You are on page 1of 17

Leading Edge

Primer

Using Genome-scale Models


to Predict Biological Capabilities
Edward J. O’Brien,1,4,6 Jonathan M. Monk,1,2,6 and Bernhard O. Palsson1,3,5,*
1Department of Bioengineering
2Department of NanoEngineering
3Department of Pediatrics
4Bioinformatics and Systems Biology Program

University of California, San Diego, La Jolla, CA 92093, USA


5Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby 2800, Denmark
6Co-first author

*Correspondence: palsson@ucsd.edu
http://dx.doi.org/10.1016/j.cell.2015.05.019

Constraint-based reconstruction and analysis (COBRA) methods at the genome scale have been
under development since the first whole-genome sequences appeared in the mid-1990s. A few
years ago, this approach began to demonstrate the ability to predict a range of cellular functions,
including cellular growth capabilities on various substrates and the effect of gene knockouts at the
genome scale. Thus, much interest has developed in understanding and applying these methods to
areas such as metabolic engineering, antibiotic design, and organismal and enzyme evolution. This
Primer will get you started.

Introduction was created for Haemophilus influenza and appeared shortly


Bottom-up approaches to systems biology rely on constructing after this first genome was sequenced (Edwards and Palsson,
a mechanistic basis for the biochemical and genetic processes 1999), and GEMs have now grown to the level where they
that underlie cellular functions. Genome-scale network recon- enable predictive biology (Bordbar et al., 2014; McCloskey
structions of metabolism are built from all known metabolic et al., 2013; Oberhardt et al., 2009). Here, we will focus on recon-
reactions and metabolic genes in a target organism. Networks structions of metabolism and the process of converting them
are constructed based on genome annotation, biochemical into GEMs to produce computational predictions of biological
characterization, and the published scientific literature on the functions.
target organism; the latter is sometimes referred to as the bib- The fundamentals of the constraints-based reconstruction
liome. DNA sequence assembly provides a useful analogy to and analysis (COBRA) approach and its uses are also described
the process of network reconstruction (Figure 1). The genome in this Primer, which lays out the constraint-based methodology
of an organism is systematically assembled from many short at four levels. First, there is a textual description of the methods
DNA stubs, called reads, using sophisticated computer algo- and their applications. Second, visualization is presented in
rithms. Similarly, the reactome of a cell is assembled, or recon- the form of detailed figures to succinctly convey the key con-
structed, from all the biochemical reactions known or predicted cepts and applications. Third, the figure captions contain more
to be present in the target microorganism. Importantly, network detailed information about the computational approaches
reconstruction includes an explicit genetic basis for each illustrated in the figures. Fourth, the Primer provides a table of
biochemical reaction in the reactome as well as information selected detailed resources to enable an in-depth review for
about the genomic location of the gene. Thus, reconstructed the keenly interested reader. The text is organized into six sec-
networks, or an assembled reactome, for a target organism tions, each addressing a grand challenge in today’s world of
represent biochemically, genetically, and genomically structured ‘‘big data’’ biology.
knowledge bases, or BiGG k-bases. Network reconstructions
have different biological scope and coverage. They may 1. Network Reconstructions Assemble Knowledge
describe metabolism, protein-protein interactions, regulation, Systematically
signaling, and other cellular processes, but they have a unifying There is a large library of scientific publications that describe
aspect: an embedded, standardized biochemical and genetic different model organisms’ specific molecular features. Molecu-
representation amenable to computational analysis. lar biology’s focus on knowing much about a limited number of
A network reconstruction can be converted into a mathemat- molecular events changed once annotated genome sequences
ical format and thus lend itself to mathematical analysis and became available, leading to the emergence of a genome-scale
computational treatment. Genome-scale models, called GEMs, point of view. Now, putting all available knowledge about the
have been under development for nearly 15 years and have molecular processes of a target organism in context and linking
now reached a high level of sophistication. The first GEM it to its genome sequence has emerged as a grand challenge.

Cell 161, May 21, 2015 ª2015 Elsevier Inc. 971


Figure 1. Network Reconstruction
An organism’s reactome can be assembled in
a way that is analogous to DNA-sequencing
assembly. (Right to left) First the interacting com-
pounds must be identified. Then, the reactions
acting on these compounds are tabulated and
the protein that catalyzes the reaction and the
corresponding open reading frame is identified
in the organism of interest. These reactions are
assembled into pathways that can be laid out
graphically to visualize a cell’s metabolic map at
the genome scale. Several tools for reactome
assembly and curation exist, including the COBRA
Toolbox (Ebrahim et al., 2013; Schellenberger
et al., 2011b), KEGG (Kanehisa et al., 2014),
EcoCyc (Keseler et al., 2013), ModelSeed (Henry
et al., 2010b), BiGG (Schellenberger et al., 2010),
Rbionet (Thorleifsson and Thiele, 2011), Subliminal
(Swainston et al., 2011), Raven toolbox (Agren
et al., 2013), and others.

Recapitulation
Network reconstructions represent an
organized process for genome-scale as-
sembly of disparate information about a
target organism. All of this information is
Genome-scale network reconstructions were a response to this put into context with the annotated genome to form a coherent
challenge. whole that, through computations, is able to recapitulate
Network Reconstructions Organize Knowledge whole-cell functions. The grand challenge of disparate data inte-
into a Structured Format gration into a coherent whole is achieved through the formulation
The reconstruction process treats individual reactions as the of a GEM. A GEM can then compute cellular states such as an
basic elements of a network, somewhat similar to a base pair be- optimal growth state. This process is further explored in the
ing the smallest element in an assembled DNA sequence next section. A detailed reading list is available in Table S1 on
(Figure 1). To implement the metabolic reconstruction process, the network reconstruction process and software tools used to
a series of questions needs to be answered for each of the en- facilitate it.
zymes in a metabolic network. (1) What are the substrates and
products? (2) What are the stoichiometric coefficients for each 2. Converting a Genome-scale Reconstruction
metabolite that participates in the reaction (or reactions) cata- to a Computational Model
lyzed by an enzyme? (3) Are these reactions reversible? (4) In Before a reconstruction can be used to compute network prop-
what cellular compartment does the reaction occur? (5) What erties, a subtle but crucial step must be taken in which a network
gene(s) encode for the protein (or protein complex), and what reconstruction is mathematically represented. This conversion
is (are) their genomic location(s)? Genes are linked to the pro- translates a reconstructed network into a chemically accurate
teins they encode and the reactions they catalyze using the mathematical format that becomes the basis for a genome-scale
gene-protein-reaction relationship (GPR). All of this information model. This conversion requires the mathematical representa-
is assembled from a range of sources, including organism-spe- tion of metabolic reactions. The core feature of this representa-
cific databases, high-throughput data, and primary literature. tion is tabulation, in the form of a numerical matrix, of the
Establishing a set of the biochemical reactions that constitute stoichiometric coefficients of each reaction (Figure 2A). These
a reaction network in the target organism culminates in a data- stoichiometries impose systemic constraints on the possible
base of chemical equations. Reactions are then organized flow patterns (called a flux map, or flux distribution) of metabo-
into pathways, pathways into sectors (such as amino acid syn- lites through the network. These concepts are detailed below.
thesis), and ultimately into genome-scale networks, akin to reads Imposition of constraints on network functions fundamentally
becoming a full DNA sequence. This process has been differentiates the COBRA approach from models described by
described in the form of a 96-step standard operating procedure biophysical equations, which require many difficult-to-measure
(Thiele and Palsson, 2010). kinetic parameters.
Today, after many years of hard work by many researchers, Constraints are mathematically represented as equations that
there exist collections of genome-scale reconstructions (some- represent balances or as inequalities that impose bounds
times called GENREs) for a number of target organisms (Monk (Figure 2B). The matrix of stoichiometries imposes flux balance
et al., 2014; Oberhardt et al., 2011), and established protocols constraints on the network, ensuring that the total amount of
for reconstruction exist (Thiele and Palsson, 2010) that can be any compound being produced must be equal to the total
partially automated (Agren et al., 2013; Henry et al., 2010a). amount being consumed at steady state. Every reaction can

972 Cell 161, May 21, 2015 ª2015 Elsevier Inc.


also be given upper and lower bounds, which define the
maximum and minimum allowable fluxes through the reactions
that, in turn, are related to the turnover number of the enzyme
and its abundance. Once imposed on a network reconstruction,
these balances and bounds define a space of allowable flux dis-
tributions in a network—the possible rates at which every metab-
olite is consumed or produced by every reaction in the network.
The flux vector, a mathematical object, is a list of all such flux
values for a single point in the space. The flux vector represents
a ‘‘state’’ of the network that is directly related to the physiolog-
ical function that the network produces. Many other constraints
such as substrate uptake rates, secretion rates, and other limits
on reaction flux can also be imposed, further restricting the
possible state that a reconstructed network can take (Reed,
2012). The computed network states that are consistent with
all imposed constraints are thus candidate physiological states
of the target organisms under the conditions considered. The
study of the properties of this space thus becomes an important
subject.
Flux Balance Analysis Calculates Candidate Phenotypes
Flux balance analysis (FBA) is the oldest COBRA method. It is a
mathematical approach for analyzing the flow of metabolites
through a metabolic network (Orth et al., 2010). This approach
relies on an assumption of steady-state growth and mass
balance (all mass that enters the system must leave). The con-
straints discussed above take the form of equalities and inequal-
ities to define a polytope (blue area within the illustration in
Figure 2B) that represents all possible flux states of the network
given the constraints imposed. Thus, many network states are
possible under the given constraints, and multiple solutions exist
that satisfy the governing equations. The blue area is therefore
often called the ‘‘solution space’’ to denote a mathematical
space that is filled with candidate solutions to the network

(B) Constraints can be added to the model, such as: (1) enforcement of mass
balance and (2) reaction flux (v) bounds. The blue polytope represents different
possible fluxes for reactions 5 and 6, consistent with stated constraints.
Those outside of the polytope violate the imposed constraints and are thus
‘‘infeasible.’’
(C) Constraint-based models predict the flow of metabolites through a defined
network. The predicted path is determined using linear programming solvers
and is termed flux balance analysis (FBA). FBA can be used to calculate
the optimal flow of metabolites from a network input to a network output. The
desired output is described by an objective function. If the objective is to
optimize flux through reaction 5, the optimal flux distribution would correspond
to the levels of flux 5 and flux 6 at the blue point circled in the figure. The
objective function can be a simple value or can draw on a combination of
outputs, such as the biomass objective shown in (E). It is important to note that
alternate optimal flux distributions may exist to reach the optimal state, as
discussed in Figure 4C.
(D) Once a network reconstruction is converted to a mathematical format,
the inputs to the system must be defined by adding consideration of the
extracellular environment. Compounds enter and exit the extracellular
Figure 2. Formulation of a Computational Model environment via ‘‘exchange’’ reactions. The GEM will not be able to import
(A) After the metabolic network has been assembled, it must be converted into compounds unless a transport reaction from the external environment to the
a mathematical representation. This conversion is performed using a stoi- inside of the cell is present.
chiometric (S) matrix in which the stoichiometry of each metabolite involved in (E) In addition to exchange reactions, the biomass objective function acts as a
a reaction is enumerated. Reactions form the columns of this matrix and drain on cellular components in the same ratios as they are experimentally
metabolites the rows. Each metabolite’s entry corresponds to its stoichio- measured in the biomass. In FBA simulations, the biomass function is used to
metric coefficient in the corresponding reaction. Negative coefficient sub- simulate cellular growth. The biomass function is composed of all necessary
strates are consumed (reactants), and positive coefficients are produced compounds needed to create a new cell, including DNA, amino acids, lipids,
(products). Converting a metabolic network reconstruction to a mathematical and polysaccharides. This is not the only physiological objective that can be
formulation can be achieved with several of the toolboxes listed in Table S1. examined using COBRA tools.

