Elsevier

Seminars in Cancer Biology

Volume 30, February 2015, Pages 21-29
Seminars in Cancer Biology

Review
Metabolic Cancer Biology: Structural-based analysis of cancer as a metabolic disease, new sights and opportunities for disease treatment

https://doi.org/10.1016/j.semcancer.2014.01.007Get rights and content

Abstract

The cancer cell metabolism or the Warburg effect discovery goes back to 1924 when, for the first time Otto Warburg observed, in contrast to the normal cells, cancer cells have different metabolism. With the initiation of high throughput technologies and computational systems biology, cancer cell metabolism renaissances and many attempts were performed to revise the Warburg effect. The development of experimental and analytical tools which generate high-throughput biological data including lots of information could lead to application of computational models in biological discovery and clinical medicine especially for cancer. Due to the recent availability of tissue-specific reconstructed models, new opportunities in studying metabolic alteration in various kinds of cancers open up. Structural approaches at genome-scale levels seem to be suitable for developing diagnostic and prognostic molecular signatures, as well as in identifying new drug targets. In this review, we have considered these recent advances in structural-based analysis of cancer as a metabolic disease view. Two different structural approaches have been described here: topological and constraint-based methods. The ultimate goal of this type of systems analysis is not only the discovery of novel drug targets but also the development of new systems-based therapy strategies.

Introduction

Cancer is a complex disease which contains multiple types of biological interactions through various physical, sequential, and biological scales. This complexity generates considerable challenges for the description of cancer biology, and inspires the study of cancer in the context of molecular, cellular, and physiological systems. A significant factor contributing to this new synthesis is the observation that several signaling pathways changed in cancer are key regulators of the human metabolic network. This specifies a rational interplay between genetic and metabolic alterations during tumorigenesis without a permanent cause–effect relationship [1]. Otto Warburg first suggested this metabolic modification based on his observations in leukemic cells that altered metabolism of glucose may lead to cancer. This effect is now referred as the “Warburg effect”. Since then, different hypotheses (Fig. 1) have been proposed to find the mechanisms responsible for the Warburg effect [2]. However, the metabolic landscape of cancer is still far from understood, and in particular its regulation. Recently, there has been a resurgence of interest in cancer metabolism [1], [3], [4]. In the last decade, there is a paradigm shift from studying individual enzymes to newer approaches that aims to comprehend altered tumor metabolism as a whole. These new efforts flourish due to increasing availability of high-throughput data from various tumor studies elucidating metabolic concentrations, fluxes and abundance and regulation of the key enzymes. The data can now be analyzed integratively using statistical models to describe cancer metabolism. Beside experimental work, a metabolic network reconstruction is a manually curated, computational framework that empowers the description of gene–protein-reaction relationships [5]. For understanding the metabolic fluxes of a cancer cell, mechanistic genome scale models of cancer metabolism are needed and first attempts are very promising. Mechanistic methods are becoming increasingly feasible not only because of more sophisticated approaches and better data, but also due to hardware improvements enabling to simulate these models on clusters with a couple of hundreds of cores. Several studies have established how such reconstructions of metabolism could guide the development of biological theories and discoveries [6], [7], [8].

In this article we have described recent advances in network-based analysis of cancer as a metabolic disease. In the first section, the topological approach has been explained. In the next section, the constraint-based method (as another network-based approach) has been considered.

Section snippets

Genome-scale metabolic model of human cancer

With the advent of genome-scale metabolic models (GEMs) of various cell types and diseases, a valuable tool to study genetic, epigenetic and metabolic events in combination, has emerged [9]. The convergence of these developments enables the researchers to predict physiological functions and the relevant growth rate of particular human cell types, tissue-specificity and cancer [10], [11], [12]. There are four generic reconstructed genome-scale human metabolic networks: Recon1 [13], Recon2 [14],

Topological approach

The topological analysis of biological networks is an important method in systems biology which allows the investigation of large scale networks such as GEMs [48]. Graph theory is the most useful framework for representation of GEMs. A mathematical representation of a network is a graph G(V,E). Its vertex set (V) consists of all nodes. Two nodes are adjacent if an edge exist between which connects them.

