn, C.; Mazure, N.; Pouysségur, J. Signalling via the hypoxia-inducible factor-1α requires multiple posttranslational modifications. Cell Signal 2005, 17, 1–9. [Google Scholar]
  103. Balamurugan, K. HIF-1 at the crossroads of hypoxia, inflammation, and cancer. Int. J. Cancer 2016, 138, 1058–1066. [Google Scholar]
  104. Pan, Z.; Ma, G.; Kong, L.; Du, G. Hypoxia-inducible factor-1: Regulatory mechanisms and drug development in stroke. Pharmacol. Res. 2021, 170, 105742. [Google Scholar] [PubMed]
  105. Eales, K.L.; Hollinshead, K.E.R.; Tennant, D.A. Hypoxia and metabolic adaptation of cancer cells. Oncogenesis 2016, 5, e190. [Google Scholar] [PubMed]
  106. Ke, Q.; Costa, M. Hypoxia-inducible factor-1 (HIF-1). Mol. Pharmacol. 2006, 70, 1469–1480. [Google Scholar]
  107. Yang, M.; Su, H.; Soga, T.; Kranc, K.R.; Pollard, P.J. Prolyl hydroxylase domain enzymes: Important regulators of cancer metabolism. Hypoxia 2014, 2, 27–142. [Google Scholar]
  108. Tahiliani, M.; Koh, K.P.; Shen, Y.; Pastor, W.A.; Bandukwala, H.; Brudno, Y.; Agarwal, S.; Iyer, L.M.; Liu, D.R.; Aravind, L.; et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 2009, 324, 930–935. [Google Scholar]
  109. Tan, L.; Shi, Y.G. Tet family proteins and 5-hydroxymethylcytosine in development and disease. Development 2012, 139, 1895–1902. [Google Scholar]
  110. Rasmussen, K.D.; Helin, K. Role of TET enzymes in DNA methylation, development, and cancer. Genes. Dev. 2016, 30, 733–750. [Google Scholar]
  111. Williams, K.; Christensen, J.; Helin, K. DNA methylation: TET proteins-guardians of CpG islands? EMBO Rep. 2012, 13, 28–35. [Google Scholar]
  112. Yang, J.; Bashkenova, N.; Zang, R.; Huang, X.; Wang, J. The roles of TET family proteins in development and stem cells. Development 2020, 147, dev183129. [Google Scholar]
  113. Labbé, R.M.; Holowatyj, A.; Yang, Z.Q. Histone lysine demethylase (kdm) subfamily 4: Structures, functions and therapeutic potential. Am. J. Transl. Res. 2014, 6, 1–15. [Google Scholar]
  114. Staehle, H.F.; Pahl, H.L.; Jutzi, J.S. The cross marks the spot: The emerging role of JmjC domain-containing proteins in myeloid malignancies. Biomolecules 2021, 11, 1911. [Google Scholar] [CrossRef]
  115. Sui, Y.; Gu, R.; Janknecht, R. Crucial functions of the JMJD1/KDM3 epigenetic regulators in cancer. Mol. Cancer Res. 2021, 19, 3–13. [Google Scholar]
  116. Sedgwick, B.; Bates, P.A.; Paik, J.; Jacobs, S.C.; Lindahl, T. Repair of alkylated DNA: Recent advances. DNA Repair 2007, 6, 429–442. [Google Scholar] [PubMed]
  117. Fedeles, B.I.; Singh, V.; Delaney, J.C.; Li, D.; Essigmann, J.M. The AlkB Family of Fe(II)/α-Ketoglutarate-dependent Dioxygenases: Repairing Nucleic Acid Alkylation Damage and Beyond. J. Biol. Chem. 2015, 290, 20734–20742. [Google Scholar] [PubMed]
  118. Alemu, E.A.; He, C.; Klungland, A. ALKBHs-facilitated RNA modifications and de-modifications. DNA Repair 2016, 44, 87–91. [Google Scholar]
  119. Schvartzman, J.M.; Thompson, C.B.; Finley, L.W.S. Metabolic regulation of chromatin modifications and gene expression. J. Cell Biol. 2018, 217, 2247–2259. [Google Scholar]
  120. Friedberg, E.C.; Walker, G.C.; Siede, W.; Wood, R.D.; Schultz, R.A.; Ellenberger, T. DNA Repair and Mutagenesis, 2nd ed.; ASM Press: Washington, DC, USA, 2006. [Google Scholar]
  121. Chatterjee, N.; Walker, G.C. Mechanisms of DNA damage, repair, and mutagenesis. Environ. Mol. Mutagen. 2017, 58, 235–263. [Google Scholar]
  122. Yang, M.; Pollard, P.J. Succinate: A new epigenetic hacker. Cancer Cell 2013, 23, 709–711. [Google Scholar]
  123. Parsons, D.W.; Jones, S.; Zhang, X.; Lin, J.C.H.; Leary, R.J.; Angenendt, P.; Mankoo, P.; Carter, H.; Siu, I.M.; Gallia, G.L.; et al. An integrated genomic analysis of human glioblastoma multiforme. Science 2008, 321, 1807–1812. [Google Scholar]
  124. Yan, H.; Parsons, D.W.; Jin, G.; McLendon, R.; Rasheed, B.A.; Yuan, W.; Kos, I.; Batinic-Haberle, I.; Jones, S.; Riggins, G.J.; et al. IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med. 2009, 360, 765–773. [Google Scholar]
  125. Ward, P.S.; Patel, J.; Wise, D.R.; Abdel-Wahab, O.; Bennett, B.D.; Coller, H.A.; Cross, J.R.; Fantin, V.R.; Hedvat, C.V.; Perl, A.E.; et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 2010, 17, 225–234. [Google Scholar]
  126. Frezza, C.; Pollard, P.J.; Gottlieb, E. Inborn and acquired metabolic defects in cancer. J. Mol. Med. 2011, 89, 213–220. [Google Scholar] [PubMed]
  127. Zhao, T.; Mu, X.; You, Q. Succinate: An initiator in tumorigenesis and progression. Oncotarget 2017, 8, 53819–53828. [Google Scholar]
  128. Jiang, S.; Yan, W. Succinate in the cancer–immune cycle. Cancer Lett. 2017, 390, 45–47. [Google Scholar] [PubMed]
  129. Zhao, Y.; Feng, F.; Guo, Q.H.; Wang, Y.P.; Zhao, R. Role of succinate dehydrogenase deficiency and oncometabolites in gastrointestinal stromal tumors. World J. Gastroenterol. 2020, 26, 5074–5089. [Google Scholar]
  130. Baysal, B.E.; Ferrell, R.E.; Willett-Brozick, J.E.; Lawrence, E.C.; Myssiorek, D.; Bosch, A.; Mey, A.V.D.; Taschner, P.E.; Rubinstein, W.S.; Myers, E.N.; et al. Mutations in SDHD, a mitochondrial complex II gene, in hereditary paraganglioma. Science 2000, 287, 848–851. [Google Scholar] [PubMed]
  131. Barletta, J.A.; Hornick, J.L. Succinate dehydrogenase-deficient tumors: Diagnostic advances and clinical implications. Adv. Anat. Pathol. 2012, 19, 193–203. [Google Scholar]
  132. Gill, A.J. Succinate dehydrogenase (SDH)-deficient neoplasia. Histopathology 2018, 72, 106–116. [Google Scholar]
  133. Buffet, A.; Burnichon, N.; Favier, J.; Gimenez-Roqueplo, A.P. An overview of 20 years of genetic studies in pheochromocytoma and paraganglioma. Best Pract. Res. Clin. Endocrinol. Metab. 2020, 34, 101416. [Google Scholar]
  134. Dalla Pozza, E.; Dando, I.; Pacchiana, R.; Liboi, E.; Scupoli, M.T.; Donadelli, M.; Palmieri, M. Regulation of succinate dehydrogenase and role of succinate in cancer. Semin. Cell Dev. Biol. 2020, 98, 4–14. [Google Scholar] [PubMed]
  135. Killian, J.K.; Kim, S.Y.; Miettinen, M.; Smith, C.; Merino, M.; Tsokos, M.; Quezado, M.; Smith Jr, W.I.; Jahromi, M.S.; Xekouki, P.; et al. Succinate dehydrogenase mutation underlies global epigenomic divergence in gastrointestinal stromal tumor. Cancer Discov. 2013, 3, 648–657. [Google Scholar]
  136. Flavahan, W.A.; Drier, Y.; Johnstone, S.E.; Hemming, M.L.; Tarjan, D.R.; Hegazi, E.; Shareef, S.J.; Javed, N.M.; Raut, C.P.; Eschle, B.K.; et al. Altered chromosomal topology drives oncogenic programs in SDH-deficient GISTs. Nature 2019, 575, 229–233. [Google Scholar]
