You are on page 1of 6

Gene 726 (2020) 144158

Contents lists available at ScienceDirect

Gene
journal homepage: www.elsevier.com/locate/gene

Review

Crucial players in glycolysis: Cancer progress T



Zaka Abbaszadeh, Selin Çeşmeli , Çığır Biray Avcı
Ege University, Medical School, Department of Medical Biology, Turkey

A R T I C LE I N FO A B S T R A C T

Keywords: Cancer is the second most important cause of death and new therapy modalities continue to be developed and
Glcolysis evolved. Cancer cells’ metabolism is far different from the normal, healthy cells; they are more metabolically
Oxidative phosphorylation (OXPHOS) active, have higher proliferation rate and could able to resist to cell death pathways like apoptosis. It is known
Cancer that in addition to increasing the expression of enzymes that are crucial in glycolysis for much more energy
Glycolytic enzymes
production, cancer cells produce energy from lactic acid fermentation after glycolysis. In 1920s, Warburg has
claimed that cancer cells are more active in glycolysis than normal cells and use much more glucose in order to
obtain more ATP for metabolic activities, then this is named as Warburg effect. After that; new methodologies
and therapeutics that target metabolism, began to be attractive subject in cancer studies. Therefore, the main
genes, enzymes and factors are begun to investigate and further studied for understanding their roles in meta-
bolism of cancer cells.

1. Introduction some cancer types (Ancey et al., 2018).


There are 14 GLUT proteins and they are grouped into three classes,
1.1. Glucose transporters these different GLUTs are responsible in development of embryo and
have different affinities for glucose, fructose and mannose. In cancer
Solute carriers (SLC) which are integral membrane transport pro- cells, glucose uptake is higher than healthy cells and it is achieved by
teins and responsible for carrying metabolites, include glucose trans- the GLUTs, specially by GLUT1. Therefore, the overexpression of
porters. Glucose transporters allow glucose and other nutrient sources GLUT1 that is maintained by growth factors, is thought to be associated
to enter from the hydrophobic cell membrane, they have two families with malignancy. In addition, as the growth factor deprivation occur in
that are secondary active Na+/glucose cotransporter (SGLT) and fa- the healthy cells, GLUT1 is internalized in lysosomes and degraded;
cilitative sugar transporter (GLUT). SGLT family has two members that therefore, glucose uptake and metabolism decrease, and cells die. On
are SGLT1 and SGLT2, and their expression is found to be decreased in the other hand, cancer cells overexpress GLUT1 and glucose

Abbreviations: Abhd5, α/β-hydrolase domain containing 5; Acetyl-CoA, acetyl coenzyme A; ADP, adenosine diphosphate; AMPK, adenosine monophosphate ac-
tivated protein kinase; ATP, adenosine triphosphate; CAV1, caveolin-1; c-Met, tyrosine-protein kinase Met or HGFR; CO2, carbon dioxide; CRC, colorectal cancer;
DERL3, derlin-3; DHAP, dihydroxyacetone phosphate; DNA, deoxyribonucleic acid; EGFR, epidermal growth factor receptor; EMT, epithelial–mesenchymal tran-
sition; ENO, enolase; FH, fumarate hydratase; F-1,6-P, fructose-1,6-biphosphate; GAPDH, glyceraldehyde phosphate dehydrogenase; GLUTs, glucose transporters;
GLUT1, glucose transporter-1; GLUT3, glucose transporter-3; GLUT4, glucose transporter-4; GRP78, glucose-regulated protein 78; GTPase, guanosine tripho-
sphatases; G3P, glyceraldehyde-3-phosphate; G6P, glucose-6-phosphate; G6PD, glucose-6-phosphate dehydrogenase; HGFR, hepatocyte growth factor receptor; HIF,
hypoxia-inducible factor; HIF-1, hypoxia-inducible factor-1; HIF-1α, hypoxia-inducible factor-1α; HK, hexokinases; HMGA1, High Mobility Group AT-Hook 1; HRE,
hypoxia responsible element; HSPs, heat shock proteins; H2B, histone 2B; IDH1, isocitrate dehydrogenase 1; LDH, lactate dehydrogenase; LncRNA, long non-coding
RNA; LUAD, lung adenocarcinoma; miRNAs, MicroRNAs; mTOR, mammalian target of rapamycin; mTORC1, mTOR complex 1; NAD, nicotinamide adenine dinu-
cleotide; NADH, nicotinamide adenine dinucleotide–hydrogen; NSCLC, non-small cell lung cancer; OXPHOS, oxidative phosphorylation; PDH, pyruvate dehy-
drogenase; PDK, pyruvate dehydrogenase kinase; PEP, phosphoenolpyruvate; PET, positron emission tomography; PFKFBs, phosphofructo-kinase/fructosebipho-
sphatases; PFKM, phosphofructokinase M; PFK2, phosphofructokinase 2; PGI, phosphoglucose isomerase; PGK, phosphoglycerate kinase; PGM, phosphoglycerate
mutase; PIP3, phosphatidylinositol (3,4,5)-trisphosphate; PI3K, phosphatidyl-inositol 3-kinase; PK, pyruvate kinase; PPP, pentose phosphate pathway; PTTG, pi-
tuitary tumour-transforming gene; ROS, reactive oxygen species; RTK, receptor tyrosine kinase; SDH, succinate dehydrogenase; SGLT, secondary active Na+/glucose
cotransporter; SGLT1, secondary active Na+/glucose cotransporter; SGLT2, secondary active Na+/glucose cotransporter; SLC, solute carriers; TCA, tricarboxylic
acid; TIGAR, TP53-induced glycolysis and apoptosis regulator; TPI, triosephosphate Isomerase; TRAP-1, TNF-receptor-associated protein-1; TSC1, TSC complex
subunit 1; TSC2, TSC complex subunit 2; VDAC, voltage-dependent anion channel; VEGF, vascular endothelial growth factor; 3′UTR, 3′-untranslated region