Cell 161, May 21, 2015 ª2015 Elsevier Inc. 973


equations given the governing constraints. FBA uses the GEMs Are Input-Output ‘‘Flow Models’’
stated objective to find the solution(s) that optimize the The inner workings of a GEM are readily understood conceptu-
objective function. The solution is found using linear program- ally. In a given environment (i.e., where the nutritional inputs
ming, and, as indicated in Figure 2C, the optimal solution lies are defined) GEMs can be used to compute network outputs.
at the edges of the solution space impinging up against govern- FBA can computationally trace a fully balanced path through
ing constrains. the reactome from the available nutrients to the prerequisite
The utility of FBA has been increasingly recognized due to its output metabolite. Such calculations are performed with an
simplicity and extensibility: it requires only the information on objective function that describes the removal of the target
metabolic reaction stoichiometry and mass balances around metabolite from the network. The synthesis of biomass in a cell
the metabolites under pseudo-steady state assumption. It com- requires the simultaneous removal of about 60–70 different me-
putes how the flux map must balance to achieve a particular tabolites. Using FBA, a GEM can also compute the balanced use
homeostatic state. However, FBA has limitations. It balances of the reactome to produce all of the prerequisite metabolites for
fluxes but cannot predict metabolite concentrations. Except growth simultaneously and does so in the correct relative
in some modified forms, FBA does not account for regulatory amounts while accounting for all of the energetic, redox, and
effects such as activation of enzymes by protein kinases or chemical interactions that must balance to enable such biomass
regulation of gene expression. More details are found in the synthesis. This exercise is one of genome-scale accounting of all
caption of Figure 2, and computational resources are summa- molecules flowing through the reactome.
rized below that can be deployed to find the optimal state and Recapitulation
to study its characteristics. Given its simplicity and utility, FBA has become one of the most
Models Impose Constraints and Allow Prediction widely employed computational techniques for the systems-
One of the most basic constraints imposed on genome-scale level analysis of living organisms (Bordbar et al., 2014; Lewis
models of metabolism is that of substrate, or nutrient, availability et al., 2012). It has been successfully applied to a multitude of
and its uptake rate (Figure 2D). Metabolites enter and leave the species for modeling their cellular metabolisms (Feist and Pals-
systems through what are termed ‘‘exchange reactions’’ (i.e., son, 2008; McCloskey et al., 2013; Oberhardt et al., 2009) and
active or passive transport mechanisms). These reactions define therefore enabled a variety of applications such as metabolic
the extracellular nutritional environment and are either left engineering for the over-production of biochemicals (Yim et al.,
‘‘open’’ (to allow a substrate to enter the system at a specified 2011; Adkins et al., 2012), identification of anti-microbial
rate) or ‘‘closed’’ (the substrate can only leave the system). drug targets (Kim et al., 2011), and the elucidation of cell-cell
Measurements of the rate of exchange with the environment interactions (Bordbar et al., 2010). Further reading and detailed
are relatively easy to perform, and they prove to be some of descriptions of FBA and sources for existing genome-scale
the more important constraints placed on the possible functions models are available in Table S1.
of reaction networks internal to the cell. More biological- and
data-derived constraints can also be imposed on a model. These 3. Validation and Reconciliation of Qualitative
advanced constraints are detailed in sections 4, 5 and 6. Model Predictions
The next step in converting a network reconstruction to a Ensuring the consistency and accuracy of all of the information
model is to define what biological function(s) the network can available for a target organism is a grand challenge of
achieve. Mathematically, such a statement takes the form of genome-scale biology. Since model predictions are based on
an ‘‘objective function.’’ For predicting growth, the objective is a network reconstruction that represents the totality of what is
biomass production—that is, the rate at which the network can known about a target organism, such predictions are a critical
convert metabolites into all required biomass constituents test of our comprehensive understanding of the metabolism for
such as nucleic acids, proteins, and lipids needed to produce the target organism. Incorrect model predictions can be used
biomass. The objective of biomass production is mathematically for biological discovery by classifying them and understanding
represented by a ‘‘biomass reaction’’ that becomes an extra their underlying causes. Performing targeted experiments to
column of coefficients in the stoichiometric matrix. One can understand failed predictions is a proven method for systematic
formulate a biomass objective function at an increasing level of discovery of new biochemical knowledge (Orth and Palsson,
detail: basic, intermediate, and advanced (Feist and Palsson, 2010b). This section will focus on evaluating qualitative model
2010; Monk et al., 2014). The biomass reaction is scaled so predictions, their outcomes, underlying causes of incorrect
that the flux through it represents the growth rate (m) of the target predictions, and how to go about correcting them. Section 4
organism. discusses the same process for quantitative model predictions.
It is important to note that the biomass objective function is Genetic and Environmental Parameters
determined from measurements of biomass composition—the Genome-scale models have many genetic and environmental
uptake and secretion rates from measuring the nutrients in the parameters that can be experimentally varied. Altering the
medium—and that the model formulation is built on a knowl- composition of the growth media changes environmental pa-
edge-based network reconstruction. Thus, the growth rate rameters. Alteration of genetic parameters is achieved through
optimization problem represents ‘‘big data’’ integrated into a genome editing methods. Both environmental and genetic pa-
structured format and the hypothesis of a biological objective: rameters are explicit in GEMs, and thus the consequence of
grow as fast as possible with the resources available. This is a both types of perturbations can be computed, predicted, and
well-defined optimization problem. analyzed. The scale of such predictions has grown steadily since

974 Cell 161, May 21, 2015 ª2015 Elsevier Inc.


the first genome-scale model of E. coli appeared in 2000 (Ed- than success, as it represents an opportunity for biological dis-
wards and Palsson, 2000). covery. False-negative predictions occur when a GEM predicts
Genome-scale gene essentiality data are available from the inability to grow in a given environment without the deleted
specific projects or organism-specific databases. One can sys- gene, but the experiments show growth. This discrepancy indi-
tematically remove genes from a reconstruction and thus the cates that the reconstructed reactome is incomplete. In contrast,
corresponding reactions from the reactome and then repeat false-positive predictions occur when a GEM predicts growth
the growth computation to predict gene essentiality; if a growth but the experiment results in no growth. This outcome indicates
state cannot be computed without a particular gene, the GEM possible errors in the knowledge on which the reactome was
predicts it to be essential (Figure 3A). Such growth rate predic- based or that a regulatory process is missing that prevents the
tions of gene deletion strains have gone from 100 predictions use of a gene product factored in the computed solution.
in the year 2000 (Edwards and Palsson, 2000) to more than An example would be regulation that either represses gene
100,000 in 2013 (Österlund et al., 2013) and may be heading expression or a metabolite-enzyme interaction that inhibits the
for more than one million predictions in just a few years (Monk function of an enzyme that the GEM used to compute the pre-
and Palsson, 2014). dicted growth state.
Both environmental and genetic parameters can be varied Prediction failures can be used to systematically (i.e., algorith-
when performing FBA. The simplicity of computing growth states mically) generate hypotheses addressing the failures. Such
(i.e., an output) as a function of media composition (i.e., the nutri- hypotheses have been shown to direct experimentation to
tional inputs) with the selective removal of genes (Figure 3B) has improve our knowledge base for the target organism. Computa-
led to a number of studies that cross environmental parameters tions that vary environmental and genetic parameters become
with gene deletions. The explicit relationship between a gene part of a workflow (Figure 3C). The outcome of the workflow is
and a reaction makes the deletion of genes and their encoding a set of qualitative model predictions of growth or no growth
reactions straightforward. You can readily do this for your target that are then compared to the experimental outcome of a growth
organism, provided that you can construct a library of gene dele- screen. Correct predictions align with experimental results,
tion strains. Improved molecular tools for generating knockout while incorrect predictions do not. The two are then compared
collection libraries (Tn-seq, CRISPR systems, etc.) and improved and classified into four categories, as shown in Figure 3C. The
high-throughput methods for measuring knockout phenotypes failure modes lead to systematic experimentation.
have enabled a massive scale-up in the number of phenotypes Discovery Using Model False Negatives
that can be measured. Reconciling such discrepancies between predicted and
Classification of Model Predictions observed growth states is now a proven approach for biological
Computational predictions of outcomes fall into four categories: discovery. A series of algorithms have been developed that have
true positives, true negatives, false positives, and false nega- been shown to compute the most likely reasons for failure of
tives. The true-positive and true-negative predictions, in which prediction that, in turn, led to a model-guided experimental
computational predictions and experimental outcomes agree, inquiry and discovery. Furthermore, high-throughput tools
have generally exceeded 80%–90% for well-characterized such as phenotypic microarrays and robotic instruments are
target organisms. Going beyond single-gene knockouts to becoming available to screen cells at high rates. Such discov-
double-gene knockouts and more, true-negative predictions eries are then incorporated into the reconstruction, leading to
are particularly significant, as they indicate model predictions its iterative improvement.
of true genetic, or epistatic, interactions. In a screen of double- The discrepancies between GEM predictions and experi-
gene yeast knockouts, Szappanos et al. found that models could mental data have been used to design targeted experiments
predict 2.8% of negative genetic interactions (Szappanos et al., that correct inaccuracies in metabolic knowledge. In this
2011). While this indicates poor recall of prediction, of these, subsection, we provide three illustrative examples that detail
50% were correct, indicating that model predictions are highly how reconciliation of model errors led to the discovery of new
precise but may miss several interactions. These missed predic- metabolic capabilities in three model organisms (Figure 3D).
tions represent cases that are currently difficult for functional Human. The activity of open reading frame 103 on chromo-
geneticists to understand. For applications where the goal is to some 9 (C9orf103) of the human genome was discovered
have true predictions, such as for antibiotic design, precision is (Rolfsson et al., 2011a) using established gap-filling protocols
more important than recall. (Orth and Palsson, 2010b; Reed et al., 2006). The authors
FBA-based models are highly precise because they are focused on unconnected, ‘‘dead-end’’ metabolites in the human
good at predicting impossible states (such as when a gene metabolic network reconstruction, Recon 1 (Duarte et al., 2007).
knockout leads to death). This assumes that the network struc- Dead-end metabolites lead to model errors by creating blocked
ture is complete, an assumption that can be a problem when reactions due to a violation of mass balance. Any flux leading
promiscuous enzyme activity arises, leading to a reaction with to them cannot leave the network. In an attempt to connect
an encoding gene that is not captured in the model. Models these dead-end metabolites, a universal database of metabolic
have lower accuracy because FBA assumes that all reactions reactions was used to predict the fewest reactions required
can happen at maximum rates. Model false positives often to fully connect all metabolites in the network. Focusing on
occur because an enzyme is either transcriptionally repressed gluconate, which is a disconnected metabolite, the authors
or does not catalyze the designated reaction at a high enough experimentally characterized C9orf103, previously identified as
rate (Table S2). Predictive failure is perhaps of more interest a candidate tumor suppressor gene, as the gene that encodes

