Feeds:
Posts
Comments

Posts Tagged ‘efflux transporter’

Extracellular evaluation of intracellular flux in yeast cells

Larry H. Bernstein, MD, FCAP, Reviewer and Curator

Leaders in Pharmaceutical Intelligence

This is the fourth article in a series on metabolomics, which is a major development in -omics, integrating transcriptomics, proteomics,  genomics, metabolic pathways analysis, metabolic and genomic regulatory control using computational mapping.  In the previous two part presentation, flux analysis was not a topic for evaluation, but here it is the major focus.  It is a study of yeast cells, and bears some relationship to the comparison of glycemia, oxidative phosphorylation, TCA cycle, and ETC in leukemia cell lines.  In the previous study – system flux was beyond the scope of analysis, and explicitly stated.  The inferences made in comparing the two lymphocytic leukemia cells was of intracellular metabolism from extracellular measurements.  The study of yeast cells is aimed at looking at cellular effluxes, which is also an important method for studying pharmacological effects and drug resistance.

Metabolomic series

1.  Metabolomics, Metabonomics and Functional Nutrition: the next step in nutritional metabolism and biotherapeutics

http://pharmaceuticalintelligence.com/2014/08/22/metabolomics-metabonomics-and-functional-nutrition-the-next-step-in-nutritional-metabolism-and-biotherapeutics/

2.  Metabolomic analysis of two leukemia cell lines. I

http://pharmaceuticalintelligence.com/2014/08/23/metabolomic-analysis-of-two-leukemia-cell-lines-_i/

3.  Metabolomic analysis of two leukemia cell lines. II.

 http://pharmaceuticalintelligence.com/2014/08/24/metabolomic-analysis-of-two-leukemia-cell-lines-ii/

4.  Extracellular evaluation of intracellular flux in yeast cells

Q1. What is efflux?

Q2. What measurements were excluded from the previous study that would not allow inference about fluxes?

Q3. Would this study bear any relationship to the Pasteur effect?

Q4 What is a genome scale network reconstruction?

Q5 What type of information is required for a network prediction model?

Q6. Is there a difference between the metabolites profiles for yeast grown under aerobic and anaerobuc conditions – under the constrainsts?

Q7.  If there is a difference in the S metabolism, would there be an effect on ATP production?

 

 

Connecting extracellular metabolomic measurements to intracellular flux
states in yeast

Monica L Mo1Bernhard Ø Palsson1 and Markus J Herrgård12*

Author Affiliations

1 Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA

2 Current address: Synthetic Genomics, Inc, 11149 N Torrey Pines Rd, La Jolla, CA 92037, USA

For all author emails, please log on.

BMC Systems Biology 2009, 3:37  doi:10.1186/1752-0509-3-37

 

The electronic version of this article is the complete one and can be found online at: http://www.biomedcentral.com/1752-0509/3/37

 

Received: 15 December 2008
Accepted: 25 March 2009
Published: 25 March 2009

© 2009 Mo et al; licensee BioMed Central Ltd.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Background

Metabolomics has emerged as a powerful tool in the

  • quantitative identification of physiological and disease-induced biological states.

Extracellular metabolome or metabolic profiling data, in particular,

  • can provide an insightful view of intracellular physiological states in a noninvasive manner.

Results

We used an updated genome-scale

  • metabolic network model of Saccharomyces cerevisiae, iMM904, to investigate
  1. how changes in the extracellular metabolome can be used
  2. to study systemic changes in intracellular metabolic states.

The iMM904 metabolic network was reconstructed based on

  • an existing genome-scale network, iND750,
  • and includes 904 genes and 1,412 reactions.

The network model was first validated by

  • comparing 2,888 in silico single-gene deletion strain growth phenotype predictions
  • to published experimental data.

Extracellular metabolome data measured

  • of ammonium assimilation pathways 
  • in response to environmental and genetic perturbations

was then integrated with the iMM904 network

  • in the form of relative overflow secretion constraints and
  • a flux sampling approach was used to characterize candidate flux distributions allowed by these constraints.

Predicted intracellular flux changes were

  • consistent with published measurements
  • on intracellular metabolite levels and fluxes.

Patterns of predicted intracellular flux changes

  • could also be used to correctly identify the regions of
  • the metabolic network that were perturbed.

Conclusion

Our results indicate that

  • integrating quantitative extracellular metabolomic profiles
  • in a constraint-based framework
  • enables inferring changes in intracellular metabolic flux states.

Similar methods could potentially be applied

  • towards analyzing biofluid metabolome variations
  • related to human physiological and disease states.

Background

“Omics” technologies are rapidly generating high amounts of data

  • at varying levels of biological detail.

In addition, there is a rapidly growing literature and

  • accompanying databases that compile this information.

This has provided the basis for the assembly of

  • genome-scale metabolic networks for various microbial and eukaryotic organisms [111].

These network reconstructions serve

  • as manually curated knowledge bases of
  • biological information as well as
  • mathematical representations of biochemical components and
  • interactions specific to each organism.

genome-scale network reconstruction is

  • structured collection of genes, proteins, biochemical reactions, and metabolites
  • determined to exist and operate within a particular organism.

This network can be converted into a predictive model

  • that enables in silico simulations of allowable network states based on
  • governing physico-chemical and genetic constraints [12,13].

A wide range of constraint-based methods have been developed and applied

  • to analyze network metabolic capabilities under
  • different environmental and genetic conditions [13].

These methods have been extensively used to

  • study genome-scale metabolic networks and have successfully predicted, for example,
  1. optimal metabolic states,
  2. gene deletion lethality, and
  3. adaptive evolutionary endpoints [1416].

Most of these applications utilize

  • optimization-based methods such as flux balance analysis (FBA)
  • to explore the metabolic flux space.

However, the behavior of genome-scale metabolic networks can also be studied

  • using unbiased approaches such as
  • uniform random sampling of steady-state flux distributions [17].

Instead of identifying a single optimal flux distribution based on

  • a given optimization criterion (e.g. biomass production),

these methods allow statistical analysis of

  • a large range of possible alternative flux solutions determined by
  • constraints imposed on the network.

Sampling methods have been previously used to study

  1. global organization of E. coli metabolism [18] as well as
  2. to identify candidate disease states in the cardiomyocyte mitochondria [19].

Network reconstructions provide a structured framework

  • to systematically integrate and analyze disparate datasets
  • including transcriptomic, proteomic, metabolomic, and fluxomic data.

Metabolomic data is one of the more relevant data types for this type of analysis as

  1. network reconstructions define the biochemical links between metabolites, and
  2. recent advancements in analytical technologies have allowed increasingly comprehensive
  • intracellular and extracellular metabolite level measurements [20,21].

The metabolome is

  1. the set of metabolites present under a given physiological condition
  2. at a particular time and is the culminating phenotype resulting from
  • various “upstream” control mechanisms of metabolic processes.

Of particular interest to this present study are

  • the quantitative profiles of metabolites that are secreted into the extracellular environment
  • by cells under different conditions.

Recent advances in profiling the extracellular metabolome (EM) have allowed

  • obtaining insightful biological information on cellular metabolism
  • without disrupting the cell itself.

This information can be obtained through various

  • analytical detection,
  • identification, and
  • quantization techniques

for a variety of systems ranging from

  • unicellular model organisms to human biofluids [2023].

Metabolite secretion by a cell reflects its internal metabolic state, and

  • its composition varies in response to
  • genetic or experimental perturbations
  • due to changes in intracellular pathway activities
  • involved in the production and utilization of extracellular metabolites [21].

Variations in metabolic fluxes can be reflected in EM changes which can

  • provide insight into the intracellular pathway activities related to metabolite secretion.

The extracellular metabolomic approach has already shown promise

  • in a variety of applications, including
  1. capturing detailed metabolite biomarker variations related to disease and
  2. drug-induced states and
  3. characterizing gene functions in yeast [2427].

However, interpreting changes in the extracellular metabolome can be challenging

  • due to the indirect relationship between the proximal cause of the change
    (e.g. a mutation)
  • and metabolite secretion.

Since metabolic networks describe

  • mechanistic,
  • biochemical links between metabolites,

integrating such data can allow a systematic approach

  • to identifying altered pathways linked to
  • quantitative changes in secretion profiles.

Measured secretion rates of major byproduct metabolites

  • can be applied as additional exchange flux constraints
  • that define observed metabolic behavior.

For example, a recent study integrating small-scale EM data

  • with a genome-scale yeast model
  • correctly predicted oxygen consumption and ethanol production capacities
  • in mutant strains with respiratory deficiencies [28].

The respiratory deficient mutant study

  • used high accuracy measurements for a small number of
  • major byproduct secretion rates
  • together with an optimization-based method well suited for such data.

Here, we expand the application range of the model-based method used in [28]

  • to extracellular metabolome profiles,
  • which represent a temporal snapshot of the relative abundance
  • for a larger number of secreted metabolites.

Our approach is complementary to

  • statistical (i.e. “top-down”) approaches to metabolome analysis [29]
  • and can potentially be used in applications such as biofluid-based diagnostics or
  • large-scale characterization of mutants strains using metabolite profiles.

This study implements a constraint-based sampling approach on

  • an updated genome-scale network of yeast metabolism
  • to systematically determine how EM level variations

are linked to global changes in intracellular metabolic flux states.

By using a sampling-based network approach and statistical methods (Figure 1),

  • EM changes were linked to systemic intracellular flux perturbations
    in an unbiased manner
  • without relying on defining single optimal flux distributions
  • used in the previously mentioned study [28].

The inferred perturbations in intracellular reaction fluxes were further analyzed

  • using reporter metabolite and subsystem (i.e., metabolic pathway) approaches [30]
  • in order to identify dominant metabolic features that are collectively perturbed (Figure 2).

The sampling-based approach also has the additional benefit of

  • being less sensitive to inaccuracies in metabolite secretion profiles than
  • optimization-based methods and can effectively be used – in biofluid metabolome analysis.
integration of exometabolomic (EM) data

integration of exometabolomic (EM) data

Figure 1. Schematic illustrating the integration of exometabolomic (EM) data with the constraint-based framework.

(A) Cells are subjected to genetic and/or environmental perturbations to secrete metabolite patterns unique to that condition.
(B) EM is detected, identified, and quantified.
(C) EM data is integrated as required secretion flux constraints to define allowable solution space.
(D) Random sampling of solution space yields the range of feasible flux distributions for intracellular reactions.
(E) Sampled fluxes were compared to sampled fluxes of another condition to determine

  • which metabolic regions were altered between the two conditions (see Figure 2).

(F) Significantly altered metabolic regions were identified.

http://www.biomedcentral.com/content/figures/1752-0509-3-37-1.jpg

 

sampling and scoring analysis to determine intracellular flux changes

sampling and scoring analysis to determine intracellular flux changes

Figure 2. Schematic of sampling and scoring analysis to determine intracellular flux changes.

(A) Reaction fluxes are sampled for two conditions.
(B & C) Sample of flux differences is calculated by selecting random flux values from each condition

  • to obtain a distribution of flux differences for each reaction.

(D) Standardized reaction Z-scores are determined, which represent

  • how far the sampled flux differences deviates from a zero flux change.

