You are on page 1of 9

Toxicology 313 (2013) 113–121

Contents lists available at ScienceDirect

Toxicology
journal homepage: www.elsevier.com/locate/toxicol

In vitro approach to predict post-translational phosphorylation response to


mixtures
Jonathan Boyd ∗ , Julie A. Vrana, Holly N. Williams
C Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV 26506, United States

a r t i c l e i n f o a b s t r a c t

Article history: While exposure to chemical mixtures is an everyday reality, an understanding of their combined effects,
Received 18 April 2012 and any potential prediction thereof, is extremely limited. Realistic exposures potentially consist of
Received in revised form hundreds to thousands of chemicals per day, but even relatively simple binary mixture interactions
19 September 2012
can be inherently difficult to predict based upon the lack of temporal and spatial mechanisms for the
Accepted 1 October 2012
individual constituents. To this end, we explore the concept of capitalizing on downstream convergence
Available online 9 November 2012
of intracellular signal transduction to experimentally simplify the means of determining xenobiotics that,
when combined, could result in increased or unexpected toxicity. In a proof of principle study, we exposed
Keywords:
Mixtures
HepG2 cells to deguelin, a natural isoflavonoid, alone and in combination with KCN, and determined the
Signal transduction relative post-translational phosphorylation responses to several key proteins related to mitochondrial
Apoptosis outer membrane permeabilization. Dose-dependent phosphorylation activity provides a clear identifi-
cation of threshold response to low-level exposures, and crosstalk amongst selected proteins correctly
forecasts mixtures interactions that may lead to increased toxicity. We then used Bliss Independence
to determine if the experimentally measured mixture phosphorylation responses could be predicted
with individual responses. Independence accurately predicted mixture interactions for deguelin and
KCN (87.5%). To more fully exhaust independence as a model for determining potential pharmacody-
namic interactions, we exposed HepG2 cells to deguelin and staurosporine, a broad kinase inhibitor;
independence accurately predicted these mixture responses (77.5%). In this study, we demonstrate the
potential of a new in vitro approach for the prediction of toxic mixtures interactions that is fundamentally
driven by the interdependence of signal transduction and apoptosis.
© 2012 Elsevier Ireland Ltd. All rights reserved.

1. Introduction Mixtures research takes many forms, but the basic conceptual
framework is simple: the physiological response to a mixture of
Exposure to chemical mixtures is an everyday reality, how- xenobiotics may be significantly different than the contributions
ever an understanding of their combined effects, and any potential of its individual constituents. These differences are the result of
prediction thereof, is extremely limited. From an environmental biochemical–xenobiotic interactions which can be pharmacoki-
perspective, realistic exposures potentially consist of hundreds to netic and/or pharmacodynamic in nature. Most current mixtures
thousands of chemicals per day. From a pharmaceutical perspec- research is focused on pharmacokinetics (absorption, distribution,
tive, adverse drug–drug interactions are of toxicological concern, metabolism and excretion) because alterations in the means by
but novel regimens involving mixtures have also shown therapeu- which the body processes a primary xenobiotic can increase or
tic potential (Ma and Waxman, 2008; Cottarel and Wierzbowski, decrease concentrations of a secondary xenobiotic in the blood
2007). Understanding and ultimately predicting chemical mixture and tissues by an order of magnitude or more (U.S. DHHS, 2006;
effects could not only elucidate potential risk factors associated for reviews see Zhang et al., 2009a, 2009b). On the other hand,
with exposures, but also offers the opportunity to develop a greater pharmacodynamic mixture effects can occur when the tissue- or
understanding of cellular mechanisms for enhanced drug discovery cellular-level response to the primary xenobiotic enhances sensi-
(Jonker et al., 2005; Fitzgerald et al., 2006). tivity to the secondary exposure; these responses are thought to
amplify or dampen the effects of secondary xenobiotics (Wu et al.,
2010; Natarajan et al., 2006).
The pharmacodynamic response of cells to xenobiotics
is primarily coordinated by signal transduction networks
∗ Corresponding author at: P.O. Box 6045, Morgantown, WV 26506, United States.
which typically follow a fundamental motif, the phospho-
Tel.: +1 304 615 9627; fax: +304 293 4904.
E-mail address: jonathan.boyd@mail.wvu.edu (J. Boyd). rylation/dephosphorylation cycle mediated by kinases and

