You are on page 1of 23

9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

Enzyme kinetics
Enzyme kinetics is the study of the rates of enzyme-
catalysed chemical reactions. In enzyme kinetics, the
reaction rate is measured and the effects of varying the
conditions of the reaction are investigated. Studying
an enzyme's kinetics in this way can reveal the
catalytic mechanism of this enzyme, its role in
metabolism, how its activity is controlled, and how a
drug or a modifier (inhibitor or activator) might affect
the rate.

An enzyme (E) is typically a protein molecule that


promotes a reaction of another molecule, its substrate
(S). This binds to the active site of the enzyme to
produce an enzyme-substrate complex ES, and is
transformed into an enzyme-product complex EP and
from there to product P, via a transition state ES*. The
series of steps is known as the mechanism:

E + S ⇄ ES ⇄ ES* ⇄ EP ⇄ E + P

This example assumes the simplest case of a reaction


with one substrate and one product. Such cases exist:
for example a mutase such as phosphoglucomutase Dihydrofolate reductase from E. coli with its two
catalyses the transfer of a phospho group from one substrates dihydrofolate (right) and NADPH (left),
position to another, and isomerase is a more general bound in the active site. The protein is shown as a
term for an enzyme that catalyses any one-substrate ribbon diagram, with alpha helices in red, beta
one-product reaction, such as triosephosphate sheathes in yellow and loops in blue. Generated
isomerase. However, such enzymes are not very from 7DFR (http://www.rcsb.org/pdb/explore.do?s
common, and are heavily outnumbered by enzymes tructureId=7DFR).
that catalyse two-substrate two-product reactions:
these include, for example, the NAD-dependent
dehydrogenases such as alcohol dehydrogenase, which catalyses the oxidation of ethanol by NAD+.
Reactions with three or four substrates or products are less common, but they exist. There is no
necessity for the number of products to be equal to the number of substrates; for example,
glyceraldehyde 3-phosphate dehydrogenase has three substrates and two products.

When enzymes bind multiple substrates, such as dihydrofolate reductase (shown right), enzyme
kinetics can also show the sequence in which these substrates bind and the sequence in which
products are released. An example of enzymes that bind a single substrate and release multiple
products are proteases, which cleave one protein substrate into two polypeptide products. Others join
two substrates together, such as DNA polymerase linking a nucleotide to DNA. Although these
mechanisms are often a complex series of steps, there is typically one rate-determining step that
determines the overall kinetics. This rate-determining step may be a chemical reaction or a
conformational change of the enzyme or substrates, such as those involved in the release of product(s)
from the enzyme.
https://en.wikipedia.org/wiki/Enzyme_kinetics 1/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

Knowledge of the enzyme's structure is helpful in interpreting kinetic data. For example, the structure
can suggest how substrates and products bind during catalysis; what changes occur during the
reaction; and even the role of particular amino acid residues in the mechanism. Some enzymes change
shape significantly during the mechanism; in such cases, it is helpful to determine the enzyme
structure with and without bound substrate analogues that do not undergo the enzymatic reaction.

Not all biological catalysts are protein enzymes: RNA-based catalysts such as ribozymes and
ribosomes are essential to many cellular functions, such as RNA splicing and translation. The main
difference between ribozymes and enzymes is that RNA catalysts are composed of nucleotides,
whereas enzymes are composed of amino acids. Ribozymes also perform a more limited set of
reactions, although their reaction mechanisms and kinetics can be analysed and classified by the same
methods.

Contents
General principles
Enzyme assays
Single-substrate reactions
Michaelis–Menten kinetics
Direct use of the Michaelis–Menten equation for time course kinetic analysis
Linear plots of the Michaelis–Menten equation
Practical significance of kinetic constants
Michaelis–Menten kinetics with intermediate
Multi-substrate reactions
Ternary-complex mechanisms
Ping–pong mechanisms
Reversible catalysis and the Haldane equation
Non-Michaelis–Menten kinetics
Pre-steady-state kinetics
Chemical mechanism
Enzyme inhibition and activation
Reversible inhibitors
Irreversible inhibitors
Philosophical discourse on reversibility and irreversibility of inhibition
Mechanisms of catalysis
History
Software
See also
Footnotes
References
Further reading
External links

https://en.wikipedia.org/wiki/Enzyme_kinetics 2/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

General principles
The reaction catalysed by an enzyme uses exactly the
same reactants and produces exactly the same
products as the uncatalysed reaction. Like other
catalysts, enzymes do not alter the position of
equilibrium between substrates and products.[1]
However, unlike uncatalysed chemical reactions,
enzyme-catalysed reactions display saturation
kinetics.[2] For a given enzyme concentration and for
relatively low substrate concentrations, the reaction
rate increases linearly with substrate concentration; As larger amounts of substrate are added to a
the enzyme molecules are largely free to catalyse the reaction, the available enzyme binding sites
reaction, and increasing substrate concentration become filled to the limit of . Beyond this limit
means an increasing rate at which the enzyme and the enzyme is saturated with substrate and the
substrate molecules encounter one another. However, reaction rate ceases to increase.
at relatively high substrate concentrations, the
reaction rate asymptotically approaches the
theoretical maximum; the enzyme active sites are almost all occupied by substrates resulting in
saturation, and the reaction rate is determined by the intrinsic turnover rate of the enzyme.[3] The
substrate concentration midway between these two limiting cases is denoted by KM. Thus, KM is the
substrate concentration at which the reaction velocity is half of the maximum velocity.[3]

The two most important kinetic properties of an enzyme are how easily the enzyme becomes
saturated with a particular substrate, and the maximum rate it can achieve. Knowing these properties
suggests what an enzyme might do in the cell and can show how the enzyme will respond to changes
in these conditions.

