SlideShare a Scribd company logo
1 of 8
Download to read offline
Recent advances in enzyme assays
Jean-Philippe Goddard and Jean-Louis Reymond
Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
Enzyme assays for high-throughput screening and
enzyme engineering, which are often based on deriva-
tives of coumarin, nitrophenol, fluorescein, nitrobenzo-
furazane or rhodamine dyes, can be divided into two
categories: those that depend on labelled substrates,
and those that depend on sensing the reactions of
unmodified substrates. Labelled substrates include, for
example, fluorogenic and chromogenic substrates that
generate a reporter molecule by b-elimination, fluor-
escence resonance energy transfer (FRET) substrates
and isotopic labels for enantioselectivity screening. By
contrast, endpoint sensing can be done using amine
reagents, fluorescent affinity labels for phosphorylated
proteins, or synthetic multifunctional pores. Sensing
assays can also be done in real time by using, for
example, aldehyde trapping to follow vinyl ester acyla-
tion in organic solvent or calcein–copper fluorescence
for sensing amino acids. The current trend is to assem-
ble many such assays in parallel for enzyme profiling
and enzyme fingerprinting.
Enzyme assays are experimental protocols that make
enzyme-catalysed chemical transformations visible. Such
assays are very important for high-throughput screening
in the context of drug discovery. Recently, much effort has
been directed at improving enzyme assays for enzyme
engineering, where they provide the functional basis for
identifying and selecting new enzymes, most often in
screens of either large sample libraries, such as micro-
organism collections from the biosphere or a series of
enzyme mutants generated by genetic recombination
methods, such as gene shuffling and error-prone PCR
[1–6]. The well-known adage ‘you get what you screen for’
clearly states what assays are all about: they must closely
reproduce the desired catalytic reaction.
An enzymatic transformation can be traced easily if the
enzyme is highly active and present in large amounts. In
most applications, however, the aim is the identification of a
dilute, and therefore weak, enzymatic activity in a reaction
medium that contains many other components. In addition
tobeingcrucialforidentifyingfunction,detectinganenzyme
by means of its catalytic action on a substrate is actually
very advantageous in terms of sensitivity, because a single
enzyme molecule can generate many product molecules
through enzymatic turnover. Thus, a signal amplification
effect is intrinsically present in any enzyme assay that is
based on substrate turnover. A good fluorogenic or chromo-
genic assay can often detect an enzyme below the protein
detection limit and even in a mixture of other proteins and
enzymes, such as a crude bacterial lysate.
For whichever application they are designed, enzyme
assays should be simple and robust. Technical parameters
must be fulfilled, such as a low rate of false-positive and
false-negative signals under the real assay conditions.
Even if these criteria are fulfilled, whether an assay will be
used or not is decided by the availability of the assay
reagents, their price, and their overall simplicity.
Here we review recent progress that has been made in
enzyme assays; we refer the reader to earlier reviews for
a more comprehensive coverage of the field [7–12]. The
examples cover a broad spectrum of possibilities that can
be exploited to build enzyme assays. We have classified
enzyme assays according to the nature of the substrate
used. We distinguish between enzyme assays based on
substrates that have been modified with a label and those
using sensors that can operate with natural, unmodified
substrates. This distinction makes a lot of sense in the
perspective of practical application, because very often a
sensor-based system does not require any synthesis and
turns out to be cheaper and more versatile. Nevertheless,
assays based on labelled substrates can be more sensitive
to low enzyme turnover and more selective.
We emphasize assays that detect enzymatic turnover,
but there is another technique for identifying enzymes
that is based on covalent labelling by active-site-directed
probes, followed by separation by gel electrophoresis. This
type of analysis is relevant to proteome analysis and has
been reviewed recently [13].
Assays with labelled substrates
Enzyme assays using labelled synthetic substrates are
advantageous in that they usually provide a very direct
connection between enzymatic activity and the signal.
Such assays are resistant to artefacts, in particular in the
context of enzyme inhibition assays. The use of labelled
substrates, however, can be undesirable when screening
for biocatalytic transformations. Synthetic labelled sub-
strates include fluorogenic and chromogenic substrates,
isotopically labelled substrates, FRET substrates, and
substrates with fluorescent labels for indirect detection.
Fluorogenic and chromogenic substrates by
b-elimination
Esters and ethers of nitrophenol and umbelliferone, and
amides of nitroaniline and aminocoumarin have been
known for many years to be chromogenic or fluorogenic
substrates for glycosidases, lipases, esterases and pro-
teases (Figure 1). Assays based on these substrates are
problematic, however, because the phenolate or aniline
Corresponding author: Jean-Louis Reymond ( jean-louis.reymond@ioc.unibe.ch).
Available online 30 April 2004
Review TRENDS in Biotechnology Vol.22 No.7 July 2004
www.sciencedirect.com 0167-7799/$ - see front matter q 2004 Elsevier Ltd. All rights reserved. doi:10.1016/j.tibtech.2004.04.005
leaving group is at least five orders of magnitude more
reactive as a leaving group than is the aliphatic alcoholate
or amine that is present in the natural substrates of the
enzymes. These substrates are therefore often unstable
and susceptible to cleavage by non-catalytic contaminants.
In addition, substrate structure cannot be diversified in
proximity to the enzyme-labile bond.
In 1998, Klein and Reymond [14] reported a new type
of fluorogenic substrate that uses phenolate release as
signal. First, the chiral secondary alcohol (S)-(4) (Figure 2)
was used as a fluorogenic substrate for alcohol dehydro-
genase. The primary enzyme reaction product is a ketone
(5), which subsequently undergoes a b-elimination reac-
tion to liberate the fluorescent umbelliferone reporter
molecule (Figure 2). Furthermore, a double indirect pro-
cedure facilitated the assay of lipases with the correspond-
ing esters [15]. This strategy efficiently decouples the
phenolate release chemistry from the enzymatic trans-
formation and facilitates structural and functional diver-
sification of the substrates. The b-elimination approach
has been used in substrates for various enzyme assays,
Figure 1. Chromogenic and fluorogenic substrates with activated leaving groups.
Substrates are shown for glycosidases (1), proteases (2) and lipases and esterases
(3). The enzyme-labile bond is shown in red and the chromophore in blue.
TRENDS in Biotechnology
H
N
N
H
O
O
NH
NH2HN
O OO
O
1
β-glucosidase
2
Trypsin
3
Lipases
NO2
O
OH
HO
HO
OH
O
NO2
Figure 2. Fluorogenic enzyme substrates with indirect fluorophore release by secondary b-elimination. The chiral secondary alcohol (S)-(4) is used as a fluorogenic
substrate for alcohol dehydrogenase (ADH). The primary enzyme reaction product is a ketone (5), which subsequently undergoes b-elimination accelerated by bovine
serum albumin (BSA) to liberate the fluorescent umbelliferone reporter molecule. This approach has been used in assays for various enzymes, including epoxide hydro-
lases (6), transadolases (7) transketolases (8) and Baeyer–Villigerases (9). An assay for b-lactamase (10) operates by a similar release mechanism for umbelliferone. In
addition, a probe for quantifying NADH and NADPH relies on hydride transfer and b-elimination (11). The enzyme-labile bond, the enzymes and (where needed) secondary
reagents are shown in red. Abbreviations: Obn, benzyloxy; TfO, trifluoromethansulfonate.
TRENDS in Biotechnology
O OO
OCoum
O
H
(S)-4
OCoum
O OO
OCoum
N
N
OCoum
N
S
H
N
O
O
Ph
OBnO
OCoum
TfO
ADH
NAD(P)+
BSA, pH>7
5
Umbelliferone anion
(blue fluorescent)
10
β-lactamase
6
Epoxide hydrolase
+ NaIO4
9
Baeyer–Villigerase,
+ esterase + ADH/NAD+
8
Transketolase
7
Transaldolase
Coum
O
CoumO OH
OH
HO OH
11
NADH/NADPH
HO OCoum
O
OH
OH
O
O
H OH
Review TRENDS in Biotechnology Vol.22 No.7 July 2004364
www.sciencedirect.com
including assays for aldolase catalytic antibodies [16–18],
epoxide hydrolases (6) [19], transadolases (7) [20], lipases
and esterases [19,21,22], phosphatases [19,23], acylases
[19], proteases [24], transketolases (8) [25] and Baeyer–
Villigerases (9) [26]. The recently reported b-lactamase
substrate (10) operates by a similar release mechanism for
umbelliferone [27] (Figure 2).
FRET substrates
One of the most efficient principles for detecting cleavage
reactions is the use of FRET substrates, whereby a fluoro-
phore quencher or fluorophore–fluorophore pair is sepa-
rated by the cleavage of an enzyme-labile bond, resulting
in either an increase in fluorescence or a shift in
wavelength. This principle was originally reported for
the detection of HIV protease activities [28].
The same approach has been used recently in an all-
protein construct to detect protease activity using two-
photon cross-correlation and FRET analysis [29]. This
irradiation technique is milder than the direct fluorophore
excitation used in classical protease assays. FRET sub-
strates are not limited to protease-type cleavage, and
an elegant system with FRET phospholipids has been
recently used for a genetic analysis of phospholipase
activity by in vivo imaging [30].
Other signalling substrates
Several other recently reported fluorogenic and chromo-
genic substrates are interesting in terms of their particu-
lar chromogenic mechanism (Figure 3). In early 1990, it
was shown that the changes in the ultraviolet/visible and
fluorescence properties of 6-methoxynaphthyl-methanol
(12) (Figure 2) on oxidation to the corresponding aldehyde
(13) could be exploited to provide a fluorogenic alcohol
dehydrogenase assay [31] (Figure 3). This assay has been
subsequently adapted for assaying retroaldolase catalytic
antibodies [18] and, together with a secondary oxidation
with sodium periodate, for lipases and epoxide hydrolases
[32]. Styrene oxides can be used directly as chromogenic
substrates for epoxide hydrolases in a similar set-up [33].
In the area of lipase assays, acyloxymethylethers of
umbelliferone such as (14) (Figure 3), which were origin-
ally developed as fluorogenic probes for catalytic anti-
bodies [34], have been recently found to be excellent
substrates for lipases and esterases [35]. The advantage of
these substrates is that the leaving group is an aliphatic
alcohol that is much less acidic than umbelliferone itself,
which reduces susceptibility to non-catalytic cleavage.
The non-fluorescent N-aryl maleimide (15) (Figure 3)
can be used to follow C–C bond formation reactions, such
as antibody-catalysed Michael additions and Diels–Alder
reactions, because the corresponding maleimide adducts
such as (16) are fluorescent [36] (Figure 3). A potential
problem with this assay is the fact that the maleimide
reagent (16) is a highly reactive electrophile and readily
adds to thiols to form a fluorescent product.
In an elegant solid-phase assay, Yeo and Mrksich [37]
have monitored the activity of the lipase cutinase by using
a mono-ester of para-hydroxyphenol as a substrate
immobilized on an electrode surface by a sulphur–gold
interaction [37]. The para-hydroxyphenol released by
hydrolysis is detected electrochemically by oxidation to
the quinone, facilitating the voltametric quantification of
enzymatic activity and thereby demonstrating a principle
that might provide a general solution for the electro-
chemical sensing of enzyme activities.
13
C- and 2
H-labelled pseudoenantiomers
One of the key problems in enzyme assays for biocatalysis
is the ability to detect enantioselectivity directly in high
throughput, but Reetz and co-workers [38–40] have pro-
posed various solutions based on isotopic labelling. For
example, enantioselective isotopic labelling of one enan-
tiomer of a chiral acetyl ester or amide has been used to
trace the enzymatic kinetic resolutions of these substrates
by enantioselective hydrolysis. Isotopic 13
C or 2
H labelling
of the acetyl group produces no chemical reactivity
changes between the enantiomers, but facilitates the
selective tracing of each enantiomeric substrate or product
by mass spectrometry [38], 1
H-NMR [39] or Fourier
transform infrared spectroscopy [40]. The methods can
be implemented in high throughput by using appropri-
ately robotized instruments to screen libraries of enzyme
mutants [41].
These approaches are certainly powerful, but they
require expensive isotopic labelling reagents and possibly
the modification of rather expensive instruments for high-
throughput use, which renders them off-limits for many
Figure 3. Additional types of fluorogenic substrate. 6-methoxynaphthylmethanol
(12) is a fluorogenic substrate for alcohol dehydrogenase. The coumarinoxymethyl
ester (14) is a low-reactivity fluorogenic substrate for lipases. The fluorogenic
maleimide probe (15) reacts with nucleophiles and allows screening of aldolase
catalytic antibodies (Ab) for C–C bond formation. Abbreviations: ADH, alcohol
dehydrogenase; Cat. Ab., catalytic antibody; MeO, methoxy.
TRENDS in Biotechnology
O
H
CoumO
ADH, NAD+
O OOO
O
MeO
OH
N
H
N
N
O
O
O
MeO
O
Ar N
O
O
O
15
Non-fluorescent
12
Non-fluorescent
14
Non-fluorescent
13
Blue fluorescent
Blue fluorescent
Lipase or esterase
Cat. Ab.
16
Fluorescent
Coum
Ar
Review TRENDS in Biotechnology Vol.22 No.7 July 2004 365
www.sciencedirect.com
laboratories. Reetz and colleagues have also implemented
other instrumental methods for following enantioselective
reactions in high throughput, including NMR analysis
of Mosher’s esters [39], and monitoring the differential