Cell 161, May 21, 2015 ª2015 Elsevier Inc. 975


Figure 3. Using Models for Qualitative Predictions and Iterative Improvement
(A) Each reaction in the network is linked to a protein and encoding gene through the gene-protein-reaction (GPR) relationship. Because each reaction in the
network corresponds to a column in the stoichiometric matrix, simply removing the column association with a particular reaction can simulate gene knockouts.
Thus, multiple KO simulations can be performed. For example, it is easy to delete every pairwise combination of 136 central carbon metabolic E. coli genes to find
double-gene knockouts that are essential for survival of the bacteria.
(B) The simplicity of altering inputs to change cellular growth environments and removing genes in silico allows one to perform simulations in millions of
experimental conditions quickly. Even on a modest laptop computer, a single FBA calculation runs in a fraction of a second, thus simulating the effect of all gene
knockouts in E. coli central metabolism in less than 10 s.
(C) Incorrect model predictions are an opportunity for biological discovery because they highlight where knowledge is missing. Targeted experiments can
be performed to discover new content that can then be added back to a model to improve its predictive accuracy. Missing model content can be discovered
using automated approaches known as ‘‘gap filling’’ (Orth and Palsson, 2010a) that query a universal database of potential reactions to restore in silico growth
to a model.
(legend continued on next page)

976 Cell 161, May 21, 2015 ª2015 Elsevier Inc.


gluconokinase, thereby consuming this metabolite and connect- dicted that the cells could route flux through the glyoxylate shunt
ing it to the rest of the human metabolic network. when ppc is removed due to the backup function of isocitrate
E. coli. Gap-filling methods combined with systematic gene lyase encoded by aceA. However, the Dppc cells were nonviable
knockouts in E. coli (Nakahigashi et al., 2009b) were used to experimentally. The protein IclR is a transcription factor that
discover new metabolic functions for the classic glycolytic en- regulates the transcription of genes involved in the glyoxylate
zymes phosphofructokinase and aldolase. Single-, double-, shunt, including aceA. Therefore a dual-knockout DppcDiclR
and triple-knockout strains of central metabolic genes were mutant was constructed. Growth was restored in this double
grown on 13 different carbon sources. Concurrently, the same mutant at 60% of the wild-type growth rate. Therefore, the
gene knockouts and growth conditions were simulated using prediction of the metabolic model of S. Typhimurium failed
the E. coli GEM. Several discrepancies between model predic- because it erroneously allowed flux through the glyoxylate
tions and experimental results were related to talAB interactions shunt when ppc was deleted due to the absence of regulatory
in the pentose phosphate pathway and could not be reconciled. information in the model.
A metabolomic analysis identified a new metabolite, sedoheptu- Adaptive laboratory evolution can also be used to reconcile
lose-1,7-bisphosphate, that had not been previously character- false-positive predictions. Often, cell populations may need
ized. Using metabolic flux analysis and in vitro enzyme assays, time to adapt to a genetic change or shift in media conditions,
the investigators confirmed that phosphofructokinase carries giving them the appearance of slow or no growth despite a
out the reaction and that glycolytic aldolase can split the model prediction of growth. However, it has been shown that
seven-carbon sugar into three- and four-carbon sugars, glycer- incorrect predictions of in silico models based on optimal per-
aldehyde-3-phosphate (G3P) and D-erythrose 4-phosphate formance criteria may be incorrect due to incomplete adaptive
(E4P), respectively. laboratory evolution under the conditions examined. It has been
Yeast. An analysis of synthetic lethal screens and gap-filling shown that E. coli K-12 grown on glycerol over 40 days
methods was used to correct incorrect pathways leading to (or about 700 generations) and subjected to a growth rate
NAD+ synthesis in yeast (Szappanos et al., 2011). The study selection pressure (passing a small fraction of the fastest
compared an experimental set of genetic interactions for growers) achieves a final growth rate that is predicted by the
metabolic genes against interactions that were predicted by GEM (Ibarra et al., 2002). The quantitative prediction of growth
FBA. Using machine-learning techniques, key changes to the rates is discussed in section 4. Thus, a false-positive result may
metabolic network that improved model accuracy were identi- indicate that the model is in fact correct, and a researcher
fied. Model refinement identified one of the two NAD+ biosyn- should be patient while the cell adapts to achieve the model-
thetic pathways from amino acids in the GEM as a source of predicted growth.
inaccurate predictions. Using growth screens with mutant Recapitulation
strains, the authors validated that the synthesis of NAD+ from Given that our knowledge of any target organism is incomplete,
amino acids was only possible from L-tryptophan (L-trp), but its network reconstruction will also be incomplete. Thus, failures
not from L-aspartate (L-asp). in GEM prediction of qualitative outcomes of growth capability
Adaptive Laboratory Evolution in the Discovery Process are informative about the completeness of a network reconstruc-
In contrast to false negatives, false positives arise when the tion and the consistency of its content. Furthermore, these
model predicts growth, but experiments show no growth approaches can be extended beyond model improvement. As
(Figure 3E). False positives occur in cases in which experimental genome editing techniques improve, in silico prediction of the
data show a particular gene to be essential but model simula- effect of multiple gene knockouts will be vital for contextualizing
tions do not. Metabolic models can be used to predict efficient results of knockout studies and engineering genomes to achieve
compensatory pathways, after which cloning and overexpres- a desired phenotype (Campodonico et al., 2014). Additionally,
sion of these pathways are performed to investigate whether reconciliation of model false negatives has been used to explore
they restore growth and to help determine why these compensa- the role that underground metabolism plays in adapting to
tory pathways are not active in mutant cells. alternate nutrient environments (Notebaart et al., 2014). The
Discovering Context-Specific Regulatory Interactions Using algorithmic procedures that have been developed to address
False-Positive Predictions. Cloning and overexpression of a failure of prediction have led to some computer-generated
false-positive associated gene has been demonstrated for a hypotheses resulting in productive experimental undertaking.
ppc knockout of Salmonella enterica serovar Typhimurium Further reading about the gap-filling process and algorithms
(Fong et al., 2013). A metabolic model of S. Typhimurium pre- for its implementation are available in Table S1.

(D) Gap-filling approaches have been used to discover new metabolic reactions in several organisms. E. coli: Two new functions for two classical glycolytic
enzymes, phosphofructokinase (PFK) and fructose-bisphosphate aldolase (FBA), were discovered (red) (Nakahigashi et al., 2009a). Human: Gluconokinase
(EC 2.7.1.12) activity was discovered based on the known presence of the metabolite 6-phosphogluconolactonate in the human reconstruction (Rolfsson et al.,
2011b) (red). Yeast: Automated model refinement suggested modifications in the NAD biosynthesis pathway. Experiments demonstrated that a parallel pathway
from aspartate thought to exist in yeast was not present (Szappanos et al., 2011).
(E) False-positive predictions can be reconciled by adding regulatory rules derived from high-throughput data (Covert et al., 2004), for example, a recent study
was able to reconcile 2,442 false-model predictions from the E. coli GEM by updating the function of just 12 genes (Barua et al., 2010). Additionally, a false-
positive growth inconsistency in the metabolic model of S. Typhimurium was reconciled by updating regulatory rules for the iclR gene product’s transcriptional
repression of aceA encoding isocitrate lyase. Transcriptional repression can also often be relieved via adaptive laboratory evolution. Such evolution drives
experimental phenotypes to achieve model predictions. Several experimental studies have shown that an organism can evolve to achieve the model-predicted
optimal growth state (Ibarra et al., 2002).