Enzyme assays
Enzyme assays are laboratory procedures that measure the
rate of enzyme reactions. Since enzymes are not consumed by
the reactions they catalyse, enzyme assays usually follow
changes in the concentration of either substrates or products
to measure the rate of reaction. There are many methods of
measurement. Spectrophotometric assays observe change in
the absorbance of light between products and reactants;
radiometric assays involve the incorporation or release of
radioactivity to measure the amount of product made over
time. Spectrophotometric assays are most convenient since
they allow the rate of the reaction to be measured
Progress curve for an enzyme reaction.
The slope in the initial rate period is the
continuously. Although radiometric assays require the
initial rate of reaction v. The Michaelis–
removal and counting of samples (i.e., they are discontinuous
Menten equation describes how this assays) they are usually extremely sensitive and can measure
slope varies with the concentration of very low levels of enzyme activity.[4] An analogous approach
substrate. is to use mass spectrometry to monitor the incorporation or
release of stable isotopes as substrate is converted into
product. Occasionally, an assay fails and approaches are
essential to resurrect a failed assay.

https://en.wikipedia.org/wiki/Enzyme_kinetics 3/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

The most sensitive enzyme assays use lasers focused through a microscope to observe changes in
single enzyme molecules as they catalyse their reactions. These measurements either use changes in
the fluorescence of cofactors during an enzyme's reaction mechanism, or of fluorescent dyes added
onto specific sites of the protein to report movements that occur during catalysis.[5] These studies are
providing a new view of the kinetics and dynamics of single enzymes, as opposed to traditional
enzyme kinetics, which observes the average behaviour of populations of millions of enzyme
molecules.[6][7]

An example progress curve for an enzyme assay is shown above. The enzyme produces product at an
initial rate that is approximately linear for a short period after the start of the reaction. As the reaction
proceeds and substrate is consumed, the rate continuously slows (so long as substrate is not still at
saturating levels). To measure the initial (and maximal) rate, enzyme assays are typically carried out
while the reaction has progressed only a few percent towards total completion. The length of the
initial rate period depends on the assay conditions and can range from milliseconds to hours.
However, equipment for rapidly mixing liquids allows fast kinetic measurements on initial rates of
less than one second.[8] These very rapid assays are essential for measuring pre-steady-state kinetics,
which are discussed below.

Most enzyme kinetics studies concentrate on this initial, approximately linear part of enzyme
reactions. However, it is also possible to measure the complete reaction curve and fit this data to a
non-linear rate equation. This way of measuring enzyme reactions is called progress-curve analysis.[9]
This approach is useful as an alternative to rapid kinetics when the initial rate is too fast to measure
accurately.

Single-substrate reactions
Enzymes with single-substrate mechanisms include isomerases such as triosephosphateisomerase or
bisphosphoglycerate mutase, intramolecular lyases such as adenylate cyclase and the hammerhead
ribozyme, an RNA lyase.[10] However, some enzymes that only have a single substrate do not fall into
this category of mechanisms. Catalase is an example of this, as the enzyme reacts with a first molecule
of hydrogen peroxide substrate, becomes oxidised and is then reduced by a second molecule of
substrate. Although a single substrate is involved, the existence of a modified enzyme intermediate
means that the mechanism of catalase is actually a ping–pong mechanism, a type of mechanism that
is discussed in the Multi-substrate reactions section below.

Michaelis–Menten kinetics

As enzyme-catalysed reactions are saturable, their rate of catalysis does not show a linear response to
increasing substrate. If the initial rate of the reaction is measured over a range of substrate
concentrations (denoted as [S]), the initial reaction rate ( ) increases as [S] increases, as shown on
the right. However, as [S] gets higher, the enzyme becomes saturated with substrate and the initial
rate reaches Vmax, the enzyme's maximum rate.

The Michaelis–Menten kinetic model of a single-substrate reaction is shown on the right. There is an
initial bimolecular reaction between the enzyme E and substrate S to form the enzyme–substrate
complex ES. The rate of enzymatic reaction increases with the increase of the substrate concentration
up to a certain level called Vmax; at Vmax, increase in substrate concentration does not cause any
increase in reaction rate as there is no more enzyme (E) available for reacting with substrate (S).
Here, the rate of reaction becomes dependent on the ES complex and the reaction becomes a
unimolecular reaction with an order of zero. Though the enzymatic mechanism for the unimolecular
https://en.wikipedia.org/wiki/Enzyme_kinetics 4/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

reaction can be quite complex, there


is typically one rate-determining enzymatic step
that allows this reaction to be modelled as a single
catalytic step with an apparent unimolecular rate
constant kcat.
If the reaction path proceeds over
one or several intermediates, kcat will be a function
of several elementary rate constants, whereas in A chemical reaction mechanism with or without
enzyme catalysis. The enzyme (E) binds substrate (S)
the simplest case of a single elementary reaction
to produce product (P).
(e.g. no intermediates) it will be identical to the
elementary unimolecular rate constant k2. The
apparent unimolecular rate constant kcat is also
called turnover number and denotes the
maximum number of enzymatic reactions
catalysed per second.

The Michaelis–Menten equation[11] describes how


the (initial) reaction rate v0 depends on the Saturation curve for an enzyme reaction showing the
position of the substrate-binding equilibrium and relation between the substrate concentration and
the rate constant k2. reaction rate.

