Review
New aspects of the Warburg effect in cancer cell biology

https://doi.org/10.1016/j.semcdb.2012.02.003Get rights and content

Abstract

Altered cellular metabolism is a defining feature of cancer [1]. The best studied metabolic phenotype of cancer is aerobic glycloysis – also known as the Warburg effect – characterized by increased metabolism of glucose to lactate in the presence of sufficient oxygen. Interest in the Warburg effect has escalated in recent years due to the proven utility of FDG-PET for imaging tumors in cancer patients and growing evidence that mutations in oncogenes and tumor suppressor genes directly impact metabolism. The goals of this review are to provide an organized snapshot of the current understanding of regulatory mechanisms important for Warburg effect and its role in tumor biology. Since several reviews have covered aspects of this topic in recent years, we focus on newest contributions to the field and reference other reviews where appropriate.

Highlights

► This review discusses regulatory mechanisms that contribute to the Warburg effect in cancer. ► We list cancers for which FDG-PET has established applications as well as those cancers for which FDG-PET has not been established. ► PKM2 is highlighted as an important integrator of diverse cellular stimuli to modulate metabolic flux and cancer cell proliferation. ► We discuss how cancer metabolism can directly influence gene expression programs. ► Contribution of aerobic glycolysis to the cancer microenvironment and chemotherapeutic resistance/susceptibility is also discussed.

Introduction

In the 1920s, Otto Warburg made his seminal observation that tumor cells metabolize more glucose to lactate than normal cells. By directly measuring lactate production and oxygen consumption rates in thin slices of rat liver carcinoma and normal liver tissue, Warburg and colleagues found that normal liver tissue exhibited the Pasteur effect (inhibition of lactate production in the presence of oxygen), whereas tumor tissue maintained lactate production regardless of oxygen tension [2], [3]. Sustained lactic acid production in the presence of oxygen was also observed in several human carcinomas. Warburg determined that cancer tissue consumes ten-fold more glucose than can be accounted for by respiration, and the amount of lactic acid produced is two orders of magnitude greater than the amount produced by normal tissue [2]. For a detailed description of Dr. Warburg's life and research, we direct the reader to an eloquent recent review by Dang and colleagues [4].

FDG-PET (2-deoxy-2-[18F]fluoro-d-glucose positron emission tomography) is a molecular imaging technique that exploits cancer cells’ preferential utilization of aerobic glycolysis. Similar to glucose, FDG enters cancer cells via glucose transporters GLUT1 and GLUT3 and is subsequently phosphorylated by hexokinase to FDG-6-phosphate. While glucose-6-phosphate undergoes further isomerization to fructose-6-phospate in the glycolytic pathway or oxidation to 6-phosphogluconolactone in the pentose phosphate pathway, FDG-6-phosphate cannot be further catabolized due to lack of an oxygen atom at the C-2 position. FDG-6-phosphate is unable to diffuse out of cells, and the rate of dephosphorylation occurs slowly; therefore, it becomes trapped and accumulates at a rate proportional to glucose utilization. Thus, FDG uptake depends on both glucose transporter expression and hexokinase activity and provides a way to assess glucose uptake rates in cells [5]. Tumors that take up more glucose than surrounding tissues can be non-invasively visualized in cancer patients by FDG-PET.

FDG-PET has been used in cancer patients since the 1980s and is now a widely used clinical imaging tool in oncology. It is approved for disease diagnosis, staging, restaging, and therapy monitoring in many but not all cancers [5]. Table 1 shows a current list of select cancers for which FDG-PET has established applications [6]. Despite its widespread use in clinical oncology, some cancers remain difficult to image by FDG-PET, such as hepatocellular carcinoma, prostate cancer, and pancreatic cancer. It is possible that these tumor types do not exhibit the Warburg effect metabolic phenotype and instead rely on alternative carbon sources than glucose to fuel proliferation. Alternatively, these tumor types may be difficult to image by FDG-PET for imaging-related reasons such as poor perfusion of the tumor by the probe, low density of tumor cells in the tumor tissue (high stromal cell to tumor cell ratio) or high background signal (such as bladder signal in the case of prostate cancer). Some cancers, such as well-differentiated hepatocellular carcinoma, exhibit high expression levels of glucose-6-phosphatase, which by dephosphorylating FDG-6-phosphate, allows efflux of FDG from cancer cells [7]. Further work is needed to address these issues in FDG-PET negative cancers. However, the large number of tumor types for which FDG-PET has proven utility has helped spur interest in the Warburg effect and its regulation and role in tumor biology.

Section snippets

Regulation of the Warburg effect

Accumulating evidence indicates that every major oncogene and tumor suppressor can affect metabolic regulation. However, the molecular mechanisms by which the cancer metabolic phenotype is accomplished by oncogenes and tumor suppressors in different tumor types varies. Below we outline some of the better characterized mechanisms by which the Warburg effect is established in cancer cells. This topic was also recently reviewed by Mak and colleagues [8].

Can the Warburg effect and cancer metabolism be programmed?

While the Warburg effect metabolic phenotype was initially identified in cancer tissue, it is now well appreciated that rapidly dividing normal tissues, such as ES cells and lymphocytes, employ aerobic glycolysis to meet their energetic and biosynthetic requirements during expansion (reviewed in [91]). These observations support the notion that aerobic glycolysis is a preferred metabolic program under conditions of rapid cellular expansion. However, it remains unclear how the Warburg effect is

Conclusion

In conclusion, the cancer metabolic program is an important component of tumor growth and survival. Renewed interest in this program has yielded significant progress over the last decade toward identifying genetic and biochemical events underlying “how” and “why” a cancer cell engages aerobic glycolysis. Importantly, these observations have reinforced our understanding of the molecular heterogeneity by which the Warburg effect and cancer metabolic phenotype is achieved. This heterogeneity also

References (117)

  • O. Minchenko et al.

