Skip to main content
Erschienen in: Cancer Cell International 1/2018

Open Access 01.12.2018 | Commentary

Citrate targets FBPase and constitutes an emerging novel approach for cancer therapy

verfasst von: Philippe Icard, Ludovic Fournel, Antoine Coquerel, Joseph Gligorov, Marco Alifano, Hubert Lincet

Erschienen in: Cancer Cell International | Ausgabe 1/2018

Abstract

Gao-Min Liu and Yao-Ming Zhang recently published a review entitled «Targeting FBPase is an emerging novel approach for cancer therapy» (Liu and Zhang in Cancer Cell Int 18:36, 2018). In this paper, the authors highlighted how the down regulation or inactivation of FBPase, a rate limiting enzyme of gluconeogenesis, can promote the Warburg effect and cancer growth. In contrast, activation of this enzyme demonstrates anti-cancer effects and may appear as emerging novel approach for cancer therapy. Among the potential activators of FBP listed by Liu and Zhang, citrate was surprisingly not mentioned although it is an activator of FBPase, also demonstrating various anti-cancer effects in pre-clinical studies. Thus, citrate should be tested as a new therapeutic strategy, in particular in clinical studies.
Drs. Gao-Min Liu and Yao-Ming Zhang must be congratulated for their review «Targeting FBPase is an emerging novel approach for cancer therapy» explaining how the down expression or inactivation of this rate limiting enzyme of gluconeogenesis promotes aerobic glycolysis, cancer cell proliferation and dedifferentiation. Therefore, activation of FBPase may have anti-cancer effects, counteracts the Warburg effect and antagonizes chemoresistance [1]. Among the potential activators of FBP listed by Liu and Zhang, citrate was surprisingly not mentioned, although this molecule plays a central role in the metabolism of cancer cells [2]. Physiologically, when energy stores are elevated, citrate is abundant and the activity of (phosphofructokinase 1) PFK1 is nearly switched off [2]. Citrate is also a potent allosteric inhibitor of PFK2 (also called PFKFB (6-phosphofructo-2-kinase/fructose-2,6-biphosphatase) in non-cancer cells (such as cardiac cells [3]) and ascites cancer cells [4]. It is likely that citrate also indirectly inhibits PFK2 through an increased palmitate synthesis [5].
By regulating key regulatory enzymes located at entrance and/or exit of glycolysis (PFK1, PFK2, PK), TCA cycle (PDH, SDH), gluconeogenesis (F1,6BPase) and fatty acid synthesis (ACC), citrate controls anabolic and catabolic pathways, and allows a close adjustment of metabolic flows to ATP production [2].
In cancer cells, the Warburg effect is associated with a decrease production of citrate by mitochondria [6]. This reprogramming metabolism is promoted by over-expression of HIF-1alpha and Myc, activation of Ras, loss of functional p53, all factors promoting the down-regulation of mitochondria [6]. Such down-regulation leads to a reduced production of citrate, ATP and CO2, such condition promoting intracellular alkaline pH, a strong activator of PFK-1 [6]. This rate-limiting enzyme of glycolysis is also controlled by a family of regulatory bifunctional enzymes PFKFB. When the kinase activity is promoted, fructose-2,6-bisphosphate (F2,6BP) increases and allosterically activates PFK-1, directing the carbon flux into glycolysis and sustaining cell survival and proliferation [7].
We showed for the first time that administration of citrate to cancer cells arrests cell growth, induces apoptosis (in particular through extinction of expression of the anti-apoptotic factor Mcl-1) and sensitizes chemoresistant cells to cisplatin [8]. Several authors have confirmed on different cancer cell lines that citrate administration (generally at ≥ 10 mM) inhibits glycolysis and ATP production, promotes apoptosis through activation of caspases, and increases response to Bcl-xL inhibitors (for additional references see [6, 9]). Recently, Ren et al. [9] confirmed that citrate decreases resistance to cisplatin (in particular by reducing the expression of Snail and MUC-1), and showed that it inhibits the proliferative IGF-1R/AKT axis, stimulates the suppressive PTEN-eIF2α pathway, and induces tumor cell differentiation (expression of E-cadherin). Importantly, these authors observed that daily oral administration of citrate at a dose of 8 g/kg/day for 1 month reduces tumor growth of several xenograft tumor models in mice (pancreatic cancer, Ras-driven lung tumor and Her2/Neu mammary cancer), increasing significantly the number of infiltrating T-cells in tumors [9].
All these experimental in vitro and in vivo studies give arguments to consider that high-dose citrate administration inhibits cancer cell development. Nonetheless, the mechanisms underlying this process remain unclear. Beside its allosteric inactivation of PFK 1 and 2, reducing or arresting the glycolytic flux, citrate may also exercise other anti-cancer effects by reducing the levels of fructose-1,6-bisphosphate (F1,6BP) and F2,6BP. Indeed, these molecules couple the glycolytic flux with activation of Ras and cell cycle progression, respectively: (i) cytosolic F1,6BP binds Son of sevenless homolog 1 (Sos1), a factor promoting the activation of Ras and its downstream targets MEK and ERK [10]; (ii) F2,6BP represses p27Kip1, a potent inhibitor of cyclins D and E regulating G1/S transition, and cyclin-dependent kinase (Cdk)-1 regulating entrance in mitosis [7].
Knowing that both isoforms of FBP (FBP1 and FBP2) are inhibited by Ca2+, very probably high concentration of citrate also inactivates these enzymes by this mechanism because citrate is a well-known chelator of calcium. This action should be particularly interesting for treatment of cancers with poor prognosis (such as gastric cancer, brain metastatic breast cancers) expressing low FBP2, because as explained by Liu and Zhang [1], FBP2 is 1000 times more sensitive to inhibition by Ca2+.
More studies are needed to nail down the mechanisms underlying citrate effect in cancer cells proliferation and strengthen the proof of principle.