    (Michaelis–Menten

equation)

with the constants

This Michaelis–Menten equation is the basis for most single-substrate enzyme kinetics.[12] Two
crucial assumptions underlie this equation (apart from the general assumption about the mechanism
only involving no intermediate or product inhibition, and there is no allostericity or cooperativity).
The first assumption is the so-called quasi-steady-state assumption (or pseudo-steady-state
hypothesis), namely that the concentration of the substrate-bound enzyme (and hence also the
unbound enzyme) changes much more slowly than those of the product and substrate and thus the
change over time of the complex can be set to zero
. The second assumption is that the
total enzyme concentration does not change over time, thus .
A complete
derivation can be found here.

The Michaelis constant KM is experimentally defined as the concentration at which the rate of the
enzyme reaction is half Vmax, which can be verified by substituting [S] = KM into the Michaelis–
Menten equation and can also be seen graphically. If the rate-determining enzymatic step is slow
compared to substrate dissociation ( ), the Michaelis constant KM is roughly the dissociation
constant KD of the ES complex.

If is small compared to then the term and also very little ES


complex is formed, thus . Therefore, the rate of product formation is
https://en.wikipedia.org/wiki/Enzyme_kinetics 5/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

Thus the product formation rate depends on the enzyme concentration as well as on the substrate
concentration, the equation resembles a bimolecular reaction with a corresponding pseudo-second
order rate constant . This constant is a measure of catalytic efficiency. The most efficient
enzymes reach a in the range of 108 – 1010  M−1  s−1. These enzymes are so efficient they
effectively catalyse a reaction each time they encounter a substrate molecule and have thus reached an
upper theoretical limit for efficiency (diffusion limit); and are sometimes referred to as kinetically
perfect enzymes.[13] But most enzymes are far from perfect: the average values of and are
about and , respectively.[14]

Direct use of the Michaelis–Menten equation for time course kinetic analysis

The observed velocities predicted by the Michaelis–Menten equation can be used to directly model
the time course disappearance of substrate and the production of product through incorporation of
the Michaelis–Menten equation into the equation for first order chemical kinetics. This can only be
achieved however if one recognises the problem associated with the use of Euler's number in the
description of first order chemical kinetics. i.e. e−k is a split constant that introduces a systematic
error into calculations and can be rewritten as a single constant which represents the remaining
substrate after each time period.[15]

In 1983 Stuart Beal (and also independently Santiago Schnell and Claudio Mendoza in 1997) derived a
closed form solution for the time course kinetics analysis of the Michaelis-Menten mechanism.[16][17]
The solution, known as the Schnell-Mendoza equation, has the form:

where W[ ] is the Lambert-W function.[18][19] and where F(t) is

This equation is encompassed by the equation below, obtained by Berberan-Santos,[20] which is also
valid when the initial substrate concentration is close to that of enzyme,

where W[ ] is again the Lambert-W function.

https://en.wikipedia.org/wiki/Enzyme_kinetics 6/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

Linear plots of the Michaelis–Menten equation

The plot of v versus [S] above is not linear;


although initially linear at low [S], it bends
over to saturate at high [S]. Before the modern
era of nonlinear curve-fitting on computers,
this nonlinearity could make it difficult to
estimate KM and Vmax accurately. Therefore,
several researchers developed linearisations of
the Michaelis–Menten equation, such as the
Lineweaver–Burk plot, the Eadie–Hofstee
diagram and the Hanes–Woolf plot. All of
these linear representations can be useful for
visualising data, but none should be used to
determine kinetic parameters, as computer Lineweaver–Burk or double-reciprocal plot of kinetic data,
software is readily available that allows for showing the significance of the axis intercepts and
more accurate determination by nonlinear gradient.
regression methods.[21]

The Lineweaver–Burk plot or double reciprocal plot is a common way of illustrating kinetic data. This
is produced by taking the reciprocal of both sides of the Michaelis–Menten equation. As shown on the
right, this is a linear form of the Michaelis–Menten equation and produces a straight line with the
equation y = mx + c with a y-intercept equivalent to 1/Vmax and an x-intercept of the graph
representing −1/KM.

Naturally, no experimental values can be taken at negative 1/[S]; the lower limiting value 1/[S] = 0
(the y-intercept) corresponds to an infinite substrate concentration, where 1/v=1/Vmax as shown at
the right; thus, the x-intercept is an extrapolation of the experimental data taken at positive
concentrations. More generally, the Lineweaver–Burk plot skews the importance of measurements
taken at low substrate concentrations and, thus, can yield inaccurate estimates of Vmax and KM.[22] A
more accurate linear plotting method is the Eadie–Hofstee plot. In this case, v is plotted against
v/[S]. In the third common linear representation, the Hanes–Woolf plot, [S]/v is plotted against [S].
In general, data normalisation can help diminish the amount of experimental work and can increase
the reliability of the output, and is suitable for both graphical and numerical analysis.[23]

Practical significance of kinetic constants

The study of enzyme kinetics is important for two basic reasons. Firstly, it helps explain how enzymes
work, and secondly, it helps predict how enzymes behave in living organisms. The kinetic constants
defined above, KM and Vmax, are critical to attempts to understand how enzymes work together to
control metabolism.