  137. Janeway, K.A.; Kim, S.Y.; Lodish, M.; Nosé, V.; Rustin, P.; Gaal, J.; Dahia, P.L.; Liegl, B.; Ball, E.R.; Raygada, M.; et al. Defects in succinate dehydrogenase in gastrointestinal stromal tumors lacking KIT and PDGFRA mutations. Proc. Natl. Acad. Sci. USA 2011, 108, 314–318. [Google Scholar]
  138. Ibrahim, A.; Chopra, S. Succinate dehydrogenase-deficient gastrointestinal stromal tumors. Arch. Pathol. Lab. Med. 2020, 144, 655–660. [Google Scholar]
  139. Blay, J.Y.; Kang, Y.K.; Nishida, T.; von Mehren, M. Gastrointestinal stromal tumours. Nat. Rev. Dis. Primers 2021, 7, 22. [Google Scholar]
  140. Aghamir, S.M.K.; Heshmat, R.; Ebrahimi, M.; Ketabchi, S.E.; Parichehreh Dizaji, S.; Khatami, F. The impact of succinate dehydrogenase gene (SDH) mutations in renal cell carcinoma (RCC): A systematic review. Onco Targets Ther. 2019, 12, 7929–7940. [Google Scholar] [PubMed]
  141. Kamai, T.; Higashi, S.; Murakami, S.; Arai, K.; Namatame, T.; Kijima, T.; Abe, H.; Jamiyan, T.; Ishida, K.; Shirataki, H.; et al. Single nucleotide variants of succinate dehydrogenase A gene in renal cell carcinoma. Cancer Sci. 2021, 112, 3375–3387. [Google Scholar]
  142. Xekouki, P.; Pacak, K.; Almeida, M.; Wassif, C.A.; Rustin, P.; Nesterova, M.; de la Luz Sierra, M.; Matro, J.; Ball, E.; Azevedo, M.; et al. Succinate dehydrogenase (SDH) D subunit (SDHD) inactivation in a growth-hormone-producing pituitary tumor: A new association for SDH? J. Clin. Endocrinol. Metab. 2012, 97, 357–366. [Google Scholar]
  143. Gill, A.J.; Toon, C.W.; Clarkson, A.; Sioson, L.; Chou, A.; Winship, I.; Robinson, B.G.; Benn, D.E.; Clifton-Bligh, R.J.; Dwight, T. Succinate dehydrogenase deficiency is rare in pituitary adenomas. Am. J. Surg. Pathol. 2014, 38, 560–566. [Google Scholar]
  144. Chen, L.; Liu, T.; Zhang, S.; Zhou, J.; Wang, Y.; Di, W. Succinate dehydrogenase subunit B inhibits the AMPK-HIF-1α pathway in human ovarian cancer in vitro. J. Ovarian Res. 2014, 7, 115. [Google Scholar]
  145. Selak, M.A.; Armour, S.M.; MacKenzie, E.D.; Boulahbel, H.; Watson, D.G.; Mansfield, K.D.; Pan, Y.; Simon, M.C.; Thompson, C.B.; Gottlieb, E. Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-α prolyl hydroxylase. Cancer Cell 2005, 7, 77–85. [Google Scholar]
  146. Frezza, C.; Zheng, L.; Folger, O.; Rajagopalan, K.N.; MacKenzie, E.D.; Jerby, L.; Micaroni, M.; Chaneton, B.; Adam, J.; Hedley, A.; et al. Haem oxygenase is synthetically lethal with the tumour suppressor fumarate hydratase. Nature 2011, 477, 225–228. [Google Scholar] [PubMed]
  147. Sciacovelli, M.; Gonçalves, E.; Johnson, T.I.; Zecchini, V.R.; Da Costa, A.S.H.; Gaude, E.; Drubbel, A.V.; Theobald, S.J.; Abbo, S.R.; Tran, M.G.B.; et al. Fumarate is an epigenetic modifier that elicits epithelial-to-mesenchymal transition. Nature 2016, 537, 544–547. [Google Scholar]
  148. Tomlinson, I.P.M.; Alam, N.A.; Rowan, A.J.; Barclay, E.; Jaeger, E.E.; Kelsell, D.; Leigh, I.; Gorman, P.; Lamlum, H.; Rahman, S.; et al. Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer the multiple leiomyoma consortium. Nat. Genet. 2002, 30, 406–410. [Google Scholar]
  149. Lehtonen, H.J.; Kiuru, M.; Ylisaukko-Oja, S.K.; Salovaara, R.; Herva, R.; Koivisto, P.A.; Vierimaa, O.; Aittomäki, K.; Pukkala, E.; Launonen, V.; et al. Increased risk of cancer in patients with fumarate hydratase germline mutation. J. Med. Genet. 2006, 43, 523–526. [Google Scholar]
  150. Bayley, J.P.; Launonen, V.; Tomlinson, I.P.M. The FH mutation database: An online database of fumarate hydratase mutations involved in the MCUL (HLRCC) tumor syndrome and congenital fumarase deficiency. BMC Med. Genet. 2008, 9, 20. [Google Scholar]
  151. Schmidt, L.S.; Linehan, W.M. Hereditary leiomyomatosis and renal cell carcinoma. Int. J. Nephrol. Renov. Dis. 2014, 7, 253–260. [Google Scholar]
  152. Crooks, D.R.; Maio, N.; Lang, M.; Ricketts, C.J.; Vocke, C.D.; Gurram, S.; Turan, S.; Kim, Y.Y.; Cawthon, G.M.; Sohelian, F.; et al. Mitochondrial DNA alterations underlie an irreversible shift to aerobic glycolysis in fumarate hydratase-deficient renal cancer. Sci. Signal. 2021, 14, eabc4436. [Google Scholar]
  153. Gleeson, J.P.; Nikolovski, I.; Dinatale, R.; Zucker, M.; Knezevic, A.; Patil, S.; Ged, Y.; Kotecha, R.R.; Shapnik, N.; Murray, S.; et al. Comprehensive Molecular Characterization and Response to Therapy in Fumarate Hydratase-Deficient Renal Cell Carcinoma. Clin. Cancer Res. 2021, 27, 2910–2919. [Google Scholar] [PubMed]
  154. Sun, G.; Zhang, X.; Liang, J.; Pan, X.; Zhu, S.; Liu, Z.; Armstrong, C.M.; Chen, J.; Lin, W.; Liao, B.; et al. Integrated molecular characterization of fumarate hydratase deficient renal cell carcinoma. Clin. Cancer Res. 2021, 27, 1734–1743. [Google Scholar] [PubMed]
  155. Zhang, Q.; Poropatich, K.; Ubago, J.; Xie, J.; Xu, X.; Frizzell, N.; Kim, J.; Kong, B.; Wei, J.J. Fumarate Hydratase Mutations and Alterations in Leiomyoma with Bizarre Nuclei. Int. J. Gynecol. Pathol. 2018, 37, 421–430. [Google Scholar] [PubMed]
  156. Castro-Vega, L.J.; Buffet, A.; De Cubas, A.A.; Cascón, A.; Menara, M.; Khalifa, E.; Amar, L.; Azriel, S.; Bourdeau, I.; Chabre, O.; et al. Germline mutations in FH confer predisposition to malignant pheochromocytomas and paragangliomas. Hum. Mol. Genet. 2014, 23, 2440–2446. [Google Scholar]
  157. Clark, G.R.; Sciacovelli, M.; Gaude, E.; Walsh, D.M.; Kirby, G.; Simpson, M.A.; Trembath, R.C.; Berg, J.N.; Woodward, E.R.; Kinning, E.; et al. Germline FH mutations presenting with pheochromocytoma. J. Clin. Endocrinol. Metab. 2014, 99, E2046–E2050. [Google Scholar] [PubMed]
  158. Schmidt, C.; Sciacovelli, M.; Frezza, C. Fumarate hydratase in cancer: A multifaceted tumour suppressor. Semin. Cell Dev. Biol. 2020, 98, 15–25. [Google Scholar]
  159. Li, S.; Qiao, C.; Yang, L.; Hong, M.; Fang, Y.; Jin, H.; Li, J.; Qian, S. Fumarate hydratase deficiency induces chronic myeloid leukemia progression. Transl. Cancer Res. 2019, 8, 592–602. [Google Scholar] [PubMed]
  160. Wang, S.; Ramamurthy, D.; Tan, J.; Liu, J.; Yip, J.; Chua, A.; Yu, Z.; Lim, T.K.; Lin, Q.; Pines, O.; et al. Post-translational Modifications of Fumarase Regulate its Enzyme Activity and Function in Respiration and the DNA Damage Response. J. Mol. Biol. 2020, 432, 6108–6126. [Google Scholar] [PubMed]
  161. Chowdhury, R.; Yeoh, K.K.; Tian, Y.M.; Hillringhaus, L.; Bagg, E.A.; Rose, N.R.; Leung, I.K.; Li, X.S.; Woon, E.C.; Yang, M.; et al. The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep. 2011, 12, 463–469. [Google Scholar] [PubMed]
  162. Xu, W.; Yang, H.; Liu, Y.; Yang, Y.; Wang, P.; Kim, S.H.; Ito, S.; Yang, C.; Wang, P.; Xiao, M.T.; et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell 2011, 19, 17–30. [Google Scholar] [PubMed]