0300-483X/$ – see front matter © 2012 Elsevier Ireland Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.tox.2012.10.010
114 J. Boyd et al. / Toxicology 313 (2013) 113–121

phosphatases (Kholodenko, 2006; Sauro and Kholodenko, 2004). 2.3. Dosing


This simple cycle is embedded in early response networks that use
Cells were seeded into clear-bottom, black-sided 96-well plates at a concen-
positive and negative feedback to generate extremely diverse func-
tration of 4 × 104 cells per well in RPMI-1640 without phenol red and allowed to
tions like signal amplification and perfect adaptation, reversible grow for 24 h before dosing. Media was then aspirated from wells and cells were
and irreversible switches, homeostasis, and oscillations (Tyson challenged with varying concentrations of single and mixed compounds (1 ␮L injec-
et al., 2003). One key irreversible switch is the mitochondrial tions) in fresh media (100 ␮L). Compounds were prepared in either DMSO (deguelin,
pathway of apoptosis, and both the signaling network and infra- staurosporine) or water (KCN) to microplate well concentrations of 0.01 ␮M to
100 ␮M. Media and injection volumes were held constant for all assays and DMSO
structure surrounding the decision to flip the switch is complex. well content was <1%.
Multiple signals, generated by either external or internal stresses,
converge upon the mitochondria and result in mitochondrial 2.4. MTT assay
outer membrane permeabilization (MOMP), which is currently
After 24 h of exposure to single compounds or mixtures, cell viability was deter-
considered the point of no return for apoptosis (i.e., irreversible
mined using the MTT assay, according to the manufacturer’s protocol. The assay is
switch) (Chipuk and Green, 2008; Galluzzi et al., 2012). based on the reduction of tetrazolium MTT to formazan by metabolically active
In the study presented here, we propose that by capitalizing cells, in part by the action of dehydrogenase enzymes, to generate reducing equiv-
on downstream convergence of signal transduction toward MOMP, alents such as NADH and NADPH. Briefly, MTT reagent was added to the wells of
one may experimentally simplify the means for determining xeno- the microplate, and after two hours of incubation at 37 ◦ C, intracellular formazan
crystals were solubilized with the provided detergent solution. Absorbance values
biotics that, when combined, could result in increased apoptosis. were obtained using the Safire2 microplate reader (Tecan US, Raleigh, NC) with a
Rather than assess all possible interactions between xenobiotics measurement wavelength of 570 nm and a reference wavelength of 700 nm, read
and multiple cellular targets, this paper suggests that it may be from the bottom. Experiments were each performed at least in triplicate on separate
possible to screen this pharmacodynamic response of cells to indi- days.
vidual compounds and map their crosstalk for the prediction of
2.5. Oxygen consumption assay
significant interactions. This approach is not reliant on the entire
xenobiotic mechanism, but instead, it monitors the cellular inter- Immediately after dosing single compounds, cellular oxygen consumption was
pretation of the exposure; thus, it has the potential to capture both assessed using the MitoXpress probe, according to manufacturer’s protocol. Briefly,
on- and off-target activity. A benefit of this type of approach is that oxygen-sensitive probe was diluted to a stock concentration of 1 ␮M, and stock
probe was diluted 1:15 in each well of a 96-well plate containing cells; 100 ␮L of pre-
it can identify doses of xenobiotics that may lead to aberrant mix-
warmed mineral oil was also added to each well to block ambient oxygen from the
ture effects, rather than focusing on the mode(s) of action of the cells. After pre-warming the plates at 37 ◦ C for 1 h, cells were challenged with varying
individual xenobiotics. concentrations of single compounds. Immediately following addition of compound,
In this initial proof of principle study, we exposed human hepa- oxygen consumption was determined by measuring fluorescence. Fluorescent signal
was obtained using the Safire2 microplate reader (Tecan US, Raleigh, NC) with exci-
tocellular carcinoma-derived cells (HepG2) to two mitochondrial
tation wavelength of 380 nm and emission wavelength of 650 nm, reading from the
inhibitors, deguelin and KCN, to test our hypothesis that spatiotem- bottom every 2 min for 24 h after dosing. Experiments were performed in triplicate
porally matched post-translational phosphorylation of proteins on separate days.
related to MOMP could be predictive of mixtures interactions that
lead to enhanced toxicity. Deguelin is a natural isoflavonoid that 2.6. Bio-plex multiplex immunoassay
inhibits NADH ubiquinone oxidoreductase (complex I), AKT (pro-
After 400 min exposure to increasing doses of deguelin (0.01, 0.1, 1.0, 10,
tein kinase B) and HSP90; further, it has shown promise as an 100 ␮M) in 1% DMSO alone and in combination with the EC50 concentrations of stau-
effective agent for a variety of cancer types, including hepatocel- rosporine (10 ␮M) or KCN (0.01–100 ␮M) obtained from Sigma Aldrich (St. Louis,
lular carcinoma (Lee et al., 2005; Peng et al., 2007; Oh et al., 2007, MO), cells were lysed using lysis buffer (BioRad, Hercules, CA) with 500 ␮M phenyl-
methanesulfonylfluoride (PMSF) (Sigma, St. Louis, MO) and phosphatase inhibitors
2008). KCN is an inhibitor of cytochrome c oxidase (complex IV) of
(BioRad, Hercules, CA). Total protein concentration was determined using the DC
the mitochondrial electron transport chain, and is often released Protein Assay (BioRad, Hercules, CA) according to the manufacturer’s instructions.
in the body following ingestion of cyanogenic glycosides which Protein phosphorylation was detected using multiplex assays analyzed with the
are found in over 2000 plants, including sorghum and cassava flow based BioPlex suspension array system (Bio-Rad, Hercules, CA). Beads con-
(Haque and Bradbury, 2002). To further evaluate the boundaries of taining antibodies against phosphorylated ERK1/2 (Thr202/Tyr204, Thr185/Tyr187),
AKT (Ser473), HSP27 (Ser78), I␬B␣ (Ser32/Ser36), JNK (Thr183/Tyr185), p38MAPK
this approach, we co-exposed HepG2 cells to deguelin and stau-
(Thr180/Tyr182), p53 (Ser15), and p90RSK (Thr359/Ser363) were obtained from Bio-
rosporine. Staurosporine is a microbial alkaloid natural product Rad (Hercules, CA). All experiments were performed on separate days in triplicate
and broad kinase inhibitor that has affinity for the ATP bind- and error bars reflect standard error of the mean.
ing site of numerous kinases (Omura et al., 1995; Fabian et al.,
2005). 2.7. Statistical analysis

