You are on page 1of 11

Kinetic Assay Methods UNIT 3.

The design and use of a particular enzyme dictate the assay procedure used. Enzyme-as-
assay is dependent on the objectives of the say methods include those based on spectro-
particular investigation. If the focus is on en- photometry, radiometry, pH-stat, turbidometry,
zyme purification or issues of clinical biochem- polarography, and fluorometry. Spectro-
istry, experimental protocols typically require photometric and fluorometric assay protocols
optimizing conditions for all substrates and rely on the differences in spectral or fluorescent
effectors—e.g., with respect to pH value, tem- properties, respectively, of the product(s) rela-
perature, buffering agent, and ionic strength. tive to the substrate(s). With radiometric pro-
However, optimal conditions may not be rea- cedures, effective means for separating labeled
sonable for all parameters and factors. For ex- substrate(s) from labeled product(s) must be
ample, substrate instability may preclude stud- available so that the amount of product formed
ies at the enzyme’s optimal pH or at the physi- at a given time can be accurately measured.
ological temperature. Thus, Silverstein and Those reactions that utilize or generate protons
Sulebele (1969) examined the exchange rates (or hydrons) can be followed by pH-stat meth-
of malate dehydrogenase at 1°C to avoid prob- ods. If the reaction studied results in a change
lems arising from the decomposition of ox- in solution turbidity (e.g., caused by aggrega-
aloacetate. Likewise, in clinical studies and tion of a macromolecular complex), then turbi-
enzyme purification, saturating concentrations dometric protocols can effectively assay for the
of substrate(s) are typically required, although protein that causes the change in turbidity. All
this may not be possible or even desirable in of these methods, as well as others, can be found
many instances, as limitations in substrate solu- in Bergmeyer (1983).
bility or substrate inhibition of the enzyme may
discourage the use of elevated concentrations. GENERAL ASPECTS OF ASSAY
The focus in clinical studies and enzyme puri- DESIGN
fication is on providing good estimates of the An enzyme’s progress curve (i.e., a plot of
activities of a specific enzyme in a number of product formation or substrate depletion as a
physiological samples with a rapid, straightfor- function of time; see Fig. 3.5.1) consists of four
ward, and reproducible assay method. How- phases that are of interest to the investigator.
ever, for the kineticist the objective is to char- The duration of the pre-steady-state region is
acterize the initial rates of the enzyme-cata- relatively short and is typically followed via
lyzed reaction under a wide set of conditions. rapid-flow or relaxation techniques. Discus-
It is through these detailed studies, often carried sion of this region of the progress curve is
out at subsaturating substrate concentrations, beyond the scope of this unit, but a review of
that the investigator is able to determine the true these techniques has recently appeared (Fierke
optimal parameters to be used in protein puri- and Hammes, 1995). The next portion of the
fication and clinical studies, as well as to probe curve is the steady-state phase, a longer phase
the dynamics and mechanism of the reaction characterized by relatively constant concentra-
and its modulation by other factors. The pur- tions of each of the individual enzyme forms
pose of this unit is to provide a brief review of (e.g., the enzyme-substrate binary complexes).
issues important in the design of initial-rate During the third phase of the progress curve—
assay methods. the post-steady-state region—the levels of
In most instances, the investigator already these enzyme complexes change rapidly, un-
has some information about the kinetic proper- til the last phase—equilibrium—has been
ties of the enzyme, obtained from preliminary reached. Each of these phases contains infor-
experiments as well as from the literature. It is mation of use to the investigator. The steady-
necessary to have an approximation of the mag- state portion of the curve typically exists for a
nitude of the Michaelis constants of all the few to several hundred seconds. It is this region
substrates in order that detailed initial rate stud- of the curve that is most accessible to the ma-
ies can be effectively designed. Certainly, in- jority of investigators. In addition, the relatively
formation on the identity of the substrates and straightforward mathematical treatment of
products is required, along with an effective steady-state kinetics provides an excellent
procedure for monitoring substrate depletion or foundation for the subsequent characterization
product accumulation. The nature of the reac- of the enzyme system. Because enzyme rate
tion and the properties of the substrates help expressions can be quite complex, a number of Detection and
Assay Methods

Contributed by R. Donald Allison 3.5.1


Current Protocols in Protein Science (1996) 3.5.1-3.5.11
Copyright © 2000 by John Wiley & Sons, Inc. Supplement 5
15

Product concentration (nmo l/ m l)


d
c
10

b
5

a
0
0 5 10 15 20
Time (sec)

Figure 3.5.1 Typical progress curve for an enzyme-catalyzed reaction, where a represents the
pre-steady-state region, b the steady-state region, c the post-steady-state region, and d the
equilibrium region.

assumptions are made to linearize the equa- cally, there is at least a hundred-fold difference
tions. The investigator should ensure that each in volume between the reaction mixture and the
of these factors are addressed when any assay initiating aliquot. If the enzyme and substrate
protocol is being designed. are stable under the assay conditions, then both
solutions should be preincubated at this tem-
Maintaining Constant Reaction perature.
Conditions It should be emphasized that valid initial rate
The status of each experimental parame- experiments are only obtained when good tem-
ter—e.g., temperature, ionic strength, and perature control is maintained. The tempera-
pH—should remain constant over the time ture-regulated water bath should have a vari-
course of the assay. ability of ≤0.1°C. When the reaction is initiated,
the solution should be thoroughly mixed. If
Temperature reaction progress is followed using an instru-
Prior to initiation of the reaction by addition ment such as a spectrophotometer, the sample
of a small aliquot of enzyme or substrate, the compartment should be thermally isolated.
reaction mixture should be thermally equili- Another issue of concern is choice of the
brated with an accurately controlled constant- temperature at which enzyme activity will be
temperature water bath for several minutes. If measured. In 1964, the International Union of
the substrate is thermally stable, this incubation Biochemistry (IUB) recommended a tempera-
should be for ≥10 min or until the desired ture of 30°C. Clinical chemists have often sug-
reaction temperature is reached. If the substrate gested a change to 37°C. However, the final
is unstable at this temperature, it may be nec- choice of temperature will be dependent on the
essary either to assay at a different temperature, system being investigated. For example, the
to correct for substrate decay with time, or to activity of glutamate dehydrogenase isolated
initiate the reaction by addition of a small from the hyperthermophile Pyrococcus furio-
aliquot of substrate solution that was freshly sus is negligible at both 30°C and 37°C; the
prepared and maintained in an ice bath. When enzyme exhibits maximal activity at 97°C
the reaction is initiated by addition of either the (Klump et al., 1992). Clearly, at some point in
enzyme or the substrate, the volume of the the investigation, the effect of temperature on
aliquot should be small enough to minimize any the catalytic activity will have to be charac-
Kinetic Assay
Methods alterations in temperature upon addition. Typi- terized.

3.5.2
Supplement 5 Current Protocols in Protein Science
Buffer selection Substrate Stability and Purity
In setting up the assay protocol, care should It is crucial to establish the degree of stability
also be exercised in selecting an appropriate of the substrate as well as that of any enzyme
buffer agent. Unfortunately, many investigators effector under the assay conditions (also see
choose a buffer solely on the basis of the desired discussion of Maintaining Constant Reaction
pH of the reaction medium. Many excellent Conditions). Substrate and effector stabilities
buffers, however, have a significant oxidation- can be tested by preparing a solution of the
reduction potential that can severely alter an compound at a concentration that will be used
enzyme’s activity. For example, cacodylate experimentally with the other components of
(pKa = 6.27) and other organoarsenicals have a the assay mixture, but without the enzyme.
potent oxidizing potential under acidic condi- Then, samples are removed at different periods
tions. Borate (pKa = 9.23) and other borate- of time, usually over a period of several hours,
based buffers form complexes with many diols, and assayed either chemically or enzymatically
polyols, carbohydrates, and ribonucleotides; (Bergmeyer, 1983). A plot of concentration as
for example, in the presence of borate ion, both a function of time will reveal if the substrate is
L- and D-serine form a transition complex and stable or if there is a decay. When present, this
strongly inhibit γ-glutamyl transpeptidase decomposition is usually a first-order process
(Tate and Meister, 1978). Hence, care should and can be described by the equation [S]t = [S]0
be exercised in selecting a buffer. e−kt, where [S]t and [S]0 are the substrate con-
In determining which buffer is optimal for centrations at time t and time 0, respectively,
a given enzyme system, it is best to examine and k is the first-order rate constant. If the
the enzyme’s activity with as many buffers as decomposition is greater than first-order or
possible. Buffers in whose presence the enzyme composed of more than one first-order process,
exhibits high activity should then be examined then the decay must be evaluated by the appro-
to determine if the buffering agent interacts priate expression, which can be found in any
with the protein. If pH and ionic strength are basic kinetics text.
held constant, the enzyme activity should be An issue related to substrate stability is sub-
independent of the buffer concentration. An- strate purity. Impurities in the substrate, effec-
other factor of importance in selecting the ap- tors, or enzyme preparation can be potential
propriate buffer is the capacity of the buffering sources of experimental error. Because alterna-
agent to bind metal ions. Ideally, a good buffer tive substrates and competitive inhibitors are
will not form any significant complexes with often structurally similar to the substrate and
metal-ion cofactors. frequently have similar chemical and physical
properties, it is not surprising to find these
Metal-ion concentrations compounds as impurities in commercial sub-
The role of metal ions is crucial with many strate preparations. Spectral and chroma-
enzymes, particularly with systems that require tographic analyses, as well as enzymatic analy-
nucleotides—e.g., phosphotransferases. Care sis, are crucial in establishing the purity of a
must be exercised in designing initial-rate as- particular biochemical substance. If impurities
says to ensure that the concentration of free are present in a reaction mixture, a kinetic
metal ions does not vary significantly with analysis will give wrong estimates of the kinetic
substrate concentration. Thus, when varying parameters (Allison and Purich, 1979). Heavy-
the concentrations of substrates such as ATP, metal ions are often found as impurities in
the concentration of free, uncomplexed Mg2+ commercial substrates. To remove these, it may
should remain relatively constant. One of two be necessary to recrystallize the substrates, pos-
methods is commonly utilized to achieve this sibly in the presence of a chelating agent such
end: either (1) maintaining the total nucleotide as EDTA.
concentration and the metal concentration at a These issues of purity and stability are also
constant stoichiometric ratio (e.g., 10:1) or (2) pertinent for any modulators of enzyme activ-
maintaining a constant excess of metal ion over ity, including the product(s). The products have
the total nucleotide concentration (e.g., 1.0 mM to be stable over the time course of the assay.
free Mg2+). The second method, which has been In some instances, the precise nature of the true
demonstrated to be the preferable of the two product of the enzyme-catalyzed reaction may
(O’Sullivan and Smithers, 1979), requires not be known. In these cases, the investigator
knowledge of the stability constant for the met- may follow the reaction via substrate depletion.
al-ion–nucleotide complex under the condi- It is frequently useful to do a kinetic analysis Detection and
tions of the assay. of freshly prepared substrate in the absence and Assay Methods

3.5.3
Current Protocols in Protein Science Supplement 5
presence of enzymatically prepared product. A for those enzymes that require reduced thiol
decent understanding of the possible presence groups for activity. Care must be exercised that
of product inhibition in initial-rate experiments the reducing agent does not produce other ef-
is always useful to the protein chemist. fects. For example, γ-glutamylcysteine synthe-
tase is inhibited by the presence of thiol re-
Enzyme Stability and Purity agents even though cysteine is a substrate
Enzymes often undergo a slow inactivation. (Seelig and Meister, 1985).
Usually, the rate of this inactivation is acceler- Enzyme purity is also an issue of concern.
ated by dilution of the stock enzyme prepara- It is important in early initial-rate studies to
tion. Efforts have to be made to eliminate or eliminate contaminating activities and to dem-
lessen the loss of enzyme activity with time. onstrate that any minor activity does not sub-
Often, multisubstrate enzymes are stabilized by stantially affect the reaction being studied. For
the presence of a substrate. For example, tubu- multisubstrate enzymes, this can be readily
lin:tyrosine ligase is stabilized by the presence accomplished by observing the stability of each
of its protein substrate (Deans et al., 1992) and substrate, effector, and product separately in the
the presence of Mg2+–ATP is known to stabilize presence of the enzyme preparation. Adenylate
a number of phosphotransferases. If the en- kinase is a common contaminant in prepara-
zyme inactivation can be retarded by the pres- tions of a wide variety of phosphotransferases
ence of a substrate, the investigator should note and other nucleotide-dependent enzymes. This
the amount of that substrate that is present with contaminating enzyme can be inhibited by per-
the enzyme so that proper corrections can be forming the assay at high ionic strength (Bowen
made when the reaction is initiated with the full and Kerwin, 1955). Addition of elevated lev-
assay mixture. els of AMP will also inhibit adenylate kinase.
Other agents—e.g., glycerol, certain salts, However, many phosphotransferases are also
high-ionic-strength solutions, and EDTA— inhibited by this mononucleotide or are them-
have also been used to stabilize proteins. Often, selves inhibited by high ionic strength. Pu-
inactivation is a result of surface denaturation rich and Fromm (1972) and Lienhard and
or adsorption of the enzyme on glass surfaces Secemski (1973) have shown that the mul-
following dilution. The presence of stabilizing tisubstrate analogs P1,P4-di(adenosine-5′)-
agents and/or use of plastic containers can less- tetraphosphate and P1,P5-di(adenosine-5′)-
en these effects significantly. The presence of pentaphosphate are potent and specific in-
serum albumin or another protein will fre- hibitors of this activity.
quently minimize the effects of enzyme dilu- In some instances it may be impossible to
tion; the investigator should test several pro- completely eliminate side reactions. Enzymes
teins when this technique is used. The stabiliz- that have branched reaction pathways (i.e.,
ing protein may also bind the substrates or the multifunctional enzymes) will exhibit two
enzyme’s cofactors; if this is the case, correc- pathways. Most multifunctional enzymes are
tions have to be made. transferases having multiple acceptor specific-
If a stabilizing agent cannot be identified, ity. Glucose-6-phosphatase catalyzes not only
the inactivation should be minimized by using the hydrolysis of glucose-6-phosphate but also
the smallest dilution possible. If practical, the its synthesis; thus, it may effect the transfer of
reaction may be initiated by the addition of one phosphoryl groups from pyrophosphate, car-
of the substrates. Corrections for loss of en- bamoyl phosphate, or other glucose-6-phos-
zyme activity can also be made by establishing phate molecules (Nordlie, 1982). This syn-
an activity-decay curve. However, it should be thetic role of glucose-6-phosphatase is not a
verified that the loss of activity is due to total minor activity; at a concentration of 100 mM
inactivation and not simply to a conformational D-glucose, the synthesis and hydrolysis activi-
change to a form having less activity (i.e., a ties are approximately equal. The enzyme γ-
form with different kinetic parameters). This glutamyl transpeptidase catalyzes the hydroly-
possibility could be tested by kinetic assays at sis of a number of γ-glutamyl-containing mole-
significantly different times following dilution cules (including glutathione and glutathione
or by two reference assays, one using a saturat- disulfide) and also exhibits a transferase activ-
ing substrate level and the other using a sub- ity in which the glutamyl moeity is transferred
saturating substrate level. to any of a number of amino acid acceptors,
The presence of thiol reagents—e.g., 2-mer- particularly cysteine, glutamine, and methion-
Kinetic Assay captoethanol, dithiothreitol, or dithioerythri- ine (Allison, 1985). The hydrolase and trans-
Methods tol—is routinely helpful in maintaining activity ferase activities of γ-glutamyl transpeptidase

3.5.4
Supplement 5 Current Protocols in Protein Science
are approximately equal in the presence of a product accumulation can lead to significant
pool of acceptor substrates at their physiologi- errors in initial-rate determinations even in
cal concentrations (Allison and Meister, 1981). those cases where the degree of conversion is
Asparagine synthetase catalyzes the glu- low. A preliminary test to assess this possibility
tamine-dependent synthesis of L-asparagine is to look at initial rates in the absence and
and also exhibits a significant glutaminase ac- presence of a known amount of product. Where
tivity (Boehlein et al., 1994). In all of these product inhibition is a problem, product can be
cases, it is possible to use inactivation studies depleted if an auxiliary enzyme system for
to observe the parallel loss of both (or all) doing this is present. Nevertheless, product
activities with time. inhibition can be an important regulatory
scheme for an enzyme. Any kinetic charac-
Enzyme Concentration terization of an enzyme should include accurate
The duration of the steady-state phase of an measurements of the dissociation constants for
enzyme-catalyzed reaction is dependent on a all products. Product-inhibition studies can
number of factors. Perhaps the most important also assist the investigator in establishing
of these is the relative concentration of the mechanistic details of the protein undergoing
enzyme and substrate. The ratio of the initial evaluation (Rudolph, 1979).
substrate concentration to the total enzyme con- In addition to establishing that the progress
centration should be ≥100:1. With lower values, curve is truly linear, demonstrating that the
the steady-state phase will be of very short degree of substrate depletion in the initial time
duration. It should be pointed out that this course of the assay is low, and assessing the
relationship holds for enzyme effectors as well. degree of product inhibition that may be pre-
Thus, when investigating the influences of an sent, the investigator should also demonstrate
activator or competitive inhibitor on enzyme in the early stages of the kinetic studies that the
activity, the total enzyme concentration should rates observed are linear with respect to the total
be much less than the concentration of the enzyme concentration ([E]total). If true steady-
effector. This may prove difficult if the effector state conditions are present, a plot of initial rates
has a very tight affinity with the enzyme. In versus [E]total (in which the enzyme concentra-
these cases, the protocols outlined by Williams tion is varied above and below the final value
and Morrison (1979) will prove useful. to be used in the initial-rate assays) should be
linear.
Initial Rate and Steady-State
Condition Substrate Concentration Ranges
In an experiment, the investigator typically A wide range of substrate concentrations
measures the rate of an enzyme-catalyzed re- should be tested in preliminary studies. Such
action by calculating the slope of the progress trials will provide an estimate of the Michaelis
curve. This procedure is only valid if the plot constant (Km) and an assessment of whether
of product concentration as a function of time substrate inhibition occurs at elevated substrate
has been demonstrated to be linear within the levels. These tests will also detect the possible
time frame of the assay. However, this is not the existence of cooperativity. Once an estimate of
only criterion that determines initial rate and the Michaelis constant has been made, a more
steady-state kinetics. Linearity has been ob- detailed kinetic study can be made, in which
served in many non-steady-state systems (Al- the substrate concentration is typically varied
lison and Purich, 1979). from ∼0.2 to 5.0 times the value of the apparent
The general rule of thumb is that initial rate constant. It should be emphasized that reason-
conditions prevail when the substrate concen- able estimates of the Km value can only be made
tration is still within 10% of its starting value. when initial rates have been obtained from
This value is clearly acceptable for reactions substrate concentrations both below and above
that are thermodynamically quite favorable and the true Km value. For a one-substrate enzyme-
that do not exhibit any significant product in- catalyzed reaction:
hibition. However, if the equilibrium constant
Km = [A](Vmax − v)/v
for the reaction is not large, an estimate of the
degree of substrate-to-product conversion where [A] is the substrate concentration, Vmax
should be made. With smaller equilibrium con- is the maximal velocity of the reaction, and v
stants, the initial-rate assay protocol necessar- is the velocity of the reaction at the substrate
ily must be more sensitive. In addition, if the concentration. A series of substrate concentra- Detection and
product has very tight affinity for the enzyme, tions with their respective initial rate velocities Assay Methods

3.5.5
Current Protocols in Protein Science Supplement 5
will provide an estimate of the Michaelis con- strate enzyme-catalyzed reaction (where the the
stant. In a standard double-reciprocal plot, the two substrates are A and B). In this example,
horizontal intercept is numerically equal to Michaelis constants for the two substrates can
−1/Km. For multisubstrate enzymes (see be- be obtained by constructing slope and/or inter-
low), estimates of the Michaelis constants will cept replots of the data. For example, plotting
be obtained from slope and intercept replots of the slopes of the five lines as a function of the
the initial rate data. In addition, appropriate reciprocal of the concentration of B will yield
statistical treatment of the raw data should be a straight line with a horizontal intercept nu-
provided (Wilkinson, 1961; Cleland, 1979a) merically equal to −1/KB, where KB is the
for both Km and Vmax values (or kcat values, Michaelis constant for B.
equal to Vmax/[E]total, if those are reported). For enzymes that utilize three or more sub-
Note that, regardless of whether Vmax or kcat strates, the protocol described above can be-
values are reported, the total protein concentra- come unwieldy. Two procedures have been
tion, as well as the specific activity of the described to address the design of kinetic stud-
enzyme used, should be reported whenever ies for these proteins. In the first method
possible. (Frieden, 1959), one of the three substrates is
With enzyme systems utilizing two sub- held constant at a concentration above its re-
strates, the concentrations of both substrates spective Michaelis constant that is still subsatu-
must be varied as described above. Typically, rating. The other two are then varied as de-
three solutions are prepared. One solution con- scribed above. This same procedure is then
tains one of the substrates at its highest level. followed with each of the remaining substrates.
A second solution contains the second substrate Thus, multisubstrate enzymes are reduced to
at its highest level. The third solution contains pseudo-two-substrate systems. One distinct ad-
the buffer, all cofactors, metal ions, auxiliary vantage of this procedure is that standard
enzymes, and any other effectors, but no sub- graphical presentations of the data thus pro-
strates. This third solution is used to prepare duced (e.g., via double-reciprocal plots) are
serial dilutions of the other two stock solutions. identical to that shown in Figure 3.5.2. How-
Thus, a matrix of initial rate measurements on ever, the concentration value for the third sub-
both substrates can be evaluated from a single strate have to be provided. In the second pro-
experimental trial. Figure 3.5.2 illustrates a cedure, the concentration of one substrate is
typical double-reciprocal plot for a two-sub- varied while the other substrates are held at a

30 5.56

7.14

20
1/v (min /mM)

10.00
[B](mM)

16.70

50.00
10

0
0 10 20
1/[A]
(1/ mM)

Figure 3.5.2 Double-reciprocal plot of a two-substrate enzyme-catalyzed reaction, where v = the


Kinetic Assay velocity of the reaction and [A] and [B] represent the concentrations of substrates A and B
Methods
respectively.
3.5.6
Supplement 5 Current Protocols in Protein Science
constant ratio, although the absolute amounts a continuous assay, the investigator must dem-
are varied. This is then repeated for all remain- onstrate that the single-point assay is a true
ing substrates until the concentration of each of measure of the steady-state velocity of the re-
the substrates has been varied (Fromm, 1967). action. In the single-point method, an aliquot
One limitation with this approach is the diffi- of the reaction mixture is removed from the
culty in obtaining true Michaelis constants. sample at a given time point after the reaction
The presence of substrate inhibition can be has been initiated (after a “blank” aliquot has
an annoyance to an investigator who is attempt- been removed from the reaction mixture prior
ing to determine the kinetic parameters of an to the initiation step). Aliquots from at least four
enzyme as well as identify the binding pattern. different time points are removed from the
However, the pattern of such inhibition can reaction solution; then the reaction is termi-
serve as a tool in understanding the kinetic and nated and the amount of product formed (or
possibly the regulatory scheme for the enzyme. substrate depleted) is determined. It should be
Substrate inhibition can occur as a result of a ascertained that the termination procedure truly
number of different causes—i.e., the formation quenches the reaction. Once the steady-state
of a dead-end complex with a particular enzyme relationship has been established, replicate ki-
form, the appearance of a different binding netic analyses at each substrate concentration
mechanism at elevated levels of substrate, the will provide sufficient data for a statistical
presence of an allosteric site for the substrate, analysis. The number of replicate samples that
or nonspecific inhibition that is due to an ele- are assayed should also be reported.
vated ionic strength or another factor. When
substrate inhibition occurs, in the case of mul- COUPLED ENZYME ASSAYS
tisubstrate enzymes, estimates of the Ki value In many instances, continuous assays are
(i.e., the dissociation constant for inhibition) made possible by the presence of auxiliary
for the substrate can be obtained from slope and enzymes. Product formation is measured by
intercept replots of the kinetic data. For en- having the product react further with another
zymes having a single substrate, the data can enzyme (or enzymes). Although such assay
be fit to an enzyme-rate expression containing systems are convenient, precautions must be
a term for substrate inhibition (v = Vmax[S]/{Km exercised to ensure that the initial rates ob-
+ [S] + [S]2/Ki}). In addition, estimates of Km, served reflect the enzyme that is undergoing
Ki, and Vmax can be obtained from double-re- investigation and not the auxiliary system. If
ciprocal plots (Cleland, 1979b). Note that in the auxiliary system is influencing the initial-
such cases, double-reciprocal plots will be non- rate results, the reported values for the kinetic
linear, with the degree of curvature dependent parameters will be in error and the double-re-
on the magnitude of the Ki value. In these cases, ciprocals may exhibit considerable nonlinear-
it may be necessary to use a computer program ity.
to fit the data. Cleland (1979a) has provided a The product or products formed by the en-
program to address substrate inhibition. In ad- zyme undergoing study clearly direct the
dition, one might consider utilizing a Marmasse choice of auxiliary enzyme(s). This coupling
plot (Marmasse, 1963). Although rarely used, system should provide turnover in excess over
this plot provides good estimates of Km and Ki. that due to the enzyme being investigated.
A Marmasse plot is a plot of 1/v versus (α + However, the degree of excess will vary de-
1/α ), where α = [A]/[A]m and [A]m represents pending upon the assay method, the nature of
the concentration of substrate at which the the coupling system, and the magnitude of the
velocity is highest (note that this is not Vmax, as kinetic parameters for each auxiliary enzyme
substrate inhibition is present.) This plot, when as measured under the same conditions being
combined with the observation that [A]m = observed with the target enzyme. Once this
(Km/Ki) ⁄2, will provide values for the kinetic
1
information is in hand, the amount of auxiliary
parameters (Cleland, 1979b). enzyme needed can be calculated (McClure,
1969; Storer and Cornish-Bowden, 1974). Test
CONTINUOUS VERSUS runs should then be undertaken using both the
STOP-TIME ASSAYS calculated and higher levels of auxiliary en-
As the best estimate for the initial rate of an zymes to insure that identical rates are obtained,
enzyme-catalyzed reaction derives from the indicating that a sufficient excess has been
slope of the steady-state region of the progress achieved. The auxiliary enzyme system should
curve, continuous assays are greatly preferable also have minimal effect on cofactors and ef- Detection and
to single-point assays. If it is not possible to use fectors required by the target enzyme. Assay Methods

3.5.7
Current Protocols in Protein Science Supplement 5
Frequently, a small amount of product may PRESENTATION OF
be present in the reaction mixture as a contami- INITIAL-RATE DATA
nant. For example, commercial preparations of The description of the assay protocols used
ATP often include a small amount of ADP. The in connection with enzyme purifications, in
contaminating product can be removed prior to clinical studies, and in all kinetic investigations
initiation of the assay by incubating the reaction should include a clear definition of the unit of
mixture with the auxiliary system. The system enzyme activity. When possible, velocities
should also be checked to insure that no activity should be expressed in terms of molarity
is observed in the absence of the auxiliary changes with time, as molarity is an intensive
enzymes. Finally, care should be exercised variable. Other units, proportional to molarity,
when using commercial preparations for the are in common use. In all cases, the conditions
auxiliary enzymes. Often these sources of pro- of the assay should be fully described, includ-
teins contain contaminating activities that may ing the volume of the initiating aliquot, the
cause serious difficulties unless they are ad- specific activity, and the amount of enzyme
dressed. used in each experiment. The international unit
(U), defined as the conversion of 1 µmol of
BINDING STUDIES substrate to product per minute, is in common
Measurements of equilibrium ligand bind- use. Another unit of catalytic activity, the katal,
ing can frequently add to the characterization corresponding to the transformation of one
of an enzyme’s binding mechanism, and also mole of substrate per second, is also in use. It
act as a tool in such procedures as molecular- has been suggested that the latter unit be re-
receptor binding studies and radioimmunoas- stricted to catalytic activities measured in a
says. A wide variety of methods have been clinical context (Dybkaer, 1979).
utilized in characterizing binding phenom- Initial-rate data is frequently best presented
ena—e.g., gel filtration and equilibrium dialy- via graphical displays. These visual lineariza-
sis. The theory underlying this valuable tool can tions of kinetic information provide a straight-
be found in any physical biochemistry text. forward evaluation of the binding sequence and
Commonly, the binding data are analyzed with mechanism, as well as a ready appraisal of the
Scatchard plots (Scatchard, 1949) in which the kinetic parameters, reversible inhibition,
number of moles of ligand bound per mole of cooperativity (both allosteric and hysteretic),
total protein (ν) are plotted versus the ratio of and many other steady-state parameters. When-
ν to the free molar concentration of the ligand. ever possible, slope and/or intercept replots
These plots are frequently used to obtain values should also be provided in the graphical dis-
for the ligand dissociation constant, to identify play, perhaps as insets of the initial rate data.
interacting sites, and to estimate the binding There are three methods frequently employed
capacity of the protein. Care should be exer- for depicting kinetic data, with the double-re-
cised in this last use of the binding data, as it is ciprocal plot (i.e., 1/v versus 1/[A]; see Fig.
easy to obtain a poor estimate of the 3.5.2) being the most common. The other com-
stoichiometry of binding from Scatchard plots mon plots are [A]/v versus [A] and v versus
(Klotz, 1982, 1983). For example, a Scatchard v/[A]. Wilkinson (1961) has discussed each of
plot of the binding of diazepam to ben- these plots in terms of statistics. When the data
zodiazepine receptors is reported to provide an are unweighted, the three graphical presenta-
incorrect total number of receptor sites. Often, tions are not statistically equivalent. However,
such problems are due to having only a few data this problem is considerably mitigated by
points in the Scatchard plot at which the ligand proper analysis using computer methods and
concentration is above the half-maximum bind- data weighting (Siano et al., 1975). As men-
ing point. Just as in kinetic data, concentrations tioned earlier, Cleland (1979a) has provided a
of the binding ligand both above and below this number of computer programs for the analysis
half-maximum point should be investigated of data. Eisenthal and Cornish-Bowden (1974)
(Klotz, 1982). Feldman (1983) has recently have also introduced a novel method for graphi-
presented a statistical analysis associated with cal treatment of data that provides a useful tool
ligand binding data and Scatchard plots. As this for evaluating experimental error. An Eisen-
paper points out, estimates of molar binding thal–Cornish-Bowden plot is based on the
capacity should always be reported with the equation (Vmax/v) − (Km/[A]) = 1. Instead of a
appropriate degree of statistical certainty. With- particular set of data (i.e., a single initial rate
Kinetic Assay out such statistical analyses, reports of the num- velocity with its respective substrate concen-
Methods ber of binding sites can be meaningless. tration) being represented by a point in a Car-

3.5.8
Supplement 5 Current Protocols in Protein Science
tesian coordinate system, the velocity (v) is initial-rate assay. In addition, the data assem-
denoted along the vertical axis and the substrate bled from these rigorous studies permit the
concentration ([A]) is denoted along the hori- development of more reliable and sensitive as-
zontal axis. The two points thus plotted are then say protocols for protein purification and iden-
connected by a line. Thus, whereas in the other tification.
graphical procedures the velocity and sub-
strate-concentration data are represented by LITERATURE CITED
points, in the Eisenthal–Cornish-Bowden plot Allison, R.D. 1985. γ-Glutamyl transpeptidase: Ki-
the same data are represented by lines. These netics and mechanism. Methods Enzymol.
113:419-437.
lines, obtained from a set of initial-rate experi-
ments, will intersect about a point equivalent to Allison, R.D. and Meister, A. 1981. Evidence that
transpeptidation is a significant function of γ-
v = Vmax and [A] = Km. Because median values
glutamyl transpeptidase. J. Bio l. Ch em.
of intersection points are used, outlier (i.e., 256:2988-2992.
aberrant) data will have minimal effects on
Allison, R.D. and Purich, D.L. 1979. Practical con-
estimates of the kinetic parameters. In addition, siderations in the design of initial velocity en-
this plot is very useful in that calculations are zyme rate assays. Methods Enzymol. 63:3-22.
not required and data do not have to be Beechem, J.M. 1992. Global analysis of biochemi-
weighted. However, it does have limitations cal and biophysical data. Methods Enzymol.
(Cornish-Bowden and Endrenyi, 1981, 1986). 210:37-54.
For example, the plot can become quite un- Bergmeyer, H.U. (ed.) 1983. Methods of Enzymatic
wieldly with multisubstrate enzymes and in Analysis, 3rd ed., Vols. 1-4. Verlag Chemie,
inhibition studies. Deerfield Beach, Florida.
It has recently been suggested (Brand and Boehlein, S.K., Richards, N.G.J., and Schuster,
Johnson, 1992; Johnson and Brand, 1994) that S.M. 1994. Glutamine-dependent nitrogen trans-
fer in Escherichia coli asparagine synthetase B.
all of the graphical methods described above
J. Biol. Chem. 269:7450-7457.
may be unnecessary now that personal comput-
Bowen, W.J. and Kerwin, T.P. 1955. The purification
ers and associated new methods of data analysis
of myokinase with an ion exchange resin. Arch.
are available. For example, global analysis of Biochem. Biophys. 57:522-524.
biochemical data is a powerful tool that may
Brand, L. and Johnson, M.L. (eds.). 1992. Numeri-
prove to be quite useful in the analysis of cal computer methods. Methods Enzymol. Vol.
enzyme kinetics (Beechem, 1992). However, 210.
these numerical methods have not yet been Cleland, W.W. 1979a. Statistical analysis of enzyme
commonly applied to studies such as those of kinetic data. Methods Enzymol. 63:103-138.
multisubstrate systems, equilibrium ex- Cleland, W.W. 1979b. Substrate inhibition. Methods
changes, or product inhibition. At this time, Enzymol. 63:500-513.
with proper statistical weighting of the data, Cornish-Bowden, A. and Endrenyi, L. 1981. Fitting
multisubstrate systems are more readily ap- of enzyme kinetic data without prior knowledge
praised by one of the common graphical pro- of weights. Biochem. J. 193:1005-1008.
cedures. More importantly, these visual depic- Cornish-Bowden, A, and Endrenyi, L. 1986. Robust
tions of initial-rate data are a clear aid to any regression of enzyme kinetic data. Biochem. J.
individual reviewing the kinetic experiments. 234:21-29.
Deans, N.L., Allison, R.D., and Purich, D.L. 1992.
CONCLUDING REMARKS Steady-state kinetic mechanism of bovine brain
tubulin:tyrosine ligase. Biochem. J. 286:243-
Initial-rate assays are extremely useful tools
251.
in the detailed characterization of an enzyme.
Dybkaer, R.. 1979. International Union of Pure and
The behavior, mechanism, and regulatory prop-
Applied Chemistry and International Federation
erties of an enzyme can be readily assessed by of Clinical Chemistry. IUPAC Section of Clinical
straightforward assays. Such assays can fre- Chemistry; Commission on Quantities and Units
quently be completed within a day, once pre- in Clinical Chemistry and IFCC Committee on
liminary runs have identified the magnitude of Standards, Expert Panel on Quantities and Units;
Approved Recommendation (1978) Quantities
the kinetic parameters and revealed those fac-
and units in clinical chemistry. Clin. Chim. Acta
tors that affect activity. However, the details of 96:157F-183F.
the kinetic mechanism and regulatory proper-
Eisenthal, R. and Cornish-Bowden, A. 1974. The
ties of the enzyme can easily be obscured by direct linear plot. A new graphical procedure for
poor assay design. The structural/functional estimating enzyme kinetic parameters. Biochem.
role of an enzyme, as well as its regulation and J. 139:715-720. Detection and
control, can only be assessed by a reliable Assay Methods

3.5.9
Current Protocols in Protein Science Supplement 5
Feldman, H.A. 1983. Statistical limits in Scatchard hexokinase type III isozyme. Arch. Biochem.
analysis. J. Biol. Chem. 258:12865-12867. Biophys. 170:587-600.
Fierke, C.A. and Hammes, G. 1995. Transient ki- Silverstein, E. and Sulebele, G. 1969. Catalytic
netic approaches to enzyme mechanisms. Meth- mechanism of pig heart mitochondrial malate
ods Enzymol. 249:1-32. dehydrogenase studied by kinetics at equilib-
Frieden, C. 1959. Glutamate dehydrogenase III. The rium. Biochemistry 8:2543-2550.
order of substrate addition in the enzymatic re- Storer, A.C. and Cornish-Bowden, A. 1974. The
action. J. Biol. Chem. 234:2391-2396. kinetics of coupled enzyme reactions. Biochem.
Fromm, H.J. 1967. The use of competitive inhibitors J. 141:205-209.
in studying the mechanism of action of some Tate, S.S. and Meister, A. 1978. Serine-borate com-
enzyme systems utilizing three substrates. Bio- plex as a transition-state inhibitor of γ-glutamyl
chim. Biophys. Acta 139:221-230. transpeptidase. Proc. Natl. Acad. Sci. U.S.A.
Johnson, M.L. and Brand, L. (eds.). 1994. Numeri- 75:4806-4809.
cal computer methods, part B. Methods Enzymol. Wilkinson, G.N. 1961. Statistical estimations in en-
240:xi-xiii, 1-36, 51-170, 181-198, 311-322, zyme kinetics. Biochem. J. 80:324-332.
781-816. Williams, J.W., and Morrison, J.F. 1979. The kinet-
Klotz, I.M. 1982. Numbers of receptor sites from ics of reversible tight-binding inhibition. Meth-
Scatchard graphs: Facts and fantasies. Science ods Enzymol. 63:437-467.
217:1247-1249.
Klotz, I.M. 1983. Number of receptor sites from KEY REFERENCES
Scatchard and Klotz graphs: A constructive cri- Bergmeyer, 1983. See above.
tique. Science 220:981. Detailed collection of enzyme assay protocols and
Klump, H., Di Ruggiero, J., Kessel, M., Park, J.-B., methods for a wide variety of enzymes, including
Adams, M.W.W., and Robb, F.T. 1992. Gluta- descriptions of many special techniques: e.g.,
mate dehydrogenase from the hyperthermophile fluorometry, turbidimetry, luminometry, and micro-
Pyrococcus furiosus. J. Biol. Chem. 267:22681- techniques.
22685.
Dixon, M. and Webb, E.C. 1979. Enzymes, 3rd ed.
Lienhard, G.E. and Secemski, I.I. 1973. P1, P5- Academic Press, New York.
Di(adenosine-5′)pentaphosphate, a potent mul-
tisubstrate inhibitor of adenylate kinase. J. Biol. Classic text on the basics of enzymology; Chapter 4
Chem. 248:1121-1123. deals specifically with enzyme kinetics.
Marmasse, C. 1963. Enzyme inhibition by excess Fromm, H.J. 1975. Initial Rate Enzyme Kinetics.
substrate. Biochim. Biophys. Acta 77:530-535. Springer-Verlag, New York.
McClure, W.R. 1969. A kinetic analysis of coupled Valuable introductory text with considerable details
enzyme assays. Biochem. 8:2782-2786. on experimental design.
Nordlie, R.C. 1982. Kinetic examination of enzyme
mechanisms involving branched reaction path- Kuby, S.A. 1990. A Study of Enzymes, vol. I. CRC
ways: A detailed consideration of multifunc- Press, Boca Raton, Florida
tional glucose-6-phosphatase. Methods Enzy- Detailed text on steady-state kinetics. Contains use-
mol. 87:319-353. ful chapters on the effects of metal cofactors and
O’Sullivan, W.J. and Smithers, G.W. 1979. Stability transient phase kinetics.
constants for biologically important metal-li- Purich, D.L. (ed.) 1979. Enzyme kinetics and
gand complexes. Methods Enzymol. 63:294-336. mechanism, part A. Methods Enzymol. Vol. 63.
Purich, D.L. and Fromm, H.J. 1972. Inhibition of Describes a variety of initial rate methods and in-
rabbit muscle adenylate kinase by the transition hibitor studies for characterizing enzyme-catalyzed
state analogue, P1,P4-di(adenosine-5′)tetraphos- reactions.
phate. Biochim. Biophys. Acta 276:563-567.
Rudolph, F.B. 1979. Product inhibition and abortive Purich, D.L. (ed.) 1980. Enzyme kinetics and
complex formation. Methods Enzymol. 63: 411- mechanism, part B. Methods Enzymol. Vol. 64.
436. Describes a number of isotopic probes for studying
Scatchard, G. 1949. The attraction of proteins for enzyme mechanisms and contains valuable chapters
small molecules and ions. Ann. N.Y. Acad. Sci. on allosterism, hysteresis, immobilized systems, and
51:660-673. processivity.
Seelig, G.F. and Meister, A. 1985. Glutathione bio- Purich, D. L. (ed.) 1982. Enzyme kinetics and
synthesis: γ-Glutamylcysteine synthetase from mechanism, part C. Methods Enzymol. Vol. 87.
rat kidney. Methods Enzymol. 113:379-390.
Describes methods for characterizing intermediates
Siano, D.B., Zyskind, J.W., and Fromm, H.J. 1975. in enzyme-catalyzed reactions and using stereo-
A computer program for fitting and statistically chemical probes for enzyme mechanisms; includes
analyzing initial rate data applied to bovine additional initial-rate and inhibitor methods and
Kinetic Assay discusses further uses of isotopic probes.
Methods

3.5.10
Supplement 5 Current Protocols in Protein Science
Purich, D.L. (ed.) 1995. Enzyme kinetics and
mechanism, part D. Methods in Enzymol. 249:3-
662.
Contributed by R. Donald Allison
University of Florida
Covers in detail specialized topics in enzyme kinet- Gainesville, Florida
ics including transition-state approaches, kinetic
probes with site-directed mutagenesis, partition
analysis, positional isotope exchange, interfacial
catalysis, and hydrogen tunneling.

Detection and
Assay Methods

3.5.11
Current Protocols in Protein Science Supplement 5

You might also like