Authors’ contributions

PI: conception, writing of the commentary and revision. LF: conception, editing and submission. AC: conception and commentary design, reviewing. JG: commentary design and correction. MA: conception and reviewing. HL: design, final supervision. All authors read and approved the final manuscript.

Acknowledgements

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

Not applicable.
Not applicable.
Not applicable.

Funding

No funding was provided for this commentary.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Liu GM, Zhang YM. Targeting FBPase is an emerging novel approach for cancer therapy. Cancer Cell Int. 2018;18:36.CrossRef Liu GM, Zhang YM. Targeting FBPase is an emerging novel approach for cancer therapy. Cancer Cell Int. 2018;18:36.CrossRef
2.
Zurück zum Zitat Icard P, Poulain L, Lincet H. Understanding the central role of citrate in the metabolism of cancer cells. Biochim Biophys Acta. 2012;1825:111–6.PubMed Icard P, Poulain L, Lincet H. Understanding the central role of citrate in the metabolism of cancer cells. Biochim Biophys Acta. 2012;1825:111–6.PubMed
3.
Zurück zum Zitat Crochet RB, Kim JD, Lee H, Yim YS, Kim SG, Neau D, Lee YH. Crystal structure of heart 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB2) and the inhibitory influence of citrate on substrate binding. Proteins. 2017;85:117–24.CrossRef Crochet RB, Kim JD, Lee H, Yim YS, Kim SG, Neau D, Lee YH. Crystal structure of heart 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB2) and the inhibitory influence of citrate on substrate binding. Proteins. 2017;85:117–24.CrossRef
4.
Zurück zum Zitat Nissler K, Petermann H, Wenz I, Brox D. Fructose 2,6-bisphosphate metabolism in Ehrlich ascites tumor cells. J Cancer Res Clin Oncol. 1995;121:739–45.CrossRef Nissler K, Petermann H, Wenz I, Brox D. Fructose 2,6-bisphosphate metabolism in Ehrlich ascites tumor cells. J Cancer Res Clin Oncol. 1995;121:739–45.CrossRef
5.
Zurück zum Zitat Hue L, Maisin L, Rider MH. Palmitate inhibits liver glycolysis. Involvement of fructose 2,6-bisphosphate in the glucose/fatty acid cycle. Biochem J. 1988;251:541–5.CrossRef Hue L, Maisin L, Rider MH. Palmitate inhibits liver glycolysis. Involvement of fructose 2,6-bisphosphate in the glucose/fatty acid cycle. Biochem J. 1988;251:541–5.CrossRef
6.
Zurück zum Zitat Icard P, Lincet H. The reduced concentration of citrate in cancer cells: an indicator of cancer aggressiveness and a possible therapeutic target. Drug Resist Updates. 2016;29:47–53.CrossRef Icard P, Lincet H. The reduced concentration of citrate in cancer cells: an indicator of cancer aggressiveness and a possible therapeutic target. Drug Resist Updates. 2016;29:47–53.CrossRef
7.
Zurück zum Zitat Yalcin A, Clem BF, Simmons A, Lane A, Nelson K, Clem AL, et al. Nuclear targeting of 6-phosphofructo-2-kinase (PFKFB3) increases proliferation via cyclin-dependent kinases. J Biol Chem. 2009;284:24223–32.CrossRef Yalcin A, Clem BF, Simmons A, Lane A, Nelson K, Clem AL, et al. Nuclear targeting of 6-phosphofructo-2-kinase (PFKFB3) increases proliferation via cyclin-dependent kinases. J Biol Chem. 2009;284:24223–32.CrossRef
8.
Zurück zum Zitat Zhang X, Varin E, Allouche S, Lu Y, Poulain L, Icard P. Effect of citrate on malignant pleural mesothelioma cells: a synergistic effect with cisplatin. Anticancer Res. 2009;29:1249–54.PubMed Zhang X, Varin E, Allouche S, Lu Y, Poulain L, Icard P. Effect of citrate on malignant pleural mesothelioma cells: a synergistic effect with cisplatin. Anticancer Res. 2009;29:1249–54.PubMed
9.
Zurück zum Zitat Ren JG, Seth P, Ye H, Guo K, Hanai JL, Husain Z, Sukhatme VP. Citrate suppresses tumor growth in multiple models through inhibition of glycolysis, the tricarboxylic acid cycle and the IGF-1R pathway. Sci Rep. 2017;7:4537.CrossRef Ren JG, Seth P, Ye H, Guo K, Hanai JL, Husain Z, Sukhatme VP. Citrate suppresses tumor growth in multiple models through inhibition of glycolysis, the tricarboxylic acid cycle and the IGF-1R pathway. Sci Rep. 2017;7:4537.CrossRef
10.
Zurück zum Zitat Peeters K, Van Leemputte F, Fischer B, Bonini BM, Quezada H, Tsytlonok M, et al. Fructose-1,6-bisphosphate couples glycolytic flux to activation of Ras. Nat Commun. 2017;8:922.CrossRef Peeters K, Van Leemputte F, Fischer B, Bonini BM, Quezada H, Tsytlonok M, et al. Fructose-1,6-bisphosphate couples glycolytic flux to activation of Ras. Nat Commun. 2017;8:922.CrossRef
Metadaten
Titel
Citrate targets FBPase and constitutes an emerging novel approach for cancer therapy
verfasst von
Philippe Icard
Ludovic Fournel
Antoine Coquerel
Joseph Gligorov
Marco Alifano
Hubert Lincet
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2018
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-018-0676-y

Weitere Artikel der Ausgabe 1/2018

Cancer Cell International 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.