There are many parameters which could be calculated by graph theory method such as clustering

Flux balance analysis

The potential of mathematical computational modeling tools in description, exploration, and prediction of metabolic networks has been realized in recent years [73]. One of the most widely used network analysis approaches is Flux Balance Analysis (FBA), which is based on derivation of the steady-state metabolic capabilities of a system with appropriate constraints and without the need for accurate kinetic data [74]. The main principle of the FBA is that the system will reach a steady-state under

Drug-target prediction and structural analysis

GEMs provide a useful tool for the study of diseases and the development of drugs. Several simulations and modeling methods have been developed to address the issues of drug-target prediction [92], [93], [94], [95], [96]. The structural features of metabolic networks contribute to the robustness and flexibility of the complex biosystems and may explain, in general, the fact that many drug candidates are ineffective (the drug effect is compensated by other pathways in the network) or show

Outline

Aggressive cancers established evidence of a metabolic shift, including upregulation of the pentose phosphate pathway (PPP) and the glutamine transporter genes, downregulation of genes contributed in the TCA cycle, increased acetyl-CoA carboxylase protein, decreased AMPK and PTEN protein levels, and altered promoter methylation of miR-21 and GRB10 [106]. As far as we know, different strategies are available to target metabolic enzymes for cancer therapy including nucleic acid synthesis, amino

Conflict of interest

No conflict of interest exists.

Funding source

No funding.

References (114)

  • R.K. Nibbe et al.

    Discovery and scoring of protein interaction subnetworks discriminative of late stage human colon cancer

    Mol Cell Proteomics

    (2009)
  • K.J. Kauffman et al.

    Advances in flux balance analysis

    Curr Opin Biotechnol

    (2003)
  • M. Csete et al.

    Bow ties, metabolism and disease

    Trends Biotechnol

    (2004)
  • G. Zimmermann et al.

    Multi-target therapeutics: when the whole is greater than the sum of the parts

    Drug Discov Today

    (2007)
  • M.G. Vander Heiden et al.

    Understanding the Warburg effect: the metabolic requirements of cell proliferation

    Science

    (2009)
  • M.A. Oberhardt et al.

    Metabolic network analysis of Pseudomonas aeruginosa during chronic cystic fibrosis lung infection

    J Bacteriol

    (2010)
  • M.I. Sigurdsson et al.

    A detailed genome-wide reconstruction of mouse metabolism based on human Recon 1

    BMC Syst Biol

    (2010)
  • R.L. Chang et al.

    Metabolic network reconstruction of Chlamydomonas offers insight into light-driven algal metabolism

    Mol Syst Biol

    (2011)
  • A.M. Feist et al.

    The growing scope of applications of genome-scale metabolic reconstructions using Escherichia coli

    Nat Biotechnol

    (2008)
  • I. Thiele et al.

    A protocol for generating a high-quality genome-scale metabolic reconstruction

    Nat Protoc

    (2010)
  • N.C. Duarte et al.

    Global reconstruction of the human metabolic network based on genomic and bibliomic data

    Proc Natl Acad Sci U S A

    (2007)
  • I. Thiele et al.

    A community-driven global reconstruction of human metabolism

    Nat Biotechnol

    (2013)
  • T. Hao et al.

    Compartmentalization of the Edinburgh Human Metabolic Network

    BMC Bioinform

    (2010)
  • P. Romero et al.

    Computational prediction of human metabolic pathways from the complete human genome

    Genome Biol

    (2005)
  • C. Gille et al.

    HepatoNet1: a comprehensive metabolic reconstruction of the human hepatocyte for the analysis of liver physiology

    Mol Syst Biol

    (2010)
  • R.L. Chang et al.

    Drug off-target effects predicted using structural analysis in the context of a metabolic network model

    PLoS Comput Biol

    (2010)
  • N.E. Lewis et al.

    Large-scale in silico modeling of metabolic interactions between cell types in the human brain

    Nat Biotechnol

    (2010)
  • A. Bordbar et al.

    iAB-RBC-283: a proteomically derived knowledge-base of erythrocyte metabolism that can be used to simulate its physiological and patho-physiological states

    BMC Syst Biol

    (2011)
  • A. Bordbar et al.

    Insight into human alveolar macrophage and M. tuberculosis interactions via metabolic reconstructions

    Mol Syst Biol

    (2010)
  • O. Resendis-Antonio et al.

    Modeling core metabolism in cancer cells: surveying the topology underlying the Warburg effect

    PLoS ONE

    (2010)
  • T. Shlomi et al.

    Network-based prediction of human tissue-specific metabolism

    Nat Biotechnol

    (2008)
  • L. Jerby et al.

    Computational reconstruction of tissue-specific metabolic models: application to human liver metabolism

    Mol Syst Biol

    (2010)
  • A. Vazquez et al.

    Catabolic efficiency of aerobic glycolysis: the Warburg effect revisited

    BMC Syst Biol

    (2010)
  • T. Shlomi et al.

    Genome-scale metabolic modeling elucidates the role of proliferative adaptation in causing the Warburg effect

    PLoS Comput Biol

    (2011)
  • O. Folger et al.

    Predicting selective drug targets in cancer through metabolic networks

    Mol Syst Biol

    (2011)
  • R. Agren et al.

    Reconstruction of genome-scale active metabolic networks for 69 human cell types and 16 cancer types using INIT

    PLoS Comput Biol

    (2012)
  • A.I. Su et al.