Reaction scores can be used in

  1. visualizing perturbation subnetworks and
  2. analyzing reporter metabolites and subsystems.

http://www.biomedcentral.com/content/figures/1752-0509-3-37-2.jpg

This study was divided into two parts and describes:

(i) the reconstruction and validation of an expanded S. cerevisiae metabolic network, iMM904; and
(ii) the systematic inference of intracellular metabolic states from

  • two yeast EM data sets using a constraint-based sampling approach.

The first EM data set compares wild type yeast to the gdh1/GDH2 (glutamate dehydrogenase) strain [31],

  • which indicated good agreement between predicted metabolic changes
  • of intracellular metabolite levels and fluxes [31,32].

The second EM data set focused on secreted amino acid measurements

  • from a separate study of yeast cultured in different
    ammonium and potassium concentrations [33].

We analyzed the EM data to gain further insight into

  • perturbed ammonium assimilation processes as well as
  1. metabolic states relating potassium limitation and
  2. ammonium excess conditions to one another.

The model-based analysis of both

  • separately published extracellular metabolome datasets
  • suggests a relationship between
  1. glutamate,
  2. threonine and
  3. folate metabolism,
  • which are collectively perturbed when
    ammonium assimilation processes are broadly disrupted
  1. either by environmental (excess ammonia) or
  2. genetic (gene deletion/overexpression) perturbations.

The methods herein present an approach to

  • interpreting extracellular metabolome data and
  • associating these measured secreted metabolite variations
  • to changes in intracellular metabolic network states.

Additional file 1. iMM904 network content.

The data provided represent the content description of the iMM904 metabolic network and
detailed information on the expanded content.

Format: XLS Size: 2.7MB Download file

This file can be viewed with: Microsoft Excel Viewer

Additional file 2. iMM904 model files.

The data provided are the model text files of the iMM904 metabolic network
that is compatible with the available COBRA Toolbox [13]. The model structure
can be loaded into Matlab using the ‘SimPhenyPlus’ format with GPR and compound information.

Format: ZIP Size: 163KB Download file

Conversion of the network to a predictive model

The network reconstruction was converted to a constraint-based model using established procedures [13].

Network reactions and metabolites were assembled into a stoichiometric matrix 

  • containing the stoichiometric coefficients of the reactions in the network.

The steady-state solution space containing possible flux distributions

  • is determined by calculating the null space of S= 0,

where is the reaction flux vector.

Minimal media conditions were set through constraints on exchange fluxes

  • corresponding to the experimental measured substrate uptake rates.

All the model-based calculations were done using the Matlab COBRA Toolbox [13]

  • utilizing the glpk or Tomlab/CPLEX (Tomopt, Inc.) optimization solvers.

Chemostat growth simulations

The iMM904 model was initially validated by

  1. simulating wild type yeast growth in aerobic and anaerobic
    carbon-limited chemostat conditions
  2. and comparing the simulation results to published experimental data

on substrate uptake and byproduct secretion in these conditions [34].

The study was performed following the approach taken to validate the iFF708 model in a previous study [35].

The predicted glucose uptake rates were determined

  1. by setting the in silico growth rate to the measured dilution rate,
    – equivalent under continuous culture growth,
  2. and minimizing the glucose uptake rate.

The accuracy of in silico predictions of

  • substrate uptake and byproduct secretion by the iMM904 model
  • was similar to the accuracy obtained using the iFF708 model
  • and results are shown in Figure S1 [see Additional file 3].

Additional file 3. Supplemental figures. 

The file provides the supplemental figures and descriptions of S1, S2, S3, and S4.

Format: PDF Size: 513KB Download file

This file can be viewed with: Adobe Acrobat Reader

Genome-scale gene deletion phenotype predictions

The iMM904 network was further validated by

  • performing genome-scale gene lethality computations
  • following established procedures to determine growth phenotypes
  1. under minimal medium conditions and
  2. compared to published data.

A modified version of the biomass function used in previous iND750 studies

  1. was set as the objective to be maximized and
  2. gene deletions were simulated by

setting the flux through the corresponding reaction(s) to zero.

The biomass function was based on the experimentally measured

  1. composition of major cellular constituents
  2. during exponential growth of yeast cells and
  3. was reformulated to include trace amounts of
  4. additional cofactors and metabolites
  5. with the assumed fractional contribution of 10-.

These additional biomass compounds were included

according to the biomass formulation used in the iLL672 study

  • to improve lethality predictions through
  • the inclusion of additional essential biomass components [3].

The model was constrained by limiting

  1. the carbon source uptake to 10 mmol/h/gDW
  2. and oxygen uptake to 2 mmol/h/gDW.

Ammonia, phosphate, and sulfate were assumed to be non-limiting.

The experimental phenotyping data was obtained

  • using strains that were auxotrophic for
  1. methionine,
  2. leucine,
  3. histidine, and
  4. uracil.

These auxotrophies were simulated

  1. by deleting the appropriate genes from the model and
  2. supplementing the in silico strain with the appropriate supplements
  3. at non-limiting, but low levels.

Furthermore, trace amounts of essential nutrients that are present

  • in the experimental minimal media formulation
  1. 4-aminobenzoate,
  2. biotin,
  3. inositol,
  4. nicotinate,
  5. panthothenate,
  6. thiamin)
  • were supplied in the simulations [3].

Three distinct methods to simulate the outcome of gene deletions were utilized:

  1. Flux-balance analysis (FBA) [36-38],
  2. Minimization of Metabolic Adjustment (MoMA) [39], and
  3. a linear version of MoMA (linearMoMA).

In the linearMoMA method, minimization of the quadratic objective function
of the original MoMA algorithm

  • was replaced by minimization of the corresponding 1-norm objective function
    (i.e. sum of the absolute values of the differences of wild type FBA solution
    and the knockout strain flux solution).

The computed results were then compared to growth phenotype data
(viable/lethal) from a previously published experimental gene deletion study [3].

The comparison between experimental and in silico deletion phenotypes involved

  • choosing a threshold for the predicted relative growth rate of
  • a deletion strain that is considered to be viable.

We used standard ROC curve analysis

  • to assess the accuracy of different prediction methods and models
  • across the full range of the viability threshold parameter,
    results shown in Figure S2 [see Additional file 3].

The ROC curve plots the true viable rate against the false viable rate

  • allowing comparison of different models and methods
  • without requiring arbitrarily choosing this parameter a priori [40].

The optimal prediction performance corresponds to

  • the point closest to the top left corner of the ROC plot
    (i.e. 100% true viable rate, 0% false viable rate).

Table 1

Table 1 Comparison of iMM904 and iLL672 gene deletion predictions and experimental data under minimal media conditions
Media Model Method True viable False viable False lethal True lethal True viable % False viable % MCC
Glucose iMM904 full FBA 647 10 32 33 95.29 23.26 0.6
iMM904 full linMOMA 644 10 35 33 94.85 23.26 0.58
iMM904 full MOMA 644 10 35 33 94.85 23.26 0.58
iMM904 red FBA 440 9 28 33 94.02 21.43 0.61
iMM904 red linMOMA 437 9 31 33 93.38 21.43 0.6
iMM904 red MOMA 437 9 31 33 93.38 21.43 0.6
iLL672 full MOMA 433 9 35 33 92.52 21.43 0.57
Galactose iMM904 full FBA 595 32 36 59 94.29 35.16 0.58
iMM904 full linMOMA 595 32 36 59 94.29 35.16 0.58
iMM904 full MOMA 595 32 36 59 94.29 35.16 0.58
iMM904 red FBA 409 12 33 56 92.53 17.65 0.67
iMM904 red linMOMA 409 12 33 56 92.53 17.65 0.67
iMM904 red MOMA 409 12 33 56 92.53 17.65 0.67
iLL672 full MOMA 411 19 31 49 92.99 27.94 0.61
Glycerol iMM904 full FBA 596 43 36 47 94.3 47.78 0.48
iMM904 full linMOMA 595 44 37 46 94.15 48.89 0.47
iMM904 full MOMA 598 44 34 46 94.62 48.89 0.48
iMM904 red FBA 410 20 34 46 92.34 30.3 0.57
iMM904 red linMOMA 409 21 35 45 92.12 31.82 0.56
iMM904 red MOMA 412 21 32 45 92.79 31.82 0.57
iLL672 full MOMA 406 20 38 46 91.44 30.3 0.55
Ethanol iMM904 full FBA 593 45 29 55 95.34 45 0.54
iMM904 full linMOMA 592 45 30 55 95.18 45 0.54
iMM904 full MOMA 592 44 30 56 95.18 44 0.55
iMM904 red FBA 408 21 27 54 93.79 28 0.64
iMM904 red linMOMA 407 21 28 54 93.56 28 0.63
iMM904 red MOMA 407 20 28 55 93.56 26.67 0.64
iLL672 full MOMA 401 13 34 62 92.18 17.33 0.68
MCC, Matthews correlation coefficient (see Methods). Note that the iLL672 predictions were obtained directly from [3] and thus the viability threshold was not optimized using the maximum MCC approach.
Mo et al. BMC Systems Biology 2009 3:37  http://dx.doi.org:/10.1186/1752-0509-3-37

 

The values reported in Table 1 correspond to selecting

  • the optimal viability threshold based on this criterion.

We summarized the overall prediction accuracy of a model and method

  • using the Matthews Correlation Coefficient (MCC) [40].

The MCC ranges from -1 (all predictions incorrect) to +1 (all predictions correct) and

  • is suitable for summarizing overall prediction performance

in our case where there are substantially more viable than lethal gene deletions.

ROC plots were produced in Matlab (Mathworks, Inc.).

 

Table 1. Comparison of iMM904 and iLL672

  • gene deletion predictions and
  • experimental data

Inferring perturbed metabolic regions based on EM profiles

The method implemented in this study is shown schematically in Figures 1 and 2

Constraining the iMM904 network 

Relative levels of quantitative EM data were incorporated into the constraint-based framework

  • as overflow secretion exchange fluxes to simulate the required low-level production of
  • experimentally observed excreted metabolites.

The primary objective of this study is to associate

  • relative metabolite levels that are generally measured for metabonomic or biofluid analyses
  • to the quantitative ranges of intracellular reaction fluxes required to produce them.

However, without detailed kinetic information or dynamic metabolite measurements available,

  • we approximated EM datasets of relative quantitative metabolite levels
  • to be proportional to the rate in which they are secreted and detected
  • (at a steady state) – into the extracellular media.

This approach is analogous to approximating uptake rates based

  • on metabolite concentrations from a previous study performing sampling analysis
  • on a cardiomyocyte mitochondrial network
  • to identify differential flux distribution ranges

for various environmental (i.e. substrate uptake) conditions [19].

The raw data was normalized by the raw maximum value of the dataset
(thus the maximum secretion flux was 1 mmol/hr/gDW) with

  • an assumed error of 10%
  • to set the lower and upper bounds and thus
  • inherently accounting for sampling calculation sensitivity.

The gdh1/GDH2 strains were flask cultured under minimal glucose media conditions; thus,

  • glucose and oxygen uptake rates were set at 15 and 2 mmol/hr/gDW, respectively,
  • for the gdh1/GDH2 strain study.

