You are on page 1of 7

© 2021 IJRAR January 2021, Volume 8, Issue 1 www.ijrar.

org (E-ISSN 2348-1269, P- ISSN 2349-5138)

Glycolytic Pyruvate Kinase Isoform PKM2 in


Cancer
Jaspreet Singh* and Balwinder Singh
P.G. Department of Biotechnology, Khalsa College, Amritsar – 143002, Punjab

Abstract :
Pyruvate kinase a glycolytic enzyme that catalyses the dephosphorylation of phosphoenolpyruvate into
pyruvate exists in four isoforms in mammals ie. PKL in the liver, PKR in erythrocytes, PKM1 in somatic cells
and PKM2 in cancer cells. PKM2 has potential to revive cellular homeostatic state to proliferative state,
thereby, converting normal cell to cancerous cell. PKM2 activity is regulated by various metabolites
contributing towards elevated levels of glycolytic intermediates which are to be utilized in anabolic reactions
leading to nucleic acid biosynthesis. Fructose-1,6-bisphosphate and triiodothyronine are allosterically
modulating the activity of PKM2. In cancer cells all other Pyruvate kinase isoforms are replaced by PKM2
which is encoded by alternative splicing of the PKM gene. Cancer cells appreciate PKM2 isoform because of
its uniqueness from the others by providing selective advantage in tumor growth. PKM2 activity supports the
survival of cancer cells under oxidative stress conditions. PKM2 comes out to be a critical player in
reprogramming the cellular metabolism facilitating the cancer cell growth. Understanding the role of this
enzymic isoform can pave the way for an early-stage cancer diagnosis and therapy, lighting a ray of hope to
ailing humanity.

Key words: Cancer, Pyruvate Kinase, PKM gene, PKM2 isoform.


1. INTRODUCTION
An unregulated quickened pace of growth and division of cancer cells is attributed to the altered metabolism
as compared to normal cells (Zhou et al., 2012). The cellular proliferation needs adequate metabolic
intermediates that can ensure the synthesis of cell building components along with sufficient energy (Mazurek,
2011). To carry out anabolic processes, the cancer cells use glucose excessively (Hsu and Sabatini, 2008); and
the consistent switch over to energy production pathways ie. from oxidative to glycolysis, an important
characteristic of cancerous cell metabolism (Kondoh et al., 2007). Hence, aerobic glycolysis appeared as a
hallmark of cancer cell growth (Toschi et al., 2010). Glycolysis generates energy for rapid tumor growth
promoting cancer metastasis (Gao and Chen, 2015). Along with infrequent mitochondrial defects studied in
the cancer cells (Koppenol et al., 2011; Ward & Thompson, 2012), A key glycolytic enzyme pyruvate kinase
isoform M2 (PKM2) that regulate the glycolysis pathway appeared as a component necessary for tumor
growth (Christofk et al., 2008a).

2. GLYCOLYSIS
Glycolysis is a metabolic pathway through which glucose is catabolized to pyruvate producing four
ATPs (Li et al., 2015). Normally, in cells the glycolysis is prioritized only in the limited supply of oxygen.
In the presence of oxygen, pyruvate undergoes the tricarboxylic acid cycle (TCA cycle) in mitochondria
for fully oxidation to CO2 and H2O (oxidative pathway) resulting production of 36 ATP molecules approx.
(Annibaldi and Widmann, 2010). after complete oxidation one glucose molecule produces 38 ATPs. In
tumor cells, the pyruvate oxidation is replaced by lactate production yielding no ATP, rection being
catalyzed by lactate dehydrogenase (LDH); thereby, only two ATPs per glucose are generated (Li et al.,
2015).