cell growth in media containing either enantiomer of a
fluoroacetate ester that releases the toxic fluoroacetic acid
on hydrolysis [42].
Fluorogenic substrates for kinases and phosphatases
Several kinase and phosphatase assays based on labelled
substrates have been reported recently, and these deserve
to be discussed separately. Interest in kinases and phos-
phatases is mainly linked to screening for inhibitors of
these enzymes, which control signal transduction cascades
for gene activation. The family of kinases and phospha-
tases represents up to 5% of the human genome and thus is
an important target for drug discovery [43].
One kinase assay is based on the activity of an
aminopeptidase for the fluorogenic release of rhodamine
from a phosphorylated labelled peptide (18), which is
diminished relative to its activity towards the non-
phosphorylated peptide (17) [44] (Figure 4). This set-up
is similar to a patented assay based on the differential
protease sensitivity of phosphorylatable FRET peptides,
which has been commercialized by Invitrogen under the
trade name Z0
-Lyte [45]. Similarly, the differential reac-
tivity of chymotrypsin has been used to follow cis–trans
prolyl isomerase activity [46–48].
Whereas the above assays are based on the reactivity of
a second enzyme to trigger fluorophore release, a much
simpler solution for assaying kinases has been developed
by Lawrence and co-workers [49], who detect intracellular
protein kinase activity in the form of a short synthetic
peptide (19) containing a caged serine residue in the
vicinity of a nitrobenzofurazane fluorophore (Figure 4).
After irradiation, phosphorylation of the liberated serine
residue by the kinase induces a threefold increase in
fluorescence in the fluorophore. Shults and Imperiali [50]
have reported another very elegant solution for assaying
kinases. They found that peptides containing a phosphory-
latable amino acid followed by a b-turn and a ‘Sox’ amino
acid operate as fluorogenic kinase substrates owing to the
phosphorylation-induced binding of magnesium ions by
the Sox chromophore, which results in an increase in
fluorescence [50].
In a completely different assay design, a fluorescent
label in a peptide substrate has been used to report binding
of the phosphorylated peptide to a macromolecule via
induced changes in fluorescence polarization. This assay
enables researchers to detect the activity of proteases,
kinases and phosphatases by following changes in the
fluorescence polarization induced by binding of the phos-
phorylated peptides to poly-arginine [51]. The similar,
immobilized metal assay platform method detects an
increase in fluorescence polarization in fluorescently
labelled peptides when they bind, in their phosphorylated
form, to nanoparticles via metal coordination [52]. Other
kinase assays based on indirect sensing are discussed below.
Sensing reactions of unmodified substrates
Many enzyme assays have been reported that come very
close to the ideal situation: that is, they detect transform-
ation of the enzyme’s natural or targeted substrate itself.
This is often realizable by employing an analytical
approach such as gas chromatography, high-performance
liquid chromatography, mass spectrometry or NMR, and
the use of these methods is very common within the
constraints of industrial applications.
Below we focus on methods that produce a colour or a
fluorescent change by a chemical process within the test
solution and have the potential for very high throughput at
low cost. These assays are based on chemosensors that
respond to product formation independently of any label in
the substrate or product during the enzymatic transform-
ation. The prototypical application of this idea is the use of
a chromogenic pH indicator to follow ester hydrolysis by
lipases – a concept that has been elegantly implemented
by Kazlauskas and co-workers [53,54] to screen for enan-
tioselective lipases. Such sensors have also been used in
the context of enzymes encapsulated in a sol-gel [55].
Endpoint assays
The endpoint determination of substrate consumption or
product formation is almost always sufficient for assaying
enzyme activities, and it can be realized by applying a
Figure 4. Fluorogenic probes for kinases. The bis-peptidyl rhodamine substrate
undergoes fluorogenic cleavage by aminopeptidase in its non-phosphorylated
state (17), but not in its phosphorylated state (18), enabling indirect quantification
of phosphorylation by kinases. On phosphorylation by kinases, the caged serine
peptide (19) is deprotected photochemically and becomes fluorescent at the nitro-
benzofurazane chromophore on. Other kinase assays are discussed in the text.
TRENDS in Biotechnology
O2N OMe
OMe
O
N
H
Phe-Arg-Arg-Arg-Arg-Lys-NH2
O
N
O N
O2N
O
O
O
H
N
H
N
Leu-...-Ser-Leu-Gly Gly-Leu-Ser-...-Leu
OH2N NH2
+
CO2
–
2 Aminopeptidase
Rhodamine
(red fuorescent)
18 (X = PO3H–
)
hν
19
O O
17 (X = H)
1 Kinase
X X
Kinase phosphorylation site
Review TRENDS in Biotechnology Vol.22 No.7 July 2004366
www.sciencedirect.com
reagent that specifically detects a functional group that is
formed in the reaction. A straightforward application of
this principle has been reported recently by Henke and
Bornscheuer [56] for the detection of amidase activities.
The amine product that is formed is detected by the fluoro-
genic amine-reagent 4-nitro-7-chloro-benzofurazane. This
method has been applied to the enantioselective screening
of different hydrolases. The broadly applicable probe for
quantifying NADH and NADPH, which relies on an ele-
gant hydride-transfer/b-elimination sequence as discussed
above, represents a more subtle application of the principle
of product detection by quantitative reaction [57] (Figure 2).
Classically, the activity of protein kinases has been
analysed by incubating immobilized proteins or peptides
with radiolabelled ATP, or by staining the product with
antibodies specific for the phosphoprotein. A recent improve-
ment in protein kinase activity detection has been made
with the development of a phosphoprotein-selective
fluorescent dye called Pro-Q Diamond. This dye is suitable
for staining protein microarrays as an endpoint treatment
after phosphorylation with ATP and a kinase [58], and
thus should facilitate the identification of kinase sub-
strates. A similar dye that is based on a zinc fluorescein
complex and binds binding to phosphorylated peptides has
been reported recently [59]. All of these assays require an
immobilized substrate, usually a whole protein, because
the excess ATP reagent and the kinase must be washed
away before the phosphate-staining reagent can be applied.
Matile and co-workers [60,61] have reported an elegant
assay for substrate versus product quantification that uses
synthetic multifunctional pores (SMPs) consisting of syn-
thetic peptide-appended oligophenylene building blocks
and can be used to monitor the activity of various enzymes.
The SMPs are incorporated into the membrane of vesicles
loaded with fluorescein, where they function as channels
for the escape of this fluorophore, which results in an
increase in fluorescence because dilution removes auto-
quenching. Substrate/product ratios in an enzymatic
reaction can be monitored whenever substrate and pro-
duct differentially modulate the flow offluorescein through
the SMPs – for example, during the hydrolysis of DNA by
DNases or of peptides by proteases. Although this set-up is
elegant, the availability of the pore component, which
must be prepared by complex multistep syntheses, and the
robustness of the system to unselective pore blockage by
contaminants such as the enzymes and cofactors them-
selves represent potential limitations.
Indirect product detection as an endpoint is also
possible by the principle of back-titration. Wahler and
Reymond [62] have demonstrated this principle in a
versatile colorimetric assay based on the quantification
of periodate-sensitive reaction products formed by the
enzymatic hydrolysis of periodate-resistant substrates
(Figure 5). Sodium periodate is either consumed by the
product formed or used to oxidize adrenaline (20) to form
the deeply red coloured adrenochrome (21) (Figure 5).
Product formation is proportional to the decrease in
adrenochrome formation.
The assay is compatible with various operating con-
ditions, including co-solvents and extreme pH values, and
has been used to test the hydrolysis of vegetal oil and
tributyrin by lipases and esterase, the opening of various
aliphatic and aromatic epoxides by epoxide hydrolases,
and the hydrolysis of phytic acid by phytases. It has been
applied to the rapid screening of esterases against an array
of carbohydrate acetates [63]. This so-called ‘adrenaline
test’ for enzyme is surprisingly versatile and works with
inexpensive reagents, so that it can be implemented
almost anywhere. One of its limitations is that the
periodate reagents also reacts with any other diol that
might be present in the medium, in particular glycerol,
which is often used for enzyme cryopreservation.
It is also possible to detect the carbonyl products of
periodate cleavage by using a colorimetric Schiff base
reagent, and this has been demonstrated for an epoxide
hydrolase assay [64]. It must be mentioned, however,
that detecting volatile aliphatic aldehydes is problematic
in a microtitre plate owing to cross-diffusion. The assay
has a detection limit in the 0.01 M range, which is 10- to
50-fold less sensitive that the adrenaline test based on
back-titration.
Real-time assays
A few sensor systems enable enzymatic reactions to be
monitored in real time. This is possible whenever the
sensor does not interact with the enzymatic reaction. For
example, Konarzycka–Bessler and Bornscheuer [65] have
reported an assay for the lipase-catalysed esterification of
alcohols by vinyl acetate in organic solvent. This assay is
based on the real-time trapping of the released acet-
aldehyde by non-fluorescent 4-hydrazino-7-nitro-benzo-
furazane to form a fluorescent hydrazone. The activity of
Figure 5. The adrenaline test for enzymes. Sodium periodate is consumed by
oxidation of either an oxidizable diol or an amino alcohol product of the enzyme
reaction. Back-titration of the unreacted adrenaline (20) to form the deeply coloured
adrenochrome (21) enables quantification of the extent of product formation.
TRENDS in Biotechnology
S P
Step 1: Enzyme reaction
NaIO4 resistant substrate:
Tributyrin (esterases)
Triglycerides (lipases)
Epoxides (EH)
Phytic acid (phytases)
NaIO4-sensitive
product:
1, 2-diol
NaIO3
+ H2O
NaIO4
OH
HO
MeHN
OH
20
Colorless O
O
N
HO
Me
21
Deep red color
Step 3: Add
adrenaline
Step 2:
Product
oxidation
Review TRENDS in Biotechnology Vol.22 No.7 July 2004 367
www.sciencedirect.com
telomerase, a DNA polymerase whose detection might be
useful in cancer diagnosis and which is a possible target for
chemotherapy, has been followed on a biosensor chip by
detecting elongation of a surface immobilized telomeric
repeat primer directly using surface plasmon resonance
(Biacore) [66]. DNA nucleases, also called ‘DNases’, have
been recently assayed using the double-helix-selective dye
PicoGreen (which is also used in real-time PCR) by
following the fluorescence intensity decrease induced by
dye release [67].
Building on earlier work with a quinacridone fluoro-
phore [68–69], we and our co-workers [70] have recently
found that a non-fluorescent calcein–copper complex, a
fluorescein derivative, can be used as a selective dynamic
sensor for amino acids. The sensor is used at a micromolar
or submicromolar concentration and responds to amino
acids in the 0.1–1 mM range by showing a tenfold increase
in its intensity of green fluorescence as the quenching
copper ion is displaced from the calcein fluorophore (22)
(Figure 6) by the chelating amino acid. This fluorescent
Figure 6. Calcein–copper as a selective dynamic sensor of amino acids. (a) Principle of detecting amino acid products by calcein–copper. Copper(II) ions quench the
fluorescence of calcein when bound to the fluorophore. Calcein (22), used in the 1027
–1026
M concentration range, is released when copper ions (5 £ 1026
M) are chelated
by amino acids (AA) in the 1024
–1023
M concentration range. This assay facilitates the direct detection of aminopeptidase, acylase and protease activities. (b) Time course
of fluorescence increase in a calcein–copper assay for the hydrolysis of N-acetyl L-methionine to L-methionine by acylase I. Conditions: 10 mM N-acetyl-L-methionine in
aqueous 5 mM Bis-tris buffer (pH 7.2), 25 8C, 1 mM calcein, 5 mM CuCl2, lem ¼ 530 ^ 25 nm, lex ¼ 450 ^ 50 nm, and either no enzyme (black) or acylase I at 2.5 mg/ml
(green), 5.0 mg/ml (blue) or 10 mg/ml (red). Abbreviations: lem, emission wavelength; lex, excitation wavelength.
TRENDS in Biotechnology
OHO O
N CO2H
CO2H
NHO2C
HO2C
CO2H
H2N
O
OH
R
AA
[CuAA2]
Non-chelating
derivative (N-acyl amino
acid, amino acid amide,
protein)
Enzyme
22
Fluorescent
[Cu.22]
Non-fluorescent
22
(λex = 450 nm, λem = 530 nm)
(a)
(b)
0
10000
5000
15000
0 3600
Time (s)
7200
Fluorecence
Review TRENDS in Biotechnology Vol.22 No.7 July 2004368
www.sciencedirect.com
sensor system has been used to monitor the activity of
acylases, aminopeptidases and proteases (Figure 6).
Indeed, these enzymes release free amino acids from
non-chelating amide precursor substrates such as
N-acetyl-methionine, L-leucinamide and whole bovine
serum albumin (BSA) protein.
Similar to the adrenaline test discussed above, the
calcein–copper assay uses only commercially available,
inexpensive reagents and can be implemented almost
anywhere. It cannot be used in the presence of metal-
chelating species such as EDTA, however, which also
induce a fluorescence increase. If the enzyme is sensitive
to copper ions, the assay can be used as an endpoint
measurement – for example, it can be used to assay
cysteine proteases.
Outlook: enzyme activity profiles and fingerprints
An enzyme assay not only detects the enzyme, but
naturally also indicates the enzyme type by the substrate
that is used. Currently, the general trend beyond the
development of enzyme assays is to assemble many such
assays in a parallel format to generate the complete
reactivity profile of an enzyme [71]. Multienzyme profiling
with chromogenic substrates was developed in the 1960s
as a tool for identifying microorganisms [72], and today it
forms the basis for medical diagnostics of infectious
diseases in hospitals.
In the 1990s the substrate specificity of proteases
was investigated by using combinatorial libraries of
fluorogenic peptides [73,74]. Recently, we and our
co-workers [32,62,75] have shown that assays with
multiple fluorogenic substrates can be carried out rapidly
in microtitre-plate format to generate activity profiles of
many different hydrolytic enzymes. Because these profiles
are recorded easily and reproducibly, they can be con-
sidered ‘fingerprints’ of these enzymes. They also provide a
rapid and versatile tool for the functional classification of
enzymes such as various lipases and esterases [76].
The operational simplicity of these fingerprinting
experiments can be improved by using substrates arrayed
on a solid support such as a glass surface, as shown
recently by Ellman and co-workers [77] for peptide micro-
arrays for determining protease substrate specificity.
Furthermore, Yao and co-workers [78] have adapted
periodate/b-elimination-triggered fluorogenic substrates
for epoxide hydrolases, amidases, esterases and phospha-
tases to a similar glass-supported format. Such enzyme
fingerprints might form the basis for a new type of quality
control and diagnostic application.
Concluding remarks
The field of enzyme assays has undergone tremendous
developement in recent years due to the growing import-
ance of high-throughput screening in drug discovery and
enzyme engineering. Enzyme assays are conceptually
close to the general problem of designing selective chemo-
sensors for small molecules. Enzyme assays have provided
a fertile ground for the realization of creative ideas in
system design, and a surprising diversity of formats have
been imagined to make enzyme reactions visible. Thus,
time-honored ‘classics’ such as nitrophenyl esters and
ethers and alcohol dehydrogenase-coupled assays, which
a few years ago represented almost the entire known
chemistry of enzyme assays, have made room for a broad
variety of labels, triggering chemistries, and signalling
systems. There is no doubt that the field will continue to
evolve as newer and better methods appear for screening
enzymes. Challenges abound because many reaction types
are still difficult to assay in high-throughput or with a
sufficient level of accuracy.
Acknowledgements
This work was supported by the Swiss National Science Foundation,
COST Action D16, the Swiss OFES and Prote´us SA, Nıˆmes, France.
References
1 Marrs, B. et al. (1999) Novel approaches for discovering industrial
enzymes. Curr. Opin. Microbiol. 2, 241–245
2 Stemmer, W.P.C. (1994) DNA shuffling by random fragmentation and
reassembly: in vitro recombination for molecular evolution. Proc. Natl.
Acad. Sci. U. S. A. 91, 10747–10751
3 Stemmer, W.P.C. (1994) Rapid evolution of a protein in vitro by DNA
shuffling. Nature 370, 389–391
4 You, L. and Arnold, F.H. (1996) Directed evolution of subtilisin E in
Bacillus subtilis to enhance total activity in aqueous dimethylforma-
mide. Protein Eng. 9, 77–83
5 Reetz, M.T. and Jaeger, K-E. (2000) Enantioselective enzymes for
organic synthesis created by directed evolution. Chemistry 6, 407–412
6 Taylor, S.V. et al. (2001) Investigating and engineering enzymes by
genetic selection. Angew. Chem. Int. Ed. Engl. 40, 3310–3335
7 Eisenthal, R. and Danson, M. eds (2002) Enzyme Assays: a Practical
Approach, Oxford University Press
8 Gul, S. et al. (1998) Enzyme Assays Essential Data, John Wiley & Sons
9 Reetz, M.T. (2001) Combinatorial and evolution-based methods in the
creation of enantioselective catalysts. Angew. Chem. Int. Ed. Engl. 40,
284–310
10 Wahler, D. and Reymond, J-L. (2001) Novel methods for biocatalysts
screening. Curr. Opin. Chem. Biol. 5, 152–158
11 Beisson, F. et al. (2000) Methods for lipase detection and assay:
a critical review. Eur. J. Lipid Sci. Technol. 2, 133–153
12 Wahler, D. and Reymond, J-L. (2001) High-throughput screening for
biocatalysts. Curr. Opin. Biotechnol. 12, 535–544
13 Speer, A.E. and Cravatt, B.F. (2004) Chemical strategies for activity-
based proteomics. ChemBioChem 5, 41–47
14 Klein, G. and Reymond, J-L. (1998) An enantioselective fluorimetric
assay for alcohol dehydrogenases using albumin-catalyzed b-elimin-
ation of umbelliferone. Bioorg. Med. Chem. Lett. 8, 1113–1116
15 Klein, G. and Reymond, J.-L. (1999) Enantioselective fluorogenic assay
of acetate hydrolysis for detecting lipase catalytic antibodies. Helv.
Chim. Acta 3, 400–408
16 Jourdain, N. et al. (1998) A stereoselective fluorogenic assay for
aldolases: detection of an anti-selective aldolase catalytic antibody.
Tetrahedron Lett. 51, 9415
17 Pe´rez Carlo`n, R. et al. (2000) Fluorogenic polypropionate fragments for
detecting stereoselective aldolases. Chemistry 6, 4154–4162
18 List, B. et al. (1998) Aldol sensors for the rapid generation of tunable
fluorescence by antibody catalysis. Proc. Natl. Acad. Sci. U. S. A. 95,
15351–15355
19 Badalassi, F. et al. (2000) A versatile periodate-coupled fluorogenic
assay for hydrolytic enzymes. Angew. Chem. Int. Ed. Engl. 39,
4067–4070
20 Gonza´lez-Garcı´a, E. et al. (2003) Fluorogenic stereochemical probes for
transaldolases. Chemistry 9, 893–899
21 Leroy, E. et al. (2003) Fluorogenic cyanohydrin esters as chiral probes
for esterase and lipase activity. Adv. Synth. Catal. 6 þ 7, 859–865
22 Nyfeler, E. et al. (2003) A sensitive and selective high-throughput
screening fluorescence assay for lipases and esterases. Helv. Chim.
Acta 8, 2919–2927
23 Gonza´lez-Garcia´, E.M. et al. (2003) Synthesis and evaluation of
chromogenic and fluorogenic analogs of glycerol for enzyme assays.
Helv. Chim. Acta 7, 2458–2470
Review TRENDS in Biotechnology Vol.22 No.7 July 2004 369
www.sciencedirect.com
24 Badalassi, F. et al. (2002) A selective HIV-protease assay based on a
chromogenic amino acid. Helv. Chim. Acta 10, 3090–3098
25 Sevestre, A. et al. (2003) A fluorogenic assay for transketolase from
Saccharomyces cerevisiae. Tetrahedron lett. 4, 827–830
26 Gutierrez, M.C. et al. (2003) The first fluorogenic assay for detecting a
Baeyer–Villigerase activity on microbial cells. Org. Biomol. Chem. 1,
3500–3506
27 Gao, W. et al. (2003) Novel fluorogenic substrates for imaging
b-lactamase gene expression. J. Am. Chem. Soc. 37, 11146–11147
28 Matayoshi, E.D. et al. (1990) Novel fluorogenic substrates for assay-
ing retroviral proteases by resonance energy transfer. Science 247,
954–958
29 Kohl, T. et al. (2002) A protease assay for two-photon crosscorrelation
and FRET analysis based solely on fluorescent proteins. Proc. Natl.
Acad. Sci. U. S. A. 99, 12161–12166
30 Farber, S.A. et al. (2001) Genetic analysis of digestive physiology using
fluorescent phospholipid reporters. Science 292, 1385–1388
31 Vallee, B.L. (1992) Methods of measuring isozymes and isozyme
classes of alcohol dehydrogenase. US Patent, 5,162,203
32 Wahler, D. et al. (2002) Enzyme fingerprints of activity, stereo- and
enantioselectivity from fluorogenic and chromogenic substrate arrays.
Chemistry 8, 3211–3228
33 Mateo, C. et al. (2003) A spectrophotometric assay for measuring and
detecting an epoxide hydrolase activity. Anal. Biochem. 314, 135–141
34 Bensel, N. et al. (2001) Pivalase catalytic antibodies: towards
abzymatic activation of prodrugs. Chemistry 7, 4604–4612
35 Leroy, E. et al. (2003) A low background high-throughput screening
(HTS) fluorescence assay for lipases and esterases using acyloxy-
methylethers of umbelliferone. Bioorg. Med. Chem. Lett. 13, 2105–2108
36 Tanaka, F. et al. (2003) Fluorescent detection of carbon–carbon bond
formation. J. Am. Chem. Soc. 125, 8523–8528
37 Yeo, W-S. and Mrksich, M. (2003) Self-assembled monolayers that
transduce enzymatic activities to electrical signals. Angew. Chem. Int.
Ed. Engl. 42, 3121–3124
38 Reetz, M.T. et al. (1999) A method for high-throughput screening of
enantioselective catalysts. Angew. Chem. Intl. Ed. 12, 1758–1761
39 Reetz, M.T. et al. (2002) A practical NMR-based high-throughput assay
for screening enantioselective catalysts and biocatalysts. Adv. Synth.
Catal. 9, 1008–1016
40 Tielmann, P. et al. (2003) A practical high-throughput screening
system for enantioselectivity by using FT-IR spectroscopy. Chemistry
9, 3882–3887
41 Cedrone, F. et al. (2003) Directed evolution of the epoxide hydrolase
from aspergillus niger. Biocatalysis and Biotransformation 6, 357–364
42 Reetz, M.T. and Ru¨ggeberg, C.J. (2002) A screening system for
enantioselective enzymes based on differential cell growth. Chem.
Commun. 7, 1428–1429
43 Cohen, P. (2001) The role of protein phosphorylation in human health
and disease. Eur. J. Biochem. 268, 5001–5010
44 Kupcho, K. et al. (2003) A homogeneous, nonradioactive high-
throughput fluorogenic protein kinase assay. Anal. Biochem. 317,
210–217
45 Hamman, B.D. et al. (2002) Optical probes and assays. US Patent
60
4100
255, 2002-06-25
46 Garcya-Echeverrya, C. et al. (1992) Continuous fluorimetric direct
(uncoupled) assay for peptidyl prolyl cis-trans isomerases. J. Am.
Chem. Soc. 7, 2758–2759
47 Kofron, J.L. et al. (1991) Determination of kinetic constants for
peptidyl prolyl cis–trans isomerases by an improved spectrophoto-
metric assay. Biochemistry 30, 6127–6134
48 Yli-Kauhaluoma, J.T. et al. (1996) Catalytic antibodies with peptidyl-
prolyl cis-trans isomerase activity. J. Am. Chem. Soc. 23, 5496–5497
49 Velhuyzen, W.F. et al. (2003) A light-activated probe of intracellular
protein kinase activity. J. Am. Chem. Soc. 125, 13358–13359
50 Shults, M.D. and Imperiali, B. (2003) Versatile fluorescence probes of
protein kinase activity. J. Am. Chem. Soc. 125, 14248–14249
51 Simeonov, A. et al. (2002) Enzyme assays by fluorescence polarization
in the presence of polyarginine: study of kinase, phosphatases and
protease reactions. Anal. Biochem. 304, 193–199
52 Gaudet, E. et al. (2003) A homogeneous fluorescence polarization assay
adaptable for a range of protein serine/threonine and tyrosine kinases.
J. Biomol. Screen. 8, 164–175
53 Janes, L.E. and Kazlauskas, R.J. (1997) Quick E. A fast spectro-
photometric method to measure the enantioselectivity of hydrolases.
J. Org. Chem. 14, 4560–4561
54 Janes, L.E. et al. (1998) Quantitative screening of hydrolase libraries
using pH indicators: identifying active and enantioselective hydro-
lases. Chem. Eur. J. 11, 2324–2331
55 Park, C.B. and Clark, D.S. (2002) Sol-gel-encapsulated enzyme arrays
for high-throughput screening of biocatalytic activity. Biotechnol.
Bioeng. 78, 229–235
56 Henke, E. and Bornscheuer, U.T. (2003) Fluorophoric assay for the
high-throughput determination of amidase activity. Anal. Chem. 75,
255–260
57 Roeschlaub, C.A. et al. (1999) A fluorescent probe for the detection of
NAD(P)H. Chem. Commun., 1637–1638
58 Martin, K. et al. (2003) Quantitative analysis of protein phosphory-
lationstatus and protein kinase activity on microarrays using a novel
fluorescent phosphorylation sensor dye. Proteomics 3, 1244–1255
59 Ojida, A. et al. (2002) First artificial receptors and chemosensors
toward phosphorylated peptide in aqueous solution. J. Am. Chem. Soc.
124, 6256–6258
60 Sorde`, N. et al. (2003) Enzyme screening with synthetic multi-
functional pores: focus on biopolymers. Proc. Natl. Acad. Sci. U. S. A.
100, 11964–11969
61 Das, G. et al. (2002) Fluorometric detection of enzyme activity with
synthetic supramolecular pores. Science 298, 1600–1602
62 Wahler, D. and Reymond, J-L. (2002) The adrenaline test for enzymes.
Angew. Chem. Int. Ed. Engl. 41, 1229–1232
63 Wahler, D. et al. (2004) Adrenaline profiling of lipases and esterases
with 1,2-diol and carbohydrate acetates. Tetrahedron 3, 703–710
64 Doderer, K. et al. (2003) Spectrophotometric assay for epoxide
hydrolase activity toward any epoxide. Anal. Biochem. 321, 131–134
65 Konarzycka-Bessler, M. and Bornscheuer, U.T. (2003) A high-
throughput screening method for determining the synthetic activity
of hydrolases. Angew. Chem. Int. Ed. Engl. 42, 1418–1420
66 Maesawa, C. et al. (2003) A rapid biosensor chip assay for measuring of
telomerase activity using surface plasmon resonance. Nucleic Acids
Res. 31, E4
67 Tolun, G. and Myers, R.S. (2003) A real-time DNase assay (ReDA)
based on PicoGreen fluorescence. Nucleic Acids Res. 31, E111
68 Klein, G. et al. (2001) A fluorescent metal sensor based on macrocyclic
chelation. Chem. Commun., 561
69 Klein, G. and Reymond, J-L. (2001) An enzyme assay using pM.
Angew. Chem. Int. Ed. Engl. 40, 1771–1773
70 Dean, K.E.S. et al. (2003) A green fluorescent chemosensor for amino
acids provides a versatile high-throughput screening (HTS) assay for
proteases. Bioorg. Med. Chem. Lett. 13, 1653–1656
71 Reymond, J-L. and Wahler, D. (2002) Substrate arrays as enzyme
fingerprinting tools. ChemBioChem 3, 701–708
72 Bussie`re, J. et al. (1967) Usage de substrats synthetiques pour l’e´tude
de l’e´quipement enzymatique de microorganismes. C. R. Acad. Sc.
Paris 264D, 415–417
73 Meldal, M. et al. (1994) Portion-mixing peptide libraries of quenched
fluorogenic substrates for complete subsite mapping of endoprotease
specificity. Proc. Natl. Acad. Sci. USA 8, 3314–3318
74 Maly, D.J. et al. (2002) Combinatorial strategies for targeting protein
families: application to the proteases. Chem. BioChem. 1, 16–37
75 Wahler, D. et al. (2001) Enzyme fingerprints by fluorogenic and
chromogenic substrate arrays. Angew. Chem. Int. Ed. Engl. 40,
4457–4460
76 Grognux, J. and Reymond, J-L. Classifying enzymes from selectivity
fingerprints. ChemBioChem (in press)
77 Salisbury, C.M. et al. (2002) Peptide microarrays for the determination
of protease substrate specificity. J. Am. Chem. Soc. 124, 14868–14870
78 Zhu, Q. et al. (2003) Enzymatic profiling system in a small-molecule
microarray. Org. Lett. 5, 1257–1260
Review TRENDS in Biotechnology Vol.22 No.7 July 2004370
www.sciencedirect.com

More Related Content

What's hot

Protein Electrophoresis & Gas Liquid Chromatography & HPLC Applications
 Protein Electrophoresis  & Gas Liquid Chromatography &  HPLC Applications  Protein Electrophoresis  & Gas Liquid Chromatography &  HPLC Applications
Protein Electrophoresis & Gas Liquid Chromatography & HPLC Applications Amany Elsayed
 
Affinity chromatography
Affinity chromatographyAffinity chromatography
Affinity chromatographyRavi kumar
 
ElogDoct: A Tool for Lipophilicity Determination in Drug Discovery. 2. Basic ...
ElogDoct: A Tool for Lipophilicity Determination in Drug Discovery. 2. Basic ...ElogDoct: A Tool for Lipophilicity Determination in Drug Discovery. 2. Basic ...
ElogDoct: A Tool for Lipophilicity Determination in Drug Discovery. 2. Basic ...Brian Bissett
 
Affinity Chromoatograpy
Affinity ChromoatograpyAffinity Chromoatograpy
Affinity ChromoatograpyNaresh sah
 
Antibody Glycan Analysis with Normal Phase PhyTip Columns
Antibody Glycan Analysis with Normal Phase PhyTip ColumnsAntibody Glycan Analysis with Normal Phase PhyTip Columns
Antibody Glycan Analysis with Normal Phase PhyTip ColumnsChris Suh
 
Application of chromatography in pharmaceutics
Application of chromatography in pharmaceuticsApplication of chromatography in pharmaceutics
Application of chromatography in pharmaceuticsMina John
 
Guest Molecule-Responsive Functional CaPhosphonates proton conductivity (JACS)
Guest Molecule-Responsive Functional CaPhosphonates proton conductivity (JACS)Guest Molecule-Responsive Functional CaPhosphonates proton conductivity (JACS)
Guest Molecule-Responsive Functional CaPhosphonates proton conductivity (JACS)maria papadaki
 
OMalley2013BMCL_SS
OMalley2013BMCL_SSOMalley2013BMCL_SS
OMalley2013BMCL_SSSina Sareth
 
Antibody purification – what you need to know to use antibodies effectively
Antibody purification – what you need to know to use antibodies effectivelyAntibody purification – what you need to know to use antibodies effectively
Antibody purification – what you need to know to use antibodies effectivelyExpedeon
 
Seminar on Food and Wine Metabolomics
Seminar on Food and Wine MetabolomicsSeminar on Food and Wine Metabolomics
Seminar on Food and Wine MetabolomicsPanagiotis Arapitsas
 
Trecker_summer_2015_poster
Trecker_summer_2015_posterTrecker_summer_2015_poster
Trecker_summer_2015_posterJohn Trecker
 
Affinity chromatography
Affinity chromatographyAffinity chromatography
Affinity chromatographyasim sami
 
Layer by-layer films of poea- chitosan nanoparticles and their application in...
Layer by-layer films of poea- chitosan nanoparticles and their application in...Layer by-layer films of poea- chitosan nanoparticles and their application in...
Layer by-layer films of poea- chitosan nanoparticles and their application in...Grupo de Pesquisa em Nanoneurobiofisica
 

What's hot (20)

Affinity Chromatography
Affinity ChromatographyAffinity Chromatography
Affinity Chromatography
 
Protein Electrophoresis & Gas Liquid Chromatography & HPLC Applications
 Protein Electrophoresis  & Gas Liquid Chromatography &  HPLC Applications  Protein Electrophoresis  & Gas Liquid Chromatography &  HPLC Applications
Protein Electrophoresis & Gas Liquid Chromatography & HPLC Applications
 
Size exclusion chromatography
Size exclusion chromatographySize exclusion chromatography
Size exclusion chromatography
 
Affinity chromatography
Affinity chromatographyAffinity chromatography
Affinity chromatography
 
ElogDoct: A Tool for Lipophilicity Determination in Drug Discovery. 2. Basic ...
ElogDoct: A Tool for Lipophilicity Determination in Drug Discovery. 2. Basic ...ElogDoct: A Tool for Lipophilicity Determination in Drug Discovery. 2. Basic ...
ElogDoct: A Tool for Lipophilicity Determination in Drug Discovery. 2. Basic ...
 
Ef
EfEf
Ef
 
Affinity Chromoatograpy
Affinity ChromoatograpyAffinity Chromoatograpy
Affinity Chromoatograpy
 
Antibody Glycan Analysis with Normal Phase PhyTip Columns
Antibody Glycan Analysis with Normal Phase PhyTip ColumnsAntibody Glycan Analysis with Normal Phase PhyTip Columns
Antibody Glycan Analysis with Normal Phase PhyTip Columns
 
Application of chromatography in pharmaceutics
Application of chromatography in pharmaceuticsApplication of chromatography in pharmaceutics
Application of chromatography in pharmaceutics
 
Affinity Chromatography
Affinity ChromatographyAffinity Chromatography
Affinity Chromatography
 
Protein purification
Protein purificationProtein purification
Protein purification
 
Affinity chromatography
Affinity chromatographyAffinity chromatography
Affinity chromatography
 
Guest Molecule-Responsive Functional CaPhosphonates proton conductivity (JACS)
Guest Molecule-Responsive Functional CaPhosphonates proton conductivity (JACS)Guest Molecule-Responsive Functional CaPhosphonates proton conductivity (JACS)
Guest Molecule-Responsive Functional CaPhosphonates proton conductivity (JACS)
 
OMalley2013BMCL_SS
OMalley2013BMCL_SSOMalley2013BMCL_SS
OMalley2013BMCL_SS
 
Antibody purification – what you need to know to use antibodies effectively
Antibody purification – what you need to know to use antibodies effectivelyAntibody purification – what you need to know to use antibodies effectively
Antibody purification – what you need to know to use antibodies effectively
 
Seminar on Food and Wine Metabolomics
Seminar on Food and Wine MetabolomicsSeminar on Food and Wine Metabolomics
Seminar on Food and Wine Metabolomics
 
Trecker_summer_2015_poster
Trecker_summer_2015_posterTrecker_summer_2015_poster
Trecker_summer_2015_poster
 
Protein purification
Protein purification Protein purification
Protein purification
 
Affinity chromatography
Affinity chromatographyAffinity chromatography
Affinity chromatography
 
Layer by-layer films of poea- chitosan nanoparticles and their application in...
Layer by-layer films of poea- chitosan nanoparticles and their application in...Layer by-layer films of poea- chitosan nanoparticles and their application in...
Layer by-layer films of poea- chitosan nanoparticles and their application in...
 

Similar to Recent advances in_enzyme_assays_2004

Technologies to study kinases
Technologies to study kinasesTechnologies to study kinases
Technologies to study kinasesCreative-Bioarray
 
Lecture 1 enzyme assays nov02 2007
Lecture 1 enzyme assays nov02 2007Lecture 1 enzyme assays nov02 2007
Lecture 1 enzyme assays nov02 2007mdroslan09
 
Ligand interaction by kk sahu
Ligand interaction by kk sahuLigand interaction by kk sahu
Ligand interaction by kk sahuKAUSHAL SAHU
 
biophysical &biomedical analysis.pptx
biophysical  &biomedical analysis.pptxbiophysical  &biomedical analysis.pptx
biophysical &biomedical analysis.pptxbreenaawan
 
Application of UV-vis in natural product research
Application of UV-vis in natural product researchApplication of UV-vis in natural product research
Application of UV-vis in natural product researchZarTaj2
 
Analysis of bioreactor parameters online and offline
Analysis of bioreactor parameters online and offlineAnalysis of bioreactor parameters online and offline
Analysis of bioreactor parameters online and offlinevikash_94
 
New high thoughput screening copy
New high thoughput screening   copyNew high thoughput screening   copy
New high thoughput screening copyMOHANLAL CHOUDHARY
 
Bioanalytical Method Development and Validation for Simultaneous Estimation o...
Bioanalytical Method Development and Validation for Simultaneous Estimation o...Bioanalytical Method Development and Validation for Simultaneous Estimation o...
Bioanalytical Method Development and Validation for Simultaneous Estimation o...BRNSSPublicationHubI
 
Apoptosis assays detection and methods
Apoptosis assays   detection and methodsApoptosis assays   detection and methods
Apoptosis assays detection and methodsPrabhu Thirusangu
 
Reporter gene by kk sahu sir
Reporter gene by kk sahu sirReporter gene by kk sahu sir
Reporter gene by kk sahu sirKAUSHAL SAHU
 
5 Reasons Aptamers for Lateral Flow
5 Reasons Aptamers for Lateral Flow5 Reasons Aptamers for Lateral Flow
5 Reasons Aptamers for Lateral FlowLisa Thurston
 
Chromatography and types Likhith K
Chromatography and types Likhith KChromatography and types Likhith K
Chromatography and types Likhith KLIKHITHK1
 
Lecture 1 enzyme assays nov02 2007
Lecture 1 enzyme assays nov02 2007Lecture 1 enzyme assays nov02 2007
Lecture 1 enzyme assays nov02 2007Aleesha Ashila
 

Similar to Recent advances in_enzyme_assays_2004 (20)

Technologies to study kinases
Technologies to study kinasesTechnologies to study kinases
Technologies to study kinases
 
Lecture 1 enzyme assays nov02 2007
Lecture 1 enzyme assays nov02 2007Lecture 1 enzyme assays nov02 2007
Lecture 1 enzyme assays nov02 2007
 
Ligand interaction by kk sahu
Ligand interaction by kk sahuLigand interaction by kk sahu
Ligand interaction by kk sahu
 
ABZYMES 2.4.16
ABZYMES 2.4.16ABZYMES 2.4.16
ABZYMES 2.4.16
 
biophysical &biomedical analysis.pptx
biophysical  &biomedical analysis.pptxbiophysical  &biomedical analysis.pptx
biophysical &biomedical analysis.pptx
 
Protein gen
Protein genProtein gen
Protein gen
 
Application of UV-vis in natural product research
Application of UV-vis in natural product researchApplication of UV-vis in natural product research
Application of UV-vis in natural product research
 
Analysis of bioreactor parameters online and offline
Analysis of bioreactor parameters online and offlineAnalysis of bioreactor parameters online and offline
Analysis of bioreactor parameters online and offline
 
IB.pptx
IB.pptxIB.pptx
IB.pptx
 
New high thoughput screening copy
New high thoughput screening   copyNew high thoughput screening   copy
New high thoughput screening copy
 
Bioanalytical Method Development and Validation for Simultaneous Estimation o...
Bioanalytical Method Development and Validation for Simultaneous Estimation o...Bioanalytical Method Development and Validation for Simultaneous Estimation o...
Bioanalytical Method Development and Validation for Simultaneous Estimation o...
 
Kinase Screening
Kinase ScreeningKinase Screening
Kinase Screening
 
Apoptosis assays detection and methods
Apoptosis assays   detection and methodsApoptosis assays   detection and methods
Apoptosis assays detection and methods
 
Hplc final
Hplc finalHplc final
Hplc final
 
Reporter gene by kk sahu sir
Reporter gene by kk sahu sirReporter gene by kk sahu sir
Reporter gene by kk sahu sir
 
Bmm480 Enzymology lecture-6
Bmm480 Enzymology lecture-6Bmm480 Enzymology lecture-6
Bmm480 Enzymology lecture-6
 
5 Reasons Aptamers for Lateral Flow
5 Reasons Aptamers for Lateral Flow5 Reasons Aptamers for Lateral Flow
5 Reasons Aptamers for Lateral Flow
 
Chromatography and types Likhith K
Chromatography and types Likhith KChromatography and types Likhith K
Chromatography and types Likhith K
 
Lecture 1 enzyme assays nov02 2007
Lecture 1 enzyme assays nov02 2007Lecture 1 enzyme assays nov02 2007
Lecture 1 enzyme assays nov02 2007
 
Enzyme labeling
Enzyme labelingEnzyme labeling
Enzyme labeling
 

Recently uploaded

main PPT.pptx of girls hostel security using rfid
main PPT.pptx of girls hostel security using rfidmain PPT.pptx of girls hostel security using rfid
main PPT.pptx of girls hostel security using rfidNikhilNagaraju
 
Architect Hassan Khalil Portfolio for 2024
Architect Hassan Khalil Portfolio for 2024Architect Hassan Khalil Portfolio for 2024
Architect Hassan Khalil Portfolio for 2024hassan khalil
 
MANUFACTURING PROCESS-II UNIT-2 LATHE MACHINE
MANUFACTURING PROCESS-II UNIT-2 LATHE MACHINEMANUFACTURING PROCESS-II UNIT-2 LATHE MACHINE
MANUFACTURING PROCESS-II UNIT-2 LATHE MACHINESIVASHANKAR N
 
Introduction to IEEE STANDARDS and its different types.pptx
Introduction to IEEE STANDARDS and its different types.pptxIntroduction to IEEE STANDARDS and its different types.pptx
Introduction to IEEE STANDARDS and its different types.pptxupamatechverse
 
Microscopic Analysis of Ceramic Materials.pptx
Microscopic Analysis of Ceramic Materials.pptxMicroscopic Analysis of Ceramic Materials.pptx
Microscopic Analysis of Ceramic Materials.pptxpurnimasatapathy1234
 
Software Development Life Cycle By Team Orange (Dept. of Pharmacy)
Software Development Life Cycle By  Team Orange (Dept. of Pharmacy)Software Development Life Cycle By  Team Orange (Dept. of Pharmacy)
Software Development Life Cycle By Team Orange (Dept. of Pharmacy)Suman Mia
 
Call Girls Delhi {Jodhpur} 9711199012 high profile service
Call Girls Delhi {Jodhpur} 9711199012 high profile serviceCall Girls Delhi {Jodhpur} 9711199012 high profile service
Call Girls Delhi {Jodhpur} 9711199012 high profile servicerehmti665
 
Extrusion Processes and Their Limitations
Extrusion Processes and Their LimitationsExtrusion Processes and Their Limitations
Extrusion Processes and Their Limitations120cr0395
 
Coefficient of Thermal Expansion and their Importance.pptx
Coefficient of Thermal Expansion and their Importance.pptxCoefficient of Thermal Expansion and their Importance.pptx
Coefficient of Thermal Expansion and their Importance.pptxAsutosh Ranjan
 
(MEERA) Dapodi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts
(MEERA) Dapodi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts(MEERA) Dapodi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts
(MEERA) Dapodi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escortsranjana rawat
 
the ladakh protest in leh ladakh 2024 sonam wangchuk.pptx
the ladakh protest in leh ladakh 2024 sonam wangchuk.pptxthe ladakh protest in leh ladakh 2024 sonam wangchuk.pptx
the ladakh protest in leh ladakh 2024 sonam wangchuk.pptxhumanexperienceaaa
 
SPICE PARK APR2024 ( 6,793 SPICE Models )
SPICE PARK APR2024 ( 6,793 SPICE Models )SPICE PARK APR2024 ( 6,793 SPICE Models )
SPICE PARK APR2024 ( 6,793 SPICE Models )Tsuyoshi Horigome
 
What are the advantages and disadvantages of membrane structures.pptx
What are the advantages and disadvantages of membrane structures.pptxWhat are the advantages and disadvantages of membrane structures.pptx
What are the advantages and disadvantages of membrane structures.pptxwendy cai
 
GDSC ASEB Gen AI study jams presentation
GDSC ASEB Gen AI study jams presentationGDSC ASEB Gen AI study jams presentation
GDSC ASEB Gen AI study jams presentationGDSCAESB
 
High Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur EscortsHigh Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur Escortsranjana rawat
 
Call Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur Escorts
Call Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur EscortsCall Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur Escorts
Call Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur EscortsCall Girls in Nagpur High Profile
 
High Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur EscortsHigh Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur EscortsCall Girls in Nagpur High Profile
 

Recently uploaded (20)

main PPT.pptx of girls hostel security using rfid
main PPT.pptx of girls hostel security using rfidmain PPT.pptx of girls hostel security using rfid
main PPT.pptx of girls hostel security using rfid
 
Architect Hassan Khalil Portfolio for 2024
Architect Hassan Khalil Portfolio for 2024Architect Hassan Khalil Portfolio for 2024
Architect Hassan Khalil Portfolio for 2024
 
MANUFACTURING PROCESS-II UNIT-2 LATHE MACHINE
MANUFACTURING PROCESS-II UNIT-2 LATHE MACHINEMANUFACTURING PROCESS-II UNIT-2 LATHE MACHINE
MANUFACTURING PROCESS-II UNIT-2 LATHE MACHINE
 
Introduction to IEEE STANDARDS and its different types.pptx
Introduction to IEEE STANDARDS and its different types.pptxIntroduction to IEEE STANDARDS and its different types.pptx
Introduction to IEEE STANDARDS and its different types.pptx
 
Microscopic Analysis of Ceramic Materials.pptx
Microscopic Analysis of Ceramic Materials.pptxMicroscopic Analysis of Ceramic Materials.pptx
Microscopic Analysis of Ceramic Materials.pptx
 
Software Development Life Cycle By Team Orange (Dept. of Pharmacy)
Software Development Life Cycle By  Team Orange (Dept. of Pharmacy)Software Development Life Cycle By  Team Orange (Dept. of Pharmacy)
Software Development Life Cycle By Team Orange (Dept. of Pharmacy)
 
Exploring_Network_Security_with_JA3_by_Rakesh Seal.pptx
Exploring_Network_Security_with_JA3_by_Rakesh Seal.pptxExploring_Network_Security_with_JA3_by_Rakesh Seal.pptx
Exploring_Network_Security_with_JA3_by_Rakesh Seal.pptx
 
Call Girls Delhi {Jodhpur} 9711199012 high profile service
Call Girls Delhi {Jodhpur} 9711199012 high profile serviceCall Girls Delhi {Jodhpur} 9711199012 high profile service
Call Girls Delhi {Jodhpur} 9711199012 high profile service
 
Extrusion Processes and Their Limitations
Extrusion Processes and Their LimitationsExtrusion Processes and Their Limitations
Extrusion Processes and Their Limitations
 
Coefficient of Thermal Expansion and their Importance.pptx
Coefficient of Thermal Expansion and their Importance.pptxCoefficient of Thermal Expansion and their Importance.pptx
Coefficient of Thermal Expansion and their Importance.pptx
 
(MEERA) Dapodi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts
(MEERA) Dapodi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts(MEERA) Dapodi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts
(MEERA) Dapodi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts
 
the ladakh protest in leh ladakh 2024 sonam wangchuk.pptx
the ladakh protest in leh ladakh 2024 sonam wangchuk.pptxthe ladakh protest in leh ladakh 2024 sonam wangchuk.pptx
the ladakh protest in leh ladakh 2024 sonam wangchuk.pptx
 
SPICE PARK APR2024 ( 6,793 SPICE Models )
SPICE PARK APR2024 ( 6,793 SPICE Models )SPICE PARK APR2024 ( 6,793 SPICE Models )
SPICE PARK APR2024 ( 6,793 SPICE Models )
 
What are the advantages and disadvantages of membrane structures.pptx
What are the advantages and disadvantages of membrane structures.pptxWhat are the advantages and disadvantages of membrane structures.pptx
What are the advantages and disadvantages of membrane structures.pptx
 
Roadmap to Membership of RICS - Pathways and Routes
Roadmap to Membership of RICS - Pathways and RoutesRoadmap to Membership of RICS - Pathways and Routes
Roadmap to Membership of RICS - Pathways and Routes
 
GDSC ASEB Gen AI study jams presentation
GDSC ASEB Gen AI study jams presentationGDSC ASEB Gen AI study jams presentation
GDSC ASEB Gen AI study jams presentation
 
High Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur EscortsHigh Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur Escorts
 
Call Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur Escorts
Call Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur EscortsCall Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur Escorts
Call Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur Escorts
 
★ CALL US 9953330565 ( HOT Young Call Girls In Badarpur delhi NCR
★ CALL US 9953330565 ( HOT Young Call Girls In Badarpur delhi NCR★ CALL US 9953330565 ( HOT Young Call Girls In Badarpur delhi NCR
★ CALL US 9953330565 ( HOT Young Call Girls In Badarpur delhi NCR
 
High Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur EscortsHigh Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur Escorts
 

Recent advances in_enzyme_assays_2004

  • 1. Recent advances in enzyme assays Jean-Philippe Goddard and Jean-Louis Reymond Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland Enzyme assays for high-throughput screening and enzyme engineering, which are often based on deriva- tives of coumarin, nitrophenol, fluorescein, nitrobenzo- furazane or rhodamine dyes, can be divided into two categories: those that depend on labelled substrates, and those that depend on sensing the reactions of unmodified substrates. Labelled substrates include, for example, fluorogenic and chromogenic substrates that generate a reporter molecule by b-elimination, fluor- escence resonance energy transfer (FRET) substrates and isotopic labels for enantioselectivity screening. By contrast, endpoint sensing can be done using amine reagents, fluorescent affinity labels for phosphorylated proteins, or synthetic multifunctional pores. Sensing assays can also be done in real time by using, for example, aldehyde trapping to follow vinyl ester acyla- tion in organic solvent or calcein–copper fluorescence for sensing amino acids. The current trend is to assem- ble many such assays in parallel for enzyme profiling and enzyme fingerprinting. Enzyme assays are experimental protocols that make enzyme-catalysed chemical transformations visible. Such assays are very important for high-throughput screening in the context of drug discovery. Recently, much effort has been directed at improving enzyme assays for enzyme engineering, where they provide the functional basis for identifying and selecting new enzymes, most often in screens of either large sample libraries, such as micro- organism collections from the biosphere or a series of enzyme mutants generated by genetic recombination methods, such as gene shuffling and error-prone PCR [1–6]. The well-known adage ‘you get what you screen for’ clearly states what assays are all about: they must closely reproduce the desired catalytic reaction. An enzymatic transformation can be traced easily if the enzyme is highly active and present in large amounts. In most applications, however, the aim is the identification of a dilute, and therefore weak, enzymatic activity in a reaction medium that contains many other components. In addition tobeingcrucialforidentifyingfunction,detectinganenzyme by means of its catalytic action on a substrate is actually very advantageous in terms of sensitivity, because a single enzyme molecule can generate many product molecules through enzymatic turnover. Thus, a signal amplification effect is intrinsically present in any enzyme assay that is based on substrate turnover. A good fluorogenic or chromo- genic assay can often detect an enzyme below the protein detection limit and even in a mixture of other proteins and enzymes, such as a crude bacterial lysate. For whichever application they are designed, enzyme assays should be simple and robust. Technical parameters must be fulfilled, such as a low rate of false-positive and false-negative signals under the real assay conditions. Even if these criteria are fulfilled, whether an assay will be used or not is decided by the availability of the assay reagents, their price, and their overall simplicity. Here we review recent progress that has been made in enzyme assays; we refer the reader to earlier reviews for a more comprehensive coverage of the field [7–12]. The examples cover a broad spectrum of possibilities that can be exploited to build enzyme assays. We have classified enzyme assays according to the nature of the substrate used. We distinguish between enzyme assays based on substrates that have been modified with a label and those using sensors that can operate with natural, unmodified substrates. This distinction makes a lot of sense in the perspective of practical application, because very often a sensor-based system does not require any synthesis and turns out to be cheaper and more versatile. Nevertheless, assays based on labelled substrates can be more sensitive to low enzyme turnover and more selective. We emphasize assays that detect enzymatic turnover, but there is another technique for identifying enzymes that is based on covalent labelling by active-site-directed probes, followed by separation by gel electrophoresis. This type of analysis is relevant to proteome analysis and has been reviewed recently [13]. Assays with labelled substrates Enzyme assays using labelled synthetic substrates are advantageous in that they usually provide a very direct connection between enzymatic activity and the signal. Such assays are resistant to artefacts, in particular in the context of enzyme inhibition assays. The use of labelled substrates, however, can be undesirable when screening for biocatalytic transformations. Synthetic labelled sub- strates include fluorogenic and chromogenic substrates, isotopically labelled substrates, FRET substrates, and substrates with fluorescent labels for indirect detection. Fluorogenic and chromogenic substrates by b-elimination Esters and ethers of nitrophenol and umbelliferone, and amides of nitroaniline and aminocoumarin have been known for many years to be chromogenic or fluorogenic substrates for glycosidases, lipases, esterases and pro- teases (Figure 1). Assays based on these substrates are problematic, however, because the phenolate or aniline Corresponding author: Jean-Louis Reymond ( jean-louis.reymond@ioc.unibe.ch). Available online 30 April 2004 Review TRENDS in Biotechnology Vol.22 No.7 July 2004 www.sciencedirect.com 0167-7799/$ - see front matter q 2004 Elsevier Ltd. All rights reserved. doi:10.1016/j.tibtech.2004.04.005
  • 2. leaving group is at least five orders of magnitude more reactive as a leaving group than is the aliphatic alcoholate or amine that is present in the natural substrates of the enzymes. These substrates are therefore often unstable and susceptible to cleavage by non-catalytic contaminants. In addition, substrate structure cannot be diversified in proximity to the enzyme-labile bond. In 1998, Klein and Reymond [14] reported a new type of fluorogenic substrate that uses phenolate release as signal. First, the chiral secondary alcohol (S)-(4) (Figure 2) was used as a fluorogenic substrate for alcohol dehydro- genase. The primary enzyme reaction product is a ketone (5), which subsequently undergoes a b-elimination reac- tion to liberate the fluorescent umbelliferone reporter molecule (Figure 2). Furthermore, a double indirect pro- cedure facilitated the assay of lipases with the correspond- ing esters [15]. This strategy efficiently decouples the phenolate release chemistry from the enzymatic trans- formation and facilitates structural and functional diver- sification of the substrates. The b-elimination approach has been used in substrates for various enzyme assays, Figure 1. Chromogenic and fluorogenic substrates with activated leaving groups. Substrates are shown for glycosidases (1), proteases (2) and lipases and esterases (3). The enzyme-labile bond is shown in red and the chromophore in blue. TRENDS in Biotechnology H N N H O O NH NH2HN O OO O 1 β-glucosidase 2 Trypsin 3 Lipases NO2 O OH HO HO OH O NO2 Figure 2. Fluorogenic enzyme substrates with indirect fluorophore release by secondary b-elimination. The chiral secondary alcohol (S)-(4) is used as a fluorogenic substrate for alcohol dehydrogenase (ADH). The primary enzyme reaction product is a ketone (5), which subsequently undergoes b-elimination accelerated by bovine serum albumin (BSA) to liberate the fluorescent umbelliferone reporter molecule. This approach has been used in assays for various enzymes, including epoxide hydro- lases (6), transadolases (7) transketolases (8) and Baeyer–Villigerases (9). An assay for b-lactamase (10) operates by a similar release mechanism for umbelliferone. In addition, a probe for quantifying NADH and NADPH relies on hydride transfer and b-elimination (11). The enzyme-labile bond, the enzymes and (where needed) secondary reagents are shown in red. Abbreviations: Obn, benzyloxy; TfO, trifluoromethansulfonate. TRENDS in Biotechnology O OO OCoum O H (S)-4 OCoum O OO OCoum N N OCoum N S H N O O Ph OBnO OCoum TfO ADH NAD(P)+ BSA, pH>7 5 Umbelliferone anion (blue fluorescent) 10 β-lactamase 6 Epoxide hydrolase + NaIO4 9 Baeyer–Villigerase, + esterase + ADH/NAD+ 8 Transketolase 7 Transaldolase Coum O CoumO OH OH HO OH 11 NADH/NADPH HO OCoum O OH OH O O H OH Review TRENDS in Biotechnology Vol.22 No.7 July 2004364 www.sciencedirect.com
  • 3. including assays for aldolase catalytic antibodies [16–18], epoxide hydrolases (6) [19], transadolases (7) [20], lipases and esterases [19,21,22], phosphatases [19,23], acylases [19], proteases [24], transketolases (8) [25] and Baeyer– Villigerases (9) [26]. The recently reported b-lactamase substrate (10) operates by a similar release mechanism for umbelliferone [27] (Figure 2). FRET substrates One of the most efficient principles for detecting cleavage reactions is the use of FRET substrates, whereby a fluoro- phore quencher or fluorophore–fluorophore pair is sepa- rated by the cleavage of an enzyme-labile bond, resulting in either an increase in fluorescence or a shift in wavelength. This principle was originally reported for the detection of HIV protease activities [28]. The same approach has been used recently in an all- protein construct to detect protease activity using two- photon cross-correlation and FRET analysis [29]. This irradiation technique is milder than the direct fluorophore excitation used in classical protease assays. FRET sub- strates are not limited to protease-type cleavage, and an elegant system with FRET phospholipids has been recently used for a genetic analysis of phospholipase activity by in vivo imaging [30]. Other signalling substrates Several other recently reported fluorogenic and chromo- genic substrates are interesting in terms of their particu- lar chromogenic mechanism (Figure 3). In early 1990, it was shown that the changes in the ultraviolet/visible and fluorescence properties of 6-methoxynaphthyl-methanol (12) (Figure 2) on oxidation to the corresponding aldehyde (13) could be exploited to provide a fluorogenic alcohol dehydrogenase assay [31] (Figure 3). This assay has been subsequently adapted for assaying retroaldolase catalytic antibodies [18] and, together with a secondary oxidation with sodium periodate, for lipases and epoxide hydrolases [32]. Styrene oxides can be used directly as chromogenic substrates for epoxide hydrolases in a similar set-up [33]. In the area of lipase assays, acyloxymethylethers of umbelliferone such as (14) (Figure 3), which were origin- ally developed as fluorogenic probes for catalytic anti- bodies [34], have been recently found to be excellent substrates for lipases and esterases [35]. The advantage of these substrates is that the leaving group is an aliphatic alcohol that is much less acidic than umbelliferone itself, which reduces susceptibility to non-catalytic cleavage. The non-fluorescent N-aryl maleimide (15) (Figure 3) can be used to follow C–C bond formation reactions, such as antibody-catalysed Michael additions and Diels–Alder reactions, because the corresponding maleimide adducts such as (16) are fluorescent [36] (Figure 3). A potential problem with this assay is the fact that the maleimide reagent (16) is a highly reactive electrophile and readily adds to thiols to form a fluorescent product. In an elegant solid-phase assay, Yeo and Mrksich [37] have monitored the activity of the lipase cutinase by using a mono-ester of para-hydroxyphenol as a substrate immobilized on an electrode surface by a sulphur–gold interaction [37]. The para-hydroxyphenol released by hydrolysis is detected electrochemically by oxidation to the quinone, facilitating the voltametric quantification of enzymatic activity and thereby demonstrating a principle that might provide a general solution for the electro- chemical sensing of enzyme activities. 13 C- and 2 H-labelled pseudoenantiomers One of the key problems in enzyme assays for biocatalysis is the ability to detect enantioselectivity directly in high throughput, but Reetz and co-workers [38–40] have pro- posed various solutions based on isotopic labelling. For example, enantioselective isotopic labelling of one enan- tiomer of a chiral acetyl ester or amide has been used to trace the enzymatic kinetic resolutions of these substrates by enantioselective hydrolysis. Isotopic 13 C or 2 H labelling of the acetyl group produces no chemical reactivity changes between the enantiomers, but facilitates the selective tracing of each enantiomeric substrate or product by mass spectrometry [38], 1 H-NMR [39] or Fourier transform infrared spectroscopy [40]. The methods can be implemented in high throughput by using appropri- ately robotized instruments to screen libraries of enzyme mutants [41]. These approaches are certainly powerful, but they require expensive isotopic labelling reagents and possibly the modification of rather expensive instruments for high- throughput use, which renders them off-limits for many Figure 3. Additional types of fluorogenic substrate. 6-methoxynaphthylmethanol (12) is a fluorogenic substrate for alcohol dehydrogenase. The coumarinoxymethyl ester (14) is a low-reactivity fluorogenic substrate for lipases. The fluorogenic maleimide probe (15) reacts with nucleophiles and allows screening of aldolase catalytic antibodies (Ab) for C–C bond formation. Abbreviations: ADH, alcohol dehydrogenase; Cat. Ab., catalytic antibody; MeO, methoxy. TRENDS in Biotechnology O H CoumO ADH, NAD+ O OOO O MeO OH N H N N O O O MeO O Ar N O O O 15 Non-fluorescent 12 Non-fluorescent 14 Non-fluorescent 13 Blue fluorescent Blue fluorescent Lipase or esterase Cat. Ab. 16 Fluorescent Coum Ar Review TRENDS in Biotechnology Vol.22 No.7 July 2004 365 www.sciencedirect.com
  • 4. laboratories. Reetz and colleagues have also implemented other instrumental methods for following enantioselective reactions in high throughput, including NMR analysis of Mosher’s esters [39], and monitoring the differential cell growth in media containing either enantiomer of a fluoroacetate ester that releases the toxic fluoroacetic acid on hydrolysis [42]. Fluorogenic substrates for kinases and phosphatases Several kinase and phosphatase assays based on labelled substrates have been reported recently, and these deserve to be discussed separately. Interest in kinases and phos- phatases is mainly linked to screening for inhibitors of these enzymes, which control signal transduction cascades for gene activation. The family of kinases and phospha- tases represents up to 5% of the human genome and thus is an important target for drug discovery [43]. One kinase assay is based on the activity of an aminopeptidase for the fluorogenic release of rhodamine from a phosphorylated labelled peptide (18), which is diminished relative to its activity towards the non- phosphorylated peptide (17) [44] (Figure 4). This set-up is similar to a patented assay based on the differential protease sensitivity of phosphorylatable FRET peptides, which has been commercialized by Invitrogen under the trade name Z0 -Lyte [45]. Similarly, the differential reac- tivity of chymotrypsin has been used to follow cis–trans prolyl isomerase activity [46–48]. Whereas the above assays are based on the reactivity of a second enzyme to trigger fluorophore release, a much simpler solution for assaying kinases has been developed by Lawrence and co-workers [49], who detect intracellular protein kinase activity in the form of a short synthetic peptide (19) containing a caged serine residue in the vicinity of a nitrobenzofurazane fluorophore (Figure 4). After irradiation, phosphorylation of the liberated serine residue by the kinase induces a threefold increase in fluorescence in the fluorophore. Shults and Imperiali [50] have reported another very elegant solution for assaying kinases. They found that peptides containing a phosphory- latable amino acid followed by a b-turn and a ‘Sox’ amino acid operate as fluorogenic kinase substrates owing to the phosphorylation-induced binding of magnesium ions by the Sox chromophore, which results in an increase in fluorescence [50]. In a completely different assay design, a fluorescent label in a peptide substrate has been used to report binding of the phosphorylated peptide to a macromolecule via induced changes in fluorescence polarization. This assay enables researchers to detect the activity of proteases, kinases and phosphatases by following changes in the fluorescence polarization induced by binding of the phos- phorylated peptides to poly-arginine [51]. The similar, immobilized metal assay platform method detects an increase in fluorescence polarization in fluorescently labelled peptides when they bind, in their phosphorylated form, to nanoparticles via metal coordination [52]. Other kinase assays based on indirect sensing are discussed below. Sensing reactions of unmodified substrates Many enzyme assays have been reported that come very close to the ideal situation: that is, they detect transform- ation of the enzyme’s natural or targeted substrate itself. This is often realizable by employing an analytical approach such as gas chromatography, high-performance liquid chromatography, mass spectrometry or NMR, and the use of these methods is very common within the constraints of industrial applications. Below we focus on methods that produce a colour or a fluorescent change by a chemical process within the test solution and have the potential for very high throughput at low cost. These assays are based on chemosensors that respond to product formation independently of any label in the substrate or product during the enzymatic transform- ation. The prototypical application of this idea is the use of a chromogenic pH indicator to follow ester hydrolysis by lipases – a concept that has been elegantly implemented by Kazlauskas and co-workers [53,54] to screen for enan- tioselective lipases. Such sensors have also been used in the context of enzymes encapsulated in a sol-gel [55]. Endpoint assays The endpoint determination of substrate consumption or product formation is almost always sufficient for assaying enzyme activities, and it can be realized by applying a Figure 4. Fluorogenic probes for kinases. The bis-peptidyl rhodamine substrate undergoes fluorogenic cleavage by aminopeptidase in its non-phosphorylated state (17), but not in its phosphorylated state (18), enabling indirect quantification of phosphorylation by kinases. On phosphorylation by kinases, the caged serine peptide (19) is deprotected photochemically and becomes fluorescent at the nitro- benzofurazane chromophore on. Other kinase assays are discussed in the text. TRENDS in Biotechnology O2N OMe OMe O N H Phe-Arg-Arg-Arg-Arg-Lys-NH2 O N O N O2N O O O H N H N Leu-...-Ser-Leu-Gly Gly-Leu-Ser-...-Leu OH2N NH2 + CO2 – 2 Aminopeptidase Rhodamine (red fuorescent) 18 (X = PO3H– ) hν 19 O O 17 (X = H) 1 Kinase X X Kinase phosphorylation site Review TRENDS in Biotechnology Vol.22 No.7 July 2004366 www.sciencedirect.com
  • 5. reagent that specifically detects a functional group that is formed in the reaction. A straightforward application of this principle has been reported recently by Henke and Bornscheuer [56] for the detection of amidase activities. The amine product that is formed is detected by the fluoro- genic amine-reagent 4-nitro-7-chloro-benzofurazane. This method has been applied to the enantioselective screening of different hydrolases. The broadly applicable probe for quantifying NADH and NADPH, which relies on an ele- gant hydride-transfer/b-elimination sequence as discussed above, represents a more subtle application of the principle of product detection by quantitative reaction [57] (Figure 2). Classically, the activity of protein kinases has been analysed by incubating immobilized proteins or peptides with radiolabelled ATP, or by staining the product with antibodies specific for the phosphoprotein. A recent improve- ment in protein kinase activity detection has been made with the development of a phosphoprotein-selective fluorescent dye called Pro-Q Diamond. This dye is suitable for staining protein microarrays as an endpoint treatment after phosphorylation with ATP and a kinase [58], and thus should facilitate the identification of kinase sub- strates. A similar dye that is based on a zinc fluorescein complex and binds binding to phosphorylated peptides has been reported recently [59]. All of these assays require an immobilized substrate, usually a whole protein, because the excess ATP reagent and the kinase must be washed away before the phosphate-staining reagent can be applied. Matile and co-workers [60,61] have reported an elegant assay for substrate versus product quantification that uses synthetic multifunctional pores (SMPs) consisting of syn- thetic peptide-appended oligophenylene building blocks and can be used to monitor the activity of various enzymes. The SMPs are incorporated into the membrane of vesicles loaded with fluorescein, where they function as channels for the escape of this fluorophore, which results in an increase in fluorescence because dilution removes auto- quenching. Substrate/product ratios in an enzymatic reaction can be monitored whenever substrate and pro- duct differentially modulate the flow offluorescein through the SMPs – for example, during the hydrolysis of DNA by DNases or of peptides by proteases. Although this set-up is elegant, the availability of the pore component, which must be prepared by complex multistep syntheses, and the robustness of the system to unselective pore blockage by contaminants such as the enzymes and cofactors them- selves represent potential limitations. Indirect product detection as an endpoint is also possible by the principle of back-titration. Wahler and Reymond [62] have demonstrated this principle in a versatile colorimetric assay based on the quantification of periodate-sensitive reaction products formed by the enzymatic hydrolysis of periodate-resistant substrates (Figure 5). Sodium periodate is either consumed by the product formed or used to oxidize adrenaline (20) to form the deeply red coloured adrenochrome (21) (Figure 5). Product formation is proportional to the decrease in adrenochrome formation. The assay is compatible with various operating con- ditions, including co-solvents and extreme pH values, and has been used to test the hydrolysis of vegetal oil and tributyrin by lipases and esterase, the opening of various aliphatic and aromatic epoxides by epoxide hydrolases, and the hydrolysis of phytic acid by phytases. It has been applied to the rapid screening of esterases against an array of carbohydrate acetates [63]. This so-called ‘adrenaline test’ for enzyme is surprisingly versatile and works with inexpensive reagents, so that it can be implemented almost anywhere. One of its limitations is that the periodate reagents also reacts with any other diol that might be present in the medium, in particular glycerol, which is often used for enzyme cryopreservation. It is also possible to detect the carbonyl products of periodate cleavage by using a colorimetric Schiff base reagent, and this has been demonstrated for an epoxide hydrolase assay [64]. It must be mentioned, however, that detecting volatile aliphatic aldehydes is problematic in a microtitre plate owing to cross-diffusion. The assay has a detection limit in the 0.01 M range, which is 10- to 50-fold less sensitive that the adrenaline test based on back-titration. Real-time assays A few sensor systems enable enzymatic reactions to be monitored in real time. This is possible whenever the sensor does not interact with the enzymatic reaction. For example, Konarzycka–Bessler and Bornscheuer [65] have reported an assay for the lipase-catalysed esterification of alcohols by vinyl acetate in organic solvent. This assay is based on the real-time trapping of the released acet- aldehyde by non-fluorescent 4-hydrazino-7-nitro-benzo- furazane to form a fluorescent hydrazone. The activity of Figure 5. The adrenaline test for enzymes. Sodium periodate is consumed by oxidation of either an oxidizable diol or an amino alcohol product of the enzyme reaction. Back-titration of the unreacted adrenaline (20) to form the deeply coloured adrenochrome (21) enables quantification of the extent of product formation. TRENDS in Biotechnology S P Step 1: Enzyme reaction NaIO4 resistant substrate: Tributyrin (esterases) Triglycerides (lipases) Epoxides (EH) Phytic acid (phytases) NaIO4-sensitive product: 1, 2-diol NaIO3 + H2O NaIO4 OH HO MeHN OH 20 Colorless O O N HO Me 21 Deep red color Step 3: Add adrenaline Step 2: Product oxidation Review TRENDS in Biotechnology Vol.22 No.7 July 2004 367 www.sciencedirect.com
  • 6. telomerase, a DNA polymerase whose detection might be useful in cancer diagnosis and which is a possible target for chemotherapy, has been followed on a biosensor chip by detecting elongation of a surface immobilized telomeric repeat primer directly using surface plasmon resonance (Biacore) [66]. DNA nucleases, also called ‘DNases’, have been recently assayed using the double-helix-selective dye PicoGreen (which is also used in real-time PCR) by following the fluorescence intensity decrease induced by dye release [67]. Building on earlier work with a quinacridone fluoro- phore [68–69], we and our co-workers [70] have recently found that a non-fluorescent calcein–copper complex, a fluorescein derivative, can be used as a selective dynamic sensor for amino acids. The sensor is used at a micromolar or submicromolar concentration and responds to amino acids in the 0.1–1 mM range by showing a tenfold increase in its intensity of green fluorescence as the quenching copper ion is displaced from the calcein fluorophore (22) (Figure 6) by the chelating amino acid. This fluorescent Figure 6. Calcein–copper as a selective dynamic sensor of amino acids. (a) Principle of detecting amino acid products by calcein–copper. Copper(II) ions quench the fluorescence of calcein when bound to the fluorophore. Calcein (22), used in the 1027 –1026 M concentration range, is released when copper ions (5 £ 1026 M) are chelated by amino acids (AA) in the 1024 –1023 M concentration range. This assay facilitates the direct detection of aminopeptidase, acylase and protease activities. (b) Time course of fluorescence increase in a calcein–copper assay for the hydrolysis of N-acetyl L-methionine to L-methionine by acylase I. Conditions: 10 mM N-acetyl-L-methionine in aqueous 5 mM Bis-tris buffer (pH 7.2), 25 8C, 1 mM calcein, 5 mM CuCl2, lem ¼ 530 ^ 25 nm, lex ¼ 450 ^ 50 nm, and either no enzyme (black) or acylase I at 2.5 mg/ml (green), 5.0 mg/ml (blue) or 10 mg/ml (red). Abbreviations: lem, emission wavelength; lex, excitation wavelength. TRENDS in Biotechnology OHO O N CO2H CO2H NHO2C HO2C CO2H H2N O OH R AA [CuAA2] Non-chelating derivative (N-acyl amino acid, amino acid amide, protein) Enzyme 22 Fluorescent [Cu.22] Non-fluorescent 22 (λex = 450 nm, λem = 530 nm) (a) (b) 0 10000 5000 15000 0 3600 Time (s) 7200 Fluorecence Review TRENDS in Biotechnology Vol.22 No.7 July 2004368 www.sciencedirect.com
  • 7. sensor system has been used to monitor the activity of acylases, aminopeptidases and proteases (Figure 6). Indeed, these enzymes release free amino acids from non-chelating amide precursor substrates such as N-acetyl-methionine, L-leucinamide and whole bovine serum albumin (BSA) protein. Similar to the adrenaline test discussed above, the calcein–copper assay uses only commercially available, inexpensive reagents and can be implemented almost anywhere. It cannot be used in the presence of metal- chelating species such as EDTA, however, which also induce a fluorescence increase. If the enzyme is sensitive to copper ions, the assay can be used as an endpoint measurement – for example, it can be used to assay cysteine proteases. Outlook: enzyme activity profiles and fingerprints An enzyme assay not only detects the enzyme, but naturally also indicates the enzyme type by the substrate that is used. Currently, the general trend beyond the development of enzyme assays is to assemble many such assays in a parallel format to generate the complete reactivity profile of an enzyme [71]. Multienzyme profiling with chromogenic substrates was developed in the 1960s as a tool for identifying microorganisms [72], and today it forms the basis for medical diagnostics of infectious diseases in hospitals. In the 1990s the substrate specificity of proteases was investigated by using combinatorial libraries of fluorogenic peptides [73,74]. Recently, we and our co-workers [32,62,75] have shown that assays with multiple fluorogenic substrates can be carried out rapidly in microtitre-plate format to generate activity profiles of many different hydrolytic enzymes. Because these profiles are recorded easily and reproducibly, they can be con- sidered ‘fingerprints’ of these enzymes. They also provide a rapid and versatile tool for the functional classification of enzymes such as various lipases and esterases [76]. The operational simplicity of these fingerprinting experiments can be improved by using substrates arrayed on a solid support such as a glass surface, as shown recently by Ellman and co-workers [77] for peptide micro- arrays for determining protease substrate specificity. Furthermore, Yao and co-workers [78] have adapted periodate/b-elimination-triggered fluorogenic substrates for epoxide hydrolases, amidases, esterases and phospha- tases to a similar glass-supported format. Such enzyme fingerprints might form the basis for a new type of quality control and diagnostic application. Concluding remarks The field of enzyme assays has undergone tremendous developement in recent years due to the growing import- ance of high-throughput screening in drug discovery and enzyme engineering. Enzyme assays are conceptually close to the general problem of designing selective chemo- sensors for small molecules. Enzyme assays have provided a fertile ground for the realization of creative ideas in system design, and a surprising diversity of formats have been imagined to make enzyme reactions visible. Thus, time-honored ‘classics’ such as nitrophenyl esters and ethers and alcohol dehydrogenase-coupled assays, which a few years ago represented almost the entire known chemistry of enzyme assays, have made room for a broad variety of labels, triggering chemistries, and signalling systems. There is no doubt that the field will continue to evolve as newer and better methods appear for screening enzymes. Challenges abound because many reaction types are still difficult to assay in high-throughput or with a sufficient level of accuracy. Acknowledgements This work was supported by the Swiss National Science Foundation, COST Action D16, the Swiss OFES and Prote´us SA, Nıˆmes, France. References 1 Marrs, B. et al. (1999) Novel approaches for discovering industrial enzymes. Curr. Opin. Microbiol. 2, 241–245 2 Stemmer, W.P.C. (1994) DNA shuffling by random fragmentation and reassembly: in vitro recombination for molecular evolution. Proc. Natl. Acad. Sci. U. S. A. 91, 10747–10751 3 Stemmer, W.P.C. (1994) Rapid evolution of a protein in vitro by DNA shuffling. Nature 370, 389–391 4 You, L. and Arnold, F.H. (1996) Directed evolution of subtilisin E in Bacillus subtilis to enhance total activity in aqueous dimethylforma- mide. Protein Eng. 9, 77–83 5 Reetz, M.T. and Jaeger, K-E. (2000) Enantioselective enzymes for organic synthesis created by directed evolution. Chemistry 6, 407–412 6 Taylor, S.V. et al. (2001) Investigating and engineering enzymes by genetic selection. Angew. Chem. Int. Ed. Engl. 40, 3310–3335 7 Eisenthal, R. and Danson, M. eds (2002) Enzyme Assays: a Practical Approach, Oxford University Press 8 Gul, S. et al. (1998) Enzyme Assays Essential Data, John Wiley & Sons 9 Reetz, M.T. (2001) Combinatorial and evolution-based methods in the creation of enantioselective catalysts. Angew. Chem. Int. Ed. Engl. 40, 284–310 10 Wahler, D. and Reymond, J-L. (2001) Novel methods for biocatalysts screening. Curr. Opin. Chem. Biol. 5, 152–158 11 Beisson, F. et al. (2000) Methods for lipase detection and assay: a critical review. Eur. J. Lipid Sci. Technol. 2, 133–153 12 Wahler, D. and Reymond, J-L. (2001) High-throughput screening for biocatalysts. Curr. Opin. Biotechnol. 12, 535–544 13 Speer, A.E. and Cravatt, B.F. (2004) Chemical strategies for activity- based proteomics. ChemBioChem 5, 41–47 14 Klein, G. and Reymond, J-L. (1998) An enantioselective fluorimetric assay for alcohol dehydrogenases using albumin-catalyzed b-elimin- ation of umbelliferone. Bioorg. Med. Chem. Lett. 8, 1113–1116 15 Klein, G. and Reymond, J.-L. (1999) Enantioselective fluorogenic assay of acetate hydrolysis for detecting lipase catalytic antibodies. Helv. Chim. Acta 3, 400–408 16 Jourdain, N. et al. (1998) A stereoselective fluorogenic assay for aldolases: detection of an anti-selective aldolase catalytic antibody. Tetrahedron Lett. 51, 9415 17 Pe´rez Carlo`n, R. et al. (2000) Fluorogenic polypropionate fragments for detecting stereoselective aldolases. Chemistry 6, 4154–4162 18 List, B. et al. (1998) Aldol sensors for the rapid generation of tunable fluorescence by antibody catalysis. Proc. Natl. Acad. Sci. U. S. A. 95, 15351–15355 19 Badalassi, F. et al. (2000) A versatile periodate-coupled fluorogenic assay for hydrolytic enzymes. Angew. Chem. Int. Ed. Engl. 39, 4067–4070 20 Gonza´lez-Garcı´a, E. et al. (2003) Fluorogenic stereochemical probes for transaldolases. Chemistry 9, 893–899 21 Leroy, E. et al. (2003) Fluorogenic cyanohydrin esters as chiral probes for esterase and lipase activity. Adv. Synth. Catal. 6 þ 7, 859–865 22 Nyfeler, E. et al. (2003) A sensitive and selective high-throughput screening fluorescence assay for lipases and esterases. Helv. Chim. Acta 8, 2919–2927 23 Gonza´lez-Garcia´, E.M. et al. (2003) Synthesis and evaluation of chromogenic and fluorogenic analogs of glycerol for enzyme assays. Helv. Chim. Acta 7, 2458–2470 Review TRENDS in Biotechnology Vol.22 No.7 July 2004 369 www.sciencedirect.com
  • 8. 24 Badalassi, F. et al. (2002) A selective HIV-protease assay based on a chromogenic amino acid. Helv. Chim. Acta 10, 3090–3098 25 Sevestre, A. et al. (2003) A fluorogenic assay for transketolase from Saccharomyces cerevisiae. Tetrahedron lett. 4, 827–830 26 Gutierrez, M.C. et al. (2003) The first fluorogenic assay for detecting a Baeyer–Villigerase activity on microbial cells. Org. Biomol. Chem. 1, 3500–3506 27 Gao, W. et al. (2003) Novel fluorogenic substrates for imaging b-lactamase gene expression. J. Am. Chem. Soc. 37, 11146–11147 28 Matayoshi, E.D. et al. (1990) Novel fluorogenic substrates for assay- ing retroviral proteases by resonance energy transfer. Science 247, 954–958 29 Kohl, T. et al. (2002) A protease assay for two-photon crosscorrelation and FRET analysis based solely on fluorescent proteins. Proc. Natl. Acad. Sci. U. S. A. 99, 12161–12166 30 Farber, S.A. et al. (2001) Genetic analysis of digestive physiology using fluorescent phospholipid reporters. Science 292, 1385–1388 31 Vallee, B.L. (1992) Methods of measuring isozymes and isozyme classes of alcohol dehydrogenase. US Patent, 5,162,203 32 Wahler, D. et al. (2002) Enzyme fingerprints of activity, stereo- and enantioselectivity from fluorogenic and chromogenic substrate arrays. Chemistry 8, 3211–3228 33 Mateo, C. et al. (2003) A spectrophotometric assay for measuring and detecting an epoxide hydrolase activity. Anal. Biochem. 314, 135–141 34 Bensel, N. et al. (2001) Pivalase catalytic antibodies: towards abzymatic activation of prodrugs. Chemistry 7, 4604–4612 35 Leroy, E. et al. (2003) A low background high-throughput screening (HTS) fluorescence assay for lipases and esterases using acyloxy- methylethers of umbelliferone. Bioorg. Med. Chem. Lett. 13, 2105–2108 36 Tanaka, F. et al. (2003) Fluorescent detection of carbon–carbon bond formation. J. Am. Chem. Soc. 125, 8523–8528 37 Yeo, W-S. and Mrksich, M. (2003) Self-assembled monolayers that transduce enzymatic activities to electrical signals. Angew. Chem. Int. Ed. Engl. 42, 3121–3124 38 Reetz, M.T. et al. (1999) A method for high-throughput screening of enantioselective catalysts. Angew. Chem. Intl. Ed. 12, 1758–1761 39 Reetz, M.T. et al. (2002) A practical NMR-based high-throughput assay for screening enantioselective catalysts and biocatalysts. Adv. Synth. Catal. 9, 1008–1016 40 Tielmann, P. et al. (2003) A practical high-throughput screening system for enantioselectivity by using FT-IR spectroscopy. Chemistry 9, 3882–3887 41 Cedrone, F. et al. (2003) Directed evolution of the epoxide hydrolase from aspergillus niger. Biocatalysis and Biotransformation 6, 357–364 42 Reetz, M.T. and Ru¨ggeberg, C.J. (2002) A screening system for enantioselective enzymes based on differential cell growth. Chem. Commun. 7, 1428–1429 43 Cohen, P. (2001) The role of protein phosphorylation in human health and disease. Eur. J. Biochem. 268, 5001–5010 44 Kupcho, K. et al. (2003) A homogeneous, nonradioactive high- throughput fluorogenic protein kinase assay. Anal. Biochem. 317, 210–217 45 Hamman, B.D. et al. (2002) Optical probes and assays. US Patent 60 4100 255, 2002-06-25 46 Garcya-Echeverrya, C. et al. (1992) Continuous fluorimetric direct (uncoupled) assay for peptidyl prolyl cis-trans isomerases. J. Am. Chem. Soc. 7, 2758–2759 47 Kofron, J.L. et al. (1991) Determination of kinetic constants for peptidyl prolyl cis–trans isomerases by an improved spectrophoto- metric assay. Biochemistry 30, 6127–6134 48 Yli-Kauhaluoma, J.T. et al. (1996) Catalytic antibodies with peptidyl- prolyl cis-trans isomerase activity. J. Am. Chem. Soc. 23, 5496–5497 49 Velhuyzen, W.F. et al. (2003) A light-activated probe of intracellular protein kinase activity. J. Am. Chem. Soc. 125, 13358–13359 50 Shults, M.D. and Imperiali, B. (2003) Versatile fluorescence probes of protein kinase activity. J. Am. Chem. Soc. 125, 14248–14249 51 Simeonov, A. et al. (2002) Enzyme assays by fluorescence polarization in the presence of polyarginine: study of kinase, phosphatases and protease reactions. Anal. Biochem. 304, 193–199 52 Gaudet, E. et al. (2003) A homogeneous fluorescence polarization assay adaptable for a range of protein serine/threonine and tyrosine kinases. J. Biomol. Screen. 8, 164–175 53 Janes, L.E. and Kazlauskas, R.J. (1997) Quick E. A fast spectro- photometric method to measure the enantioselectivity of hydrolases. J. Org. Chem. 14, 4560–4561 54 Janes, L.E. et al. (1998) Quantitative screening of hydrolase libraries using pH indicators: identifying active and enantioselective hydro- lases. Chem. Eur. J. 11, 2324–2331 55 Park, C.B. and Clark, D.S. (2002) Sol-gel-encapsulated enzyme arrays for high-throughput screening of biocatalytic activity. Biotechnol. Bioeng. 78, 229–235 56 Henke, E. and Bornscheuer, U.T. (2003) Fluorophoric assay for the high-throughput determination of amidase activity. Anal. Chem. 75, 255–260 57 Roeschlaub, C.A. et al. (1999) A fluorescent probe for the detection of NAD(P)H. Chem. Commun., 1637–1638 58 Martin, K. et al. (2003) Quantitative analysis of protein phosphory- lationstatus and protein kinase activity on microarrays using a novel fluorescent phosphorylation sensor dye. Proteomics 3, 1244–1255 59 Ojida, A. et al. (2002) First artificial receptors and chemosensors toward phosphorylated peptide in aqueous solution. J. Am. Chem. Soc. 124, 6256–6258 60 Sorde`, N. et al. (2003) Enzyme screening with synthetic multi- functional pores: focus on biopolymers. Proc. Natl. Acad. Sci. U. S. A. 100, 11964–11969 61 Das, G. et al. (2002) Fluorometric detection of enzyme activity with synthetic supramolecular pores. Science 298, 1600–1602 62 Wahler, D. and Reymond, J-L. (2002) The adrenaline test for enzymes. Angew. Chem. Int. Ed. Engl. 41, 1229–1232 63 Wahler, D. et al. (2004) Adrenaline profiling of lipases and esterases with 1,2-diol and carbohydrate acetates. Tetrahedron 3, 703–710 64 Doderer, K. et al. (2003) Spectrophotometric assay for epoxide hydrolase activity toward any epoxide. Anal. Biochem. 321, 131–134 65 Konarzycka-Bessler, M. and Bornscheuer, U.T. (2003) A high- throughput screening method for determining the synthetic activity of hydrolases. Angew. Chem. Int. Ed. Engl. 42, 1418–1420 66 Maesawa, C. et al. (2003) A rapid biosensor chip assay for measuring of telomerase activity using surface plasmon resonance. Nucleic Acids Res. 31, E4 67 Tolun, G. and Myers, R.S. (2003) A real-time DNase assay (ReDA) based on PicoGreen fluorescence. Nucleic Acids Res. 31, E111 68 Klein, G. et al. (2001) A fluorescent metal sensor based on macrocyclic chelation. Chem. Commun., 561 69 Klein, G. and Reymond, J-L. (2001) An enzyme assay using pM. Angew. Chem. Int. Ed. Engl. 40, 1771–1773 70 Dean, K.E.S. et al. (2003) A green fluorescent chemosensor for amino acids provides a versatile high-throughput screening (HTS) assay for proteases. Bioorg. Med. Chem. Lett. 13, 1653–1656 71 Reymond, J-L. and Wahler, D. (2002) Substrate arrays as enzyme fingerprinting tools. ChemBioChem 3, 701–708 72 Bussie`re, J. et al. (1967) Usage de substrats synthetiques pour l’e´tude de l’e´quipement enzymatique de microorganismes. C. R. Acad. Sc. Paris 264D, 415–417 73 Meldal, M. et al. (1994) Portion-mixing peptide libraries of quenched fluorogenic substrates for complete subsite mapping of endoprotease specificity. Proc. Natl. Acad. Sci. USA 8, 3314–3318 74 Maly, D.J. et al. (2002) Combinatorial strategies for targeting protein families: application to the proteases. Chem. BioChem. 1, 16–37 75 Wahler, D. et al. (2001) Enzyme fingerprints by fluorogenic and chromogenic substrate arrays. Angew. Chem. Int. Ed. Engl. 40, 4457–4460 76 Grognux, J. and Reymond, J-L. Classifying enzymes from selectivity fingerprints. ChemBioChem (in press) 77 Salisbury, C.M. et al. (2002) Peptide microarrays for the determination of protease substrate specificity. J. Am. Chem. Soc. 124, 14868–14870 78 Zhu, Q. et al. (2003) Enzymatic profiling system in a small-molecule microarray. Org. Lett. 5, 1257–1260 Review TRENDS in Biotechnology Vol.22 No.7 July 2004370 www.sciencedirect.com