10 1016@j Coisb 2017 05 011

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Available online at www.sciencedirect.

com Current Opinion in

ScienceDirect Systems Biology

The importance of post-translational modifications


in systems biology approaches to identify
therapeutic targets in cancer metabolism
Alfonso Martín-Bernabé1,2,3,4,5, Cristina Balcells1,5,
Josep Tarragó-Celada1,5, Carles Foguet1,5,
Sandrine Bourgoin-Voillard2,3,4, Michel Seve2,3,4 and
Marta Cascante1

Abstract Introduction
Cancer metabolism is reprogrammed to fulfill the needs of Cancer is a complex and heterogeneous disease, which
proliferation and migration, which is accomplished through involves alteration of multiple biological processes.
different levels of regulation. In recent years, new advances in Some of these alterations are geared towards metabolic
protein post-translational modifications (PTMs) research have reprogramming, which provides advantages to cancer
revealed a complex layer of regulatory mechanisms through cells in terms of energy production and synthesis of
which PTMs control cell signaling and metabolic pathways, biomolecules and is essential for tumor progression.
contributing to the diverse metabolic phenotypes found in Understanding metabolic reprogramming in heteroge-
cancer. Despite the efficacy of current modeling approaches neous tumor cell populations is key to identify meta-
to study cancer metabolism they still lack the capacity to inte- bolic vulnerabilities in cancer that can be exploited in
grate PTMs in their predictions. Here we will review the impor- therapy [1].
tance of PTMs in cancer metabolic reprogramming and
suggest ways in which computational predictions could be Metabolism is a complex network of biochemical re-
enhanced through the integration of PTMs. actions that requires a systemic view. Computational
models have emerged as platforms to integrate multi-
Addresses omics data (genomics, transcriptomics, proteomics,
1
Department of Biochemistry and Molecular Biomedicine, Universitat
metabolomics, etc.), and therefore to understand the
de Barcelona and Institute of Biomedicine of Universitat de Barcelona
(IBUB), Barcelona, Spain underlying metabolic phenotype. However current
2
Univ. Grenoble Alpes, LBFA et BEeSy, PROMETHEE Proteomic modeling approaches have a limited capacity to inte-
Platform, Grenoble, France grate modifications that occur at a post-translational
3
Inserm, U1055, PROMETHEE Proteomic Platform, Grenoble, France level hence limiting their usefulness at analyzing
4
CHU de Grenoble, Institut de Biologie et de Pathologie, PROM-
cancer metabolic reprogramming.
ETHEE Proteomic Platform, Grenoble, France

Corresponding author: Cascante, Marta (martacascante@ub.edu) Here, we review recent studies regarding the role of
Email addresses: alf.martin@gmail.com (A. Martín-Bernabé), post-translational modifications (PTMs) in cancer
crisgatsu@gmail.com (C. Balcells), jtarragocelada@ub.edu metabolic reprogramming, mainly focusing on central
(J. Tarragó-Celada), cfoguet@ub.edu (C. Foguet),
sandrine.bourgoin@univ-grenoble-alpes.fr (S. Bourgoin-Voillard),
carbon metabolism. In this context, we assess how
michel.seve@univ-grenoble-alpes.fr (M. Seve) computational predictions through genome-scale meta-
5
These authors contributed equally to this work. bolic models (GSMMs) and kinetic models could be
enhanced through the integration of PTMs.
Current Opinion in Systems Biology 2017, 3:161–169
Metabolic reprogramming in cancer
This review comes from a themed issue on Clinical and translational The most common metabolic adaptation in cancer is the
systems biology (2017)
Warburg effect, consisting of high glucose uptake,
Edited by Jesper Tegnér and David Gomez-Cabrero glycolytic activity, and lactate production even under
For a complete overview see the Issue and the Editorial aerobic conditions [2]. However, metabolic reprogram-
Available online 19 May 2017 ming is not limited to the Warburg effect and usually
http://dx.doi.org/10.1016/j.coisb.2017.05.011 involves the enhancement of other essential metabolic
pathways including pentose phosphate pathway (PPP),
2452-3100/© 2017 Elsevier Ltd. All rights reserved.
glutaminolysis, and amino acid and lipid metabolisms [3].

Tumor cells display metabolic flexibility, which allows


them to undergo metabolic switches and use different
energy and carbon sources depending on their