IJRAR21A1559 International Journal of Research and Analytical Reviews (IJRAR) www.ijrar.org 956
© 2021 IJRAR January 2021, Volume 8, Issue 1 www.ijrar.org (E-ISSN 2348-1269, P- ISSN 2349-5138)

3. PYRUVATE KINASE
Pyruvate kinase (PK) dephosphorylates the phosphoenolpyruvate to pyruvate as a rate-limiting step in
glycolysis (Lyssiotis et al., 2012; Lu, 2012a), releases a significant amount of energy for generation of one
ATP (Mazurek et al., 2002; Dombrauckas et al., 2005). Divalent cations (Mg2+ or Mn2+) are required for PK
activity. An essentially irreversible reaction under normal cellular conditions, is critical to control the central
metabolic flux, as both the substrate Phosphoenolpyruvate and its product pyruvate are involved in various
other metabolic pathways also (Valentini et al., 2000).

Pyruvate kinase
Phosphoenolpyruvate + ADP -------------------→ Pyruvate + ATP

4. PYRUVATE KINASE ISOFORMS


Four PK isoforms of (R, L, M1 and M2) exist in the mammals (Zhou et al., 2012a). Two isoforms Pyruvate
kinase R (PKR) and Pyruvate kinase L (PKL) are products of the common PKL gene but transcribed involving
different promoters (Wu & Le, 2013). PKR is found in erythrocytes whereas PKL is present in organs
performing gluconeogenesis like liver (Chen et al., 2010). Pyruvate kinase M1 (PKM1) is located found in
brain and adult skeletal muscles; while Pyruvate kinase M2 (PKM2) predominates in proliferating cells both
being the alternative splicing products of same PKM gene (Figure 1). Existence of PK isoforms helps
glycolysis to cope with variable metabolic situations in different cells/tissues (Iqbal et al., 2014).

Gene PKL PKM

Isoform PKL PKR PKM1 PKM

Figure 1. Genes and isoforms of pyruvate kinase

5. ISOFORM PKM2
Various factors ie. splicing repressors, polypyrimidine tract binding protein (hnRNPI) as well as
heterogeneous nuclear ribonucleoprotein A1& A2 (hnRNP A1/A2) control the PKM gene splicing and the
expression of repressors is upregulated by oncoprotein (Clower et al., 2010; David et al., 2010). These proteins
repress PKM1 mRNA splicing and contributes towards the increased expression of PKM2 isoform (Chen et
al., 2012). The isoform PKM2 exists in two states 1) a low-active dimeric state or 2) highly active tetrameric
state (Dang, 2009). During tumorigenesis, other isoforms of pyruvate kinase are replaced by PKM2, playing
an essential role in aerobic glycolysis (Oremek et al., 1999). PKM2 also acts like an on/off switch and its
expression in the cancer cells paves way to cellular proliferation. The shift of isoform PKM2 from the dimer
state to tetramer state changes its activity that regulates the speed of celllar proliferation (Wu and Le, 2013).
Cancer cells predominantly express the dimer with low-activity (Hitosugi et al., 2009) but the normal
proliferating cells express the tetramer with high-activity (Mazurek et al., 2005). Slow active dimer drives
glycolysis while the more active tetramer drives the tricarboxylic acid cycle (TCA) (Christofk et al., 2008b;
Vander Heiden et al., 2009). Each subunit in the homotetramer of four identical subunits is composed of 4
domains (Gupta and Bamezai, 2010).

6. PKM2 ACTIVITY
Low activity of PKM2 allowing the smooth operation of glycolysis customarily promotes the cellular
proliferation and help the cancer cells to stockpile metabolites upstream of PEP, thereby, supply sufficient
amounts of metabolites for the synthesis of precursors (Mazurek, 2011). The glycolytic intermediates
produced due to the low activity of PKM2, enters the glycerol biosynthesis and pentose phosphate pathway
(glycolysis branch pathways) generating reducing power NADPH for nucleotide biosynthesis and to suppress
ROS production also (Jiang et al., 2010; Hamanaka and Chandel, 2012).