In the anaerobic case the oxygen uptake rate was set to zero, and

  • sterols and fatty acids were provided as in silico supplements as described in [35].

For the potassium limitation/ammonium toxicity study

  • the growth rate was set at 0.17 1/h, and
  • the glucose uptake rate was minimized
  • to mimic experimental chemostat cultivation conditions.

These input constraints were constant for each perturbation and comparative wild-type condition

  • such that the calculated solution spaces between the conditions
  • differed based only on variations in the output secretion constraints.

FBA optimization of EM-constrained networks

A modified FBA method with minimization of the 1-norm objective function

  • between two optimal flux distributions was used
  • to determine optimal intracellular fluxes
  • based on the EM-constrained metabolic models.

This method determines two optimal flux distributions simultaneously

  • for two differently constrained models (e.g. wild type vs. mutant) –
  • these flux distributions maximize biomass production in each case and
  • the 1-norm distance between the distributions is as small as possible
  • given the two sets of constraints.

This approach avoids problems with

  • alternative optimal solutions when comparing two FBA-computed flux distributions
  • by assuming minimal rerouting of flux distibution between a perturbed network and its reference network.

Reaction flux changes from the FBA optimization results were determined

  • by computing the relative percentage fold change for each reaction
  • between the mutant and wild-type flux distributions.

Random sampling of the steady-state solution space

We utilized artificial centering hit-and-run (ACHR) Monte Carlo sampling [19,41]

  • to uniformly sample the metabolic flux solution space
  • defined by the constraints described above.

Reactions, and their participating metabolites, found to participate in intracellular loops [42]

  • were discarded from further analysis as these reactions can have arbitrary flux values.

The following sections describe the approaches used for the analysis of the different datasets.

Sampling approach used in the gdh1/GDH2 study

Due to the overall shape of the metabolic flux solution space,

  • most of the sampled flux distributions resided close to the minimally allowed growth rate
    (i.e. biomass production) and
  • corresponded to various futile cycles that utilized substrates but
  • did not produce significant biomass.

In order to study more physiologically relevant portions of the flux space

  • we restricted the sampling to the part of the solution space
  • where the growth rate was at least 50% of the maximum growth rate
  • for the condition as determined by FBA.

This assumes that cellular growth remains an important overall objective by the yeast cells

  • even in batch cultivation conditions, but
  • that the intracellular flux distributions
  • may not correspond to maximum biomass production [43].

To test the sensitivity of the results to the minimum growth rate threshold,

  • separate Monte Carlo samples were created for each minimum threshold
  • ranging from 50% to 100% at 5% increments.

We also tested the sensitivity of the results

  • to the relative magnitude of the extracellular metabolite secretion rates
  • by performing the sampling at three different relative levels

(0 corresponding to no extracellular metabolite secretion, maximum rate of 0.5 mmol/hr/gDW,
and maximum rate of 1.0 mmol/hr/gDW).

For each minimum growth rate threshold and extracellular metabolite secretion rate,

  • the ACHR sampler was run for 5 million steps and
  • a flux distribution was stored every 5000 steps.

The sensitivity analysis results are presented in Figures S3 and S4 [see Additional File 3], and

  • the results indicate that the reaction Z-scores (see below) are not significantly affected by
  1. either the portion of the solution space sampled or
  2. the exact scaling of secretion rates.

The final overall sample used was created by combining the samples for all minimum growth rate thresholds

  • for the highest extracellular metabolite secretion rate (maximum 1 mmol/hr/gDW).

This approach allowed biasing the sampling towards

  • physiologically relevant parts of the solution space
  • without imposing the requirement of strictly maximizing a predetermined objective function.

The samples obtained with no EM data were used as control samples

  • to filter reporter metabolites/subsystems whose scores were significantly high
  • due to only random differences between sampling runs.

Sampling approach used in the potassium limitation/ammonium toxicity study

Since the experimental data used in this study was generated in chemostat conditions, and

  • previous studies have indicated that chemostat flux patterns predicted by FBA are
  • close to the experimentally measured ones [43],
  • we assumed that sampling of the optimal solution space was appropriate for this study.

In order to sample a physiologically reasonable range of flux distributions,

  • samples for four different oxygen uptake rates
    (1, 2, 3, and 4 mmol/hr/gDW with 5 million steps each)
  • were combined in the final analysis.

Standardized scoring of flux differences between perturbation and control conditions

Z-score based approach was implemented to quantify differences in flux samples between two conditions (Figure 2).
First, two flux vectors were chosen randomly,

  • one from each of the two samples to be compared and
  • the difference between the flux vectors was computed.

This approach was repeated to create a sample of 10,000 (n) flux difference vectors

  • for each pair of conditions considered (e.g. mutant or perturbed environment vs. wild type).

Based on this flux difference sample, the sample mean (μdiff,i) and standard deviation (σdiff,i)

  • between the two conditions was calculated for each reaction i. The reaction Z-score was calculated as:

 

reaction Z-score

reaction Z-score

which describes the sampled mean difference deviation

  • from a population mean change of zero (i.e. no flux difference
    between perturbation and wild type).

Note that this approach allows accounting for uncertainty in the

  • flux distributions inferred based on the extracellular metabolite secretion constraints.

This is in contrast to approaches such as FBA or MoMA that would predict

  • a single flux distribution for each condition and thus potentially
  • overestimate differences between conditions.

The reaction Z-scores can then be further used in analysis

  • to identify significantly perturbed regions of the metabolic network
  • based on reporter metabolite [44] or subsystem [30] Z-scores.

These reporter regions indicate, or “report”, dominant perturbation features

  • at the metabolite and pathway levels for a particular condition.

The reporter metabolite Z-score for any metabolite can be derived from the reaction Z-scores

  • of the reactions consuming or producing j (set of reactions denoted as Rj) as:

 

reporter z-score for any metabolite j

reporter z-score for any metabolite j

where Nis the number of reactions in Rand mmet,is calculated as

 

distributional correction for m_met,j SQRT

distributional correction for m_met,j SQRT

To account and correct for background distribution, the metabolite Z-score was normalized

  • by computing μmet,Nj and σmet,,Nj corresponding to the mean mmet and
  • its standard deviation for 1,000 randomly generated reaction sets of size Nj.

Z-scores for subsystems were calculated similarly by considering the set of reactions R

  • that belongs to each subsystem k.

Hence, positive metabolite and subsystem scores indicate a significantly perturbed metabolic region

  • relative to other regions, whereas
  • a negative score indicate regions that are not perturbed
  • more significantly than what is expected by random chance.

Perturbation subnetworks of reactions and connecting metabolites were visualized using Cytoscape [45].

Results and discussion

  1. Reconstruction and validation of iMM904 network iMM904 network content 

A previously reconstructed S. cerevisiae network, iND750,

  • was used as the basis for the construction of the expanded iMM904 network.
  • Prior to its presentation here, the
    iMM904 network content was the basis for a consensus jamboree network that was recently published
  • but has not yet been adapted for FBA calculations [46].

The majority of iND750 content was carried over and

  • further expanded on to construct iMM904, which accounts for
  1. 904 genes,
  2. 1,228 individual metabolites, and
  3. 1,412 reactions of which
  •                       395 are transport reactions.

Both the number of gene-associated reactions and the number of metabolites

  • increased in iMM904 compared with the iND750 network.

Additional genes and reactions included in the network primarily expanded the

  • lipid,
  • transport, and
  • carbohydrate subsystems.

The lipid subsystem includes

  • new genes and
  • reactions involving the degradation of sphingolipids and glycerolipids.

Sterol metabolism was also expanded to include

  • the formation and degradation of steryl esters, the
  •                      storage form of sterols.

The majority of the new transport reactions were added

  • to connect network gaps between intracellular compartments
  • to enable the completion of known physiological functions.

We also added a number of new secretion pathways

  • based on experimentally observed secreted metabolites [31].

A number of gene-protein-reaction (GPR) relationships were modified

  • to include additional gene products that are required to catalyze a reaction.

For example, the protein compounds

  • thioredoxin and
  • ferricytochrome C

were explicitly represented as compounds in iND750 reactions, but

  • the genes encoding these proteins were not associated with their corresponding GPRs.

Other examples include glycogenin and NADPH cytochrome p450 reductases (CPRs),

  1. which are required in the assembly of glycogen and
  2. to sustain catalytic activity in cytochromes p450, respectively.

These additional proteins were included in iMM904 as

  • part of protein complexes to provide a more complete
  • representation of the genes and
  • their corresponding products necessary for a catalytic activity to occur.

Major modifications to existing reactions were in cofactor biosynthesis, namely in

  • quinone,
  • beta-alanine, and
  • riboflavin biosynthetic pathways.

Reactions from previous S. cerevisiae networks associated with

  • quinone,
  • beta-alanine, and
  • riboflavin biosynthetic pathways

were essentially inferred from known reaction mechanisms based on

  • reactions in previous network reconstructions of E. coli [2,47].

These pathways were manually reviewed

  • based on current literature and subsequently replaced by
  • reactions and metabolites specific to yeast.

Additional changes in other subsystems were also made, such as

  1. changes to the compartmental location of a gene and
  2. its corresponding reaction(s),
  3. changes in reaction reversibility and cofactor specificity, and
  4. the elucidation of particular transport mechanisms.

A comprehensive listing of iMM904 network contents as well as

  • a detailed list of changes between iND750 and iMM904 is included
    [see Additional file 1].

Predicting deletion growth phenotypes

The updated genome-scale iMM904 metabolic network was validated

  • by comparing in silico single-gene deletion predictions to
  • in vivo results from a previous study used
  • to analyze another S. cerevisiae metabolic model, iLL672 [3].

This network was constructed based on the iFF708 network [22],

  • which was also the starting point for
  • reconstructing the iND750 network [2].

The experimental data used to validate the iLL672 model consisted of

3,360 single-gene knockout strain phenotypes evaluated

  • under minimal media growth conditions with
  1. glucose,
  2. galactose,
  3. glycerol, and
  4. ethanol

as sole carbon sources. Growth phenotypes for the iMM904 network were predictedusing

  1. FBA [3234],
  2. MoMA [35], and
  3. linear MoMA methods

as described in Methods and subsequently compared to the experimental data (Table 1).

Each deleted gene growth prediction comparison was classified as

  1. true lethal,
  2. true viable,
  3. false lethal, or
  4. false viable.

The growth rate threshold for considering a prediction viable was chosen

  • for each condition and method separately
  • to optimize the tradeoff between true viable and false viable predictions
    (maximum Matthews correlation coefficient, see Methods).

Since iMM904 has 212 more genes than iLL672 with experimental data, we also present results

  • for the subset of iMM904 predictions with genes included in iLL672 (reduced iMM904 set).

When the same gene sets are compared, iMM904 improves gene lethality predictions under

  • glucose,
  • galactose, and
  • glycerol conditions

over iLL672 somewhat, but is less accurate

  • at predicting growth phenotypes under the ethanol condition.

It should be noted that the iLL672 predictions were obtained directly from [3]

  • thus the growth rate threshold was not optimized similarly to iMM904 predictions.