Making these predictions is not trivial, even for simple systems. For example, oxaloacetate is formed
by malate dehydrogenase within the mitochondrion. Oxaloacetate can then be consumed by citrate
synthase, phosphoenolpyruvate carboxykinase or aspartate aminotransferase, feeding into the citric
acid cycle, gluconeogenesis or aspartic acid biosynthesis, respectively. Being able to predict how much

https://en.wikipedia.org/wiki/Enzyme_kinetics 7/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

oxaloacetate goes into which pathway requires knowledge of the concentration of oxaloacetate as well
as the concentration and kinetics of each of these enzymes. This aim of predicting the behaviour of
metabolic pathways reaches its most complex expression in the synthesis of huge amounts of kinetic
and gene expression data into mathematical models of entire organisms. Alternatively, one useful
simplification of the metabolic modelling problem is to ignore the underlying enzyme kinetics and
only rely on information about the reaction network's stoichiometry, a technique called flux balance
analysis.[24][25]

Michaelis–Menten kinetics with intermediate

One could also consider the less simple case

where a complex with the enzyme and an intermediate exists and the intermediate is converted into
product in a second step. In this case we have a very similar equation[26]

but the constants are different

We see that for the limiting case , thus when the last step from is much faster
than the previous step, we get again the original equation. Mathematically we have then
and .

Multi-substrate reactions
Multi-substrate reactions follow complex rate equations that describe how the substrates bind and in
what sequence. The analysis of these reactions is much simpler if the concentration of substrate A is
kept constant and substrate B varied. Under these conditions, the enzyme behaves just like a single-
substrate enzyme and a plot of v by [S] gives apparent KM and Vmax constants for substrate B. If a set
of these measurements is performed at different fixed concentrations of A, these data can be used to
work out what the mechanism of the reaction is. For an enzyme that takes two substrates A and B and
turns them into two products P and Q, there are two types of mechanism: ternary complex and ping–
pong.

Ternary-complex mechanisms

https://en.wikipedia.org/wiki/Enzyme_kinetics 8/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

In these enzymes, both substrates bind to the


enzyme at the same time to produce an EAB ternary
complex. The order of binding can either be random
(in a random mechanism) or substrates have to bind
in a particular sequence (in an ordered mechanism).
When a set of v by [S] curves (fixed A, varying B)
from an enzyme with a ternary-complex mechanism
are plotted in a Lineweaver–Burk plot, the set of
lines produced will intersect.
Random-order ternary-complex mechanism for an
Enzymes with ternary-complex mechanisms include enzyme reaction. The reaction path is shown as a
line and enzyme intermediates containing
glutathione S-transferase,[27] dihydrofolate
[28] substrates A and B or products P and Q are written
reductase and DNA polymerase.[29] The below the line.
following links show short animations of the ternary-
complex mechanisms of the enzymes dihydrofolate
reductase[β] and DNA polymerase[γ].

Ping–pong mechanisms

As shown on the right, enzymes with a


ping-pong mechanism can exist in two
states, E and a chemically modified Ping–pong mechanism for an enzyme reaction. Intermediates
form of the enzyme E*; this modified contain substrates A and B or products P and Q.
enzyme is known as an intermediate.
In such mechanisms, substrate A
binds, changes the enzyme to E* by, for example, transferring a chemical group to the active site, and
is then released. Only after the first substrate is released can substrate B bind and react with the
modified enzyme, regenerating the unmodified E form. When a set of v by [S] curves (fixed A, varying
B) from an enzyme with a ping–pong mechanism are plotted in a Lineweaver–Burk plot, a set of
parallel lines will be produced. This is called a secondary plot.

Enzymes with ping–pong mechanisms include some oxidoreductases such as thioredoxin


peroxidase,[30] transferases such as acylneuraminate cytidylyltransferase[31] and serine proteases
such as trypsin and chymotrypsin.[32] Serine proteases are a very common and diverse family of
enzymes, including digestive enzymes (trypsin, chymotrypsin, and elastase), several enzymes of the
blood clotting cascade and many others. In these serine proteases, the E* intermediate is an acyl-
enzyme species formed by the attack of an active site serine residue on a peptide bond in a protein
substrate. A short animation showing the mechanism of chymotrypsin is linked here.[δ]

Reversible catalysis and the Haldane equation


External factors may limit the ability of an enzyme to catalyse a reaction in both directions (whereas
the nature of a catalyst in itself means that it cannot catalyse just one direction, according to the
principle of microscopic reversibility). We consider the case of an enzyme that catalyses the reaction
in both directions:

https://en.wikipedia.org/wiki/Enzyme_kinetics 9/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

The steady-state, initial rate of the reaction is

is positive if the reaction proceed in the forward direction ( ) and negative otherwise.

Equilibrium requires that , which occurs when . This shows that

thermodynamics forces a relation between the values of the 4 rate constants.

The values of the forward and backward maximal rates, obtained for , , and ,
, respectively, are and , respectively. Their ratio is not
equal to the equilibrium constant, which implies that thermodynamics does not constrain the ratio of
the maximal rates. This explains that enzymes can be much "better catalysts" (in terms of maximal
rates) in one particular direction of the reaction.[33]

On can also derive the two Michaelis constants and .

The Haldane equation is the relation .

Therefore, thermodynamics constrains the ratio between the forward and backward
values, not the ratio of values.