www.sciencedirect.com Current Opinion in Systems Biology 2017, 3:161–169


162 Clinical and translational systems biology (2017)

availability. Moreover, the metabolic phenotype is In addition to AKT signaling, both AS160 and PFK2 also
influenced by the interplay between tumor cells and the receive signaling inputs through phosphorylation by
surrounding stroma, resulting in distinct metabolic sig- AMP-activated protein kinase (AMPK). AMPK con-
natures, as an adaptation to a particular microenviron- tributes to achieve a fine-tuned control over the glyco-
ment [4]. lytic rate as a response to low energy levels. On the one
hand, enhancing glucose uptake via AS160 [12] and, on
Post-translational modifications the other, balancing the glycolysis/gluconeogenesis ratio
Post-translational modifications (PTMs) can directly by regulating PFK2, promoting energy homeostasis and
regulate metabolic enzymes employing different survival in lung and breast cancer [13]. Moreover, AMPK
mechanisms such as leading to proteineprotein in- is also an important regulator of lipid metabolism,
teractions that can result in the formation of multi- targeting acetyl-CoA carboxylase (ACC), responsible for
protein complexes, triggering conformational changes the synthesis of the lipid precursor malonyl-CoA [14].
between active and inactive states, causing translocation Indeed, the interplay between AKT pathway and AMPK
to a different cellular compartment or targeting for in driving central energy metabolism is complex, as is
degradation. The signal transduction effectors respon- their contribution to oncogenic metabolic reprogram-
sible for these modifications, such as kinases/phospha- ming. As described recently for HeLa cells, PI3K/AKT
tases or acetylases/deacetylases are also regulated at the pathway is also responsible for the regulation of trans-
same level generating a complex signaling circuitry that ketolase (TKT), one important node of the non-
responds quickly to cellular needs or external stimuli oxidative branch of PPP. AKT directly phosphorylates
(Figure 1) [5]. TKT, increasing its activity and the carbon flow through
the non-oxidative branch of PPP, thus having a direct
Furthermore, metabolites can also alter signal trans- impact on purine synthesis [15].
duction pathways, inducing metabolically sensitive
PTMs, which in turn can modulate the activities of Another prominent family of kinases with direct impact
signaling proteins, metabolic enzymes and transcrip- on cancer metabolic reprogramming is the pyruvate
tional regulators [6]. This constitutes an important dehydrogenase kinase (PDHK) family. Phosphorylation,
mechanism by which signal transduction pathways are and thereby inactivation, of pyruvate dehydrogenase by
modulated through feedback control of multiple types PDHKs certainly contributes to maintaining an
of metabolite-derived PTMs acting as “nutritional sen- increased glycolytic rate by redirecting pyruvate to
sors” [7,8]. lactate production, consequently supporting the
Warburg effect. An increased relative abundance of
Recent advances in post-translational phosphorylated pyruvate dehydrogenase (PDH) has
modifications in cancer metabolic been linked to higher metastatic and proliferative ca-
reprogramming pabilities in a prostate cancer model of increased and
Phosphorylation suppressed metastatic capacity of PC-3 [16].
Protein phosphorylation is a well-studied regulatory
mechanism of metabolism with an important role in Another phosphorylation event that promotes the
most cellular processes, and its dysfunction is a major Warburg effect has been elucidated recently. This new
contributor to cancer. Protein kinases and phosphatases mechanism involves the enhancement of hexokinases 1
modulate the capability of a protein to perform its and 2 (HK1/2) activities by tyrosine kinase c-Src phos-
function in the cellular metabolic and signaling net- phorylation, thus activating mechanisms that lead to
works, by covalently binding or hydrolyzing phosphate tumorigenesis and metastasis in different cancer cell
groups from the hydroxyl-containing residues of pro- types such as lung, ovarian, breast or colon, among others
teins, serine, tyrosine and threonine. In particular, (Figure 2A) [17]. Since phosphorylation has such a
different signaling cascades of phosphorylation regulate profound impact on the regulation of metabolic en-
central carbon metabolism in a direct manner [9]. For zymes, the addition of this layer of data is crucial. In this
instance, crosstalk between insulin receptor tyrosine fashion, some examples are already available in the
kinase (IRTK) and PI3K/AKT serine/threonine kinase literature using phosphoproteomic approaches, focusing
pathways regulates insulin-stimulated metabolism. on the signaling pathways mentioned above that exert
Insulin-driven glucose uptake is regulated by insulin control over metabolism [18e20].
receptor substrates IRS1 and IRS2 that rely on PI3K
activation and, besides, glucose transporter GLUT4 is Acetylation
regulated via AS160 phosphorylation by AKT [10]. Protein lysine acetylation is another well-studied PTM,
Moreover, AKT is also responsible for phosphofructoki- which consists on transferring or removing an acetyl
nase 2 (PFK2) phosphorylation, an enzyme that directly group from acetyl-CoA to an ε-N-lysine. The responsible
regulates glycolysis through fructose-2,6-bisphosphate enzymes for this PTM are lysine acetyltransferases
levels, as found in HeLa cells [11]. (KATs) and lysine deacetylases (KDACs). It was first

Current Opinion in Systems Biology 2017, 3:161–169 www.sciencedirect.com


Post-translational modifications as therapeutic targets in cancer metabolism Martín-Bernabé et al. 163

Figure 1

Simplified diagram of central carbon metabolism depicting some of the metabolic enzymes directly regulated by post-translational modifica-
tions (PTMs) and involved in the cancer metabolic reprogramming. Each metabolic enzyme is regulated by different PTMs: phosphorylation (P),
acetylation (+Ac), deacetylation (−Ac), glycosylation (G), hydroxylation (OH), oxidation (Ox), ubiquitination (+Ub), deubiquitination (−Ub) and SUMOylation
(SU). The modifications are represented in boxes next to the metabolic enzymes of the following pathways: glycolysis/gluconeogenesis (blue), pentose
phosphate pathway (orange), lactate metabolism (purple), TCA cycle (green) and lipid metabolism (yellow). Each box is split in three: left box indicates the
type of PTM, right box indicates the effect on the metabolic enzyme and the responsible effectors are enclosed between them. 6PGD, 6-phosphogluconate
dehydrogenase; ACAT1, acetyl-CoA acetyltransferase 1; ACAT2, acetyl-CoA acetyltransferase 2; ACC1, acetyl-CoA carboxylase 1; ACC2, acetyl-CoA
carboxylase 2; ACLY, ATP citrate lyase; AMPK, AMP-activated protein kinase; c-Src, proto-oncogene tyrosine-protein kinase; CK2, casein kinase 2;
DLAT, dihydrolipoamide S-acetyltransferase; ENO1/2, enolase 1/2; ERK1/2, extracellular signal-regulated kinase 1/2; FASN, fatty acid synthase;
FGFR1, fibroblast growth factor receptor 1; G6PD, glucose-6-phosphate dehydrogenase; GAPDH, glyceraldehyde-3-phosphate dehydrogenase;
GLUT1, glucose transporter 1; HK1/2, hexokinase 1/2; LDHA, lactate dehydrogenase A; IDH2, isocitrate dehydrogenase 2; KDAC3, histone deacetylase
3; KAT, lysine acetyltransferase; LCAD, long chain fatty acid acyl-CoA dehydrogenase; MDH, malate dehydrogenase; OGDH, 2-oxoglutarate dehydroge-
nase; OGT, O-linked N-acetylglucosamine (GlcNAc) transferase; PCAF, p300/CBP-associated factor; PDH, pyruvate dehydrogenase; PDH3, prolyl hy-
droxylase 3; PEPCK1, phosphoenolpyruvate carboxykinase; PFK1/2, phosphofructokinase1/2; PGAM1, phosphoglycerate mutase 1; PGI,
phosphoglucose isomerase; PIAS3, protein inhibitor of activated STAT (signal transducer and activator of transcription); PI3K/AKT,
phosphatidylinositol-3 kinase, serine/threonine protein kinase; PKA, protein kinase A; PKM2, pyruvate kinase M2; ROS, reactive oxygen species;
SDH, succinate dehydrogenase; SIRT1, sirtuin 1; SIRT2, sirtuin 2; TKT, transketolase; USP13, ubiquitin-specific peptidase 13; USP20, ubiquitin-
specific peptidase 20.