Overall, when viability cutoff is chosen

  • as indicated above for each method separately,
  • the three prediction methods perform similarly
  1. FBA,
  2. MOMA, and
  3. linear MOMA) .

While the full gene complement in iMM904 greatly increased

  • the number of true viable predictions,
  • the full model also made significantly more false viable predictions
  • compared with reduced iMM904 and iLL672 predictions.

However, it is important to note that 143 reactions involved in dead-end biosynthetic pathways were actually

  • removed from iFF708 to build the iLL672 reconstruction [3].

These dead-ends are considered “knowledge gaps” in pathways

  • that have not been fully characterized and, as a result,
  • lead to false viable predictions when determining gene essentiality
  • if the pathway is in fact required for growth under a certain condition [2,26].

As more of these pathways are elucidated and

  • included in the model to
  • fill in existing network gaps,
  • we can expect false viable prediction rates to consequently decrease.

Thus, while a larger network has a temporarily reduced capacity to accurately predict gene deletion phenotypes,

  • it captures a more complete picture of currently known metabolic functions and
  • provides a framework for network expansion as new pathways are elucidated [48].

 

Inferring intracellular perturbation states from metabolic profiles – Aerobic and anaerobic gdh1/GDH2 mutant behavior

The gdh1/GDH2 mutant strain was previously developed [49,50]

  • to lower NADPH consumption in ammonia assimilation, which would
  • favor the NADPH-dependent fermentation of xylose.

In this strain, the NADPH-dependent glutamate dehydrogenase, Gdh1, was

  • deleted and the NADH-dependent form of the enzyme, Gdh2,
  •                     was overexpressed.

The net effect is to allow efficient assimilation of ammonia

  • into glutamate using NADH instead of NADPH as a cofactor.

While growth characteristics remained unaffected,

  • relative quantities of secreted metabolites differed between the wild-type and mutant strain
  • under aerobic and anaerobic conditions.

We analyzed EM data for the gdh1/GDH2 and wild-type strains reported

  • in [31] under aerobic and anaerobic conditions separately using
  • both FBA optimization and
  • sampling-based approaches as described in Methods.

43 measured extracellular and intracellular metabolites from the original dataset [31],

  • primarily of central carbon and amino acid metabolism,
  • were explicitly represented in the iMM904 network [see Additional file 4].

Extracellular metabolite levels were used

  • to formulate secretion constraints and
  • differential intracellular metabolites were used
  • to compare and validate the intracellular flux predictions.

Perturbed reactions from the FBA results were

  • determined by calculating relative flux changes, and
  • reaction Z-scores were calculated from the sampling analysis
  • to quantify flux changes between the mutant and wild-type strains,
  • with Z reaction > 1.96 corresponding to a two-tailed p-value < 0.05 and
  • considered to be significantly perturbed [see Additional file 4].

Additional file 4. Gdh mutant aerobic and anaerobic analysis results. 

The data provided are the full results for the exometabolomic analysis of aerobic and anerobic gdh1/GDH2 mutant.

Format: XLS Size: 669KB Download file

This file can be viewed with: Microsoft Excel Viewer

To validate the predicted results, reaction flux changes from both FBA and sampling methods were compared to differential intracellular metabolite level data measured from the same study. Intracellular metabolites involved in highly perturbed reactions (i.e. reactants and products) predicted from FBA and sampling analyses were identified and
compared to metabolites that were experimentally identified as significantly changed (< 0.05) between mutant and wild-type. Statistical measures of recall, accuracy, and
precision were calculated and represent the predictive sensitivity, exactness, and reproducibility respectively. From the sampling analysis, a considerably larger number of
significantly perturbed reactions are predicted in the anaerobic case (505 reactions, or 70.7% of active reactions) than in aerobic (394 reactions, or 49.8% of active reactions). The top percentile of FBA flux changes equivalent to the percentage of significantly perturbed sampling reactions were compared to the intracellular data. Results from both analyses are summarized in Table 2. Sampling predictions were considerably higher in recall than FBA predictions for both conditions, with respective ranges of 0.83–1
compared to 0.48–0.96. Accuracy was also higher in sampling predictions; however, precision was slightly better in the FBA predictions as expected due to the smaller
number of predicted changes. Overall, the sampling predictions of perturbed intracellular metabolites are strongly consistent with the experimental data and significantly
outperforms that of FBA optimization predictions in accurately predicting differential metabolites involved in perturbed intracellular fluxes.

Table 2. Statistical comparison of the differential intracellular metabolite data set (< 0.05) with metabolites involved in perturbed reactions predicted by FBA optimization and sampling analyses for aerobic and anaerobic gdh1/GDH2 mutant.

 

Table 2 Statistical comparison of the differential intracellular metabolite data set (p < 0.05)
with metabolites involved in perturbed reactions predicted by FBA optimization and
sampling analyses for aerobic and anaerobic gdh1/GDH2 mutant.
                           Aerobic                         Anaerobic                             Overall
FBA Sampling FBA Sampling FBA
Recall 0.48 0.83 0.96 1 0.71 0.91
Accuracy 0.55 0.62 0.64 0.64 0.6 0.63
Precision 0.78 0.69 0.64 0.63 0.68 0.66
Overall statistics indicate combined results of both conditions.
Mo et al. BMC Systems Biology 2009 3:37   http://dx.doi.org:/10.1186/1752-0509-3-37


Figure 3.
 Perturbation reaction subnetwork of gdh1/GDH2 mutant under aerobic conditions.

The network illustrates a simplified subset of highly perturbedPerturbation subnetworks can be drawn to visualize predicted significantly perturbed intracellular reactions and illustrate their connection to the observed secreted metabolites in the aerobic and anaerobic gdh1/GDH2 mutants.

Perturbation reaction subnetwork of gdh1.GDH2 mutant under aerobic conditions.

Perturbation reaction subnetwork of gdh1.GDH2 mutant under aerobic conditions.

Figure 3 shows an example of a simplified aerobic perturbation subnetwork consisting primarily of proximal pathways connected directly to a subset of major secreted
metabolites

  • glutamate,
  • proline,
  • D-lactate, and
  • 2-hydroxybuturate.

Figure 4 displays anaerobic reactions with Z-scores of similar magnitude to the perturbed reactions in Figure 3. The same subset of metabolites is also present in the
larger anaerobic perturbation network and indicates that the NADPH/NADH balance perturbation induced by the gdh1/GDH2 manipulation has widespread effects
beyond just altering glutamate metabolism anaerobically.

Interestingly, it is clear that the majority of the secreted metabolite pathways involve connected perturbed reactions that broadly converge on glutamate.

Note that Figures 3 and 4 only show the subnetworks that consisted of two or more connected reactions  for a number of secreted metabolites no contiguous perturbed pathway could be identified by the sampling approach. This indicates that the secreted metabolite pattern alone is not sufficient to determine which specific
production and secretion pathways are used by the cell for these metabolites.

Reactions connected to aerobically-secreted metabolites predicted from the sampling analysis of the gdh1/GDH2 mutant strain.
The major secreted metabolites

  • glutamate,
  • proline,
  • D-lactate, and
  • 2-hydroxybuturate

were also detected in the anaerobic condition. Metabolite abbreviations are found in Additional file 1.

Figure 4.

Perturbation reaction subnetwork of gdh1/GDH2 mutant under anaerobic conditions.

Perturbation reaction subnetwork of gdh1.GDH2 mutant under anaerobic conditions

Perturbation reaction subnetwork of gdh1.GDH2 mutant under anaerobic conditions

Subnetwork illustrates the highly perturbed anaerobic reactions of similar Z-reaction magnitude to the reactions in Figure 3.

A significantly larger number of reactions indicates mutant metabolic effects are more widespread in the anaerobic environment.
The network shows that perturbed pathways converge on glutamate, the main site in which the gdh1/GDH2 modification was introduced, which
suggests that the direct genetic perturbation effects are amplified under this environment. Metabolite abbreviations are found in Additional file 1.

To further highlight metabolic regions that have been systemically affected by the gdh1/GDH2 modification, reporter metabolite and subsystem methods [30] were used to
summarize reaction scores around specific metabolites and in specific metabolic subsystems. The top ten significant scores for metabolites/subsystems associated with more
than three reactions are summarized in Tables 3 (aerobic) and 4 (anaerobic), with Z > 1.64 corresponding to < 0.05 for a one-tailed distribution. Full data for all reactions,
reporter metabolites, and reporter subsystems is included [see Additional file 4].

Table 3. List of the top ten significant reporter metabolite and subsystem scores for the gdh1/GDH2 vs. wild type comparison in aerobic conditions.

Table 3
List of the top ten significant reporter metabolite and subsystem scores for the gdh1/GDH2 vs. wild type comparison in aerobic conditions.
Reporter metabolite Z-score No of reactions*
L-proline [c] 2.71 4
Carbon dioxide [m] 2.51 15
Proton [m] 2.19 51
Glyceraldehyde 3-phosphate [c] 1.93 7
Ubiquinone-6 [m] 1.82 5
Ubiquinol-6 [m] 1.82 5
Ribulose-5-phosphate [c] 1.8 4
Uracil [c] 1.74 4
L-homoserine [c] 1.72 4
Alpha-ketoglutarate [m] 1.71 8
Reporter subsystem Z-score No of reactions
Citric Acid Cycle 4.58 7
Pentose Phosphate Pathway 3.29 12
Glycine and Serine Metabolism 2.69 17
Alanine and Aspartate Metabolism 2.65 6
Oxidative Phosphorylation 1.79 8
Thiamine Metabolism 1.54 8
Arginine and Proline Metabolism 1.44 20
Other Amino Acid Metabolism 1.28 5
Glycolysis/Gluconeogenesis 0.58 14
Anaplerotic reactions 0.19 9
*Number of reactions categorized in a subsystem or found to be neighboring each metabolite
Mo et al. BMC Systems Biology 2009 3:37   http://dx.doi.org:/10.1186/1752-0509-3-37

Table 4. List of top ten significant reporter metabolite and subsystem scores for the gdh1/GDH2 vs. wild type comparison in anaerobic conditions.

 

Table 4
List of top ten significant reporter metabolite and subsystem scores for the gdh1/GDH2 vs. wild type comparison in anaerobic conditions.
Reporter metabolite Z-score No of reactions
Glutamate [c] 4.52 35
Aspartate [c] 3.21 11
Alpha-ketoglutarate [c] 2.66 17
Glycine [c] 2.65 7
Pyruvate [m] 2.56 7
Ribulose-5-phosphate [c] 2.43 4
Threonine [c] 2.28 6
10-formyltetrahydrofolate [c] 2.27 5
Fumarate [c] 2.27 5
L-proline [c] 2.04 4
Reporter subsystem Z-score No of reactions
Valine, Leucine, and Isoleucine Metabolism 3.97 15
Tyrosine, Tryptophan, and Phenylalanine Metabolism 3.39 23
Pentose Phosphate Pathway 3.29 11
Purine and Pyrimidine Biosynthesis 3.08 40
Arginine and Proline Metabolism 2.96 19
Threonine and Lysine Metabolism 2.74 14
NAD Biosynthesis 2.66 7
Alanine and Aspartate Metabolism 2.65 6
Histidine Metabolism 2.24 10
Cysteine Metabolism 1.85 10
Mo et al. BMC Systems Biology 2009 3:37   http://dx.doi.org:/10.1186/1752-0509-3-37
Open Data

Perturbations under aerobic conditions largely consisted of pathways involved in mediating the NADH and NADPH balance. Among the highest scoring aerobic subsystems
are TCA cycle and pentose phosphate pathway – key pathways directly involved in the generation of NADH and NADPH. Reporter metabolites involved in these
subsystems –

  • glyceraldehyde-3-phosphate,
  • ribulose-5-phosphate, and
  • alpha-ketoglutarate – were also identified.