Cell 161, May 21, 2015 ª2015 Elsevier Inc. 977


4. Quantitative Phenotype Prediction through Optimality Flux Variability Analysis Calculates Possible Flux States
Principles Flux balance analysis computes an optimal objective value and
The previous section treated qualitative predictions that relate a flux state that is consistent with that objective (and all of
to the presence or absence of parts from a reconstruction. the imposed constraints). While the objective value is unique,
Quantitative predictions of phenotypic functions are more chal- multiple flux states can typically support the same objective
lenging but possible. The ability to compute quantitative organ- value in genome-scale models. For this reason, flux variability
ism functions from a genome-scale model represents a grand analysis (FVA) is used to determine the possible ranges for
challenge in systems biology. Quantitative predictions are each reaction flux (Mahadevan and Schilling, 2003). With FVA,
achievable with GEMs (even if they are based on incomplete the objective value is set to be equal to its maximum value,
reconstructions) by deploying cellular optimality principles. and each reaction is maximized and minimized. For some
Evolutionary arguments underlie the deployment of optimality- fluxes, their maximum value will be equal to their minimum,
based hypotheses. Phenotypes maximizing a hypothesized enabling a specific prediction. For others, there may be a
fitness function (as represented by an objective function) can wide range of possible values due to alternative pathways.
be computed with constrained-optimization methods (Orth Often, a parsimonious flux state is also assumed and computed
et al., 2010). with parsimonious FBA (pFBA) (Lewis et al., 2010a). With pFBA,
As for qualitative binary predictions of possible growth states, the sum of fluxes across the entire network is minimized
incorrect quantitative predictions often lead to new biological (again, subject to the optimal objective value determined);
hypotheses and understanding. However, the discoveries pFBA will eliminate some alternative pathways. Typically,
arising from quantitative phenotype predictions are typically of many reaction fluxes can be uniquely predicted with optimality
a different nature than qualitative predictions. Rather than and parsimony assumptions. Additional biological constraints in
relating to missing reconstruction content (section 3), the discov- next-generation models (section 6) reduce the possible flux
eries from quantitative phenotype prediction often relate to states further (Lerman et al., 2012).
broad, fundamental organismal constraints (Beg et al., 2007; Types of Possible and Evolutionarily Optimal
Zhuang et al., 2011b) and evolutionary objectives and trade- Quantitative Predictions
offs (Shoval et al., 2012). The simplest type of quantitative phenotype predictable with
Quantitative phenotype prediction has also proven to be constraint-based models is nutrient utilization. While metabolic
a useful capability for bioengineering applications. By opti- models do not predict absolute rates of nutrient uptake, they
mizing an engineering (instead of evolutionary) objective, predict the optimal ratios at which nutrients are utilized. For
the best possible performance of an engineered biological sys- example, metabolic models predict an optimal oxygen uptake
tem can be determined. Furthermore, the specific flux states rate relative to the carbon source uptake rate (resulting in a pre-
needed to achieve high performance can guide engineering dicted optimal ratio between the two nutrients). In an early
design. study, the ratios of oxygen and carbon uptake were shown to
Workflow for Quantitative Phenotype Prediction be predictable for a number of carbon sources in E. coli
Quantitative phenotypes can be predicted through the same (Edwards et al., 2001). In a later study, E. coli was evolved in
computational procedures used for qualitative growth predic- the laboratory on a carbon source (glycerol) for which the
tions (Figure 4A). An objective (either evolutionary or engineer- wild-type strain did not match the predicted nutrient utilization;
ing) is assumed and maximized computationally (subject to after evolution, the strain exhibited the optimal uptake rates
flux balance and other constraints). The flux state(s) that predicted theoretically (Figure 4B) (Ibarra et al., 2002). Compar-
maximize the objective are then the predicted quantitative ison of experimental and predicted phenotypes therefore
fluxes. These predictions can then be compared to experi- reveals the environments to which an organism has been evolu-
mental measurements. In cases of agreement, the evolu- tionary exposed.
tionary hypothesis is supported. In cases of a disagreement Metabolic fluxes for central carbon metabolism can be esti-
between experimental and theoretical predictions, either the mated with 13C carbon labeling experiments, making them
biological system has not been exposed to the selection candidates for quantitative prediction (Figure 4B). Since the
pressure to reach the theoretical optimum (i.e., the assumed dimensionality of carbon labeling data is larger than that for
evolutionary objective is incorrect or partially correct) or nutrient uptake, there is more opportunity to dissect the differ-
there are missing biological constraints that affect the theoret- ences in computed and measured fluxes to better understand
ical predictions (i.e., the relevant biological constraints are the multiple objectives and constraints underlying microbial
incomplete). metabolism. Impressively, the biomass objective function can
Experimental evolution can discriminate between these alter- explain a large amount of the variability of fluxes (Schuetz
natives (Ibarra et al., 2002; Schuetz et al., 2012) by exposing the et al., 2007). Failure modes in prediction have led to the appreci-
biological system to the appropriate selection pressure, leading ation of the importance of protein cost (O’Brien et al., 2013) and
it to evolve toward the stated optimum. For example, in one membrane (Zhuang et al., 2011b) and cytoplasmic spatial con-
study, strains carrying deletions of one of six metabolic genes straints (Beg et al., 2007), which affect the optimal flux state
were evolved on four different carbon sources. A total of 78% (Figure 4C). Furthermore, failure modes have led to the under-
of strains tested reached the metabolic model predicted optimal standing that metabolism is simultaneously subject to multiple
growth rate after adaptive laboratory evolution after 40 days of competing evolutionary objectives, resulting in trade-offs (e.g.,
passage (Fong and Palsson, 2004). growth versus maintenance) employed by different species

978 Cell 161, May 21, 2015 ª2015 Elsevier Inc.


Figure 4. Quantitative Phenotype Prediction
Using Optimization
(A) Quantitative phenotype prediction is an
iterative workflow. First, hypothesized biological
constraints and objectives are formulated
mathematically, and computational optimization
is used to determine optimal phenotypic states
(see section 2). The predicted phenotypic
states can then be compared to experimental
measurements to identify where predictions
are consistent. When consistent, the hypothe-
sized evolutionary objective and constraints are
validated. When inconsistent, laboratory evolu-
tion can be used to gain further insight as to
why the computed and measured states differ.
Examples of validation of quantitative pheno-
types are detailed in (B), and further hypotheses
derived from incorrect predictions are detailed
in (C).
(B) The generic workflow in (A) has been
successfully applied to several classes of phe-
notypes. (1) Nutrient utilization ratios can be
predicted by maximizing biomass flux (Edwards
et al., 2001). (2) Central carbon metabolism fluxes
can be predicted; for some organisms, much of
the variability in flux can be attributed to biomass
flux maximization (Schuetz et al., 2012). (3) The
ratio of organism abundances and nutrient ex-
changes can be predicted for both natural and
synthetic communities. Note that one important
feature of quantitative phenotype predictions is
that optimal flux solutions are often not unique.
To address this, flux variability analysis (FVA)
(Mahadevan and Schilling, 2003) can be used to
identify the ranges of possible fluxes. It should be
noted that non-uniqueness is not necessarily a
handicap of COBRA, as biological evolution can
come up with alternate solutions (Fong et al.,
2005).
(C) Inconsistencies with model predictions
have led to the appreciation of new constraints
and objectives underlying cellular phenotypes.
(1) Inconsistent predictions in by-product
secretion have led to the hypothesis that
membrane space limits membrane protein
abundance and metabolic flux (Zhuang et al.,
2011b). (2) The range of metabolic fluxes
observed across different environments has led
to the realization that fluxes can be understood
as simultaneously satisfying multiple competing
objectives, such as growth and cellular mainte-
nance. Multi-objective optimization algorithms
find solutions that maximize multiple competing
objectives.
(D) Accurate prediction of quantitative pheno-
types has led to prospective design of biological
functions. A number of algorithms have been
developed that predict genetic and/or environ-
mental perturbations required to achieve a bioengineering objective. Relevant bioengineering objectives have included biosensing, bioremediation, bio-
production, the creation of synthetic ecologies, and the intracellular production of reaction oxygen species (ROS) to potentiate antibiotic effects.

(Figure 4C). In this way, outliers in quantitative predictions can interactions. For a number of cases of communities composed
improve the understanding of constraints and objectives under- of two or three members, the optimal rate of nutrient exchange
lying a particular organism’s metabolism. and the ratio of the species in the population (Wintermute and
Optimality principles from stoichiometric models have also Silver, 2010) can be predicted. The effects of spatial organization
been expanded from single populations of cells to microbial of community members are also being uncovered (Harcombe
communities. To model microbial communities, multiple species et al., 2014). The constraints on nutrient flow between organisms
are linked together through the exchange of nutrients extra-cell- (e.g., diffusion) have proven to be important for predicting com-
ularly (Stolyar et al., 2007) or through direct electron transfer munity composition and behavior, highlighting the importance
(Nagarajan et al., 2013). The secretion rate from one species of abiotic constraints and community structure in the behavior
limits the uptake rate for others, resulting in balanced species of biological communities.

Cell 161, May 21, 2015 ª2015 Elsevier Inc. 979


Figure 5. Data Integration and Exploration of Possible Cellular Phenotypes
(A) The general workflow for multi-omic data integration begins with the conversion of the experimental data into model constraints (see B). This procedure results
in cell-type- (e.g., neuron, macrophage) and condition-specific (e.g., healthy versus diseased) models that represent the metabolic capabilities of those specific
cells (see C). Several computational procedures can then be used to explore the metabolic capabilities and determine achievable phenotypes systematically (see
D). Evaluation of these phenotypic capabilities and comparison of different cells or environments leads to identification of their molecular differences (see E).
Additionally, if the original experimental data cannot precisely distinguish between certain metabolic states, additional targeted experiments can be designed and
integrated as further constraints.
(B) Numerous data types can be integrated into metabolic models. Some directly affect model structure and variables (e.g., growth rate, biomass composition,
exchange fluxes, internal fluxes, and reaction directionality). Standard processing of these data types allows for integration into the model. Other data types affect
metabolic fluxes more indirectly. As such, different computational methods exist for formulating the appropriate constraints (Table S1).
(C) Experimental data are integrated to construct cell-type- and/or condition-specific models. These models represent the metabolic capabilities in a certain state
and are then used for further inquiry (see D and E). Specific algorithms for building cell-type-specific models from gene expression data include MBA (Jerby et al.,
2010) and GIMME (Becker and Palsson, 2008).
(legend continued on next page)