discovered in histones, but it is now known to act on is widely studied: acetylation at K305 decreases its
other proteins, including metabolic enzymes [21]. Since enzyme activity and chaperone-mediated degradation
its discovery, acetylation has been extensively explored, [25] while acetylation at K433 promotes its nuclear
and new methods to detect and quantify it have been translocation [26]. In both cases, cancer cell prolifera-
developed [22,23] and virtually, almost every enzyme tion is favored through glycolytic intermediates accu-
from the central carbon metabolism is susceptible to be mulation. Another well characterized example of direct
acetylated [24]. acetylation of an enzyme is LDH isoform A. In contrast
with the previous example, in this case acetylation at K5
Among the most relevant enzymes in cancer meta- impairs tumor growth by inhibiting LDHA enzyme ac-
bolism, the case of pyruvate kinase isoform M2 (PKM2) tivity [27]. Fatty acid synthesis is another important

www.sciencedirect.com Current Opinion in Systems Biology 2017, 3:161–169


164 Clinical and translational systems biology (2017)

Figure 2

Schematic examples of recent post-translational modifications involved in cancer metabolic reprogramming with potential therapeutic
implications. (A) Phosphorylation. Hexokinases (HK1/2) catalyze the rate-limiting step of glycolysis. Zhang et al. found that s-Src promotes glycolytic
flux by phosphorylating and activating HK, which stimulates the Warburg effect, cell proliferation, and metastasis [17]. (B) Acetylation. Cluntun et al.
proved that the glycolytic rate correlates with histone acetylation sites leading to transcriptional activation of glycolytic and TCA enzymes, which in
turn enhances the Warburg effect [29]. This is mediated by the high levels of Acetyl-CoA induced by glycolysis that can be reverted in a caloric restriction
diet, which can be a complement to cancer treatment [33]. (C) Glycosylation. The redirection of glucose from glycolysis through the pentose phosphate
pathway (PPP) provides a growth advantage to tumors. Rao et al. identified a new mechanism by which O-GlcNAcylation of glucose-6-phosphate de-
hydrogenase (G6PD) promotes the PPP, which has clear therapeutic implications [41]. (D) Ubiquitination. ATP citrate lyase (ACLY) and oxoglutarate
dehydrogenase (OGDH) are key metabolic enzymes involved in mitochondrial respiration, glutaminolysis, and fatty acid synthesis. Han et al. found a
positive correlation between up-regulation of ACLY and OGDH and USP13 (ubiquitin specific peptidase 13) overexpression which, is frequently amplified
in human ovarian cancer [48].

pathway for cancer metabolism affected by acetylation: manner. This allows cells to proliferate even under the
It has been recently reported that FASN proteasome inhibition of the epidermal growth factor receptor VIII
degradation is promoted by KAT8 acetylation while (EGFRVIII) unveiling a new mechanism for targeted
HDAC3 deacetylation prevents FASN degradation, therapy resistance [30].
promoting tumor cell growth by de novo lipogenesis [28].
However, the levels of acetyl-CoA are not only depen-
Protein acetylation is highly sensitive to acetyl-CoA dent on glycolysis but also on synthesis from acetate or
levels, the substrate for lysine acetyltransferases. A pos- fatty acid and amino acid catabolism. In certain cancers,
itive correlation has been found between acetyl-CoA where cells that are usually glucose-deprived or suffer
levels and histone acetylation which results in from acidosis, fatty acids and acetate constitute signifi-
increased expression of glycolytic enzymes in HCT-116 cant energetic substrates [31]. In this context, mito-
colon cancer cells (Figure 2B) [29]. Another effect of chondrial proteins such as components of the respiratory
high acetyl-CoA levels is found in glioblastoma cells complex I are down-regulated by hyperacetylation,
where acetylation of a component of the mTORC2 while histones in the nucleus are deacetylated to
signaling complex, Rictor, occurs in a glucose-dependent maintain high acetate levels [32]. Additionally, reduced