These results are consistent with flux and enzyme activity measurements

  • of the gdh1/GDH2 strain under aerobic conditions [32],
  1. which reported significant reduction in the pentose phosphate pathway flux
  2. with concomitant changes in other central metabolic pathways.

Levels of several TCA cycle intermediates (e.g. fumarate, succinate, malate) were also elevated

  • in the gdh1/GDH2 mutant according to the differential intracellular metabolite data.

Altered energy metabolism, as indicated by

  • reporter metabolites (i.e. ubiquinone- , ubiquinol, mitochondrial proton)
  • and subsystem (oxidative phosphorylation),

is certainly feasible as NADH is a primary reducing agent for ATP production.

Pentose phosphate pathway and NAD biosynthesis also appears

  • among the most perturbed anaerobic subsystems, further suggesting
  • perturbed cofactor balance as a common, dominant effect under both conditions.

Glutamate dehydrogenase is a critical enzyme of amino acid biosynthesis as it acts as

  • the entry point for ammonium assimilation via glutamate.

Consequently, metabolic subsystems involved in amino acid biosynthesis were broadly perturbed

  • as a result of the gdh1/GDH2 modification in both aerobic and anaerobic conditions.

For example, the proline biosynthesis pathway that uses glutamate as a precursor

  • was significantly perturbed in both conditions,
  • with significantly changed intracellular and extracellular levels.

There were differences, however, in that more amino acid related subsystems were

  • significantly affected in the anaerobic case (Table 4),
  • further highlighting that altered ammonium assimilation in the mutant
  • has a more widespread effect under anaerobic conditions.

This effect is especially pronounced for

  • threonine and nucleotide metabolism,
  • which were predicted to be significantly perturbed only in anaerobic conditions.

Intracellular threonine levels were amongst the most significantly reduced

  • relative to other differential intracellular metabolites in the anaerobically grown gdh1/GDH2 strain
    (see [31] and Additional file 4), and
  • the relationship between threonine and nucleotide biosynthesis is further supported

by threonine’s recently discovered role as a key precursor in yeast nucleotide biosynthesis [51].

Other key anaerobic reporter metabolites are

  • glycine and 10-formyltetrahydrofolate,
  • both of which are involved in the cytosolic folate cycle (one-carbon metabolism).

Folate is intimately linked to biosynthetic pathways of

  • glycine (with threonine as its precursor) and purines
  • by mediating one-carbon reaction transfers necessary in their metabolism and
  • is a key cofactor in cellular growth [52].

Thus, the anaerobic perturbations identified in the analysis emphasize the close relationship

  • between threonine, folate, and nucleotide metabolic pathways as well as
  • their potential connection to perturbed ammonium assimilation processes.

Interestingly, this association has been previously demonstrated at the transcriptional level

  • as yeast ammonium assimilation (via glutamine synthesis) was found to be
  • co-regulated with genes involved in glycine, folate, and purine synthesis [53].

In summary, the overall differences in predicted gdh1/GDH2 mutant behavior

  • under aerobic and anaerobic conditions show that changes in flux states
  • directly related to modified ammonium assimilation pathway
  1. are amplified anaerobically whereas the
  2. indirect effects through NADH/NADPH balance are more significant aerobically.

Perturbed metabolic regions under aerobic conditions were predominantly

  • in central metabolic pathways involved in responding to the changed NADH/NADPH demand
  • and did not necessarily emphasize that glutamate dehydrogenase was the site of the genetic modification.

The majority of affected anaerobic pathways were involved directly

  • in modified ammonium assimilation as evidenced by

1) significantly perturbed amino acid subsystems,

2) a broad perturbation subnetwork converging on glutamate (Figure 4), and

3) glutamate as the most significant reporter metabolite (Table 4).

Potassium-limited and excess ammonium environments

A recent study reported that potassium limitation resulted in significant

  • growth retardation effect in yeast due to excess ammonium uptake
  • when ammonium was provided as the sole nitrogen source [33].

The proposed mechanism for this effect was that ammonium

  • could to be freely transported through potassium channels
  • when potassium concentrations were low in the media environment, thereby
  • resulting in excess ammonium uptake [33].

As a result, yeast incurred a significant metabolic cost

  • in assimilating ammonia to glutamate and
  • secreting significant amounts of glutamate and other amino acids
  • in potassium-limited conditions as a means to detoxify the excess ammonium.

A similar effect was observed when yeast was grown

  • with no potassium limitation,
  • but with excess ammonia in the environment.

While the observed effect of both environments (low potassium or excess ammonia) was similar,

  • quantitatively unique amino acid secretion profiles suggested that
  • internal metabolic states in these conditions are potentially different.

In order to elucidate the differences in internal metabolic states, we utilized

  • the iMM904 model and the EM profile analysis method to analyze amino acid secretion profiles
  • for a range of low potassium and high ammonia conditions reported in [33].

As before, we utilized amino acid secretion patterns as constraints to the iMM904 model,

  1. sampled the allowable solution space,
  2. computed reaction Z-scores for changes from a reference condition (normal potassium and ammonia), and
  3. finally summarized the resulting changes using reporter metabolites.

Figure 5 shows a clustering of the most significant reporter metabolites (Z ≥ 1.96 in any of the four conditions studied)

  • obtained from this analysis across the four conditions studied.

Interestingly, the potassium-limited environment perturbed only a subset of

  • the significant reporter metabolites identified in the high ammonia environments.

Both low potassium environments shared a consistent pattern of

  • highly perturbed amino acids and related precursor biosynthesis metabolites
    (e.g. pyruvate, PRPP, alpha-ketoglutarate)
  • with high ammonium environments.

The amino acid perturbation pattern (indicated by red labels in Figure 5) was present in

  • the ammonium-toxic environments, although the pattern was
  • slightly weaker for the lower ammonium concentration.

Nevertheless, the results clearly indicate that a similar

  • ammonium detoxifying mechanism that primarily perturbs pathways
  • directly related to amino acid metabolism
  • exists under both types of media conditions.

Figure 5.

Clustergram of top reporter metabolites - y in ammonium-toxic and potassium-limited conditions

Clustergram of top reporter metabolites – y in ammonium-toxic and potassium-limited conditions

Clustergram of top reporter metabolites (i.e. in yellow) in ammonium-toxic and potassium-limited conditions.

Amino acid perturbation patterns (shown in red) were shown to be consistently scored across conditions, indicating that potassium-limited environments K1 (lowest
concentration) and K2 (low concentration) elicited a similar ammonium detoxification response as ammonium-toxic environments N1 (high concentration) and N2
(highest concentration). Metabolites associated with folate metabolism (highlighted in green) are also highly perturbed in ammonium-toxic conditions. Metabolite
abbreviations are found in Additional file 1.

In addition to perturbed amino acids, a secondary effect notably appears at high ammonia levels in which metabolic regions related to folate metabolism are significantly affected. As highlighted in green in Figure 3, we predicted significantly perturbed key metabolites involved in the cytosolic folate cycle. These include tetrahydrofolate derivatives and other metabolites connected to the folate pathway, namely glycine and the methionine-derived methylation cofactors S-adenosylmethionine and S-adenosyl-homocysteine. Additionally, threonine was identified to be a key perturbed metabolite in excess ammonium conditions. These results further illustrate the close
connection between threonine biosynthesis, folate metabolism involving glycine derived from its threonine precursor, and nucleotide biosynthesis [51] that was discussed in
conjunction with the gdh1/GDH2 strain data. Taken together with the anaerobic gdh1/GDH2 data, the results consistently suggest highly perturbed threonine and folate
metabolism when amino acid-related pathways are broadly affected.

In both ammonium-toxic and potassium-limited environments, impaired cellular growth was observed, which can be attributed to high energetic costs of increased
ammonium assimilation to synthesize and excrete amino acids. However, under high ammonium environments, reporter metabolites related to threonine and folate
metabolism indicated that their perturbation, and thus purine supply, may be an additional factor in decreasing cellular viability as there is a direct relationship between
intracellular folate levels and growth rate [54]. Based on these results, we concluded that while potassiumlimited growth in yeast indeed shares physiological features with
growth in ammonium excess, its effects are not as detrimental as actual ammonium excess. The effects on proximal amino acid metabolic pathways are similar in both
environments as indicated by the secretion of the majority of amino acids. However, when our method was applied to analyze the physiological basis behind differences in
secretion profiles between low potassium and high ammonium conditions, ammonium excess was predicted to likely disrupt physiological ammonium assimilation processes,
which in turn potentially impacts folate metabolism and associated cellular growth.

Conclusion

The method presented in this study presents an approach to connecting intracellular flux states to metabolites that are excreted under various physiological conditions. We
showed that well-curated genome-scale metabolic networks can be used to integrate and analyze quantitative EM data by systematically identifying altered intracellular
pathways related to measured changes in the extracellular metabolome. We were able to identify statistically significant metabolic regions that were altered as a result of
genetic (gdh1/GD2 mutant) and environmental (excess ammonium and limited potassium) perturbations, and the predicted intracellular metabolic changes were consistent
with previously published experimental data including measurements of intracellular metabolite levels and metabolic fluxes. Our reanalysis of previously published EM data
on ammonium assimilation-related genetic and environmental perturbations also resulted in testable hypotheses about the role of threonine and folate pathways in mediating
broad responses to changes in ammonium utilization. These studies also demonstrated that the samplingbased method can be readily applied when only partial secreted
metabolite profiles (e.g. only amino acids) are available.

With the emergence of metabolite biofluid biomarkers as a diagnostic tool in human disease [55,56] and the availability of genome-scale human metabolic networks [1],
extensions of the present method would allow identifying potential pathway changes linked to these biomarkers. Employing such a method for studying yeast metabolism was possible as the metabolomic data was measured under controllable environmental conditions where the inputs and outputs of the system were defined. Measured metabolite biomarkers in a clinical setting, however, is far from a controlled environment with significant variations in genetic, nutritional, and environmental factors between different
patients. While there are certainly limitations for clinical applications, the method introduced here is a progressive step towards applying genome-scale metabolic networks
towards analyzing biofluid metabolome data as it 1) avoids the need to only study optimal metabolic states based on a predetermined objective function, 2) allows dealing with noisy experimental data through the sampling approach, and 3) enables analysis even with limited identification of metabolites in the data. The ability to establish potential
connections between extracellular markers and intracellular pathways would be valuable in delineating the genetic and environmental factors associated with a particular
disease.

Authors’ contributions

Conceived and designed the experiments: MLM MJH BOP. Performed experiments: MLM MJH. Analyzed the data: MLM MJH. Wrote the paper: MLM MJH BOP. All authors have read and approved the final manuscript.