    A gene atlas of the mouse and human protein-encoding transcriptomes

    Proc Natl Acad Sci U S A

    (2004)
  • D.S. Wishart et al.

    HMDB: the human metabolome database

    Nucl Acids Res

    (2007)
  • P.J. Park

    ChIP-seq: advantages and challenges of a maturing technology

    Nat Rev Genet

    (2009)
  • Z. Wang et al.

    RNA-Seq: a revolutionary tool for transcriptomics

    Nat Rev Genet

    (2009)
  • J.L. Reed

    Shrinking the metabolic solution space using experimental datasets

    PLoS Comput Biol

    (2012)
  • S.A. Becker et al.

    Context-specific metabolic networks are consistent with experiments

    PLoS Comput Biol

    (2008)
  • C. Colijn et al.

    Interpreting expression data with metabolic flux models: predicting Mycobacterium tuberculosis mycolic acid production

    PLoS Comput Biol

    (2009)
  • J.F. Moxley et al.

    Linking high-resolution metabolic flux phenotypes and transcriptional regulation in yeast modulated by the global regulator Gcn4p

    Proc Natl Acad Sci U S A

    (2009)
  • P.A. Jensen et al.

    Functional integration of a metabolic network model and expression data without arbitrary thresholding

    Bioinformatics

    (2011)
  • J. Kim et al.

    RELATCH: relative optimality in metabolic networks explains robust metabolic and regulatory responses to perturbations

    Genome Biol

    (2012)
  • J.P.v.B. Rogier

    Predicting Metabolic Fluxes Using Gene Expression Differences As Constraints

    IEEE/ACM Trans Comput Biol Bioinform

    (2011)
  • A. Bordbar et al.

    Model-driven multi-omic data analysis elucidates metabolic immunomodulators of macrophage activation

    Mol Syst Biol

    (2012)
  • R. Agren et al.

    Reconstruction of genome-scale active metabolic networks for 69 human cell types and 16 cancer types using INIT

    PLoS Comput Biol

    (2012)
  • M.J. Herrgard et al.

    Integrated analysis of regulatory and metabolic networks reveals novel regulatory mechanisms in Saccharomyces cerevisiae

    Genome Res

    (2006)
  • Cited by (34)

    • The high-resolution proteomic analysis of protein composition of rat spleen lymphocytes stimulated by Concanavalin A; a comparison with morphine-treated cells

      2020, Journal of Neuroimmunology
      Citation Excerpt :

      It also participates in gluconeogenesis by catalyzing the reverse reaction - hydration of phosphoenolpyruvate to 2-phospho-D-glycerate. The NSE was found to be expressed mostly in neurons and cells of the neuroendocrine system and, as a tumor marker, was widely used to diagnose cancer (Levin et al., 2010; Yan et al., 2011; Masoudi-Nejad and Asgari, 2015). The C-terminal moiety of NSE, which is not directly involved in catalytical activity, was shown to promote survival of neuronal cells by regulating NGF receptor dependent signaling pathways resulting in extensive remodeling of actin cytoskeleton (Vizin and Kos, 2015).

    • Transcriptomic changes associated with PCK1 overexpression in hepatocellular carcinoma cells detected by RNA-seq

      2020, Genes and Diseases
      Citation Excerpt :

      In recent years, the role of metabolism in tumorigenesis received extensive attention. A tumor is not only considered to be a genetic disease but also a metabolic disease.2 Tumor cells are achieved by reprogramming their own metabolic patterns to balance between energy and synthesis.3

    • Unravelling the complexity of signalling networks in cancer: A review of the increasing role for computational modelling

      2017, Critical Reviews in Oncology/Hematology
      Citation Excerpt :

      Further information on circos and its models can be found at http://circos.ca/in_literature/scientific/. However, to return to the section, modelling can be based on theoretical “equilibrium/constraint” states, where, for example Flux Balance Analysis uses a computed “steady-state” to construct a network; or on graphical representations of reaction sequences (as in signalling) (Nagrath et al., 2011; Masoudi-Nejad and Asgari, 2015). Of particular interest for this article is network thermodynamics (Soh and Hatzimanikatis, 2010).

    • Epithelial mesenchymal transition induces aberrant glycosylation through hexosamine biosynthetic pathway activation

      2016, Journal of Biological Chemistry
      Citation Excerpt :

      The Warburg effect consists of an increase of glucose uptake for producing energy by a high rate of glycolysis followed by lactic acid fermentation even under high oxygen tension (“aerobic glycolysis”). Understanding the metabolism of tumors remains a topic of intense study with important therapeutic potential (2, 3). Several advances in cancer metabolism research over past years have enhanced our understanding of how aerobic glycolysis and other metabolic shifts support the anabolic demands of high growth rate (4).

    View all citing articles on Scopus
    View full text