  163. Isaacs, J.S.; Jung, Y.J.; Mole, D.R.; Lee, S.; Torres-Cabala, C.; Chung, Y.L.; Merino, M.; Trepel, J.; Zbar, B.; Toro, J.; et al. HIF overexpression correlates with biallelic loss of fumarate hydratase in renal cancer: Novel role of fumarate in regulation of HIF stability. Cancer Cell 2005, 8, 143–153. [Google Scholar]
  164. O’Flaherty, L.; Adam, J.; Heather, L.C.; Zhdanov, A.V.; Chung, Y.L.; Miranda, M.X.; Croft, J.; Olpin, S.; Clarke, K.; Pugh, C.W.; et al. Dysregulation of hypoxia pathways in fumarate hydratase-deficient cells is independent of defective mitochondrial metabolism. Hum. Mol. Genet. 2010, 19, 3844–3851. [Google Scholar]
  165. Yang, M.; Soga, T.; Pollard, P.J. Oncometabolites: Linking altered metabolism with cancer. J. Clin. Investig. 2013, 123, 3652–3658. [Google Scholar]
  166. Koyasu, S.; Kobayashi, M.; Goto, Y.; Hiraoka, M.; Harada, H. Regulatory mechanisms of hypoxia-inducible factor 1 activity: Two decades of knowledge. Cancer Sci. 2018, 109, 560–571. [Google Scholar]
  167. Laukka, T.; Mariani, C.J.; Ihantola, T.; Cao, J.Z.; Hokkanen, J.; Kaelin, W.G.; Godley, L.A.; Koivunen, P. Fumarate and succinate regulate expression of hypoxia-inducible genes via TET enzymes. J. Biol. Chem. 2016, 291, 4256–4265. [Google Scholar]
  168. Wang, Y.P.; Li, J.T.; Qu, J.; Yin, M.; Lei, Q.Y. Metabolite sensing and signaling in cancer. J. Biol. Chem. 2020, 295, 11938–11946. [Google Scholar] [PubMed]
  169. Young, L.C.; McDonald, D.W.; Hendzel, M.J. Kdm4b histone demethylase is a DNA damage response protein and confers a survival advantage following γ-irradiation. J. Biol. Chem. 2013, 288, 21376–21388. [Google Scholar]
  170. Shmakova, A.; Batie, M.; Druker, J.; Rocha, S. Chromatin and oxygen sensing in the context of JmjC histone demethylases. Biochem. J. 2014, 462, 385–395. [Google Scholar]
  171. Sciacovelli, M.; Frezza, C. Metabolic reprogramming and epithelial-to-mesenchymal transition in cancer. FEBS J. 2017, 284, 3132–3144. [Google Scholar]
  172. Sciacovelli, M.; Frezza, C. Fumarate drives EMT in renal cancer. Cell Death Differ. 2017, 24, 1–2. [Google Scholar]
  173. Røsland, G.V.; Dyrstad, S.E.; Tusubira, D.; Helwa, R.; Tan, T.Z.; Lotsberg, M.L.; Pettersen, I.K.; Berg, A.; Kindt, C.; Hoel, F.; et al. Epithelial to mesenchymal transition (EMT) is associated with attenuation of succinate dehydrogenase (SDH) in breast cancer through reduced expression of SDHC. Cancer Metab. 2019, 7, 6. [Google Scholar]
  174. Friedrich, M.; Hahn, M.; Michel, J.; Sankowski, R.; Kilian, M.; Kehl, N.; Günter, M.; Bunse, T.; Pusch, S.; von Deimling, A.; et al. Dysfunctional dendritic cells limit antigen-specific T cell response in glioma. Neuro-Oncology 2023, 25, 263–276. [Google Scholar] [PubMed]
  175. Jeridi, A.; Kapellos, T.S.; Yildirim, A.Ö. Fumarate hydratase: A new checkpoint of metabolic regulation in inflammatory macrophages. Signal Transduct. Target. Ther. 2023, 8, 332. [Google Scholar]
  176. Arts, R.J.; Novakovic, B.; Ter Horst, R.; Carvalho, A.; Bekkering, S.; Lachmandas, E.; Rodrigues, F.; Silvestre, R.; Cheng, S.C.; Wang, S.Y.; et al. Glutaminolysis and Fumarate Accumulation Integrate Immunometabolic and Epigenetic Programs in Trained Immunity. Cell Metab. 2016, 24, 807–819. [Google Scholar]
  177. Jackson, S.P.; Bartek, J. The DNA-damage response in human biology and disease. Nature 2009, 461, 1071–1078. [Google Scholar] [PubMed]
  178. Ciccia, A.; Elledge, S.J. The DNA Damage Response: Making It Safe to Play with Knives. Mol. Cell 2010, 40, 179–204. [Google Scholar] [PubMed]
  179. Laurini, E.; Marson, D.; Fermeglia, A.; Aulic, S.; Fermeglia, M.; Pricl, S. Role of Rad51 and DNA repair in cancer: A molecular perspective. Pharmacol. Ther. 2020, 208, 107492. [Google Scholar] [PubMed]
  180. Carusillo, A.; Mussolino, C. DNA Damage: From Threat to Treatment. Cells 2020, 9, 1665. [Google Scholar] [CrossRef]
  181. Pilié, P.G.; Tang, C.; Mills, G.B.; Yap, T.A. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat. Rev. Clin. Oncol. 2019, 16, 81–104. [Google Scholar]
  182. Mognato, M.; Burdak-Rothkamm, S.; Rothkamm, K. Interplay between DNA replication stress, chromatin dynamics and DNA-damage response for the maintenance of genome stability. Mutat. Res. 2021, 787, 108346. [Google Scholar]
  183. Shimizu, I.; Yoshida, Y.; Suda, M.; Minamino, T. DNA damage response and metabolic disease. Cell Metab. 2014, 20, 967–977. [Google Scholar]
  184. Mallette, F.A.; Mattiroli, F.; Cui, G.; Young, L.C.; Hendzel, M.J.; Mer, G.; Sixma, T.K.; Richard, S. RNF8- and RNF168-dependent degradation of KDM4A/JMJD2A triggers 53BP1 recruitment to DNA damage sites. EMBO J. 2012, 31, 1865–1878. [Google Scholar]
  185. Ceccaldi, R.; Rondinelli, B.; D’Andrea, A.D. Repair Pathway Choices and Consequences at the Double-Strand Break. Trends Cell Biol. 2016, 26, 52–64. [Google Scholar]
  186. Rossetto, D.; Truman, A.W.; Kron, S.J.; Côté, J. Epigenetic modifications in double-strand break DNA damage signaling and repair. Clin. Cancer Res. 2010, 16, 4543–4552. [Google Scholar]
  187. Schipler, A.; Iliakis, G. DNA double-strand-break complexity levels and their possible contributions to the probability for error-prone processing and repair pathway choice. Nucleic Acids Res. 2013, 41, 7589–7605. [Google Scholar]
  188. Symington, L.S.; Gautier, J. Double-strand break end resection and repair pathway choice. Annu. Rev. Genet. 2011, 45, 247–271. [Google Scholar]
  189. Mladenov, E.; Magin, S.; Soni, A.; Iliakis, G. DNA double-strand break repair as determinant of cellular radiosensitivity to killing and target in radiation therapy. Front. Oncol. 2013, 3, 113. [Google Scholar]
  190. Wright, W.D.; Shah, S.S.; Heyer, W.D. Homologous recombination and the repair of DNA double-strand breaks. J. Biol. Chem. 2018, 293, 10524–10535. [Google Scholar] [PubMed]
  191. Scully, R.; Panday, A.; Elango, R.; Willis, N.A. DNA double-strand break repair-pathway choice in somatic mammalian cells. Nat. Rev. Mol. Cell Biol. 2019, 20, 698–714. [Google Scholar]
  192. Shibata, A.; Jeggo, P.A. Roles for 53BP1 in the repair of radiation-induced DNA double strand breaks. DNA Repair 2020, 93, 102915. [Google Scholar] [PubMed]
  193. Cejka, P.; Symington, L.S. DNA End Resection: Mechanism and Control. Annu. Rev. Genet. 2021, 55, 285–307. [Google Scholar] [PubMed]
  194. Zheng, L.; Cardaci, S.; Jerby, L.; MacKenzie, E.D.; Sciacovelli, M.; Johnson, T.I.; Gaude, E.; King, A.; Leach, J.D.; Edrada-Ebel, R.; et al. Fumarate induces redox-dependent senescence by modifying glutathione metabolism. Nat. Commun. 2015, 6, 6001. [Google Scholar]
  195. Lombard, D.B.; Chua, K.F.; Mostoslavsky, R.; Franco, S.; Gostissa, M.; Alt, F.W. DNA repair, genome stability, and aging. Cell 2005, 120, 497–512. [Google Scholar]