IJRAR21A1559 International Journal of Research and Analytical Reviews (IJRAR) www.ijrar.org 957
© 2021 IJRAR January 2021, Volume 8, Issue 1 www.ijrar.org (E-ISSN 2348-1269, P- ISSN 2349-5138)

7. DIMER/TETRAMER SWITCHING
Homotetrameric isoform PKM2 can form nexus with additional enzymes too and upon convertion of
phosphoenolpyruvate to pyruvate, the lactic acid is formed quickly fulfilling the cellular energy needs
(Mazurek et al., 2001). The shifting of PKM2 from the dimer to tetramer state changes its activity that alters
the ratio of the generated energy to metabolic precursor and thereby, regulate the cellular proliferation pace
(Wu and Le, 2013). A number of factors effect the dimeric/tetrameric switching (Mazurek et al., 2007; Wu et
al., 2008) and fructose-1,6-bisphosphate, serine, tyrosine kinase, oncoproteins, reactive oxygen species (ROS)
are the major ones (Tamada et al., 2012). When PKM2 is in the inactive dimeric state, glucose carbon skeleton
is streamed into biosynthetic processes and hence, are no longer available for energy generation under
glycolysis. That necessitates the demand for an alternative source of energy production (Mazurek et al., 2005).
Consequently, cancer cells utilise glutamine excessively and thereby, proliferate even at diminished glucose
concentrations (Wu and Le, 2013).

8. DOMAIN STRUCTURE
Characterized from a large number of prokaryotic as well as eukaryotic sources, Pyruvate kinase is
considered as an evolutionary conserved enzyme. has been and in most of the cases, it has been found that the
composed of 4 identical subunits, the enzyme usually exist as a homotetramer. With each subunit composed
of 500 - 600 amino acids (approx.) depending upon the enzyme source. PKs from different species show high
degree of structural homology (Wang et al., 2001). As crystal structures of PKs from variable reports show
similar architecture. Each subunit shows 3 domains: A domain - (α/β)8 barrel, B domain - β-stranded
(irregular β-barrel folds) and C domain - (α/β) open-sheet (α+β organization) (Valentini et al., 2000). An
auxillary N-terminal fourth domain in eukaryotic PK, is however, not present in the prokaryotic ones (Jurica
et al., 1998).

9. LIGAND BINDING SITES


PK tetramers comprise three distinct ligand binding sites- (i) active site, (ii) effector site along with (iii)
amino acid binding site and exhibit cooperative binding for the substrate Phosphoenolpyruvate that may
enhance the glycolytic flux in response to Phosphoenolpyruvate concentration (Morgan et al., 2014). Location
wise active site is situated on the C-terminal end of A-domain (α/β)8 barrel, whereas, Fructose-1,6-
bisphosphate binding site is on C-domain (Jurica et al., 1998). Assembling of all four subunits result in a
tetrameric enzyme (Valentini et al., 2000).

10. PK ACTIVITY REGULATION


Two different mechanisms regulating PK activity in mammals are allosteric control and expression control.
In spite of the comparatively small size of catalytic site, the allosteric control is extended over a wide range
of regulatory molecules that involves reciprocal rotations of A- and C-domains within each subunit of tetramer
(Takenaka et al., 1991). The amino acid residues at domain and subunit interfaces function critically in
imparting the allosteric communicative signal from and to the catalytic as well as regulatory sites (Gupta and
Bamezai, 2010). However, the transmission of the allosteric communicative signal between the
Phosphoenolpyruvate and Fructose-1,6-bisphosphate binding sites may be through intra-subunit interface
between both A- and C-domains. (Valentini et al., 2000). Owing to the involvement of a variety of effector
molecules in allosteric response, the enzyme adopts two different conformations: R-state (active form) and T-
state (inactive from) (Wang et al., 2001). R and T forms propose two-state model showing differential
affinities for the ligands (Wooll et al., 2001). Binding of effector molecules trigger the transition between both
R and T-states (ie. Phosphoenolpyruvate homotropically). The multidomain architecture of pyruvate kinase is
instrumental in its regulation and activation that involves both domain and subunit rotations along with active
site geometry alterations. Amino acid residues on domain and subunit interfaces crucially communicate
between activator-binding site and catalytic centre (Valentini et al., 2002). During T-state to R-state transition,
domains of the functional tetramer modify their respective orientation which is coupled to the conformational
swap in the active centre; and that upon transition from R-state to the T-state there occurs a distortion in the
Phosphoenolpyruvate binding site (Valentini et al., 2000). The Phosphoenolpyruvate affinity as well as