Current Opinion in Systems Biology 2017, 3:161–169 www.sciencedirect.com


Post-translational modifications as therapeutic targets in cancer metabolism Martín-Bernabé et al. 165

acetylation associated with low acetyl-CoA levels has and lactate transporters. Besides the post-translational
been shown to drive autophagy, evidencing caloric re- regulation of HIF-1, PKM2 and PDH and, more
striction as a potential complement to cancer therapy recently acetyl-CoA carboxylase 2 (ACC2) have also
[33]. Therefore, it is clear that acetylation links nutrient been found to be susceptible to hydroxylation [43e45].
status with gene expression and metabolic regulation. Certainly, due to the high importance of hypoxia and
ROS in cancer metabolism, hydroxylation will probably
Lysine deacetylases are also possible targets for cancer attract more attention in the future.
treatment. The inhibitors of lysine deacetylases have
been broadly studied, and some of them are even clin- Ubiquitination and SUMOylation are also worth
ically approved. In HeLa and A549 cells, one of the ef- mentioning, since they are important regulators of pro-
fects of these inhibitors is the acetylation of tein function and fate. They involve the attachment of
mitochondrial proteins [34,35]. It is also observed that the polypeptides ubiquitin or SUMO (small ubiquitin-
ketogenic diets maintain high acetylation levels due to like modifier) to lysine residues. In the context of
the inhibitory effect of beta-hydroxybutyrate to lysine cancer, increasing evidence has linked dysregulation of
deacetylases [36]. these two PTMs with cancer development and meta-
bolic reprogramming [46e48]. There is a recent example
Glycosylation where Parkin, a ubiquitin E3 ligase, has an important role
Protein glycosylation is another widespread PTM, which in cancer metabolism by inhibiting glycolysis through
involves the addition of glycans to proteins. This ubiquitination of PKM2 and therefore, suppressing
modification is mostly associated with membrane pro- tumor growth [49]. Another example is given by the
teins but also occurs at intracellular level. It is now well upregulation of ATP citrate lyase (ACLY) and oxogluta-
established that protein glycosylation modulates tumor rate dehydrogenase (OGDH), which control mitochon-
growth and malignant transformation and glycans might drial function, lipid metabolism, and glutaminolysis. In
play a role in every well-recognized cancer hallmark this case, the event responsible of the metabolic rewiring
[37]. Among the various forms of protein glycosylations, is the overexpression of the ubiquitin-specific peptidase
intracellular O-GlcNAcylation (O-GlcNAc), which is 13 (USP13), a deubiquitination enzyme activating both
abundant in proteins involved in cell signaling, stress metabolic enzymes, thereby conferring a tumor growth
responses and energy metabolism, plays a relevant role advantage (Figure 2D) [48]. Additionally, the nuclear
in cancer development by regulating signaling pathways receptor Nur77 has been shown to activate gluconeo-
[38]. In addition, O-GlcNAc glycosylation and expres- genesis through an attenuation of the SUMOylation of
sion of O-GlcNAc-cycling enzymes have also been phosphoenolpyruvate carboxykinase 1 (PEPCK1) in he-
widely described as being dysregulated in many types of patocellular carcinoma [50].
cancer, activating multiple transcription factors and key
metabolic enzymes [39]. In lung cancer, for example, However, the scope of PTMs is rapidly expanding with
the O-GlcNAcylations of PFK1 and glucose-6- the identification of new PTMs, and this is the case of
phosphate dehydrogenase (G6PD), enzymes catalyzing protein lysine acylations such as malonylation, succiny-
rate-limiting steps in glycolysis and PPP, respectively, lation, and glutarylation. They are believed to modify
are dynamically controlled in response to different metabolic enzymes using intermediates of energy
cellular conditions including hypoxia and nutrient metabolism, but their roles in the regulation of cellular
levels. The inhibition of PFK1 and activation of G6PD metabolism remain largely unknown [51e55].
by O-GlcNAc control directly the flux of PPP which may
represent a potential therapeutic target in cancer Finally, it is important to mention that PTMs are rarely
(Figure 2C) [40,41]. single independent events and they likely occur in a
coordinated manner through various and abundant
Other protein post-translational modifications PTMs crosstalks, but this will not be discussed here.
Metabolic enzymes are also regulated through other
PTMs such as hydroxylation, which is the transfer of PTMs in metabolic modeling
hydroxyl groups to proline or lysine residues. Hydrox- PTMs in genome-scale metabolic models
ylation of proline, mostly catalyzed by prolyl hydroxylase GSMMs are mathematical representations of the entire
domain (PHD) enzymes, has a prominent role in cancer metabolic reaction complement encoded by an organ-
[42]. For instance, the metabolic adaptation of cancer ism’s genome. An essential part of GSMMs is the Gene
cells to hypoxia is predominantly executed through the Protein Reaction (GPR) associations, a set of rules that
activation of the hypoxia-induced factor-1 (HIF-1) define the gene products needed to catalyze each of the
pathway, which is degraded and inactivated in the reactions included in the model. The GPR rules allow
presence of oxygen through hydroxylation. In hypoxia, GSMMs to serve as platforms where transcriptomics and
HIF-1 activity enhances glycolysis through the up- proteomics can be integrated to construct condition-
regulation of almost all glycolytic genes and glucose specific metabolic models. There are several algorithms