  196. Jongen, J.M.J.; van der Waals, L.M.; Trumpi, K.; Laoukili, J.; Peters, N.A.; Schenning-van Schelven, S.J.; Govaert, K.M.; Rinkes, I.H.B.; Kranenburg, O. Downregulation of DNA repair proteins and increased DNA damage in hypoxic colon cancer cells is a therapeutically exploitable vulnerability. Oncotarget 2017, 8, 86296–86311. [Google Scholar]
  197. Scanlon, S.E.; Glazer, P.M. Multifaceted control of DNA repair pathways by the hypoxic tumor microenvironment. DNA Repair 2015, 32, 180–189. [Google Scholar]
  198. Kaplan, A.R.; Glazer, P.M. Impact of hypoxia on DNA repair and genome integrity. Mutagenesis 2020, 35, 61–68. [Google Scholar] [PubMed]
  199. Silas, Y.; Singer, E.; Das, K.; Lehming, N.; Pines, O. A combination of Class-I fumarases and metabolites (α-ketoglutarate and fumarate) signal the DNA damage response in Escherichia coli. Proc. Natl. Acad. Sci. USA 2021, 118, 2021–2022. [Google Scholar]
  200. Kaina, B.; Christmann, M.; Naumann, S.; Roos, W.P. MGMT: Key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents. DNA Repair 2007, 6, 1079–1099. [Google Scholar]
  201. Riemenschneider, M.J.; Hegi, M.E.; Reifenberger, G. MGMT promoter methylation in malignant gliomas. Target. Oncol. 2010, 5, 161–165. [Google Scholar]
  202. Ricci, R.; Martini, M.; Ravegnini, G.; Cenci, T.; Milione, M.; Lanza, P.; Pierconti, F.; Santini, D.; Angelini, S.; Biondi, A.; et al. Preferential MGMT methylation could predispose a subset of KIT/PDGFRA-WT GISTs, including SDH-deficient ones, to respond to alkylating agents. Clin. Epigenetics 2019, 11, 2. [Google Scholar] [PubMed]
  203. Dinglay, S.; Trewick, S.C.; Lindahl, T.; Sedgwick, B. Defective processing of methylated single-stranded DNA by E. coli AlkB Mutants. Genes. Dev. 2000, 14, 2097–2105. [Google Scholar] [PubMed]
  204. Gerson, S.L. MGMT: Its role in cancer aetiology and cancer therapeutics. Nat. Rev. Cancer 2004, 4, 296–307. [Google Scholar] [PubMed]
  205. Mielecki, D.; Grzesiuk, E. Ada response—A strategy for repair of alkylated DNA in bacteria. FEMS Microbiol. Lett. 2014, 355, 1–11. [Google Scholar] [PubMed]
  206. Wang, P.; Wang, Y. Cytotoxic and mutagenic properties of O6-alkyl-2′-deoxyguanosine lesions in Escherichia coli cells. J. Biol. Chem. 2018, 293, 15033–15042. [Google Scholar]
  207. Margison, G.P.; Santibáñez-Koref, M.F. O6-alkylguanine-DNA alkyltransferase: Role in carcinogenesis and chemotherapy. Bioessays 2002, 24, 255–266. [Google Scholar] [PubMed]
  208. Puyo, S.; Montaudon, D.; Pourquier, P. From old alkylating agents to new minor groove binders. Crit. Rev. Oncol. Hematol. 2014, 89, 43–61. [Google Scholar] [PubMed]
  209. Sharma, S.; Salehi, F.; Scheithauer, B.W.; Rotondo, F.; Syro, L.V.; Kovacs, K. Role of MGMT in tumor development, progression, diagnosis, treatment and prognosis. Anticancer. Res. 2009, 29, 3759–3768. [Google Scholar]
  210. Haque, W.; Thong, E.; Andrabi, S.; Verma, V.; Brian Butler, E.; Teh, B.S. Prognostic and predictive impact of MGMT promoter methylation in grade 3 gliomas. J. Clin. Neurosci. 2021, 85, 115–121. [Google Scholar]
  211. Smits, A.; Lysiak, M.; Magnusson, A.; Rosell, J.; Söderkvist, P.; Malmström, A. Sex Disparities in MGMT Promoter Methylation and Survival in Glioblastoma: Further Evidence from Clinical Cohorts. J. Clin. Med. 2021, 10, 556. [Google Scholar] [CrossRef] [PubMed]
  212. Horbinski, C.; McCortney, K.; Stupp, R. MGMT promoter methylation is associated with patient age and 1p/19q status in IDH-mutant gliomas. Neuro Oncol. 2021, 23, 858–860. [Google Scholar] [PubMed]
  213. Butler, M.; Pongor, L.; Su, Y.T.; Xi, L.; Raffeld, M.; Quezado, M.; Trepel, J.; Aldape, K.; Pommier, Y.; Wu, J. MGMT Status as a Clinical Biomarker in Glioblastoma. Trends Cancer 2020, 6, 380–391. [Google Scholar]
  214. Mansouri, A.; Hachem, L.D.; Mansouri, S.; Nassiri, F.; Laperriere, N.J.; Xia, D.; Lindeman, N.I.; Wen, P.Y.; Chakravarti, A.; Mehta, M.P.; et al. MGMT promoter methylation status testing to guide therapy for glioblastoma: Refining the approach based on emerging evidence and current challenges. Neuro Oncol. 2019, 21, 167–178. [Google Scholar]
  215. Binabaj, M.M.; Bahrami, A.; ShahidSales, S.; Joodi, M.; Joudi Mashhad, M.; Hassanian, S.M.; Anvari, K.; Avan, A. The prognostic value of MGMT promoter methylation in glioblastoma: A meta-analysis of clinical trials. J. Cell Physiol. 2018, 233, 378–386. [Google Scholar] [PubMed]
  216. Zhang, Z.; Xin, S.; Gao, M.; Cai, Y. Promoter hypermethylation of MGMT gene may contribute to the pathogenesis of gastric cancer: A PRISMA-compliant meta-analysis. Medicine 2017, 96, e6708. [Google Scholar] [PubMed]
  217. Lou, L.; Zhang, W.; Li, J.; Wang, Y. Abnormal MGMT Promoter Methylation in Gastrointestinal Stromal Tumors: Genetic Susceptibility and Association with Clinical Outcome. Cancer Manag. Res. 2020, 12, 9941–9952. [Google Scholar]
  218. Chen, B.; Ying, X.; Bao, L. MGMT gene promoter methylation in humoral tissue as biomarker for lung cancer diagnosis: An update meta-analysis. Thorac. Cancer 2021, 12, 3194–3200. [Google Scholar]
  219. Jank, P.; Gehlhaar, C.; Bianca, L.; Caterina, F.; Andreas, S.; Karn, T.; Marmé, F.; Sinn, H.P.; van Mackelenbergh, M.; Sinn, B.; et al. MGMT promoter methylation in triple negative breast cancer of the GeparSixto trial. PLoS ONE 2020, 15, e0238021. [Google Scholar]
  220. Chai, R.; Li, G.; Liu, Y.; Zhang, K.; Zhao, Z.; Wu, F.; Chang, Y.; Pang, B.; Li, J.; Li, Y.; et al. Predictive value of MGMT promoter methylation on the survival of TMZ treated IDH-mutant glioblastoma. Cancer Biol. Med. 2021, 18, 272–282. [Google Scholar]
  221. Mulholland, S.; Pearson, D.M.; Hamoudi, R.A.; Malley, D.S.; Smith, C.M.; Weaver, J.M.; Jones, D.T.; Kocialkowski, S.; Bäcklund, L.M.; Collins, V.P.; et al. MGMT CpG island is invariably methylated in adult astrocytic and oligodendroglial tumors with IDH1 or IDH2 mutations. Int. J. Cancer 2012, 131, 1104–1113. [Google Scholar]
  222. Abe, H.; Natsumeda, M.; Kanemaru, Y.; Watanabe, J.; Tsukamoto, Y.; Okada, M.; Yoshimura, J.; Oishi, M.; Fujii, Y. MGMT Expression Contributes to Temozolomide Resistance in H3K27M-Mutant Diffuse Midline Gliomas and MGMT Silencing to Temozolomide Sensitivity in IDH-Mutant Gliomas. Neurol. Med. Chir. 2018, 58, 290–295. [Google Scholar]
  223. Baldewpersad Tewarie, N.M.; Burgers, I.A.; Dawood, Y.; den Boon, H.C.; den Brok, M.G.; Klunder, J.H.; Koopmans, K.B.; Rademaker, E.; van den Broek, H.B.; van den Bersselaar, S.M.; et al. NADP+ -dependent IDH1 R132 mutation and its relevance for glioma patient survival. Med. Hypotheses 2013, 80, 728–731. [Google Scholar] [PubMed]