Corresponding author.
E-mail address: selcesmeli.sc@gmail.com (S. Çeşmeli).

https://doi.org/10.1016/j.gene.2019.144158
Received 20 May 2019; Received in revised form 3 October 2019; Accepted 4 October 2019
Available online 17 October 2019
0378-1119/ © 2019 Elsevier B.V. All rights reserved.
Z. Abbaszadeh, et al. Gene 726 (2020) 144158

Fig. 1. GLUT1 glucose transportation into the cell. Figure shows the differences in glycolytic process between normal and cancer cells. In normal cells, glucose is
converted to ATP and CO2 through oxidative phosphorylation. But in cancer cells; after the conversion of glucose to pyruvate, lactate is produced.

metabolism is maintained hence the cells become resistant to apoptosis. Interleukin-3, GTPase Rab11a and phosphatidyl-inositol 3-kinase
There is also another glucose transporter, GLUT3, which is considered (PI3K) are other factors that play role in modulating GLUT1 expression
as the second most important transporter in cancer cells. GLUT1 and by trafficking it through the cell (Gonzalez-Menendez, 2018).
GLUT3 have amino acid sequences with 64% similarity and GLUT3 is
shown to be overexpressed in bladder cancer. In general, GLUT1 and 1.3. Glut3
GLUT3 express highly in most cancer types and result in poor survival
rates (Gonzalez-Menendez, 2018). Several factors and signalling pathways are known to participate in
GLUT3 regulation. Caveolin-1 (CAV1) stimulates GLUT3 transcription
1.2. Glut1 by activating nuclear localization of High Mobility Group AT-Hook 1
(HMGA1) which binds to promoter region of GLUT3 in colon cancer cell
GLUT1 is activated in response to HIF-1α (hypoxia-inducible factor- lines. Moreover, mammalian target of rapamycin (mTOR) pathway
1α), and other HIF-1α related factors such as (vascular endothelial upregulates GLUT3 and via NF-ĸB pathway, mTOR complex 1
growth factor) VEGF receptor and calcium channel transactivation help (mTORC1) enhances GLUT3 expression. In lung adenocarcinoma
the synthesis of GLUT1. GLUT1 is over expressed in Burkitt’s lymphoma (LUAD), it was shown that there is a link between EGFR signalling and
as a result of MYC-originated chromosomal translocations and muta- GLUT3 expression, mutated EGFR causes elevated glucose uptake and
tions in both K-Ras and epidermal growth factor receptor (EGFR) on- glycolysis with gain-of-function property. In addition, OCT4 protein has
cogenes result in increased expression of GLUT1 in non-small cell lung shown to be involved in the regulation of GLUT3 because of its co-
cancer (NSCLC). In K-Ras or BRAF mutant colorectal cancer (CRC) cells operating functions with HIF-1α.
which is advantage for these mutant cells with providing survival ca- As in the GLUT1, an EMT transcription factor ZEB1 is involved in
pacity in low glucose conditions, GLUT1 expression is higher than non- binding and activating enhancer of GLUT3, shown in hepatocellular
mutant or wild type CRC cells (Fig. 1). and NSCLC cells.
There are microRNAs (miRNAs) that regulate GLUT1; miR-144 and In NSCLC cell lines, GLUT3 expression is induced after treatment
miR-132 are two of them and they are shown to decrease GLUT1 ex- with histone deacetylase inhibitors. 3′-untranslated region (3′UTR) is
pression in lung and prostate cancer cells, decreased expression of targeted bymiR-195-5p in bladder cancer; glucose uptake and cell
GLUT1, increases glucose uptake and glycolysis. Another factor reg- growth are decreased.
ulating GLUT1 is DNA methylation; in a study, Derlin-3 (DERL3) – a
tumour suppressor – was transcriptionally silenced by hypermethyla- 1.4. Gluts and cancer therapies
tion of its promoter CpG island. The study has shown that DERL3 hy-
permethylation and loss is associated with increase in GLUT1 expres- GLUTs have essential roles in detection and diagnosis of cancer
sion. This shortened the relapse-free survival in CRC and Warburg effect diseases in some ways. In positron emission tomography (PET) scan,
was seen in HCT-116 CRC cells. which is one of the non-invasive techniques for examining tumours, is
Another way for controlling glucose uptake in cancer cells is the based on glucose uptake and glycolysis rate in tumour cells. PET scan
regulation of the GLUT1 modifications. In cancer cells, mutated p53 uses radiotracer that is a glucose-analogue 2-deoxy-2-[fluorine-18]
causes GLUT1 translocation to the plasma membrane and provoke fluoro-D-glucose and this radiotracer is crossed the membrane of cells
Warburg effect, whereas wild type p53 inhibits GLUT1 expression. by the help of GLUTs.