www.sciencedirect.com Current Opinion in Systems Biology 2017, 3:161–169


166 Clinical and translational systems biology (2017)

capable of integrating these omics data, but mostly, they annotation should provide the following information: 1)
are all based on the assumption that a reaction catalyzed which residues of a given enzyme can be modified, 2)
by a highly expressed enzyme is most likely to be active the specific PTMs that are possible at each residue and
than a reaction catalyzed by a lowly expressed one [56]. 3) a set of rules capable of predicting the likely effect of
These algorithms have been extensively used to possible combinations of PTMs on enzyme activity.
generate cancer-specific metabolic reconstructions with Secondly, we must develop a new generation of algo-
the goal of identifying vulnerabilities that can be rithms capable of predicting flux distributions using not
exploited in therapy [57,58]. only transcriptomic or proteomic data but also the
relative abundances of the modified and unmodified
However, PTMs are one of the major limitations of the forms for each enzyme (Figure 3). As not all PTMs
current generation of algorithms as highly expressed might be characterized in a given study and some PTMs
enzymes can be inactivated due to PTMs, which may might be undetectable (e.g. due to low abundance or
lead to errors on the resulting metabolic reconstructions. short half-life), such algorithm should be robust enough
Therefore, a major challenge in the coming years will be to deal with incomplete PTM experimental data.
to overcome this limitation and improve their predictive
capacity through the integration of PTM measure- PTMs in kinetic modeling
ments. To achieve this, GSMMs will firstly need to be Kinetic modeling is based on using a system of ordinary
expanded to include annotations about PTMs. Such differential equations to predict the evolution of a

Figure 3

Example of condition-specific reconstructions performed with a current and a hypothetical next generation algorithms. Consider three metabolic pathways
(P1, P2, and P3) in which a set of enzymes (E1, E2, …, E9) catalyze the conversion of a series of metabolites (M1, M2, …, M8). Additionally, assume that
E7 can be completely inhibited by a PTM. Furthermore, consider that the heatmap summarizes gene expression evidence of the enzymes in the system
(green = lowly expressed, red = highly expressed) and the relative abundance of E7 post-translationally modified (red = 100% abundance compared to
the total E7). A current generation algorithm would only be able to integrate gene expression data and might predict that P1 is inactive (as its reactions are
catalyzed by lowly expressed enzymes) and that pathway P2 and P3 can be active. A hypothetical next generation algorithm would also integrate gene
expression, but would also be able to integrate PTM data (E7-PTM) predicting that P3 is inactive as E7 is inactivated by PTMs. Should production of M8
be necessary for cell proliferation/survival, the reliance on P2 to produce M8 would be a vulnerability that could be exploited in cancer therapy.

Current Opinion in Systems Biology 2017, 3:161–169 www.sciencedirect.com


Post-translational modifications as therapeutic targets in cancer metabolism Martín-Bernabé et al. 167

metabolic network in time. A key part of kinetic Conclusions


modeling is kinetic laws, mathematical representations Oncogenic activation alters post-translational signaling
of the kinetic properties of enzymes in the network (i.e. cascades that contribute, together with changes in
substrate affinity, maximal velocity, cooperativity, etc.). transcription and epigenetics, to a complete reprog-
Consequently, the effect of a PTM that alters the ki- ramming of metabolism. However, limited information
netic properties of an enzyme can be modeled by using is available on post-translational events and their precise
different kinetic laws for the modified and unmodified contribution to metabolic reprogramming.
enzymes. Then, the apparent kinetic law of the enzyme
can be formulated as the linear combination of kinetic Understanding the roles of the diverse PTMs governing
laws using the fraction of modified and unmodified cancer metabolism is still challenging due to the com-
enzyme as coefficients (Figure 4). This approach was plex and heterogeneous nature of tumors. Thus, further
successfully used by König et al. [59] to build a kinetic research and PTM data integration are necessary to
model of hepatic metabolism with hormonal control develop more accurate modeling strategies, which may
represented as changes in the phosphorylation state of offer new approaches to target cancer metabolism. In
key enzymes. the horizon, a high-throughput characterization of
PTMs and its integration in genome-scale modeling
The main challenge to simulating PTMs in kinetic approaches is likely to help to identify novel personal-
models is that the effect of PTMs on kinetic properties ized diagnostic and therapeutic applications.
has only been characterized on a very small subset of
enzymes. Acknowledgements
This work was supported by grants to M.C. from the “Agència de Gestió
Nevertheless, the development of genome-scale kinetic d’Ajuts Universitaris i de Recerca (AGAUR)” e Generalitat de Catalunya,
(2014SGR1017), ICREA Foundation (ICREA Acadèmia 2015), the
models [60,61] is sparking a renewed interest in kinetic “Ministerio de Economı́a y Competitividad” from the Spanish Government
modeling, and it is likely that we will see the kinetic and FEDER funds from the European Union “una manera de hacer
Europa” (SAF2014-56059-R, SAF2015-70270-REDT). A.M.B. acknowl-
characterization of many PTMs in the coming years. edges the “Ministère de l’Enseignement supérieur et de la Recherche”
from the French Government. C.B. acknowledges the AGAUR. J.T.C. ac-
knowledges the “Ministerio de Educación, Cultura y Deporte”. C.F. ac-
knowledges “La Caixa” Foundation.

Figure 4 References
Papers of particular interest, published within the period of review,
have been highlighted as:

 of special interest
 of outstanding interest

1. Tarrado-Castellarnau M, De Atauri P, Cascante M: Oncogenic


regulation of tumor metabolic reprogramming. Oncotarget
2016, 7.
2. Liberti MV, Locasale JW: The Warburg effect: how does it
benefit cancer cells? Trends Biochem Sci 2016, 41:211–218.
3. Pavlova NN, Thompson CB: The emerging hallmarks of cancer
metabolism. Cell Metab 2016, 23:27–47.
4. Martinez-Outschoorn UE, Lisanti MP, Sotgia F: Catabolic
cancer-associated fibroblasts transfer energy and biomass
to anabolic cancer cells, fueling tumor growth. Semin Cancer
Biol 2014, 25:47–60.
5. Hitosugi T, Chen J: Post-translational modifications and the
Warburg effect. Oncogene 2014, 33:4279–4285.
6. Wellen KE, Thompson CB: A two-way street: reciprocal regu-
lation of metabolism and signalling. Nat Rev Mol Cell Biol
2012, 13:270–276.
7. Yuan HX, Xiong Y, Guan KL: Nutrient sensing, metabolism,
Example of how the effect of PTM can be simulated in kinetic models. and cell growth control. Mol Cell 2013, 49:379–387.
Consider, the phosphofructokinase 2 (PFK2) activity in the liver which is
8. Xu W, Li Y, Liu C, Zhao S, Ho Jun L, Lang PT, Fortune SM,
partially inhibited by phosphorylation. This inhibition can be simulated by Sassetti CM, Alber T, Friedmann DR, et al.: Protein lysine
using different kinetic laws for the dephosphorylated and phosphorylated acetylation guards metabolic homeostasis to fight against
enzyme (Kdp and Kp, respectively). Then, the apparent kinetic properties cancer. Oncogene 2013, 33:5570–5581.
of PFK2 can be simulated as the sum of the phosphorylated and
9. Humphrey SJ, James DE, Mann M: Protein phosphorylation: a
dephosphorylated kinetic laws weighted by the fraction of phosphorylated major switch mechanism for metabolic regulation. Trends
(p) and dephosphorylated forms of the enzyme (1-p) [59]. This is exempli- Endocrinol Metab 2015, 26:676–687.
fied in this figure, where the reaction rate (V) of PFK2 is plotted as function
fructose 6-phosphate (Fru6P) at 0%, 33%, 66% and 100% fractions of the
phosphorylated enzyme.