980 Cell 161, May 21, 2015 ª2015 Elsevier Inc.


Evolution is a natural counterpart to optimality-based predic- 5. Multi-Omic Data Integration: Constraining
tions with constraint-based methods. Constraint-based opti- and Exploring Possible Phenotypic States
mality predictions have focused on predicting the endpoints of With the expanding quantity of omics and other phenotypic data,
short-term experimental evolution. However, this scope of appli- there is an increasing need to integrate these data sets to drive
cation has increased in recent years to study long-term pheno- further understanding and hypothesis generation. Phenotypic
typic and enzyme evolution (Nam et al., 2012; Plata et al., 2015). data types can be integrated with metabolic GEMs to determine
From Optimality Principles to Prospective Design condition-specific capabilities and flux states in the absence of
Quantitative phenotype prediction via optimization is also assumed objectives (section 4). Computational methods that
commonly used for bioengineering applications (Figure 4D). identify the possible range of phenotypic states given the
For example, in metabolic engineering, optimal pathway yields measured data allow one to quantify the degree of (un)certainty
are used to prioritize pathways to be built into a production strain in metabolic fluxes. Some types of data are quantitative and
and to benchmark their performance. Furthermore, the flux directly indicative of metabolic fluxes, whereas other data are
states required to achieve these optima (and how they differ qualitative or indirectly related to metabolic fluxes. By layering
from wild-type growth states) can guide strain design (Cvijovic different data types, the true state of a biological system can
et al., 2011). be determined with increased precision. The need for formal
A number of design algorithms have been built to work with integration of disparate data types represents a grand challenge
metabolic models and predict the genetic and environmental that has been termed Big Data to Knowledge (BD2K, http://
modifications to increase performance (Burgard et al., 2003; bd2k.nih.gov).
Ranganathan et al., 2010). While many design algorithms and Workflow for Multi-omic Data Integration
applications have been focused on metabolite production (e.g., The overall procedure for multi-omic integration with genome-
for production of fuels and chemicals), metabolic models have scale models is an iterative workflow (Figure 5A). Once experi-
also been utilized for the design of biosensors (Tepper and mental data from the particular biological system under study
Shlomi, 2011) and biodegradation (Scheibe et al., 2009; Zhuang is obtained, it is converted into constraints on model function
et al., 2011a). Also, design has expanded beyond single popula- (Figure 5B). The successive application of experimentally
tions to microbial communities/ecosystems (Klitgord and Segrè, derived constraints to the reaction network results in the gener-
2010). ation of a cell-type- and condition-specific model (Figure 5C).
Recapitulation Several computational procedures can then be used to explore
Quantitative phenotype predictions initially focused on simple the metabolic capabilities and achievable phenotypes of the
physiological predictions and are still expanding to more com- experimentally constrained model (Figure 5D). Evaluation of
plex phenotypes, biological systems (Levy and Borenstein, these phenotypic capabilities and comparison of different cells
2013), and environments. Although there have been notable suc- or environments lead to identification of their molecular differ-
cesses of quantitative phenotype prediction, certain phenotypes ences (Figure 5E), providing biological insight and driving further
are still difficult to predict. Historically, difficult predictions have hypotheses.
led to the development of new computational methods and an Converting Data to Model Constraints
appreciation of new biological constraints. Table S2 summarizes Successive imposition of constraints is a basic principle of
several types of predictions and the approximate performance of COBRA (Palsson, 2000). Some data types can be directly con-
constraint-based methods utilized to date. The expansion in the verted into constraints on model variables. Biomass composi-
scope and accuracy of predictions continues today, with models tion and growth rate affect the metabolic demands of cellular
of increased scope (Chang et al., 2013a; O’Brien et al., 2013), growth (Feist and Palsson, 2010). Time-course exo-metabolo-
discussed in section 6. mics can be used to set the uptake and secretion rates of nutri-
Thus far, quantitative phenotypes have been limited primarily ents (Mo et al., 2009). Intracellular quantitative metabolomics
to microbial systems and, more recently, plants (Collakova combined with reaction free energies can discern condition-
et al., 2012; Williams et al., 2010). For multi-cellular organisms, specific reaction directionalities (Henry et al., 2007). Isotopomer
specialized cell types support the fitness of the entire organism. distributions from cellular biomass or metabolite pools can be
Cell-type-specific ‘‘objectives’’ have been constructed (Chang used to infer and constrain intracellular fluxes (Zamboni et al.,
et al., 2010), though they typically are used for qualitative (sec- 2009). These data can be used separately or combined to iden-
tion 3) rather than quantitative phenotype prediction. Instead, tify with increasing precision the true state of the cell.
quantitative phenotypes in multi-cellular organisms are typically Other data types affect metabolism more qualitatively. In the-
determined through model-driven analysis of experimental data, ory, quantitative metabolite, transcript, and protein levels can be
discussed in section 5. used to constrain metabolism quantitatively, but in practice,

(D) After adding constraints to the model, computational procedures are used to assess the implication of the experimental data on metabolic fluxes. The two
main methods for querying the consequences of the measured data on a cell’s phenotypes are flux variability analysis (FVA) and Markov-chain Monte-Carlo
(MCMC) sampling. (1) FVA determines the maximum and minimum values of all metabolic fluxes. (2) MCMC sampling randomly samples feasible metabolic flux
vectors (usually resulting in tens to hundreds of thousands of flux vectors). These sampled flux vectors can then be used to derive the distribution of possible flux
values for a given metabolic reaction.
(E) Often a comparative approach is employed in which experimental data from two conditions are used to generate two condition-specific models. Then, the
achievable phenotypes of the two states are compared (e.g., though MCMC sampling, see D).

Cell 161, May 21, 2015 ª2015 Elsevier Inc. 981


this requires many parameters that are hard to measure and are (Schellenberger et al., 2010), whereas others create auto-gener-
organism specific. Instead, these data types can be used as ated layouts and new tools allow for the drawing of maps based
qualitative constraints relating to gene product or metabolite on flux solutions (King and Ebrahim, 2014). Finally, identifying
presence/absence; that is, if a metabolite is present, a reaction reactions or subsystems that remain partially identified (e.g.,
must be active that produces it (Shlomi et al., 2008), and if a based on a large FVA range) can guide further experimentation,
gene product is absent, its catalyzed reactions cannot carry resulting in an iterative computational and experimental elucida-
flux (Jerby et al., 2010; Schmidt et al., 2013). Similarly, regulatory tion of a cell’s state.
interactions can be added to affect the presence/absence of Recapitulation
a gene product based on condition-specific activity of a tran- GEMs can be used to integrate numerous data types. In fact,
scription factor (Chandrasekaran and Price, 2010). as more experimentally derived constraints are successively
Cell-Type- and Condition-Specific Models imposed, analysis often becomes easier (as the range of
Starting from a large reconstructed reaction network (e.g., repre- possible solutions shrinks [Reed, 2012]) instead of more chal-
senting all metabolic reactions encoded in the human genome lenging, as often occurs with statistically based data integration
[Thiele et al., 2013]), the imposition of experimental data results procedures. A current challenge with metabolic GEMs is the
in the generation of cell-type- and condition-specific models. explicit integration of data types that do not directly reflect meta-
Experimentally derived constraints pare down the achievable bolic fluxes (e.g., transcriptomics, proteomics, and regulatory
phenotypes from those encoded by the totality of the cell’s interactions). This challenge is primarily due to the fact that
genome. By eliminating phenotypes that cannot be achieved, these processes are not explicitly described in metabolic
this new model represents the capabilities of the particular cell models. Expansion of metabolic models to encompass gene
type and environment assayed. This model summarizes the expression hold promise to address this challenge and are
experimental data in a self-consistent and integrated format discussed in section 6.
and forms the starting point for further computational and biolog-
ical inquiry (Agren et al., 2012; Shlomi et al., 2008) (see Figures 6. Moving beyond Metabolism to Molecular Biology
5D and 5E). Up to this point, this Primer has focused on metabolic models,
Quantifying Uncertainty or M models. M models have reached a high degree of sophisti-
Once a cell-type- and condition-specific model is created, cation after 15 years of development, resulting in standard
computational methods are used to determine the possible operating procedures for their construction (Thiele and Palsson,
flux states of the cell. FVA (which is described in section 4) 2010) and use (Schellenberger et al., 2011a). However, M
(Mahadevan and Schilling, 2003) can be used to determine the models are limited in their explicit coverage to metabolic fluxes.
range of fluxes that are consistent with the experimental data. Thus, a grand challenge in the field has been to expand the con-
A more refined approach is flux sampling (Schellenberger and cepts of constraint-based models of metabolism to other cellular
Palsson, 2009) (typically with Markov Chain Monte Carlo processes to formally include more disparate data types in
[MCMC] methods), which determines the distribution of fluxes genome-scale models (Reed and Palsson, 2003).
for all reactions (instead of simply the range). When no cellular Computing Properties of the Proteome
objective is assumed, the feasible flux space is very uncon- The process of addressing this grand challenge has begun
strained and a particular reaction could be operating at nearly (Figure 6A). Recently, genome-scale network reconstructions
any flux value. As more data are layered, the feasible flux space have expanded to encompass aspects of molecular biology.
decreases. When no objective is assumed, fluxes are rarely Two significant expansions are genome-scale models integrated
precisely known, and many will remain completely unknown. with protein structures, GEM-PRO, and integrated models of
However, an imprecisely known flux space is often sufficient metabolism and protein expression, ME models. GEM-PRO al-
to discern differences between two environments/states as lows for structural bioinformatics analysis to be performed
discussed in the following subsection. from a systems-level perspective and to have those results in
Using Computed States to Drive Discovery turn affect network simulations. ME models allow for the simula-
Once the range of possible phenotypic states is quantified, they tion of proteome synthesis and account for the capacity and
must be analyzed to gain biological insights. Often a compara- metabolic requirements of gene expression.
tive approach is employed, in which two experimental states A Structural Biology View of Cellular Networks
(e.g., neurons from Alzheimer’s disease patients compared to GEM-PRO reconstructions can have varying degrees of detail,
healthy controls [Lewis et al., 2010b]) are compared. Reactions which affects the types of analysis possible. So far, GEM-PRO
that have a non-overlapping FVA range must be different be- reconstructions have been created for T. maritima (Zhang
tween the two states and can be indicative of important meta- et al., 2009) and E. coli (Chang et al., 2013a; Chang et al.,
bolic changes. In cases in which the FVA ranges are overlapping, 2013b). Initial reconstructions have focused on single peptide
the flux distributions from MCMC sampling can still be different; chains (Zhang et al., 2009) and have utilized homology modeling
that is, the reactions are likely different between the two states, to fill in gaps where organism-specific structures have not been
but the current experimental data are insufficient to guarantee it. identified. Further reconstruction detail has included protein-
Pathway visualization is also helpful in gaining insight into ligand complexes (Chang et al., 2013a) and quaternary protein
changes in cell states—fluxes (or flux ranges) are most com- assemblies (Chang et al., 2013b). To link the structures to the
prehensible in a network context. A few tools exist for the visual- metabolic model, structural data directly reference the GPRs in
ization of metabolic fluxes; some are based on static maps the metabolic reconstruction. For cases of protein-metabolite

982 Cell 161, May 21, 2015 ª2015 Elsevier Inc.


Figure 6. Expansion of Genome-scale
Models to Encompass Molecular Biology
(A) Metabolic models have been expanded to
encompass the processes of proteome synthesis
and localization as well as data on protein struc-
tures. Models including protein synthesis and
localization are referred to as ME models, which
stands for metabolism and gene expression. GEM-
PRO refers to genome-scale models integrated
with protein structures. For GEM-PRO, a combi-
nation of structural data directly references the
GPRs in the metabolic reconstruction; structures
can be obtained from experimental databases or
homology modeling. The E. coli ME model mech-
anistically accounts for 80% of the proteome
mass in conditions of exponential growth and
100% of other major cell constituents (DNA, RNA,
cell wall, lipids, etc.).
(B) Addition of cellular processes vastly increases
the predictive scope of models. ME models
can predict biomass composition, abundances
of protein across subsystems, and differential
gene expression in certain environmental shifts
(in addition to the predictions possible with M
models); like FBA, these were predicted by
assuming growth maximization as an evolutionary
objective, though the specific optimization algo-
rithm differs due to the addition of coupling con-
straints. GEM-PRO has been used to predict
the metabolic bottlenecks and growth defects of
changes in temperature on protein stability and
catalysis; protein stability is predicted with struc-
tural bioinformatics methods and is then used
to limit the catalyzed metabolic flux. The uses of
these integrated models are just beginning to be
explored.