IJRAR21A1559 International Journal of Research and Analytical Reviews (IJRAR) www.ijrar.org 958
© 2021 IJRAR January 2021, Volume 8, Issue 1 www.ijrar.org (E-ISSN 2348-1269, P- ISSN 2349-5138)

activity of the PKM2 isoform depends upon its quaternary shape and both metabolic intermediates as well as
the oncogenes work in its regulation (Mazurek, 2011).

11. PKM GENE SPLICING


Human PKM gene organisation contains 12 exons as well as 11 introns (Hasan, 2012). Mutually exclusive
alternative splicing of the PKM pre-mRNA results in two isoforms viz. PKM1 and PKM2, reflecting the
insertion of either exon-9 or exon-10 resp. (Yang and Lu, 2013). ie. PKM1 splice variant holds exon-9, while
PKM2 holds exon-10 (Hasan, 2012). Out of 531 amino acids, both PKM1 and PKM2 proteins vary only in 23
amino acids (Noguchi et al., 1986). Both mRNAs are composed of 1593 base pairs and differ from one another
by only 160 nucleotides from site 1143-1303 base pairs that corresponds to a variation of 23 amino acid
residues (Hasan, 2012). These 23 amino acids are concentrated in a stretch of 56 amino acid ranging from
378-434 amino acid residues at the carboxyl end of the PKM2 protein. out of the 56 amino acid long stretch
the 44 residues, that belong to the C-domain of the PKM2 protein participate in the association of the two
dimers to yield a tetrameric state (Dombrauckas et al., 2005).

12. PKM GENE EXPRESSION


Numerous conditions regulate PKM gene expression (Yamada and Noguchi, 1999b) as follows.
a) PKM1 is expressed in tissues/organs demanding high energy ie. brain and skeletal muscles. It possesses the
highest affinity towards substrate phosphoenolpyruvate and is not prone to allosteric regulation or
phosphorylation (Mazurek, 2011).
b) PKM2 is expressed in tissues that involves excessive nucleic acid biosynthesis ie. fetal tissues. Initially,
identified in the hepatoma cell line, its raised levels are recorded in the cancerous cells (Kato et al., 1989;
Obata et al., 1989; Dombrauckas et al., 2005). The promoter sequence of the PKM gene consists of the three
cis-acting regions: region A (-279 to -265), region B (-256 to -242) and region C (-235 to -216), downstream
at -279 to -216 residues (Wang et al., 1994). Additionally, there are 3 GC boxes: Box 1 (-48 to -39), Box 2 (-
86 to -77) and Box 3 (-133 to -124), which reside -133 bp residue from the transcription initiation site (Netzker
et al., 1997). Mutation in either box 1 or 3 results in a 50% reduction in the promoter activity (Yamada and
Noguchi, 1999a).