  224. Lu, Y.; Kwintkiewicz, J.; Liu, Y.; Tech, K.; Frady, L.N.; Su, Y.T.; Bautista, W.; Moon, S.I.; MacDonald, J.; Ewend, M.G.; et al. Chemosensitivity of IDH1-Mutated Gliomas Due to an Impairment in PARP1-Mediated DNA Repair. Cancer Res. 2017, 77, 1709–1718. [Google Scholar]
  225. Miller, J.J.; Cahill, D.P. MGMT promoter methylation and hypermutant recurrence in IDH mutant lower-grade glioma. Neuro Oncol. 2020, 22, 1553–1554. [Google Scholar]
  226. Lin, L.; Cai, J.; Tan, Z.; Meng, X.; Li, R.; Li, Y.; Jiang, C. Mutant IDH1 Enhances Temozolomide Sensitivity via Regulation of the ATM/CHK2 Pathway in Glioma. Cancer Res. Treat. 2021, 53, 367–377. [Google Scholar]
  227. Śledzińska, P.; Bebyn, M.G.; Furtak, J.; Kowalewski, J.; Lewandowska, M.A. Prognostic and Predictive Biomarkers in Gliomas. Int. J. Mol. Sci. 2021, 22, 10373. [Google Scholar] [CrossRef] [PubMed]
  228. Hadoux, J.; Favier, J.; Scoazec, J.Y.; Leboulleux, S.; Al Ghuzlan, A.; Caramella, C.; Déandreis, D.; Borget, I.; Loriot, C.; Chougnet, C.; et al. SDHB mutations are associated with response to temozolomide in patients with metastatic pheochromocytoma or paraganglioma. Int. J. Cancer 2014, 135, 2711–2720. [Google Scholar] [PubMed]
  229. Giger, O.T.; Ten Hoopen, R.; Shorthouse, D.; Abdullahi, S.; Bulusu, V.R.; Jadhav, S.; Maher, E.R.; Casey, R.T. Preferential MGMT hypermethylation in SDH-deficient wild-type GIST. J. Clin. Pathol. 2023, 77, 34–39. [Google Scholar]
  230. Müller, T.; Gessi, M.; Waha, A.; Isselstein, L.J.; Luxen, D.; Freihoff, D.; Freihoff, J.; Becker, A.; Simon, M.; Hammes, J.; et al. Nuclear exclusion of TET1 is associated with loss of 5-hydroxymethylcytosine in IDH1 wild-type gliomas. Am. J. Pathol. 2012, 181, 675–683. [Google Scholar]
  231. Turcan, S.; Rohle, D.; Goenka, A.; Walsh, L.A.; Fang, F.; Yilmaz, E.; Campos, C.; Fabius, A.W.; Lu, C.; Ward, P.S.; et al. IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 2012, 483, 479–483. [Google Scholar]
  232. Malta, T.M.; de Souza, C.F.; Sabedot, T.S.; Silva, T.C.; Mosella, M.S.; Kalkanis, S.N.; Snyder, J.; Castro, A.V.B.; Noushmehr, H. Glioma CpG island methylator phenotype (G-CIMP): Biological and clinical implications. Neuro Oncol. 2018, 20, 608–620. [Google Scholar]
  233. Madala, H.R.; Punganuru, S.R.; Arutla, V.; Misra, S.; Thomas, T.J.; Srivenugopal, K.S. Beyond Brooding on Oncometabolic Havoc in IDH-Mutant Gliomas and AML: Current and Future Therapeutic Strategies. Cancers 2018, 10, 49. [Google Scholar] [CrossRef]
  234. Ajalla Aleixo, M.A.; Rangel, V.L.; Rustiguel, J.K.; de Pádua, R.A.P.; Nonato, M.C. Structural, biochemical and biophysical characterization of recombinant human fumarate hydratase. FEBS J. 2019, 286, 1925–1940. [Google Scholar]
  235. Chen, T.; Wang, T.; Liang, W.; Zhao, Q.; Yu, Q.; Ma, C.M.; Zhuo, L.; Guo, D.; Zheng, K.; Zhou, C.; et al. PAK4 Phosphorylates Fumarase and Blocks TGFβ-Induced Cell Growth Arrest in Lung Cancer Cells. Cancer Res. 2019, 79, 1383–1397. [Google Scholar]
  236. Xu, Y.; Ayrapetov, M.K.; Xu, C.; Gursoy-Yuzugullu, O.; Hu, Y.; Price, B.D. Histone H2A.Z controls a critical chromatin remodeling step required for DNA double-strand break repair. Mol. Cell 2012, 48, 723–733. [Google Scholar]
  237. Williamson, E.A.; Wray, J.W.; Bansal, P.; Hromas, R. Overview for the histone codes for DNA repair. Prog. Mol. Biol. Transl. Sci. 2012, 110, 207–227. [Google Scholar]
  238. Saatchi, F.; Kirchmaier, A.L. Tolerance of DNA replication stress is promoted by fumarate through modulation of histone demethylation and enhancement of replicative intermediate processing in Saccharomyces cerevisiae. Genetics 2019, 212, 631–654. [Google Scholar]
  239. Singer, E.; Silas, Y.B.; Ben-Yehuda, S.; Pines, O. Bacterial fumarase and L-malic acid are evolutionary ancient components of the DNA damage response. eLife 2017, 6, e30927. [Google Scholar] [PubMed]
  240. Yip, J.; Wang, S.; Tan, J.; Lim, T.K.; Lin, Q.; Yu, Z.; Karmon, O.; Pines, O.; Lehming, N. Fumarase affects the deoxyribonucleic acid damage response by protecting the mitochondrial desulfurase Nfs1p from modification and inactivation. iScience 2021, 24, 103354. [Google Scholar]
  241. Karakaidos, P.; Karagiannis, D.; Rampias, T. Resolving DNA damage: Epigenetic regulation of DNA repair. Molecules 2020, 25, 2496. [Google Scholar] [CrossRef] [PubMed]
  242. Dabin, J.; Fortuny, A.; Polo, S.E. Epigenome Maintenance in Response to DNA Damage. Mol. Cell 2016, 62, 712–727. [Google Scholar] [PubMed]
  243. Filipp, F.V. Crosstalk between epigenetics and metabolism-Yin and Yang of histone demethylases and methyltransferases in cancer. Brief. Funct. Genomics 2017, 16, 320–325. [Google Scholar] [PubMed]
  244. Khoury-Haddad, H.; Guttmann-Raviv, N.; Ipenberg, I.; Huggins, D.; Jeyasekharan, A.D.; Ayoub, N. PARP1-dependent recruitment of KDM4D histone demethylase to DNA damage sites promotes double-strand break repair. Proc. Natl. Acad. Sci. USA 2014, 111, E728–E737. [Google Scholar] [PubMed]
  245. Efimova, E.V.; Takahashi, S.; Shamsi, N.A.; Wu, D.; Labay, E.; Ulanovskaya, O.A.; Weichselbaum, R.R.; Kozmin, S.A.; Kron, S.J. Linking Cancer Metabolism to DNA Repair and Accelerated Senescence. Mol. Cancer Res. 2016, 14, 173–184. [Google Scholar]
  246. Wentzel, J.F.; Lewies, A.; Bronkhorst, A.J.; Van Dyk, E.; Du Plessis, L.H.; Pretorius, P.J. Exposure to high levels of fumarate and succinate leads to apoptotic cytotoxicity and altered global DNA methylation profiles in vitro. Biochimie 2017, 135, 28–34. [Google Scholar] [PubMed]
  247. Wang, Y.; Wild, A.T.; Turcan, S.; Wu, W.H.; Sigel, C.; Klimstra, D.S.; Ma, X.; Gong, Y.; Holland, E.C.; Huse, J.T.; et al. Targeting therapeutic vulnerabilities with PARP inhibition and radiation in IDH-mutant gliomas and cholangiocarcinomas. Sci. Adv. 2020, 6, eaaz3221. [Google Scholar] [PubMed]
  248. Johnson, T.I.; Costa, A.S.H.; Ferguson, A.N.; Frezza, C. Fumarate hydratase loss promotes mitotic entry in the presence of DNA damage after ionising radiation. Cell Death Dis. 2018, 9, 913. [Google Scholar] [PubMed]
  249. Sun, Y.; Jiang, X.; Xu, Y.; Ayrapetov, M.K.; Moreau, L.A.; Whetstine, J.R.; Price, B.D. Histone H3 methylation links DNA damage detection to activation of the tumour suppressor Tip60. Nat. Cell Biol. 2009, 11, 1376–1382. [Google Scholar]
  250. Bondarev, A.D.; Attwood, M.M.; Jonsson, J.; Chubarev, V.N.; Tarasov, V.V.; Schiöth, H.B. Recent developments of HDAC inhibitors: Emerging indications and novel molecules. Br. J. Clin. Pharmacol. 2021, 87, 4577–4597. [Google Scholar]
  251. Biersack, B.; Polat, S.; Höpfner, M. Anticancer properties of chimeric HDAC and kinase inhibitors. Semin. Cancer Biol. 2022, 83, 472–486. [Google Scholar]