www.sciencedirect.com Current Opinion in Systems Biology 2017, 3:161–169


168 Clinical and translational systems biology (2017)

10. Guo S: Insulin signaling, resistance, and metabolic syn- 26. Lv L, Xu Y, Zhao D, Li F, Wang W, Sasaki N, Jiang Y, Zhou X:
drome: insights from mouse models into disease mecha- Mitogenic and oncogenic stimulation of K433 acetylation
nisms. J Endocrinol 2014, 220:T1–T13. promotes PKM2 protein kinase activity and nuclear localiza-
tion. Mol Cell 2013, 52:340–352.
11. Mulukutla BC, Yongky A, Daoutidis P, Hu WS: Bistability in
glycolysis pathway as a physiological switch in energy 27. Zhao D, Zou SW, Liu Y, Zhou X, Mo Y, Wang P, Xu YH, Dong B,
metabolism. PLoS One 2014, 9. Xiong Y, Lei QY, et al.: Lysine-5 acetylation negatively regu-
lates lactate dehydrogenase a and is decreased in pancreatic
12. Treebak JT, Taylor EB, Witczak CA, An D, Toyoda T, Koh H-J, cancer. Cancer Cell 2013, 23:464–476.
Xie J, Feener EP, Wojtaszewski JFP, Hirshman MF, et al.:
Identification of a novel phosphorylation site on TBC1D4 28. Lin H, Cheng Z, He R, Song L, Tian M, Zhou L, Groh BS, Liu W,
regulated by AMP-activated protein kinase in skeletal Ji M, Ding C: Destabilization of fatty acid synthase by acety-
muscle. Am J Physiol Cell Physiol 2010, 298:C377–C385. lation inhibits de novo lipogenesis and tumor cell growth.
Cancer Res 2016, 76:6924–6936.
13. Chaube B, Malvi P, Singh SV, Mohammad N, Viollet B, Bhat MK:
AMPK maintains energy homeostasis and survival in cancer 29. Cluntun AA, Huang H, Dai L, Liu X, Zhao Y, Locasale JW: The
cells via regulating p38/PGC-1a-mediated mitochondrial rate of glycolysis quantitatively mediates specific histone
biogenesis. Cell Death Discov 2015, 1:15063. acetylation sites. Cancer Metab 2015, 3:10.
14. Li W, Saud SM, Young MR, Chen G, Hua B: Targeting AMPK for 30. Masui K, Tanaka K, Ikegami S, Villa GR, Yang H, Yong WH,
cancer prevention and treatment. Oncotarget 2015, 6: Cloughesy TF, Yamagata K, Arai N, Cavenee WK, et al.:
7365–7378. Glucose-dependent acetylation of Rictor promotes targeted
cancer therapy resistance. Proc Natl Acad Sci U S A 2015, 112:
15. Saha A, Connelly S, Jiang J, Zhuang S, Amador DT, Phan T, 9406–9411.
Pilz RB, Boss GR: Article Akt phosphorylation and regulation
of transketolase is a nodal point for amino acid control of 31. Mashimo T, Pichumani K, Vemireddy V, Hatanpaa KJ, Singh DK,
purine synthesis. Mol Cell 2014, 55:264–276. Sirasanagandla S, Nannepaga S, Piccirillo SG, Kovacs Z,
Foong C, et al.: Acetate is a bioenergetic substrate for human
16. Aguilar E, Marin de Mas I, Zodda E, Marin S, Morrish F, glioblastoma and brain metastases. Cell 2014, 159:
 Selivanov V, Meca-Cortés O, Delowar H, Pons M, Izquierdo I, 1603–1614.
et al.: Metabolic reprogramming and dependencies associ-
ated with epithelial cancer stem cells independent of the 32. Cyril Corbet A, Pinto A, Martherus R, Pedro Santiago de Jesus J,
epithelial-mesenchymal transition program. Stem Cells 2016,  Corbet C, Polet F, Feron O: Acidosis drives the reprogram-
33:1163–1176. ming of fatty acid metabolism in cancer cells through
This study unveils the metabolic differences between two sub- changes in mitochondrial and histone acetylation. Cell Metab
populations of a dual cell line model of prostate cancer. Phosphoryla- 2016, 24:311–323.
tion of pyruvate dehydrogenase is increased in the metastatic Description of a new cancer metabolic reprogramming strategy in
subpopulation over the non-metastatic one. acidosis context based on acetylation of mitochondrial proteins and
histone deacetylation.
17. Zhang J, Wang S, Jiang B, Huang L, Ji Z, Li X, Zhou H, Han A,
Chen A, Wu Y, et al.: c-Src phosphorylation and activation of 33. Mariño G, Pietrocola F, Eisenberg T, Kong Y, Malik SA,
hexokinase promotes tumorigenesis and metastasis. Nat Andryushkova A, Schroeder S, Pendl T, Harger A, Niso-
Commun 2017, 8:1–16. Santano M, et al.: Regulation of autophagy by cytosolic acetyl-
coenzyme A. Mol Cell 2014, 53:710–725.
18. Humphrey SJ, Azimifar SB, Mann M: High-throughput phos-
phoproteomics reveals in vivo insulin signaling dynamics. 34. Wu Q, Cheng Z, Zhu J, Xu W, Peng X, Chen C, Li W, Wang F,
Nat Biotechnol 2015, 33:990–995. Cao L, Yi X, et al.: Suberoylanilide hydroxamic acid treatment
reveals crosstalks among proteome, ubiquitylome and
19. Kim J-Y, Welsh EA, Fang B, Bai Y, Kinose F, Eschrich SA, acetylome in non-small cell lung cancer A549 cell line. Sci
Koomen JM, Haura EB: Phosphoproteomics reveals MAPK Rep 2015, 5:9520.
inhibitors enhance MET- and EGFR-driven AKT signaling in
KRAS-mutant lung cancer. Mol Cancer Res 2016, 217: 35. Schölz C, Weinert BT, Wagner SA, Beli P, Miyake Y, Qi J,
1019–1029.  Jensen LJ, Streicher W, McCarthy AR, Westwood NJ, et al.:
Acetylation site specificities of lysine deacetylase inhibitors
20. Hu C-W, Hsu C-L, Wang Y-C, Ishihama Y, Ku W-C, Huang H-C, in human cells. Nat Biotechnol 2015, 33:415–423.
Juan H-F: Temporal phosphoproteome dynamics induced by A systemic approach of lysine deacetylases inhibitors specificities,
an ATP synthase inhibitor citreoviridin. Mol Cell Proteomics which permits to study the function of acetylation.
2015, 14:3284–3298.
36. Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le
21. Kim SC, Sprung R, Chen Y, Xu Y, Ball H, Pei J, Cheng T, Kho Y, Moan N, Grueter CA, Lim H, Saunders LR, Stevens RD,
Xiao H, Xiao L, et al.: Substrate and functional diversity of Newgard CB, Farese Jr RV, de Cabo R, Ulrich S, Akassoglou K,
lysine acetylation revealed by a proteomics survey. Mol Cell Verdin E: Suppression of oxidative stress by b-hydrox-
2006, 23:607–618. ybutyrate, an endogenous histone deacetylase inhibitor.
Science 2013 Jan 11, 339:211–214.
22. Mertins P, Qiao JW, Patel J, Udeshi ND, Clauser KR, Mani DR,
Burgess MW, Gillette MA, Jaffe JD, Carr SA: Integrated prote- 37. Munkley J, Elliott DJ: Hallmarks of glycosylation in cancer.
omic analysis of post-translational modifications by serial Oncotarget 2016, 7:1–12.
enrichment. Nat Methods 2013, 10:634–637.
38. Fardini Y, Dehennaut V, Lefebvre T, Issad T: O-GlcNAcylation: a
23. Xu L, Wang F, Xu Y, Wang Y, Zhang C, Qin X, Yu H, Yang P: An new cancer hallmark? Front Endocrinol (Lausanne) 2013, 4:
MRM-based workflow for absolute quantitation of lysine- 1–14.
acetylated metabolic enzymes in mouse liver. Analyst 2015,
140:7868–7875. 39. Ma Z, Vosseller K: Cancer metabolism and elevated
O-GlcNAc in oncogenic signaling. J Biol Chem 2014, 289:
24. Zhao S, Xu W, Jiang W, Yu W, Lin Y, Zhang T, Yao J, Zhou L, 34457–34465.
Zeng Y, Li H, Li Y, Shi J, An W, Hancock SM, He F, Qin L, Chin J,
Yang P, Chen X, Lei Q, Xiong Y, Guan KL: Regulation of cellular 40. Yi W, Clark PM, Mason DE, Keenan MC, Hill C, Goddard 3rd WA,
metabolism by protein lysine acetylation. Science 2010 Feb Peters EC, Driggers EM, Hsieh-Wilson LC: Phosphofructoki-
19, 327:1000–1004. nase 1 glycosylation regulates cell growth and metabolism.
Science 2012 Aug 24, 337:975–980.
25. Lv L, Li D, Zhao D, Lin R, Chu Y, Zhang H, Zha Z, Liu Y, Li Z,
Xu Y, et al.: Acetylation targets the M2 isoform of pyruvate 41. Rao X, Duan X, Mao W, Li X, Li Z, Li Q, Zheng Z, Xu H, Chen M,
kinase for degradation through chaperone-mediated auto- Wang PG, et al.: O-GlcNAcylation of G6PD promotes the
phagy and promotes tumor growth. Mol Cell 2011, 42: pentose phosphate pathway and tumor growth. Nat Commun
719–730. 2015, 6:8468.