Non-Michaelis–Menten kinetics
Many different enzyme systems follow non Michaelis-
Menten behavior.[34] A select few examples include
kinetics of self-catalytic enzymes, cooperative and
allosteric enzymes, interfacial and intracellular
enzymes, processive enzymes and so forth. Some
enzymes produce a sigmoid v by [S] plot, which often
indicates cooperative binding of substrate to the active
site. This means that the binding of one substrate
molecule affects the binding of subsequent substrate
molecules. This behavior is most common in
multimeric enzymes with several interacting active
sites.[35][36] Here, the mechanism of cooperation is
Saturation curve for an enzyme reaction showing similar to that of hemoglobin, with binding of
sigmoid kinetics. substrate to one active site altering the affinity of the
other active sites for substrate molecules. Positive
cooperativity occurs when binding of the first
substrate molecule increases the affinity of the other active sites for substrate. Negative cooperativity
occurs when binding of the first substrate decreases the affinity of the enzyme for other substrate
molecules.

Allosteric enzymes include mammalian tyrosyl tRNA-synthetase, which shows negative


cooperativity,[37] and bacterial aspartate transcarbamoylase[38] and phosphofructokinase,[39] which
show positive cooperativity.

https://en.wikipedia.org/wiki/Enzyme_kinetics 10/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

Cooperativity is surprisingly common and can help regulate the responses of enzymes to changes in
the concentrations of their substrates.[35] Positive cooperativity makes enzymes much more sensitive
to [S] and their activities can show large changes over a narrow range of substrate concentration.
Conversely, negative cooperativity makes enzymes insensitive to small changes in [S].

The Hill equation[40] is often used to describe the degree of cooperativity quantitatively in non-
Michaelis–Menten kinetics. The derived Hill coefficient n measures how much the binding of
substrate to one active site affects the binding of substrate to the other active sites. A Hill coefficient of
<1 indicates negative cooperativity and a coefficient of >1 indicates positive cooperativity.

Pre-steady-state kinetics
In the first moment after an enzyme is mixed with
substrate, no product has been formed and no
intermediates exist. The study of the next few
milliseconds of the reaction is called pre-steady-state
kinetics. Pre-steady-state kinetics is therefore
concerned with the formation and consumption of
enzyme–substrate intermediates (such as ES or E*)
until their steady-state concentrations are reached.

This approach was first applied to the hydrolysis


reaction catalysed by chymotrypsin.[41] Often, the
detection of an intermediate is a vital piece of evidence Pre-steady state progress curve, showing the
in investigations of what mechanism an enzyme burst phase of an enzyme reaction.
follows. For example, in the ping–pong mechanisms
that are shown above, rapid kinetic measurements can
follow the release of product P and measure the formation of the modified enzyme intermediate
E*.[42] In the case of chymotrypsin, this intermediate is formed by an attack on the substrate by the
nucleophilic serine in the active site and the formation of the acyl-enzyme intermediate.

In the figure to the right, the enzyme produces E* rapidly in the first few seconds of the reaction. The
rate then slows as steady state is reached. This rapid burst phase of the reaction measures a single
turnover of the enzyme. Consequently, the amount of product released in this burst, shown as the
intercept on the y-axis of the graph, also gives the amount of functional enzyme which is present in
the assay.[43]

Chemical mechanism
An important goal of measuring enzyme kinetics is to determine the chemical mechanism of an
enzyme reaction, i.e., the sequence of chemical steps that transform substrate into product. The
kinetic approaches discussed above will show at what rates intermediates are formed and inter-
converted, but they cannot identify exactly what these intermediates are.

Kinetic measurements taken under various solution conditions or on slightly modified enzymes or
substrates often shed light on this chemical mechanism, as they reveal the rate-determining step or
intermediates in the reaction. For example, the breaking of a covalent bond to a hydrogen atom is a
common rate-determining step. Which of the possible hydrogen transfers is rate determining can be
shown by measuring the kinetic effects of substituting each hydrogen by deuterium, its stable isotope.
The rate will change when the critical hydrogen is replaced, due to a primary kinetic isotope effect,
https://en.wikipedia.org/wiki/Enzyme_kinetics 11/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

which occurs because bonds to deuterium are harder to break than bonds to hydrogen.[44] It is also
possible to measure similar effects with other isotope substitutions, such as 13C/12C and 18O/16O, but
these effects are more subtle.[45]

Isotopes can also be used to reveal the fate of various parts of the substrate molecules in the final
products. For example, it is sometimes difficult to discern the origin of an oxygen atom in the final
product; since it may have come from water or from part of the substrate. This may be determined by
systematically substituting oxygen's stable isotope 18O into the various molecules that participate in
the reaction and checking for the isotope in the product.[46] The chemical mechanism can also be
elucidated by examining the kinetics and isotope effects under different pH conditions,[47] by altering
the metal ions or other bound cofactors,[48] by site-directed mutagenesis of conserved amino acid
residues, or by studying the behaviour of the enzyme in the presence of analogues of the
substrate(s).[49]

Enzyme inhibition and activation


Enzyme inhibitors are molecules that reduce or
abolish enzyme activity, while enzyme activators are
molecules that increase the catalytic rate of enzymes.
These interactions can be either reversible (i.e.,
removal of the inhibitor restores enzyme activity) or
irreversible (i.e., the inhibitor permanently inactivates
the enzyme).

Kinetic scheme for reversible enzyme inhibitors.