2
Z. Abbaszadeh, et al. Gene 726 (2020) 144158

Fig. 2. The genes and factors involved in glycolytic pathway in cancer cells. The role of each gene/factor is described in the related section.

Chemotherapy is principal method that has been applied for tu- lonidamine, astragalin, resveratrol, chrysin, GEN-27 and benserazide.
mours for many years, but cells could become resistant to this therapy 3-bromopyruvate is also able to decrease ATP level and activate cell
with many ways just as drug degradation, drug inhibition by several death; for example, in breast cancer it promotes apoptosis (Akins et al.,
mechanisms and alterations in cellular export. Studies showed that 2018).
GLUTs are also involved in these mechanisms; it was seen that in Except glycolysis, hexokinases also have other functions; for anti-
bevacizumab-resistant tumours show higher glucose uptake, glycolysis apoptotic activity they translocate to mitochondrial outer membrane
and these cells also overexpress GLUT3. and by interacting Bax protein and voltage- dependent anion channel
Similar results were claimed for GLUT1 in radiotherapy resistance; (VDAC), inhibit cytochrome c releasing and caspase-9-dependent
this study showed that the overexpression of GLUT1 is associated with apoptosis. In addition, glioma studies showed that in the absence of
radiation-resistance in cervical squamous cell carcinoma. These out- HK2, cell proliferation and angiogenesis decrease; cell death is in-
comes show that the expression of glucose transporters must be ex- creased (Pastorino et al., 2002; Wolf et al., 2011; Bayley and Devilee,
amined before the therapy and the clinicians must consider their pos- 2012).
sible effects on the treatment method (Ancey et al., 2018).
2.1.2. Phosphoglucose isomerase (PGI)
2. Genes and factors in cancer glycolysis On the other hand, another enzyme takes place in the glycolytic
mechanism which is phosphoglucose isomerase (PGI) and it converts
2.1. Genes in glycolytic pathway G6P to fructose-6-phosphate (Akins et al., 2018).

Glycolysis is regulated by several genes and enzymes, and many 2.1.3. Phosphofructo-kinase/fructose biphosphatases (PFKFBs)
regulatory factors are involved in this process (Fig. 2). These key factors Fructose-1,6-biphosphate (F-1,6-P) formation is carried out by
are known to be altered and expressed abnormally in many cancers. As PFKFBs and this enzyme is found to be overexpressed in many cancer
mentioned before, GLUTs are first regulator of glycolytic pathway be- cells. The overexpression of PFKBs are depend on not only HIF-1α, Ras,
cause of its role in taking glucose into the cells and they are upregulated c-Myc or p53; also, microenvironment, ATP and reactive oxygen species
by c-Myc, Ras oncoproteins and HIF-1; Ras molecule, which is an on- (ROS) levels and pH could control the PFKBs’ expression.
cogene, provokes glycolytic pathway in mutated form (Yu et al., 2016).
2.1.4. Aldolase (ALDO)
2.1.1. Hexokinases (HKs) Then fructose-bisphosphate is converted into glyceraldehyde 3-
Hexokinases (HKs) are enzymes that have role in phosphorylating phosphate by the enzyme, ALDO and it is proved that a form of ALDO
glucose and generating glucose-6-phosphate (G6P) which is the first enzymes, ALDO B is overexpressed in cancer cells (Zhang et al., 2017).
step of both glycolysis and pentose phosphate pathway (PPP). HK2 is a
common type of HKs and its expression is controlled by c-Myc and HIF- 2.1.5. Glyceraldehyde phosphate dehydrogenase (GAPDH)
1; also, in malignant tissues HK2′s activity is found to be upregulated. Next, 1,3-bisphosphoglycerate is formed by dehydrogenation by the
The inhibition of HK2 is taken into consideration in many studies help of GAPDH which is upregulated in the breast and colorectal can-
and many agents such as 3-bromopyruvate, 2-deoxyglucose, cers (Dai et al., 2015; Du et al., 2016). GAPDH is also able to translocate