Acknowledgements

We thank Jens Nielsen for providing the raw metabolome data for the mutant strain, and Jan Schellenberger and Ines Thiele for valuable discussions. This work was supported by NIH grant R01 GM071808. BOP serves on the scientific advisory board of Genomatica Inc.

 

References

  1. Duarte NC, Becker SA, Jamshidi N, Thiele I, Mo ML, Vo TD, Srivas R, Palsson BO: Global reconstruction of the human metabolic network based on genomic and bibliomic data. 

Proc Natl Acad Sci USA 2007, 104(6):1777-1782. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Duarte NC, Herrgard MJ, Palsson B: Reconstruction and Validation of Saccharomyces cerevisiae iND750, a Fully Compartmentalized Genome-Scale Metabolic Model. 

Genome Res 2004, 14(7):1298-1309. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Kuepfer L, Sauer U, Blank LM: Metabolic functions of duplicate genes in Saccharomyces cerevisiae. 

Genome Res 2005, 15(10):1421-1430. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Nookaew I, Jewett MC, Meechai A, Thammarongtham C, Laoteng K, Cheevadhanarak S, Nielsen J, Bhumiratana S: The genome-scale metabolic model iIN800 of Saccharomyces cerevisiae and its validation: a scaffold to query lipid metabolism. 

BMC Syst Biol 2008, 2:71. PubMed Abstract | BioMed Central Full Text | PubMed Central Full Text

  1. Edwards JS, Palsson BO: Systems properties of the Haemophilus influenzae Rd metabolic genotype. 

J biol chem 1999, 274(25):17410-17416. PubMed Abstract | Publisher Full Text

  1. Edwards JS, Palsson BO: The Escherichia coli MG1655 in silico metabolic genotype: Its definition, characteristics, and capabilities. 

Proc Natl Acad Sci USA 2000, 97(10):5528-5533. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Thiele I, Vo TD, Price ND, Palsson B: An Expanded Metabolic Reconstruction of Helicobacter pylori (IT341 GSM/GPR): An in silico genome-scale characterization of single and double deletion mutants. 

J Bacteriol 2005, 187(16):5818-5830. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Vo TD, Greenberg HJ, Palsson BO: Reconstruction and functional characterization of the human mitochondrial metabolic network based on proteomic and biochemical data. 

J Biol Chem 2004, 279(38):39532-39540. PubMed Abstract | Publisher Full Text

  1. Reed JL, Vo TD, Schilling CH, Palsson BO: An expanded genome-scale model of Escherichia coli K-12 (iJR904 GSM/GPR). 

Genome Biology 2003, 4(9):R54.51-R54.12. BioMed Central Full Text

  1. Feist AM, Henry CS, Reed JL, Krummenacker M, Joyce AR, Karp PD, V H, Palsson BO: A genome-scale metabolic reconstruction for Escherichia coli K-12 MG1655 that accounts for 1261 ORFs and thermodynamic information. 

Molecular Systems Biology 2007, 3:121. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Suthers PF, Dasika MS, Kumar VS, Denisov G, Glass JI, Maranas CD: A genome-scale metabolic reconstruction of Mycoplasma genitalium, iPS189. 

PLos Comp Biol 2009, 5(2):e1000285. Publisher Full Text

  1. Price ND, Reed JL, Palsson BO: Genome-scale models of microbial cells: evaluating the consequences of constraints. 

Nat Rev Microbiol 2004, 2(11):886-897. PubMed Abstract | Publisher Full Text

  1. Becker SA, Feist AM, Mo ML, Hannum G, Palsson BO, Herrgard MJ: Quantitative Prediction of Cellular Metabolism with Constraint-based Models: The COBRA Toolbox. 

Nature protocols 2007, 2(3):727-738. PubMed Abstract | Publisher Full Text

  1. Reed JL, Palsson BO: Genome-Scale In Silico Models of E. coli Have Multiple Equivalent Phenotypic States: Assessment of Correlated Reaction Subsets That Comprise Network States. 

Genome Res 2004, 14(9):1797-1805. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Fong SS, Palsson BO: Metabolic gene deletion strains of Escherichia coli evolve to computationally predicted growth phenotypes. 

Nature Genetics 2004, 36(10):1056-1058. PubMed Abstract | Publisher Full Text

  1. Ibarra RU, Edwards JS, Palsson BO: Escherichia coli K-12 undergoes adaptive evolution to achieve in silico predicted optimal growth. 

Nature 2002, 420(6912):186-189. PubMed Abstract | Publisher Full Text

  1. Schellenberger J, Palsson BØ: Use of randomized sampling for analysis of metabolic networks. 

J Biol Chem 2009, 284(9):5457-5461. PubMed Abstract | Publisher Full Text

  1. Almaas E, Kovács B, Vicsek T, Oltvai ZN, Barabási AL: Global organization of metabolic fluxes in the bacterium Escherichia coli. 

Nature 2004, 427(6977):839-843. PubMed Abstract | Publisher Full Text

  1. Thiele I, Price ND, Vo TD, Palsson BO: Candidate metabolic network states in human mitochondria: Impact of diabetes, ischemia, and diet. 

J Biol Chem 2005, 280(12):11683-11695. PubMed Abstract | Publisher Full Text

  1. Kell DB: Metabolomics and systems biology: making sense of the soup. 

Curr Opin Microbiol 2004, 7(3):296-307. PubMed Abstract | Publisher Full Text

  1. Kell DB, Brown M, Davey HM, Dunn WB, Spasic I, Oliver SG: Metabolic footprinting and systems biology: the medium is the message. 

Nat Rev Microbiol 2005, 3(7):557-565. PubMed Abstract | Publisher Full Text

  1. Goodacre R, Vaidyanathan S, Dunn WB, Harrigan GG, Kell DB: Metabolomics by numbers: acquiring and understanding global metabolite data. 

Trends Biotechnol 2004, 22(5):245-252. PubMed Abstract | Publisher Full Text

  1. Lenz EM, Bright J, Wilson ID, Morgan SR, Nash AF: A 1H NMR-based metabonomic study of urine and plasma samples obtained from healthy human subjects. 

J Pharm Biomed Anal 2003, 33(5):1103-1115. PubMed Abstract | Publisher Full Text

  1. Allen J, Davey HM, Broadhurst D, Heald JK, Rowland JJ, Oliver SG, Kell DB: High-throughput classification of yeast mutants for functional genomics using metabolic footprinting. 

Nat Biotech 2003, 21(6):692-696. Publisher Full Text

  1. Nicholson JK, Connelly J, Lindon JC, Holmes E: Metabonomics: a platform for studying drug toxicity and gene function. 

Nat Rev Drug Discov 2002, 1(2):153-161. PubMed Abstract | Publisher Full Text

  1. Mortishire-Smith RJ, Skiles GL, Lawrence JW, Spence S, Nicholls AW, Johnson BA, Nicholson JK: Use of metabonomics to identify impaired fatty acid metabolism as the mechanism of a drug-induced toxicity. 

Chem Res Toxicol 2004, 17(2):165-173. PubMed Abstract | Publisher Full Text

  1. Sabatine MS, Liu E, Morrow DA, Heller E, McCarroll R, Wiegand R, Berriz GF, Roth FP, Gerszten RE: Metabolomic identification of novel biomarkers of myocardial ischemia. 

Circulation 2005, 112(25):3868-3875. PubMed Abstract | Publisher Full Text

  1. Cakir T, Efe C, Dikicioglu D, Hortaçsu AKB, Oliver SG: Flux balance analysis of a genome-scale yeast model constrained by exometabolomic data allows metabolic system identification of genetically different strains. 

Biotechnol Prog 2007, 23(2):320-326. PubMed Abstract | Publisher Full Text

  1. Bang JW, Crockford DJ, Holmes E, Pazos F, Sternberg MJ, Muggleton SH, Nicholson JK: Integrative top-down system metabolic modeling in experimental disease states via data-driven Bayesian methods. 

J Proteome Res 2008, 7(2):497-503. PubMed Abstract | Publisher Full Text

  1. Oliveira AP, Patil KR, Nielsen J: Architecture of transcriptional regulatory circuits is knitted over the topology of bio-molecular interaction networks. 

BMC Syst Biol 2008, 2:17. PubMed Abstract | BioMed Central Full Text | PubMed Central Full Text

  1. Villas-Boas SG, Moxley JF, Akesson M, Stephanopoulos G, Nielsen J: High-throughput metabolic state analysis: the missing link in integrated functional genomics of yeasts. 

Biochem J 2005, 388(Pt 2):669-677. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Moreira dos Santos M, Thygesen G, Kötter P, Olsson L, Nielsen J: Aerobic physiology of redox-engineered Saccharomyces cerevisiae strains modified in the ammonium assimilation for increased NADPH availability. 

FEMS Yeast Res 2003, 4(1):59-68. PubMed Abstract | Publisher Full Text

  1. Hess DC, Lu W, Rabinowitz JD, Botstein D: Ammonium toxicity and potassium limitation in yeast. 

PLoS Biol 2006, 4(11):e351. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Nissen TL, Schulze U, Nielsen J, Villadsen J: Flux distributions in anaerobic, glucose-limited continuous cultures of Saccharomyces cerevisiae. 

Microbiology 1997, 143(Pt 1):203-218. PubMed Abstract | Publisher Full Text

  1. Famili I, Forster J, Nielsen J, Palsson BO: Saccharomyces cerevisiae phenotypes can be predicted by using constraint-based analysis of a genome-scale reconstructed metabolic network. 

Proc Natl Acad Sci USA 2003, 100(23):13134-13139. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Bonarius HPJ, Schmid G, Tramper J: Flux analysis of underdetermined metabolic networks: The quest for the missing constraints. 

Trends in Biotechnology 1997, 15(8):308-314. Publisher Full Text

  1. Edwards JS, Palsson BO: Metabolic flux balance analysis and the in silico analysis of Escherichia coli K-12 gene deletions. 

BMC Bioinformatics 2000, 1:1. PubMed Abstract | BioMed Central Full Text | PubMed Central Full Text

  1. Varma A, Palsson BO: Metabolic Flux Balancing: Basic concepts, Scientific and Practical Use. 

Nat Biotechnol 1994, 12:994-998. Publisher Full Text

  1. Segre D, Vitkup D, Church GM: Analysis of optimality in natural and perturbed metabolic networks. 

Proc Natl Acad Sci USA 2002, 99(23):15112-15117. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Baldi P, Brunak S, Chauvin Y, Andersen CA, Nielsen H: Assessing the accuracy of prediction algorithms for classification: an overview. 

Bioinformatics 2000, 16(5):412-424. PubMed Abstract | Publisher Full Text

  1. Price ND, Schellenberger J, Palsson BO: Uniform Sampling of Steady State Flux Spaces: Means to Design Experiments and to Interpret Enzymopathies. 

Biophysical Journal 2004, 87(4):2172-2186. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Price ND, Thiele I, Palsson BO: Candidate states of Helicobacter pylori’s genome-scale metabolic network upon application of “loop law” thermodynamic constraints. 

