ArticlePDF Available

Optimization of a Non-Radioactive High-Throughput Assay for Decarboxylase Enzymes

Authors:

Abstract and Figures

Herein, we describe the optimization of a linked enzyme assay suitable for high-throughput screening of decarboxylases, a target family whose activity has historically been difficult to quantify. Our approach uses a commercially available bicarbonate detection reagent to measure decarboxylase activity. The assay is performed in a fully enclosed automated screening system under inert nitrogen atmosphere to minimize perturbation by exogenous CO2. Receiver operating characteristic (ROC) analysis following a pilot screen of a small library of approximately 3,600 unique molecules for inhibitors of Trypanosoma brucei ornithine decarboxylase quantitatively demonstrates that the assay has excellent discriminatory power (area under the curve = 0.90 with 95% confidence interval between 0.82 and 0.97).
Optimization of assay conditions. ( A ) Increase in background signal under normal atmosphere. The signal seen here in green is solely from atmospheric CO 2 (orange = nitrogen atmosphere, no ornithine; green = standard atmosphere, no ornithine; blue = nitrogen atmosphere, 625 μM ornithine; red = standard atmosphere, 625 μM ornithine). Quadruplicate data were collected in a 384-well microplate as described in the presence of the indicated concentrations of ornithine. All other reagent concentrations were identical to the optimized conditions described in Materials and Methods. The nitrogen atmosphere was maintained as described with O 2 levels under 2.5%. ( B ) Optimization of enzyme levels. ▼ = Z ′ values, ■ = reaction rates. Dashed line indicates standard Z ′ cutoff value of 0.5. Data were collected in a 384-well microplate using optimized assay conditions with varied fi nal ornithine decarboxylase (ODC) concentrations. Z ′ Values were calculated using 8 positive (1 mM difl uoromethylornithine (DFMO)) and 8 negative (DMSO) controls. The signal window at 150 nM ODC is ∼5-fold. ( C ) Determination of ODC K m at 150 nM ODC and 60 μM PLP. Data were collected in a 384-well plate as described and fi t to the Michaelis–Menten equation. ( D ) Optimization of Infi nity CO 2 percentage (■ = 60% Infi nity CO 2 , ▲ = 45% Infi nity CO 2 , ▼ = 30% Infi nity CO 2 , ♦ = 15% Infi nity CO 2 , @BULLET = 7.5% Infi nity CO 2 ). Data were collected in 384-well plates as described at optimized assay conditions with varied Infi nity CO 2 . Data points were taken every 15 s in quadruplicate. The plot of AU 340 vs. time at 60% Infi nity CO 2 represents a typical data set under optimized assay conditions with a ∆AU 340 of ∼0.4 AU.
… 
Content may be subject to copyright.
© MARY ANN LIEBERT, INC. VOL. 8 NO. 2 APRIL 2010 ASSAY and Drug Development Technologies 175
DOI: 10.1089/adt.2009.0249
D a v i d C . S m i t h s o n , 1,2 A n a n g A . S h e l a t , 2 J e f f r e y B a l d w i n , 3
M a r g a r e t A . P h i l l i p s , 3 a n d R . K i p l i n G u y 2
1 Graduate Program in Chemistry and Chemical Biology, University
of California, San Francisco, California.
2 Department of Chemical Biology and Therapeutics, St. Jude
Children’s Research Hospital, Memphis, Tennessee.
3 Department of Pharmacology, The University of Texas
S o u t h w e s t e r n M e d i c a l C e n t e r a t D a l l a s , D a l l a s , T e x a s .
A B S T R A C T
Herein, we describe the optimization of a linked enzyme assay suit-
able for high-throughput screening of decarboxylases, a target fam-
ily whose activity has historically been dif ficult to quantif y. Our
approach u ses a commerci ally available bicarbonate detection reagent
to measure decarbox ylase activity. The assay is performed in a fully
enclosed automated screening system under inert nitrogen atmo-
sphere to minimize perturbation by exogenous CO
2 . Receiver operat-
ing characteristic (ROC) analysis following a pilot screen of a small
library of 3,600 unique molec ules for inhibitors of Tr ypanosoma
brucei ornithine decarboxylase quantitatively demonstrates that the
assay has excellent discriminatory power (area under the curve =
0.90 with 95% confidence interval between 0.82 and 0.97).
INTRODUCTION
D ecarboxylase enzy mes represent a significant target fam-
ily for the development of therapeutic agent s. Ornithine
decarboxylase (ODC) and S -adenosylmethionine decar-
boxylase control the polyamine biosynthetic pathway,
a therapeutic target in both oncology and parasitology.
1,2 Other
medically relevant decarboxylases include histidine decarboxyl-
ase, a target in the inflammation pathway, DOPA decarboxylase,
a Parkinson’s disease target, and diaminopimelate decarboxylase,
a potential antibiotic target.
3–5 In spite of the importance of the
enzyme class, few decarboxylases have been subjected to large-
scale drug discovery efforts, in part due to difficulty in quantifying
thei r activities in a ma nner c ompatible with moder n high- th rough-
put approaches. Classical methods for assaying decarboxylase func-
tion often involve capture of
14 CO
2 from radiolabeled substrates or
de ri va ti zation of t he e nzy me p ro duc ts f ol lowed by HP LC a naly si s.
6,7
Neither of these techniques is suitable for high-throughput screen-
ing (H TS) efforts, as they involve either formation of radioactive
gas or leng thy, resource intensive detection procedures. Use of a
commercial low-throughput method linking the production of CO
2
to the consumption of NADH using phosphoenolpy ruvate carbox-
ylase (PEPC) and malate dehydrogenase (MDH) to track decarbox-
ylase activities has been reported
8 ( Scheme 1 ). Reaction progress
was measured by the decrease in NADH absorbance at 340 nm. We
believed this assay was a good candidate for adaptation to HTS.
The target chosen for optimization of this assay system was
ODC from Tr y pa no so ma br ucei , the causative agent of human
African trypanosomiasis. T his disease is fatal if left untreated
and is a major health problem in much of central Africa.
9 The
only clinica lly validated molecular target for treatment of T.
brucei infections is ODC, which catalyzes the f irst step in poly-
amine metabolism, the decarbox ylation of ornithine to produce
putrescine. Putrescine and the other polyamines, sper midine and
sper mine, are necessar y for cellular reproduction, making their
regulatory enzymes attractive d rug targets in both parasitology
and oncology. The biological roles of polyam ines are numerous,
and they have been implicated in the regulation of a wide range
of important genes, including P53 and c-myc .
10,11
Optimization of a Non-Radioactive
High-Throughput Assay for Decarboxylase
Enzymes
ABBREVIATIONS: DFMO, di uoromethyl ornithine; DTT, dithiothreitol; HTS, high throughput screening; MDH, malate dehydrogenase; ODC, ornithine decarboxylase;
PEPC, phosphoenol pyruvate carboxylase; PLP, pyridoxal-5-phosphate; ROC, receiver operator characteristic.
adt.2009.0249.indd 175 4/19/2010 11:37:43 AM
SMITHSON ET AL.
176 ASSAY and Drug Development Technologies APRIL 2010
In mammalian cells, ODC is highly regulated and possesses a
very short half-life of 10 to 20 min.
12 However, in protozoal para-
sites, par ticularly T. brucei brucei and T. b ru ce i g am bi ense , the
enzyme is longer lived (4 h).
13 This difference allows the use of
ir rever sible and non select ive inhibitor s of ODC such as difluorom-
ethylornithine (DFMO) in clinical treatments for African trypano-
somiasis.
14 Unfortunately, DFMO possesses poor pharmacokinetic
proper ties, resulting in the need for multi-day high-dosage intra-
venous treatment regimens that limit its use in the third world.
15
To date, most d rug discover y efforts directed toward ODC
inh ibitors have focused on analogs of or nith ine (such as DFMO),
putrescine, or pyridoxal-5-phosphate (PLP), a cofactor necessar y
for ODC activity shared by many other enzymes.
16,17 None of these
have proved as effective as DFMO for treatment of T. b rucei infec-
tions. No large-scale efforts to discover inhibitors based on scaf-
folds other than substrate, product, or cofactor analogs have been
repor ted. The lack of large-scale drug discover y effor ts has been
due in part to the difficulty in assay ing ODC activity in a high-
throughput manner.
Therefore, we have optim ized a commercial enzy me-linked
bicarbonate detection system for use in H TS of ODC. We report
the results of this optimization and the performance of the assay
on a proof-of-concept screen of 3,600 unique molecu les.
MATERIALS AND METHODS
Materials
All chemicals assayed in this st udy were purchased from ven-
dors without further purification. DI water was filtered with a
MilliQ Synthesis Ultra-Pu re water system (Millipore, Billerica,
MA) immediately before use. Infinity™ Carbon Dioxide Liquid
Stable Reagent was purchased from Thermo Fisher Scientific
(Waltham, M A).
L -Ornithine, pyridoxal 5-phosphate (PLP), and
dithiothr eitol (DTT ) were purchased from Sigma-Aldrich (St. Louis,
MO). DFMO-HCl was purchased from Chem-Impex International
(Wood Dale, IL). All plate-based enzymatic assays were perfor med
in 384-well black-sided, clear-bottomed polystyrene microplates
(#3,702) from Cor ning Life Sciences (Acton, M A). The bioac-
tive compound screening librar y at St. Jude Children’s Research
Hospital was assembled from commercially available collections
including the Prestwick Chemical Library (Prestwick Chemical,
Ill kirch, France); the LOPAC Collection (Sigma-Aldrich, St. Louis,
MO); and the Spectrum Collection, the N INDS Collection, the
Natural Product Collection, and t he Killer Plate Collection from
Microsource (Microsource Discovery systems, Gaylordsville, CT).
The tota l bioactive test set contains 7,300 compounds, including
many internal replicates. The total number of unique compounds
in the collection is 3,600 molecules.
Purification of T. br u ce i ODC
ODC was expressed as an N-terminal 6×His-tag fusion protein
in Escherichia coli BL21 (DE3) cells as described.
8 Protein was
purified by Ni
2+ -NTA-agarose column followed by Superdex 200
gel-filtration column chromatography. Fractions containing the
desired protein were identified by SDS-PAGE. Those conta ining
pure ODC were combined and concentrated using an Amicon-Ultra
centrifugal filter device (10 kDa cutoff, UFC901024; Millipore,
Bi ller ica, M A) to concentrat ions of 40 mg/mL. Yields of purified
ODC were generally 7 to 13 mg/L of cultured cells.
HO
NH2
HCO3
O
OO
O
OO
O
OOO
OOH
OO
P
HO
NH2
NH2+CO2
H2N
O
ODC
PEPC
Monitored via Decrease in
Absorbance at 340 nm
MDH
NADH NAD+
OO
S c h e m e 1 . Linked a ssay mechanism. Ornithine decarbox ylase (ODC) catalyzes the dec arboxylation of or nithine, releasing CO
2 , which is then
captured by the basic buffer (pH 8.05) as bicarbonate. Phosphoenolpyruvate carboxylase (PEPC) uses this bicarbonate to generate oxa-
loacetate from phosphoenolpyruvate. The oxaloacetate is then reduced by malate dehydrogenase (MDH) to malate in a NADH-dependent
fashion. There is a 1:1 relationship between the amount of CO
2 produced by ODC and t he amount of NADH ox idized by MDH, a llowing kinetic
parameters for ODC to be calculated from obser ving NADH levels as a function of time. The assay system has been optimized such that ODC
is the rate-limiting step.
adt.2009.0249.indd 176 4/19/2010 11:37:44 AM
DECARBOXYLASE ENZYME HTS
© MARY ANN LIEBERT, INC. • VOL . 8 NO. 2 • APRIL 2010 ASSAY and Drug Development Technologies 177
Assay Automation and Nitrogen Atmosphere Generation
All screening data were generated on a High Resolution
Engineering (Woburn, MA) integrated screening system using
Liconic plate incubators (Woburn, MA) and a Staubli T60 robotic
arm (Staubli, SC). This s ystem is enclosed in a gas-tight Plex iglas
enclosure allowing a nitrogen atmosphere to be generated by
continual purging with 30 psi nitrogen through t win 8-mm
inner diameter tubes. Nitrogen was obtained by boil-of f from liq-
uid nitrogen usi ng an in-house dry nitrogen system supplied by
NexAir (Memphis, TN). Nitrogen consumption during screening
was estimated to be 200 L/min (12,000 L/h or 1,200 L per plate
for a kinetic read). Percent oxygen within the enclosure was mon-
itored using an Air Aware oxygen detector (model 6810-0056;
Industrial Scientific, Oakdale, PA) and maintained at <2.5%
throughout all high-t hroughput assays. Percent carbon diox-
ide was below detectable levels (0.5%) as measured by Liconic
STR240 series incubators (Liconic Instruments, Woburn, MA).
Assay solutions were dispensed using Mat rix Wellmates (Matrix
Technologies, NH) equipped with 1 μL rated tubes. Plates were
centrifuged after all liquid additions using a Vspin plate cen-
trifuge (Velocit y11, Menlo Park, CA). Compound transfers were
performed using a 384-well pin tool equipped wit h 10 nL slot-
ted hydrophobic surface-coated pins (V&P Scientific, San Diego,
CA). This allowed delivery of 25 nL of DMSO stock solution
with CVs of <10%. All absorbance data were measured using an
EnVision Multilabel Plate Reader equipped with a 340 nm narrow
bandwidth fi lter (Perkin Elmer, 2100-5740). During automation,
the screening system was operated offline and individual instru-
ments were accessed using manual operation of the robot arm.
ODC-PEPC-MDH-Linked Assay
This assay was performed under nitrogen atmosphere. Assay
buffers were prepared under normal atmosphere while f lushing
with a stream of nitrogen and transferred to enclosed nitrogen
atmosphere upon complet ion. Assay buffer (66 mM Tris, 25 mM
NaCl, 8 mM MgSO
4 , 0.01% Triton-X, pH 8.05) was prepared daily.
The assay reaction was prepared using 2 master mixes A and B,
which were prepared immediately before use. Mix A contained
ornithine (0–10 mM), PLP (0–937 μM), and 5.7 mM DTT in assay
buffer. Mix A was prepared immediately before testing from fro-
zen stocks prepared in water (0.5 M Orn, pH 7.5, 20 mM PLP, and
1 M DTT). Mix B contained I nfinity Carbon Dioxide Liquid Stable
Reagent (Infinity CO
2 , Thermo Fisher Scientific) and ODC (0–300
nM). Mix B was prepared immediately prior to testing from fresh
Inf init y Carbon Dioxide Liquid Stable Reagent and frozen ODC
stocks. For testing, 15 μL Mix B was added to appropriate wells in a
384-well clear-bottomed plate followed by compounds transferred
by pin. T he 10 μL Mix A was then added to star t the reaction.
Final optim ized assay concentrations were 2.3 mM DT T, 600 μM
ornithi ne, 60 μ M PLP, 60% I nfinit y Carbon Dioxide Liquid Stable
Reagent, 150 nM ODC, 10 μM test compound, and 0.01% Triton-X,
in a 25-μ L final volume unless otherwise specified. Reaction pro-
gress was monitored by following absorbance at 340 nm using
an Env ision plate reader (Perk in Elmer) equipped with a narrow
bandwidth 340 nm filter (Perkin Elmer, 2100-5740). Absorbance
was monitored for 20 min, with time points taken every minute.
Data from 15 to 20 min after addition of Mix A were fit to a linear
model using statistical methods described below. The resulting
slope of this fit was taken as the rate of the reaction and used as
the endpoint for the assay.
Compounds for screening were placed in 384-well poly propyl-
ene plates (Corning Life Sciences, Acton, MA) at 10 mM concen-
tration in DMSO with columns 1, 2, 13, and 14 empty. Positive
controls (DFMO, 1 M, 10 μL) were placed in a separate 384-well
polypropylene plate in wells A2, B2, C2, D2, E14, F14, G14, H14, I2,
J2, K2, L2, M14, N14, O14, and P14. Negative controls (DMSO, 10
μL) were placed in the same plate as positive controls in wells A14,
B14, C14, D14, E2, F2, G2, H2, I14, J14, K14, L14, M2, N2, O2, and
P2. All microplate compound transfers were accomplished using
a 384-well pin tool equipped with 10 nL hydrophobic surface-
coated pins (V&P Scientific, San Diego, CA). This allowed deliv-
ery of 25 nL of DMSO stock solution with CVs of <10%.
Cuvette assays were performed as described above with the
following minor mod ifications; the f inal assay volume in cuvettes
was 500 μL at 40% Infinity Carbon Dioxide Liquid Stable Reagent,
50 μM PLP, 50 μM DTT, 1% DMSO, and varied ornithine concen-
tration s from 10 mM to 100 μM. As with microplate assays, assay
buffer (66 mM Tris, 25 mM NaCl, 8 mM MgSO
4 , 0.01% Triton-X,
pH 8.05) was prepared fresh daily.
PEPC-MDH-Linked Assay—Microplate
For assay of the linking enzymes, assay buffer (66 mM Tris, 25
mM NaCl, 8 mM MgSO
4 , 0.01% Triton-X, pH 8.05) was prepared
daily using water. The assay reaction was prepared in 2 master
mixes. Mix A contained 1.25 mM sodium bicarbonate (Sigma-
Aldrich), 100 μM PLP, and 5.7 mM DTT in assay buffer. Mix B was
100% Infinity Carbon Dioxide Liquid Stable Reagent. For testing
15 μL, Mix B was added to appropriate wells of a 384-well clear-
bottomed microplate followed by pin-transfer compound DMSO
stocks. Compounds were allowed to equilibrate in the presence of
enzymes for 20 min before substrate was added. The reaction was
star ted by addition of 10 μL Mix A and reaction progress was mon-
itored by absorbance at 340 nm. Positive controls were wells with
no sodium bicarbonate added, negative controls were DMSO.
Final assay concentrations were 2.3 m M DTT, 60 μM PLP, 0.75
mM sodium bicarbonate, 60% Infinity Carbon Dioxide Liquid
adt.2009.0249.indd 177 4/19/2010 11:37:44 AM
SMITHSON ET AL.
178 ASSAY and Drug Development Technologies APRIL 2010
Stable Reagent, and 0.01% Triton-X. Reaction progress was mon-
itored by decrease in absorbance at 340 nm using an Envision
plate reader (Perkin Elmer) equipped with a nar row bandwidth
340 nm f ilter (Perkin Elmer, 2100-5740) for 10 min with time
points taken every 1 min. Data from 1 to 5 min were fit to a linear
model using statistical methods as descr ibed below and normal-
ized to the positive and negative controls.
Primary Screening Data Analysis and Reaction
Rate Calculation
Pr imar y screening data analysis was perfor med using cus-
tom protocols w ritten in Pipeline Pilot (v. 6.1.1, Accelr ys) and the
R program (htt p://www.r-project.org/, v. 2.7.0).
18 For rate deter-
mination, kinetic data were fit using 3 met hods: a linear model
using least squares (“linear,” calculated using the lm function in
robustbase R package, v. 0.2-7), a linear model using only the
difference between the first and last time points (“delta”), and a
robust linear model using an iteratively re-weighted least squares
algorithm (“robust,” calcu lated using the lmrob function in robust
ba se R p ac ka ge , v. 0. 2-7).
19 For hit identification, the fina l 5 min of
data (6 data points in total including the 15 min time point) were
fit using the “linear” method. The fitted rates were the values
used in the calculations of all plate statistics. Only plates passing
the minimum Z and Z -factor thresholds of 0.4 and 0.4, respec-
tively, were accepted. Initial screening hits were determined on
a plate-by-plate basis by identifying compounds with activities
that were simultaneously outliers from the negative control and
variable compound populations. The outlier cutoffs were calcu-
lated as t he upper fourth plus 1.5 times the fourth spread (the
upper fourth and fourt h spread are similar to the third quartile
and interquartile range, respectively), a robust statistic that cor-
responds to a P value of 0.005 for normal distr ibutions.
20 For
Z calculations, 16 positive and 16 negative controls were used
unless other wise stated. Reaction rate was transformed from AU/
min to m M NADH/min using an ex tinction coeff icient of 6.349
AU/mM/cm for NADH and an approximate path length of 0.4 cm
for assays performed at 25 μL f inal volume in a 384-well plate.
Reaction rates for dose–response data were calculated as
descr ibed above. Rates were then normalized to DMSO and DFMO
controls and sigmoidal cur ves with variable slopes were fit using
Graphpad Prism 4.03. No constraints were used when fitt ing this
data.
Receiver Operating Characteristic Analysis
Receiver operating characteristic (ROC) analysis was per-
formed using custom R scripts (http://www.r-project.org/, v. 2.9.0)
and the R rocr package (v. 1.0.2). The ROC test set was composed
of 63 compounds that uniformly sample from the distribution of
observed primary screen activities (7% to 103%). A compound
was considered a true positive if it inhibited ODC in a dose-depen-
dent fashion, had no effect in the PEPC-MDH assay system, and
showed a 2-fold increase in reaction r ate when screened with 300
nM ODC. The ROC curve was formed by plotting the true positive
rate vs. the false positive rate as a function of decreasing prima ry
screen activity. Conf idence intervals for the ROC area under the
cur ve (AUC) were calculated from 200 bootstrap simulations.
R E S U L T S
Optimization of ODC-PEPC-MDH-Linked Assay
The assay system was chosen in part because the linking sys-
tem was available from Ther mo Fisher Scientif ic, simplifying
quality control and large-scale reagent sourcing. This system was
designed for clinical detection of bicarbonate in bod ily f luids, and
is extremely sensitive to low levels of dissolved CO
2 , which is in
equilibrium with bicarbonate in aqueous solutions. Unfortunately,
this sensitivit y is a disadvantage in HTS applications. The back-
ground signal produced by atmospheric CO
2 reduces the assay
linear time and causes reagents placed on the screening deck
to rapidly degrade. However, if reagents are stored, dispensed,
incubated, and read under a nitrogen atmosphere, this liability is
largely mitigated. When the assay is performed under nitrogen,
the signal-to-noise ratio increases from 2.5 to 8.1 and t here is a
significant increase in reagent stability (see Fig. 1A ). For this rea-
son, all optimization and screening experiments were performed
under an inert nitrogen atmosphere as described.
The assay rate was linear with respect to ODC concentration
from 20 to 600 nM. A final concentration of 150 nM ODC was
chosen for the primary screen, based upon signal-to-noise ratio
and Z score (>4 and >0.5, respectively, Fig. 1B ). The final PLP
concentration chosen was 60 μM, 300-fold over the literature
reported K
m ,
21 to minimize the probability of finding compounds
that interf er ed di rec tl y w it h P LP, o r comp et ed for th e PLP-bi nd ing
site. The assay buffer was not optimized, and was formulated to
match the commercial linking buffer in order to maintain perfor-
mance of the linking system. Several methods to remove dissolved
CO
2 from the buffer were tested including vacuum degassing and
nitrogen sparging. None offered significant reductions in back-
ground signal over using fresh MilliQ Sy nthesis grade water (data
not shown). To ensure that the assay system was performing com-
parably to other literature methods, the K
m for ornithine was mea-
sured. The va lues obtained (420 ± 22 μM, Fig. 1C ) were consistent
with liter ature values (370–50 0 μM).
22 ,
23 T he relatively low orni-
thine concentration chosen for screening (625 μM, 1.5 × K
m ) was
selected to maximize the probability of finding or nith ine compet-
itive inhibitors. The k
cat determined under screening conditions
(0.71 s
1 at 22°C) is 10-fold lower than the reported literature
adt.2009.0249.indd 178 4/19/2010 11:37:45 AM
DECARBOXYLASE ENZYME HTS
© MARY ANN LIEBERT, INC. • VOL . 8 NO. 2 • APRIL 2010 ASSAY and Drug Development Technologies 179
values.
21 This was determined to be a temperature effect, and it
was possible to replicate literat ure values by performing the assay
at 37°C. Due to limitations with our integrated plate reader, all
HTS was performed at room temperature (22°C).
Finally, the amount of linking enzyme mix (Infinity CO
2 ) was
optimized ( Fig. 1D ). While the signal remained linear over the
20-min assay window for both 60% and 45% Infinity CO
2 , 60%
was chosen in order to maximize the linear time available during
screening and reduce dependence on precise timing during the
process. The percentage was not increased past 60% due to the
fact that absorbance values were already approaching 1.2 at this
concentration. The assay system’s tolerance for DMSO was also
tested, and was shown to be up to 10% at optimized screening
conditions (data not shown).
Optimization of Secondary PEPC-MDH Assay
In light of the fact that this assay is dependent upon the activit y
of the 2 linking enzy mes as well as that of ODC, a secondar y assay
0
01020304050
Time (min)
60 70 80 90
0 0.0
0.00
0.25
0.50
0.75
1.00
1.25
AU340
2.5 5.0 7.5 10.0
Time (min)
Optimization of Infinity CO2 Percentage
12.5 15.0 17.5 20.0
0.0000
0.0025
0.0050
Rate (mM/min)
0.0075
5,000 10,000
Determination of Orn Km
Km = 420 ± 22 μM
kcat = 0.78 ± 0.02 s1
15,000 20,000 25,000
Orn (μM)
0.00 0.0
0.1
0.2
0.3
0.4
0.5
0.6
Z Value
0.7
0.8
0.9
0.01
0.02
0.03
0.04
250 500 750 1,000 1,250 1,500
0.00
0.25
0.50
0.75
1.00
[ODC (nM)]
Optimization of ODC Levels
Reaction Rate (mM NADH/min)
Rate = 0.020 AU/min
Rate = 0.0162 AU/min
Rate = 0.008 AU/min
Rate = 0.002 AU/min
Effect of Nitrogen Atmosphere
A
CD
B
AU
F i g . 1 . O p t i m i z a t i o n o f a s s a y c o n d i t i o n s . ( A ) Increase in background signal under normal atmosphere. The signal seen here in green is
solely from atmospheric CO
2 (orange = nitrogen atmosphere, no ornithine; green = standard atmosphere, no ornithine; blue = nitrogen
atmosphere, 625 μM ornithine; red = standard atmosphere, 625 μM ornithine). Quadruplicate data were collected in a 384-well microplate
as described in the presence of the indicated concentrations of ornithine. All other reagent concentrations were identical to the optimized
conditions described in Materials and Methods. The nitrogen atmosphere was maintained as described with O
2 levels under 2.5%. ( B )
Optimization of enzyme levels. = Z values, = reac tion r ates. Dashed line indicates st andar d Z cutoff value of 0.5. Da ta wer e collected
in a 384-well microplate using optimized assay conditions with varied fi nal ornithine decarboxylase (ODC) concentrations. Z Values were
calculated using 8 positive (1 mM difl uoromethylornithine (DFMO)) and 8 negative (DMSO) controls. The signal window at 150 nM ODC is
5-fold. ( C ) Determination of ODC K m at 150 nM ODC and 60 μM PLP. Data were collected in a 384-well plate as described and fi t to the
Michaelis–Menten equation. ( D ) Optimization of Infi nity CO
2 percentage ( = 60% Infi nity CO
2 , = 45% Infi nity CO
2 , = 30% Infi nity CO
2 ,
= 15% Infi nity CO
2 , = 7.5% Infi nity CO
2 ). Data were collected in 384-well plates as described at optimized assay conditions with varied
Infi nity CO
2 . Data points were taken every 15 s in quadruplicate. The plot of AU
340 vs. time at 60% Infi nity CO
2 represents a typical data set
under optimized assay conditions with a AU
340 of 0.4 AU.
adt.2009.0249.indd 179 4/19/2010 11:37:45 AM
SMITHSON ET AL.
180 ASSAY and Drug Development Technologies APRIL 2010
to test the effects of hits on the linking enzymes was designed.
Titration of exogenous sodium bicarbonate into the reaction mix-
ture showed a K
mapp of 0.47 ± 0.04 mM, which is consistent with
repor ted literature values for the K
m of PEPC from E. coli (0.1–0.3
mM)
24 ,
25 ( Fig. 2A ). A bicarbonate concentration of 1.5 × K
mapp
(0.75 mM) was chosen for testing inhibitors. A final concentration
of 60% Infinity CO
2 was used in order to mimic the primary assay
screening conditions. At these reagent concentrations, a linear
time of 7 min was observed with a AU of 0.8 ( Fig. 2B ). A signal
window of 26 was observed and a Z of 0.75 was calculated using
no bicarbonate reagent as the positive control.
Testing of Known Inhibitors and Selection of Positive
Controls
The only know n ODC inhibitor available in large enough quan-
tities for use as a HTS control compound is DFMO. At optimized
assay conditions in the ODC-PEPC-MDH-linked system, DFMO
exhibited an IC
50 of 200 ± 40 μM, which is consistent with the
literature K
iapp value of 160 μM ( Fig. 3A ).
26 No effect on the IC
50
value was seen when the amounts of link ing enzyme present were
changed. Additionally, it was possible to make 1 M stock solutions
of DFMO in DMSO that were stable for several months at room
temperature. Stock solutions at this concentration allowed effi-
cient pin transfer of DFMO into the control wells in nanoliter vol-
umes and permitted th is compound to be used as positive control
in screening at a final concentration of 1 mM. Inhibitors of the
linking enzymes were ineffective under nor mal screening condi-
tions ( Fig. 3B and 3C ). Neither baicalein, a PEPC inhibitor, nor
isoquinoline, a MDH inhibitor, had any effect at concentrations
up to 1 mM under optimized assay conditions, well past their
reported K
i values of 0.79 and 200 μM, respectively.
27 ,
28 However,
if the percentage of Infinity CO
2 was reduc ed , inh ib it ion by b aica-
le in beca me app ar en t, wi th IC
50 values approaching literature val-
ues. Isoquinoline was not observed to have any inhibitor y effects
under any conditions tested; t his is unsurprising given its poor
potency.
In the PEPC-MDH secondar y assay system, DFMO did not
have any effect below 10 mM concentrations. This inhibition is
the result of a pH decrease in the assay solution due to the fact
that the hydrochloride salt of DFMO was u sed to make inhibitor
stock solutions. Baicalein, the PEPC inhibitor, was not obser ved
to have any effect on the secondary assay at norma l screening
levels of Infinity CO
2 . However, as in the case of the ODC-PEPC-
MDH system, when linking enzyme levels were dropped, inhibi-
tion by baicalein was detected. Isoquinoline had no effect on the
2 enzyme system under any conditions tested.
Optimization of the High-Throughput Assay
Before perfor ming a ny screening, the stability of the assay
was tested using the fully automated system. Location-dependent
effects were measured across a microplate prepared using the
automated assay system. The only significant effect seen was a
slight increase in high signal for the outside columns ( Fig. 4A and
4B ). This did not affect Z values significantly due to t he fact that
negative control wells were placed on both interior and edge col-
umns (8 negative controls in columns 1 and 2, and 8 in columns
13 and 14). In order to
maximize the amount
of data gathered across
a wide range of variable
compounds, a 20-min
kinetic read with time
points every minute was
used during the initial
screening efforts.
Primary Screen
and Optimization
of Kinetic Fitting
Parameters
For the initial proof-
of-concept screen, the
bioactive collection
at St. Jude Children’s
Research Hospital (8,832
data points for 3,600
0
0.00
0.01
0.02
0.03
0.04
0.05
0.06
0.07
0.08
0.09
AB
123 4
Rate (mM NADH/min)
5
[HCO3(mM)]
6789 0
0.0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0.9
1.0
1.1
1.2
51015
Time (min)
Rate = 0.087 AU/min
Rate = 0.002 AU/min
AU
20 25
Km = 0.47 ± 0.04 mM
F i g . 2 . Optimization of PEPC-MDH assay system. Data were collected under nitrogen as described in Materials
and Methods. All data were collected under nitrogen in 384-well microplates. ( A ) Determination of the K m appar-
ent with respect to bicarbonate. The value obtained is consistent with literature values.
24,25 Data were fi t to the
Michaelis–Menten equation. ( B ) Determination of assay linear time ( = 0.75 mM HCO
3 , = no HCO
3 ). Under
optimized assay conditions, the bicarbonate signal was linear for 7 m i n .
adt.2009.0249.indd 180 4/19/2010 11:37:46 AM
DECARBOXYLASE ENZYME HTS
© MARY ANN LIEBERT, INC. • VOL . 8 NO. 2 • APRIL 2010 ASSAY and Drug Development Technologies 181
unique molecules in 23 384-well plates) was selected for testing.
Absolute replicate number for each compound varied from 1 to 7
depending on vendor library composition and plating. The com-
pounds were screened at 10 μM against the ODC-PEPC-MDH sys-
tem using a 20-min kinetic read. Eight posit ive controls (1 mM
DFMO) and 8 negative controls (DMSO) were placed in colum ns 1,
2 and 13, 14 to help detect possible edge effects. The scatter plot
for percent activ ity across the library is shown in Figure 4C . The
screening r un took 8.7 h to complete using our automated system.
We tested 3 different met hods for determining the assay rates
( Fig. 5 ). Fitting using the later portion of the data (time points
10–20) improved Z . The “delta” model performed similarly to the
linear model on average, albeit with slightly higher variance.
The “robust” model performed the best when all time points were
included, but fai led to converge without at least 15 time points.
The “ linea r” me thod using ti me points fr om 15 t o 20 min wa s c ho -
sen to calculate endpoint values as a compromise between assay
run time and Z .
0.1
25
0
25
50
75
100
ABC
% Inhibition
0
25
50
75
100
D
% Inhibition
0
25
50
75
100
E
% Inhibition
0
25
50
75
100
F
% Inhibition
25
25
0
25
50
75
100
% Inhibition
25
0
25
50
75
100
% Inhibition
1 10 100
[Inhibitor (μM)]
DFMO
ODC Inhibitor, Ki = 160 μM*
ODC-PEPC-MDH
DFMO
PEPC-MDH
Baicalein
PEPC-MDH
Isoquinoline
PEPC-MDH
Baicalein
PEPC Inhibitor, Ki = 0.79 μM**
ODC-PEPC-MDH
Isoquinoline
MDH Inhibitor, Ki = 200 μM***
ODC-PEPC-MDH
1,000 10,000
0.1 1 10 100
[Inhibitor (μM)]
1,000 10,000 0.10.01 1 10 100
[Inhibitor (μM)]
1,000 10,000 0.10.01 1 10 100
[Inhibitor (μM)]
1,000 10,000100,000
0.1 1 10 100
[Inhibitor (μM)]
1,000 0.1 1 10 100
[Inhibitor (μM)]
1,000 10,000
F i g . 3 . Performance of known inhibitors in the ODC-PEPC-MDH and PEPC-MDH assays ( = 60% Infi nity CO
2 , = 45% Infi nity CO
2 , =
30% Infi nity CO
2 , = 15% Infi nity CO
2 ). Data were collected under optimized screening conditions as described unless otherwise noted.
Compounds were allowed to equilibrate with enzymes for 20 min prior to addition of substrate. Percent inhibition is defi ned as (1 v i / v o ) ×
100. At Infi nity, CO
2 percentages lower than 45% reaction rates were calculated from the fi rst 10 min of the reaction rather than the normal
20 min used at higher percentages due to limitations in assay linear time (see Fig. 1D ). For PEPC-MDH assay results, rates were determined
from the fi rst 5 min (see Fig. 2B ). ( A ) Performance of difl uoromethylornithine (DFMO), a known ornithine decarboxylase (ODC) inhibitor, in
the ODC-PEPC-MDH assay with varying amount s of linking enzymes. Note that linking enzyme concentrations do not effect the per formance
of DFMO. ( B ) Performance of baicalein, a known inhibitor of PEPC, at several different fi nal linking mix concentrations in the ODC-PEPC-MDH
assay. At normal screening conditions (60% Infi nity CO
2 ) no inhibition is seen, even from this submicromolar inhibitor. However, upon dilu-
tion of the linking mix, inhibition becomes apparent . ( C ) Perfor mance of isoquinoline, a known inhibitor of MDH in the ODC-PEPC-MDH assay
at several Infi nity CO
2 percentages. No inhibition was seen in any condition tested. Reaction rates at Infi nity CO
2 concentrations lower than
45% were calculated from the fi rst 10 min of data due to limitations in assay linear time (see Fig. 1D ). ( D ) Performance of DFMO in the PEPC-
MDH assay system at varying levels of Infi nity CO
2 . ( E ) Performance of baicalein in the PEPC-MDH assay system at varying levels of Infi nity
CO
2 . As with the ODC-PEPC-MDH system, a dependency on the amount of linking enzymes present greatly effects the amount of inhibition
seen. ( F ) Performance of isoquinoline in the PEPC-MDH assay. No inhibition was observed under any conditions tested. *Literature value, 26
**Literature value,
27 ***Literature value. 28
adt.2009.0249.indd 181 4/19/2010 11:37:46 AM
SMITHSON ET AL.
182 ASSAY and Drug Development Technologies APRIL 2010
The overall data quality for t he 3-enzyme assay was satisfac-
tory, with an average Z value of 0.68 across all plates. The robust
outlier cutoff yielded 84 pri mary hits, of which 52 were unique.
Internal duplication of compounds afforded an assessment of
assay reproducibility. Active repli-
cates were detected 86% of the time
with an average activity CV of 16%,
and 97% of the time with an average
act ivity C V of on ly 3% when restrict-
ing replicates to those from the same
vendor.
Secondary Testing
To further characterize the hits,
aliquots of each unique sample were
cherry-picked and subjected to full
dose–response st udies consisting of
10 points in a 1:3 dilution series (top
= 100 μM). Thirty t hree of the initial
52 unique hits displayed a full dose
response while 15 displayed partial
dose responses in the ODC-PEPC-
MDH system—a 92% confirmation
rate. None of the samples signif i-
cantly affected the PEPC-MDH sys-
tem at concentrations up to 100 μM
at 60% Infinity CO
2 under optimized
assay conditions. To further confirm
that inhibition of ODC was the rate-
limiting step in the reaction cascade,
the samples were re-screened at 300
nM ODC, and the rates at the IC
50 were
compared to those determined at 150
nM ODC. A ll samples displayed the
predicted 2-fold increase at the higher
enzyme concentration, confirming
that inhibition of ODC was the rate-
limiting step. These compounds were
designated as true positives for the
ROC analysis.
ROC Analysis
ROC analysis provides an assess-
ment of the discriminatory power of
an assay that is nonparametric (ie,
does not assume Gaussian distribu-
tion) and that is independent of both
the number of true positives and the
threshold for classifying hits. The ROC AUC is the probabi lity
that the assay will ra nk a randomly chosen true positive a head
of a randomly chosen true negative. A random assay—one that
cannot discriminate true positives from tr ue negatives—has an
0
0.0000
0.0009
0.0019
Reaction Rate (mM NADH/min)
0.0029
0.0039
0.0049
A
C
0.0000
0.0009
0.0019
Reaction Rate (mM NADH/min)
0.0029
0.0039
0.0049
B
64 128 192
Well Number by Row
0
0
50
100
% Activity
150
2,000 4,000
Compounds in Order Screened
Scatterplot of High Throughput Data
6,000 8,000
256 320 384 0 64 128 192
Well Number by Column
256 320 384
F i g . 4 . ODC-PEPC-MDH-linked plate geographic effects and high-throughput data scatter plot.
Assays were performed in 384-well plates as described and rates were calculated from the fi nal
6 data points from a 20-min observation. = High signal (DMSO), = mid signal (200 μM
difl uoromethylornithine (DFMO)), = low signal (1 mM DFMO). ( A ) Column effects resulting in
a slight increase in signal on the outer columns. ( B ) Data from 3A arranged by row, showing that
signal across rows is constant. The signal window for the assay is also apparent in these fi gures
and is 4.5. ( C ) Data from the small-scale high-throughput assay of the bioactive compound col-
lection at St. Jude Children’s Research Hospital. Green circles = positive control (1 mM DFMO),
red circles = negative control (DMSO), blue circles = hit compound, black circles = inactive
compound. Magenta line = 99th percentile cutoff, orange line = 95th percentile cutoff. Percent
activity was calculated by normalizing kinetic reaction rates (20-min data collection with the
nal 6 data points fi t as described in Materials and Methods) to positive and negative controls.
The 8,832 total data points were collected from 3,600 unique compounds. Absolute replicate
number for each compound varied from 1 to 7, depending on vendor library composition and
plating. The assay took 8.7 h to complete using our automation system.
adt.2009.0249.indd 182 4/19/2010 11:37:47 AM
DECARBOXYLASE ENZYME HTS
© MARY ANN LIEBERT, INC. • VOL . 8 NO. 2 • APRIL 2010 ASSAY and Drug Development Technologies 183
AUC = 0.5. The ROC AUC for the ODC assay is 0.90 ( Fig. 6 , 0.82–
0.97, 95% confidence inter val). Setting the cutoff at >30% in the
primary screen would retu rn >70% of all t rue positives, while
yielding 10% of all true negatives.
Hit Compounds
Due to t he nature of the compound librar y, this proof-of-concept
screen was not expected to yield many well-behaved inhibitors w ith
drug-like scaffolds. Indeed, a la rge number of quinone-containing
compound s were identi fied, a long with se veral compou nd series con-
taining Michael acceptors and other reactive groups. Though these
compounds are not likely to be good lead candidates, they do inhibit
the enzyme and were useful in the validation of the assay proto-
col. ODC has an active site cysteine residue and is known to require
large amounts of reducing agent to be active in vitro . Therefore, it
would be expected to be sensitive to potential alkylators as well as
oxidizing compounds. ODC has historically been a difficult target
to inhibit, and it is likely that screening a larger compound library
wil l be necessary to find well-behaved inhibitors.
D I S C U S S I O N
This study represents the first effort to develop an assay suit-
able for screening decarbox ylases in a high-throughput manner.
Alt hough it is a linked enzy me assay, the lac k of susceptibility to
known inhibitors of the li nking enzymes suggests that relatively
few false positive hits will be identified due to inhibition of the
other enzymes in the mix. Baicalein, a known inhibitor of PEPC
with a K
i of 0.79 μM , did not affect the assay appreciably until the
linking enzyme mix had been diluted 2-fold and even then only
inh ibited the reaction by 50%. Isoquinoline, a know n inhibitor
of MDH with a literature K
i of 200 μM, did not i nhibit the assay
at any tested condition. Furthermore, none of the hits identified
at 10 μM in the primary screen affected the dual enzyme link-
ing system at concentrations up to 100 μM. Although the exact
concentrations of the link ing enzymes in the mi xture are not dis-
closed by the manufacturer, these observations suggest that high
concentrations of both PEPC and M DH are present, and that the
assay is extremely resistant to inhibitors of the link ing enzymes
under optimized conditions (60% Infinity CO
2 , 600 μM Orn, 60
μM PLP, and 0.01% Triton-X).
However, the know n ODC inhibitor, DFMO, exhibited an IC
50
value of 206 ± 40 μM af ter 20 min of preincubation, which is
consistent with the literature K
iapp of 160 μM. ROC analysis, based
on a definition of true positive that requires ODC specificity,
quantitatively demonstrates the high discriminatory power of
the assay. Moreover, the high degree of agreement found between
1
1
0.8
0.6
0.4
0.2
0
0.2
0.4
Z
0.6
0.8
Start (min)
End (min)
111 555
Non-Robust Linear Curve Fitting 2 Point Delta Values Robust Linear Curve Fitting
10 10 15
5 10 15 20 10 15 20 15 20 20 5 10 15 20 10 15 20 15 20 20 5 10 15 20 10 15 20 15 20 20
1111 5551010151111 5 55101015
F i g . 5 . Effect of var ying curve fi tting methods on average Z values for scaling screen. = Median, = mean, solid line = standard Z cutoff
of 0.5. Data were collected as described in Materials and Methods under optimized assay conditions. Time points were taken ever y minute
for 20 min. Note that the fi rst 5 to 10 min are poorly behaved and that eliminating them dramatically increases the average Z for the run.
This is primarily a result of a decrease in scatter rather than an increase in signal, since the average rates calculated were consistent for all
methods tested.
adt.2009.0249.indd 183 4/19/2010 11:37:49 AM
SMITHSON ET AL.
184 ASSAY and Drug Development Technologies APRIL 2010
replicate samples in the library f urt her underscores the robust-
ness of the assay system.
While the HTS assay requires a CO
2 -free atmosphere in order
to stabilize the reagents, other engineer ing controls may allevi-
ate this need. For example, sealed reagent bottles equipped with
nitrogen lines feeding bulk liquid dispensers would allow large
amounts of the Inf init y CO
2 reagent to be loaded into automated
systems without degrading. It shou ld be noted that performing
the assay under nor mal atmosphere decreases linear t ime from
60 to 40 min, meaning that timing in the screening run must
be monitore d close ly. T he effect of mov ing to norm al atmospheric
conditions for screening is primarily an increase in background
signal. This decreases the average Z values observed from 0.7
to 0.5, a typical lower limit for high-th roughput assay systems.
Additionally, since t his assay is based on absorbance values, it
cannot be used in an endpoint fashion due to absorbance inter-
ference from the test compounds. To compensate for t his, it is
necessary to use a kinetic read. However, throughput was max-
imized by using a short kinetic read as it minimized movement
of the plate, which is often rate limiting in high-t hroughput
screens on integrated systems. The throughput for t his assay is
10 plates per hou r using ou r automated systems, when using a
5-min kinetic read.
The relatively high prima ry hit rate of 1.4% ref lects both the
liberal primary hit cutoff in and the nature of the bioactive collec-
tion. It is likely that the h it rate would decline signif icantly for a
more diverse compound libra ry, particular ly one curated to r emove
electrophiles and redox-active compounds. The ident ification of
active scaffold series known to be redox-active compounds
and/or nonspecif ic alkylators highlights the need for second-
ary assays that will address both of these liabilities in any
true HTS-effort aimed at discovering novel inhibitors of ODC.
However, since none of these compounds were found to effect
the linking enzymes, this sensitiv ity is due to the specific
decarboxylase screened, not the assay met hod itself.
In conclusion, this study describes a quick and accurate
method to screen ODC and other decarbox ylases in high
throughput. T he system is robust, generates hits with the
desired biochemical effects, and is superior to ex isting assays
that rely on either radiolabeled substrate or HPLC methods.
A C K N O W L E D G M E N T S
This work was supported by the American Lebanese
Syrian Associated Charities (ALSAC) and St. Jude Children’s
Research Hospital (SJCRH). A portion of this research was
performed while on appoint ment as a U.S. Department of
Homeland Secur ity (DHS) Fellow under the DHS Scholarship
and Fellowship Program, a program administered by the Oak
Ridge Institute for Science and Education (ORISE) for DHS
through an interagency agreement with the U.S. Department of
Energ y. ORISE is managed by Oak Ridge Associated Universities
under DOE contract number DE-AC05-00OR22750. A ll opinions
expressed in the article are the author’s and do not necessari ly
ref lect the policies and views of DHS, DOE, or ORISE. Additional
funding provided by National Institutes of Health grants (R01
AI34432) (to M.A .P.), and the Welch Foundation grant I-1257 (to
M.A.P.).
REFERENCES
1 . C a s e r o R A , J r , M a r t o n L J : T a r g e t i n g p o l y a m i n e m e t a b o l i s m a n d f u n c t i o n i n c a n c e r
and other hyperproliferative diseases . Nat Rev Drug Discov 2 0 0 7 ; 6 : 3 7 3 3 9 0 .
2 . H e b y O , R o b e r t s S C , U l l m a n B : P o l y a m i n e b i o s y n t h e t i c e n z y m e s a s d r u g t a r g e t s
in parasitic protozoa . Biochem Soc Trans 2 0 0 3 ; 3 1 : 4 1 5 4 1 9 .
3 . M o y a - G a r c í a A A , P i n o - A n g e l e s A , G i l - R e d o n d o R , M o r r e a l e A , S á n c h e z - J i m é n e z
F : Structural features of mammalian histidine decarboxylase reveal the basis
for speci c inhibition . Br J Pharmacol 2 0 0 9 ; 1 5 7 : 4 1 3 .
4 . B o n i f a t i V , M e c o G : N e w , s e l e c t i v e c a t e c h o l - O - m e t h y l t r a n s f e r a s e i n h i b i t o r s a s
therapeutic agents in Parkinson’s disease . Pharmacol Ther 1 9 9 9 ; 8 1 : 1 3 6 .
5 . R a y S S , B o n a n n o J B , R a j a s h a n k a r K R , P i n h o M G , H e G , D e L e n c a s t r e H , et al . :
Cocrystal structures of diaminopimelate decarboxylase: mechanism, evo-
lution, and inhibition of an antibiotic resistance accessory factor . Structure
2 0 0 2 ; 1 0 : 1 4 9 9 1 5 0 8 .
6 . B i t o n t i A J , M c C a n n P P , S j o e r d s m a A : R e s t r i c t i o n o f b a c t e r i a l g r o w t h b y i n h i b i t i o n
of polyamine biosynthesis by using mono uoromethylornithine, di uorometh-
ylarginine and dicyclohexylammonium sulphate . Biochem J 1 9 8 2 ; 2 0 8 : 4 3 5 4 4 1 .
7 . T h y s s e n S M , L i b e r t u n C : Q u a n t i t a t i o n o f p o l y a m i n e s i n h y p o t h a l a m u s a n d p i t u -
itary of female and male developing rats . Neurosci Lett 2 0 0 2 ; 3 2 3 : 6 5 6 9 .
8 . O s t e r m a n A , G r i s h i n N V , K i n c h L N , P h i l l i p s M A : F o r m a t i o n o f f u n c t i o n a l c r o s s -
species heterodimers of ornithine decarboxylase . Biochemistry 1 9 9 4 ; 3 3 :
1366 2 – 13667 .
0.0
0.0 6.51
15.83
38.16
60.5
82.83
105.17
0.2
0.4
True Positive Rate
0.6
0.8
1.0
0.2 0.4
False Positive Rate
Heat Map by Single Point % Activity
ROC AUC = 0.90 (0.82–0.97)
0.6 0.8 1.0
F i g . 6 . Receiver operating characteristic (ROC) curve for the ornithine
decarboxylase (ODC) assay. The ROC area under the curve (AUC) is 0.90
(0.82– 0.97, 95% confi dence interval). Bootstrap simulation cur ves are in
light gray. The dotted line is the curve for a random assay.
adt.2009.0249.indd 184 4/19/2010 11:37:50 AM
DECARBOXYLASE ENZYME HTS
© MARY ANN LIEBERT, INC. • VOL . 8 NO. 2 • APRIL 2010 ASSAY and Drug Development Technologies 185
9 . W H O . African Trypanosomiasis (sleeping sickness) . http://www.who.int/media-
centre/factsheets/fs259/en/ (accessed November 26, 2007).
1 0 . R a y R M , Z i m m e r m a n B J , M c C o r m a c k S A , P a t e l T B , J o h n s o n L R : P o l y a m i n e
depletion arrests cell cycle and induces inhibitors p21(Waf1/Cip1), p27(Kip1),
and p53 in IEC-6 cells . Am J P hysiol 1 9 9 9 ; 2 7 6 : C 6 8 4 C 6 9 1 .
1 1 . C e l a n o P , B a y l i n S B , G i a r d i e l l o F M , N e l k i n B D , C a s e r o R A , J r : Effect of polyamine
depletion on c-myc expression in human colon carcinoma cells . J Biol Chem
1 9 8 8 ; 2 6 3 : 5 4 9 1 5 4 9 4 .
1 2 . I w a m i K , W a n g J Y , J a i n R , M c C o r m a c k S , J o h n s o n L R : I n t e s t i n a l o r n i -
thine decarboxylase: half-life and regulation by putrescine . Am J Physiol
1 9 9 0 ; 2 5 8 : G 3 0 8 G 3 1 5 .
1 3 . P h i l l i p s M A , C o f n o P , W a n g C C : C l o n i n g a n d s e q u e n c i n g o f t h e o r n i t h i n e d e c a r -
boxylas e gene from Trypanosoma bruc ei. Implications for enzyme turnover and
selective di uoromethylornithine inhibition . J Biol Chem 1 9 8 7 ; 2 6 2 : 8 7 2 1 8 7 2 7 .
1 4 . V a n N i e u w e n h o v e S , S c h e c h t e r P J , D e c l e r c q J , B o n é G , B u r k e J , S j o e rdsma
A : Treatment of gambiense sleeping sickness in the Sudan with oral DFMO
(DL-alpha-di uoromethylornithine), an inhibitor of ornithine decarboxylase;
rst  eld trial . Trans R Soc Trop Med Hyg 1 9 8 5 ; 7 9 : 6 9 2 6 9 8 .
1 5 . A b e l o f f M D , S l a v i k M , L u k G D , G r i f n C A , H e r m a n n J , B l a n c O , et al .: Phase I trial
and pharmacokinetic studies of alpha-di uoromethylornithine--an inhibitor of
polyamine biosynthesis . J Clin Oncol 1 9 8 4 ; 2 : 1 2 4 1 3 0 .
1 6 . B e y P , D a n z i n C , J u n g M : I n h i b i t i o n o f b a s i c a m i n o a c i d d e c a r b o x y l a s e s i n v o l v e d
in polyamine biosynthesis . In: Inhibition of Polyamine Metabolism—Biological
Signi cance and Basis for New Therapies ( M c C a n n P P , P e g g A E , S j o e r d s m a A ,
e d s . ) , p p . 1 3 1 . A c a d e m i c P r e s s , S a n D i e g o , 1 9 8 7 .
1 7 . A m a d a s i A , B e r t o l d i M , C o n t e s t a b i l e R , B e t t a t i S , C e l l i n i B , d i S a l v o M L , et al . :
Pyridoxal 5’-phosphate enzymes as targets for therapeutic agents . Curr Med
Chem 2 0 0 7 ; 1 4 : 1 2 9 1 1 3 2 4 .
1 8 . T e a m R D C : R: A Language and Environment for Statistical Computing . R
Foundation for Statistical Computing , Vienna , 2007 .
1 9 . P e t e r R o u s s e e u w C C , T o d o r o v V , R u c k s t u h l A , S a l i b i a n - B a r r e r a M , M a e c h l e r M :
Robustbase:Basic Robust Statistics R package 0.2-7; 2006.
2 0 . H o a g l i n D C , M o s t e l l e r F , T u k e y J W : Understanding Robust and Ex ploratory Data
Analysis . John Wiley and Sons, Inc. , New York , 1983 .
2 1 . O s t e r m a n A L , B r o o k s H B , R i z o J , P h i l l i p s M A : R o l e o f A r g - 2 7 7 i n t h e b i n d i n g
of pyridoxal 5-phosphate to Trypanosoma brucei ornithine decarboxylase .
Biochemistry 1 9 9 7 ; 3 6 : 4 5 5 8 4 5 6 7 .
2 2 . M y e r s D P , J a c k s o n L K , I p e V G , M u r p h y G E , P h i l l i p s M A : L o n g - r a n g e i nteractions
in the dimer interface of ornithine decarboxylase are important for enzyme
f u n c t i o n . Biochemistry 2 0 0 1 ; 4 0 : 1 3 2 3 0 1 3 2 3 6 .
2 3 . J a c k s o n L K , B a l d w i n J , A k e l l a R , G o l d s m i t h E J , P h i l l i p s M A : M u l t i p le active site
conformations revealed by distant site mutation in ornithine decarbox ylase .
Biochemistry 2 0 0 4 ; 4 3 : 1 2 9 9 0 1 2 9 9 9 .
2 4 . T e r a d a K , I z u i K : S i t e - d i r e c t e d m u t a g e n e s i s o f t h e c o n s e r v e d h i s t i d i n e r e s i d u e
of phosphoenolpyruvate carbox ylase. His138 is essential for the second partial
r e a c t i o n . Eur J Biochem 1 9 9 1 ; 2 0 2 : 7 9 7 8 0 3 .
2 5 . Y a n o M , T e r a d a K , U m i j i K , I z u i K : C a t a l y t i c r o l e o f a n a r g i n i n e r e s i d u e i n the
highly conserved and unique sequence of phosphoenolpyruvate carboxylase . J
Biochem 1 9 9 5 ; 1 1 7 : 1 1 9 6 1 2 0 0 .
2 6 . B i t o n t i A J , B a c c h i C J , M c C a n n P P , S j o e r d s m a A : C a t a l y t i c i r r e v e r s i b l e i n h i b i tion
of Trypanosoma brucei brucei ornithine decarboxylase by substrate and prod-
uct analogs and their effects on murine trypanosomiasis . Biochem Pharmacol
1 9 8 5 ; 3 4 : 1 7 7 3 1 7 7 7 .
2 7 . P a i r o b a C F , C o l o m b o S L , A n d r e o C S : F l a v o n o i d s a s i n h i b i t o r s o f N A D P - m a l i c
enzyme and PEP carboxylase from C4 plants . Biosci Biotechnol Biochem
1 9 9 6 ; 6 0 : 7 7 9 7 8 3 .
2 8 . K a p p E , W h i t e l e y C : P r o t e i n l i g a n d i n t e r a c t i o n s : i s o q u i n o l i n e a l k a l o i d s a s i n h i b i -
tors for lactate and malate dehydrogenase . J En zym Inhib 1 9 9 1 ; 4 : 2 3 3 2 4 3 .
Address correspondence to:
Dr. R. Kiplin Guy
Department of Chemical Biology and Therapeutics
St. Jude Children’s Research Hospital
262 Danny Thomas Place
Memphis, TN 38105
E-mail: kip.guy@stjude.org
adt.2009.0249.indd 185 4/19/2010 11:37:51 AM
... It can be performed in an automated system with 384-well microplates. It has already been used to analyze more than 3,000 compounds Need a commercial system for bicarbonate detection and N 2 atmosphere for analysis Scriven et al. (1988), Smithson et al. (2010b), Liao et al. (2015), Chai et al. (2020), Smithson et al. (2010a) The cost of the kit can make analysis by HTS unfeasible Putrescine oxidation: ...
... The presence of CO 2 in the reaction system leads to a decrease in light absorbance, used to assess the production of CO 2 (Scriven et al., 1988). These reactions can be performed using commercial kits, standardized for CO 2 detection (Smithson et al., 2010b;Liao et al., 2015;Chai et al., 2020). This assay allows the monitoring of the reaction in real-time and is considered by some authors as a good candidate for adaptation to HTS (Smithson et al., 2010b;Chai et al., 2020). ...
... These reactions can be performed using commercial kits, standardized for CO 2 detection (Smithson et al., 2010b;Liao et al., 2015;Chai et al., 2020). This assay allows the monitoring of the reaction in real-time and is considered by some authors as a good candidate for adaptation to HTS (Smithson et al., 2010b;Chai et al., 2020). Smithson et al. (Smithson et al., 2010b), optimized this assay using a commercial system for the detection of bicarbonate, carried out the experiments in N 2 atmosphere, performed the analyzes in an automated system in 384-well microplates, and optimized all the conditions reactions to evaluate 3,600 compounds. ...
Article
Full-text available
Ornithine decarboxylase (ODC) catalyzes the decarboxylation of ornithine to produce putrescine, the first step in the metabolism of polyamines (putrescine, spermidine, and spermine), which are essential growth factors in eukaryotic cells. ODC is active as a homodimer and depends on pyridoxal 5′-phosphate (PLP) as a cofactor. An increase in the concentration of polyamines has been associated with carcinogenesis. Therefore, there is much interest in identifying inhibitors of this pathway as potential chemotherapeutic and chemopreventive agents. The best-known inhibitor of mammalian ODC is α -difluoromethylornithine (DFMO), a highly selective compound that alkylates Cys-360 (a residue of the ODC active site). Although DFMO was initially developed for the treatment of cancer, the World Health Organization recommends its use in combination with nifurtimox for the treatment of human African trypanosomiasis. Considering the importance of ODC as a promising target for the treatment of various types of cancer and other infectious diseases, choosing the right method for screening potential inhibitors can help to accelerate the discovery of new drugs. Several methods for the determination of ODC activity are found in the literature. Among these, we can mention analysis with radioactive markers, colorimetric assays using auxiliary enzymes to detect CO 2 or H 2 O 2 release, chromatographic separations with putrescine derivatization, mass spectrometry, and spectroscopic techniques. In this review, the main analysis methods used will be described, highlighting their advantages and disadvantages, as well as identifying the most promising methods for high-throughput screening.
... As an enzyme family, decarboxylases have been challenging for the development of facile generic screens amenable for high throughput screening (23). One approach to developing screens for PSDs is to use agents that selectively react with the PE product of the enzyme reaction. ...
... The fluorescence assay for PSD provides an important new gateway for application in high throughput screening platforms. Large scale screening assays for decarboxylase enzymes have been generally challenging (23). One approach has been to couple CO 2 production by decarboxylases to enzymes that fix CO 2 to substrates (e.g. ...
... phosphoenolpyruvate carboxylase), and are further coupled to subsequent enzyme reactions involved in the oxidation of NADH (e.g. malate dehydrogenase) (23). This latter approach also requires screening in environments where ambient CO 2 levels are greatly reduced. ...
Article
Full-text available
Phosphatidylserine decarboxylases (PSDs) are central enzymes in phospholipid metabolism that produce phosphatidylethanolamine (PE) in bacteria, protists, plants and animals. We developed a fluorescence-based assay for selectively monitoring production of PE, in reactions using a maltose binding protein fusion with Plasmodium knowlesi PSD (MBP-His6-∆34PkPSD) as the enzyme. The PE detection by fluorescence (λex = 403 nm, λem = 508 nm ) occurred after the lipid reacted with a water soluble distyrylbenzene-bis-aldehyde (DSB-3), and provided strong discrimination against the PS substrate. The reaction conditions were optimized for enzyme, substrate, product and DSB-3 concentrations with the purified enzyme and also tested with crude extracts and membrane fractions from bacteria and yeast. The assay is readily amenable to application in 96-well and 384-well microtiter plates and should prove useful for high throughput screening for inhibitors of PSD enzymes across diverse phyla.
... S4, S5, S6) we found even a SP run with a robust Z′ factor alone could not provide a precise inhibition cutoff for selecting putative hits for confirmation. To better understand how discovery rates were affected by different cutoffs, the SP and DR data were subjected to Receiver-Operator Characteristic (ROC) analyses 79 . As noted above, toxic compounds often resulted in ambiguous activity at toxic doses. ...
Article
Full-text available
Improved control of Plasmodium vivax malaria can be achieved with the discovery of new antimalarials with radical cure efficacy, including prevention of relapse caused by hypnozoites residing in the liver of patients. We screened several compound libraries against P. vivax liver stages, including 1565 compounds against mature hypnozoites, resulting in one drug-like and several probe-like hits useful for investigating hypnozoite biology. Primaquine and tafenoquine, administered in combination with chloroquine, are currently the only FDA-approved antimalarials for radical cure, yet their activity against mature P. vivax hypnozoites has not yet been demonstrated in vitro. By developing an extended assay, we show both drugs are individually hypnozonticidal and made more potent when partnered with chloroquine, similar to clinically relevant combinations. Post-hoc analyses of screening data revealed excellent performance of ionophore controls and the high quality of single point assays, demonstrating a platform able to support screening of greater compound numbers. A comparison of P. vivax liver stage activity data with that of the P. cynomolgi blood, P. falciparum blood, and P. berghei liver stages reveals overlap in schizonticidal but not hypnozonticidal activity, indicating that the delivery of new radical curative agents killing P. vivax hypnozoites requires an independent and focused drug development test cascade.
... Kinetic calculations were made on data fit to a linear model, and the slope of this fit was taken as the rate of the reaction to obtain mean velocity V (absorbance units (AU)/min) using the automatic function of the plate reader. Reaction rate was transformed from AU/min to mM NADH/min using an extinction coefficient of 6.349 AU/mM/cm for NADH (66) and an approximate path length of 0.58 cm for assays performed in 200 µl final volume in a Corning Costar flat bottom 96-well plate. ...
Article
Full-text available
Polyamines are fundamental molecules of life, and their deep evolutionary history is reflected in extensive biosynthetic diversification. The polyamines putrescine, agmatine and cadaverine, are produced by pyridoxal 5'-phosphate-dependent L-ornithine, L-arginine and L-lysine decarboxylases (ODC, ADC, LDC), respectively, from both the alanine racemase (AR) and aspartate aminotransferase (AAT) folds. Two homologous forms of AAT-fold decarboxylase are present in bacteria: an ancestral form and a derived, acid-inducible extended form containing an N-terminal fusion to the receiver-like domain of a bacterial response regulator. Only ADC was known from the ancestral form, and limited to the Firmicutes phylum, whereas extended forms of ADC, ODC and LDC are present in Proteobacteria and Firmicutes. Here, we report the discovery of ancestral form ODC, LDC, and bifunctional O/LDC, and extend the phylogenetic diversity of functionally characterized ancestral ADC, ODC and LDC to include phyla Fusobacteria, Caldiserica, Nitrospirae and Euryarchaeota. Using purified recombinant enzymes, we show that these ancestral forms have a nascent ability to decarboxylate kinetically less preferred amino acid substrates with low efficiency, and that product inhibition primarily affects preferred substrates. We also note a correlation between the presence of ancestral ODC and ornithine/arginine auxotrophy, and link this with a known symbiotic dependence on exogenous ornithine produced by species using the arginine deiminase system. Finally, we show that ADC, ODC and LDC activities emerged independently, in parallel, in the homologous AAT-fold ancestral and extended forms. The emergence of the same ODC, ADC, and LDC activities in the non-homologous AR-fold, suggests that polyamine biosynthesis may be inevitable.
... To better understand how discovery rates were affected by different cutoffs, the SP and DR data were subjected to Receiver-Operator Characteristic (ROC) analyses 46 . As noted above, toxic compounds often resulted in ambiguous activity at toxic doses. ...
Preprint
Full-text available
Improved control of malaria caused by Plasmodium vivax can be achieved with the discovery of new antimalarials with radical cure efficacy, including prevention of relapse caused by hypnozoites residing in the liver of vivax patients. We screened several compound libraries against P. vivax liver stages, including 1566 compounds against mature hypnozoites, resulting in one drug-like and several probe-like hits useful for investigating hypnozoite biology. Primaquine and tafenoquine, administered in combination with chloroquine, are currently the only FDA-approved antimalarials for radical cure, yet their activity against mature P. vivax hypnozoites has not yet been demonstrated in vitro. By developing an extended assay, we show both drugs are individually hypnozonticidal and made more potent when partnered with chloroquine, similar to clinically-relevant combinations. Post-hoc analyses of screening data revealed excellent performance of ionophore controls and the high quality of single point assays, demonstrating a platform able to support the screening of greater compound numbers. A comparison of P. vivax liver stage activity data with that of the P. cynomolgi blood, P. falciparum blood, and P. berghei liver stages reveals little activity overlap, indicating that the delivery of new radical curative agents killing P. vivax hypnozoites requires an independent, focused drug development test cascade.
... ODC1, or ornithine decarboxylase, is frequently upregulated in cancer as a direct target of Myc-driven transcription, is a promoter of polyamine synthesis-dependent proliferation in a variety of settings, and is one of the most tightly regulated enzymes in the human proteome (Agarwal, Katiyar, Zaidi, & Mukhtar, 1992;Funakoshi-Tago, Sumi, Kasahara, & Tago, 2013;He et al., 2017;Hogarty et al., 2008;Jang et al., 2017;Kim et al., 2017;Mangold & Leberer, 2005;Mohammed et al., 2014;Patel, Loboda, Showe, Showe, & McMahon, 2004;Pegg, 2006;Singh et al., 2018). While there are long-standing, low-throughput assays for endogenous ODC1 activity (Gupta, Ahmad, Mohan, Husain, & Mukhtar, 1999;Verma & Boutwell, 1977;Verma, Shapas, Rice, & Boutwell, 1979), more recent approaches for ligand discovery in the literature focus on either the active site of the enzyme (Smithson, Shelat, Baldwin, Phillips, & Guy, 2010) or on molecular docking studies in silico (Kim et al., 2016). Given the attractiveness of array-based platforms for novel ligand discovery mentioned earlier and the short half-life of this therapeutically relevant protein, we present to the reader a protocol for screening ODC1 by SMM as an illustrative screen for the method. ...
Chapter
Many promising therapeutic protein targets were previously considered “undruggable” due to a deficit in structural information to guide drug design and/or a lack of an obvious binding pocket. Fortunately, array-based methods for evaluating protein binding against large chemical libraries, such as small-molecule microarray screening, have provided one of several emerging inroads to ligand discovery for these elusive targets. Despite the advance in the area of ligand discovery for poorly structured and intrinsically disordered proteins provided by array-based technologies involving cell lysates, the extension of this technology for screening proteins with short half-lives in physiologically relevant conformations has been technically challenging. In this chapter we present a protocol for leveraging in vitro translation strategies to enable array-based screening of short-lived proteins against large small-molecule libraries for ligand discovery.
... We previously described an alternative decarboxylase assay that enzymatically couples CO 2 production to NADH oxidation, and this assay was optimized for HTS and used for the identification of T. brucei ODC inhibitors. 29 However, in preliminary work we found this assay was unsuitable for detection of AdoMetDC activity in HTS format due to low signal-to-background ratio. Thus, a new HTS-compatible assay for AdoMetDC was required. ...
Article
Human African trypanosomiasis (HAT) is a fatal infectious disease caused by the eukaryotic pathogen Trypanosoma brucei. Available treatments are difficult to administer and have significant safety issues. S-Adenosylmethionine decarboxylase (AdoMetDC) is an essential enzyme in the parasite polyamine biosynthetic pathway. Previous attempts to develop TbAdoMetDC inhibitors into anti-HAT therapies failed due to poor brain exposure. Here, we describe a large screening campaign of two small-molecule libraries (~400,000 compounds) employing a new high-throughput (~7 s per sample) mass spectrometry-based assay for AdoMetDC activity. As a result of primary screening, followed by hit confirmation and validation, we identified 13 new classes of reversible TbAdoMetDC inhibitors with low-micromolar potency (IC50) against both TbAdoMetDC and T. brucei parasite cells. The majority of these compounds were >10-fold selective against the human enzyme. Importantly, compounds from four classes demonstrated high propensity to cross the blood-brain barrier in a cell monolayer assay. Biochemical analysis demonstrated that compounds from eight classes inhibited intracellular TbAdoMetDC in the parasite, though evidence for a secondary off-target component was also present. The discovery of several new TbAdoMetDC inhibitor chemotypes provides new hits for lead optimization programs aimed to deliver a novel treatment for HAT.
Article
S-Adenosylmethionine decarboxylase (AdoMetDC/SpeD) is a key polyamine biosynthetic enzyme required for conversion of putrescine to spermidine. Autocatalytic self-processing of the AdoMetDC/SpeD proenzyme generates a pyruvoyl cofactor from an internal serine. Recently, we discovered that diverse bacteriophages encode AdoMetDC/SpeD homologs that lack AdoMetDC activity and instead decarboxylate L-ornithine or L-arginine. We reasoned that neofunctionalized AdoMetDC/SpeD homologs were unlikely to have emerged in bacteriophages and were probably acquired from ancestral bacterial hosts. To test this hypothesis, we sought to identify candidate AdoMetDC/SpeD homologs encoding L-ornithine and L-arginine decarboxylases in bacteria and archaea. We searched for the anomalous presence of AdoMetDC/SpeD homologs in the absence of its obligatory partner enzyme spermidine synthase, or the presence of two AdoMetDC/SpeD homologs encoded in the same genome. Biochemical characterization of candidate neofunctionalized genes confirmed lack of AdoMetDC activity, and functional presence of L-ornithine or L-arginine decarboxylase activity in proteins from phyla Actinomycetota, Armatimonadota, Planctomycetota, Melainabacteria, Perigrinibacteria, Atribacteria, Chloroflexota, Sumerlaeota, Omnitrophota, Lentisphaerota and Euryarchaeota, the bacterial candidate phyla radiation and DPANN archaea, and the δ-Proteobacteria class. Phylogenetic analysis indicated that L-arginine decarboxylases emerged at least three times from AdoMetDC/SpeD, whereas L-ornithine decarboxylases arose only once, potentially from the AdoMetDC/SpeD-derived L-arginine decarboxylases, revealing unsuspected polyamine metabolic plasticity. Horizontal transfer of the neofunctionalized genes appears to be the more prevalent mode of dissemination. We identified fusion proteins of bona fide AdoMetDC/SpeD with homologous L-ornithine decarboxylases that possess two, unprecedented internal protein-derived pyruvoyl cofactors. These fusion proteins suggest a plausible model for the evolution of the eukaryotic AdoMetDC.