3
Z. Abbaszadeh, et al. Gene 726 (2020) 144158

for DNA replication, in oxidative stress conditions. activated, cell division speed increases due to the inhibition of genes
that are antiproliferative functions (Miller et al., 2012). In addition, c-
2.1.6. Phosphoglycerate kinase (PGK) Myc can promote apoptotic pathway.
PGKtransfers one phosphate group from 1,3-bisphosphoglycerate to The expression of c-Myc is regulated by some signals. When positive
ADP and result in the production of ATP and 3-phosphoglycerate. With signals are present which is observed especially in tumour cells, c-Myc
increasing rate of glycolysis and tumour invasion in pancreatic and transcription and protein levels increase, then proliferation is induced.
gastric adenocarcinoma, increased PGK1 expression has been thought In addition to the positive signals, post-translational modifications,
(Xu et al., 2005; Xu et al., 2017). including mutations, stimulate overexpression of c-Myc. On the other
hand, miRNAs regulate c-Myc expression and, they are also targeted by
2.1.7. Phosphoglycerate mutase (PGM) and Enolase (ENO) c-Myc.
Then, 3-phosphoglycerate is converted to 2-phosphoglycerate by C-Myc targets many genes involved in cellular functions. These
PGM enzyme and ENO transform 2-phosphoglycerate to phosphoe- genes include cyclins A2, D2, E1 and ENO, LDHA, serine hydro-
nolpyruvate (PEP). These two enzymes are also upregulated in cancer xymethyl transferase, cadherin, integrin and others that have role in
cells such as melanoma and breast cancer. metabolism, translation, transformation and proliferation.
One of the most important duties of c-Myc is in glycolysis, LDHA is
2.1.8. Pyruvate kinase (PK) targeting. In several studies, LDHA is showed to be downstream target
By transferring phosphate group from PEP to ADP, PK enzyme that stimulates Warburg effect. Human pituitary tumour-transforming
generates pyruvate and an ATP molecule. PKL and PKM genes code four gene (PTTG) represses c-Myc, PKM2, GLUT1 and LDHA. However,
PKs consisting of L, R, M1 and M2 iso enzymes and it is known that c- overexpressed c-Myc can inhibit PTTG and this proves that PTTG is able
Myc and HIF-1 upregulate the expression of PKM2 that is overexpressed to modulate the metabolism in c-Myc-dependent manner (Yu et al.,
in cancer cells. 2016).
P53 gene, which is a tumour suppressor, represses PGM’s activity In the absence of c-Myc expression, it downregulates LDHA and
and this results in senescence and limitless proliferation. A substrate of lactate production decreases. Also; GLUT1, HK2, ENO and phospho-
PK, PEP phosphorylates proteins for example PGM and when PKM2 is fructokinase M (PFKM) are other targets that result in Warburg effect.
absent it generates pyruvate. Thus, PKM2 overexpression is thought to Under hypoxic conditions, ENO1 causes the generation of MBP-1 that is
plays role in other functions in tumour cells (Hu et al., 2014). The a translation initiation product and negative regulator of c-Myc and
identification of hypoxia responsible element (HRE) within the PKM2′s therefore this results in negative feedback loop (Miller et al., 2012). c-
intron showed that activity of PKM2 is regulated by HIF-1 (Courtnay Myc also controls the expression of phosphofructokinase 2 (PFK2), PK
et al., 2015). Reduction of PKM2 shown in mice, increases the tumour and glucokinase that are involved in carbohydrate metabolism (Yu
formation and thus it was understood that PKM2 is not required for cell et al., 2016). Because of its roles in essential functions of cells, c-Myc is
proliferation in cancers (Israelsen et al., 2013). According to the stu- considered in developing new cancer therapies.
dies, PKM2 switches to PKM1 to reverse Warburg effect and this
showed that aerobic glycolysis is depend on PKM2 (Courtnay et al., 2.2.2. HIF
2015). HIF is the regulator of cell proliferation and it is involved in tran-
scriptional regulation. As is evident from its name, HIF is stimulated in
2.1.9. Lactate dehydrogenase (LDH) hypoxia. When oxygen is present, HIF degradation occurs by ubiquiti-
In addition to these enzymes, cancer cells use LDHsfor reducing nation (Jun et al., 2017). But in hypoxic conditions, HIF-α and HIF-β
pyruvate to lactate. subunits are form dimer and result in HIF-1 product. After the activa-
LDH regulates histone 2B (H2B) and cell cycle by binding to Oct-1 tion of HIF-1, oxygen levels increase and angiogenesis is induced thus
trans activator in normal conditions; but when the NAD+/NADH level the tumour cells take much more nutrient. On the other hand, HIF-1
becomes low, lactate production is started by LDH (19). Reversely, high regulates the GLUTs activity and increase the expression of HK, PDH
ratio of NAD+/NADH causes movement of LDH to the nucleus and and LDH. PDH is responsible for converting pyruvate to acetyl-CoA for
progression in cell cycle by the help of transcription factors (Sun et al., tricarboxylic acid (TCA) cycle and PDK-1 inhibits the activity of PDH by
2011). phosphorylating then prevents pyruvate to enter TCA cycle (Yu et al.,
LDHA-encoded M subunit of LDH is found abundantly in tumours. 2016). Therefore, PDK-1 inhibitors are thought to be efficient in cancer
Inhibition of LDH’s activity causes the translocation of it to the nucleus, therapies.
cell proliferation increases. On the other hand, as LDH activity increases De-functioning of long non-coding RNAs (lncRNAs) and miRNAs
lactate production increases too and cancer cells are killed due to may also control HIF-1α and stimulate aerobic glycolysis. In addition,
acidosis. LDH is also used in diagnosis by using as a biomarker in some hypoxic conditions and ROS is other parameter that regulates HIF-1α
diseases like cancer (Hardie et al., 2012). expression (Yu et al., 2017).
In addition, 14-3-3ζ-mediated upregulation of LDHA in breast cells HIF-1α is the main factor in activating Warburg effect and it is
showed to increase glycolysis rate and therefore is thought to cause stimulated by other mutated factors, then GLUTs and enzymes parti-
breast cancer formation (Chang et al., 2016). cipate in glycolysis. During this process, tumour development begins by
In addition to the enzymes take place in glycolytic pathway and at regulating the PI3K-Akt-mTOR pathway and its elements. PI3K and Akt
the end of glycolysis, pyruvate dehydrogenase (PDH) is inactivated by have essential role in glucose metabolism. PI3K can be activated by
pyruvate dehydrogenase kinase (PDK), which is upregulated by HIF and several factors like receptor tyrosine kinases (RTKs), platelet-derived
c-Myc. Also, acetyl coenzyme A (acetyl-CoA) formation is inhibited by growth factor receptor (PDGFR), insulin-like growth factor receptor and
blocking pyruvate import into the mitochondrial matrix (Bayley and EGFR; it is deregulated by Pten phosphatase and Pten therefore inhibits
Devilee, 2012). glycolysis. Akt, which is a serine/threonine kinase, regulates HK2,
GLUT1 and PFK2 and provides the activation of aerobic glycolysis and
2.2. Other genes and factors in glycolysis growth of cancer cells. mTOR, that is a serine/threonine kinase just like
Akt and involved in tumorigenesis (Yu et al., 2016). In consideration of
2.2.1. c-Myc oncogene these factors, many researchers has focused on metabolism in cancer
c-Myc gene, which is activated by many pathways in cancer cells, is studies. In this year; William Kaelin Jr., Sir Peter Ratcliffe and Gregg
the master regulatory element in metabolism and cell proliferation; Semenza, who are proffessors from different universities currently,
also, it causes malignancy with changes in metabolism. When c-Myc is have been awarded Nobel Prize in Medicine for their discoveries of how