  252. Daško, M.; de Pascual-Teresa, B.; Ortín, I.; Ramos, A. HDAC Inhibitors: Innovative Strategies for Their Design and Applications. Molecules 2022, 27, 715. [Google Scholar] [CrossRef] [PubMed]
  253. Sutendra, G.; Kinnaird, A.; Dromparis, P.; Paulin, R.; Stenson, T.H.; Haromy, A.; Hashimoto, K.; Zhang, N.; Flaim, E.; Michelakis, E.D. A nuclear pyruvate dehydrogenase complex is important for the generation of acetyl-CoA and histone acetylation. Cell 2014, 158, 84–97. [Google Scholar] [PubMed]
  254. Park, S.; Mossmann, D.; Chen, Q.; Wang, X.; Dazert, E.; Colombi, M.; Schmidt, A.; Ryback, B.; Ng, C.K.; Terracciano, L.M.; et al. Transcription factors TEAD2 and E2A globally repress acetyl-CoA synthesis to promote tumorigenesis. Mol. Cell 2022, 82, 4246–4261.e11. [Google Scholar] [PubMed]
  255. He, W.; Li, Q.; Li, X. Acetyl-CoA regulates lipid metabolism and histone acetylation modification in cancer. Biochim. Biophys. Acta Rev. Cancer 2023, 1878, 188837. [Google Scholar]
  256. Izzo, L.T.; Trefely, S.; Demetriadou, C.; Drummond, J.M.; Mizukami, T.; Kuprasertkul, N.; Farria, A.T.; Nguyen, P.T.; Murali, N.; Reich, L.; et al. Acetylcarnitine shuttling links mitochondrial metabolism to histone acetylation and lipogenesis. Sci. Adv. 2023, 9, eadf0115. [Google Scholar] [PubMed]
  257. Cleary, J.M.; Aguirre, A.J.; Shapiro, G.I.; D’Andrea, A.D. Biomarker-Guided Development of DNA Repair Inhibitors. Mol. Cell 2020, 78, 1070–1085. [Google Scholar] [PubMed]
  258. Makovec, T. Cisplatin and beyond: Molecular mechanisms of action and drug resistance development in cancer chemotherapy. Radiol. Oncol. 2019, 53, 148–158. [Google Scholar] [PubMed]
  259. Bukowski, K.; Kciuk, M.; Kontek, R. Mechanisms of Multidrug Resistance in Cancer Chemotherapy. Int. J. Mol. Sci. 2020, 21, 3233. [Google Scholar] [CrossRef] [PubMed]
  260. Rocha, C.R.R.; Silva, M.M.; Quinet, A.; Cabral-Neto, J.B.; Menck, C.F.M. DNA repair pathways and cisplatin resistance: An intimate relationship. Clinics 2018, 73 (Suppl. S1), e478s. [Google Scholar]
  261. Fox, M.; Roberts, J.J. Drug resistance and DNA repair. Cancer Metastasis Rev. 1987, 6, 261–281. [Google Scholar]
  262. Stordal, B.; Pavlakis, N.; Davey, R. A systematic review of platinum and taxane resistance from bench to clinic: An inverse relationship. Cancer Treat. Rev. 2007, 33, 688–703. [Google Scholar] [PubMed]
  263. Galluzzi, L.; Vitale, I.; Michels, J.; Brenner, C.; Szabadkai, G.; Harel-Bellan, A.; Kroemer, G. Systems biology of cisplatin resistance: Past, present and future. Cell Death Dis. 2014, 5, e1257. [Google Scholar]
  264. Kara, A.; Özgür, A.; Nalbantoğlu, S.; Karadağ, A. DNA repair pathways and their roles in drug resistance for lung adenocarcinoma. Mol. Biol. Rep. 2021, 48, 3813–3825. [Google Scholar] [PubMed]
  265. Zhang, J.; Stevens, M.F.; Bradshaw, T.D. Temozolomide: Mechanisms of action, repair and resistance. Curr. Mol. Pharmacol. 2012, 5, 102–114. [Google Scholar]
  266. Amaya, M.L.; Pollyea, D.A. Targeting the IDH2 Pathway in Acute Myeloid Leukemia. Clin. Cancer Res. 2018, 24, 4931–4936. [Google Scholar]
  267. Nassereddine, S.; Lap, C.J.; Tabbara, I.A. Evaluating ivosidenib for the treatment of relapsed/refractory AML: Design, development, and place in therapy. Onco Targets Ther. 2018, 12, 303–308. [Google Scholar]
  268. Majchrzak-Celińska, A.; Warych, A.; Szoszkiewicz, M. Novel Approaches to Epigenetic Therapies: From Drug Combinations to Epigenetic Editing. Genes 2021, 12, 208. [Google Scholar] [CrossRef]
  269. Sabatier, M.; Boet, E.; Zaghdoudi, S.; Guiraud, N.; Hucteau, A.; Polley, N.; Cognet, G.; Saland, E.; Lauture, L.; Farge, T.; et al. Activation of Vitamin D Receptor Pathway Enhances Differentiating Capacity in Acute Myeloid Leukemia with Isocitrate Dehydrogenase Mutations. Cancers 2021, 13, 5243. [Google Scholar] [CrossRef] [PubMed]
  270. Stuani, L.; Sabatier, M.; Saland, E.; Cognet, G.; Poupin, N.; Bosc, C.; Castelli, F.A.; Gales, L.; Turtoi, E.; Montersino, C.; et al. Mitochondrial metabolism supports resistance to IDH mutant inhibitors in acute myeloid leukemia. J. Exp. Med. 2021, 218, e20200924. [Google Scholar]
  271. Yao, K.; Liu, H.; Yin, J.; Yuan, J.; Tao, H. Synthetic lethality and synergetic effect: The effective strategies for therapy of IDH-mutated cancers. J. Exp. Clin. Cancer Res. 2021, 40, 263. [Google Scholar]
  272. Gatto, L.; Franceschi, E.; Tosoni, A.; Di Nunno, V.; Maggio, I.; Lodi, R.; Brandes, A.A. IDH Inhibitors and Beyond: The Cornerstone of Targeted Glioma Treatment. Mol. Diagn. Ther. 2021, 25, 457–473. [Google Scholar]
  273. Oltvai, Z.N.; Harley, S.E.; Koes, D.; Michel, S.; Warlick, E.D.; Nelson, A.C.; Yohe, S.; Mroz, P. Assessing acquired resistance to IDH1 inhibitor therapy by full-exon IDH1 sequencing and structural modeling. Cold Spring Harb. Mol. Case Stud. 2021, 7, a006007. [Google Scholar]
  274. Godel, M.; Ortone, G.; Anobile, D.P.; Pasino, M.; Randazzo, G.; Riganti, C.; Kopecka, J. Targeting Mitochondrial Oncometabolites: A New Approach to Overcome Drug Resistance in Cancer. Pharmaceutics 2021, 13, 762. [Google Scholar] [CrossRef]
  275. Kim, G.H.; Choi, S.Y.; Oh, T.I.; Kan, S.Y.; Kang, H.; Lee, S.; Oh, T.; Ko, H.M.; Lim, J.H. IDH1R132H Causes Resistance to HDAC Inhibitors by Increasing NANOG in Glioblastoma Cells. Int. J. Mol. Sci. 2019, 20, 2679. [Google Scholar] [CrossRef] [PubMed]
  276. Lavacchi, D.; Caliman, E.; Rossi, G.; Buttitta, E.; Botteri, C.; Fancelli, S.; Pellegrini, E.; Roviello, G.; Pillozzi, S.; Antonuzzo, L. Ivosidenib in IDH1-mutated cholangiocarcinoma: Clinical evaluation and future directions. Pharmacol. Ther. 2022, 237, 108170. [Google Scholar]
  277. Intlekofer, A.M.; Shih, A.H.; Wang, B.; Nazir, A.; Rustenburg, A.S.; Albanese, S.K.; Patel, M.; Famulare, C.; Correa, F.M.; Takemoto, N.; et al. Acquired resistance to IDH inhibition through trans or cis dimer-interface mutations. Nature 2018, 559, 125–129. [Google Scholar] [PubMed]
  278. Du, Z.; Liu, X.; Chen, T.; Gao, W.; Wu, Z.; Hu, Z.; Wei, D.; Gao, C.; Li, Q. Targeting a Sirt5-Positive Subpopulation Overcomes Multidrug Resistance in Wild-Type Kras Colorectal Carcinomas. Cell Rep. 2018, 22, 2677–2689. [Google Scholar]
  279. Gao, M.; Yi, J.; Zhu, J.; Minikes, A.M.; Monian, P.; Thompson, C.B.; Jiang, X. Role of Mitochondria in Ferroptosis. Mol. Cell 2019, 73, 354–363.e3. [Google Scholar]
  280. Wang, T.X.; Liang, J.Y.; Zhang, C.; Xiong, Y.; Guan, K.L.; Yuan, H.X. The oncometabolite 2-hydroxyglutarate produced by mutant IDH1 sensitizes cells to ferroptosis. Cell Death Dis. 2019, 10, 755. [Google Scholar] [PubMed]