Biophysical J 2006, 90(11):3919-3928. Publisher Full Text

  1. Schuetz R, Kuepfer L, Sauer U: Systematic evaluation of objective functions for predicting intracellular fluxes in Escherichia coli. 

Mol Syst Biol 2007, 3:119. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Patil KR, Nielsen J: Uncovering transcriptional regulation of metabolism by using metabolic network topology. 

Proc Natl Acad Sci USA 2005, 102(8):2685-2689. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T: Cytoscape: a software environment for integrated models of biomolecular interaction networks. 

Genome Res 2003, 13(11):2498-2504. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Herrgard MJ, Swainston N, Dobson P, Dunn WB, Arga KY, Arvas M, Bluthgen N, Borger S, Costenoble R, Heinemann M, et al.: A consensus yeast metabolic network reconstruction obtained from a community approach to systems biology. 

Nat Biotech 2008, 26:1155-1160. Publisher Full Text

  1. Forster J, Famili I, Fu PC, Palsson BO, Nielsen J: Genome-Scale Reconstruction of the Saccharomyces cerevisiae Metabolic Network. 

Genome Research 2003, 13(2):244-253. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Reed JL, Patel TR, Chen KH, Joyce AR, Applebee MK, Herring CD, Bui OT, Knight EM, Fong SS, Palsson BO: Systems Approach to Genome Annotation: Prediction and Validation of Metabolic Functions. 

Proc Natl Acad Sci USA 2006, 103(46):17480-17484. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Nissen TL, Kielland-Brandt MC, Nielsen J, Villadsen J: Optimization of ethanol production in Saccharomyces cerevisiae by metabolic engineering of the ammonium assimilation. 

Metab Eng 2000, 2(1):69-77. PubMed Abstract | Publisher Full Text

  1. Roca C, Nielsen J, Olsson L: Metabolic engineering of ammonium assimilation in xylose-fermenting Saccharomyces cerevisiae improves ethanol production. 

Appl Environ Microbiol 2003, 69(8):4732-4736. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Hartman JL IV: Buffering of deoxyribonucleotide pool homeostasis by threonine metabolism. 

Proc Natl Acad Sci USA 2007, 104(28):11700-11705. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Gelling CL, Piper MD, Hong SP, Kornfeld GD, Dawes IW: Identification of a novel one-carbon metabolism regulon in Saccharomyces cerevisiae. 

J Biol Chem 2004, 279(8):7072-7081. PubMed Abstract | Publisher Full Text

  1. Denis V, Daignan-Fornier B: Synthesis of glutamine, glycine and 10-formyl tetrahydrofolate is coregulated with purine biosynthesis in Saccharomyces cerevisiae. 

Mol Gen Genet 1998, 259(3):246-255. PubMed Abstract | Publisher Full Text

  1. Hjortmo S, Patring J, Andlid T: Growth rate and medium composition strongly affect folate content in Saccharomyces cerevisiae. 

Int J Food Microbiol 2008, 123(1–2):93-100. PubMed Abstract | Publisher Full Text

  1. Kussmann MRF, Affolter M: OMICS-driven biomarker discovery in nutrition and health. 

J Biotechnol 2006, 124(4):758-787. PubMed Abstract | Publisher Full Text

  1. Serkova NJ, Niemann CU: Pattern recognition and biomarker validation using quantitative 1H-NMR-based metabolomics. 

Expert Rev Mol Diagn 2006, 6(5):717-731. PubMed Abstract | Publisher Full Text

 

Read Full Post »

Curation, HealthCare System in the US, and Calcium Signaling Effects on Cardiac Contraction, Heart Failure, and Atrial Fibrillation, and the Relationship of Calcium Release at the Myoneural Junction to Beta Adrenergic Release

Curation, HealthCare System in the US, and Calcium Signaling Effects on Cardiac Contraction, Heart Failure, and Atrial Fibrillation, and the Relationship of Calcium Release at the Myoneural Junction to Beta Adrenergic Release

Curator and e-book Contributor: Larry H. Bernstein, MD, FCAP
Curator and BioMedicine e-Series Editor-in-Chief: Aviva Lev Ari, PhD, RN

and 

Content Consultant to Six-Volume e-SERIES A: Cardiovascular Diseases: Justin Pearlman, MD, PhD, FACC

This portion summarises what we have covered and is now familiar to the reader.  There are three related topics, and an extension of this embraces other volumes and chapters before and after this reading.  This approach to the document has advantages over the multiple authored textbooks that are and have been pervasive as a result of the traditional publication technology.  It has been stated by the founder of ScoopIt, that amount of time involved is considerably less than required for the original publications used, but the organization and construction is a separate creative process.  In these curations we amassed on average five articles in one curation, to which, two or three curators contributed their views.  There were surprises, and there were unfulfilled answers along the way.  The greatest problem that is being envisioned is the building a vision that bridges and unmasks the hidden “dark matter” between the now declared “OMICS”, to get a more real perspective on what is conjecture and what is actionable.  This is in some respects unavoidable because the genome is an alphabet that is matched to the mino acid sequences of proteins, which themselves are three dimensional drivers of sequences of metabolic reactions that can be altered by the accumulation of substrates in critical placements, and in addition, the proteome has functional proteins whose activity is a regulatory function and not easily identified.  In the end, we have to have a practical conception, recognizing the breadth of evolutionary change, and make sense of what we have, while searching for more.

We introduced the content as follows:

1. We introduce the concept of curation in the digital context, and it’s application to medicine and related scientific discovery.

Topics were chosen were used to illustrate this process in the form of a pattern, which is mostly curation, but is significantly creative, as it emerges in the context of this e-book.

  • Alternative solutions in Treatment of Heart Failure (HF), medical devices, biomarkers and agent efficacy is handled all in one chapter.
  • PCI for valves vs Open heart Valve replacement
  • PDA and Complications of Surgery — only curation could create the picture of this unique combination of debate, as exemplified of Endarterectomy (CEA) vs Stenting the Carotid Artery (CAS), ischemic leg, renal artery stenosis.

2. The etiology, or causes, of cardiovascular diseases consist of mechanistic explanations for dysfunction relating to the heart or vascular system. Every one of a long list of abnormalities has a path that explains the deviation from normal. With the completion of the analysis of the human genome, in principle all of the genetic basis for function and dysfunction are delineated. While all genes are identified, and the genes code for all the gene products that constitute body functions, there remains more unknown than known.

3. Human genome, and in combination with improved imaging methods, genomics offers great promise in changing the course of disease and aging.

4. If we tie together Part 1 and Part 2, there is ample room for considering clinical outcomes based on individual and organizational factors for best performance. This can really only be realized with considerable improvement in information infrastructure, which has miles to go.

Curation

Curation is an active filtering of the web’s  and peer reviewed literature found by such means – immense amount of relevant and irrelevant content. As a result content may be disruptive. However, in doing good curation, one does more than simply assign value by presentation of creative work in any category. Great curators comment and share experience across content, authors and themes.
Great curators may see patterns others don’t, or may challenge or debate complex and apparently conflicting points of view.  Answers to specifically focused questions comes from the hard work of many in laboratory settings creatively establishing answers to definitive questions, each a part of the larger knowledge-base of reference. There are those rare “Einstein’s” who imagine a whole universe, unlike the three blindmen of the Sufi tale.  One held the tail, the other the trunk, the other the ear, and they all said this is an elephant!
In my reading, I learn that the optimal ratio of curation to creation may be as high as 90% curation to 10% creation. Creating content is expensive. Curation, by comparison, is much less expensive.  The same source says “Scoop.it is my content marketing testing “sandbox”. In sharing, he says that comments provide the framework for what and how content is shared.

Healthcare and Affordable Care Act

We enter year 2014 with the Affordable Care Act off to a slow start because of the implementation of the internet signup requiring a major repair, which is, unfortunately, as expected for such as complex job across the US, and with many states unwilling to participate.  But several states – California, Connecticut, and Kentucky – had very effective state designed signups, separate from the federal system.  There has been a very large rush and an extension to sign up. There are many features that we can take note of:

1. The healthcare system needed changes because we have the most costly system, are endowed with advanced technology, and we have inexcusable outcomes in several domains of care, including, infant mortality, and prenatal care – but not in cardiology.

2. These changes that are notable are:

  • The disparities in outcome are magnified by a large disparity in highest to lowest income bracket.
  • This is also reflected in educational status, and which plays out in childhood school lunches, and is also affected by larger class size and cutbacks in school programs.
  • This is not  helped by a large paralysis in the two party political system and the three legs of government unable to deal with work and distraction.
  • Unemployment is high, and the banking and home construction, home buying, and rental are in realignment, but interest rates are problematic.

3.  The  medical care system is affected by the issues above, but the complexity is not to be discounted.

  •  The medical schools are unable at this time to provide the influx of new physicians needed, so we depend on a major influx of physicians from other countries
  • The technology for laboratories, proteomic and genomic as well as applied medical research is rejuvenating the practice in cardiology more rapidly than any other field.
  • In fields that are imaging related the life cycle of instruments is shorter than the actual lifetime use of the instruments, which introduces a shortening of ROI.
  • Hospitals are consolidating into large consortia in order to maintain a more viable system for referral of specialty cases, and also is centralizing all terms of business related to billing.
  • There is reduction in independent physician practices that are being incorporated into the hospital enterprise with Part B billing under the Physician Organization – as in Partners in Greater Boston, with the exception of “concierge” medical practices.
  • There is consolidation of specialty laboratory services within state, with only the most specialized testing going out of state (Quest, LabCorp, etc.)
  • Medicaid is expanded substantially under the new ACA.
  • The federal government as provider of services is reducing the number of contractors for – medical devices, diabetes self-testing, etc.
  • The current rearrangements seeks to provide a balance between capital expenses and fixed labor costs that it can control, reduce variable costs (reagents, pharmaceutical), and to take in more patients with less delay and better performance – defined by outside agencies.

Cardiology, Genomics, and calcium ion signaling and ion-channels in cardiomyocyte function in health and disease – including heart failure, rhythm abnormalities, and the myoneural release of neurotransmitter at the vesicle junction.

This portion is outlined as follows:

2.1 Human Genome: Congenital Etiological Sources of Cardiovascular Disease

2.2 The Role of Calcium in Health and Disease

2.3 Vasculature and Myocardium: Diagnosing the Conditions of Disease

Genomics & Genetics of Cardiovascular Disease Diagnoses

actin cytoskeleton

wall stress, ventricular workload, contractile reserve

Genetic Base of Atherosclerosis and Loss of Arterial Elasticity with Aging

calcium and actin skeleton, signaling, cell motility

hypertension & vascular compliance

Genetics of Conduction Disease

Ca+ stimulated exostosis: calmodulin & PKC (neurotransmitter)

complications & MVR

disruption of Ca2+ homeostasis cardiac & vascular smooth muscle

synaptotagmin as Ca2+ sensor & vesicles

atherosclerosis & ion channels


It is increasingly clear that there are mutations that underlie many human diseases, and this is true of the cardiovascular system.  The mutations are mistakes in the insertion of a purine nucleotide, which may or may not have any consequence.  This is why the associations that are being discovered in research require careful validation, and even require demonstration in “models” before pursuing the design of pharmacological “target therapy”.  The genomics in cardiovascular disease involves very serious congenital disorders that are asserted early in life, but the effects of and development of atherosclerosis involving large and medium size arteries has a slow progression and is not dominated by genomic expression.  This is characterized by loss of arterial elasticity. In addition there is the development of heart failure, which involves the cardiomyocyte specifically.  The emergence of regenerative medical interventions, based on pleuripotent inducible stem cell therapy is developing rapidly as an intervention in this sector.