4
Z. Abbaszadeh, et al. Gene 726 (2020) 144158

cells sense and adapt to oxygen availability. The idea of cells’ need for helps the cells in both death and repair decisions. However, mutated
energy is a crystal-clear fact, however the way they use it and effect in p53 could not bind DNA and therefore the cell division occur in un-
changes in the oxygen levels on the cells were not known until the desirable way and resulting in tumorigenic cells with damaged DNA
argument of this precious scientists. While Ratcliffe were investigating (Courtnay et al., 2015). Moreover, glucose metabolism that is regulated
the effect of oxygen on the regulation of EPO which is a gene normally by p53 could be depend on the NF-ĸB transcription factor. GLUT1,
expressed in kidneys, Gregg Semenza has studied the EPO gene and its GLUT3 and GLUT4′s expression is downregulated depend on p53 and
regulatory ways in different oxygen levels. In Semenza’s Specific DNA PGM degradation and TP53-induced glycolysis and apoptosis regulator
segments are located the region near to EPO gene, which claimed the (TIGAR) is regulated by p53. p53 inhibits glucose-6-phosphate dehy-
response to hypoxic conditions. These studies has shown that the ex- drogenase (G6PD) and this way regulates metabolism. Furthermore;
istence of oxygen sensing mechanisms in all tissues (Semenza, 1991). activation of certain genes such as Pten, adenosine monophosphate
After that Semenza identified a complex called hypoxia- inducible activated protein kinase (AMPK) by p53, causes negative regulation of
factor (HIF) that binds to specific DNA by depending on oxygen PI3K/Akt/mTOR signalling. Pten leads to PI3K/Akt signal inhibition,
(Semenza, 1995). In the light of latter studies, they began to search for whereas AMPK causes mTOR downregulation (Yu et al., 2016).
the answer about the binding of ubiquitin to HIF -1α, and Kaelin come AMPK regulates metabolism between anabolic and catabolic pro-
to the help. Because he showed that VHL, that is amutated gene in von cesses; when it is activated anabolism is turned off and protein pro-
Hippel-Lindau’s disease, is able to prevent to onset of cancer by en- duction is repressed by inhibiting translation that is depend on mTOR.
coding a protein. In addition, Kaelin proved that hypoxia regulated These proteins include HIF-1α, therefore the enzymes involved in gly-
genes are highly found in cancer cells that lacks of functional VHL gene. colysis is downregulated and oxidative phosphorylation (OXPHOS) is
This showed that VHL involve in the controlling the responses to hy- preferred by the cell (Yu et al., 2016).
poxic conditions. Then Ratcliff and his friends claimed that VHL can
interact with HIF-1α and also in normal oxygen levels, it is essential for 2.2.5. TIGAr
the degradation (Ratcliffe, 1999). In 2001, they revealed that hydroxyl Another factor that is involved in regulation of glycolytic pathway is
groups are added to HIF-1α at specific positions under normal oxygen TIGAR, which also controls oxidative stress and glycolysis in a cell.
conditions and this modiciation is named as prolyl hydroxylation which TIGAR is translated from the TIGAR gene which is located on chro-
allows recognition and binding of HIF-1α by VHL. More research mosome 12p13-3. Apoptotic signals that are associated to ROS are
showed that oxygen-dependent hydroxylation controls the gene acti- decreased because of lowering in ROS levels by TIGAR, thus it is con-
vation carried out by HIF-1 (Kaelin, 2001; Ratcliffe, 2001). The studies sidered as a mediator of the tumour suppression by p53. In addition to
of these three nobel laureates have marked an era for upcoming studies decreasing ROS levels and controlling survival, TIGAR plays role in
to fight with diseases like cancer, anemia with these pioneer dis- DNA repair (Bensaad et al., 2006).
coveries. TIGAR, as a tumour suppressor, degrades fructose-2,6-bisphosphate
and block glycolysis and causes the G6P precursor accumulation, hence
2.2.3. PI3K/Akt/mTOR glucose catabolism turns into PPP. When TIGAR is in an oncogene role,
PI3K molecule includes lipid enzymes that are able to phosphorylate for the protection the carcinoma cells from the apoptosis; this process is