for its synthesis. This represents an inte-


grated view of metabolic biochemistry
and the core processes of molecular
biology. As with GEM-PRO, the first ME
complexes, the metabolites also need to be properly annotated models were formulated for T. maritima (Lerman et al., 2012)
in the structural data. The structural reconstruction therefore and E. coli (O’Brien et al., 2013; Thiele et al., 2012).
provides a physical embodiment of the gene-protein-reaction The reconstruction of a ME model starts with the formation of
relationship. reactions for gene expression and enzyme synthesis (Thiele
There are a few notable cases demonstrating the unique anal- et al., 2009). The processes explicitly accounted for in ME
ysis possible with the combination of protein structures and models are very detailed, including transcription units and
network models. In T. maritima, network context and protein initiation and termination factors for transcription, tRNAs and
fold annotations were combined to test alternative models for chaperones needed for translation and protein folding, and metal
pathway evolution (Zhang et al., 2009). The T. maritima GEM- ion and prosthetic group requirements for catalysis. In other
PRO supported the patchwork model for genesis of new meta- words, the reconstructions strive to match as closely as possible
bolic pathways. In E. coli, the effect of temperature on protein all of the biochemical processes required to synthesize fully
stability and enzyme activity was simulated at the systems level, functional enzymes. To create a ME model, the reactions for
recapitulating the effects of temperature on growth (Chang et al., enzyme synthesis are coupled to the totality of metabolic
2013a). Also in E. coli, protein-ligand interactions were combined reactions with pseudo-kinetic constraints, termed ‘‘coupling
with gene essentiality predictions to discover new antibiotic constraints’’ (Lerman et al., 2012; Thiele et al., 2010). These con-
leads and off-targets (Chang et al., 2013b). These examples straints relate the abundance of an enzyme (or any ‘‘recyclable’’
just scratch the surface of analyses made possible with the inte- chemical species, e.g., mRNA, tRNA) to its degradation rate and
gration of network and structural biology. catalytic capacity.
Modeling Molecular Biology and Metabolism ME models thus significantly expand the scope of possible
ME models formalize all of the requirements for biosynthesis phenotype predictions to include aspects of transcription and
of the functional proteome (Figure 6B). They compute the translation. RNA and protein biomass composition are variables
proteome composition and its integrated function to produce in ME models and are no longer set a priori (as in the biomass
phenotypic states and all of the metabolic processes needed objective function of M models). ME models predict the

Cell 161, May 21, 2015 ª2015 Elsevier Inc. 983


experimentally observed linear changes in the ratio of RNA-to- ME models (Sun et al., 2013) and structural simulations for
protein mass fractions as a consequence of changes in protein GEM-PRO have been rapid and are likely to ameliorate these
synthesis demands (O’Brien et al., 2013). Furthermore, the challenges.
mass fractions of protein subsystems agree well with those Like discoveries enabled by comparing M model predictions
predicted by the ME model. This shows that the broad distri- to experimental data, we anticipate that much biology can be
bution of protein subsystem abundance is predictable using learned from comparing in silico and in vivo proteome allocation
optimality principles, and the comparison reveals that some (O’Brien and Palsson, 2015), leading to increasingly predictive
subsystems were under-predicted, thus identifying them as models. The E. coli ME model currently encompasses many
gaps in knowledge and targets for further reconstruction and key cellular functions, covering 80% of the proteome by
model refinement (Liu et al., 2014). While the quantitative predic- mass in conditions of exponential growth; the remaining prote-
tion of individual protein abundances is currently beyond the ome mass outside of the scope of the model can guide model
scope of the ME model (as these demands depend on expansion. In addition to DNA replication and cell division (Karr
enzyme-specific kinetics), the ME model has been shown to et al., 2012), much of the remaining proteome mass involves
accurately predict differential expression across certain environ- cellular stress responses (e.g., pH, osmolarity, osmotic); like
mental shifts due to the differential requirements of proteins with temperature, GEM-PRO will aid in modeling these cellular
across conditions (a more qualitative than quantitative predic- stresses.
tion) (Lerman et al., 2012).
A recent expansion to the ME model includes the addition of
Perspective
protein translocation, allowing for the localization of protein to
Genome-scale models have been under development since
be computed (Liu et al., 2014) (i.e., into cytoplasm, periplasm,
the first annotated genome sequences appeared in the late
and inner and outer membrane). Translocase abundances and
1990s. For most of this history, the focus of GEMs has been
compartmentalized proteome mass were accurately predicted
on metabolism. After initial successes with metabolic GEMs,
from the bottom up based on optimality principles. Addition of
it became clear that the same approach could be applied to
compartmentalization also allows for membrane area and cyto-
other cellular process that could be reconstructed in biochem-
plasmic volume constraints to be formalized, which, if combined
ically accurate detail. Thus, a vision was laid out in 2003
with GEM-PRO, approaches a digital embodiment of a three-
that the path to whole-cell models was conceptually possible
dimensional cell.
and that such models could be used as a context for mecha-
Recapitulation
nistically integrating disparate omic data types (Reed and
The predictive ability of metabolic models are dictated by the
Palsson, 2003). This vision is now being realized. This Primer
scope of the reconstruction. Nearly all of the predictions of meta-
shows how six grand challenges in cell, molecular, and
bolic models outlined in the previous sections can be refined and
systems biology can be addressed using GEMs. A surprising
expanded with GEM-PRO or ME models. Advances to include
range of cellular functions and phenotypic states can now be
protein structures and protein synthesis open new vistas for
dealt with.
constraint-based modeling.
We now have the tools at hand to develop quantitative geno-
The scope of genetic perturbations (section 2) that can be
type-phenotype relationships from first principles and at the
simulated is significantly larger due to the inclusion of genes
genome scale. Current models of prokaryotes account for meta-
for gene expression (and accounting for protein cost) and the
bolism, transcription, translation, protein localization, and pro-
effects of coding mutations on protein structures; GEM-PRO
tein structure. Processes not described in the current ME models
also expands the scope of environmental perturbation to enable
will be systematically reconstructed over the coming years to
simulation of changes in temperature. GEM-PRO allows for
gain a more and more comprehensive description of cellular
new gap-filling approaches (section 3) based on structural bioin-
functions. Biology can thus look forward to the continued
formatics methods. ME models expand the scope of quantitative
development and use of a mechanistic framework for the study
molecular phenotypes to include transcript and protein levels
of biological phenomena just as physics and chemistry have
(section 4), and transcriptomics and proteomics can be analyzed
enjoyed for over a century.
in mechanistic detail (section 5).
With the added capabilities of GEM-PRO and ME models also
comes additional computational challenges. While single-opti- SUPPLEMENTAL INFORMATION
mization calculations with M models take less than a second
Supplemental Information includes two tables and can be found with this
on a modest laptop computer, growth maximization with a ME article online at http://dx.doi.org/10.1016/j.cell.2015.05.019.
model can take more than an hour. The ME model also requires
specialized high-precision solvers. Many promising applications
AUTHOR CONTRIBUTIONS
of GEM-PRO will require simulation of protein dynamics with
molecular dynamics (MD) and hybrid quantum mechanics/ E.J.O., J.M.M., and B.O.P. conceived, wrote, and edited the manuscript.
molecular mechanics (QM/MM) simulations on protein struc-
tures. High-performance computing environments are required
ACKNOWLEDGMENTS
for such simulations, and there is a pervasive trade-off between
the precision of simulations and the scope of structural This work was supported by The Novo Nordisk Foundation and the National
coverage. However, advances in high-precision solvers for Institutes of Health grant number R01 GM057089.