Finally, it is incumbent on me to call attention to the huge contribution that research on calcium (Ca2+) signaling has made toward the understanding of cardiac contraction and to the maintenance of the heart rhythm.  The heart is a syncytium, different than skeletal and smooth muscle, and the innervation is by the vagus nerve, which has terminal endings at vesicles which discharge at the myocyte junction.  The heart specifically has calmodulin kinase CaMK II, and it has been established that calmodulin is involved in the calcium spark that triggers contraction.  That is only part of the story.  Ion transport occurs into or out of the cell, the latter termed exostosis.  Exostosis involves CaMK II and pyruvate kinase (PKC), and they have independent roles.  This also involves K+-Na+-ATPase.  The cytoskeleton is also discussed, but the role of aquaporin in water transport appears elsewhere, as the transport of water between cells.  When we consider the Gibbs-Donnan equilibrium, which precedes the current work by a century, we recall that there is an essential balance between extracellular Na+ + Ca2+ and the intracellular K+ + Mg2+, and this has been superceded by an incompletely defined relationship between ions that are cytoplasmic and those that are mitochondrial.  The glass is half full!

 

Read Full Post »

Author and Curator: Ritu Saxena, Ph.D

Although cancer stem cells constitute only a small percentage of the tumor burden, their self-renewal capacity and possible link with recurrence of cancer post treatment makes them a sought after therapeutic target in cancer. The post on cancer stem cells published on the 22nd of March, 2013, describes the identity of CSCs, their functional characteristics, possible cell of origin and biomarkers. This post focuses on the therapeutic potential of CSCs, their resistance to conventional anti-tumor therapies and current therapeutic targets including biomarkers, signaling pathways and niches.

CSCs Are Resistant to conventional anticancer therapies including chemotherapy, radiotherapy and surgery that are used either alone or in combination. However, these strategies have failed several times to eradicate CSCs resulting in metastasis and relapse, hence, a fatal disease outcome.

The properties of CSCs that contribute to or lead to chemoresistance include:

Quiescent Phenotype

Chemotherapeutic agents target fast-growing cells; however, some CSCs that remain in the dormant or quiescent stage are spared from lethal damage. Later, when the dormant CSCs enter cell cycle, tumor proliferation is stimulated.

Antiapoptosis

Antiapoptotic proteins such as BCL-2 and some self-renewal pathways such as transforming growth factor β, Wnt/ β -catenin or BMI-1 are activated in CSCs. Consequently, DNA damage repair capability of CSCs is enhanced after genotoxic stress or activation of autocrine loops through the production of growth factors like epidermal growth factor (Moserle L, Cancer Lett, 1 Feb 2010;288(1):1-9).

Expression of Drug Efflux Pumps

CSCs express some proteins that have typically been known to contribute to multidrug resistance. The proteins are drug efflux pumps ABCC1, ABCG2 or MDR1. Multidrug resistance-associated proteins (ABCC subfamily) are members of the ATP-binding cassette (ABC) superfamily of transport proteins and act as cellular efflux transporters for a wide variety of substrates, in particular glutathione, glucuronide and sulfate conjugates of diverse compounds.

Radiotherapy is mainly used in breast cancer and glioblastoma multiforme. In glioblastoma multiforme, the properties of CSCs that contribute to radiotherapy resistance is the presence of CD133 marker. CD133+ CSCs preferentially activate DNA damage repair pathway and significantly induced checkpoint kinases that leads to reduced apoptosis in CSCs compared to the CD133- tumor cells (Bao S, Nature, 7 Dec 2006;444(7120):756-60).

Radiotherapy resistance in breast cancer is due to reduced levels of reactive oxygen species in CSCs. In addition, radiation resistance of progenitor cells in an immortalized breast cancer cell line was mediated by the Wnt/β catenin pathway proteins (Diehn M, et al, Nature, 9 Apr 2009;458(7239):780-3; Chen MS, et al, J Cell Sci, 1 Feb 2007;120(Pt 3):468-77).

As mentioned in the previous post on CSCs, CSC targeting therapy could either eliminate CSCs by either killing them after differentiating them from other tumor population, and/or by disrupting their niche. Efficient eradication of CSCs may require the combined ablation of CSCs themselves and their niches. Thus, identification of appropriate and specific markers of CSCs is crucial for targeting them and preventing tumor relapse. Table 1 (adapted from a review article on CSCs by Zhao et al) describes the currently used biomarkers for CSC-targeted therapy (Zhao L, et al, Eur Surg Res, 2012;49(1):8-15).

Table 1

Specific Target Cancer type Marker properties and therapy
Targeting cell markers
CD24+CD44+ESA+ Pancreatic cancer Pancreatic CSCs, elevated during tumorigenesis
CD44+CD24–ESA+ Breast cancer Breast CSCs
EpCAM high CD44+CD166+ Colorectal cancer
CD34+CD38– AML broad use as a target for chemotherapy
CD133+ Prostate cancer and breast cancer 5-transmembrane domain cell surface glycoprotein,also a marker for neuron epithelial, hematopoietic and endothelialprogenitor cells
Stro1+CD105+CD44+ Bone sarcoma
Nodal/activin Knockdown or pharmacological inhibition of its receptorAlk4/7 abrogated self-renewal capacity and in vivo tumorigenicity of CSCs.
Targeting signaling pathways
Hedgehog signaling Upregulated in several cancer types inhibitors: GDC-0449,PF04449913, BMS-833923, IPI-926 and TAK-441
Wnt/β-catenin signaling CML, squamous cell carcinoma Be required for CSC self-renewal and tumor growthinhibitors: PRI-724, WIF-1 and telomerase
Notch signaling Several cancer types An important regulator in normal development, adult stem cell maintenance,and tumorigenesis in multiple organs,inhibitors: RO4929097, BMS-906024, IPI-926 and MK0752
PI3K/Akt/PTEN/mTOR, Several cancer types The pathway is deregulated in many tumors and used to preferentially target CSCsinhibitors: temsirolimus, everolimus FDA-approved therapy for renal cell carcinoma
Targeting CSC Niche
Angiogenesis Niche Colon cancer, breast cancer, NSCLC Inhibitor: bevacizumab results in a disruption of the CSC niche, depleted vasculature and a dramatic reduction in the number of CSCs.
Hypoxia (HIF pathway) Ovarian cancer, lung cancer, cervical cancer Inhibitors: topotecan and digoxin have been approved for ovarian, lung and cervical cancer
Targeting Micro RNA
miR-200 family Inhibits EMT and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2
Let-7 family Regulates BT-IC stem cell-like properties by silencing more than one target
miR-124 Related to neuronal differentiation, targets laminin γ1 and integrin β1.
miR-21 Suppresses the self-renewal of embryonic stem cells

The challenge is to develop an effective treatment regimen that prevents survival, self-renewal and differentiation of CSCs and also disturbs their niche without damaging normal stem cells. In order to evaluate the efficiency of CSC-targeting therapies, in vitro models and mouse xenotransplantation models have been used for preclinical studies. Some potential CSC targeting agents in preclinical stages include notch inhibitors for glioblastoma stem cells and telomerase peptide vaccination after chemoradiotherapy of non-small cell lung cancer stem cells Stem Cells (Hovinga KE, et al, Jun 2010;28(6):1019-29; Serrano D, Mol Cancer, 9 Aug 2011;10:96). In addition, several phase II and phase III trials are currently underway to test CSC-targeting drugs focusing on efficacy and safety of treatment.

Reference:

Bao S, Nature, 7 Dec 2006;444(7120):756-60).

Diehn M, et al, Nature, 9 Apr 2009;458(7239):780-3

Chen MS, et al, J Cell Sci, 1 Feb 2007;120(Pt 3):468-77

Zhao L, et al, Eur Surg Res, 2012;49(1):8-15

Hovinga KE, et al, Jun 2010;28(6):1019-29

Serrano D, Mol Cancer, 9 Aug 2011;10:96

Pharmaceutical Intelligence posts:

http://pharmaceuticalintelligence.com/2013/03/22/in-focus-identity-of-cancer-stem-cells/ Author and curator: Ritu Saxena, PhD

http://pharmaceuticalintelligence.com/2012/08/15/to-die-or-not-to-die-time-and-order-of-combination-drugs-for-triple-negative-breast-cancer-cells-a-systems-level-analysis/ Authors: Anamika Sarkar, PhD and Ritu Saxena, PhD

http://pharmaceuticalintelligence.com/2013/03/07/the-importance-of-cancer-prevention-programs-new-perceptions-for-fighting-cancer/ Author: Ziv Raviv, PhD

http://pharmaceuticalintelligence.com/2013/03/03/treatment-for-metastatic-her2-breast-cancer/ Reporter: Larry H Bernstein, MD

http://pharmaceuticalintelligence.com/2013/03/02/recurrence-risk-for-breast-cancer/ Larry H Bernstein, MD

http://pharmaceuticalintelligence.com/2013/02/14/prostate-cancer-androgen-driven-pathomechanism-in-early-onset-forms-of-the-disease/ Curator: Aviva Lev-Ari, PhD, RN

http://pharmaceuticalintelligence.com/2013/01/15/exploring-the-role-of-vitamin-c-in-cancer-therapy/ Curator: Ritu Saxena, PhD

http://pharmaceuticalintelligence.com/2013/01/12/harnessing-personalized-medicine-for-cancer-management-prospects-of-prevention-and-cure-opinions-of-cancer-scientific-leaders-httppharmaceuticalintelligence-com/ Curator: Aviva Lev-Ari, PhD, RN

http://pharmaceuticalintelligence.com/2013/01/10/the-molecular-pathology-of-breast-cancer-progression/ Author and reporter: Tilda Barliya PhD

http://pharmaceuticalintelligence.com/2012/11/30/histone-deacetylase-inhibitors-induce-epithelial-to-mesenchymal-transition-in-prostate-cancer-cells/ Reporter and Curator: Stephen J. Williams, PhD

http://pharmaceuticalintelligence.com/2012/10/22/blood-vessel-generating-stem-cells-discovered/ Reporter: Ritu Saxena, PhD

http://pharmaceuticalintelligence.com/2012/10/17/stomach-cancer-subtypes-methylation-based-identified-by-singapore-led-team/ Reporter: Aviva Lev-Ari, PhD, RN

http://pharmaceuticalintelligence.com/2012/09/17/natural-agents-for-prostate-cancer-bone-metastasis-treatment/ Reporter: Ritu Saxena, PhD

http://pharmaceuticalintelligence.com/2012/08/28/cardiovascular-outcomes-function-of-circulating-endothelial-progenitor-cells-cepcs-exploring-pharmaco-therapy-targeted-at-endogenous-augmentation-of-cepcs/ Aviva Lev-Ari, PhD, RN

 

Read Full Post »