Dose–response curves for MTT assays were generated by best-fit Hill-plot


regression of scatter plot data using Prism V5 (Graphpad Software, San Diego, CA).
2. Materials and methods Oxygen consumption curves were generated by choosing the best-fit polynomial
regression of scatter plot data using Prism V5 (Graphpad Software, San Diego, CA).
2.1. Materials The time point of interest (400 min) was selected using SAS JMP V8 (Cary, NC) where
the change in the slope of the curve reached a minimum for most exposures. Sta-
Deguelin, potassium cyanide, and staurosporine were obtained from Sigma tistical significance was assessed by using a two-way analysis of variance (ANOVA)
Aldrich (St. Louis, MO). RPMI-1640 containing phenol red, RPMI-1640 without with Bonferroni post-test. A difference at P < 0.05 level was considered statistically
phenol red, sodium pyruvate, HEPES, l-glutamine, fetal bovine serum, and penicillin- significant. For all graphs, error bars reflect standard error of the mean.
streptomycin were obtained from Invitrogen (Carlsbad, CA). Cell lines and MTT
assay kits were obtained from American Type Culture Collection (Manassas, VA).
MitoXpress oxygen probe was obtained from Luxcel Corporation (Cork, Ireland). 3. Results and discussion

3.1. KCN enhances deguelin toxicity


2.2. Cell culture
For this initial study, deguelin and KCN were selected due to
Human hepatocellular carcinoma-derived HepG2 cells were cultured in RPMI- their intracellular activity: we hypothesize that deguelin, a com-
1640, supplemented with 1 mM sodium pyruvate, 5 mM HEPES, 2 mM l-glutamine,
10% fetal bovine serum, 100 U/ml penicillin, and 100 ␮g/ml streptomycin. Cells were
plex I, AKT and HSP90 inhibitor, and KCN, a complex IV inhibitor,
maintained in a humidified atmosphere at 37 ◦ C, 5% CO2 and passaged at 80% con- will have both convergent and divergent post-translational phos-
fluence. phorylation responses that may be used to determine mixtures
J. Boyd et al. / Toxicology 313 (2013) 113–121 115

Fig. 1. KCN enhances the toxicity of deguelin in HepG2 cells. HepG2 viability dose–response curves for deguelin and KCN, alone and in combination. Viability was measured
by an MTT assay and is shown as percent viability. Percent viability was calculated relative to control cells which received dosing vehicle (<1% DMSO), but no deguelin or
KCN. Mixture responses found to be significantly different (p < 0.05) from individual deguelin or KCN are marked with * and # , respectively. Vertical dashed line indicates the
deguelin EC50 with gray highlight representing standard error of the mean (SEM).