–OH groups on the plasma membrane. Several studies have showed the stimulated by c-Met (Hu et al., 2014). However, whether presence or
roles of PI3K in metabolism, survival and cancer progression because of the level of TIGAR expression affects tumour formation is not fully
regulating glucose uptake and PI3K via Akt moderate the expression of known yet and upcoming studies is necessary.
GLUT1. PI3K classified in several categories and class I subclass 1A is
one of them, which is activated by RTKs. PI3K helps phosphoinositide- 2.2.6. Triosephosphate isomerase (TPI)
dependent kinase-1 (PDK1) and protein kinase b (Akt) for binding to Triosephosphate isomerase, also known as TPI, is a homodimeric
phosphatidylinositol (3,4,5)-trisphosphate (PIP3) molecule, results in enzyme that is translated from TPI gene and it participates in invasion
uncovering the phosphorylation sites of Akt and then the first phos- and migration. TPI is abundantly expressed in many cancer cells in-
phorylation of it. mTOR, which is activated by PI3K allows the second cluding urinary cancer, lung cancer and squamous cell lung carcinoma,
phosphorylation of Akt and its complete activation process. Active Akt and it is located on 12p13, near to TIGAR (Lincet and Icard, 2015). In
is involved in cell cycle progression, growth, cell death and GLUT1- addition to glycolytic activity by conversion of dihydroxyacetone
mediated glucose uptake regulation (Courtnay et al., 2015). mTOR phosphate (DHAP) to glyceraldehyde-3-phosphate (G3P) or vice versa,
regulates the expression of c-Myc, NF-ĸB and HIF-1α and it also pro- TPI plays role in cell growth (Chen et al., 2017). Activation of TPI en-
mote PKM2 which is critical in growth and aerobic glycolysis. PKM2 zyme results in ATP production and inhibition of the enzyme result in
disruption occurs by the help of c-Mycand inhibits tumorigenesis that is the orientation dihydroxyacetone to PPP. Transcription of gene ex-
mediated by mTOR. In addition, tumour suppressor TSC complex sub- pression under stress conditions is achieved by ribose that is produced
unit 1 (TSC1) and TSC complex subunit 2 (TSC2) gene complex reg- by PPP and this is the regulation of metabolic pathway controlled by
ulates GLUT3 expression and upregulation of GLUT3 is achieved with TPI. When phosphoenolpyruvate accumulation TPI supresses activity
the help of mTORC1 and by NF-ĸB signalling (Yu et al., 2016). thus promotes PPP pathway and protect cells from ROS. In addition, TPI
Moreover; Ras, isocitrate dehydrogenase 1 (IDH1), fumarate hy- upregulation can be involved in reversing multidrug-resistance pheno-
dratase (FH) and succinate dehydrogenase (SDH) are linked to glyco- type and therefore it is thought to be an anti-drug-resistant enzyme in
lysis with the HIF-1 activation. The activation of these, result in in- some cancers such as gastric cancer (Lincet and Icard, 2015; Chen et al.,
creased expression of genes involved in glycolysis (Miller et al., 2012). 2017).
K-Ras activation results in dysfunctional mitochondria and thereby
switch to glycolysis. When K-Ras is mutated, GLUT1 expression in- 3. Other regulators of glycolysis
creases and so glycolysis and lactate production (Yu et al., 2016).
In addition to the genes, enzymes and several factors; there are
2.2.4. p53 other regulators involved in glycolytic pathway (Neugent et al., 2018).
p53 is a tumour suppressor gene that is involved in both tran- Wnt signalling is one of them and it interacts PDK-1 in controlling
scription and regulation of cellular processes, importantly cell death. It glycolysis. When Wnt is repressed, overexpressed PDK-1 recovers the
causes the initiation of cellular cell death pathways or DNA repair after cell in both glycolysis and growth of vessels. There is also focal adhe-
binding to DNA (Ozaki and Nakagawara, 2011). sion kinase (FAK) which helps glycolysis by inhibiting ENO, LDH,
p53 is an essential for functions of cell and when DNA is damaged, it PKM2 and other proteins. An ATP-gated channel, P2X7 receptor is

5
Z. Abbaszadeh, et al. Gene 726 (2020) 144158

responsible in aerobic glycolysis and regulates many glycolytic enzymes doi.org/10.2174/1568026618666180523111351.