Chapter
Malaria, trypanosomiasis, and leishmaniasis are life-threatening parasitic diseases for which safe and more effective medicines are urgently needed. The polyamine biosynthetic pathway in protozoan pathogens is a validated target for the development of drugs, as demonstrated by the current use of a-difluoromethylornithine (DFMO) to treat patients diagnosed with human African trypanosomias (HAT). These parasites have evolved novel polyamine metabolic pathways that are considerably different from those of the human host. For example, trypanosomes contain a novel spermidine/glutathione conjugate termed trypanothione, and uniquely in these parasites S -adenosylmethionine decarboxylase (AdoMetDC) is activated by heterodimer formation with a catalytically dead homologue. In Plasmodium parasites, AdoMetDC and ODC are fused as a bifunctional protein. Trypanosoma cruzi lacks ODC and relies on polyamine transporters. In this chapter, we discuss these differences and highlight additional aspects of these organisms that could be exploited as potential therapeutic strategies, including differences in protein turnover rates and polyamine transport, the latter of which could be used for delivery of cytotoxic compounds.
Article
Tools and approaches for understanding the essentiality of kinase targets in Trypanosoma brucei that causes HAT, T. cruzi that causes Chagas disease, and Leishmania spp. that cause the various types of leishmaniasis are reviewed. Parasite kinase inhibitor discovery has employed two complementary approaches, target-based, which focuses on selective inhibition of specific essential parasite kinases, and phenotype-based, which focuses on inhibition of cellular growth or other observable phenotypes that result from treatment with an investigational agent. Combined target and cell-based approaches during drug optimization are useful for avoiding the apparent loss of potency that is typical when a compound's activity is compared between biochemical and cellular assays. This can reflect characteristics of cellular permeability, competition with the inhibitor by cellular molecules such as ATP, and/or compensation by targets or pathways. Thus, driving medicinal chemistry optimization in a cellular assay factors in cellular permeability and provides a physiologically relevant system for testing the inhibitor.
Article
Full-text available
Ornithine decarboxylase of the African trypanosome Trypanosoma brucei brucei had an estimated native molecular weight of 100,000 by gel filtration and a subunit molecular weight of 45,000 by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. The gene encoding this enzyme, present in a single copy in T. brucei, was identified by mouse ornithine decarboxylase cDNA under relatively stringent conditions of hybridization and subcloned in a 5.9-kilobase (kb) SstI fragment from a cosmid clone into the plasmid pUC 19. This clone encompassed a 2.8-kb SstII fragment that contained the entire T. brucei ornithine decarboxylase gene. The 2.8-kb SstII fragment hybridized to a 2.4-kb mRNA that presumably encodes the parasite enzyme. The 2.8-kb SstII fragment was partially sequenced and found to contain an open reading frame of 445 amino acids that has 61.5% homology with the corresponding sequence of the mouse enzyme. The only major discrepancies between the two enzymes are the addition of a 20-amino acid N-terminal peptide and the deletion of a 36-amino acid C-terminal peptide and the T. brucei ornithine decarboxylase. The C terminus has been postulated to be one of the structural factors associated with rapid in vivo turnover of mammalian ornithine decarboxylase. The absence of this C-terminal peptide in T. brucei ornithine decarboxylase predicts a slow turnover for the parasite enzyme in vivo, and this is supported by our experimental data. The lack of turnover of ornithine decarboxylase in trypanosomes may constitute the basis of selective antitrypanosomal action of the irreversible enzyme inhibitor DL-alpha-difluoromethylornithine.
Article
Cocrystal structures of Methanococcus jannaschii diaminopimelate decarboxylase (DAPDC) bound to a substrate analog, azelaic acid, and its L-lysine product have been determined at 2.6 Å and 2.0 Å, respectively. This PLP-dependent enzyme is responsible for the final step of L-lysine biosynthesis in bacteria and plays a role in β-lactam antibiotic resistance in Staphylococcus aureus. Substrate specificity derives from recognition of the L-chiral center of diaminopimelate and a system of ionic “molecular rulers” that dictate substrate length. A coupled-enzyme assay system permitted measurement of kinetic parameters for recombinant DAPDCs and inhibition constants (Ki) for azelaic acid (89 μM) and other substrate analogs. Implications for rational design of broad-spectrum antimicrobial agents targeted against DAPDCs of drug-resistant strains of bacterial pathogens, such as Staphylococcus aureus, are discussed.
Conference Paper
Recursive spatial domain filtering is often used in image restoration. The techniques range from deterministic inverse filter algorithms [1] to stochastic Kalman filters [2,3]. However, all the techniques have in common the problem of choosing the appropriate boundary values. It is the purpose of this paper to demonstrate the importance of the boundary values in image restoration and to show how we can improve the transient response of the steady-state Kalman filters in [2] and [3] with better choices for the boundary values.
Article
Levodopa remains the most effective drug for Parkinson’s disease (PD). However, its benefits are limited owing to extensive metabolism by catechol-O-methyltransferase (COMT), especially if levodopa is used in combination with peripheral dopa-decarboxylase inhibitors. A new generation of potent, orally active, selective, and reversible COMT inhibitors has become available recently. Among these, tolcapone and entacapone have been best characterised. Preclinical and clinical studies have shown that COMT inhibitors markedly enhance levodopa availability and prolong its plasma half-life. In recent large clinical trials they proved to be able to ameliorate motor fluctuations, reduce disability, and decrease levodopa requirements in PD patients. The tolerability profiles of entacapone and tolcapone are good. COMT inhibition promises to become an important means of extending the benefits of levodopa therapy in PD.
Article
For a long time the structural and molecular features of mammalian histidine decarboxylase (EC 4.1.1.22), the enzyme that produces histamine, have evaded characterization. We overcome the experimental problems for the study of this enzyme by using a computer-based modelling and simulation approach, and have now the conditions to use histidine decarboxylase as a target in histamine pharmacology. In this review, we present the recent (last 5 years) advances in the structure-function relationship of histidine decarboxylase and the strategy for the discovery of new drugs.
Article
Histidine residues have previously been suggested to be essential for the activity of phosphoenolpyruvate carboxylase as demonstrated by chemical modification of these residues. Although the location of these residues on the primary structure is not known, a comparison of nine phosphoenolpyruvate (P-pyruvate) carboxylases sequenced recently revealed that there are only two conserved histidine residues (His138 and His579, coordinates from the E. coli enzyme). Site-directed mutagenesis of these residues were undertaken with the E. coli P-pyruvate carboxylase and the properties of purified mutant enzymes were investigated. Mutation of His138 to asparagine (H138N) produced a protein which did not show carboxylase activity. However, this mutant enzyme catalyzed the bicarbonate-dependent dephosphorylation (Vmax = 1.4 mumol.min-1.mg-1) of the P-pyruvate. Since this reaction is due to one of the two partial reactions proposed for this enzyme, the results indicate that His138 is obligatory for the second-step reaction, i.e. the carboxylation of the enolate form of pyruvate by carboxyphosphate. Mutation of His579 to asparagine (H579N) produced an enzyme which had 69% of the wild-type carboxylase activity, but its affinity for P-pyruvate was decreased by 24-fold.
Article
Kinetic analysis has shown that isoquinoline, papaverine and berberine act as reversible competitive inhibitors to muscle lactate dehydrogenase and mitochondrial malate dehydrogenase with respect to the coenzyme NADH. The inhibitor constants Ki vary from 7.5 microM and 12.6 microM berberine interaction with malate dehydrogenase and lactate dehydrogenase respectively to 91.4 microM and 196.4 microM with papaverine action on these two enzymes. Isoquinoline was a poor inhibitor with Ki values of 200 microM (MDH) to 425 microM (LDH). No inhibition was observed for both enzymes in terms of their respective second substrate (oxaloacetic acid - malate dehydrogenase; pyruvate - lactate dehydrogenase). A fluorimetric analysis of the binding of the three alkaloids show that the dissociation constants (Kd) for malate dehydrogenase are 2.8 microM (berberine), 46 microM (papaverine) and 86 microM (isoquinoline); the corresponding values for lactate dehydrogenase are 3.1 microM, 52 microM and 114 microM. In all cases the number of binding sites averaged at 2 (MDH) and 4 (LDH). The binding of the alkaloids takes place at sites close to the coenzyme binding site. No conformational non equivalence of subunits is evident.
Article
Ornithine decarboxylase (ODC) is the primary rate-limiting enzyme for polyamine synthesis. ODC levels are increased in most tissues, including the intestinal mucosa, by growth-promoting agents. This enzyme has a brief half-life of from 5 to 30 min in mammalian tissues and is regulated by its product; putrescine. The current study examines the turnover and regulation of ODC in the mucosa of the small intestine. With the use of scraped intestinal mucosa from cycloheximide-treated rats, the time course of the decline in ODC activity yielded a half-life of approximately 22 min. Labeling enzyme protein with [3H]difluoromethylornithine (DFMO) resulted in a nearly identical estimation of half-life. ODC activity of mucosa from isolated gut segments stimulated by luminal glycine (0.1-0.4 M) was enhanced 60-100% by 10 mM putrescine administered luminally. Putrescine alone had no effect on ODC. In contrast, 10(-7) M putrescine prevented 80% of the ODC activity stimulated by asparagine in IEC-6 cells (a rat intestinal crypt cell line). The half-life of ODC in unstimulated IEC-6 cells was 20 min and increased to 35 min in cells exposed to 10 mM asparagine. These data demonstrate that ODC of nonproliferating villous cells is regulated differently from the identical enzyme in proliferating crypt cells. Therefore, conclusions regarding mucosal growth should not be based totally on ODC activity from whole mucosa, since it is essentially a measure of only the enzyme present in the villous cells.