13. PKM2 EXPRESSION REGULATION


The isoform PKM2 expression is achieved by a multiplex of signalling pathways that operate at different
levels. PKM2 expression is induced by the insulin in adipocytes, by activating PI3K and the MAPK signalling
pathways (Mazurek, 2011). Insulin promotes the PKM2 expression independently of the glucose and the
glucosamine (Asai et al., 2003). In contrary to the PKM2 expression upregulation, the insulin decreases its
activity and thereby, encourage cancer metabolism (Iqbal et al., 2013). As the insulin signalling is implicated
in the cancer (Gallagher and LeRoith, 2011), therefore insulin is an important contributes in cancer metabolism
(Iqbal et al., 2013). Glucose also favours the PKM2 expression in rat hepatoma cells as well as isolated
adipocytes (Schafer et al., 1997). Additionally, hormones viz. Triiodothyronine and Glucocorticoids are also
known to regulate the PKM gene expression (Iqbal et al., 2014). PKM2 expression is also investigated to get
increased in the proliferating thymocytes by interleukin-2 stimulation (Netzker et al., 1992).

14. CONCLUSION
1. PKM2 a critical enzyme which control the speed restricting step of glycolysis and plays a major role not
only in the metabolic reprogramming during cancer but also in the direct synchronization of the gene
expression.
2. Improved expression of the PKM2 results in more glucose uptake and thereby, building up of glycolytic
intermediates helping the cancer cell survival.
3. Receiving the impact of proliferative signals, the PKM2 can split glycolytic products in two lineages-
(i) for energy requirements, (ii) for transformation into precursor for biosynthesis.
4. PKM2 can switch between an active tetrameric state and an inactive dimeric state, serving as a critical
regulator of the Warburg effect and metabolic budget in the tumor cells. In this way, PKM2 plays a dual
role ie. as an on/off switch as well as a pace controller in the cancer metabolism.
IJRAR21A1559 International Journal of Research and Analytical Reviews (IJRAR) www.ijrar.org 959
© 2021 IJRAR January 2021, Volume 8, Issue 1 www.ijrar.org (E-ISSN 2348-1269, P- ISSN 2349-5138)

5. PKM2 also serves as a key diagnostic biomarker in the cancer diagnosis. Quantification of the tumor
PKM2 in plasma allows an early tumor detection.
6. PKM2 can be a potential therapeutic target to inhibit the cancer cell proliferation.
7. PKM2 play multiple roles in the cancer and therefore, is a more important in comparison to other
isozymes of pyruvate kinase.