(Chatterjee and Burns, 2017). In addition, HSP40 and TNF-receptor- Pastorino, J.G., Shulga, N., Hoek, J.B., 2002. Mitochondrial binding of hexokinase II
inhibits Bax-induced cytochrome c release and apoptosis. J. Biol. Chem. https://doi.
associated protein-1 (TRAP-1) that are members of heat shock proteins org/10.1074/jbc.M109950200.
(HSPs), participate in switching OXPHOS to aerobic glycolysis and vice Wolf, A., et al., 2011. Hexokinase 2 is a key mediator of aerobic glycolysis and promotes
versa. Moreover; glucose-regulated protein 78 (GRP78), ZBTB7A, sur- tumor growth in human glioblastoma multiforme. J. Exp. Med. https://doi.org/10.
1084/jem.20101470.
vivin, KLF4, α/β-hydrolase domain containing 5 (Abhd5) and GRIM-19 Bayley, J.-P., Devilee, P., 2012. The Warburg effect in 2012. Curr. Opin. Oncol. https://
gene are involved in the regulation of glycolysis with various ways (Yu doi.org/10.1097/CCO.0b013e32834deb9e.
et al., 2016). Zhang, F., et al., 2017. Elevated transcriptional levels of aldolase A (ALDOA) associates
with cell cycle-related genes in patients with NSCLC and several solid tumors.
BioData Min. https://doi.org/10.1186/s13040-016-0122-4.
4. Conclusion Dai, W., et al., 2015. By reducing hexokinase 2, resveratrol induces apoptosis in HCC cells
addicted to aerobic glycolysis and inhibits tumor growth in mice. Oncotarget.
https://doi.org/10.18632/oncotarget.3800.
As the incidence of cancer patients has increased and no exact cure
Du, Q., et al., 2016. Chemopreventive activity of GEN-27, a genistein derivative, in colitis-
has found; studies have focussed on different hallmarks of cancer. One associated cancer is mediated by p65-CDX2-β-catenin axis. Oncotarget. https://doi.
and the most important characteristics of cancer is hidden in cell me- org/10.18632/oncotarget.7554.
tabolism. After Warburg’s hypothesis, cancer researches in worldwide Xu, R.H., et al., 2005. Inhibition of glycolysis in cancer cells: A novel strategy to overcome
drug resistance associated with mitochondrial respiratory defect and hypoxia. Cancer
has been focussed on glucose metabolism and scientists have begun to Res.
investigate the reason behind the choosing aerobic glycolysis by the Xu, D., et al., 2017. Chrysin inhibited tumor glycolysis and induced apoptosis in hepa-
cancer cells. In the light of these studies, many oncogenes and tumour tocellular carcinoma by targeting hexokinase-2. J. Exp. Clin. Cancer Res. https://doi.
org/10.1186/s13046-017-0514-4.
suppressor, signalling pathways and factors involved in these pathways Hu, Z.Y., Xiao, L., Bode, A.M., Dong, Z., Cao, Y., 2014. Glycolytic genes in cancer cells are
have been discovered. Therefore, these genes are targeted in many re- more than glucose metabolic regulators. J. Mol. Med. https://doi.org/10.1007/
searches and various molecules, chemical, nanomedicines, inhibitors, s00109-014-1174-x.
Courtnay, R., et al., 2015. Cancer metabolism and the Warburg effect: the role of HIF-1
their analogues and also miRNAs are developed against them. However, and PI3K. Mol. Biol. Rep. https://doi.org/10.1007/s11033-015-3858-x.
mechanism behind the glycolytic switch and the role of the main player Israelsen, W.J., et al., 2013. XPKM2 isoform-specific deletion reveals a differential re-
should be determined in addition to the consideration of specificity of quirement for pyruvate kinase in tumor cells. Cell. https://doi.org/10.1016/j.cell.
2013.09.025.
this player and the agent toxicity before approval. As the 2019 Nobel Ratcliffe, P.J., 1999. The tumour suppressor protein VHL targets hypoxia-inducible fac-
Prize in physiology or medicine has awarded to Kaelin, Ratcliffe and tors for oxygen-dependent proteolysis. Nature. https://doi.org/10.1038/20459.
Semenza for their studies on cell metabolism and oxygen sensation and Ratcliffe, P.J., et al., 2001. Targeting of HIF-α to the von Hippel-Lindau ubiquitylation
complex by O2-regulated prolyl hydroxylation. Science. https://doi.org/10.1126/
adaptation to oxygen level alterations. They studied on the ways to
science.1059796.
sense oxygen by the cell and cell’s adaptation to alterions in oxygen Semenza, G.L., 1991. Hypoxia-inducible nuclear factors bind to an enhancer element