interactions. To establish if these disparate xenobiotics lead to KCN initially inhibit oxygen consumption (as compared to controls
increased toxicity when combined as a mixture, we first measured which have been normalized to a value of 1). However, with the
viability of HepG2 cells in response to 24 h exposures of deguelin exception of 10 and 100 ␮M doses of deguelin, the cells appear to
and KCN, alone and in combination (Fig. 1). Individually, these recover from both deguelin and KCN induced oxygen consumption
compounds showed greatly different dose–response curves with inhibition and are shown to consume oxygen at rates equal to, or
calculated effective concentrations resulting in 50% viability (EC50 ) greater than, controls around 400 min post-exposure, implicating
of 40.2 ± 1.2 ␮M for deguelin and >100 ␮M (i.e., non-toxic at the this time-point as a key signaling event.
doses measured) for KCN. However, when combined, the mixture With this critical signaling time-point in hand, we simulta-
dose–response curves dramatically change. Even the lowest con- neously measured the phosphorylation of ERK1/2 (Thr202/Tyr204,
centration of KCN tested (0.01 ␮M) enhanced deguelin toxicity, as Thr185/Tyr187), AKT (Ser473), HSP27 (Ser78), I␬B␣ (Ser32/Ser36),
shown in Fig. 1. For all mixture combinations with KCN, the average JNK (Thr183/Tyr185), p38MAPK (Thr180/Tyr182), p53 (Ser15),
deguelin EC50 was 18.1 ± 3.0 ␮M. and p90RSK (Thr359/Ser363) at 400 min post-exposure to both
deguelin and KCN, alone and in combination. Our aim with
3.2. Observed phosphorylation responses the selection of these protein targets was to cast a wide net
with respect to signal transduction proteins relevant to MOMP,
We next explored the signal transduction response, by means and each of these proteins has demonstrated regulatory activ-
of phosphorylation activity, of HepG2 cells to each xenobiotic. ity over BCL-2 proteins that ultimately govern this process (Cory
Unfortunately to date, there is not an assay that can monitor et al., 2003; Green and Kroemer, 2004). Fig. 2A shows the rel-
post-translational phosphorylation in real-time without poten- ative phosphorylation response of HepG2 cells to exposures of
tially interfering with the response of our protein targets of interest. increasing doses of deguelin and KCN, individually (normalized
Therefore, a method for the temporal estimation of critical signaling to controls). Two-way ANOVA with Bonferroni post-test deter-
events is required. The ideal approach to determine key time points mined significant differences (p < 0.05) between deguelin and
in signal transduction response following exposure to deguelin and control for ERK1/2, p38MAPK, and p90RSK at 1 ␮M; ERK1/2,
KCN would involve an extracellular method, so as to decrease the p38MAPK and HSP27 at 10 ␮M; p38MAPK, I␬B␣ and p53 at 100 ␮M
risk of additional intracellular interference. Both of these com- (Fig. 2A). Thus, a sub-lethal threshold response exists for HepG2
pounds inhibit mitochondrial electron transport chain activity, cells exposed to deguelin at doses between 0.1 and 1 ␮M. KCN
which directly impacts the availability of ATP derived from oxida- was not found to be significantly different from control for any
tive phosphorylation. Relative rates of oxidative phosphorylation protein–dose combination, which is in agreement with viability
can be readily estimated extracellularly by the measurement of data that demonstrated all doses of KCN that were tested are
oxygen consumption. Measuring oxygen consumption in response non-toxic.
to inhibitors is a proven technique that has been used to identify To identify signal transduction proteins that may be related to
key temporal events relevant to both signal transduction (Jannsen- the toxicity of the mixture of deguelin and KCN, we compared the
Heininger et al., 2008) and apoptosis progression (Sung et al., 2010; observed mixture protein phosphorylation responses to those of
Tuttle et al., 2007). Thus, we measured the oxygen consumption its individual contributors. Results are shown in Fig. 2B but briefly,
of HepG2 cells in response to the individual compounds, deguelin the phosphoprotein response of the mixture was found to be sig-
and KCN (Appendix 1). From this assay, doses of deguelin and nificantly different (p < 0.05) from deguelin for 17 protein-dose
116 J. Boyd et al. / Toxicology 313 (2013) 113–121

Fig. 2. The relative phosphorylation response of eight proteins from HepG2 cells exposed to deguelin, KCN and their mixture. (A) Phosphorylation response of protein targets
(from cell lysates) to 400 min exposures of deguelin and KCN, individually. Phosphorylation was measured in HepG2 cell lysates (following exposures) using a multiplex
bead-based ELISA assay. Phosphorylation is shown relative to control cells, which received dosing vehicle (<1% DMSO), but no deguelin or KCN. Phosphorylation responses
found to be significantly different (p < 0.05) from controls are marked by * . (B) Phosphorylation response of protein targets to 400 min exposures of mixtures of deguelin and
KCN. Phosphorylation responses found to be significantly different (p < 0.05) from deguelin or KCN are marked with * and # , respectively.