15. FUTURE PROPECTUS


Further studies on cancer metabolism are required to translate Glycolytic Pyruvate Kinase Isoform PKM2
effective therapy.
16. REFERENCES
1. Annibaldi A. and Widmann C. (2010) Glucose metabolism in cancer cells. Curr. Opin. Clin. Nutr.
Metab. Care, 13(4): 466-470.
2. Asai Y., Yamada K., Watanabe T., Keng V.W. and Noguchi T. (2003) Insulin stimulates expression
of the pyruvate kinase M gene in 3T3-L1 adipocytes. Biosci. Biotechnol. Biochem., 67(6): 1272-1277.
3. Chen M., David C.J. and Manley J.L. (2012) Concentration-dependent control of pyruvate kinase M
mutually exclusive splicing by hnRNP proteins. Nat. Struct. Mol. Biol., 19(3): 346-354.
4. Chen M., Zhang J. and Manley J.L. (2010) Turning on a fuel switch of cancer - hnRNP proteins
regulate alternative splicing of pyruvate kinase mRNA. Cancer Res., 70(22): 8977-8980.
5. Christofk H.R., Vander Heiden M.G., Harris M.H., Ramanathan A., Gerszten R.E., Wei R., Fleming
M.D., Schreiber S.L. and Cantley L.C. (2008a) The M2 splice isoform of pyruvate kinase is important
for cancer metabolism and tumour growth. Nature, 452(7184): 230-233.
6. Christofk H.R., Vander Heiden M.G., Wu N., Asara J.M. and Cantley L.C. (2008b) Pyruvate kinase
M2 is a phosphotyrosine-binding protein. Nature, 452 (7184): 181-186.
7. Clower C.V., Chatterjee D., Wang Z., Cantley L.C., Vander Heiden M.G. and Krainer A.R. (2010)
The alternative splicing repressors hnRNP A1/A2 and PTB influence pyruvate kinase isoform
expression and cell metabolism. Proc. Natl. Acad. Sci. USA, 107(5): 1894-1899.
8. Dang C.V. (2009) PKM2 tyrosine phosphorylation and glutamine metabolism signal a different view
of the Warburg effect. Sci. Signal., 2(97): pe75.
9. David C.J., Chen M., Assanah M., Canoll P. and Manley J.L. (2010) HnRNP proteins controlled by c-
Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature, 463(7279): 364-368.
10. Dombrauckas J.D., Santarsiero B.D. and Mesecar A.D. (2005) Structural basis for tumor pyruvate
kinase M2 allosteric regulation and catalysis. Biochem., 44(27): 9417-9429.
11. Gao J.L. and Chen Y.G. (2015) Natural Compounds Regulate Glycolysis in Hypoxic Tumor
Microenvironment. Biomed. Res. Int., 2015: 1-8.
12. Gupta V. and Bamezai R.N.K. (2010) Human pyruvate kinase M2: A multi functional protein. Protein
Sci., 19: 2031-2044.
13. Hamanaka R.B. and Chandel N.S. (2012) Targeting glucose metabolism for cancer therapy. J. Exp.
Med., 209(2): 211-215.
14. Hasan D. (2012) Hypoxic regulation and selective silencing of pyruvate kinase isoforms PKM1 and
PKM2 by siRNA. pp: 1-72.
15. Hitosugi T., Kang S., Vander Heiden M.G., Chung T.W., Elf S., Lythgoe K., Dong S., Lonial S., Wang
X., Chen G.Z., Xie J., Gu T.L., Polakiewicz R.D., Roesel J.L., Boggon T.J., Khuri F.R., Gilliland D.G.,
Cantley L.C., Kaufman J. and Chen J. (2009) Tyrosine phosphorylation inhibits PKM2 to promote the
warburg effect and tumor growth. Sci. Signal., 2(97): 1-8.
16. Hsu P.P. and Sabatini D.M. (2008) Cancer cell metabolism: Warburg and beyond. Cell, 134(5): 703-
707.
17. Iqbal M.A., Gupta V., Gopinath P., Mazurek S. and Bamezai R.N.K. (2014) Pyruvate kinase M2 and
cancer: an updated assessment. FEBS Lett., 588 (16): 2685-2692.

IJRAR21A1559 International Journal of Research and Analytical Reviews (IJRAR) www.ijrar.org 960
© 2021 IJRAR January 2021, Volume 8, Issue 1 www.ijrar.org (E-ISSN 2348-1269, P- ISSN 2349-5138)