    Hypoxic regulation of the 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase gene family (PFKFB-1–4) expression in vivo

    FEBS Lett

    (2003)
  • R.B. Robey et al.

    Is Akt the Warburg kinase? – Akt-energy metabolism interactions and oncogenesis

    Semin Cancer Biol

    (2009)
  • D.P. Bartel

    MicroRNAs: genomics, biogenesis, mechanism, and function

    Cell

    (2004)
  • A. Ventura et al.

    MicroRNAs and cancer: short RNAs go a long way

    Cell

    (2009)
  • V.R. Fantin et al.

    Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance

    Cancer Cell

    (2006)
  • S. Bonnet et al.

    A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth

    Cancer Cell

    (2007)
  • S. Weinhouse

    Oxidative metabolism of neoplastic tissues

    Adv Cancer Res

    (1955)
  • C. Frezza et al.

    Mitochondria in cancer: not just innocent bystanders

    Semin Cancer Biol

    (2009)
  • A. Jacobsson et al.

    Mitochondrial uncoupling protein from mouse brown fat. Molecular cloning, genetic mapping, and mRNA expression

    J Biol Chem

    (1985)
  • G. Baffy

    Uncoupling protein-2 and cancer

    Mitochondrion

    (2010)
  • P.S. Ward et al.

    The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate

    Cancer Cell

    (2010)
  • M.E. Figueroa et al.

    Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation

    Cancer Cell

    (2010)
  • W. Xu et al.

    Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases

    Cancer Cell

    (2011)
  • G. Hatzivassiliou et al.

    ATP citrate lyase inhibition can suppress tumor cell growth

    Cancer Cell

    (2005)
  • L. Cai et al.

    Acetyl-CoA induces cell growth and proliferation by promoting the acetylation of histones at growth genes

    Mol Cell

    (2011)
  • D. Hanahan et al.

    Hallmarks of cancer: the next generation

    Cell

    (2011)
  • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell...
  • O. Warburg et al.

    Über den stoffwechsel der carcinomzelle

    Biochem Z

    (1924)
  • S. Minami

    Versuche an überlebendem carcinomgewebe

    Biochem Z

    (1923)
  • Koppenol WH, Bounds PL, Dang CV. Otto Warburg's contributions to current concepts of cancer metabolism. Nat Rev Cancer...
  • G.J. Kelloff et al.

    Progress and promise of FDG-PET imaging for cancer patient management and oncologic drug development

    Clin Cancer Res

    (2005)
  • Personal communication from Dr. Johannes Czernin,...
  • B.M. Gallagher et al.

    Metabolic trapping as a principle of oradiopharmaceutical design: some factors resposible for the biodistribution of [18F] 2-deoxy-2-fluoro-d-glucose

    J Nucl Med

    (1978)
  • Cairns RA, Harris IS, Mak TW. Regulation of cancer cell metabolism. Nat Rev Cancer...
  • P.K. Majumder et al.

    mTOR inhibition reverses Akt-dependent prostate intraepithelial neoplasia through regulation of apoptotic and HIF-1-dependent pathways

    Nat Med

    (2004)
  • D.B. Shackelford et al.

    mTOR and HIF-1alpha-mediated tumor metabolism in an LKB1 mouse model of Peutz-Jeghers syndrome

    Proc Natl Acad Sci USA

    (2009)
  • Palaskas N, Larson SM, Schultz N, Komisopoulou E, Wong J, Rohle D et al. 18F-fluorodeoxy-glucose positron emission...
  • Semenza GL. HIF-1: upstream and downstream of cancer metabolism. Curr Opin Genet Dev...
  • J.A. Bertout et al.

    The impact of O2 availability on human cancer

    Nat Rev Cancer

    (2008)
  • H. Shim et al.

    c-Myc transactivation of LDH-A: implications for tumor metabolism and growth

    Proc Natl Acad Sci USA

    (1997)
  • C.V. Dang et al.

    The interplay between MYC and HIF in cancer

    Nat Rev Cancer

    (2008)
  • P. Gao et al.

    c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism

    Nature

    (2009)
  • Dang CV. Rethinking the Warburg effect with Myc micromanaging glutamine metabolism. Cancer Res...
  • S. Matoba et al.

    p53 regulates mitochondrial respiration

    Science

    (2006)
  • K.H. Vousden et al.

    p53 and metabolism

    Nat Rev Cancer

    (2009)
  • R.D. Michalek et al.

    Estrogen-related receptor-alpha is a metabolic regulator of effector T-cell activation and differentiation

    Proc Natl Acad Sci USA

    (2011)
  • H.R. Christofk et al.

    The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth

    Nature

    (2008)
  • Luo W, Hu H, Chang R, Zhong J, Knabel M, O’Meally R, et al. Pyruvate kinase M2 is a PHD3-stimulated coactivator for...
  • J.D. Dombrauckas et al.

    Structural basis for tumor pyruvate kinase M2 allosteric regulation and catalysis

    Biochemistry

    (2005)
  • H.R. Christofk et al.

    Pyruvate kinase M2 is a phosphotyrosine-binding protein

    Nature

    (2008)
  • Cited by (0)

    View full text