984 Cell 161, May 21, 2015 ª2015 Elsevier Inc.


REFERENCES Edwards, J.S., and Palsson, B.O. (2000). The Escherichia coli MG1655 in silico
metabolic genotype: its definition, characteristics, and capabilities. Proc. Natl.
Adkins, J., Pugh, S., McKenna, R., and Nielsen, D.R. (2012). Engineering Acad. Sci. USA 97, 5528–5533.
microbial chemical factories to produce renewable ‘‘biomonomers’’. Front Edwards, J.S., Ibarra, R.U., and Palsson, B.O. (2001). In silico predictions of
Microbiol 3, 313. Escherichia coli metabolic capabilities are consistent with experimental
Agren, R., Bordel, S., Mardinoglu, A., Pornputtapong, N., Nookaew, I., and data. Nat. Biotechnol. 19, 125–130.
Nielsen, J. (2012). Reconstruction of genome-scale active metabolic networks Feist, A.M., and Palsson, B.O. (2008). The growing scope of applications of
for 69 human cell types and 16 cancer types using INIT. PLoS Comput. Biol. 8, genome-scale metabolic reconstructions using Escherichia coli. Nat. Bio-
e1002518. technol. 26, 659–667.
Agren, R., Liu, L., Shoaie, S., Vongsangnak, W., Nookaew, I., and Nielsen, J. Feist, A.M., and Palsson, B.O. (2010). The biomass objective function. Curr.
(2013). The RAVEN toolbox and its use for generating a genome-scale meta- Opin. Microbiol. 13, 344–349.
bolic model for Penicillium chrysogenum. PLoS Comput. Biol. 9, e1002980.
Fong, S.S., and Palsson, B.O. (2004). Metabolic gene-deletion strains of
Barua, D., Kim, J., and Reed, J.L. (2010). An automated phenotype-driven
Escherichia coli evolve to computationally predicted growth phenotypes.
approach (GeneForce) for refining metabolic and regulatory models. PLoS
Nat. Genet. 36, 1056–1058.
Comput. Biol. 6, e1000970.
Fong, S.S., Joyce, A.R., and Palsson, B.O. (2005). Parallel adaptive evolution
Becker, S.A., and Palsson, B.O. (2008). Context-specific metabolic networks
cultures of Escherichia coli lead to convergent growth phenotypes with
are consistent with experiments. PLoS Comput. Biol. 4, e1000082.
different gene expression states. Genome Res. 15, 1365–1372.
Beg, Q.K., Vazquez, A., Ernst, J., de Menezes, M.A., Bar-Joseph, Z., Barabási,
Fong, N.L., Lerman, J.A., Lam, I., Palsson, B.O., and Charusanti, P. (2013).
A.L., and Oltvai, Z.N. (2007). Intracellular crowding defines the mode and
Reconciling a Salmonella enterica metabolic model with experimental data
sequence of substrate uptake by Escherichia coli and constrains its metabolic
confirms that overexpression of the glyoxylate shunt can rescue a lethal ppc
activity. Proc. Natl. Acad. Sci. USA 104, 12663–12668.
deletion mutant. FEMS Microbiol. Lett. 342, 62–69.
Bordbar, A., Lewis, N.E., Schellenberger, J., Palsson, B.O., and Jamshidi, N.
Harcombe, W.R., Riehl, W.J., Dukovski, I., Granger, B.R., Betts, A., Lang, A.H.,
(2010). Insight into human alveolar macrophage and M. tuberculosis interac-
Bonilla, G., Kar, A., Leiby, N., Mehta, P., et al. (2014). Metabolic resource
tions via metabolic reconstructions. Mol. Syst. Biol. 6, 422.
allocation in individual microbes determines ecosystem interactions and
Bordbar, A., Monk, J.M., King, Z.A., and Palsson, B.O. (2014). Constraint- spatial dynamics. Cell Rep. 7, 1104–1115.
based models predict metabolic and associated cellular functions. Nat. Rev.
Henry, C.S., Broadbelt, L.J., and Hatzimanikatis, V. (2007). Thermodynamics-
Genet. 15, 107–120.
based metabolic flux analysis. Biophys. J. 92, 1792–1805.
Burgard, A.P., Pharkya, P., and Maranas, C.D. (2003). Optknock: a bilevel pro-
Henry, C.S., DeJongh, M., Best, A.A., Frybarger, P.M., Linsay, B., and
gramming framework for identifying gene knockout strategies for microbial
Stevens, R.L. (2010a). High-throughput generation, optimization and analysis
strain optimization. Biotechnol. Bioeng. 84, 647–657.
of genome-scale metabolic models. Nat. Biotechnol. 28, 977–982.
Campodonico, M.A., Andrews, B.A., Asenjo, J.A., Palsson, B.O., and Feist,
Henry, C.S., DeJongh, M., Best, A.A., Frybarger, P.M., Linsay, B., and
A.M. (2014). Generation of an atlas for commodity chemical production in
Stevens, R.L. (2010b). High-throughput generation, optimization and analysis
Escherichia coli and a novel pathway prediction algorithm, GEM-Path. Metab.
of genome-scale metabolic models. Nat. Biotechnol. 28, 977–982.
Eng. 25, 140–158.
Chandrasekaran, S., and Price, N.D. (2010). Probabilistic integrative modeling Ibarra, R.U., Edwards, J.S., and Palsson, B.O. (2002). Escherichia coli K-12
of genome-scale metabolic and regulatory networks in Escherichia coli and undergoes adaptive evolution to achieve in silico predicted optimal growth.
Mycobacterium tuberculosis. Proc. Natl. Acad. Sci. USA 107, 17845–17850. Nature 420, 186–189.

Chang, R.L., Xie, L., Xie, L., Bourne, P.E., and Palsson, B.Ø. (2010). Drug off- Jerby, L., Shlomi, T., and Ruppin, E. (2010). Computational reconstruction of
target effects predicted using structural analysis in the context of a metabolic tissue-specific metabolic models: application to human liver metabolism.
network model. PLoS Comput. Biol. 6, e1000938. Mol. Syst. Biol. 6, 401.

Chang, R.L., Andrews, K., Kim, D., Li, Z., Godzik, A., and Palsson, B.O. Kanehisa, M., Goto, S., Sato, Y., Kawashima, M., Furumichi, M., and Tanabe,
(2013a). Structural systems biology evaluation of metabolic thermotolerance M. (2014). Data, information, knowledge and principle: back to metabolism in
in Escherichia coli. Science 340, 1220–1223. KEGG. Nucleic Acids Res. 42, D199–D205.

Chang, R.L., Xie, L., Bourne, P.E., and Palsson, B.O. (2013b). Antibacterial Karr, J.R., Sanghvi, J.C., Macklin, D.N., Gutschow, M.V., Jacobs, J.M., Bolival,
mechanisms identified through structural systems pharmacology. BMC B., Jr., Assad-Garcia, N., Glass, J.I., and Covert, M.W. (2012). A whole-cell
Syst. Biol. 7, 102. computational model predicts phenotype from genotype. Cell 150, 389–401.
Collakova, E., Yen, J.Y., and Senger, R.S. (2012). Are we ready for genome- Keseler, I.M., Mackie, A., Peralta-Gil, M., Santos-Zavaleta, A., Gama-Castro,
scale modeling in plants? Plant Sci. 191-192, 53–70. S., Bonavides-Martı́nez, C., Fulcher, C., Huerta, A.M., Kothari, A., Krumme-
nacker, M., et al. (2013). EcoCyc: fusing model organism databases with sys-
Covert, M.W., Knight, E.M., Reed, J.L., Herrgard, M.J., and Palsson, B.O.
tems biology. Nucleic Acids Res. 41, D605–D612.
(2004). Integrating high-throughput and computational data elucidates bacte-
rial networks. Nature 429, 92–96. Kim, H.U., Kim, S.Y., Jeong, H., Kim, T.Y., Kim, J.J., Choy, H.E., Yi, K.Y., Rhee,
J.H., and Lee, S.Y. (2011). Integrative genome-scale metabolic analysis of
Cvijovic, M., Bordel, S., and Nielsen, J. (2011). Mathematical models of cell
Vibrio vulnificus for drug targeting and discovery. Mol. Syst. Biol. 7, 460.
factories: moving towards the core of industrial biotechnology. Microb. Bio-
technol. 4, 572–584. King, Z.A., and Ebrahim, A. (2014). escher: Escher 1.0.0 Beta 3. ZENODO.
Duarte, N.C., Becker, S.A., Jamshidi, N., Thiele, I., Mo, M.L., Vo, T.D., Srivas, Klitgord, N., and Segrè, D. (2010). Environments that induce synthetic micro-
R., and Palsson, B.O. (2007). Global reconstruction of the human metabolic bial ecosystems. PLoS Comput. Biol. 6, e1001002.
network based on genomic and bibliomic data. Proc. Natl. Acad. Sci. USA Lerman, J.A., Hyduke, D.R., Latif, H., Portnoy, V.A., Lewis, N.E., Orth, J.D.,
104, 1777–1782. Schrimpe-Rutledge, A.C., Smith, R.D., Adkins, J.N., Zengler, K., and Palsson,
Ebrahim, A., Lerman, J.A., Palsson, B.O., and Hyduke, D.R. (2013). COBRApy: B.O. (2012). In silico method for modelling metabolism and gene product
COnstraints-Based Reconstruction and Analysis for Python. BMC Syst. Biol. expression at genome scale. Nat. Commun. 3, 929.
7, 74. Levy, R., and Borenstein, E. (2013). Metabolic modeling of species interaction
Edwards, J.S., and Palsson, B.O. (1999). Systems properties of the Haemophi- in the human microbiome elucidates community-level assembly rules. Proc.
lus influenzae Rd metabolic genotype. J. Biol. Chem. 274, 17410–17416. Natl. Acad. Sci. USA 110, 12804–12809.