18. Iqbal M.A., Siddiqui F.A., Gupta V., Chattopadhyay S., Gopinath P., Kumar B., Manvati S., Chaman
N. and Bamezai R.N. (2013) Insulin enhances metabolic capacities of cancer cells by dual regulation
of glycolytic enzyme pyruvate kinase M2. Mol. Cancer, 12(72): 1-12.
19. Jiang J.K., Boxer M.B., Vander Heiden M.G., Shen M., Skoumbourdis A.P., Southall N., Veith H.,
Leister W., Austin C., Park H., Inglese J., Cantley L., Auld D. and Thomas C. (2010) Evaluation of
thieno[3,2-b]pyrrole[3,2-d]pyridazinones as activators of the tumor cell specific M2 isoform of
pyruvate kinase. Bioorg. Med. Chem. Lett., 20(11): 3387-3393.
20. Jurica M.S., Mesecar A., Heath P.J., Shi W., Nowak T. and Stoddard B.L. (1998) The allosteric
regulation of pyruvate kinase by fructose-1,6-bisphosphate. Struct., 6: 195-210.
21. Kondoh H., Lleonart M.E., Bernard D. and Gil J. (2007). Protection from oxidative stress by enhanced
glycolysis: a possible mechanism of cellular immortalization. Histol. Histopathol., 22(1): 85-90.
22. Koppenol W.H., Bounds P.L. and Dang C.V. (2011) Otto Warburg’s contributions to current concepts
of cancer metabolism. Nat. Rev. Cancer, 11: 325-337.
23. Li X.B., Gu J.D. and Zhou Q.H. (2015) Review of aerobic glycolysis and its key enzymes - new targets
for lung cancer therapy. Thorac. Cancer, 6: 17-24.
24. Lu Z. (2012a) Nonmetabolic functions of pyruvate kinase isoform M2 in controlling cell cycle
progression and tumorigenesis. Chin. J. Cancer., 31(1): 5-7.
25. Lyssiotis C.A., Anastasiou D., Locasale J.W., Vander Heiden M.G., Christofk H.R. and Cantley L.C.
(2012) Cellular Control Mechanisms that Regulate Pyruvate Kinase M2 Activity and Promote Cancer
Growth. Biomed. Res., 23: SI 213-217.
26. Mazurek S., Drexler H.C., Troppmair J., Eigenbrodt E. and Rapp U.R. (2007) Regulation of pyruvate
kinase type M2 by A-Raf: a possible glycolytic stop or go mechanism. Anticancer Res., 27: 3963-
3971.
27. Mazurek S. (2011) Pyruvate kinase type M2: A key regulator of the metabolic budget system in tumor
cells. Int. J. Biochem. Cell Biol., 43(7): 969-980.
28. Mazurek S., Boschek C.B., Hugo F. and Eigenbrodt E. (2005) Pyruvate kinase type M2 and its role in
tumor growth and spreading. Semin. Cancer. Biol., 15: 300-308.
29. Mazurek S., Grimm H., Boschek C.B., Vaupel P. and Eigenbrodt E. (2002) Pyruvate kinase type M2:
a crossroad in the tumor metabolome. Brit. J. Nutr., 87(1): S23-S29.
30. Mazurek S., Zwerschke W., Jansen-Durr P. and Eigenbrodt E. (2001) Effects of the human papilloma
virus HPV-16E7 oncoprotein on glycolysis and glutamino- lysis: Role of pyruvate kinase type M2 and
the glycolytic enzyme complex. Biochem. J., 356: 247-256.
31. Morgan H.P., Zhong W., Iainw M., Michels P.A.M., Fothergill-Gilmore L.A. and Walkinshaw M.D.
(2014) Structures of pyruvate kinases display evolutionarily divergent allosteric strategies. R. Soc.
Open Sci., 1(140120): 1-14.
32. Netzker R., Greiner E., Eigenbrodt E., Noguchi T., Tanaka T. and Brand K. (1992) Cell cycle-
associated expression of M2-type isozyme of pyruvate kinase in proliferating rat thymocytes. J. Biol.
Chem., 267(9): 6421-6424.
33. Netzker R., Weigert C. and Brand K. (1997) Role of the stimulatory proteins Sp1 and Sp3 in the
regulation of transcription of the rat pyruvate kinase M gene. Eur. J. Biochem., 245(1): 174-181.
34. Noguchi T., Inoue H. and Tanaka T. (1986) The M1- and M2- type isozymes of rat pyruvate kinase
are produced from the same gene by alternative RNA splicing. J. Biol. Chem., 261: 13807-13812.
35. Oremek G.M., Teigelkamp S., Kramer W., Eigenbrodt E. and Usadel K.H. (1999) The pyruvate kinase
isoenzyme tumor M2 (Tu M2-PK) as a tumor marker for renal carcinoma. Anticancer Res., 19(4A):
2599-2601.
36. Schafer D., Hamm-Kunzelmann B. and Brand K. (1997) Glucose regulates the promoter activity of
aldolase A and pyruvate kinase M2 via dephosphorylation of Sp1. FEBS Lett., 417: 325-328.
37. Takenaka M., Noguchi T., Sadahiro S., Hirai H., Yamada K., Matsuda T., Imai E. and Tanaka T.
(1991) Isolation and characterization of the human pyruvate kinase M gene. Eur. J. Biochem., 198:
101-106.
IJRAR21A1559 International Journal of Research and Analytical Reviews (IJRAR) www.ijrar.org 961
© 2021 IJRAR January 2021, Volume 8, Issue 1 www.ijrar.org (E-ISSN 2348-1269, P- ISSN 2349-5138)