combinations that involved JNK (0.01 ␮M DEG + 100 ␮M KCN), I␬B␣ response of the mixture was found to be significantly different
(0.01 ␮M DEG + 100 ␮M KCN; 0.10 ␮M DEG + 0.10–10 ␮M KCN), (p < 0.05) from KCN for many more protein-dose combinations
HSP27 (10 ␮M DEG + 0.01 ␮M KCN), p90RSK (10 ␮M DEG + 100 ␮M (65 total, see Fig. 2B). I␬B␣, JNK, and p38MAPK phosphorylation
KCN), and p38MAPK (10,100 ␮M DEG + 0.01–100 ␮M KCN). Due to responses from mixtures were significantly different from both
the general lack of protein phosphorylation activity in response deguelin and KCN, individually, and these may be contributing fac-
to individual KCN, it was not surprising that the phosphoprotein tors to the increased toxicity of the mixture.
J. Boyd et al. / Toxicology 313 (2013) 113–121 117

Fig. 3. Observed and predicted relative phosphorylation responses of HepG2 cells exposed to mixtures of deguelin and KCN. Phosphoprotein responses are shown as rows,
and mixture combinations are shown in columns. Observed responses found to be significantly different (p < 0.05) from predicted responses are marked with * .

3.3. Using independence to determine signal transduction Several mathematical techniques for predicting pharmacodynamic
mixture effects interactions have been proposed (Jonker et al., 2005; Tallarida,
2006, 2007), but most focus on interactions at specific receptor
In addition to experimental collection of signal transduction sites. Anderson and Dennison (2004) introduce the concept of
responses to disparate chemicals (spatiotemporally matched signal transduction cascades as a means for prediction of potential
by protein target and time course), understanding and ulti- pharmacodynamic interactions, but Jonker et al. (2005) extends
mately predicting mixtures effects requires mathematical models. this to a mathematical expression based upon the location where
118 J. Boyd et al. / Toxicology 313 (2013) 113–121

Fig. 4. Viability and phosphorylation responses of HepG2 cells to mixtures of deguelin and staurosporine. (A) HepG2 viability in response to mixtures of deguelin and 10 ␮M
staurosporine. Viability, shown as % viability, was measured as above and was calculated relative to control cells which received vehicle (<1% DMSO), but no deguelin or
staurosporine. Mixture viability responses found to be significantly different (p < 0.05) from deguelin alone are marked with * . (B) Phosphorylation response of HepG2 cells
to exposures of 10 ␮M staurosporine (EC50 dose). Phosphorylation was measured in cell lysates (following exposures) as stated above. Phosphorylation is shown relative to
control cells which received vehicle (<1% DMSO). (C) Observed and predicted relative phosphorylation responses of HepG2 cells exposed to mixtures of deguelin plus 10 ␮M
staurosporine. Observed responses found to be significantly different (p < 0.05) from predicted responses are marked with * .
J. Boyd et al. / Toxicology 313 (2013) 113–121 119