levels. And they described a machinery that regulates the genes’ activity located 3′ to the human erythropoietin gene. Proceedings of the National Academy of
in the case of changes in oxygen level. They drawn the attention of Sciences of the United States of America. https://doi.org/10.1073/pnas.88.13.5680.
Semenza, G.L., et al., 1995. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS
other scientists who are studying various fields of health and studies heterodimer regulated by cellular O2 tension. Proceedings of the National Academy
especially in cancer research concentrate on the metabolism. Further of Sciences of the United States of America. https://doi.org/10.1073/pnas.92.12.
studies justify the hope that immunotherapy may be a key point in 5510.
Sun, Q., et al., 2011. Mammalian target of rapamycin up-regulation of pyruvate kinase
future cancer treatments. Although the progressions in researches, there isoenzyme type M2 is critical for aerobic glycolysis and tumor growth. Proc. Natl.
are still dark areas in Warburg effect and thus still needed to further Acad. Sci. U.S.A. https://doi.org/10.1073/pnas.1014769108.
studies. But it is known that conventional chemotherapy with glycolytic Hardie, D.G., Ross, F.A., Hawley, S.A., 2012. AMPK: a nutrient and energy sensor that
maintains energy homeostasis. Nat. Rev. Mol. Cell Biol. https://doi.org/10.1038/
inhibitors is much more efficient in the both beginning and progression nrm3311.
of tumours and surpassing the drug resistance. Therefore, knowledge of Chang, C.-C., et al., 2016. Upregulation of lactate dehydrogenase a by 14-3-3ζ leads to
glycolytic players and inhibition of these elements will continue to be increased glycolysis critical for breast cancer initiation and progression. Oncotarget.
https://doi.org/10.1080/00288330.1991.9516485.
essential in cancer therapies.
Miller, D.M., Thomas, S.D., Islam, A., Muench, D., Sedoris, K., 2012. c-Myc and cancer
metabolism. Clin. Cancer Res. https://doi.org/10.1158/1078-0432.CCR-12-0977.
Declaration of Competing Interest Jun, C., Rathore, A., Younas, H., Gilkes, D., Polotsky, Y.V., 2017. Hypoxia-inducible
factors and cancer. Curr. Sleep Med. Rep.
Yu, L., Chen, X., Sun, X., Wang, L., Chen, S., 2017. The glycolytic switch in tumors: How
The authors declare that they have no known competing financial many players are involved? J. Cancer. https://doi.org/10.7150/jca.21125.
interests or personal relationships that could have appeared to influ- Ozaki, T., Nakagawara, A., 2011. Role of p53 in cell death and human cancers. Cancers.
ence the work reported in this paper. https://doi.org/10.3390/cancers3010994.
Bensaad, K., et al., 2006. TIGAR, a p53-inducible regulator of glycolysis and apoptosis.
Cell. https://doi.org/10.1016/j.cell.2006.05.036.
References Kaelin, J., et al., 2001. HIFα targeted for VHL-mediated destruction by proline hydro-
xylation: Implications for O2 sensing. Science. https://doi.org/10.1126/science.
1059817.
Ancey, P.B., Contat, C., Meylan, E., 2018. Glucose transporters in cancer – from tumor Lincet, H., Icard, P., 2015. How do glycolytic enzymes favour cancer cell proliferation by
cells to the tumor microenvironment. FEBS J. https://doi.org/10.1111/febs.14577. nonmetabolic functions? Oncogene. https://doi.org/10.1038/onc.2014.320.
Gonzalez-Menendez, P., et al., 2018. GLUT1 protects prostate cancer cells from glucose Chen, T., et al., 2017. Clinical significance and prognostic value of Triosephosphate
deprivation-induced oxidative stress. Redox Biol. https://doi.org/10.1016/j.redox. isomerase expression in gastric cancer. Medince (United States). https://doi.org/10.
2018.03.017. 1097/MD.0000000000006865.
Yu, L., Chen, X., Wang, L., Chen, S., 2016. The sweet trap in tumors: aerobic glycolysis Neugent, M.L., et al., 2018. A new perspective on the heterogeneity of cancer glycolysis.
and potential targets for therapy. Oncotarget. https://doi.org/10.18632/oncotarget. Biomol. Ther. https://doi.org/10.4062/biomolther.2017.210.
7676. Chatterjee, S., Burns, T.F., 2017. Targeting heat shock proteins in cancer: a promising
Akins, N.S., Nielson, T.C., Le, H.V., 2018. Inhibition of glycolysis and glutaminolysis: an therapeutic approach. Int. J. Mol. Sci. https://doi.org/10.3390/ijms18091978.
emerging drug discovery approach to combat cancer. Curr. Top. Med. Chem. https://

You might also like