38. Tamada M., Suematsu M. and Saya H. (2012) Pyruvate Kinase M2: Multiple Faces for Conferring
Benefits on Cancer Cells. Clin. Cancer Res., 18(20): 5554-5561.
39. Toschi A., Lee E., Thompson S., Gadir N., Yellen P., Drain C.M., Ohh M. and Foster D.A. (2010)
Phospholipase D-mTOR requirement for the Warburg effect in human cancer cells. Cancer Lett.,
299(1): 72-79.
40. Valentini G., Chiarelli L., Fortin R., Speranza M.L., Galizzi A. and Mattevi A. (2000) The allosteric
regulation of pyruvate kinase - A site-directed mutagenesis study. J. Biol. Chem., 275(24): 18145-
18152.
41. Valentini G., Chiarelli L.R., Fortin R., Dolzan M., Galizzi A., Abraham D.J., Wang C., Bianchi P.,
Zanella A. and Mattevi A. (2002) Structure and function of human erythrocyte pyruvate kinase. J. Biol.
Chem., 277(26): 23807-23814.
42. Vander Heiden M.G., Cantley L.C. and Thompson C.B. (2009) Understanding the Warburg effect: the
metabolic requirements of cell proliferation. Sci., 324(5930): 1029-1033.
43. Wang C., Chiarelli L.R., Bianchi P., Abraham D.J., Galizzi A., Mattevi A., Zanella A. and Valentini
G. (2001) Human erythrocyte pyruvate kinase: characterization of the recombinant enzyme and a
mutant form (R510Q) causing nonspherocytic hemolytic anemia. Blood, 98: 3113-3120.
44. Ward P.S. and Thompson C.B. (2012) Metabolic reprogramming: a cancer hallmark even warburg did
not anticipate. Cancer Cell, 21: 297-308.
45. Wooll J.O., Friesen R.H.E., White M.A., Watowich S.J., Fox R.O., Lee J.C. and Czerwinski E.W.
(2001) Structural and functional linkages between subunit interfaces in mammalian pyruvate kinase.
J. Mol. Biol., 312: 525-540.
46. Wu S. and Le H. (2013) Dual roles of PKM2 in cancer metabolism. Acta Biochim. Biophys. Sin., 45:
27-35.
47. Wu X., Zhou Y., Zhang K., Liu Q. and Guo D. (2008) Isoform-specific interaction of pyruvate kinase
with hepatitis C virus NS5B. FEBS Lett., 582: 2155-2160.
48. Yamada K. and Noguchi T. (1999a) Nutrient and hormonal regulation of pyruvate kinase gene
expression. Biochem. J., 337(1): 1-11.
49. Yamada K. and Noguchi T. (1999b) Regulation of pyruvate kinase M gene expression. Biochem.
Biophys. Res. Commun., 256(2): 257-262.
50. Yang W. and Lu Z. (2013) Regulation and function of pyruvate kinase M2 in cancer. Cancer lett.,
339(2): 153-158.
51. Zhou W., Liotta L.A. and Petricoin E.F. (2012) Cancer Metabolism: What we Can Learn from
Proteomic Analysis by Mass Spectrometry. Cancer Genomics Proteomics, 9: 373-382.

IJRAR21A1559 International Journal of Research and Analytical Reviews (IJRAR) www.ijrar.org 962

You might also like