Cell 161, May 21, 2015 ª2015 Elsevier Inc. 985


Lewis, N.E., Hixson, K.K., Conrad, T.M., Lerman, J.A., Charusanti, P., Polpi- Orth, J.D., Thiele, I., and Palsson, B.O. (2010). What is flux balance analysis?
tiya, A.D., Adkins, J.N., Schramm, G., Purvine, S.O., Lopez-Ferrer, D., et al. Nat. Biotechnol. 28, 245–248.
(2010a). Omic data from evolved E. coli are consistent with computed optimal Österlund, T., Nookaew, I., Bordel, S., and Nielsen, J. (2013). Mapping
growth from genome-scale models. Mol. Syst. Biol. 6, 390. condition-dependent regulation of metabolism in yeast through genome-scale
Lewis, N.E., Schramm, G., Bordbar, A., Schellenberger, J., Andersen, M.P., modeling. BMC Syst. Biol. 7, 36.
Cheng, J.K., Patel, N., Yee, A., Lewis, R.A., Eils, R., et al. (2010b). Large-scale Palsson, B. (2000). The challenges of in silico biology. Nat. Biotechnol. 18,
in silico modeling of metabolic interactions between cell types in the human 1147–1150.
brain. Nat. Biotechnol. 28, 1279–1285.
Plata, G., Henry, C.S., and Vitkup, D. (2015). Long-term phenotypic evolution
Lewis, N.E., Nagarajan, H., and Palsson, B.O. (2012). Constraining the meta- of bacteria. Nature 517, 369–372.
bolic genotype-phenotype relationship using a phylogeny of in silico methods.
Ranganathan, S., Suthers, P.F., and Maranas, C.D. (2010). OptForce: an opti-
Nat. Rev. Microbiol. 10, 291–305.
mization procedure for identifying all genetic manipulations leading to targeted
Liu, J.K., O’Brien, E.J., Lerman, J.A., Zengler, K., Palsson, B.O., and Feist, overproductions. PLoS Comput. Biol. 6, e1000744.
A.M. (2014). Reconstruction and modeling protein translocation and compart-
Reed, J.L. (2012). Shrinking the metabolic solution space using experimental
mentalization in Escherichia coli at the genome-scale. BMC Syst. Biol. 8, 110.
datasets. PLoS Comput. Biol. 8, e1002662.
Mahadevan, R., and Schilling, C.H. (2003). The effects of alternate optimal
Reed, J.L., and Palsson, B.O. (2003). Thirteen years of building constraint-
solutions in constraint-based genome-scale metabolic models. Metab. Eng.
based in silico models of Escherichia coli. J. Bacteriol. 185, 2692–2699.
5, 264–276.
Reed, J.L., Patel, T.R., Chen, K.H., Joyce, A.R., Applebee, M.K., Herring, C.D.,
McCloskey, D., Palsson, B.O., and Feist, A.M. (2013). Basic and applied uses
Bui, O.T., Knight, E.M., Fong, S.S., and Palsson, B.O. (2006). Systems
of genome-scale metabolic network reconstructions of Escherichia coli. Mol.
approach to refining genome annotation. Proc. Natl. Acad. Sci. USA 103,
Syst. Biol. 9, 661.
17480–17484.
Mo, M.L., Palsson, B.O., and Herrgård, M.J. (2009). Connecting extracellular
Rolfsson, O., Palsson, B.O., and Thiele, I. (2011a). The human metabolic
metabolomic measurements to intracellular flux states in yeast. BMC Syst.
reconstruction Recon 1 directs hypotheses of novel human metabolic func-
Biol. 3, 37.
tions. BMC Syst. Biol. 5, 155.
Monk, J., and Palsson, B.O. (2014). Genetics. Predicting microbial growth.
Rolfsson, O., Palsson, B.O., and Thiele, I. (2011b). The human metabolic
Science 344, 1448–1449.
reconstruction Recon 1 directs hypotheses of novel human metabolic func-
Monk, J., Nogales, J., and Palsson, B.O. (2014). Optimizing genome-scale tions. BMC Syst. Biol. 5, 155.
network reconstructions. Nat. Biotechnol. 32, 447–452.
Scheibe, T.D., Mahadevan, R., Fang, Y., Garg, S., Long, P.E., and Lovley, D.R.
Nagarajan, H., Embree, M., Rotaru, A.E., Shrestha, P.M., Feist, A.M., Palsson, (2009). Coupling a genome-scale metabolic model with a reactive transport
B.O., Lovley, D.R., and Zengler, K. (2013). Characterization and modelling model to describe in situ uranium bioremediation. Microb. Biotechnol. 2,
of interspecies electron transfer mechanisms and microbial community dy- 274–286.
namics of a syntrophic association. Nat. Commun. 4, 2809.
Schellenberger, J., and Palsson, B.O. (2009). Use of randomized sampling for
Nakahigashi, K., Toya, Y., Ishii, N., Soga, T., Hasegawa, M., Watanabe, H., analysis of metabolic networks. J. Biol. Chem. 284, 5457–5461.
Takai, Y., Honma, M., Mori, H., and Tomita, M. (2009a). Systematic phenome
Schellenberger, J., Park, J.O., Conrad, T.M., and Palsson, B.O. (2010). BiGG: a
analysis of Escherichia coli multiple-knockout mutants reveals hidden reac-
Biochemical Genetic and Genomic knowledgebase of large scale metabolic
tions in central carbon metabolism. Mol. Syst. Biol. 5, 306.
reconstructions. BMC Bioinformatics 11, 213.
Nakahigashi, K., Toya, Y., Ishii, N., Soga, T., Hasegawa, M., Watanabe, H.,
Schellenberger, J., Que, R., Fleming, R.M., Thiele, I., Orth, J.D., Feist, A.M.,
Takai, Y., Honma, M., Mori, H., and Tomita, M. (2009b). Systematic phenome
Zielinski, D.C., Bordbar, A., Lewis, N.E., Rahmanian, S., et al. (2011a). Quan-
analysis of Escherichia coli multiple-knockout mutants reveals hidden reac-
titative prediction of cellular metabolism with constraint-based models: the
tions in central carbon metabolism. Mol. Syst. Biol. 5, 306.
COBRA Toolbox v2.0. Nat. Protoc. 6, 1290–1307.
Nam, H., Lewis, N.E., Lerman, J.A., Lee, D.H., Chang, R.L., Kim, D., and
Schellenberger, J., Que, R., Fleming, R.M., Thiele, I., Orth, J.D., Feist, A.M.,
Palsson, B.O. (2012). Network context and selection in the evolution to
Zielinski, D.C., Bordbar, A., Lewis, N.E., Rahmanian, S., et al. (2011b). Quan-
enzyme specificity. Science 337, 1101–1104.
titative prediction of cellular metabolism with constraint-based models: the
Notebaart, R.A., Szappanos, B., Kintses, B., Pál, F., Györkei, Á., Bogos, B., COBRA Toolbox v2.0. Nat. Protoc. 6, 1290–1307.
} , B., Wagner, A., et al. (2014). Network-level archi-
Lázár, V., Spohn, R., Csörgo
Schmidt, B.J., Ebrahim, A., Metz, T.O., Adkins, J.N., Palsson, B.O., and
tecture and the evolutionary potential of underground metabolism. Proc. Natl.
Hyduke, D.R. (2013). GIM3E: condition-specific models of cellular metabolism
Acad. Sci. USA 111, 11762–11767.
developed from metabolomics and expression data. Bioinformatics 29, 2900–
O’Brien, E.J., and Palsson, B.O. (2015). Computing the functional proteome: 2908.
recent progress and future prospects for genome-scale models. Curr. Opin.
Schuetz, R., Kuepfer, L., and Sauer, U. (2007). Systematic evaluation of objec-
Biotechnol. 34C, 125–134.
tive functions for predicting intracellular fluxes in Escherichia coli. Mol. Syst.
O’Brien, E.J., Lerman, J.A., Chang, R.L., Hyduke, D.R., and Palsson, B.O. Biol. 3, 119.
(2013). Genome-scale models of metabolism and gene expression extend
Schuetz, R., Zamboni, N., Zampieri, M., Heinemann, M., and Sauer, U. (2012).
and refine growth phenotype prediction. Mol. Syst. Biol. 9, 693.
Multidimensional optimality of microbial metabolism. Science 336, 601–604.
Oberhardt, M.A., Palsson, B.O., and Papin, J.A. (2009). Applications of Shlomi, T., Cabili, M.N., Herrgård, M.J., Palsson, B.O., and Ruppin, E. (2008).
genome-scale metabolic reconstructions. Mol. Syst. Biol. 5, 320. Network-based prediction of human tissue-specific metabolism. Nat. Bio-
Oberhardt, M.A., Pucha1ka, J., Martins dos Santos, V.A., and Papin, J.A. technol. 26, 1003–1010.
(2011). Reconciliation of genome-scale metabolic reconstructions for compar- Shoval, O., Sheftel, H., Shinar, G., Hart, Y., Ramote, O., Mayo, A., Dekel, E.,
ative systems analysis. PLoS Comput. Biol. 7, e1001116. Kavanagh, K., and Alon, U. (2012). Evolutionary trade-offs, Pareto optimality,
Orth, J.D., and Palsson, B.O. (2010a). Systematizing the generation of missing and the geometry of phenotype space. Science 336, 1157–1160.
metabolic knowledge. Biotechnol. Bioeng. 107, 403–412. Stolyar, S., Van Dien, S., Hillesland, K.L., Pinel, N., Lie, T.J., Leigh, J.A., and
Orth, J.D., and Palsson, B.O. (2010b). Systematizing the generation of missing Stahl, D.A. (2007). Metabolic modeling of a mutualistic microbial community.
metabolic knowledge. Biotechnol. Bioeng. 107, 403–412. Mol. Syst. Biol. 3, 92.

986 Cell 161, May 21, 2015 ª2015 Elsevier Inc.


Sun, Y., Fleming, R.M., Thiele, I., and Saunders, M.A. (2013). Robust flux A community-driven global reconstruction of human metabolism. Nat. Bio-
balance analysis of multiscale biochemical reaction networks. BMC Bioinfor- technol. 31, 419–425.
matics 14, 240.
Thorleifsson, S.G., and Thiele, I. (2011). rBioNet: A COBRA toolbox extension
Swainston, N., Smallbone, K., Mendes, P., Kell, D., and Paton, N. (2011). The for reconstructing high-quality biochemical networks. Bioinformatics 27,
SuBliMinaL Toolbox: automating steps in the reconstruction of metabolic 2009–2010.
networks. J. Integr. Bioinform. 8, 186.
Williams, T.C., Poolman, M.G., Howden, A.J., Schwarzlander, M., Fell, D.A.,
Szappanos, B., Kovács, K., Szamecz, B., Honti, F., Costanzo, M., Baryshni-
Ratcliffe, R.G., and Sweetlove, L.J. (2010). A genome-scale metabolic model
kova, A., Gelius-Dietrich, G., Lercher, M.J., Jelasity, M., Myers, C.L., et al.
accurately predicts fluxes in central carbon metabolism under stress condi-
(2011). An integrated approach to characterize genetic interaction networks
tions. Plant Physiol. 154, 311–323.
in yeast metabolism. Nat. Genet. 43, 656–662.
Tepper, N., and Shlomi, T. (2011). Computational design of auxotrophy- Wintermute, E.H., and Silver, P.A. (2010). Emergent cooperation in microbial
dependent microbial biosensors for combinatorial metabolic engineering metabolism. Mol. Syst. Biol. 6, 407.
experiments. PLoS ONE 6, e16274. Yim, H., Haselbeck, R., Niu, W., Pujol-Baxley, C., Burgard, A., Boldt, J., Khan-
Thiele, I., and Palsson, B.O. (2010). A protocol for generating a high-quality durina, J., Trawick, J.D., Osterhout, R.E., Stephen, R., et al. (2011). Metabolic
genome-scale metabolic reconstruction. Nat. Protoc. 5, 93–121. engineering of Escherichia coli for direct production of 1,4-butanediol. Nat.
Thiele, I., Jamshidi, N., Fleming, R.M., and Palsson, B.O. (2009). Genome- Chem. Biol. 7, 445–452.
scale reconstruction of Escherichia coli’s transcriptional and translational Zamboni, N., Fendt, S.M., Rühl, M., and Sauer, U. (2009). (13)C-based meta-
machinery: a knowledge base, its mathematical formulation, and its functional bolic flux analysis. Nat. Protoc. 4, 878–892.
characterization. PLoS Comput. Biol. 5, e1000312.
Zhang, Y., Thiele, I., Weekes, D., Li, Z., Jaroszewski, L., Ginalski, K., Deacon,
Thiele, I., Fleming, R.M., Bordbar, A., Schellenberger, J., and Palsson, B.O.
A.M., Wooley, J., Lesley, S.A., Wilson, I.A., et al. (2009). Three-dimensional
(2010). Functional characterization of alternate optimal solutions of Escheri-
structural view of the central metabolic network of Thermotoga maritima.
chia coli’s transcriptional and translational machinery. Biophys. J. 98, 2072–
Science 325, 1544–1549.
2081.
Thiele, I., Fleming, R.M., Que, R., Bordbar, A., Diep, D., and Palsson, B.O. Zhuang, K., Izallalen, M., Mouser, P., Richter, H., Risso, C., Mahadevan, R.,
(2012). Multiscale modeling of metabolism and macromolecular synthesis in and Lovley, D.R. (2011a). Genome-scale dynamic modeling of the competition
E. coli and its application to the evolution of codon usage. PLoS ONE 7, between Rhodoferax and Geobacter in anoxic subsurface environments.
e45635. ISME J. 5, 305–316.
Thiele, I., Swainston, N., Fleming, R.M., Hoppe, A., Sahoo, S., Aurich, M.K., Zhuang, K., Vemuri, G.N., and Mahadevan, R. (2011b). Economics of mem-
Haraldsdottir, H., Mo, M.L., Rolfsson, O., Stobbe, M.D., et al. (2013). brane occupancy and respiro-fermentation. Mol. Syst. Biol. 7, 500.

Cell 161, May 21, 2015 ª2015 Elsevier Inc. 987

You might also like