the interaction is occuring- sequential vs. dual (i.e. convergent) (p < 0.05) from controls, which received dosing vehicle (1% DMSO)
pathways, which appears to be conceptually based upon Loewe only. Only one phosphoprotein response (p53) was signifi-
additivity (Loewe, 1928) and Bliss independence (Bliss, 1939). Bliss cantly increased, whereas all other responses were significantly
Independence (also known as response addition) states that the decreased from control. As a mixture, deguelin (0.01–100 ␮M)
response from the combination of chemicals in a mixture is equal with staurosporine (10 ␮M) also showed disparate phosphopro-
to the conditional sum of component responses as defined for tein responses of various magnitudes (Fig. 4C). As shown in Fig. 4C,
the sum of independent event probabilities (Bliss, 1939; U.S. EPA, most phosphoprotein responses were lower than controls; how-
2000). Independence represents a simple probabilistic model that ever, p53 phosphoprotein responses were significantly increased
calculates the predicted effects f (xmix ) of a mixture with a known over controls for deguelin (0.01 and 0.1 ␮M) with staurosporine.
composition using the expression We determined the predicted phosphoprotein mixture
 responses for deguelin with staurosporine from their individ-
f (xmix ) = 1 − (1 − F(xi )) (1) ual responses using independence, as calculated from Eq. (1). For
i the 40 protein–dose combinations (8 proteins and 5 different
where F(xi ) can be any measured quantal response of an indi- mixture doses), independence correctly predicted 31 phospho-
vidual chemical. If one uses the phosphorylation of signal protein responses (Fig. 4C), when compared to observed mixture
transduction proteins for F(xi ), and the phosphoprotein tar- responses. By using deguelin in combination with staurosporine,
gets are sufficiently downstream of any direct activity, then a broad kinase inhibitor exhibiting activity at many kinases,
independence may be able to predict potential interactions we found that independence was a reasonable model for the
between two or more xenobiotics given the appropriate temporal prediction of post-translational phosphorylation activity with an
scale. accuracy of 77.5%.
To determine if the phosphoprotein response of a mixture of In summary, monitoring a limited number of convergent path-
deguelin and KCN follows independence, predicted values deter- ways, such as MOMP/apoptosis, representative of the cellular
mined from Eq. (1) were tested against observed mixture responses. interpretation of most on- or off-target activity, allows one to
For a full factorial dosing design (5 × 5) with 8 protein targets, screen disparate xenobiotics for potential mixtures interactions.
observed responses (200 total) were found to be significantly differ- We believe that this approach may be particularly suited for the
ent (p < 0.05) from predicted in only 25 protein–dose combinations identification of potentially toxic xenobiotic combinations based
(see Fig. 3). This results in an overall 87.5% prediction rate. The upon dose, rather than mode(s) of action. By determining pharma-
differences included 7 out of 8 proteins tested, but most discrepan- codynamic mixture responses of xenobiotics that exhibit activity
cies involved either ERK1/2 (6) or p38MAPK (11). While changes in at similar sites (deguelin with KCN; electron transport chain) or
JNK and p38MAPK phosphorylation that may be relevant to mix- xenobiotics with disparate activity at various magnitudes (deguelin
ture viability were not effectively predicted with independence, with staurosporine), independence was thoroughly examined as
I␬B␣ may be contributing to enhanced toxicity (see Fig. 1) and was a model to predict mixture phosphoprotein interactions. Predic-
accurately predicted at doses that are relevant to our viability data. tions based upon Bliss Independence can give insight as to potential
Interestingly, AKT is the only protein tested that is known to be post-translational phosphorylation responses that could lead to
directly inhibited by deguelin (Dell’Eva et al., 2007), and indepen- increased or unexpected mixture effects, which may be a signif-
dence correctly predicted phosphorylation activity of AKT across icant first step toward understanding, controlling, and ultimately
all dosing combinations. predicting mixtures effects.

3.4. Signal transduction independence with a broad kinase Conflict of interest statement
inhibitor
The authors declare that there are no conflicts of interest.
While independence was 87.5% accurate for predicting deguelin
and KCN mixture phosphorylation responses, we wanted to explore Acknowledgements
the limits of this model by using a broad kinase inhibitor that has
affinity for a large number of kinases. Staurosporine is a microbial The author would like to thank the Defense Advanced Research
alkaloid that has activity at many different kinases via interactions Projects Agency, the Johns Hopkins University Applied Physics
with their ATP-binding site, and thus we hypothesize that there Laboratory, and West Virginia University whose funding sup-
will be a vast range of direct phosphorylation responses for the ported portions of this research. Further, the author wishes to
8 proteins measured that are relevant to viability. We first deter- thank K.K., K.S., and N.B. for their thoughtful discussions and
mined viability of staurosporine alone at its manufacturer reported reviews of the manuscript, as well as countless others who dis-
EC50 (10 ␮M), and in combination with deguelin (Fig. 4A). For stau- cussed the theory of using signal transduction to predict mixtures
rosporine alone, HepG2 viability was found to be 61.58 ± 2.09% interactions.
when normalized to controls. Mixtures of deguelin (0.01–100 ␮M)
with staurosporine (10 ␮M) were statistically different (p < 0.05) Appendix A.
when compared to deguelin alone for the 0.01–10 ␮M doses and
decreased viability by 50% over this range. Oxygen consumption response of HepG2 cells to deguelin
We next determined the post-translational phosphorylation and KCN (individually) measured over 24 h. Oxygen consump-
response of the same 8 proteins (as described in Section 3.2) tion was measured using MitoXpress extracellular fluorescent
to exposures of staurosporine alone and in combination with probes (Luxcel Corp, Cork, Ireland) per manufacturer’s proto-
deguelin. The post-translational phosphorylation responses of cols. Oxygen consumption is shown relative to control cells
staurosporine alone (10 ␮M), as shown in Fig. 4B, varied in mag- which received vehicle (<1% DMSO), but no deguelin or KCN
nitude and all responses measured were statistically different (Fig. A1).
120 J. Boyd et al. / Toxicology 313 (2013) 113–121

Fig. A1. Oxygen consumption response of HepG2 cells to deguelin and KCN (individually) measured over 24 h. Oxygen consumption was measured using MitoXpress
extracellular fluorescent probes (Luxcel Corp, Cork, Ireland) per manufacturer’s protocols. Oxygen consumption is shown relative to control cells which received vehicle (less
than 1% DMSO), but no deguelin or KCN. Solid curves indicate oxygen consumption with respective error (dotted curves representing ± S.E.M.). Vertical dashed line indicates
the selected 400 min time-point.

References Fabian, M.A., Biggs 3rd, W.H., Treiber, D.K., Atteridge, C.E., Azimioara, M.D., Benedetti,
M.G., Carter, T.A., Ciceri, P., Edeen, P.T., Floyd, M., Ford, J.M., Galvin, M., Gerlach,
Anderson, M.E., Dennison, J.F., 2004. Mechanistic approaches for mixture risk J.L., Grotzfeld, R.M., Herrgard, S., Insko, D.E., Insko, M.A., Lai, A.G., Lélias, J.M.,
assessments-present capabilities with simple mixtures and future directions. Mehta, S.A., Milanov, Z.V., Velasco, A.M., Wodicka, L.M., Patel, H.K., Zarrinkar,
Environ. Tox. Pharm. 16, 1–11. P.P., Lockhart, D.J., 2005. A small-molecule kinase interaction map for clinical
Bliss, C.I., 1939. The toxicity of poisons applied jointly. Ann. Appl. Biol. 26, 585–615. kinase inhibitors. Nat. Biotechnol. 23, 329–336.
Chipuk, J.E., Green, D.R., 2008. How do BCL-2 proteins induce mitochondrial outer Fitzgerald, J.B., Schoeberl, B., Nielsen, U.B., Orger, P.K., 2006. Systems biology and
membrane permeabilization? Trends Cell Biol. 18, 157–164. combination therapy in the quest for clinical efficacy. Nat. Chem. Biol. 2,
Cory, S., Huang, D.C., Adams, J.M., 2003. The Bcl-2 family: roles in cell survival and 458–466.
oncogenesis. Oncogene 24, 8590–8607. Galluzzi, L., Vitale, I., Abrams, J.M., Alnemri, E.S., Baehrecke, E.H., Blagosklonny, M.V.,
Cottarel, G., Wierzbowski, J., 2007. Combination drugs, an emerging option for Dawson, T.M., Dawson, V.L., El-Deiry, W.S., Fulda, S., Gottlieb, E., Green, D.R.,
antibacterial therapy. Trends Biotechnol. 25, 548–554. Hengartner, M.O., Kepp, O., Knight, R.A., Kumar, S., Lipton, S.A., Lu, X., Madeo,
Dell’Eva, R., Ambrosini, C., Minghelli, S., Noonan, D.M., Albini, A., Ferrari, N., 2007. The F., Malorni, W., Mehlen, P., Nuñez, G., Peter, M.E., Piacentini, M., Rubinsztein,
Akt inhibitor deguelin, is an angiopreventive agent also acting on the NF-kappaB D.C., Shi, Y., Simon, H.U., Vandenabeele, P., White, E., Yuan, J., Zhivotovsky, B.,
pathway. Carcinogenesis 28, 404–413. Melino, G., Kroemer, G., 2012. Molecular definitions of cell death subroutines:
J. Boyd et al. / Toxicology 313 (2013) 113–121 121

recommendations of the Nomenclature Committee on Cell Death 2012. Cell Peng, X.H., Karna, P., O’Regan, R.M., 2007. Down-regulation of inhibitor of apoptosis
Death Differ. 19, 107–120. proteins by deguelin selectively induces apoptosis in breast cancer cells. Mol.
Green, D.R., Kroemer, G., 2004. The pathophysiology of mitochondrial cell death. Pharmacol. 71, 101–111.
Science 305, 626–629. Sauro, H.M., Kholodenko, B.N., 2004. Quantitative analysis of signaling networks.
Haque, M.R., Bradbury, J.H., 2002. Total cyanide determination of plants and foods Prog. Biophys. Mol. Biol. 86, 5–43.
using the picrate and acid hydrolysis methods. Food Chem. 77, 107–114. Sung, H.J., Ma, W., Wang, P.Y., Hynes, J., O’Riordan, T.C., Combs, C.A., McCoy, J.P., Bunz,
Jannsen-Heininger, Y.M.W., Mossman, B.T., Heintz, N., Forman, H.J., Kalyanaraman, F., Kang, J.G., Hwang, P.M., 2010. Mitochondrial respiration protects against
B., Finkel, T., Stamler, J.S., Rhee, S.G., van der Vliet, A., 2008. Redox-based reg- oxygen-associated DNA damage. Nat. Commun. 1, 1–8.
ulation of signal transduction principles, pitfalls, and promises. Free Rad. Biol. Tyson, J.J., Chen, K.C., Novak, B., 2003. Sniffers, buzzers, toggles and blinkers: dynam-
Med. 45, 1–17. ics of regulatory and signaling pathways in the cell. Curr. Opin. Cell Biol. 15,
Jonker, D.M., VIsser, S.A.G., van der Graaf, P.H., Voskuyl, R.A., Danhof, M., 2005. 221–231.
Towards a mechanism-based analysis of pharmacodynamic drug–drug inter- Tallarida, R.J., 2006. An overview of drug combination analysis with isobolograms.
actions in vivo. Pharmacol. Ther. 106, 1–18. J. Pharmacol. Exp. Ther. 319, 1–7.
Kholodenko, B.N., 2006. Cell signaling dynamics in time and space. Nat. Rev. Mol. Tallarida, R.J., 2007. Interactions between drugs and occupied receptors. Pharma-
Cell Biol. 7, 165–176. col.Ther. 113, 197–209.
Lee, H-Y., Oh, S-H., Woo, J.K., 2005. Chemopreventive effects of deguelin, a novel Tuttle, S.W., Maity, A., Oprysko, P.R., Kachur, A.V., Ayene, I.S., Bigalow, J., Koch, C.J.,
Akt inhibitor, on tobacco-induced lung tumorigenesis. J. Natl. Cancer Inst. 97, 2007. Detection of reactive oxygen species via endogenous oxidative pentose
1695–1699. phosphate cycle activity in response to oxygen concentration: implications for
Loewe, S., 1928. Die quantitativen probleme der pharmakologie. Ergebn. Physiol. 27, the mechanism of HIF-1alpha stabilization under moderate hypoxia. J. Biol.
47–187. Chem. 282, 36790–36796.
Ma, J., Waxman, D.J., 2008. Combination of antiangiogenesis with chemotherapy for U.S. Dept of Health and Human Services, Food and Drug Administration, Center for
more effective cancer treatment. Mol. Cancer Ther. 7, 3670–3684. Drug Evaluation and Research, Center for Biologics Evaluation and Research.
Natarajan, M., Lin, K.M., Hsueh, R.C., Sternweis, P.C., Ranganathan, R., 2006. A global Guidance for Industry: Drug Interaction Studies – Study Design, Data Analysis,
analysis of cross-talk in a mammalian cellular signalling network. Nat. Cell Biol. and Implications for Dosing and Labeling. September 2006.
8, 571–580. U.S. EPA. Supplementary Guidance for Conducting Health Risk Assessment of Chem-
Oh, S.H., Woo, J.K., Jin, Q., 2008. Identification of novel antiangiogenic anticancer ical Mixtures (2000).
activitieso f deguelin targeting hypoxia-inducible factor-1␣. Int. J. Cancer 122, Wu, Z., Zhao, X.-M., Chen, L., 2010. A systems biology approach to identify effective
5–14. cocktail drugs. BMC Syst. Biol. 4, S2–S7.
Oh, S.H., Woo, J.K., Yazici, Y.D., Myers, J.N., Kim, W.-Y., Jin, Q., Hong, S.S., Park, H.-J., Zhang, L., Zhang, Y., Huang, S.M., 2009a. Scientific and regulatory perspectives on
Suh, Y.-G., Kim, K.-W., Hong, W.K., Lee, H.-Y., 2007. Structural basis for depletion metabolizing enzyme-transporter interplay and its role in drug interactions:
of heat shock protein 90 client proteins by deguelin. J. Natl. Cancer Inst. 99, challenges in predicting drug interactions. Mol. Pharm. 6, 1766–1774.
949–961. Zhang, L., Zhang, Y.D., Zhao, P., Huang, S.M., 2009b. Predicting drug–drug interac-
Omura, S., Sasaki, Y., Iwai, Y., Takeshima, H., 1995. Staurosporine, a potentially tions: an FDA perspective. AAPS J. 11, 300–306.
important gift from a microorganism. J. Antibiot. (Tokyo) 48, 535–548.

You might also like