Reversible inhibitors

Traditionally reversible enzyme inhibitors have been classified as competitive, uncompetitive, or non-
competitive, according to their effects on KM and Vmax. These different effects result from the
inhibitor binding to the enzyme E, to the enzyme–substrate complex ES, or to both, respectively. The
division of these classes arises from a problem in their derivation and results in the need to use two
different binding constants for one binding event. The binding of an inhibitor and its effect on the
enzymatic activity are two distinctly different things, another problem the traditional equations fail to
acknowledge. In noncompetitive inhibition the binding of the inhibitor results in 100% inhibition of
the enzyme only, and fails to consider the possibility of anything in between.[50] In noncompetitive
inhibition, the inhibitor will bind to an enzyme at its allosteric site; therefore, the binding affinity, or
inverse of KM, of the substrate with the enzyme will remain the same. On the other hand, the Vmax
will decrease relative to an uninhibited enzyme. On a Lineweaver-Burk plot, the presence of a
noncompetitive inhibitor is illustrated by a change in the y-intercept, defined as 1/Vmax. The x-
intercept, defined as −1/KM, will remain the same. In competitive inhibition, the inhibitor will bind to
an enzyme at the active site, competing with the substrate. As a result, the KM will increase and the
Vmax will remain the same.[51] The common form of the inhibitory term also obscures the relationship
between the inhibitor binding to the enzyme and its relationship to any other binding term be it the
Michaelis–Menten equation or a dose response curve associated with ligand receptor binding. To
demonstrate the relationship the following rearrangement can be made:

https://en.wikipedia.org/wiki/Enzyme_kinetics 12/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

Adding zero to the bottom ([I]-[I])

Dividing by [I]+Ki

This notation demonstrates that similar to the Michaelis–Menten equation, where the rate of reaction
depends on the percent of the enzyme population interacting with substrate, the effect of the inhibitor
is a result of the percent of the enzyme population interacting with inhibitor. The only problem with
this equation in its present form is that it assumes absolute inhibition of the enzyme with inhibitor
binding, when in fact there can be a wide range of effects anywhere from 100% inhibition of substrate
turn over to just >0%. To account for this the equation can be easily modified to allow for different
degrees of inhibition by including a delta Vmax term.

or

This term can then define the residual enzymatic activity present when the inhibitor is interacting
with individual enzymes in the population. However the inclusion of this term has the added value of
allowing for the possibility of activation if the secondary Vmax term turns out to be higher than the
initial term. To account for the possibly of activation as well the notation can then be rewritten
replacing the inhibitor "I" with a modifier term denoted here as "X".

While this terminology results in a simplified way of dealing with kinetic effects relating to the
maximum velocity of the Michaelis–Menten equation, it highlights potential problems with the term
used to describe effects relating to the KM. The KM relating to the affinity of the enzyme for the
https://en.wikipedia.org/wiki/Enzyme_kinetics 13/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

substrate should in most cases relate to potential changes in the binding site of the enzyme which
would directly result from enzyme inhibitor interactions. As such a term similar to the one proposed
above to modulate Vmax should be appropriate in most situations:[52]

A few examples of reversible inhibition belonging to the competitive and uncompetitive models have
been discussed in the following papers.[53][54][55]

Irreversible inhibitors

Enzyme inhibitors can also irreversibly inactivate enzymes, usually by covalently modifying active site
residues. These reactions, which may be called suicide substrates, follow exponential decay functions
and are usually saturable. Below saturation, they follow first order kinetics with respect to inhibitor.
Irreversible inhibition could be classified into two distinct types. Affinity labelling is a type of
irreversible inhibition where a functional group that is highly reactive modifies a catalytically critical
residue on the protein of interest to bring about inhibition. Mechanism-based inhibition, on the other
hand, involves binding of the inhibitor followed by enzyme mediated alterations that transform the
latter into a reactive group that irreversibly modifies the enzyme.

Philosophical discourse on reversibility and irreversibility of inhibition

Having discussed reversible inhibition and irreversible inhibition in the above two headings, it would
have to be pointed out that the concept of reversibility (or irreversibility) is a purely theoretical
construct exclusively dependent on the time-frame of the assay, i.e., a reversible assay involving
association and dissociation of the inhibitor molecule in the minute timescales would seem
irreversible if an assay assess the outcome in the seconds and vice versa. There is a continuum of
inhibitor behaviors spanning reversibility and irreversibility at a given non-arbitrary assay time
frame. There are inhibitors that show slow-onset behavior[53] and most of these inhibitors, invariably,
also show tight-binding to the protein target of interest.[53][54]

Mechanisms of catalysis
The favoured model for the enzyme–substrate interaction is the induced fit model.[56] This model
proposes that the initial interaction between enzyme and substrate is relatively weak, but that these
weak interactions rapidly induce conformational changes in the enzyme that strengthen binding.
These conformational changes also bring catalytic residues in the active site close to the chemical
bonds in the substrate that will be altered in the reaction.[57] Conformational changes can be
measured using circular dichroism or dual polarisation interferometry. After binding takes place, one
or more mechanisms of catalysis lower the energy of the reaction's transition state by providing an
alternative chemical pathway for the reaction. Mechanisms of catalysis include catalysis by bond
strain; by proximity and orientation; by active-site proton donors or acceptors; covalent catalysis and
quantum tunnelling.[42][58]

Enzyme kinetics cannot prove which modes of catalysis are used by an enzyme. However, some
kinetic data can suggest possibilities to be examined by other techniques. For example, a ping–pong
mechanism with burst-phase pre-steady-state kinetics would suggest covalent catalysis might be
https://en.wikipedia.org/wiki/Enzyme_kinetics 14/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

important in this enzyme's mechanism. Alternatively,


the observation of a strong pH effect on Vmax but not
KM might indicate that a residue in the active site
needs to be in a particular ionisation state for catalysis
to occur.