Current Opinion in Systems Biology 2017, 3:161–169 www.sciencedirect.com


Post-translational modifications as therapeutic targets in cancer metabolism Martín-Bernabé et al. 169

42. Nguyen TL, Durán RV: Prolyl hydroxylase domain enzymes 52. Hirschey MD, Zhao Y: Metabolic regulation by lysine malony-
and their role in cell signaling and cancer metabolism. Int J lation, succinylation, and glutarylation. Mol Cell Proteomics
Biochem Cell Biol 2016, 80:71–80. 2015, 14:2308–2315.
43. Luo W, Hu H, Chang R, Zhong J, Knabel M, O’Meally R, Cole RN, 53. Du Y, Cai T, Li T, Xue P, Zhou B, He X, Wei P, Liu P, Yang F,
Pandey A, Semenza GL: Pyruvate kinase M2 is a PHD3- Wei T: Lysine malonylation is elevated in type 2 diabetic
stimulated coactivator for hypoxia-inducible factor 1. Cell mouse models and enriched in metabolic associated pro-
2011, 145:732–744. teins. Mol Cell Proteomics 2015, 14:227–236.
44. Kikuchi D, Minamishima YA, Nakayama K: Prolyl-hydroxylase 54. Xu H, Chen X, Xu X, Shi R, Suo S, Cheng K, Zheng Z, Wang M,
PHD3 interacts with pyruvate dehydrogenase (PDH)-E1b and Wang L, Zhao Y, et al.: Lysine acetylation and succinylation in
regulates the cellular PDH activity. Biochem Biophys Res HeLa cells and their essential roles in response to UV-
Commun 2014, 451:288–294. induced stress. Sci Rep 2016, 6:30212.
45. German NJ, Yoon H, Yusuf RZ, Scadden DT, Kaelin WG, 55. Zhou L, Wang F, Sun R, Chen X, Zhang M, Xu Q, Wang Y,
Haigis MC: PHD3 loss in cancer enables metabolic reliance on Wang S, Xiong Y, Guan K: SIRT5 promotes IDH2 desucciny-
fatty acid oxidation via deactivation of ACC2. Mol Cell 2016, lation and G6PD deglutarylation to enhance cellular antioxi-
63:1006–1020. dant defense. EMBO Rep 2016, 36:1–12.
46. Theurillat JP, Udeshi ND, Errington WJ, Svinkina T, Baca SC, 56. Marín de Mas I, Aguilar E, Jayaraman A, Polat IH, Martín-
Pop M, Wild PJ, Blattner M, Groner AC, Rubin MA, Moch H, Bernabé A, Bharat R, Foguet C, Milà E, Papp B, Centelles JC,
Privé GG, Carr SA, Garraway LA: Prostate cancer. Ubiq- et al.: Cancer cell metabolism as new targets for novel
uitylome analysis identifies dysregulation of effector sub- designed therapies. Future Med Chem 2014, 6:1791–1810.
strates in SPOP-mutant prostate cancer. Science 2014 Oct 3,
346:85–89. 57. Megchelenbrink W, Katzir R, Lu X, Ruppin E, Notebaart R a:
 Synthetic dosage lethality in the human metabolic
47. Xu YM, Wang HJ, Chen F, Guo WH, Wang YY, Li HY, Tang JH, network is highly predictive of tumor growth and cancer
Ding Y, Shen YC, Li M, et al.: HRD1 suppresses the growth and patient survival. Proc Natl Acad Sci U S A 2015, 112:
metastasis of breast cancer cells by promoting IGF-1R 12217–12222.
degradation. Oncotarget 2015, 6:42854–42867. This study presents a method that can sistematically identify metabolic
vulnerabilities specific for cancer cells overexpressing a given meta-
48. Han C, Yang L, Choi HH, Baddour J, Achreja A, Liu Y, Li Y, Li J, bolic gene.
Wan G, Huang C, et al.: Amplification of USP13 drives ovarian
cancer metabolism. Nat Commun 2016, 7:13525. 58. Yizhak K, Chaneton B, Gottlieb E, Ruppin E: Modeling cancer
metabolism on a genome scale. Mol Syst Biol 2015, 11.
49. Liu K, Li F, Han H, Chen Y, Mao Z, Luo J, Zhao Y, Zheng B, 817–817.
 Gu W, Zhao W: Parkin regulates the activity of pyruvate
kinase M2. J Biol Chem 2016, 291:10307–10317. 59. König M, Bulik S, Holzhütter H-G: Quantifying the contribution
This work elucidated the role of Parkin in tumor cell metabolism by of the liver to glucose homeostasis: a detailed kinetic model
ubiquitination of PKM2. of human hepatic glucose metabolism. PLoS Comput Biol
2012, 8:e1002577.
50. Bian X, Chen H, Yang P, Li Y, Zhang F, Zhang J, Wang W,
Zhao W, Zhang S, Chen Q, et al.: Nur77 suppresses hepato- 60. Stanford NJ, Lubitz T, Smallbone K, Klipp E, Mendes P,
cellular carcinoma via switching glucose metabolism toward Liebermeister W: Systematic construction of kinetic models
gluconeogenesis through attenuating phosphoenolpyruvate from genome-scale metabolic networks. PLoS One 2013, 8.
carboxykinase sumoylation. Nat Commun 2017, 8:1–14.
61. Khodayari A, Maranas CD: A genome-scale Escherichia coli
51. Tan M, Peng C, Anderson KA, Chhoy P, Xie Z, Dai L, Park J, kinetic metabolic model satisfying flux data for multiple
Chen Y, Huang H, Zhang Y, et al.: Lysine glutarylation is a mutant strains. Nat Commun 2016, 7:1–12.
protein posttranslational modification regulated by SIRT5.
Cell Metab 2014, 19:605–617.

www.sciencedirect.com Current Opinion in Systems Biology 2017, 3:161–169

You might also like