  281. Kes, M.M.G.; Van den Bossche, J.; Griffioen, A.W.; Huijbers, E.J.M. Oncometabolites lactate and succinate drive pro-angiogenic macrophage response in tumors. Biochim. Biophys. Acta Rev. Cancer 2020, 1874, 188427. [Google Scholar]
  282. van der Merwe, M.; van Niekerk, G.; Fourie, C.; du Plessis, M.; Engelbrecht, A.M. The impact of mitochondria on cancer treatment resistance. Cell Oncol. 2021, 44, 983–995. [Google Scholar]
  283. Xu, Y.; Li, Y.; Chen, X.; Xiang, F.; Deng, Y.; Li, Z.; Wei, D. TGF-β protects osteosarcoma cells from chemotherapeutic cytotoxicity in a SDH/HIF1α dependent manner. BMC Cancer 2021, 21, 1200. [Google Scholar]
  284. Bunse, L.; Pusch, S.; Bunse, T.; Sahm, F.; Sanghvi, K.; Friedrich, M.; Alansary, D.; Sonner, J.K.; Green, E.; Deumelandt, K.; et al. Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate. Nat. Med. 2018, 24, 1192–1203. [Google Scholar]
  285. Oizel, K.; Gratas, C.; Nadaradjane, A.; Oliver, L.; Vallette, F.M.; Pecqueur, C. D-2-Hydroxyglutarate does not mimic all the IDH mutation effects, in particular the reduced etoposide-triggered apoptosis mediated by an alteration in mitochondrial NADH. Cell Death Dis. 2015, 6, e1704. [Google Scholar]
  286. Giallongo, S.; Costa, F.; Longhitano, L.; Giallongo, C.; Ferrigno, J.; Tropea, E.; Vicario, N.; Li Volti, G.; Parenti, R.; Barbagallo, I.; et al. The Pleiotropic Effects of Fumarate: From Mitochondrial Respiration to Epigenetic Rewiring and DNA Repair Mechanisms. Metabolites 2023, 13, 880. [Google Scholar] [CrossRef] [PubMed]
  287. Inoue, S.; Li, W.Y.; Tseng, A.; Beerman, I.; Elia, A.J.; Bendall, S.C.; Lemonnier, F.; Kron, K.J.; Cescon, D.W.; Hao, Z.; et al. Mutant IDH1 Downregulates ATM and Alters DNA Repair and Sensitivity to DNA Damage Independent of TET2. Cancer Cell 2016, 30, 337–348. [Google Scholar]
  288. Su, R.; Dong, L.; Li, C.; Nachtergaele, S.; Wunderlich, M.; Qing, Y.; Deng, X.; Wang, Y.; Weng, X.; Hu, C.; et al. R-2HG Exhibits Anti-tumor Activity by Targeting FTO/m6A/MYC/CEBPA Signaling. Cell 2018, 172, 90–105.e23. [Google Scholar]
  289. Cairncross, J.G.; Wang, M.; Jenkins, R.B.; Shaw, E.G.; Giannini, C.; Brachman, D.G.; Buckner, J.C.; Fink, K.L.; Souhami, L.; Laperriere, N.J.; et al. Benefit from procarbazine, lomustine, and vincristine in oligodendroglial tumors is associated with mutation of IDH. J. Clin. Oncol. 2014, 32, 783–790. [Google Scholar]
  290. Hartmann, C.; Hentschel, B.; Simon, M.; Westphal, M.; Schackert, G.; Tonn, J.C.; Loeffler, M.; Reifenberger, G.; Pietsch, T.; Von Deimling, A.; et al. German Glioma Network. Long-term survival in primary glioblastoma with versus without isocitrate dehydrogenase mutations. Clin. Cancer Res. 2013, 19, 5146–5157. [Google Scholar]
  291. Ohba, S.; Mukherjee, J.; See, W.L.; Pieper, R.O. Mutant IDH1-driven cellular transformation increases RAD51-mediated homologous recombination and temozolomide resistance. Cancer Res. 2014, 74, 4836–4844. [Google Scholar]
  292. Tran, A.N.; Lai, A.; Li, S.; Pope, W.B.; Teixeira, S.; Harris, R.J.; Woodworth, D.C.; Nghiemphu, P.L.; Cloughesy, T.F.; Ellingson, B.M. Increased sensitivity to radiochemotherapy in IDH1 mutant glioblastoma as demonstrated by serial quantitative MR volumetry. Neuro Oncol. 2014, 16, 414–420. [Google Scholar] [PubMed]
  293. Chan, S.M.; Thomas, D.; Corces-Zimmerman, M.R.; Xavy, S.; Rastogi, S.; Hong, W.J.; Zhao, F.; Medeiros, B.C.; Tyvoll, D.A.; Majeti, R. Isocitrate dehydrogenase 1 and 2 mutations induce BCL-2 dependence in acute myeloid leukemia. Nat. Med. 2015, 21, 178–184. [Google Scholar]
  294. Minniti, G.; Scaringi, C.; Arcella, A.; Lanzetta, G.; Di Stefano, D.; Scarpino, S.; Bozzao, A.; Pace, A.; Villani, V.; Salvati, M.; et al. IDH1 mutation and MGMT methylation status predict survival in patients with anaplastic astrocytoma treated with temozolomide-based chemoradiotherapy. J. Neuro-Oncol. 2014, 118, 377–383. [Google Scholar] [PubMed]
  295. Kaina, B.; Christmann, M. DNA repair in personalized brain cancer therapy with temozolomide and nitrosoureas. DNA Repair 2019, 78, 128–141. [Google Scholar]
  296. Nay, S.L.; Lee, D.H.; Bates, S.E.; O’Connor, T.R. Alkbh2 protects against lethality and mutation in primary mouse embryonic fibroblasts. DNA Repair 2012, 11, 502–510. [Google Scholar]
  297. Johannessen, T.C.; Prestegarden, L.; Grudic, A.; Hegi, M.E.; Tysnes, B.B.; Bjerkvig, R. The DNA repair protein ALKBH2 mediates temozolomide resistance in human glioblastoma cells. Neuro Oncol. 2013, 15, 269–278. [Google Scholar]
  298. Nie, S.; Zhang, L.; Liu, J.; Wan, Y.; Jiang, Y.; Yang, J.; Sun, R.; Ma, X.; Sun, G.; Meng, H.; et al. ALKBH5-HOXA10 loop-mediated JAK2 m6A demethylation and cisplatin resistance in epithelial ovarian cancer. J. Exp. Clin. Cancer Res. 2021, 40, 284. [Google Scholar]
  299. Fukumoto, T.; Zhu, H.; Nacarelli, T.; Karakashev, S.; Fatkhutdinov, N.; Wu, S.; Liu, P.; Kossenkov, A.V.; Showe, L.C.; Jean, S.; et al. N6-Methylation of Adenosine of FZD10 mRNA Contributes to PARP Inhibitor Resistance. Cancer Res. 2019, 79, 2812–2820. [Google Scholar] [PubMed]
  300. Shi, D.D.; Anand, S.; Abdullah, K.G.; McBrayer, S.K. DNA damage in IDH-mutant gliomas: Mechanisms and clinical implications. J. Neuro-Oncol. 2023, 162, 515–523. [Google Scholar]
  301. Monferrer, E.; Sanegre, S.; Vieco-Martí, I.; López-Carrasco, A.; Fariñas, F.; Villatoro, A.; Abanades, S.; Mañes, S.; de la Cruz-Merino, L.; Noguera, R.; et al. Immunometabolism Modulation in Therapy. Biomedicines 2021, 9, 798. [Google Scholar] [CrossRef]
  302. Chen, C.; Wang, Z.; Qin, Y. Connections between metabolism and epigenetics: Mechanisms and novel anti-cancer strategy. Front. Pharmacol. 2022, 13, 935536. [Google Scholar]
  303. Kitagawa, Y.; Kobayashi, A.; Cahill, D.P.; Wakimoto, H.; Tanaka, S. Molecular biology and novel therapeutics for IDH mutant gliomas: The new era of IDH inhibitors. Biochim. Biophys. Acta. Rev. Cancer 2024, 1879, 189102. [Google Scholar]
Figure 1. TCA cycle. Represented with the chemical formulas of the substrates/products of each reaction, indicating the catalyzing enzyme and whether the reaction generates energy-related molecules (NADH, FADH2 or GPT). Abbreviations: CS—Citrate Synthase (EC 2.3.3.1); Aconitase—Aconitate Hydratase (EC 4.2.1.3); IDH—Isocitrate Dehydrogenase (EC 1.1.1.42 and EC 1.1.1.41), α-KGDH—α-Ketoglutarate Dehydrogenase (EC 1.2.4.2); SCS—Succinyl-CoA Synthetase or Succinate Thiokinase (EC 6.2.1.4); SDH—Succinate Dehydrogenase (EC 1.3.5.1); FH—Fumarate Hydratase, or Fumarase (EC 4.2.1.2); MDH—Malate Dehydrogenase (EC 1.1.1.37). Formulas were obtained from PubChem (https://pubchem.ncbi.nlm.nih.gov/) (Accessed on 20 February 2024).