History
In 1902 Victor Henri proposed a quantitative theory of
enzyme kinetics,[59] but at the time the experimental
significance of the hydrogen ion concentration was not
yet recognized. After Peter Lauritz Sørensen had
defined the logarithmic pH-scale and introduced the The energy variation as a function of reaction
concept of buffering in 1909[60] the German chemist coordinate shows the stabilisation of the transition
Leonor Michaelis and Dr. Maud Leonora Menten (a state by an enzyme.
postdoctoral researcher in Michaelis's lab at the time)
repeated Henri's experiments and confirmed his
equation, which is now generally referred to as Michaelis-Menten kinetics (sometimes also Henri-
Michaelis-Menten kinetics).[61] Their work was further developed by G. E. Briggs and J. B. S.
Haldane, who derived kinetic equations that are still widely considered today a starting point in
modeling enzymatic activity.[62]

The major contribution of the Henri-Michaelis-Menten approach was to think of enzyme reactions in
two stages. In the first, the substrate binds reversibly to the enzyme, forming the enzyme-substrate
complex. This is sometimes called the Michaelis complex. The enzyme then catalyzes the chemical
step in the reaction and releases the product. The kinetics of many enzymes is adequately described
by the simple Michaelis-Menten model, but all enzymes have internal motions that are not accounted
for in the model and can have significant contributions to the overall reaction kinetics. This can be
modeled by introducing several Michaelis-Menten pathways that are connected with fluctuating
rates,[63][64][65] which is a mathematical extension of the basic Michaelis Menten mechanism.[66]

Software
ENZO (Enzyme Kinetics) is a graphical interface tool for building kinetic models of enzyme catalyzed
reactions. ENZO automatically generates the corresponding differential equations from a stipulated
enzyme reaction scheme. These differential equations are processed by a numerical solver and a
regression algorithm which fits the coefficients of differential equations to experimentally observed
time course curves. ENZO allows rapid evaluation of rival reaction schemes and can be used for
routine tests in enzyme kinetics.[67]

See also
Protein dynamics
Diffusion limited enzyme
Langmuir adsorption model

Footnotes
https://en.wikipedia.org/wiki/Enzyme_kinetics 15/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

α. ^ Link: Interactive Michaelis–Menten kinetics tutorial (Java required) (https://web.archive.org/


web/20120616044431/http://cti.itc.virginia.edu/~cmg/Demo/scriptFrame.html)

β. ^ Link: dihydrofolate reductase mechanism (Gif) (https://web.archive.org/web/200608200622


58/http://chem-faculty.ucsd.edu/kraut/dhfr.html)

γ. ^ Link: DNA polymerase mechanism (Gif) (https://web.archive.org/web/20060514070741/htt


p://chem-faculty.ucsd.edu/kraut/dNTP.html)

δ. ^ Link: Chymotrypsin mechanism (Flash required) (https://web.archive.org/web/20070319235


224/http://courses.cm.utexas.edu/jrobertus/ch339k/overheads-2/06_21_chymotrypsin.html)

References
1. Wrighton MS, Ebbing DD (1993). General chemistry (4th ed.). Boston: Houghton Mifflin.
ISBN 978-0-395-63696-1.
2. Srinivasan, Bharath (27 September 2020). "Words of advice: teaching enzyme kinetics" (https://do
i.org/10.1111%2Ffebs.15537). The FEBS Journal. 288 (7): 2068–2083. doi:10.1111/febs.15537 (ht
tps://doi.org/10.1111%2Ffebs.15537). ISSN 1742-464X (https://www.worldcat.org/issn/1742-
464X). PMID 32981225 (https://pubmed.ncbi.nlm.nih.gov/32981225).
3. Fromm H.J., Hargrove M.S. (2012) Enzyme Kinetics. In: Essentials of Biochemistry. Springer,
Berlin, Heidelberg
4. Danson M, Eisenthal R (2002). Enzyme assays: a practical approach. Oxford [Oxfordshire]:
Oxford University Press. ISBN 978-0-19-963820-8.
5. Xie XS, Lu HP (June 1999). "Single-molecule enzymology" (https://doi.org/10.1074%2Fjbc.274.2
3.15967). The Journal of Biological Chemistry. 274 (23): 15967–70. doi:10.1074/jbc.274.23.15967
(https://doi.org/10.1074%2Fjbc.274.23.15967). PMID 10347141 (https://pubmed.ncbi.nlm.nih.gov/
10347141).
6. Lu HP (June 2004). "Single-molecule spectroscopy studies of conformational change dynamics in
enzymatic reactions". Current Pharmaceutical Biotechnology. 5 (3): 261–9.
doi:10.2174/1389201043376887 (https://doi.org/10.2174%2F1389201043376887).
PMID 15180547 (https://pubmed.ncbi.nlm.nih.gov/15180547).
7. Schnell JR, Dyson HJ, Wright PE (2004). "Structure, dynamics, and catalytic function of
dihydrofolate reductase". Annual Review of Biophysics and Biomolecular Structure. 33: 119–40.
doi:10.1146/annurev.biophys.33.110502.133613 (https://doi.org/10.1146%2Fannurev.biophys.33.1
10502.133613). PMID 15139807 (https://pubmed.ncbi.nlm.nih.gov/15139807).
8. Gibson QH (1969). "[6] Rapid mixing: Stopped flow". Rapid mixing: Stopped flow. Methods in
Enzymology. 16. pp. 187–228. doi:10.1016/S0076-6879(69)16009-7 (https://doi.org/10.1016%2F
S0076-6879%2869%2916009-7). ISBN 978-0-12-181873-9.
9. Duggleby RG (1995). "[3] Analysis of enzyme progress curves by nonlinear regression". Analysis
of enzyme progress curves by non-linear regression. Methods in Enzymology. 249. pp. 61–90.
doi:10.1016/0076-6879(95)49031-0 (https://doi.org/10.1016%2F0076-6879%2895%2949031-0).
ISBN 978-0-12-182150-0. PMID 7791628 (https://pubmed.ncbi.nlm.nih.gov/7791628).
10. Murray JB, Dunham CM, Scott WG (January 2002). "A pH-dependent conformational change,
rather than the chemical step, appears to be rate-limiting in the hammerhead ribozyme cleavage
reaction". Journal of Molecular Biology. 315 (2): 121–30. doi:10.1006/jmbi.2001.5145 (https://doi.o
rg/10.1006%2Fjmbi.2001.5145). PMID 11779233 (https://pubmed.ncbi.nlm.nih.gov/11779233).
S2CID 18102624 (https://api.semanticscholar.org/CorpusID:18102624).
11. Michaelis L. and Menten M.L. Kinetik der Invertinwirkung Biochem. Z. 1913; 49:333–369 English
translation (http://web.lemoyne.edu/~giunta/menten.html) Accessed 6 April 2007

