Anda di halaman 1dari 21

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/280063067

Quantitative Enzymology

Article  in  Current Enzyme Inhibition · April 2015


DOI: 10.2174/157340801101150707124226

CITATIONS READS
3 1,855

1 author:

David J Timson
University of Brighton
208 PUBLICATIONS   3,053 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Calcium binding proteins from helminth parasites View project

Metabolic enzymes from helminth parasites View project

All content following this page was uploaded by David J Timson on 21 July 2015.

The user has requested enhancement of the downloaded file.


Send Orders for Reprints to reprints@benthamscience.ae
12 Current Enzyme Inhibition, 2015, 11, 12-31

Quantitative Enzymology
David J. Timson*

School of Biological Sciences, Queen’s University Belfast, Medical Biology Centre,


97 Lisburn Road, Belfast, BT9 7BL. UK
Abstract: Accurate measurement of the quantitative aspects of enzyme-catalysed reactions
is critical for a deeper understanding of their mechanisms, for their exploitation in biotech-
nology and for targeting enzymes by drug-like molecules. It is important to move beyond
basic enzyme kinetics as encapsulated in the Michaelis-Menten equation. The type and
magnitude of inhibition should be determined. Since the majority of enzyme-catalysed reac-
tions involve more than one substrate, it is critical to understand how to treat these reactions
quantitatively and how their kinetic behaviour depends on the type of mechanism occurring.
Some reactions do not conform to “standard” Michaelis-Menten treatment and exhibit phenomena such as co-
operativity. Again it is important to put these phenomena onto a quantitative basis. Similarly the treatment of
the effects of pH on enzymes is often vague and uninformative without a proper quantitative treatment. This
review brings together tools and approaches for dealing with enzymes quantitatively together with original
references for these approaches.
Keywords: Enzymes, cooperativity, Michaelis-Menten equation, multisubstrate, quantitative enzymology
enzyme inhibition.

INTRODUCTION and how drugs modulate activity) and biotechnol-


ogy (where exploiting and engineering enzymes
Without enzymes, there would be no life. The for use in industrial catalysis is of increasing inter-
vast majority of biologically important reactions est). Some important examples of enzymes of im-
happen far too slowly to sustain life. It is enzymes, portance or interest in these fields are listed in Ta-
with their substantial rate enhancements which ble 1.
enable these reactions to occur on meaningful
timescales. The selectivity and specificity of en- RATE, ACTIVITY AND SPECIFIC ACTIVI-
zymes also permit them to steer biological reac- TY
tions towards particular products thus enabling
metabolic pathways to convert molecules and ex- Rates of enzyme catalysed reactions are typical-
tract energy from them. Therefore, understanding ly measured as rates of change of concentration –
how enzymes work is critical to our overall under- either the appearance of product or the disappear-
standing of biology – and this understanding re- ance of substrate (e.g. in units of μM min-1). The
quires a quantitative basis. Only by understanding activity of an enzyme refers to the amount of sub-
the relationship between the concentrations of re- stance converted per unit time (e.g. μmol min-1) at a
actants and rates can we begin to gain deep, mech- given substrate and enzyme concentration. A more
anistic knowledge of a particular enzyme. When useful quantitative measure of an enzyme’s cata-
coupled with structural and biochemical data, it is lytic power is the specific activity. In general, this
possible to build up a picture of how these remark- is measured at a single substrate concentration and
able catalysts work. This insight has implications a single enzyme concentration. It is typically ex-
for basic science, medicine (both from understand- pressed as amount of substrate transformed (or
ing how mutations affect enzyme function product produced) per unit time per unit mass of
protein (e.g. μmol min-1 mg-1). These measures
*Address correspondence to this author at the School of Biological
have the advantage of being applicable to mixtures
Sciences, Queen’s University Belfast, Medical Biology Centre, 97 of proteins (for example, cell extracts). Thus they
Lisburn Road, Belfast, BT9 7BL. UK; Tel: +44(0)2890975875; can be useful in comparing the enzyme activity in
Fax: +44(0)2890975877; E-mail: d.timson@qub.ac.uk
different cells or tissues of an organism or the

1875-6662/15 $58.00+.00 © 2015 Bentham Science Publishers


Quantitative Enzymology Current Enzyme Inhibition, 2015, Vol. 11, No. 1 13

Table 1. Some examples of enzymes important in metabolism, disease, drug action and biotechnology.

Enzyme EC number Comments Ref.

Metabolism Hexokinase 2.7.1.1. Catalyses the phosphorylation of glucose, the first committed step in glycolysis. [50]
Bovine enzyme has random ternary complex mechanism.
Phosphofructoki- 2.7.1.11 Catalyses the phosphorylation of fructose 6-phosphate, a key step in glycolysis. [51]
nase Bacterial enzyme shows positive cooperativity towards fructose 6-phosphate.
Succinate dehy- 1.3.5.1 Mitochondrial membrane associated enzyme which catalyses a step in the tricar- [52]
drogenase boxylic acid cycle. Competitively inhibited by malonate.
Galactokinase 2.7.1.6 Catalyses the phosphorylation of galactose in the Leloir pathway. Has a variety of [37, 38,
ternary complex mechanisms, depending on the species. 40, 41,
53]
Galactose muta- 5.1.3.3 Catalyses the equilibrium between the - and -anomers of galactose and other [54]
rotase monosaccharides. Key catalytic residues have been identified by pH studies.
Disease Galactose 1- 2.7.7.12 Key enzyme in the Leloir pathway of galactose metabolism. Mutations in the [55-58]
phosphate uri- corresponding gene can result in type I galactosemia. Substituted enzyme mecha-
dylyltransferase nism.
NAD(P)H quinone 1.6.5.2 Enzyme involved in quinone metabolism and detoxification. A common poly- [59, 60]
oxidoreductase 1 morphism (p.P187S) is associated with an increased risk of cancer. NQO1 has a
(NQO1) substituted enzyme mechanism and is competitively inhibited by the anticoagu-
lant dicoumarol.
Coagulation factor 3.4.21.22 A glycosylated, extracellular serine protease enzyme required for blood clotting. [61, 62]
IXa Deficiency results in haemophilia type B which can be treated by injection of
recombinant factor IXa. Current research efforts are focused on increasing the
plasma half-life of this proteins.
-Secretase 1 3.4.23.46 Membrane-bound protease. Increased levels are associated with late-onset non- [63, 64]
(BACE 1) familial Alzheimer’s disease. Inhibition of this enzyme is possible, future treat-
ment.
Viral neuramini- 3.2.1.129 Catalyses the hydrolysis of sialic acid groups from complex oligosaccharides. [65-70]
dase (sialidase) Essential enzyme for release of some viruses from host cells. The enzyme is the
target of a number of antiviral drugs, including zanamivir (Relenza) and oselta-
mivir (Tamiflu).
Drug action Cyclooxygenase 1.14.99.1 Key enzyme of prostaglandin synthesis. Target of the non-steroidal anti- [19, 71]
inflammatory drugs aspirin (irreversible, covalent inhibition) and ibuprofen
(reversible, competitive inhibition)
Phosphoglycerate 2.7.2.3 Glycolytic enzyme. Likely target of the veterinary anthelminthic drug Clorsulon [72]
kinase which acts as a competitive inhibitor.
Phosphodiesterase 3.1.4.17 Catalyses the hydrolysis of cyclic AMP and cyclic GMP and thus responsible for [73, 74]
the down-regulation of a number of cellular processes. The human PDE5 isoform
is the target of sildenafil sulphate (Viagra) which acts as a competitive inhibitor.
Mitogen-activated 2.7.12.2 Cell signalling enzyme. The potential anti-cancer drug Selumetinib is an uncom- [75, 76]
protein kinase petitive inhibitor.
kinase (MAPKK)
-lactamase 3.5.2.6 Enzyme implicated in bacterial resistance to penicillin family antibiotics. Current [77-80]
interest in developing inhibitors of this enzyme to potentiate pencillins and over-
come resistance.
Biotechnology Laccase 1.10.3.2 Copper containing redox enzymes with biotechnological potential. Studies on the [81-83]
pH effects identified pKa values of functional groups involved in catalysis. Many
laccases show considerable thermal stability.
Glucose oxidase 1.1.3.4 Catalyses the oxygen dependent oxidation of glucose. Used for the detection of [84]
glucose concentrations (e.g. in blood glucose meters for diabetics).
Thermolysin 3.4.24.27 Thermostable protease. Reverse reaction exploited in the synthesis of short pep- [85-87]
tides, including the sweetener aspartame.
Urease 3.5.1.5 Catalyses the hydrolysis of urea to carbon dioxide and water. Potential for use in [88]
biosensors for urea.
Cellulase 3.2.1.4 Catalyses the hydrolysis of cellulose. Potential importance in optimising biofuel [89-91]
yields from plant based feedstocks. Vanillin (and some other phenolic com-
pounds commonly present in plant materials) is reported to be a non-competitive
inhibitor of some cellulases.
Readers are also referred to the Bremen Enzyme Database (BRENDA; http://www.brenda-enzymes.org/) for a comprehensive database of enzymes and their kinetic properties.
14 Current Enzyme Inhibition, 2015, Vol. 11, No. 1 David J. Timson

amount of recombinant enzyme produced under ble formation of the enzyme-substrate complex,
different culture conditions. However, their utility the conversion of this complex to an enzyme
is limited by the non-linear dependence of rate on product complex and the (reversible) release of the
substrate concentrations under most experimental product, regenerating the pure enzyme [4].
conditions (see below). This means, that if mean- (a)
ingful comparisons are to be made, then care must
be taken to duplicate exactly the concentrations of
reagents used.

THE MICHAELIS-MENTEN EQUATION


Leonor Michaelis and Maud Menten first de-
rived the equation (Equation 1) that bears their
names based on experimental work on invertase
and using the assumption that the enzyme and sub-
strate form an equilibrium complex prior to reac-
tion [1, 2]. Briggs and Haldane demonstrated that
this equilibrium assumption (which is unlikely to
apply to the majority of enzymes) was not neces- (b)
k+1 k2
sary and that essentially the same equation could E+S ES EP E+P
be derived using steady-state kinetic principles [3]. k1
The equation was, therefore, likely to be applica- Fig. (1). The fundamentals of steady-state enzyme kinet-
ble to a wide range of single substrate enzymes ics with a single substrate enzyme (a) An ideal Michaelis-
provided that certain assumptions are satisfied. Menten curve. In this case, Km = 30 μM and Vmax = 100 μMs-1
The substrate concentration must greatly exceed (b) A simplified kinetic mechanism for a one substrate en-
that of the enzyme and the chemical step (i.e. the zyme catalysed reaction. E, enzyme; S, substrate; P, product.
conversion of substrate to product) must be the
slowest step of the reaction. From this, the steady state constants Km and
  Vmax can be defined based on the rate constants of
 (1) the individual steps (for definitions of the rate con-
 
stants, see Fig. 1b).
Mathematically, this equation describes a rec-
tangular hyperbola. It gives rise to a curve (Fig.     (2)
1a) which initially rises steeply before plateauing.
where [E] is the total, active enzyme concentra-
Vmax is the maximum possible rate under the con-
tion. The rate constant k2 is often referred to as kcat
ditions of the experiment. The rate (v) asymptoti-
or turnover number (or catalytic rate constant).
cally approaches Vmax but never reaches it at any
The turnover number is a measure of the number
physically achievable substrate concentration [S].
of substrate molecules converted per enzyme mol-
The Michaelis constant (Km) is equal to the sub-
ecule per unit time. In general, it has units of in-
strate concentration which gives a rate equal to
verse time (e.g. min-1).
half Vmax. It thus has units of concentration. This is
often, erroneously, equated to the equilibrium con-  
  (3)
stant for the enzyme substrate complex. In fact, 
under steady-state conditions, it is a composite of
Thus, the Michaelis constant only approximates
three different rate constants (see below). Steady-
to the dissociation constant for the enzyme-
state conditions apply when the rate of enzyme- substrate complex if k2 is small compared to k-1.
substrate complex formation equals the rate of its
conversion to products; in other words, when the There is another useful constant which arises
concentration of enzyme-substrate complex is con- from the Michaelis-Menten equation and that is
stant. Thus, in a simplified mechanism for a sin- the ratio of kcat/Km. This is referred to as the speci-
gle-substrate enzyme, a series of steps can be de- ficity constant and can be used as a measure of
fined (Fig. 1b) beginning with the initial, reversi- which substrate enzyme “prefers”: the higher the
Quantitative Enzymology Current Enzyme Inhibition, 2015, Vol. 11, No. 1 15

value of the specificity constant the greater the to linearise enzyme kinetic data have been pro-
preference of the enzyme for that substrate. It has posed. Of these, the most widely used is the so-
units of inverse concentration per second, i.e. the called Lineweaver-Burk (or double reciprocal) plot
same as for second order rate constants (e.g. μM-1 in which the reciprocals of the rate and the sub-
s-1 or l mol-1 s-1). The specificity constant can be strate concentration are plotted against each other
used to rank substrate preferences of enzymes. For (Fig. 2a, b) [6, 7]. Despite its common use, this
example, human galactose mutarotase has a speci- plot suffers from some well-documented short-
ficity constant of 340,000 l mol-1 s-1 for galactose comings. The most significant of these is the ten-
and 90,000 l mol-1 s-1 for glucose; the enzyme dency to magnify errors in small quantities. Often
clearly “prefers” galactose as a substrate [5]. the smallest rates are the hardest measure and thus
subject to the greatest errors. However, in a double
DETERMINING KINETIC PARAMETERS reciprocal plot, these small values become large
FROM THE MICHAELIS-MENTEN EQUA- and often have a disproportionate effect on the fit-
TION ting of the straight line [7-11]. This can be partial-
ly mitigated by planning the experiment in “recip-
In practice, accurate estimation of Km and Vmax rocal space”, i.e. such that the points are evenly
from visual inspection of a plot of v against [S] is distributed in the Lineweaver-Burk plot.
almost impossible. Even when the substrate con-
centration is ten times the Michaelis constant, the A number of alternatives to the Lineweaver-
rate is only 0.91Vmax. Various practical issues Burke plot exist. These include the Eadie-Hofstee
make it difficult to achieve even these levels of plot (v against v/[S]) and the Hanes-Woolf plot
substrate concentration in many cases. These in- ([S]/v against [S]) (Fig. 2a, c, d) [12-15]. While
clude limited solubility of the substrate, limited these eliminate some of the issues associated with
supply of the substrate and that some substrates the Lineweaver-Burk plot, they bring the new is-
have other effects on the reaction (e.g. they affect sue that one variable is present on both axes. Thus,
pH or ionic strength) which become apparent at any errors associated with the measurement of that
higher concentrations. Therefore, several methods variable affect both the x- and y-coordinates and

(a) (b) (c)

Fig. (2). Linearisation of steady-state kinetic data (a) A plot of v against [S] showing simulated data points for which Km =
30 μM and Vmax = 100 μMs-1. (b) The same data transformed to make a Lineweaver-Burk plot. Note how the data points bunch
towards the lower left of the graph, despite being evenly spaced in (a). (c) The same data transformed to make an Eadie-
Hofstee plot. (d) The same data transformed to make a Hanes-Woolf plot. [A note on the representation of units in this and
other figures: here the convention is used to show the variable separated from the unit by a forward slash (“/”). This indicates
that the quantities shown on the axes are the result of dividing the value by the unit, resulting in a pure number. For example, if
the number 10 is shown on the axes and the axis is labelled “[S]/μM”, then this tells us that the actual value (10 μM) has been
divided by the unit (μM) leaving the pure number. This convention is used in many journals and is preferred by nomenclature
and standard setting bodies, for example the National Institute of Science and Technology (NIST, USA) [49].
16 Current Enzyme Inhibition, 2015, Vol. 11, No. 1 David J. Timson

disproportionately influence the estimate. are generally considered more accurate than those
determined by the Lineweaver-Burk plot and other
Increasingly, direct non-linear fitting of the ki- similar methods. However, it is necessary to avoid
netic data is used to estimate kinetic constants the trap of assuming that the computer program
from experimental data. Linear fitting (regression) “knows best”. Any fit should be critically as-
is widely used in science and many commonly sessed. If it clearly deviates from the data points,
used software packages include linear regression either the fit is a poor estimate or the equation to
functions. Normally, linear regression is achieved which the data are being fitted is inappropriate
by the least squares method. In this method, the (Fig. 3). Better estimates can sometimes be ob-
“best fit” line is the one with the lowest sum of the tained by altering the initial estimates of the pa-
squares of the differences between the line and the rameters (since all non-linear fitting programmes
data points. In general, there is one unique, analyt- can fail to find the best fit). The appropriateness of
ical solution to a least squares fit of any data. the equation can be tested statistically by compar-
(However, it should be noted that the fit will be ing it with fits to other possible equations. Another
meaningless if the data do not conform to a linear measure of the quality of the fit comes from calcu-
relationship.) Linear regression can be used in lation of the residuals, that is the difference be-
combination with linearized kinetic data to esti- tween the fit and the points. The residuals should
mate kinetic constants. Non-linear fitting works by be small in comparison to the measured values and
similar principles, i.e. the minimisation of the sum randomly distributed about zero (Fig. 3). It is pos-
of the squares of the differences between the line sible to test both of these statistically; however,
(model) and the experimentally determined data visual inspection of the residuals is often enough
points [16]. However, there is rarely a single, ana- to detect a poor quality fit. Ultimately, the judg-
lytical solution and the “best fit” must be found by ment lies with the experimenter and fits which are
incrementally changing the values and testing clearly poor on inspection should be rejected re-
whether they make the fit better or worse. Inevita- gardless of what the computer program says.
bly this is more computationally demanding, espe-
cially for complex equations. Nowadays a number THE NATURE AND TYPES OF ENZYME
of software packages are available which carry out INHIBITION
robust non-linear curve fitting and these can be
readily used with enzyme kinetic data. Since no Enzyme inhibitors are compounds which re-
linearization is involved, the estimates obtained duce the rate of an enzyme catalysed reaction.

(a) (b) (c)

Fig. (3). Non-linear curve fitting. (a) Simulated data fitted to the Michaelis-Menten equation (top) with small, randomly dis-
tributed residuals (bottom). (b) Simulated data fitted to the Michaelis-Menten equation (top) with large but randomly distribut-
ed residuals (bottom). (c) Simulated data fitted to the Michaelis-Menten equation (top) with small, but non-randomly distribut-
ed residuals (bottom). Note how in this case, the actual fit looks reasonable but the residuals reveal that there is a problem with
either the data or the choice of equation used for non-linear fitting.
Quantitative Enzymology Current Enzyme Inhibition, 2015, Vol. 11, No. 1 17

Normally, we are most interested in those com- tors and the nomenclature for these has become
pounds which inhibit specific enzymes or reac- confused over the years. The most commonly de-
tions. Although there are a number of compounds scribed form of reversible inhibition is competitive
which inhibit a wide range of enzymes, these nor- inhibition. Competitive inhibitors bind to enzymes
mally do so through denaturation of the protein at their active sites and sterically hinder access by
(e.g chaotropic agents such as urea [17]). Enzymes the substrate. One consequence of this molecular
which target specific enzymes or groups or related mechanism is that the degree is inhibition is con-
enzymes are more likely to be useful as leads in trolled by the relative concentrations of the sub-
drug discovery or reagents for use in laboratory strate and the inhibitor. Thus, the degree of inhibi-
investigations. tion can be reduced by increasing the concentra-
tion of substrate. The binding of a competitive in-
Careful, quantitative measurement of the effects hibitor to an enzyme can be considered as an equi-
of specific inhibitors on the rates of enzyme cata- librium process with the equilibrium constant, Kic.
lysed reactions can reveal much about the molecu- Since a purely competitive inhibitor cannot bind to
lar basis of their action. Enzyme inhibitors can be the enzyme-substrate complex (because its binding
divided into two main classes – reversible and ir- site is occupied) then it must only bind to the free
reversible. Irreversible inhibitors are those which enzyme. The equilibrium constant, Kic is also
bind so tightly to the enzyme that they cannot be known as the competitive inhibition constant and it
removed by common laboratory separation tech- is defined in Equation 4.
niques (e.g. dialysis, gel filtration chromatog-
raphy). This means that the interaction between  
  (4)
the enzyme and inhibitor can be detected by meth- 
ods which measure changes in molecular mass, for ([I] is the concentration of inhibitor and [EI] the
example, MALDI-TOF mass spectrometry. In concentration of the non-covalent enzyme-
general, irreversible inhibitors form covalent inhibitor complex).
bonds with the enzyme molecule. Thus, stoichio-
metric amounts of inhibitor can result in total inhi- Competitive inhibition is not the only type of
bition of the enzyme. The effects of irreversible inhibition encountered. Uncompetitive inhibition
inhibitors on steady state enzyme kinetics can be occurs when the inhibitor interacts with the en-
hard to quantify. In addition to the enzyme- zyme-inhibitor complex at a location other than
catalysed reaction a second chemical process is the active site. Therefore, uncompetitive inhibi-
occurring, i.e. the chemical reaction between the tors do not, directly interfere with the binding of
enzyme and the inhibitor. It is, of course, possible substrate and their effects cannot be reversed by
to treat this enzyme-inhibitor reaction as a classi- the addition of higher concentrations of substrate.
cal second order process and derive rate constants At a molecular level, uncompetitive inhibitors
for this. The widely prescribed analgesic and anti- reduce the rate of reaction due to allosteric ef-
inflammatory drug aspirin (acetylsalicylic acid) is fects: binding at one site causes changes in the
an irreversible inhibitor of cyclo-oxygenase 2 protein’s overall conformation which affects the
(COX-2) [18-21]. Irreversible inhibitors have structure, and therefore reactivity, of the active
some advantages as drugs. Since stoichiometric site. Uncompetitive inhibition is characterised by
amounts are required for inhibition, doses can be the uncompetitive inhibition constant, Kiu; like its
low. Their irreversible nature means that once in- counterpart in competitive inhibition this is also a
hibited, an enzyme molecule is unlikely to regain real equilibrium constant, defined according to
activity. Therefore, the only way the cell can re- Equation 5:
gain that activity is to synthesise more enzyme.  
However, the reactive nature of some irreversible   (5)

inhibitors means that there is a potential risk of
covalent modification of non-target proteins which ([ES] is the concentration of the enzyme-substrate
could lead to significant side effects. complex; [ESI] is the concentration of the enzyme-
substrate-inhibitor complex).
Reversible inhibitors normally interact with en-
zymes through weaker, non-covalent bonding. It is also possible to conceive of a third type of
There are various subgroups of reversible inhibi- inhibition in which the inhibitor binds both to the
18 Current Enzyme Inhibition, 2015, Vol. 11, No. 1 David J. Timson

free enzyme and to the enzyme-substrate complex. If the reciprocal rate of a competitively inhibit-
Such inhibition will be characterised by both com- ed reaction is plotted against inhibitor concentra-
petitive and uncompetitive inhibition constants and tion, the result is a straight line. Repeating the pro-
is known as mixed inhibition. Non-competitive cess at a second substrate concentration results in
inhibition is a special case of mixed inhibition in another straight line which intersects with the first
which the inhibitor binds equally strongly to the in the (-x,y) quadrant (Fig. 4c). Lines resulting
free enzyme and the enzyme-substrate complex from further substrate concentrations will also
and, thus, the competitive and uncompetitive inhi- cross at the same point and the x-value of this
bition constants are equal. Some authors doubt that point is equal to –Kic (and the y-value to 1/Vmax).
non-competitive inhibition occurs commonly in This plot (known as the Dixon plot [23]) can be
real enzyme systems [22]. In reality, mixed inhibi- used to estimate the value of the competitive inhi-
tion is likely to be by far the most common type of bition constant. However, it should be noted that
reversible inhibition. However, either the competi- intersection of these lines in the (-x,y) quadrant
tive or uncompetitive element may be far greater does not prove competitive inhibition since the
than the other and may thus dominate any analysis. same result will be seen with mixed inhibition [22,
24]. Of course, with real experimental data the in-
QUANTIFICATION OF REVERSIBLE EN- tersection of three or more lines will not be exactly
ZYME INHIBITION on the same point and the centroid of the intersec-
tions should be calculated in order to estimate Kic.
The Michaelis-Menten equation (Equation 1) The Dixon plot suffers from a number of prob-
can be modified to allow for the effects of reversi- lems, not least the need to use reciprocals of the
ble inhibition [22]. Using these equations and rate leading to similar issues to the Lineweaver-
carefully designed experiments, the inhibition con- Burk plot (see above) [25].
stants can be determined. The steady state equa-
tion for competitive inhibition (Equation 6) modi- Close examination of Equation 6 shows that it
fies the Km term, reflecting the fact that competi- is in the same form as the Michaelis-Menten equa-
tive inhibition affects only Km and not Vmax. tion. Indeed, it can be rewritten as:
Vmax [S]
Vmax [S] (6) v= (7)
v= K m,app + [S]
 [I] 
K m  1+ + [S]
 K ic  Here, Km,app (the apparent Km) replaces one of the
terms in the denominator of the fraction such that:
This equation describes a curve of similar
shape to the Michaelis-Menten equation (i.e. a 
     (8)
rectangular hyperbola) in which increasing the 
value of [I] while keeping all other values con- Therefore, if the apparent Km is determined for
stant shifts the curve to the right without affecting a range of different values of inhibitor concentra-
the limiting value of v (Fig. 4a). In theory, data tion then a plot of Km,app against [I] can be used to
can be fitted to Equation 6 by non-linear curve determine Kic (Fig. 4d). The gradient of the line
fitting to yield the value of Kic (and Km and Vmax). will be 1/Kic (and the y-intercept will be Km). This
In practice, as equations become more complex relatively straightforward example illustrates a
and the number of constants increases, non-linear general case that, where more complex equations
curve fitting is more likely to fail. It may also be can be reduced to a form similar to the Michaelis-
necessary to evaluate first which form of inhibi- Menten equation, it is then possible to construct
tion is occurring. (Some studies mistakenly as- so-called secondary plots (or replots) to determine
sume that inhibition is competitive since this is the inhibition constants.
the best known and most straightforward to ana-
lyse.) They key feature of competitive inhibition In uncompetitive inhibition both Km and Vmax
is that Vmax is invariant. Therefore, in a Lin- are both reduced by the same fraction; therefore
eweaver-Burk plot, all lines should pass through the ratio of Vmax to Km is unaffected by the pres-
the y-axis at the same point regardless of the con- ence of an uncompetitive inhibitor (Fig. 5a). In a
centration of inhibitor (Fig. 4b). Lineweaver-Burk plot, increasing concentrations
of an uncompetitive inhibitor give rise to parallel
Quantitative Enzymology Current Enzyme Inhibition, 2015, Vol. 11, No. 1 19

(a)

(b)(i) (ii)

(c) (d)

Fig. (4). Competitive inhibition (a) Michaelis-Menten plots of simulated kinetic data with , Km=30 μM, Vmax=100 μMs-1,
Kic=10 μM and (from top curve to bottom) 0, 10, 25 and 50 μM inhibitor. (b) (i) A Lineweaver-Burk plot using the same simu-
lated data as (a). (ii) a close-up of the graph shown in (b)(i) showing the y-intercept; note how all lines pass through the same
intercept because Vmax is unaffected. (c) A Dixon plot using data for three selected substrate concentrations from (a) – one be-
low Km (16.7 μM), one at Km (30 μM) and one higher than Km (70 μM). Note how the lines intercept in the (-x,y) quadrant as
indicated by the arrow. (d) A secondary plot constructed according to Equations 7 and 8 (see text for detail) and using the same
data as in (a).

lines (since the gradient in this plot is Km/Vmax) In order to produce a form which resembles the
(Fig. 5b). Parallel lines are also seen in the Dixon Michaelis-Menten equation, it is necessary to have
plot with uncompetitive inhibitors (Fig. 5c). An [S] in the denominator not subject to multiplica-
alternative form of this plot has been proposed for tion by any term. Dividing [S] (and all other
the determination of the uncompetitive inhibition terms) by (1+[I]/Kiu) results in an alternative form
constant Kiu. In this plot [S]/v is plotted against of Equations 9:
[I] for a range of different substrate concentra- 
 
tions. The result is very similar to the Dixon plot:  
the lines intersect in the (-x,y) quadrant such that   (9a)
 
the x-value at the interception point is - Kiu (and  

the y-value is equal to Km/Vmax) (Fig. 5d) [26].
The steady state rate equation for uncompetitive This is in the same form as the Michaelis-
inhibition is: Menten equation and can be rewritten as:
Vmax [S] Vmax, app [S]
v= (9) v= (9b)
 [I]  K m , app + [ S]
K m + [S]  1+
 K iu 
20 Current Enzyme Inhibition, 2015, Vol. 11, No. 1 David J. Timson

(a) (b)

(c) (d)

(e) (f)

Fig. (5). Uncompetitive inhibition (a) Michaelis-Menten plots of simulated kinetic data with , Km=30 μM, Vmax=100 μMs-1,
Kiu=10 μM and (from top curve to bottom) 0, 10, 25 and 50 μM inhibitor. (b) A Lineweaver-Burk plot using the same simulat-
ed data as (a). (c) A Dixon plot using data for three selected substrate concentrations from (a), –16, 32 and 64 μM. Note how
the lines are parallel in this type of inhibition. (d) A plot of [S]/v against [I] for the same three substrate concentrations shown
in (c). Note how the lines intercept in the (-x,y) quadrant as indicated by the arrow. (e) A secondary plot showing the depend-
ence of Km,app on inhibitor concentration, constructed according to Equation 10 (see text for detail) and using the same data as
in (a). (f) A secondary plot showing the dependence of Vmax,app on inhibitor concentration, constructed according to Equation
11 (see text for detail) and using the same data as in (a).

Therefore, in this case, there are two apparent f). There are a variety of approaches to lineariza-
kinetic constants, both of which are dependent on tion, but the most straightforward is to take recip-
the inhibitor concentration. These are defined as: rocals of the apparent kinetic constant and plot
these against the inhibitor concentration. This

   (10) yields a plot in which the gradient is 1/KiuVmax and
 the y-intercept is 1/Vmax. This is clearly somewhat

cumbersome and nonlinear curve fitting is pre-
 ferred. It is also clear from Equations 10 and 11
   (11)
 that the ratio of Vmax,app/Km,app will be equal to

Vmax/Km for all values of inhibitor concentration.
Both equations 10 and 11 can be used to deter- Since kcat is the maximum rate divided by the ac-
mine the uncompetitive inhibition constant. This tive enzyme concentration (Equation 2) it also fol-
can either be done by direct, nonlinear curve fit- lows from this that the specificity constant is unaf-
ting or by linearization of these equations (Fig. 5e, fected by an uncompetitive inhibitor.
Quantitative Enzymology Current Enzyme Inhibition, 2015, Vol. 11, No. 1 21

Mixed inhibition affects both Km and Vmax; both simplified by fixing the values of Km and/or Kiu if
constants are altered by different fractions. Thus, it these have already been determined.
requires both a competitive and uncompetitive in-
hibition constant to describe it quantitatively. Non- The approach of simplifying rate equations so
competitive inhibition can be considered as a spe- that they are in the form of the Michaelis-Menten
cial case of mixed inhibition, in which Kic and Kiu equation and then deriving equations for the ap-
are equal. This has the effect of keeping Km con- parent kinetic constants is applicable to competi-
stant and reducing Vmax (Fig. 6). The general tive, uncompetitive and mixed inhibition. In each
steady state rate equation for non-competitive and case, one or more secondary plots are required in
mixed inhibition is: which apparent kinetic constants are plotted
against the inhibitor concentration. This requires
the apparent constants to be known for a range of
inhibitor concentrations. Although the secondary
plot method is generally considered to be the “gold
standard” for determining inhibition constants,
there are sometimes practical issues. Probably the
most critical is a lack of time or reagents to per-
form the many individual assays required. In these
cases, it may be necessary to resort either to non-
linear fitting of the appropriate rate equation at one
inhibitor concentration or the use of Dixon plots
and their variants.
An alternative quantitative measure of inhibi-
Fig. (6). Noncompetitive inhibition is a special case of tion is the concentration of inhibitor required to
mixed inhibition. Michaelis-Menten plots of simulated ki- result in a 50% reduction in the rate (IC50). This
netic data with Km=30 μM, Vmax=100 μMs-1, Kic=Kiu=10 μM parameter is widely used for the screening of in-
and (from top curve to bottom) 0, 10, 25 and 50 μM inhibi- hibitors in the pharmaceutical and other industries.
tor. In noncompetitive inhibition, Km remains constant and
Vmax is reduced.
Often it is determined by using a wide range (sev-
eral orders of magnitude) of inhibitor concentra-
tion and so this is often plotted on a logarithmic
Vmax [S] (12) scale (Fig. 7a). It has the advantage of being rela-
v=
 [I]   [I]  tively straightforward to determine and interpret;
K m  1+  + [S]  1+
 K ic   K iu  IC50 is particularly useful for compiling rankings
of the inhibitory potential of large numbers of
Equation 12 can be reduced to a form similar to compounds. However, it provides no information
the Michaelis-Menten equation by a similar ap- on the underlying type of inhibition. Such infor-
proach to that adopted for Equation 9, i.e. dividing mation may be useful if further development of an
through by (1+[I]/Kiu). The definition of the appar- inhibitor is required since the type of inhibition
ent maximum rate, Vmax,app is the same as for un- provides some information on how the compound
competitive inhibition (Equation 11) and the ap- interacts with the enzyme. Another issue is that
parent Michaelis constant, Km,app is: IC50 values are highly dependent on the substrate

concentration used in the assay – and how these
  two parameters are related depends on the type of
    (13) inhibition. For example, in competitive inhibition
 
a much greater degree of rate reduction is ob-
There is no straightforward linearization of served when the substrate concentration is close to
Equation 13. However, if Kiu has already been de- Km than when it is many times Km and the rate is
termined it is possible, although cumbersome, to approaching Vmax; therefore a much higher inhibi-
determine Kic from a plot of Km,app(1+[I]/Kiu) tor concentration is required to give a 50% reduc-
against [I]. Once again, nonlinear curve fitting is tion in rate (Fig. 7b). Considerable care should be
preferable; in the case of Equation 13 it can be taken when comparing IC50 values from different
laboratories since the conditions of the assay (most
22 Current Enzyme Inhibition, 2015, Vol. 11, No. 1 David J. Timson

(a) the literature on the theoretical basis of quantita-


tive enzyme kinetics concentrates on reactions
which involve the conversion of one substrate to
one product. (The most common examples of the-
se are isomerisations; some reactions in which one
reactant is in vast excess, e.g. hydrolytic reactions,
can also be treated as being single substrate reac-
tions). Once we consider the general reaction in
which two substrates are converted to two prod-
ucts, the steady state rate equations need to be
modified to account for this. A general form of a
two substrate enzyme-catalysed reaction can be
written as:
(b)
A + B C + D
in which A and B are substrates and C and D are
products. There are several different ways in
which A and B can interact with the enzyme – and
there are different steady-state rate equations de-
pending on how this occurs [28-31].
In the substituted enzyme mechanism, A first
interacts with the enzyme and reacts with it. This
reaction normally involves the transfer of a chemi-
Fig. (7). IC50 is an alternative parameter to quantify in- cal group or electrons to the enzyme (or to a coen-
hibition. (a) A typical inhibition curve used to estimate IC50. zyme strongly associated with the enzyme). Ex-
In this case, the enzyme is undergoing competitive inhibition amples of commonly transferred groups include
with the simulated parameters Km=30 μM, Vmax=100 μMs-1 hydrides and phosphates. Since the enzyme has
and Kic=10 μM; the substrate concentration is equal to the been covalently modified by reaction with A, it
Km. (b) The effect of increasing [S] on IC50. Parameters are should be possible to detect this modification by
the same as in (a), expect for [S] which was varied as shown. analytical techniques. For example, if A transfers a
Fitted IC50 values were 12, 20, 60, 100 μM for [S]=5, 30,
150, 300 μM respectively. In both (a) and (b), v0 represents
phosphate group to the enzyme then there would
the initial rate in the absence of inhibitor. be an increase in mass of the protein, detectable by
mass spectrometry (and other methods which
critically, the value of [S]) are unlikely to be simi- measure molecular mass). Alternatively if A trans-
lar. If the type of inhibition, the Michaelis constant fers electrons to a coenzyme, this reduction is like-
and substrate concentration is known, IC50 values ly to be detectable spectrophotometrically. Once A
can be converted to inhibition constants using the has reacted with the enzyme, the remainder of the
Cheng-Prusoff relationships [27]. For example, molecule it will diffuse away from the active site
with competitive inhibitors, the relationship is: as product C. Therefore, reaction of enzyme with

A (in the absence of B) can be treated as a second
   (14) order chemical process and, often, exposing the

 enzyme to an excess of A will convert the vast ma-
Note that, in the case of competitive inhibition, jority of enzyme molecules. The enzyme then re-
when the substrate concentration is equal to Km, acts with B, transferring the group which came
this simplifies to Kic=IC50/2. from A to B. In this second step of the reaction,
the enzyme is regenerated and product D is
ENZYME-CATALYSED REACTIONS WITH formed. One feature of this mechanism is that A
MORE THAN ONE SUBSTRATE and B do not, directly, react with each other. In-
deed, they do not occupy the active site at the
The majority of enzymes catalyse reactions in- same time (Fig. 8a). This kinetic mechanism is
volving two or more substrates. However, much of sometimes known as the “ping-pong” reaction.
Quantitative Enzymology Current Enzyme Inhibition, 2015, Vol. 11, No. 1 23

Fig. (8). Kinetic mechanisms for enzymes with more than one substrate. (a) A schematic representation of a substituted
enzyme (or “ping-pong”) mechanism. The enzyme (E) first reacts with substrate A to form an adduct with part of A (A’), re-
leasing the first product, C. The enzyme adduct (EA’) then reacts with the second substrate, B generating the second product D
which is then released. (b) A hierarchical classification of the main mechanisms of bisubstrate enzyme catalysed reactions.

The steady state rate equation for the substitut- This yields equations for the apparent kinetic
ed enzyme mechanism is: parameters:
 
v=
Vmax [ A][B]
(15)     (16)

K m , B [ A] + K m, A [ B] + [A][B]
 
Although there is only one Vmax value for this     (17)


mechanism, each substrate has its own Michaelis
constant, Km,A and Km,B. If the concentration of A Both Equation 16 and Equation 17 are in the
is varied and the concentration of B is held con- same form as the Michaelis-Menten equation and
stant the dependence of v on [A] will be a rectan- so values for the kinetic constants can be extracted
gular hyperbola, just like the Michaelis-Menten by similar methods to those described above.
equation. This suggests a way to obtain the three Since the denominators of these two equations are
steady state kinetic parameters associated with this identical it follows that, as for uncompetitive in-
reaction. If the constant terms (i.e. Vmax, Km,A, Km,B hibitors, in the substituted enzyme mechanism
and, in this case, [B]) are collected together, Equa- Vmax,app/Km,app will be equal to Vmax/Km, i.e. the
tion 15 can be rewritten as: specificity constant is invariable.
(Vmax [B])[A] The other common kinetic mechanism for two
v= (15a)
K m,A [B] + [ A](K m,B + [B]) substrate reactions is the ternary complex mecha-
nism. In this mechanism both substrates bind to
Isolation of [A] in the denominator requires di- the active site at the same time to form a ternary
vision through by the constant term (Km,B+[B]) substrate complex (EAB). Once this has formed,
and gives a version of Equation 15 in the form of catalysis occurs and a ternary product complex
the Michaelis-Menten equation: (ECD) if formed. The products then dissociate
from the active site and diffuse away. Two types
 (Vmax [B]) 
  [A] of ternary complex mechanism can be distin-
(K + [B])
v=
m,B (15b) guished – random and ordered. In a random ter-
 K m,A [B] 
 + [ A] nary complex mechanism, the substrates can bind
(K + [B])
m,B
in any order. In contrast, in ordered mechanisms,
24 Current Enzyme Inhibition, 2015, Vol. 11, No. 1 David J. Timson

one substrate is required to bind before the second In practice examples of purely random or pure-
one. The most common, molecular explanations ly ordered mechanisms may be rare. A truly ran-
for ordered mechanisms are that the first substrate dom mechanism requires that the enzyme’s inter-
induces a conformational change in the enzyme action with each substrate has no effect on the af-
enabling binding to the second substrate or that the finity for the other substrate. Given the mutual
first substrate forms part of the binding site for the structural changes which generally occur on en-
second substrate. zyme-substrate binding, this is unlikely. A truly
ordered mechanism requires that the enzyme has
The steady state rate equation for both ternary no affinity for the second substrate to bind (or that
complex mechanisms is: it is only able to bind it in a non-productive way).
   Again this is unlikely when dealing with real en-
 (18) zymes. Nevertheless, well documented examples
        
exist of enzymes which conform (largely) to one
The new term in this equation, KIA is hard to in- mechanism or the other (for example, galactoki-
terpret and its precise meaning varies depending nase exhibits different mechanisms, depending on
on the type of ternary complex mechanism. Equa- the species [37-41]).
tion 18 can be reduced to a form similar to the
Michaelis-Menten equation by a similar process to EFFECTS OF pH ON ENZYME ACTIVITY
that used for the substituted enzyme mechanism
(Equation 15). Once again, if one substrate con- Hydrogen ion concentration has a variety of in-
centration is held constant and the other varied, terrelated effects on enzymes. At extremes of pH,
constant terms can be collected together. This the majority of enzymes will be denatured and,
yields the same equation for Vmax,app as for the sub- therefore, lose activity. However, the protonation
stituted enzyme mechanism (Equation 16) and the and deprotonation of amino acid residues can also
following equation for Km,app: have more subtle effects on protein structure and
    on the reactivity of active site residues. Measuring
   
(19) the effects of pH on enzyme activity is commonly
reported in the literature. Unfortunately, much of
Equation 19 describes a rectangular hyperbola what is reported is of limited value. Measurement
which intersects the y-axis at KIA. of the rate (or activity) at one substrate concentra-
tion over a range of pH values is not very informa-
Since the two types of ternary complex mecha-
tive. Although this experiment commonly gives
nism share a common rate equation, they cannot
rise to a bell-shaped curve and permits the identi-
be distinguished by the dependence of the initial
fication of an “optimum pH”, the effects observed
rate on substrate concentration. One way to diag-
often combine denaturation, structural changes and
nose the type of mechanism occurring is through
alterations in the reactivity of key active site resi-
studying the effects of product inhibition [32-35].
dues. Furthermore, alteration of the substrate con-
Specifically, the type of inhibition (i.e. competi-
centration may well alter the precise shape of the
tive, uncompetitive or mixed) of both C and D
curve or change the “optimum pH”.
must be determined. In the random mechanism,
both C and D will be competitive inhibitors with More information rich experiments are those in
respect to both substrates. However, in the ordered which the effect of pH on individual kinetic pa-
mechanism, where A is the first substrate to bind rameters is measured; the most useful parameters
and C is the first product to be released, C will be to study are the turnover number, kcat and the spec-
a mixed inhibitor with respect to A and B. In con- ificity constant, kcat/Km. Careful measurements of
trast D will be a competitive inhibitor with respect these effects can reveal some information about
to A, but show mixed inhibition with respect to B the residues involved in binding and catalysis. As-
[33, 35]. Enzymes which have substituted enzyme suming that there is no denaturation or other gross
or ordered ternary complex mechanisms may also structural changes, these experiments can be used
exhibit the phenomenon of substrate inhibition. to estimate the pKa values of key groups in the ac-
High concentrations of the second substrate to tive site. In effect a titration experiment is being
bind may act as inhibitors with respect to the first carried out and the ideal result is a sigmoidal curve
substrate [32, 36]. in which the x-value at the inflexion point(s) cor-
Quantitative Enzymology Current Enzyme Inhibition, 2015, Vol. 11, No. 1 25

responds to the pKa of the functional group being dues can be difficult or even impossible. Most pro-
titrated; if there are two of these and they are well- teins contain many groups with similar pKa values
separated then the result will be a bell-shaped (e.g. asparate, glutamate and the C-terminus all
curve (Fig. 9). Since denaturation gives rise to a have similar values) and, anyway, the pKa of a res-
similar shaped curve it is important to provide ex- idue is highly sensitive to its local environment,
perimental evidence that this is not occurring. One including other nearby amino acid residues. In-
simple test is to observe the solution and note the deed, the active sites of some enzymes are struc-
presence or absence of any precipitates resulting tured so that the pKa of catalytically important res-
from pH-dependent denaturation. Fluorescence or idues are displaced substantially for the value
circular dichroism spectra taken at pH values measured for the same amino acid in solution. Fur-
across the range tested should be similar indicating thermore, in some cases the substrate can also be
no major structural changes. For pH values which protonated and deprotonated over the pH range
result in very low rates, adjustment of the pH to being tested and, therefore, the observed effects
more optimal values should rapidly restore the may reflect changes to the substrate rather than the
rate. enzyme. ATP (and other nucleotide phosphates) is
one important group of molecules which alter their
In the ideal case, the effect of hydrogen ion ionisation state in the pH range 1-14 [43].
concentration on enzyme activity can be described
as:
EFFECTS OF TEMPERATURE ON EN-
  ZYME ACTIVITY
    
(20)
 
  
The effects of temperature on enzyme activity
(Ka1 and Ka2 are the two acid dissociation con- are another area where many published experi-
stants; kcat’’ is the value of kcat determined at a giv- ments are not very information rich. Again the is-
en pH value and kcat,lim is the limiting value of kcat) sue is confused by the various effects that tem-
Equation 19 assumes that there are two independ- perature can have on enzyme structure and func-
ent ionisable groups and that no denaturation oc- tion. In general terms, increasing the temperature
curs in the pH range studied [42, 43]. increases the amount of energy being supplied to a
reaction which tends to increase the rate. However,
Even if the experiment is done carefully and protein molecules are often only marginally stable
there is no evidence of denaturation, care is still at physiological temperatures and so, when raised
required in the interpretation of the results. Under significantly above these, tend to denature. How-
these circumstances, the pKa values obtained will ever, careful measurement of temperature-
correspond to important groups in the catalytic dependent effects on activity within the range
process. However, assigning these to specific resi- where only catalysis (and not overall structure) is

Fig. (9). The effects of pH on enzyme activity. A typical, ideal pKa curve in which there are two ionisable groups (pKa1=4.5
and pKa1=8.5) and no denaturation or substrate ionisation effects. The line was fitted to equation 20.
26 Current Enzyme Inhibition, 2015, Vol. 11, No. 1 David J. Timson

affected can reveal information about the activa- (a)


tion energy of the enzyme-catalysed reaction.
Since enzymes function by reducing the activation
energy of the reaction, this is a valuable parameter
to know. It can particularly useful in helping to
rationalise why an enzyme is more efficient at cat-
alysing reactions with one substrate compared to
another.
In principle, enzyme-catalysed reactions can be
considered using the Arrhenius equation:
 
      (21) (b)
(A is the Arrhenius constant; Ea is the activation
energy; R is the molar gas constant; T the absolute
temperature). Here, kcat’ is the kcat measured at any
given temperature and is often the most useful
constant to subject to this treatment since it reports
on the rate-determining step which is often the
chemistry of the enzyme catalysed reaction. Val-
ues for the constants can be extracted using a line-
arised form of the equation:
 
         (21a)
Fig. (10). The effects of temperature on enzyme activity.
Therefore, a plot of the logarithm of kcat’ (a) A typical, idealised Arrhenius plot showing the depend-
against 1/T will yield a straight line with negative ence of kcat on temperature assuming no denaturation or oth-
slope (Fig. 10). The gradient will be –Ea/R (and er gross structural changes occur in the temperature range
the y-intercept will be the logarithm of A). studied. Data were simulated using Equation 20 with the
parameters A=10,000 s-1 and Ea=20 kJmol-1. (b) Linearisa-
COOPERATIVITY tion of the data shown in (a) according to Equation 21a.

Not all enzymes conform to the Michaelis- Cooperative behaviour by enzymes results in
Menten equation. One common reason for this is them not having a rectangular hyperbolic relation-
the phenomenon of cooperativity. This was first ship between initial rate and substrate concentra-
well-characterised in the non-enzymatic protein tion. A modified form of the Michaelis-Menten
haemoglobin where it was observed that the bind- equation, the Hill equation [44] can be used as an
ing of oxygen molecules to the four binding sites empirical description of their kinetic behaviour:
became progressively easier as the sites were filled  
[44]. In other words, binding of oxygen at one site    (22)

in some way affects the other sites to increase their
affinity for the molecule. The same process can be In Equation 22, h is the Hill coefficient and K0.5
observed in many (but not all) enzymes which is analogous to Km in the Michaelis-Menten equa-
have more than one functional active site in the tion. However, unlike Km is cannot be represented
same protein. Cooperativity requires that there is as a composite of several discrete rate constants
some form of “communication” between the active and should simply be considered as the substrate
sites. In many cases, this is mediated in part by concentration required to give an initial rate equal
conformational changes which are transmitted to half the maximal rate. The Hill coefficient must
through the protein subunits. In recent years it has be greater than zero and does not have to be an
become increasingly clear that altered protein mo- integer. If it is greater than one, the enzyme is said
tions can also be used to transmit information to be positively cooperative and if it is less than
through enzymes [45]. one, the enzyme exhibits negative cooperativity. If
Quantitative Enzymology Current Enzyme Inhibition, 2015, Vol. 11, No. 1 27

h=1 then Equation 22 simplifies to Equation 1. In more “graded” and a change in substrate concen-
a positively cooperative enzyme, the Hill coeffi- tration will result in a smaller change in rate than
cient cannot exceed the number of binding sites in a non-cooperative enzyme (Fig. 11c). One role
for the substrate in the enzyme; for negatively co- of negatively cooperative enzymes may be to
operative enzymes h cannot be less than the recip- “buffer” critical systems against changes in sub-
rocal of the number of binding sites [46]. strate concentrations [47].
The dependence of initial rate on substrate con- Since both forms of cooperativity can be de-
centration differs between the two types of coop- scribed by Equation 22, this can be used to deter-
erativity. In positive cooperativity, the graph has a mine the Hill coefficient. Ideally, non-linear curve
sigmoidal shape which gets steeper as h increases fitting should be used; however the presence of
(Fig. 11a). This shape of graph clearly illustrates one constant as an exponent can lead to some dif-
one of the effects of positive cooperativity which ficulties (since small changes in the estimated val-
is to convert enzymes to a more “on/off switch- ue of h can have dramatic consequences for other
like” response. This is especially the case for high- values). It can take several rounds of fitting to
er values of h where there is a range of substrate produce acceptable results. An alternative method
concentrations with negligible activity, a small involves a linearised form of Equation 22:
transition zone (close to where [S] equals K0.5) and 
a region where the rate approaches Vmax (Fig. 11a).         (22a)
 
Negative cooperativity gives rise to a graph which,
superficially, looks more similar to the Michaelis- Using this form of the Hill equation, a graph of
Menten graph (Fig. 11c). However, the enzyme’s log10 (v/ (Vmax-v) against log10[S] will be a straight
response to increasing substrate concentration is line with gradient equal to h and a y-intercept of –

(a) (b)

(c) (d)

Fig. (11). Cooperativity (a) Positive cooperativity (solid line; data simulated using Equation 22 with K0.5=30 μM, Vmax=100
μMs-1 and h=2) generates a switch-like response when compared to Michaelis-Menten kinetics (dashed line; data simulated
with Km=30 μM and Vmax=100 μMs-1). The two dotted lines from the y-axis indicate 20% and 80% maximal activity. Note how
the corresponding distance on the x-axis (substrate concentration range) is much smaller for the positively cooperative case.
(b) The data from (a) linearised according to Equation 22a. (c) Negative cooperativity (solid line; data simulated using Equa-
tion 21 with K0.5=30 μM, Vmax=100 μMs-1 and h=0.5) generates a graded response when compared to Michaelis-Menten kinet-
ics (dashed line; data simulated with Km=30 μM and Vmax=100 μMs-1). The two dotted lines from the y-axis indicate 20% and
80% maximal activity. Note how the corresponding distance on the x-axis (substrate concentration range) is much larger for
the negatively cooperative case. (d) The data from (c) linearised according to Equation 22a and using similar axes to (b) to
enable comparison.
28 Current Enzyme Inhibition, 2015, Vol. 11, No. 1 David J. Timson

log10K0.5 (Fig. 11b,d). One strategy is to use the and medicinal chemistry students at Queen’s Uni-
linearised form to obtain an estimate of h which is versity, Belfast (UK).
then used as an initial value in non-linear curve
fitting. Cooperativity can also occur with inhibi- REFERENCES
tors. In such cases a modified form of Equation
22a can be used [48]: [1] Michaelis, L.; Menten, M.L. Kinetics of invertase
action. Biochem. Z., 1913, 49, 333-369.
 [2] Michaelis, L.; Menten, M.L.; Johnson, K.A.; Goody,
        (22b)
  R.S. The original Michaelis constant: translation of
the 1913 Michaelis-Menten paper. Biochemistry,
(v0 is the initial rate in the absence of inhibitor and 2011, 50, 8264-8269.
KI is a constant the exact meaning of which will [3] Briggs, G.E.; Haldane, J.B. A Note on the Kinetics of
vary depending on the type of inhibition present). Enzyme Action. Biochem. J., 1925, 19, 338-339.
[4] Brown, A.J. Enzyme action. J. Chem. Soc. Trans.,
CONCLUSIONS AND FINAL COMMENTS 1902, 81, 373-388.
[5] Timson, D.J.; Reece, R.J. Identification and charac-
The careful, quantitative analysis of enzyme terisation of human aldose 1-epimerase. FEBS Lett.,
2003, 543, 21-24.
catalysed reactions can yield useful information [6] Lineweaver, H.; Burk, D. The Determination of En-
about the process of catalysis. It is also a vital part zyme Dissociation Constants. J. Am. Chem. Soc.,
of drug discovery and other processes which re- 1934, 56, 658-666.
quire the identification and characterization of en- [7] Lineweaver, H.; Burk, D.; Deming, W.E. The Disso-
zyme inhibitors. It is important to remember that ciation Constant of Nitrogen-Nitrogenase in Azoto-
enzyme kinetics analysis is a tool to aid under- bacter. J. Am. Chem. Soc., 1934, 56, 225-230.
standing and that the aim is not solely the collec- [8] Tseng, S.J.; Hsu, J.P. A comparison of the parameter
estimating procedures for the Michaelis-Menten
tion of ever-increasing numbers of rate constants.
model. J. Theor. Biol., 1990, 145, 457-464.
A dose of realism is often required too. Constraints [9] Dowd, J.E.; Riggs, D.S. A Comparison of Estimates
(especially limited amounts of enzyme and limited of Michaelis-Menten Kinetic Constants from various
solubility of substrates) often restrict what can be Linear Transformations. J. Biol. Chem., 1965, 240,
done and impose compromises between what 863-869.
would be ideal from a quantitative enzymology [10] Atkins, G.L.; Nimmo, I.A. A comparison of seven
point of view and what is practically possible. De- methods for fitting the Michaelis-Menten equation.
spite these constraints, quantitative enzymology Biochem. J., 1975, 149, 775-777.
[11] Ochs, R.S. The Problem with Double Reciprocal
has been a key part of biochemistry for over 100 Plots. Curr. Enz. Inhib., 2010, 6, 164-169.
years. The robustness and usefulness of these [12] Hanes, C.S. Studies on plant amylases: The effect of
methods means that they are unlikely to become starch concentration upon the velocity of hydrolysis
obsolete and they will continue to make a contri- by the amylase of germinated barley. Biochem. J.,
bution for many years to come – especially when 1932, 26, 1406-1421.
combined with more modern approaches such as [13] Hofstee, B.H. Non-inverted versus inverted plots in
molecular biology and systems biology. enzyme kinetics. Nature, 1959, 184, 1296-1298.
[14] Eadie, G.S. The inhibition of cholinesterase by phy-
sostigmine and prostigmine. J. Biol. Chem., 1942,
CONFLICT OF INTEREST 146, 85-93.
[15] Hofstee, B.H. Specificity of esterases. I. Identification
The author confirms that this article content has of two pancreatic aliesterases. J. Biol. Chem., 1952,
no conflict of interest. 199, 357-364.
[16] Marquardt, D. An algorithm for least squares estima-
ACKNOWLEDGEMENTS tion of nonlinear parameters. SIAM J. Appl. Math.,
1963, 11, 431-441.
My interest in enzymes was largely stimulated [17] Cray, J.A.; Russell, J.T.; Timson, D.J.; Singhal, R.S.;
by two excellent lecturers at the University of Hallsworth, J.E. A universal measure of chaotropicity
Birmingham (UK) – Dr. Eva Hyde and Prof. Chris and kosmotropicity. Environ. Microbiol., 2013,15(1):
287-96. .
Wharton. My continuing interest has resulted from [18] Botting, R.M. Vane's discovery of the mechanism of
several research projects and from teaching quanti- action of aspirin changed our understanding of its
tative enzymology to second year biochemistry clinical pharmacology. Pharmacol. Rep., 2010, 62,
518-525.
Quantitative Enzymology Current Enzyme Inhibition, 2015, Vol. 11, No. 1 29

[19] Vane, J.R.; Botting, R.M. The mechanism of action of [37] Gulbinsky, J.S.; Cleland, W.W. Kinetic studies of
aspirin. Thromb. Res., 2003, 110, 255-258. Escherichia coli galactokinase. Biochemistry, 1968,
[20] Roth, G.J.; Majerus, P.W. The mechanism of the ef- 7, 566-575.
fect of aspirin on human platelets. I. Acetylation of a [38] Foglietti, M.J.; Percheron, F. [Purification and mech-
particulate fraction protein. J. Clin. Invest., 1975, 56, anism of action of a plant galactokinase]. Biochimie,
624-632. 1976, 58, 499-504.
[21] Blobaum, A.L.; Marnett, L.J. Structural and function- [39] Walker, D.G.; Khan, H.H. Some properties of galac-
al basis of cyclooxygenase inhibition. J. Med. Chem., tokinase in developing rat liver. Biochem. J., 1968,
2007, 50, 1425-1441. 108, 169-175.
[22] Cornish-Bowden, A. Fundamentals of Enzyme Kinet- [40] Ballard, F.J. Kinetic studies with liver galactokinase.
ics, Portland Press: London, 2004. Biochem. J., 1966, 101, 70-75.
[23] Dixon, M. The determination of enzyme inhibitor [41] Timson, D.J.; Reece, R.J. Kinetic analysis of yeast
constants. Biochem. J., 1953, 55, 170-171. galactokinase: implications for transcriptional activa-
[24] Schlamowitz, M.; Shaw, A.; Jackson, W.T. Limita- tion of the GAL genes. Biochemistry, 2002, 84, 265-
tions of the Dixon plot for ascertaining nature of en- 272.
zyme inhibition. Tex. Rep. Biol. Med., 1969, 27, 483- [42] Dixon, M. The effect of pH on the affinities of en-
488. zymes for substrates and inhibitors. Biochem. J.,
[25] Kakkar, T.; Boxenbaum, H.; Mayersohn, M. Estima- 1953, 55, 161-170.
tion of Ki in a competitive enzyme-inhibition model: [43] Tipton, K.F.; Dixon, H.B. Effects of pH on enzymes.
comparisons among three methods of data analysis. Methods Enzymol., 1979, 63, 183-234.
Drug Metab. Dispos., 1999, 27, 756-762. [44] Hill, A.V. The possible effects of the aggregation of
[26] Cornish-Bowden, A. A simple graphical method for molecules of haemoglobin on its dissociation curve.
determining the inhibition constants of mixed, un- J. Physiol. (Lond)., 1910, 40, 4-7.
competitive and non-competitive inhibitors. Biochem. [45] Goodey, N.M.; Benkovic, S.J. Allosteric regulation
J., 1974, 137, 143-144. and catalysis emerge via a common route. Nat. Chem.
[27] Cheng, Y.; Prusoff, W.H. Relationship between the Biol., 2008, 4, 474-482.
inhibition constant (KI) and the concentration of in- [46] Abeliovich, H. An empirical extremum principle for
hibitor which causes 50 per cent inhibition (I50) of an the hill coefficient in ligand-protein interactions
enzymatic reaction. Biochem. Pharmacol., 1973, 22, showing negative cooperativity. Biophys. J., 2005, 89,
3099-3108. 76-79.
[28] Alberty, R.A. The Effect of Enzyme Concentration on [47] Ferrell, J.E.,Jr Q&A: Cooperativity. J. Biol., 2009, 8,
the Apparent Equilibrium Constant for an Enzyme- 53.
catalyzed Reaction. J. Am. Chem. Soc., 1953, 75, [48] Engel, P.C. In Chapter 3: Enzyme Kinetics, Anony-
1925-1928. mous Bio Scientific Publishers: Oxford, UK, 1996.
[29] Cleland, W.W. The kinetics of enzyme-catalyzed [49] Thompson, A.; Taylor, B.N.Guide for the use of the
reactions with two or more substrates or products. II. international system of units (SI), NIST:
Inhibition: nomenclature and theory. Biochim. Bio- Gaithersburg, MD, 2008.
phys. Acta, 1963, 67, 173-187. [50] Ning, J.; Purich, D.L.; Fromm, H.J. Studies on the
[30] Cleland, W.W. The kinetics of enzyme-catalyzed kinetic mechanism and allosteric nature of bovine
reactions with two or more substrates or products. I. brain hexokinase. J. Biol. Chem., 1969, 244, 3840-
Nomenclature and rate equations. Biochim. Biophys. 3846.
Acta, 1963, 67, 104-137. [51] Berger, S.A.; Evans, P.R. Active-site mutants altering
[31] Fromm, H.J. Summary of kinetic reaction mecha- the cooperativity of E. coli phosphofructokinase. Na-
nisms. Methods Enzymol., 1979, 63, 42-53. ture, 1990, 343, 575-576.
[32] Cleland, W.W. The kinetics of enzyme-catalyzed [52] Krebs, H.A.; Eggleston, L.V. The oxidation of py-
reactions with two or more substrates or products: III. ruvate in pigeon breast muscle. Biochem. J., 1940, 34,
Prediction of initial velocity and inhibition patterns 442-459.
by inspection. Biochim. Biophys. Acta, 1963, 67, 188- [53] Dey, P.M. Galactokinase of Vicia faba seeds. Eur. J.
196. Biochem., 1983, 136, 155-159.
[33] Cooper, B.F.; Rudolph, F.B. Product inhibition appli- [54] Beebe, J.A.; Arabshahi, A.; Clifton, J.G.; Ringe, D.;
cations. Methods Enzymol., 1995, 249, 188-211. Petsko, G.A.; Frey, P.A. Galactose mutarotase: pH
[34] Alberty, R.A. On the Determination of Rate Con- dependence of enzymatic mutarotation. Biochemistry,
stants for Coenzyme Mechanisms1. J. Am. Chem. 2003, 42, 4414-4420.
Soc., 1958, 80, 1777-1782. [55] Wong, L.J.; Frey, P.A. Galactose 1-phosphate uri-
[35] Rudolph, F.B. Product inhibition and abortive com- dylyltransferase. Isolation of a uridylyl-enzyme in-
plex formation. Methods Enzymol., 1979, 63, 411- termediate. J. Biol. Chem., 1974, 249, 2322-2324.
436. [56] Wong, L.J.; Frey, P.A. Galactose-1-phosphate uri-
[36] Cleland, W.W. Substrate inhibition. Methods Enzy- dylyltransferase: rate studies confirming a uridylyl-
mol., 1979, 63, 500-513.
30 Current Enzyme Inhibition, 2015, Vol. 11, No. 1 David J. Timson

enzyme intermediate on the catalytic pathway. Bio- [72] Schulman, M.D.; Ostlind, D.A.; Valentino, D. Mech-
chemistry, 1974, 13, 3889-3894. anism of action of MK-401 against Fasciola hepati-
[57] McCorvie, T.J.; Timson, D.J. The structural and mo- ca: inhibition of phosphoglycerate kinase. Mol. Bio-
lecular biology of type I galactosemia: Enzymology chem. Parasitol., 1982, 5, 133-145.
of galactose 1-phosphate uridylyltransferase. IUBMB [73] Turko, I.V.; Ballard, S.A.; Francis, S.H.; Corbin, J.D.
Life, 2011, 63, 694-700. Inhibition of cyclic GMP-binding cyclic GMP-
[58] McCorvie, T.J.; Timson, D.J. Structural and molecu- specific phosphodiesterase (Type 5) by sildenafil and
lar biology of type I galactosemia: disease-associated related compounds. Mol. Pharmacol., 1999, 56, 124-
mutations. IUBMB Life, 2011, 63, 949-954. 130.
[59] Lajin, B.; Alachkar, A. The NQO1 polymorphism [74] Moreland, R.B.; Goldstein, I.; Traish, A. Sildenafil, a
C609T (Pro187Ser) and cancer susceptibility: a com- novel inhibitor of phosphodiesterase type 5 in human
prehensive meta-analysis. Br. J. Cancer, 2013, 109, corpus cavernosum smooth muscle cells. Life Sci.,
1325-1337. 1998, 62, PL 309-18.
[60] Hosoda, S.; Nakamura, W.; Hayashi, K. Properties [75] Davies, B.R.; Logie, A.; McKay, J.S.; Martin, P.;
and reaction mechanism of DT diaphorase from rat Steele, S.; Jenkins, R.; Cockerill, M.; Cartlidge, S.;
liver. J. Biol. Chem., 1974, 249, 6416-6423. Smith, P.D. AZD6244 (ARRY-142886), a potent in-
[61] Mannucci, P.M.; Franchini, M. Emerging drugs for hibitor of mitogen-activated protein ki-
hemophilia B. Exp. Opin. Emerg. Drugs, 2014, 19, nase/extracellular signal-regulated kinase kinase 1/2
407-414. kinases: mechanism of action in vivo, pharmacokinet-
[62] Kaufman, R.J.; Powell, J.S. Molecular approaches for ic/pharmacodynamic relationship, and potential for
improved clotting factors for hemophilia. Blood, combination in preclinical models. Mol. Cancer.
2013, 122, 3568-3574. Ther., 2007, 6, 2209-2219.
[63] Menting, K.W.; Claassen, J.A. beta-secretase inhibi- [76] Patel, S.P.; Kim, K.B. Selumetinib (AZD6244; AR-
tor; a promising novel therapeutic drug in Alzheimer's RY-142886) in the treatment of metastatic melanoma.
disease. Front. Aging Neurosci., 2014, 6, 165. Expert Opin. Investig. Drugs, 2012, 21, 531-539.
[64] Kandalepas, P.C.; Vassar, R. The normal and patho- [77] Guo, Z.; Ma, S. Recent advances in the discovery of
logic roles of the Alzheimer's beta-secretase, BACE1. metallo--lactamase inhibitors for beta-lactam antibi-
Curr. Alzheimer Res., 2014, 11, 441-449. otic-resistant reversing agents. Curr. Drug Targets,
[65] Nguyen-Van-Tam, J.S.; Venkatesan, S.; Muthuri, 2014, 15, 689-702.
S.G.; Myles, P.R. Neuraminidase inhibitors: who, [78] Chen, J.; Shang, X.; Hu, F.; Lao, X.; Gao, X.; Zheng,
when, where? Clin. Microbiol. Infect., 2015, 21, 222- H.; Yao, W. -Lactamase inhibitors: an update. Mini
225. Rev. Med. Chem., 2013, 13, 1846-1861.
[66] Cheng, C.K.; Tsai, C.H.; Shie, J.J.; Fang, J.M. From [79] Tilvawala, R.; Pratt, R.F. Covalent inhibition of ser-
neuraminidase inhibitors to conjugates: a step towards ine -lactamases by novel hydroxamic acid deriva-
better anti-influenza drugs? Future Med. Chem., tives. Biochemistry, 2013, 52, 3712-3720.
2014, 6, 757-774. [80] Simm, A.M.; Loveridge, E.J.; Crosby, J.; Avison,
[67] Burnham, A.J.; Baranovich, T.; Govorkova, E.A. M.B.; Walsh, T.R.; Bennett, P.M. Bulgecin A: a nov-
Neuraminidase inhibitors for influenza B virus infec- el inhibitor of binuclear metallo--lactamases. Bio-
tion: efficacy and resistance. Antiviral Res., 2013, chem. J., 2005, 387, 585-590.
100, 520-534. [81] Canas, A.I.; Camarero, S. Laccases and their natural
[68] Air, G.M. Influenza neuraminidase. Influenza Other mediators: biotechnological tools for sustainable eco-
Respir. Viruses, 2012, 6, 245-256. friendly processes. Biotechnol. Adv., 2010, 28, 694-
[69] Hata, K.; Koseki, K.; Yamaguchi, K.; Moriya, S.; 705.
Suzuki, Y.; Yingsakmongkon, S.; Hirai, G.; Sodeoka, [82] Jaiswal, N.; Pandey, V.P.; Dwivedi, U.N. Purification
M.; von Itzstein, M.; Miyagi, T. Limited inhibitory of a thermostable alkaline laccase from papaya (Cari-
effects of oseltamivir and zanamivir on human sial- ca papaya) using affinity chromatography. Int. J. Bi-
idases. Antimicrob. Agents Chemother., 2008, 52, ol. Macromol., 2015, 72, 326-332.
3484-3491. [83] Koudelka, G.B.; Hansen, F.B.; Ettinger, M.J. Solvent
[70] Kati, W.M.; Montgomery, D.; Carrick, R.; Gubareva, isotope effects and the pH dependence of laccase ac-
L.; Maring, C.; McDaniel, K.; Steffy, K.; Molla, A.; tivity under steady-state conditions. J. Biol. Chem.,
Hayden, F.; Kempf, D.; Kohlbrenner, W. In vitro 1985, 260, 15561-15565.
characterization of A-315675, a highly potent inhibi- [84] Ferri, S.; Kojima, K.; Sode, K. Review of glucose
tor of A and B strain influenza virus neuraminidases oxidases and glucose dehydrogenases: a bird's eye
and influenza virus replication. Antimicrob. Agents view of glucose sensing enzymes. J. Diabetes Sci.
Chemother., 2002, 46, 1014-1021. Technol., 2011, 5, 1068-1076.
[71] Gierse, J.K.; Koboldt, C.M.; Walker, M.C.; Seibert, [85] Birrane, G.; Bhyravbhatla, B.; Navia, M.A. Synthesis
K.; Isakson, P.C. Kinetic basis for selective inhibition of Aspartame by Thermolysin: An X-ray Structural
of cyclo-oxygenases. Biochem. J., 1999, 339 ( Pt 3), Study. ACS Med. Chem. Lett., 2014, 5, 706-710.
607-614.
Quantitative Enzymology Current Enzyme Inhibition, 2015, Vol. 11, No. 1 31

[86] Yang, C.P.; Su, C.S. Synthesis of aspartame precur- [89] Wang, M.; Li, Z.; Fang, X.; Wang, L.; Qu, Y. Cellu-
sor: alpha-L-aspartyl-L-phenylalanine methyl ester in lolytic enzyme production and enzymatic hydrolysis
ethyl acetate using thermolysin entrapped in polyure- for second-generation bioethanol production. Adv.
thane. Biotechnol. Bioeng., 1988, 32, 595-603. Biochem. Eng. Biotechnol., 2012, 128, 1-24.
[87] Voordouw, G.; Milo, C.; Roche, R.S. Role of bound [90] Li, Y.; Qi, B.; Wan, Y. Inhibitory effect of vanillin on
calcium ions in thermostable, proteolytic enzymes. cellulase activity in hydrolysis of cellulosic biomass.
Separation of intrinsic and calcium ion contributions Bioresour. Technol., 2014, 167, 324-330.
to the kinetic thermal stability. Biochemistry, 1976, [91] Oliva-Taravilla, A.; Tomas-Pejo, E.; Demuez, M.;
15, 3716-3724. Gonzalez-Fernandez, C.; Ballesteros, M. Inhibition of
[88] Dindar, B.; Karakus, E.; Abasiyanik, F. New urea cellulose enzymatic hydrolysis by laccase-derived
biosensor based on urease enzyme obtained from compounds from phenols. Biotechnol. Prog., 2015, In
Helycobacter pylori. Appl. Biochem. Biotechnol., press.
2011, 165, 1308-1321.

Received: March 11, 2015 Revised: May 07, 2015 Accepted: May 08, 2015

View publication stats

Anda mungkin juga menyukai