Figure 1. TCA cycle. Represented with the chemical formulas of the substrates/products of each reaction, indicating the catalyzing enzyme and whether the reaction generates energy-related molecules (NADH, FADH2 or GPT). Abbreviations: CS—Citrate Synthase (EC 2.3.3.1); Aconitase—Aconitate Hydratase (EC 4.2.1.3); IDH—Isocitrate Dehydrogenase (EC 1.1.1.42 and EC 1.1.1.41), α-KGDH—α-Ketoglutarate Dehydrogenase (EC 1.2.4.2); SCS—Succinyl-CoA Synthetase or Succinate Thiokinase (EC 6.2.1.4); SDH—Succinate Dehydrogenase (EC 1.3.5.1); FH—Fumarate Hydratase, or Fumarase (EC 4.2.1.2); MDH—Malate Dehydrogenase (EC 1.1.1.37). Formulas were obtained from PubChem (https://pubchem.ncbi.nlm.nih.gov/) (Accessed on 20 February 2024).
Ijms 25 09054 g001
Figure 2. Pathways to generate TCA cycle indirect metabolites. (A) Itaconate, obtained from cis-Aconitate when the concentration of citrate is high. (B) 2-Hydroxyglutarate isomers (D- and L-2HG), generated from α-KG as waste products that can revert to their α-KG origin or as a product of gain-of-function mutations in IDH genes for D-2HG. Abbreviations: CAD—Cis-Aconitate Decarboxylase; IDH—Isocitrate Dehydrogenase; HOT—Hydroxyacid-Oxoacid Transhydrogenase; D-2HGDH—D-2Hydroxyglutarate Dehydrogenase; L-2HGDH—L-2Hydroxyglutarate Dehydrogenase; L-MDH—L-Malate Dehydrogenase. Formulas were obtained from PubChem (https://pubchem.ncbi.nlm.nih.gov/) (Accessed on 20 February 2024).
Figure 2. Pathways to generate TCA cycle indirect metabolites. (A) Itaconate, obtained from cis-Aconitate when the concentration of citrate is high. (B) 2-Hydroxyglutarate isomers (D- and L-2HG), generated from α-KG as waste products that can revert to their α-KG origin or as a product of gain-of-function mutations in IDH genes for D-2HG. Abbreviations: CAD—Cis-Aconitate Decarboxylase; IDH—Isocitrate Dehydrogenase; HOT—Hydroxyacid-Oxoacid Transhydrogenase; D-2HGDH—D-2Hydroxyglutarate Dehydrogenase; L-2HGDH—L-2Hydroxyglutarate Dehydrogenase; L-MDH—L-Malate Dehydrogenase. Formulas were obtained from PubChem (https://pubchem.ncbi.nlm.nih.gov/) (Accessed on 20 February 2024).
Ijms 25 09054 g002
Figure 3. Relationships between TCA cycle metabolites and DNA repair in human cells: the AlkB pathway, following nitrogen alkylation damage, and the MGMT pathway, following oxygen alkylation damage. Human AlkB homologs remove N-alkylation damage from DNA and RNA, and 2HG and fumarate inhibit them (signaled in red). MGMT protein removes O-alkylation damage from DNA, and 2HG and succinate inhibit it through the methylation of its gene promoter (signaled in red). Abbreviations: ALKBs—human alkylated DNA repair B proteins; CIMP—CpG island methylator phenotype; 2HG—2-hydroxyglutarate; MGMT—methylguanine methyltransferase; N1-G, N3-C, N1-A—positions of ring nitrogen atoms in guanine, cytosine and adenine nucleobases; N6-A—nitrogen atom from the exocyclic amino group of adenine nucleobase; O6-G—oxygen atom from the keto exocyclic group of guanine nucleobase. H2C=O—formaldehyde formed as a byproduct of the reaction catalyzed by ALKBH proteins.
Figure 3. Relationships between TCA cycle metabolites and DNA repair in human cells: the AlkB pathway, following nitrogen alkylation damage, and the MGMT pathway, following oxygen alkylation damage. Human AlkB homologs remove N-alkylation damage from DNA and RNA, and 2HG and fumarate inhibit them (signaled in red). MGMT protein removes O-alkylation damage from DNA, and 2HG and succinate inhibit it through the methylation of its gene promoter (signaled in red). Abbreviations: ALKBs—human alkylated DNA repair B proteins; CIMP—CpG island methylator phenotype; 2HG—2-hydroxyglutarate; MGMT—methylguanine methyltransferase; N1-G, N3-C, N1-A—positions of ring nitrogen atoms in guanine, cytosine and adenine nucleobases; N6-A—nitrogen atom from the exocyclic amino group of adenine nucleobase; O6-G—oxygen atom from the keto exocyclic group of guanine nucleobase. H2C=O—formaldehyde formed as a byproduct of the reaction catalyzed by ALKBH proteins.
Ijms 25 09054 g003
Figure 4. Relationships between TCA cycle metabolites and DNA repair in human cells: the FH pathway. FH protein moves from cytosol to nucleus to synthesize fumarate at DSB sites, where fumarate activates the NHEJ system. Abbreviations: DSB—double-strand break; FH—fumarate hydratase; H3K36—Lys36 in H3 histone; KDM2B—histone lysine demethylase 2B; NHEJ—non-homologous end joining system. Figure partially based on Leshets et al. [32].
Figure 4. Relationships between TCA cycle metabolites and DNA repair in human cells: the FH pathway. FH protein moves from cytosol to nucleus to synthesize fumarate at DSB sites, where fumarate activates the NHEJ system. Abbreviations: DSB—double-strand break; FH—fumarate hydratase; H3K36—Lys36 in H3 histone; KDM2B—histone lysine demethylase 2B; NHEJ—non-homologous end joining system. Figure partially based on Leshets et al. [32].
Ijms 25 09054 g004
Figure 5. Relationships between TCA cycle metabolites and DNA repair in human cells: the chromatin remodeling pathway. Induction of DSBs increases trimethylation of H3 lysine K9. KDM4 demethylation to monomethyl H3K9 activates the HRR system. Oncometabolites inhibit KDM4 and impair repair by HRR. Abbreviations: DBSs—double-strand breaks; KDM4—histone lysine demethylase 4; H3K9—Lys9 in H3 histone; HRR—homologous recombination repair.
Figure 5. Relationships between TCA cycle metabolites and DNA repair in human cells: the chromatin remodeling pathway. Induction of DSBs increases trimethylation of H3 lysine K9. KDM4 demethylation to monomethyl H3K9 activates the HRR system. Oncometabolites inhibit KDM4 and impair repair by HRR. Abbreviations: DBSs—double-strand breaks; KDM4—histone lysine demethylase 4; H3K9—Lys9 in H3 histone; HRR—homologous recombination repair.
Ijms 25 09054 g005
Figure 6. Schematic representation of the main call of this work: do not remove the oncometabolites from the tumor cells—otherwise, the inhibited DNA repair pathways would be reactivated. No resistance is achieved when repair pathways are deficient (marked in red) Abbreviations: ALKB—AlkB pathway; MGMT—methylguanine methyltransferase; HRR—homologous recombination repair pathway; 2HG—2-Hydroxyglutarate; IDH—Isocitrate dehydrogenase deficient; IDH+—Isocitrate dehydrogenase wildtype.
Figure 6. Schematic representation of the main call of this work: do not remove the oncometabolites from the tumor cells—otherwise, the inhibited DNA repair pathways would be reactivated. No resistance is achieved when repair pathways are deficient (marked in red) Abbreviations: ALKB—AlkB pathway; MGMT—methylguanine methyltransferase; HRR—homologous recombination repair pathway; 2HG—2-Hydroxyglutarate; IDH—Isocitrate dehydrogenase deficient; IDH+—Isocitrate dehydrogenase wildtype.
Ijms 25 09054 g006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Álvarez-González, E.; Sierra, L.M. Tricarboxylic Acid Cycle Relationships with Non-Metabolic Processes: A Short Story with DNA Repair and Its Consequences on Cancer Therapy Resistance. Int. J. Mol. Sci. 2024, 25, 9054. https://doi.org/10.3390/ijms25169054

AMA Style

Álvarez-González E, Sierra LM. Tricarboxylic Acid Cycle Relationships with Non-Metabolic Processes: A Short Story with DNA Repair and Its Consequences on Cancer Therapy Resistance. International Journal of Molecular Sciences. 2024; 25(16):9054. https://doi.org/10.3390/ijms25169054

Chicago/Turabian Style

Álvarez-González, Enol, and Luisa María Sierra. 2024. "Tricarboxylic Acid Cycle Relationships with Non-Metabolic Processes: A Short Story with DNA Repair and Its Consequences on Cancer Therapy Resistance" International Journal of Molecular Sciences 25, no. 16: 9054. https://doi.org/10.3390/ijms25169054

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop