Sei sulla pagina 1di 9

Biochimie 135 (2017) 164e172

Contents lists available at ScienceDirect

Biochimie
journal homepage: www.elsevier.com/locate/biochi

Review

Genetic alterations in Krebs cycle and its impact on cancer


pathogenesis
Karishma Sajnani a, 1, Farhadul Islam a, b, 1, Robert Anthony Smith a, c, Vinod Gopalan a, 1,
Alfred King-Yin Lam a, *
a
Cancer Molecular Pathology, School of Medicine, Menzies Health Institute Queensland, Grifth University, Gold Coast, QLD, Australia
b
Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
c
Genomics Research Centre, Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology, Brisbane, Queensland,
Australia

a r t i c l e i n f o a b s t r a c t

Article history: Cancer cells exhibit alterations in many cellular processes, including oxygen sensing and energy meta-
Received 9 November 2016 bolism. Glycolysis in non-oxygen condition is the main energy production process in cancer rather than
Received in revised form mitochondrial respiration as in benign cells. Genetic and epigenetic alterations of Krebs cycle enzymes
14 February 2017
favour the shift of cancer cells from oxidative phosphorylation to anaerobic glycolysis. Mutations in
Accepted 16 February 2017
Available online 20 February 2017
genes encoding aconitase, isocitrate dehydrogenase, succinate dehydrogenase, fumarate hydratase, and
citrate synthase are noted in many cancers. Abnormalities of Krebs cycle enzymes cause ectopic pro-
duction of Krebs cycle intermediates (oncometabolites) such as 2-hydroxyglutarate, and citrate. These
Keywords:
Oncometabolites
oncometabolites stabilize hypoxia inducible factor 1 (HIF1), nuclear factor like 2 (Nrf2), inhibit p53 and
Mitochondria prolyl hydroxylase 3 (PDH3) activities as well as regulate DNA/histone methylation, which in turn
Krebs cycle activate cell growth signalling. They also stimulate increased glutaminolysis, glycolysis and production of
TCA cycle reactive oxygen species (ROS). Additionally, genetic alterations in Krebs cycle enzymes are involved with
Cancer metabolism increased fatty acid b-oxidations and epithelial mesenchymal transition (EMT) induction. These altered
Mutations phenomena in cancer could in turn promote carcinogenesis by stimulating cell proliferation and survival.
Overall, epigenetic and genetic changes of Krebs cycle enzymes lead to the production of oncometabolite
intermediates, which are important driving forces of cancer pathogenesis and progression. Under-
standing and applying the knowledge of these mechanisms opens new therapeutic options for patients
with cancer.
2017 Elsevier B.V. and Socit Franaise de Biochimie et Biologie Molculaire (SFBBM). All rights
reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165
2. Role of mitochondria in cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165
3. Tricarboxylic acid cycle and cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165
4. Role of metabolic enzyme of TCA cycle in cancer pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 166
4.1. Citrate synthase (CS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 166
4.2. Aconitase (Aco) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 166
4.3. Isocitrate dehydrogenase mutations (IDH) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 166
4.4. Succinate dehydrogenase (SDH) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167
4.5. Fumarate hydratase (FH) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168
5. Oncometabolites of the TCA cycle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168

* Corresponding author. Grifth Medical School, Gold Coast Campus, Gold Coast,
QLD, 4222, Australia.
E-mail address: a.lam@grifth.edu.au (A.K.-Y. Lam).
1
Equal to rst authors.

http://dx.doi.org/10.1016/j.biochi.2017.02.008
0300-9084/ 2017 Elsevier B.V. and Socit Franaise de Biochimie et Biologie Molculaire (SFBBM). All rights reserved.
K. Sajnani et al. / Biochimie 135 (2017) 164e172 165

5.1. Citrate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169


5.2. 2-Hydroxy glutarate (2HG) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169
6. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170

1. Introduction

Mitochondria are the power generator for cells. In the recent


years, they have been discovered to possess more complex func-
tions, especially in the pathogenesis of cancer [1]. Also, they are
involved in programmed cell death (apoptosis) in multicellular
organisms and regulate uncontrolled cell growth like cancer [2].
There are many therapeutic substances/drugs that can target
mitochondria and act as anticancer, immunosuppressant and
antiviral agents [3]. To make the best use of these recently available
and newly designed drugs to target mitochondria in cancer treat-
ment, we need to understand the mechanisms driven by the me-
tabolites in mitochondria in the pathogenesis of cancer.
In normal cells, glucose processing begins with oxidative
decarboxylation to generate energy and carbon dioxide (CO2) [4]. A
series of reactions, known as the tricarboxylic acid cycle (TCA cy-
cle), citric acid cycle or Krebs cycle are responsible for this oxidation
of glucose [4]. This cycle is the common metabolic pathway for fuel
molecules such as glucose, fatty acids, and amino acids [4]. Most Fig. 1. Abnormalities of Krebs cycle enzymes and metabolites associated with
energy yielding molecules can enter into the TCA cycle as acetyl cancer. Mutations in genes encoding TCA cycle enzymes result in a metabolic shift of
the cells and promote oncogenic transformation. Also, abnormalities of TCA cycle
CoA and this cycle is the principal metabolic process for cells.
enzymes causes accumulation of various oncometabolites ultimately leading to cancer
Biomolecules, for example glucose and fatty acids which have the development and progression. PDC: pyruvate dehydrogenase complex, CS: citrate
potential to be converted to acetyl groups or carboxylic groups synthase, Aco: aconitase, IDH: isocitrate dehydrogenase, OGDC: oxoglutarate dehy-
ultimately enter the TCA cycle for aerobic metabolism, especially drogenase complex, STK: succinate thiokinase, SDH: succinate dehydrogenase, FH:
for catabolism [4]. In oxygenated conditions, pyruvate produced fumarate hydratase, MDH: malate dehydrogenase, NAD/NADH: nicotinamide adenine
dinucleotide, FAD/FADH: avin adenine dinucleotide, CoA: coenzyme A, Pi: inorganic
from glucose is converted to acetyl CoA and enters into the TCA phosphate, GDP/GTP: guanosine di/tri phosphate, Q: coenzyme Q.
cycle for complete oxidation [4].
In 1956, Warburg reported that cancer cells had different
metabolic characteristics [5]. They mainly produce their energy in ago, it was reported by Otto Warburg that mitochondrial dysfunc-
the form of ATP (adenosine tri-phosphate) by the process of tion or defects could be the cause of cancer [4,11]. This observation
glycolysis (a non-oxidative breakdown of glucose in the cytosol of encouraged other researchers to investigate the role of mitochon-
cell) [5,6]. Thus, the energy metabolism of cancer cells shifts to dria in the pathogenesis of different cancers [12e15]. For example,
glycolysis from mitochondrial respiration. One possible reason that reduced mitochondrial genetic content, mitochondrial DNA
this may occur is that mutations in proto-oncogenes and tumour (mtDNA) copy number and oxidative damage to mtDNA were
suppressor genes which could create pseudohypoxia. Pseudohy- associated with the progression of non-small cell lung cancer [16].
poxia is a condition where cells activate oxygen deprivation Also, several studies reported that the expression of mitochondrial
mechanisms despite the presence of optimal oxygen concentra- genes were upregulated in different cancers [17,18].
tions in cancer. This condition results in the accumulation of un- The role of mitochondria in cancer formation appears to result
usual metabolites and other abnormalities in cancer cells [7]. from mutations in enzymes such as isocitrate dehydrogenase,
Recent advancements in genetic sequencing technologies have succinate dehydrogenase and fumarate dehydrogenase; all of them
enabled researchers to discover mutations in genes encoding are signicant TCA cycle enzymes [19,20]. Mutations of genes
metabolic enzymes that may be involved in this process [8]. Studies encoding these enzymes lead to abnormal accumulation of onco-
demonstrated that mutation of the genes involved in energy metabolites of the TCA cycle, which in turn, stimulates the forma-
metabolism (TCA cycle) caused unexpected enzymatic reactions tion and progression of cancer [9,20]. Mitochondrial dysfunctions
and abnormal accumulation of metabolites, termedoncometabo- due to abnormalities of metabolic enzymes and metabolites act as
lites [8,9] (Fig. 1). These oncometabolites in turn, regulate epige- the driving cause of cancer and can act as a second hit in the
netic mechanisms which control cell proliferation and process of cancer metabolic transformation [21,22]. Thus, the
differentiation [9]. mitochondrial abnormalities induce metabolic reprogramming in
cancer cells and increase the use of glycolysis, which in turn, fa-
vours cell survival and proliferation.
2. Role of mitochondria in cancer

Mitochondria are the power house of the cell. They produce the 3. Tricarboxylic acid cycle and cancer
major proportion of cellular energy and ROS, as well as regulating
apoptosis by modulating cytochrome C release through the The primary fuel source for the TCA cycle is acetyl CoA. The
permeability membrane pore complexes [10]. More than 70 years breakdown of one molecule of glucose yields two pyruvate
166 K. Sajnani et al. / Biochimie 135 (2017) 164e172

molecules. Then, acetyl CoA is generated from these pyruvate synthase (CS) aconitase (ACO2), succinate dehydrogenase (SDH),
molecules with the help of the pyruvate dehydrogenase complex fumarate hydratase (FH) and isocitrate dehydrogenase (IDH).
(PDC) [4]. A carboxyl group is eliminated in this reaction as CO2
molecules [23]. Acetyl groups of acetyl CoA are converted from the 4.1. Citrate synthase (CS)
rest of the two carbon moiety of pyruvate. The high energy sub-
stance nicotinamide adenine di-nucleotide (NADH; reduced) is Citrate synthase (CS) catalyses the conversion of oxaloacetate
generated in this stage, which in turn transfers a hydride ion (:H) to and acetyl CoA to citrate (the rst reaction of the Krebs cycle),
the electron transfer chain of the respiratory system [23]. The high which in turn regulates mitochondria mediated energy generation
energy thioester linkage between acetyl moiety of acetyl CoA and of cells [23]. Thus, citrate synthase can be assumed as a rate limiting
sulfhydryl moiety of b-mercaptoethylamine makes acetyl CoA a enzyme of the citric acid cycle. Mutational roles of CS in tumori-
high energy substance [4,24]. Acetyl CoA nally enters into the genesis were not studied in depth in the literature. However, Lin
TCA cycle to produce energy and CO2. and co-workers reported the functional role of citrate synthase in
In the Krebs cycle, oxaloacetate which is a four-carbon com- cancer metabolism in different types of cancer cells such as Hela,
pound is combined with another two-carbon acetyl unit to form a MCF7 and PC-3 [33]. They demonstrated that suppression of citrate
six-carbon citrate (tri-carboxylic acid). Isocitrate (an isomer of cit- synthase caused a metabolic shift of cellular bioenergetics from
rate) is then converted to a-ketoglutarate, a ve-carbon interme- mitochondrial oxidative phosphorylation to cytosolic glycolysis
diate of the TCA cycle by an oxidative decarboxylation reaction, [33]. Also, they noted that loss of citrate synthase expression was
catalysed by isocitrate dehydrogenase [23]. Succinate, a four carbon associated with the induction of epithelium mesenchymal transi-
compound, is then produced from a-ketoglutarate by oxidative tion (EMT) and increased the malignant behaviour of tumours [33].
decarboxylation. Succinate is then oxidised and formed another By contrast, it was demonstrated that citrate synthase was over-
four-carbon compound fumarate in presence of succinate dehy- expressed in pancreatic, renal and ovarian cancer when compared
drogenase. In this step, two hydrogen atoms along with their to non-neoplastic cells and tissues [34e36]. In addition, knock-
electrons retransferred to avin adenine dinucleotide (FAD), pro- down of citrate synthase in ovarian cancer cell lines inhibited cell
ducing FADH2 [23]. Subsequently, a water molecule is added to the proliferation, invasion and migration in vitro [34]. Thus, the role of
fumarate and generates malate with the help of the enzyme citrate synthase in cancer metabolism is ambiguous, but genetic
fumarase [4,23]. Finally oxaloacetate is regenerated from succinate alterations of this critical enzyme may have signicant impact in
[4]. Overall, a two-carbon acetyl moiety enters into the TCA cycle cancer pathogenesis and progression in specic cancer subtypes.
and two carbon atoms exit from the cycle as CO2 molecules. In these However, the specic role that deregulation of citrate synthase
process three hydride ions, and therefore, six electrons are trans- plays in carcinogenesis is not yet clear. The possible underlying
ferred to three NAD (nicotinamide adenine dinucleotide; oxi- mechanism may be an increase in citrate production by overex-
dised) and two hydrogen atoms (two electrons) are transferred to pressed citrate synthase which could act as a precursor for fatty
one FAD [4,23]. Thus, through the citric acid cycle, cells harvest acid/lipid synthesis for the cancer cells, which in turn promotes cell
high-energy electrons from the carbon skeleton of fuel molecules growth and proliferation. The other hypothesis is that the loss of
and utilize those electrons to generate NADH and FADH2 high- citrate synthase activity may induce mitochondrial dysfunctions
energy substances. In oxidative phosphorylation, these NADH and and could create a tumour-supporting glycolytic switch
FADH2 are re-oxidised and release the electrons [4,23]. These microenvironment.
electrons are then transferred through the electron-transport chain
and generate a proton gradient across the mitochondrial mem- 4.2. Aconitase (Aco)
brane. The generated proton gradient makes the protons ow
through ATP-synthase to produce adenosine tri-phosphate (ATP) Aconitase (Aco) is an essential enzyme located in the mito-
using adenosine di-phosphate (ADP) and inorganic phosphate (Pi) chondria. It catalyses the reversible isomerization of citrate to iso-
[4,23]. citrate in the TCA cycle [23]. Ternette and colleagues reported that
Studies demonstrated that abnormalities of the TCA cycle are fumarase hydratase (FH) decient cells had impaired aconitase
associated with different types of cancers [25e27] (Fig. 1). For synthase 2 enzyme activity [37]. These two alterations (mutation in
example, it was noted that phosphoenolpyruvate carboxykinase, a FH and impaired Aco2 function) contributed towards hereditary
regulator of TCA cycle ux, plays a crucial role in promoting cancer leiomyomatosis and renal cell carcinoma [37]. Aco2 mutation was
cell growth and proliferation, particularly in colorectal cancer cells also associated with gastric cancer [38]. Decreased levels of Aco2
[27]. Phosphoenolpyruvate carboxykinase increases glucose and have been reported in prostate cancer [39]. In addition, the
glutamine uptake in cancer cells and favours anabolic metabolism decreased level of Aco2 enzyme in prostate cancer was associated
[27]. This metabolic shift is important for highly proliferating cells, with down regulation of ZIP 1 (Zinc into prostate). ZIP 1 is a zinc
like cancer cells, which continuously require a supply of precursors transporter which could cause depletion of zinc in prostate cancer.
for the synthesis of lipids, proteins and nucleic acids [28]. Several This phenomenon leads to metabolic transformation of prostate
studies also demonstrated that dysfunction of mitochondrial cells by impairing the citrate oxidation and stimulates the conver-
metabolic pathways such as the TCA cycle and oxidative phos- sion of citrate secretory non-neoplastic epithelial cells to citrate
phorylation are associated with the conversion of non-neoplastic oxidizing prostatic carcinoma cells [39].
mammary epithelial cells to non-metastatic breast carcinoma and
then to the metastatic cancer cells [29e31]. 4.3. Isocitrate dehydrogenase mutations (IDH)

4. Role of metabolic enzyme of TCA cycle in cancer In the second steps of the TCA cycle, isocitrate dehydrogenase
pathogenesis (IDH) catalyses the conversion of isocitrate to a-ketoglutarate
which is an oxidative decarboxylation reaction [23]. Mutations in
Inherited and acquired alteration of TCA cycle enzymes have gene encoding IDH were reported in various cancers including
been demonstrated in different cancers [32]. There are numerous glioma, glioblastoma multiforme, acute myeloid leukaemia, chon-
alterations of TCA cycle enzymes and metabolites involved in drosarcoma, and intrahepatic cholangiocarcinoma [40e42]. In
pathogenesis of cancer (Fig. 2). The changes mainly occur in citrate addition, IDH mutation has been reported to regulate a set of genes
K. Sajnani et al. / Biochimie 135 (2017) 164e172 167

Fig. 2. Schematic representation of impacts of TCA cycle abnormalities in cancer. Impairment of Krebs cycle enzymes, metabolites and oncometabolites affects numerous
downstream metabolic pathways in cells which in turn promote carcinogenesis. Epigenetic and genetic alterations in TCA cycle components result in changing the cellular
behaviour towards the establishment of cancer.

at the transcriptional level, which are responsible for establishment radical and non-radical oxygen species generated from a partial
and progression of cancer [43]. Also, IDH mutations impair DNA reduction of oxygen and include, superoxide anion (O2-), hydrogen
demethylation in gliomas and generate a hypermethylation peroxide (H2O2), hydrogen radicals and similar molecules [59].
phenotype, which in turn was associated with the downregulation These ROS induce oxidative stress in cells, which in turn stimulate
of key genes implicated in differentiation of the chromafn cells carcinogenesis [60].
and the invasiveness of the paraganglioma [44]. Mutations in IDHs inhibit the generation of NADPH and were
The p53 gene plays crucial functions in multicellular organisms. also found to increase the consumption of NADPH by cells, which in
It prevents cancer formation, thus acts as a tumour suppressor turn promote the production of (R)-2-hydroxyglutarate (2HG), an
[45e47]. Genetic alteration of p53 is the most commonly studied oncometabolite [41,61]. It was noted that there was a correlation of
genetic alteration in cancer. The clinical and pathological roles of (R-)-2-hydroxyglutarate accumulation and tumorigenesis by
p53 have been demonstrated in many cancers [48e51]. Yan and co- modulating the tumour suppressor phenomenon autophagy in
worker reported that a substitution mutation at codon 132 of the cells in an oxidizing [62].
IDH gene was associated with the inactivation of p53 protein in Collagen is the key component of the extra-cellular matrix, the
glioma [52]. Another study demonstrated that IDH mutation is an network of bres that support the overall structure of tissues [63].
early event in glioma, which in turn affects p53 protein function in Collagen is also needed for the differentiation and proliferation of
the later stage of patients with glioma [53]. neurons and haematopoietic stem cells in brain and bone marrow
PI3K/AKT and RAS proteins constitute important growth sig- [64]. Sasaki and colleagues noted that IDH mutation conferred
nalling pathways, which control the fate of cells. The pathway reduced prolyl hydroxylase domain activity, which in turn leads to
mediated by these proteins is tightly regulated and controls pro- the disruption of collagen folding [65]. They also observed that this
grammed cell death, cell metabolism, proliferation and differenti- activity contributed to progression of glioma and acute myeloid
ation [54]. Abnormalities of PI3K-Akt pathway regulation can lead leukaemia [66]. Therefore, mutations in IDH can affect different
to an increase in signalling activity. This increased activation of biological processes that stimulate cancer formation and
signalling has been linked to a range of diseases including cancer progression.
[55]. It was demonstrated that IDH mutation activated the PI3K/
AKT signalling pathway in different cancers [56]. The authors also 4.4. Succinate dehydrogenase (SDH)
noted that PI3K plays a signicant role in maintaining the glycolytic
phenotype of cancers through the protein kinase B (PKB/Akt) sig- The succinate dehydrogenase complex (SDH) also called succi-
nalling network [56]. IDH mutation evokes hotspots in onco- nate ubiquinones oxydoreductase is a hetero-tetrameric
genes, such as KRAS [52]. Mutations in KRAS induce further (composed of SDHA, SDHB, SDHC and SDHD) and highly
modication in the genome which promotes progression and conserved protein. SDHA and SDHB act as catalytic subunits and
establishment of cancer [52]. SDHC and SDHD provide the binding site for ubiquinones (an
Studies reported that IDH mutation produced a high level of element of the electron transport chain) [67]. In the Krebs cycle,
reactive oxygen species (ROS) in cancer cells [57,58]. ROS consist of SDH converts succinate to fumarate [23]. Mutation of genes
168 K. Sajnani et al. / Biochimie 135 (2017) 164e172

encoding proteins involved in the SDH complex leads to the accu- at various amino residues [84]. Increased accumulation of succinate
mulation of succinate which can act as oncometabolite. Mutations facilitates the succination of protein residues. Succination is a post-
(loss of function) in SDH genes lead to hereditary paraganglioma translational modication of proteins, in this process a succinyl
and phaeochromocytoma, and other hereditary syndromes and moiety (-CO-CH2-CH-2-CO-) is added to a lysine residue of the
cancers [67,68]. Mutation of SDH genes leads to an increase state of protein molecule [85]. This posttranslational modication (adding
intracellular ROS [69]. This ROS increase in turn induces metabolic of succinyl unit to proteins) is noted in many proteins including
stress, genomic instability and promotes tumorigenesis [69]. DNA binding histone proteins [85]. The potential impact of protein
Mutations in SDH have been observed to cause accumulation of succination is not clear yet, but it is assumed that it can induce
succinate in mitochondria which subsequently leaves mitochondria notable alteration in the structure and function of proteins. This is
and inhibits the activity of enzyme HIFa prolyl hydroxylases. This because succination reverses the charge of lysine residues of pro-
inhibition permits escape of HIFa subunits from degradation and teins (from 1 to 1) and also incorporates a bigger structural unit
allows them to bind to HIFb to form a heterodimer [70]. Under (larger than acetylation or methylation) to the protein [86].
hypoxic conditions, this heterodimer form an active complex which Fumarate acts as electrophile and interacts with cysteine residues
acts on DNA sites of genes of interest. These genes in turn regulate of target proteins to generate S-(2-succino) cysteine [67,87]. These
biological processes such as cell survival, angiogenesis, cell growth, modied succinated proteins regulate various cellular responses
proliferation and glycolysis [71]. including activation of mitochondrial aconitase activity; inhibit
The HIF prolyl hydroxylases (PHDs) are central regulators for GAPDH (glyceraldehyde 3-phosphate dehydrogenase) activity,
molecular responses to oxygen availability [67]. PHD3 is an isoform activation of antioxidant pathway, and stimulation of the ROS sig-
of PHD, and is expressed in response to hypoxia. This enzyme also nalling pathway [37,88]. These cellular responses in turn regulate
causes apoptosis in neural cells in oxygenated conditions [72]. It cell survival and proliferation [37,88].
was demonstrated that succinate accumulation leads to inactiva- Accumulated of fumarate due to mutation of FH reacts with
tion of the prolyl hydroxylase domain of PHD3. Inactivation of glutathione and produces the proto-oncometabolite, succinated
PHD3 in turn reduces neuronal apoptosis [73]. It was also suggested glutathione. Succinated glutathione is a novel cancer metabolite
that failure in switching on apoptosis can contribute to the path- and can mimic the glutathione substrate [88]. Therefore, it can react
ogenesis of phaeochromocytoma/paraganglioma [73]. with glutathione reductase [88]. As a result, it consumes NADPH
G protein-coupled receptors, also called seven-transmembrane without the resultant antioxidant payoffs. Thus, FH impaired/de-
domain receptors are the largest receptor family in humans and cient cells have decreased levels of NADPH; this favours production
are involved in a number of signal transduction pathways [74]. They of excessive levels of reactive oxygen species [88]. These ROS
regulate various signalling pathways and play a crucial role in further activate cancer metabolism in cells. Reduced FH activity
different physiological processes as well as in tumour growth and leads to HIFa stabilization. Stabilized HIFa in FH mutant cells
metastasis [67]. Elevated concentration of succinate modulates G caused a metabolic shift to glycolysis from oxidative phosphoryla-
protein coupled metabolic receptor (GPR91) and stimulates the tion [89]. Thus, stabled HIFa could promote glycolysis in cancer
release of VEGF (Vascular endothelial growth factor). This VEGF in cells [89].
turn activates various subfamilies of growth factors and/or mito- Glutaminolysis (lysis of glutamine), is a biochemical process in
gens mediating kinase signalling pathways such as ERK1/2, JNK and which the amino acid glutamine is broken down to glutamate,
p38 MAPK etc. [75]. These pathways are involved in various bio- aspartate, carbon dioxide, pyruvate, lactate, alanine and citrate [23].
logical processes responsible for proliferation, oncogenesis, angio- Glutamine which is present in higher concentration than other
genesis, apoptosis and cell cycle kinetics [76e78]. Thus, genetic amino acids serves as a major bio-energetic substrate and nitrogen
alteration in genes encoding the SDH complex leads to impaired donor for cell proliferation [90]. Increase in glutaminolysis helps
function of the enzyme complex and causes excess accumulation of cell growth in FH decient cells [91]. Thus the alteration of primary
succinate. This accumulated succinate has the potential to trigger metabolism in FH decient cells contributes towards further
cancer pathogenesis and progression. oncogenic transformation. A detailed understanding of this inter-
relationship is not yet available and further research is needed [91].
4.5. Fumarate hydratase (FH) Increased accumulation of fumarate promotes the succination of
Kelch-like ECH-associated protein 1, which in turn helps stabiliza-
Fumarate is produced from succinate by the enzyme avopro- tion the nuclear factor like 2 (NrF2) [92,93]. Accumulation of NrF2
tein coupled succinate dehydrogenase [23]. Fumarate hydratase leads to the activation of various antioxidant signalling pathways.
(FH) converts fumarate to malate in the TCA cycle, an irreversible Thus, NrF2 helps FH mutant cells to tolerate high levels of oxidative
reaction. Mutation in gene encoding FH leads to abnormal accu- stress. This oxidative damage further promotes the pathogenesis of
mulation of fumarate in cells [79]. This increased fumarate can act cancer [91]. Thus, genetic alterations in the gene encoding FH
as oncometabolite and promote cancer. FH mutation was rst re- caused excess accumulation of fumarate which has the potential to
ported in the literature by Tomlinson and co-workers in renal cell act as an oncometabolite. The increased concentration of fumarate
cancer and hereditary leiomyomatosis [80]. in cells allows HIF stabilization, switches on glycolysis, causes post-
Reduced FH activity leads to HIFa stabilization. Stabilized HIFa translational modication of protein by succination, generates
in FH mutant cells caused a metabolic shift to glycolysis from succinated glutathione proto-oncometabolites and activates nu-
oxidative phosphorylation [81]. Accumulated fumarate competi- clear factors. These altered phenomena can activate different bio-
tively inhibits 2-oxoglutarate oxygenases, prolyl hydroxylase logical processes which in turn promote cancer pathogenesis.
domain and particularly the hypoxia inducible factor. These
competitive inhibitions produce hypoxia in the tissue microenvi- 5. Oncometabolites of the TCA cycle
ronment (actually they produce pseudohypoxia) and stabilize the
HIF complex. The HIF complex then potentially activates its onco- Oncometabolites are small biomolecules (or enantiomers) of
genic targets and promotes cell growth and proliferation [82]. Also, normal metabolism. Excessive accumulation of them causes
fumarate stimulates mRNA expression of HIF-1a through a non- metabolic dysregulation [91]. Consequently, cells with high onco-
canonical NF-kB-dependent pathway [83]. metabolite concentrations undergo progression to cancer [91].
FH mutation affects many proteins by the process of succination Oncometabolites of the TCA cycle are accumulated due to
K. Sajnani et al. / Biochimie 135 (2017) 164e172 169

Table 1
Abnormalities of TCA cycle enzymes and metabolites in cancer.

Enzymes/TCA cycle product Altered Phenomena Cancer Type References

Mutated Enzymes
Actionase 2 Reduced expression Prostate carcinoma [39]
Loss of function Hereditary leiomyamtosis [37]
Renal cell carcinoma
Down regulation Gastric adenocarcinoma [38]
Isocitrate dehydrogenase Loss of heterozygosity and altered function Malignant glioma [41]
Point mutation and neomorphic function Leukaemia, Glioma [42]
Chondrosarcoma, intrahepatic cholangiocarcinoma
Loss of heterozygosity Glioblastoma [106]
Loss of heterozygosity Acute myeloid leukaemia [107]
Somatic mutation Glioblastoma [52]
Site specic mutation Biliary tract carcinoma and Cholongiocarcinoma [109]
Somatic mutations Colon cancer [116]
Missense somatic mutations Prostate carcinoma [117]
Missense somatic mutations Acute lymphoblastic leukaemia [117]
Succinate dehydrogenase Loss of function mutation Paraganglioma and Pheochromocytoma [67]
Germline mutations Renal carcinoma [118]
Germline mutations Gastrointestinal stromal tumour [119]
Low expression Breast cancer [120]
Fumarate hydratase Germline mutations Hereditary leiomyamtosis and renal cell carcinoma [80]
Heterozygous germline mutations Paraganglioma and Pheochromocytoma [44]
Chromosome deletion Neuroblastoma [121]
Reduced expression Clear cell carcinoma [89]
Downregulation Glioblastoma [122]
Oncometabolites
Citrate Synthase Loss of function Cervical carcinoma [33]
Overexpression Ovarian adenocarcinoma [34]
2-hydroxy glutarate Increased accumulation Leukaemia, [42]
Chondrosarcoma, Intrahepatic
Cholangiocarcinoma
Excess accumulation Chondroscaroma/chondroma [108]
Excess accumulation Biliary tract carcinoma and cholangiocarcinoma [109]

mutations of genes involved in metabolism. Fig. 2 shows different transformed lipid metabolism observed in cancer cell shifts the
oncometabolites of the TCA cycle and enzymes which lead to the metabolic fate of lipid synthesis of the cell membrane and changes
formation of these oncometabolites. the redox potential of cells. These alterations promote cellular
The oncometabolites can be divided in two categories - (I) native processes like cell growth, proliferation, cell survival signalling and
oncometabolites that could be accumulated due to mutations (loss differentiation which in turn contributed in the tumorigenesis [99].
of function) of genes encoding the enzymes, and (ii) promiscuous High energy demand is one of the characteristic features of
oncometabolites that could be accumulated due to gain-of-function cancer cells [100]. Accumulated citrate in cancer cells stimulate
mutations in metabolic enzymes [91]. Table 1 presents the abnor- increased b oxidation of fatty acids and produced more energy, thus
malities/altered phenomena of TCA-cycle enzymes and interme- providing sustaining energy for the cancer cells. This energy further
diate products associated with different cancers. The major promotes proliferation of cancer cells [101].
oncometabolites in cancer pathogenesis are discussed in the Histones are the basic and principal proteins that regulate the
following sections. structural and functional organization of chromatin and also
regulate gene expressions [102]. These proteins undergo different
5.1. Citrate types of epigenetic and genetic modication. Acetylation of his-
tones emerges as a central regulator, which permits the inter-
Citrate accumulation in cancer environments will give rise to conversion of permissive and repressive chromatin structure in
epigenetic and non-epigenetic modications which could promote regards of transcriptional aptitudes [103]. Acetylation of histones is
the development and progression of cancer [94]. Excess citrate signicantly controlled by several classes of enzymes such as his-
reduces the activity of the mitochondrial isoform of pyruvate de- tone deacetylases and histones acetyltransferases. Both of these
hydrogenase and results in a shift of the cell's metabolism towards enzymes regulate specic genes transcription by targeting specic
glycolysis [94]. Increased accumulation of pyruvate leads cells to regions of the chromatin [101]. Excess acetyl CoA produced by
convert pyruvate to lactate and regenerate NAD which is the key accumulated citrate encourages acetyl CoA dependent acetylation
factor for glycolysis [94] (Fig. 1). Thus citrate accumulation further of histone at N-terminal lysine residues [73,104,105]. Acetylation of
favours the non-oxidative breakdown of glucose in the cells and lysine residues modies the charge ratio of the protein, which
promotes cancer growth. controls a numbers of proteins that are involved in cell metabolism,
Lipids are a class of bio-molecule with long hydrocarbon chains. cell kinetics, aging, angiogenesis, growth and development [73].
The main biological function of lipids includes storing energy, sig-
nalling and formation of membrane structure [95]. The key carbon 5.2. 2-Hydroxy glutarate (2HG)
precursor for lipid synthesis is acetyl CoA [96]. Increased accumu-
lation of citrate activates the enzyme acetyl CoA carboxylase (ACC), Mutations of IDH1 and IDH2 (cytosolic and mitochondrial iso-
which in turn increases the production of acetyl CoA and malonyl forms of IDH) lead to the accumulation of 2HG and acts as a
CoA [97,98]. This increased acetyl CoA and malonyl CoA is then pathogenic metabolite (oncometabolite) in many cancers
directed towards increased lipid/sterol synthesis [97,98]. The [42,52,106e110]. Dang and co-workers demonstrated that a point
170 K. Sajnani et al. / Biochimie 135 (2017) 164e172

mutation (replacing arginine 132 with histidine) at the active site of neck squamous cell carcinoma, Mitochondrion 24 (2015) 113e121.
[13] B.N. Nicolay, P.S. Danielian, F. Kottakis, J.D. Lapek Jr., I. Sanidas, W.O. Miles,
IDH1 (an isoform of IDH) was noted in primary glioma. They also p, J.J. Gierut, A.L. Manning, R. Morris, K. Haigis,
M. Dehnad, K. Tscho
reported that this mutation was involved in the conversion of N. Bardeesy, J.A. Lees, W. Haas, N.J. Dyson, Proteomic analysis of pRb loss
alpha-ketuglutarates to R (-)-2-hydroxyglutarates (2HGs) [41]. highlights a signature of decreased mitochondrial oxidative phosphoryla-
Excessive accumulation of these 2HGs acts as an oncometabolite tion, Genes Dev. 29 (2015) 1875e1889.
[14] C. Frezza, P.J. Pollard, E. Gottlieb, Inborn and acquired metabolic defects in
and led to the malignant progression of gliomas [41] (Fig. 1). cancer, J. Mol. Med. Berl. 89 (2011) 213e220.
DNA methylation in vertebrates acts as an epigenetic modi- [15] P.L. Pedersen, Tumor mitochondria and the bioenergetics of cancer cells,
cation event [111]. DNA structure and histone methylation systems Prog. Exp. Tumor Res. 22 (1978) 190e274.
[16] C.S. Lin, L.S. Wang, C.M. Tsai, Y.H. Wei, Low copy number and low oxidative
are highly regulated and rely mechanistically on each other for damage of mitochondrial DNA are associated with tumor progression in lung
chromatin function [111]. Alterations in DNA methylation patterns, cancer tissues after neoadjuvant chemotherapy, Interact. Cardiovasc Thorac.
such as hyper-methylation of CpG islands, are common changes Surg. 7 (2008) 954e958.
[17] R.A. LaBiche, M. Demars, G.L. Nicolson, Transcripts of the mitochondrial gene
noted in human cancers [112]. Studies reported that increased ND5 are overexpressed in highly metastatic murine large cell lymphoma
accumulated 2HG has potential tumorigenic activity and promotes cells, In Vivo 6 (1992) 317e324.
cancer growth by inhibiting the activity of DNA demethylases and [18] N. Glaichenhaus, P. Leopold, CuzinF. Increased levels of mitochondrial gene
expression in rat broblast cells immortalized or transformed by viral and
the ten-eleven translocation- 2 family of DNA demethylases cellular oncogenes, EMBO J. 5 (1986) 1261e1265.
[113,114]. Another study also demonstrated that IDH mutations [19] N. Raimundo, B.E. Baysal, G.S. Shadel, Revisiting the TCA cycle: signaling to
block histone N-lysine demethylases jumonji domain containing tumor formation, Trends Mol. Med. 17 (2011) 641e649.
[20] J.A. Menendez, T. Alarco n, J. Joven, Gerometabolites: the pseudohypoxic
2A (JMJD2A) [114].
aging side of cancer oncometabolites, Cell Cycle 13 (2014) 699e709.
HIF, the major regulator of hypoxia is a heterodimer comprising [21] C. Frezza, E. Gottlieb, Mitochondria in cancer: not just innocent bystanders,
of alpha and b subunits [115]. Subunit a is degraded in presence of Semin. Cancer Biol. 19 (2009) 4e11.
oxygen by enzyme prolyl hydroxylases [19]. It was demonstrated [22] E. Gaude, C. Frezza, Defects in mitochondrial metabolism and cancer, Cancer
Metab. 2 (2014) 10.
that 2HG stabilized HIF alpha by acting as a competitive inhibitor of [23] Nelson and Cox, Lehninger Principles of Biochemsitry, New York, United
the prolyl hydroxylase enzyme [58]. States of America, W H Freeman, 2004.
[24] B.L. Fatland, J. Ke, M.D. Anderson, W.I. Mentzen, L.W. Cui, C.C. Allred,
J.L. Johnston, B.J. Nikolau, WurteleES.Molecular characterization of a het-
6. Concluding remarks eromeric ATP-citrate lyase that generates cytosolic acetyl-coenzyme A in
arabidopsis, Plant Physiol. 130 (2002) 740e756.
Genetic alterations in the TCA cycle as well as the oncometa- [25] C. Yuan, C.B. Clish, C. Wu, J.R. Mayers, P. Kraft, M.K. Townsend, M. Zhang,
S.S. Tworoger, Y. Bao, Z.R. Qian, D.A. Rubinson, K. Ng, E.L. Giovannucci,
bolites generated are important in the pathogenesis of many can- S. Ogino, M.J. Stampfer, J.M. Gaziano, J. Ma, H.D. Sesso, G.L. Anderson,
cers. From the growing body of knowledge on the subject, it is clear B.B. Cochrane, J.E. Manson, M.E. Torrence, A.C. Kimmelman,
that research in the eld of oncometabolite is a promising area for L.T. Amundadottir, M.G. Vander Heiden, C.S. Fuchs, B.M. Wolpin, Circulating
metabolites and survival among patients with pancreatic cancer, J. Natl.
scientists to develop targeted therapies for cancer. The exact Cancer Inst. (2016) 108 djv409.
mechanisms of oncometabolites such as malate and oxaloacetate, [26] N.N. Pavlova, C.B. Thompson, The emerging hallmarks of cancer metabolism,
etc. leading to cancer are not clear. Therefore, investigation in this Cell Metab. 23 (2016) 27e47.
[27] E.D. Montal, R. Dewi, K. Bhalla, L. Ou, B.J. Hwang, A.E. Ropell, C. Gordon,
eld in future will aid in improving our understanding of the role of W.J. Liu, R.J. DeBerardinis, J. Sudderth, W. Twaddel, L.G. Boros, K.R. Shroyer,
the TCA cycle enzymes and oncometabolites in the pathogenesis S. Duraisamy, R. Drapkin, R.S. Powers, J.M. Rohde, M.B. Boxer, K.K. Wong,
and development of cancer. G.D. Girnun, PEPCK coordinates the regulation of central carbon metabolism
to promote cancer cell growth, Mol. Cell 60 (2015) 571e583.
[28] E. Desideri, R. Vegliante, M.R. Ciriolo, Mitochondrial dysfunctions in cancer:
Acknowledgements genetic defects and oncogenic signaling impinging on TCA cycle activity,
Cancer Lett. 356 (2015) 217e223.
[29] X. Lu, B. Bennet, E. Mu, J. Rabinowitz, Y. Kang, Metabolomic changes
The project was supported by the student scholarship from accompanying transformation and acquisition of metastatic potential in a
Grifth University and funding from School of Medical Science and syngeneic mouse mammary tumor model, J. Biol. Chem. 285 (2010)
Menzies Health Institute Queensland. 9317e9321.
[30] C.S. Ahn, C.M. Metallo, Mitochondria as biosynthetic factories for cancer
proliferation, Cancer Metab. 3 (2015) 1.
References [31] J.P. Harrelson, M.W. Lee, Expanding the view of breast cancer metabolism:
promising molecular targets and therapeutic opportunities, Pharmacol. Ther.
[1] M. Rogalinska, The role of mitochondria in cancer induction, progression and 167 (2016) 60e73.
changes in metabolism, Mini Rev. Med. Chem. 16 (2016) 524e530. [32] S. Cardaci, M.R. Ciriolo, TCA cycle defects and cancer: when metabolism
[2] D.R. Green, Means to an End: Apoptosis and Other Cell Death Mechanisms, tunes redox state, Int. J. Cell Biol. 2012 (2012) 161837.
Cold Spring Harbor Laboratory Press, 2011. [33] C.C. Lin, T.L. Cheng, W.H. Tsai, H.J. Tsai, K.H. Hu, H.C. Chang, C.W. Yeh,
[3] A. Szewczyk, L. Wojtczak, Mitochondria as a pharmacological target, Phar- Y.C. Chen, C.C. Liao, W.T. Chang, Loss of the respiratory enzyme citrate
macol. Rev. 54 (2002) 101e127. synthase directly links the Warburg effect to tumor malignancy, Sci. Rep. 2
[4] J.M. Breg, J.L. Tymoczko, G.J. Gatto Jr., L. Stryer, Biochemistry, New York, (2012) 785.
United States of America, W H Freeman, 2015. [34] L. Chen, T. Liu, J. Zhou, Y. Wang, X. Wang, W. Di, S. Zhang, Citrate synthase
[5] O. Warburg, On the origin of the Cancer cells, Science 123 (1956) 309e314. expression affects tumor phenotype and drug resistance in human ovarian
[6] A. Vazquez, J. Liu, Y. Zhou, OltvaiZN. Catabolic efciency of aerobic glycolysis: carcinoma, PLoS One 9 (2014) e115708.
the Warburg effect revisited, BMC Syst. Biol. 4 (2010) 58. [35] B. Schlichtholz, J. Turyn, E. Goyke, M. Biernacki, K. Jaskiewicz, Z. Sledzinski,
[7] P.P. Hsu, D.M. Sabatini, Cancer cell metabolism: Warburg and beyond, Cell J. Swierczynski, Enhanced citrate synthase activity in human pancreatic
134 (2008) 703e707. cancer, Pancreas 30 (2005) 99e104.
[8] S. Nowicki, E. Gottlieb, Oncometabolites: tailoring our genes, FEBS J. 282 [36] H. Simonnet, N. Alazard, K. Pfeiffer, C. Gallou, C. Be roud, J. Demont,
(2015) 2796e2805. R. Bouvier, H. Sch agger, C. Godinot, Low mitochondrial respiratory chain
[9] H. Nam, M. Campodonico, A. Bordbar, D.R. Hyduke, S. Kim, D.C. Zielinski, et content correlates with tumor aggressiveness in renal cell carcinoma,
al., A systems approach to predict oncometabolites via context-specic Carcinogenesis 23 (2002) 759e768.
genome-scale metabolic networks, PLoS Comput. Biol. 10 (2014) e1003837. [37] N. Ternette, M. Yang, M. Laroyia, M. Kitagawa, L. O'Flaherty, K. Wolhulter,
[10] D.C. Wallace, The mitochondrial genome in human adaptive radiation and K. Igarashi, K. Saito, K. Kato, R. Fischer, A. Berquand, B.M. Kessler, T. Lappin,
disease: on the road to therapeutics and performance enhancement, Gene N. Frizzell, T. Soga, J. Adam, P.J. Pollard, Inhibition of mitochondrial aconitase
354 (2005) 169e180. by succination in fumarate hydratase deciency, Cell Rep. 3 (2013) 689e700.
[11] M.G. Vander Heiden, L.C. Cantley, C.B. Thompson, Understanding the War- [38] P. Wang, C. Mai, Y.L. Wei, J.J. Zhao, Y.M. Hu, Z.L. Zeng, J. Yang, W.H. Lu,
burg effect: the metabolic requirements of cell proliferation, Science 324 R.H. Xu, P. Huang, Decreased expression of the mitochondrial metabolic
(2009) 1029e1033. enzyme aconitase (ACO2) is associated with poor prognosis in gastric cancer,
[12] E.C. Koc, E. Haciosmanoglu, P.P. Claudio, A. Wolf, L. Califano, M. Friscia, Med. Oncol. 30 (2013) 552.
A. Cortese, H. Koc, Impaired mitochondrial protein synthesis in head and [39] K.K. Singh, M.M. Desouki, R.B. Franklin, L.C. Costello, Mitochondrialaconitase
K. Sajnani et al. / Biochimie 135 (2017) 164e172 171

and citrate metabolism in malignant and non-malignant human prostate [64] A.H. Shih, R.L. Levine, IDH1 mutations disrupt blood, brain, and barriers,
tissues, Mol. Cancer 5 (2006) 14. Cancer Cell 22 (2012) 285e287.
[40] J. Balss, J. Meyer, W. Mueller, A. Korshunov, C. Hartmann, A. von Deimling, [65] M. Sasaki, C.B. Knobbe, M. Itsumi, A.J. Elia, I.S. Harris, Chio II, R.A. Cairns,
Analysis of the IDH1 codon 132 mutation in brain tumors, Acta Neuropathol. S. McCracken, A. Wakeham, J. Haight, A.Y. Ten, B. Snow, T. Ueda, S. Inoue,
116 (2008) 597e602. K. Yamamoto, M. Ko, A. Rao, K.E. Yen, S.M. Su, T.W. Mak, D-2-
[41] L. Dang, D.W. White, S. Gross, B.D. Bennett, M.A. Bittinger, E.M. Driggers, hydroxyglutarate produced by mutant IDH1 perturbs collagen maturation
V.R. Fantin, H.G. Jang, S. Jin, M.C. Keenan, K.M. Marks, R.M. Prins, P.S. Ward, and basement membrane function, Genes Dev. 26 (2012) 2038e2049.
K.E. Yen, L.M. Liau, J.D. Rabinowitz, L.C. Cantley, C.B. Thompson, M.G. Vander [66] M. Sasaki, C.B. Knobbe, J.C. Munger, E.F. Lind, D. Brenner, A. Brstle,
Heiden, S.M. Su, Cancer-associated IDH1 mutations produce 2- I.S. Harris, R. Holmes, A. Wakeham, J. Haight, A. You-Ten, W.Y. Li, S. Schalm,
hydroxyglutarate, Nature 462 (2009) 739e744. S.M. Su, C. Virtanen, G. Reifenberger, P.S. Ohashi, D.L. Barber, M.E. Figueroa,
[42] S.J. Parker, C.M. Metallo, Metabolic consequences of oncogenic IDH muta- A. Melnick, J.C. Zn ~ iga-Pcker, T.W. Mak, IDH1 (R132H) mutation increases
tions, PharmacolTher 152 (2015) 54e62. murine haematopoietic progenitors and alters epigenetics, Nature 488
[43] W. Xu, H. Yang, Y. Liu, Y. Yang, P. Wang, S.H. Kim, S. Ito, C. Yang, P. Wang, (2012) 656e659.
M.T. Xiao, L.X. Liu, W.Q. Jiang, J. Liu, J.Y. Zhang, B. Wang, S. Frye, Y. Zhang, [67] C. Bardella, P.J. Pollard, I. Tomlinson, SDH mutations in cancer, Biochim.
Y.H. Xu, Q.Y. Lei, K.L. Guan, S.M. Zhao, Y. Xiong, Oncometabolite 2- Biophys. Acta 2011 (1807) 1432e1443.
hydroxyglutarate is a competitive inhibitor of a-ketoglutarate-dependent [68] A.K. Lam, Update on paragangliomas and pheochromocytomas, Turk Patoloji
dioxygenases, Cancer Cell 19 (2011) 17e30. Derg. 31 (S1) (2015) 105e112.
[44] E. Letouze , C. Martinelli, C. Loriot, N. Burnichon, N. Abermil, C. Ottolenghi, [69] B.G. Slane, N. Aykin-Burns, B.J. Smith, A.L. Kalen, P.C. Goswami, F.E. Domann,
M. Janin, M. Menara, A.T. Nguyen, P. Benit, A. Buffet, C. Marcaillou, D.R. Spitz, Mutation of succinate dehydrogenase subunit C results in
J. Bertherat, L. Amar, P. Rustin, A. De Reynie s, A.P. Gimenez-Roqueplo, increased O2.-, oxidative stress, and genomic instability, Cancer Res. 66
J. Favier, SDH mutations establish a hypermethylator phenotype in para- (2006) 7615e7620.
ganglioma, Cancer Cell. 23 (2013) 739e752. [70] M.A. Selak, S.M. Armour, E.D. MacKenzie, H. Boulahbel, D.G. Watson,
[45] G. Matlashewski, P. Lamb, D. Pim, J. Peacock, L. Crawford, S. Benchimol, K.D. Manseld, Y. Pan, M.C. Simon, C.B. Thompson, E. Gottlieb, Succinate links
Isolation and characterization of a human p53 cDNA clone: expression of the TCA cycle dysfunction to oncogenesis by inhibiting HIF-alpha prolyl hy-
human p53 gene, EMBO J. 3 (1984) 3257e3262. droxylase, Cancer Cell 7 (2005) 77e85.
[46] M. Isobe, B.S. Emanuel, D. Givol, M. Oren, C.M. Croce, Localization of gene for [71] W.G. Kaelin Jr., P.J. Ratcliffe, Oxygen sensing by metazoans: the central role of
human p53 tumour antigen to band 17p13, Nature 320 (1986) 84e85. the HIF hydroxylase pathway, Mol. Cell 30 (2008) 393e402.
[47] J.C. Bourdon, K. Fernandes, F. Murray-Zmijewski, G. Liu, A. Diot, [72] K. Rantanen, J. Pursiheimo, H. Ho gel, V. Himanen, E. Metzen, P.M. Jaakkola,
D.P. Xirodimas, M.K. Saville, D.P. Lane, p53 isoforms can regulate p53 tran- Prolyl hydroxylase PHD3 activates oxygen-dependent protein aggregation,
scriptional activity, Genes Dev. 19 (2005) 2122e2137. Mol. Biol. Cell 19 (2008) 2231e2240.
[48] K.Y. Lam, S.W. Tsao, D. Zhang, S. Law, D. He, L. Ma, J. Wong, Prevalence and [73] S. Lee, Post-translational modication of proteins in toxicological research:
predictive value of p53 mutation in patients with esophageal squamous cell focus on lysine acylation, Toxicol. Res. 29 (2013) 81e86.
carcinomas: a prospective clinicopatholgical study and survival analysis of [74] B. Trzaskowski, D. Latek, S. Yuan, U. Ghoshdastider, A. Debinski, S. Filipek,
70 patients, Int. J. Cancer 74 (1997) 212e219. Action of molecular switches in GPCRsetheoretical and experimental
[49] K.Y. Lam, C.Y. Lo, K.W. Chan, K.Y. Wan, Insular and anaplastic carcinoma of studies, Curr. Med. Chem. 19 (2012) 1090e1109.
the thyroid: a 45-year comparative study at a single institution and a review [75] J. Hu, T. Li, S. Du, Y. Chen, S. Wang, F. Xiong, Q. Wu, The MAPK signaling
of the signicance of p53 and p21, Ann. Surg. 231 (2000) 329e338. pathway mediates the GPR91-dependent release of VEGF from RGC-5 cells,
[50] K.Y. Lam, C.Y. Lo, N.M.S. Wat, J.M. Luk, K.S.L. Lam, The clinicopathological Int. J. Mol. Med. 36 (2015) 130e138.
features and importance of p53, Rb and mdm2 expression in phaeochro- [76] S. Yoon, SegerR, The extracellular signal-regulated kinase: multiple sub-
mocytomas and paragangliomas, J. Clin. Pathol. 54 (2001) 443e448. strates regulate diverse cellular functions, Growth Factors. 4 (2006) 21e24.
[51] K. Boslooper, A.K.Y. Lam, J. Gao, S. Weinstein, N. Johnson, The clinicopatho- [77] H. Nakabayashi, K. Shimizu, HA1077, a Rho kinase inhibitor, suppresses
logical roles of alpha-B-crystallin and p53 expression in patients with head glioma-induced angiogenesis by targeting the Rho-ROCK and the mitogen-
and neck squamous cell carcinoma, Pathology 40 (2008) 500e504. activated protein kinase kinase/extracellular signal-regulated kinase (MEK/
[52] H. Yan, D.W. Parsons, G. Jin, R. McLendon, B.A. Rasheed, W. Yuan, I. Kos, ERK) signal pathways, Cancer Sci. 102 (2011) 393e399.
I. Batinic-Haberle, S. Jones, G.J. Riggins, H. Friedman, A. Friedman, D. Reardon, [78] D.N. Dhanasekaran, G.L. Johnson, MAPKs: function, regulation, role in cancer
J. Herndon, K.W. Kinzler, V.E. Velculescu, B. Vogelstein, D.D. Bigner, IDH1 and and therapeutic targeting, Oncogene 26 (2007) 3097e3099.
IDH2 mutations in gliomas, N. Engl. J. Med. 360 (2009) 765e773. [79] A. Morin, E. Letouze , A.P. Gimenez-Roqueplo, J. Favier, Oncometabolites-
[53] T. Watanabe, S. Nobusawa, P. Kleihues, H. Ohgaki, IDH1 mutations are early driven tumorigenesis: from genetics to targeted therapy, Int. J. Cancer 135
events in the development of astrocytomas and oligodendrogliomas, Am. J. (2014) 2237e2248.
Pathol. 174 (2009) 1149e1153. [80] I.P. Tomlinson, N.A. Alam, A.J. Rowan, E. Barclay, E.E. Jaeger, D. Kelsell,
[54] A. Carnero, C. Blanco-Aparicio, O. Renner, W. Link, J.F. Leal, The PTEN/PI3K/ I. Leigh, P. Gorman, H. Lamlum, S. Rahman, R.R. Roylance, S. Olpin, S. Bevan,
AKT signalling pathway in cancer, therapeutic implications, Curr. Cancer K. Barker, N. Hearle, R.S. Houlston, M. Kiuru, R. Lehtonen, A. Karhu, S. Vilkki,
Drug Targets 8 (2008) 187e198. P. Laiho, C. Eklund, O. Vierimaa, K. Aittoma ki, M. Hietala, P. Sistonen,
[55] A. Carracedo, L. Ma, J. Teruya-Feldstein, F. Rojo, L. Salmena, A. Alimonti, A. Paetau, R. Salovaara, R. Herva, V. Launonen, L.A. Aaltonen, Multiple Leio-
A. Egia, A.T. Sasaki, G. Thomas, S.C. Kozma, A. Papa, C. Nardella, L.C. Cantley, myoma Consortium. Germline mutations in FH predispose to dominantly
J. Baselga, P.P. Pandol, Inhibition of mTORC1 leads to MAPK pathway acti- inherited uterine broids, skin leiomyomata and papillary renal cell cancer,
vation through a PI3K-dependent feedback loop in human cancer, J. Clin. Nat. Genet. 30 (2002) 406e410.
Invest 118 (2008) 3065e3074. [81] S. Sudarshan, C. Sourbier, H.S. Kong, K. Block, V.A. Valera Romero, Y. Yang,
[56] EngelmanJA, Targeting PI3K signalling in cancer: opportunities, challenges C. Galindo, M. Mollapour, B. Scroggins, N. Goode, M.J. Lee, C.W. Gourlay,
and limitations, Nat. Rev. Cancer 9 (2009) 550e562. J. Trepel, W.M. Linehan, L. Neckers, Fumarate hydratase deciency in renal
[57] J. Shi, H. Zuo, L. Ni, L. Xia, L. Zhao, M. Gong, D. Nie, P. Gong, D. Cui, W. Shi, cancer induces glycolytic addiction and hypoxia-inducible transcription
J. Chen, An IDH1 mutation inhibits growth of glioma cells via GSH depletion factor 1 alpha stabilization by glucose-dependent generation of reactive
and ROS generation, NeurolSci 35 (2014) 839e845. oxygen species, Mol. Cell Biol. 29 (2009) 4080e4090.
[58] Z.J. Reitman, H. Yan, Isocitrate dehydrogenase 1 and 2 mutations in cancer: [82] J.S. Isaacs, Y.J. Jung, D.R. Mole, S. Lee, C. Torres-Cabala, Y.L. Chung, M. Merino,
alterations at a crossroads of cellular metabolism, J. Natl. Cancer Inst. 102 J. Trepel, B. Zbar, J. Toro, P.J. Ratcliffe, W.M. Linehan, L. Neckers, HIF over-
(2010) 932e941. expression correlates with biallelic loss of fumarate hydratase in renal can-
[59] M.P. Murphy, How mitochondria produce reactive oxygen species, Biochem. cer: novel role of fumarate in regulation of HIF stability, Cancer Cell 8 (2005)
J. 417 (2009) 1e13. 143e153.
[60] D. Trachootham, J. Alexandre, P. Huang, Targeting cancer cells by ROS- [83] K. Shanmugasundaram, B. Nayak, E.H. Shim, C.B. Livi, K. Block, S. Sudarshan,
mediated mechanisms: a radical therapeutic approach? Nat. Rev. Drug Dis- The oncometabolite fumarate promotes pseudohypoxia through noncanon-
cov. 8 (2009) 579e591. ical activation of NF-kappaBsignaling, J. Biol. Chem. 289 (2014)
[61] D.R. Wise, P.S. Ward, J.E. Shay, J.R. Cross, J.J. Gruber, U.M. Sachdeva, J.M. Platt, 24691e24699.
R.G. DeMatteo, M.C. Simon, C.B. Thompson, Hypoxia promotes isocitrate [84] M. Yang, N. Ternette, H. Su, R. Dabiri, B.M. Kessler, J. Adam, B.T. Teh,
dehydrogenase-dependent carboxylation of a-ketoglutarate to citrate to P.J. Pollard, The succinated proteome of FH-mutant tumours, Metabolites 4
support cell growth and viability, Proc. Natl. AcadSci U. S. A. 108 (2011) (2014) 640e654.
19611e19616. [85] Z. Xie, J. Dai, L. Dai, M. Tan, Z. Cheng, Y. Wu, J.D. Boeke, Y. Zhao, Lysine
[62] S. Gross, R.A. Cairns, M.D. Minden, E.M. Driggers, M.A. Bittinger, H.G. Jang, succinylation and lysine malonylation in histones, Mol. Cell Proteom. 11
M. Sasaki, S. Jin, D.P. Schenkein, S.M. Su, L. Dang, V.R. Fantin, Mak.Cancer- (2012) 100e107.
associated metabolite 2-hydroxyglutarate accumulates in acute myeloge- [86] Z. Zhang, M. Tan, Z. Xie, L. Dai, Y. Chen, Y. Zhao, Identication of lysine
nous leukemia with isocitrate dehydrogenase 1 and 2 mutations, J. Exp. Med. succinylation as a new post-translational modication, Nat. Chem. Biol. 7
207 (2010) 339e344. (2011) 58e63.
[63] G.A. Di-Lullo, S.M. Sweeney, J. Korkko, L. Ala-Kokko, J.D. San Antonio, Map- [87] N.L. Alderson, Y. Wang, M. Blatnik, N. Frizzell, M.D. Walla, T.J. Lyons, N. Alt,
ping the ligand-binding sites and disease-associated mutations on the most J.A. Carson, R. Nagai, S.R. Thorpe, J.W. Baynes, S-(2-Succinyl) cysteine: a novel
abundant protein in the human, type I collagen, J. Biol. Chem 277 (2002) chemical modication of tissue proteins by a Krebs cycle intermediate, Arch.
4223e4231. Biochem. Biophys. 450 (2006) 1e8.
172 K. Sajnani et al. / Biochimie 135 (2017) 164e172

[88] L.B. Sullivan, E. Martinez-Garcia, H. Nguyen, A.R. Mullen, E. Dufour, R.E. Ries, J.E. Payton, P. Westervelt, M.H. Tomasson, M.A. Watson, J. Baty,
S. Sudarshan, et al., The proto-oncometabolite fumarate binds glutathione to S. Heath, W.D. Shannon, R. Nagarajan, D.C. Link, M.J. Walter, T.A. Graubert,
amplify ROS-dependent signaling, Mol. Cell 51 (2013) 236e248. J.F. DiPersio, R.K. Wilson, T.J. Ley, Recurring mutations found by sequencing
[89] S. Sudarshan, K. Shanmugasundaram, S.L. Naylor, S. Lin, C.B. Livi, C.F. O'Neill, an acute myeloid leukemia genome, N. Engl. J. Med. 361 (2009) 1058e1066.
D.J. Parekh, I.T. Yeh, L.Z. Sun, K. Block, Reduced expression of fumarate [108] M.F. Amary, K. Bacsi, F. Maggiani, S. Damato, D. Halai, F. Berisha, R. Pollock,
hydratase in clear cell renal cancer mediates HIF-2alpha accumulation and P. O'Donnell, A. Grigoriadis, T. Diss, M. Eskandarpour, N. Presneau,
promotes migration and invasion, PLoS One 6 (2011) e21037. P.C. Hogendoorn, A. Futreal, R. Tirabosco, A.M. Flanagan, IDH1 and IDH2
[90] R.J. DeBerardinis, A. Mancuso, E. Daikhin, I. Nissim, M. Yudkoff, S. Wehrli, mutations are frequent events in central chondrosarcoma and central and
C.B. Thompson, Beyond aerobic glycolysis: transformed cells can engage in periosteal chondromas but not in other mesenchymal tumours, J. Pathol. 224
glutamine metabolism that exceeds the requirement for protein and (2011) 334e343.
nucleotide synthesis, Proc. Natl. Acad. Sci. U. S. A. 104 (2007) 19345e19350. [109] D.R. Borger, K.K. Tanabe, K.C. Fan, H.U. Lopez, V.R. Fantin, K.S. Straley,
[91] M. Yang, T. Soga, P.J. Pollard, J. Adam, The emerging role of fumarate as an D.P. Schenkein, A.F. Hezel, M. Ancukiewicz, H.M. Liebman, E.L. Kwak,
oncometabolite, Front. Oncol. 2 (2012) 85. J.W. Clark, D.P. Ryan, V. Deshpande, D. Dias-Santagata, L.W. Ellisen, A.X. Zhu,
[92] J. Adam, E. Hatipoglu, L. O'Flaherty, N. Ternette, N. Sahgal, H. Lockstone, A.J. Iafrate, Frequent mutation of isocitrate dehydrogenase (IDH)1 and IDH2
D. Baban, E. Nye, G.W. Stamp, K. Wolhuter, M. Stevens, R. Fischer, in cholangiocarcinoma identied through broad-based tumor genotyping,
P. Carmeliet, P.H. Maxwell, C.W. Pugh, N. Frizzell, T. Soga, B.M. Kessler, M. El- Oncologist 17 (2012) 72e79.
Bahrawy, P.J. Ratcliffe, P.J. Pollard, Renal cyst formation in Fh1-decient mice [110] P.S. Ward, C.B. Thompson, Metabolic reprogramming: a cancer hallmark
is independent of the Hif/Phd pathway: roles for fumarate in KEAP1 succi- even Warburg did not anticipate, Cancer Cell 21 (2012) 297e308.
nation and Nrf2 signaling, Cancer Cell 20 (2011) 524e537. [111] M. Ehrlich, M.A. Gama-Sosa, L.H. Huang, R.M. Midgett, K.C. Kuo, R.A. McCune,
[93] A. Ooi, J.C. Wong, D. Petillo, D. Roossien, V. Perrier-Trudova, D. Whitten, C. Gehrke, Amount and distribution of 5-methylcytosine in human DNA from
B.W. Min, M.H. Tan, Z. Zhang, X.J. Yang, M. Zhou, B. Gardie, V. Molinie , different types of tissues of cells, Nucleic Acids Res. 10 (1982) 2709e2721.
S. Richard, P.H. Tan, B.T. Teh, FurgeKA.An antioxidant response phenotype [112] Y. Hamai, N. Oue, Y. Mitani, H. Nakayama, R. Ito, K. Matsusaki, K. Yoshida,
shared between hereditary and sporadic type 2 papillary renal cell carci- T. Toge, W. Yasui, DNA hypermethylation and histone hypoacetylation of the
noma, Cancer Cell 20 (2011) 511e523. HLTF gene are associated with reduced expression in gastric carcinoma,
[94] P. Icard, L. Poulain, H. Lincet, Understanding the central role of citrate in the Cancer Sci. 94 (2003) 692e698.
metabolism of cancer cells, Biochim. Biophys. Acta 2012 (1825) 111e116. [113] M.E. Figueroa, O. Abdel-Wahab, C. Lu, P.S. Ward, J. Patel, A. Shih, Y. Li,
[95] E. Fahy, S. Subramaniam, R.C. Murphy, M. Nishijima, C.R. Raetz, T. Shimizu, N. Bhagwat, A. Vasanthakumar, H.F. Fernandez, M.S. Tallman, Z. Sun,
F. Spener, G. van Meer, M.J. Wakelam, Dennis EA.Update of the LIPID MAPS K. Wolniak, J.K. Peeters, W. Liu, S.E. Choe, V.R. Fantin, E. Paietta,
comprehensive classication system for lipids, J. Lipid Res. 50 (2009) s9es14. B. Lo wenberg, J.D. Licht, L.A. Godley, R. Delwel, P.J. Valk, C.B. Thompson,
[96] J. Nielsen, Systems biology of lipid metabolism: from yeast to human, FEBS R.L. Levine, A. Melnick, Leukemic IDH1 and IDH2 mutations result in a
Lett. 583 (2009) 3905e3913. hypermethylation phenotype, disrupt TET2 function, and impair hemato-
[97] L. Tong, Acetyl-coenzyme A carboxylase: crucial metabolic enzyme and poietic differentiation, Cancer Cell 18 (2010) 553e567.
attractive target for drug discovery, Cell Mol. Life Sci. 62 (2005) 1784e1803. [114] R. Chowdhury, K.K. Yeoh, Y.M. Tian, L. Hillringhaus, E.A. Bagg, N.R. Rose,
[98] C.W. Kim, Y.A. Moon, S.W. Park, D. Cheng, H.J. Kwon, J.D. Horton, Induced I.K. Leung, X.S. Li, E.C. Woon, M. Yang, M.A. McDonough, O.N. King, I.J. Clifton,
polymerization of mammalian acetyl-CoA carboxylase by MIG12 provides a R.J. Klose, T.D. Claridge, P.J. Ratcliffe, C.J. Schoeld, A. Kawamura, The onco-
tertiary level of regulation of fatty acid synthesis, Proc. Natl. AcadSci U. S. A. metabolite 2-hydroxyglutarate inhibits histone lysine demethylases, EMBO
107 (2010) 9626e9631. Rep. 12 (2011) 463e469.
[99] C.R. Santos, A. Schulze, Lipid metabolism in cancer, FEBS J. 279 (2012) [115] A.J. Majmundar, W.J. Wong, M.C. Simon, Hypoxia-inducible factors and the
2610e2623. response to hypoxic stress, Mol. Cell 40 (2010) 294e309.
[100] M. Jang, S.S. Kim, J. Lee, Cancer cell metabolism: implications for therapeutic [116] T. Sjoblom, S. Jones, L.D. Wood, D.W. Parsons, J. Lin, T.D. Barber,
targets, ExpMol Med. 45 (2013) e45. D. Mandelker, R.J. Leary, J. Ptak, N. Silliman, S. Szabo, P. Buckhaults, C. Farrell,
[101] P. Icard, H. Lincet, A global view of the biochemical pathways involved in the P. Meeh, S.D. Markowitz, J. Willis, D. Dawson, J.K. Willson, A.F. Gazdar,
regulation of the metabolism of cancer cells, Biochim. Biophys. Acta 2012b J. Hartigan, L. Wu, C. Liu, G. Parmigiani, B.H. Park, K.E. Bachman,
(1826) 423e433. N. Papadopoulos, B. Vogelstein, K.W. Kinzler, V.E. Velculescu, The consensus
[102] L. Marin ~ o-Ramrez, M.G. Kann, B.A. Shoemaker, D. Landsman, Histone coding sequences of human breast and colorectal cancers, Science 314
structure and nucleosome stability, Expert Rev. Proteom. 2 (2005) 719e729. (2006) 268e274.
[103] A. Eberharter, P.B. Becker, Histone acetylation: a switch between repressive [117] M.R. Kang, M.S. Kim, J.E. Oh, Y.R. Kim, S.Y. Song, S.I. Seo, J.Y. Lee, N.J. Yoo,
and permissive chromatin. Second in review series on chromatin dynamics, S.H. Lee, Mutational analysis of IDH1 codon 132 in glioblastomas and other
EMBO Rep. 3 (2002) 224e229. common cancers, Int. J. Cancer 125 (2009) 353e355.
[104] B. Polevoda, F. Sherman, N-terminal acetyltransferases and sequence re- [118] C. Ricketts, E.R. Woodward, P. Killick, M.R. Morris, D. Astuti, F. Latif,
quirements for N-terminal acetylation of eukaryotic proteins, J. Mol. Biol. E.R. Maher, Germline SDHB mutations and familial renal cell arcinoma,
325 (2003) 595e622. J. Natl. Cancer Inst. 100 (2008) 1260e1262.
[105] X.J. Yang, E. Seto, Lysine acetylation: codied crosstalk with other post- [119] K.A. Janeway, S.Y. Kim, M. Lodish, V. Nose, P. Rustin, J. Gaal, P.L. Dahia,
translational modications, Mol. Cell 31 (2008) 449e461. B. Liegl, E.R. Ball, M. Raygada, A.H. Lai, L. Kelly, J.L. Hornick, N.I.H. Pediatric,
[106] D.W. Parsons, S. Jones, X. Zhang, J.C. Lin, R.J. Leary, P. Angenendt, P. Mankoo, C. Wild-Type GIST, M. O'Sullivan, R.R. de Krijger, W.N. Dinjens, G.D. Demetri,
H. Carter, I.M. Siu, G.L. Gallia, A. Olivi, R. McLendon, B.A. Rasheed, S. Keir, C.R. Antonescu, J.A. Fletcher, L. Helman, C.A. Stratakis, Defects in succinate
T. Nikolskaya, Y. Nikolsky, D.A. Busam, H. Tekleab, L.A. Diaz Jr., J. Hartigan, dehydrogenase in gastrointestinal stromal tumors lacking KIT and PDGFRA
D.R. Smith, R.L. Strausberg, S.K. Marie, S.M. Shinjo, H. Yan, G.J. Riggins, mutations, Proc. Natl. Acad. Sci. U. S. A. 108 (2011) 314e318.
D.D. Bigner, R. Karchin, N. Papadopoulos, G. Parmigiani, B. Vogelstein, [120] S. Kim, D.H. Kim, W.H. Jung, J.S. Koo, Succinate dehydrogenase expression in
V.E. Velculescu, K.W. Kinzler, An integrated genomic analysis of human breast cancer, Springerplus 2 (2013) 299.
glioblastoma multiforme, Science 321 (2008) 1807e1812. [121] A. Fieuw, C. Kumps, A. Schramm, F. Pattyn, B. Menten, F. Antonacci,
[107] E.R. Mardis, L. Ding, D.J. Dooling, D.E. Larson, M.D. McLellan, K. Chen, P. Sudmant, J.H. Schulte, N. Van Roy, S. Vergult, P.G. Buckley, A. De Paepe,
D.C. Koboldt, R.S. Fulton, K.D. Delehaunty, S.D. McGrath, L.A. Fulton, R. Noguera, R. Versteeg, R. Stallings, A. Eggert, J. Vandesompele, K. De Preter,
D.P. Locke, V.J. Magrini, R.M. Abbott, T.L. Vickery, J.S. Reed, J.S. Robinson, F. Speleman, Identication of a novel recurrent 1q42.2-1qter deletion in high
T. Wylie, S.M. Smith, L. Carmichael, J.M. Eldred, C.C. Harris, J. Walker, risk MYCN single copy 11q deleted neuroblastomas, Int. J. Cancer 130 (2012)
J.B. Peck, F. Du, A.F. Dukes, G.E. Sanderson, A.M. Brummett, E. Clark, 2599e2606.
J.F. McMichael, R.J. Meyer, J.K. Schindler, C.S. Pohl, J.W. Wallis, X. Shi, L. Lin, [122] A.A. Khalil, Biomarker discovery: a proteomic approach for brain cancer
H. Schmidt, Y. Tang, C. Haipek, M.E. Wiechert, J.V. Ivy, J. Kalicki, G. Elliott, proling, Cancer Sci. 98 (2007) 201e213.

Potrebbero piacerti anche