https://en.wikipedia.org/wiki/Enzyme_kinetics 16/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

12. Srinivasan, Bharath (2021). "A Guide to the Michaelis-Menten equation: Steady state and
beyond" (https://doi.org/10.1111%2Ffebs.16124). The FEBS Journal. n/a (n/a).
doi:10.1111/febs.16124 (https://doi.org/10.1111%2Ffebs.16124). ISSN 1742-4658 (https://www.wo
rldcat.org/issn/1742-4658). PMID 34270860 (https://pubmed.ncbi.nlm.nih.gov/34270860).
13. Stroppolo ME, Falconi M, Caccuri AM, Desideri A (September 2001). "Superefficient enzymes".
Cellular and Molecular Life Sciences. 58 (10): 1451–60. doi:10.1007/PL00000788 (https://doi.org/
10.1007%2FPL00000788). PMID 11693526 (https://pubmed.ncbi.nlm.nih.gov/11693526).
S2CID 24874575 (https://api.semanticscholar.org/CorpusID:24874575).
14. Bar-Even A, Noor E, Savir Y, Liebermeister W, Davidi D, Tawfik DS, Milo R (May 2011). "The
moderately efficient enzyme: evolutionary and physicochemical trends shaping enzyme
parameters". Biochemistry. 50 (21): 4402–10. doi:10.1021/bi2002289 (https://doi.org/10.1021%2F
bi2002289). PMID 21506553 (https://pubmed.ncbi.nlm.nih.gov/21506553).
15. Walsh R, Martin E, Darvesh S (January 2010). "A method to describe enzyme-catalyzed reactions
by combining steady state and time course enzyme kinetic parameters". Biochimica et Biophysica
Acta (BBA) - General Subjects. 1800 (1): 1–5. doi:10.1016/j.bbagen.2009.10.007 (https://doi.org/1
0.1016%2Fj.bbagen.2009.10.007). PMID 19840832 (https://pubmed.ncbi.nlm.nih.gov/19840832).
16. Beal SL (December 1983). "Computation of the explicit solution to the Michaelis-Menten
equation". Journal of Pharmacokinetics and Biopharmaceutics. 11 (6): 641–57.
doi:10.1007/BF01059062 (https://doi.org/10.1007%2FBF01059062). PMID 6689584 (https://pubm
ed.ncbi.nlm.nih.gov/6689584). S2CID 32571415 (https://api.semanticscholar.org/CorpusID:32571
415).
17. Schnell S, Mendoza C (1997). "Closed Form Solution for Time-dependent Enzyme Kinetics".
Journal of Theoretical Biology. 187 (2): 207–212. Bibcode:1997JThBi.187..207S (https://ui.adsab
s.harvard.edu/abs/1997JThBi.187..207S). doi:10.1006/jtbi.1997.0425 (https://doi.org/10.1006%2F
jtbi.1997.0425).
18. Goudar CT, Sonnad JR, Duggleby RG (January 1999). "Parameter estimation using a direct
solution of the integrated Michaelis-Menten equation" (https://www.webcitation.org/6cuXxfqIj?url=
http://rdbiotech.drivehq.com/Biblio/Pubs/Pub103.pdf) (PDF). Biochimica et Biophysica Acta (BBA)
- Protein Structure and Molecular Enzymology. 1429 (2): 377–83. doi:10.1016/s0167-
4838(98)00247-7 (https://doi.org/10.1016%2Fs0167-4838%2898%2900247-7). PMID 9989222 (ht
tps://pubmed.ncbi.nlm.nih.gov/9989222). Archived from the original (http://rdbiotech.drivehq.com/
Biblio/Pubs/Pub103.pdf) (PDF) on 9 November 2015.
19. Goudar CT, Harris SK, McInerney MJ, Suflita JM (December 2004). "Progress curve analysis for
enzyme and microbial kinetic reactions using explicit solutions based on the Lambert W function".
Journal of Microbiological Methods. 59 (3): 317–26. doi:10.1016/j.mimet.2004.06.013 (https://doi.o
rg/10.1016%2Fj.mimet.2004.06.013). PMID 15488275 (https://pubmed.ncbi.nlm.nih.gov/1548827
5).
20. Berberan-Santos MN (2010). "A General Treatment of Henri Michaelis Menten Enzyme Kinetics:
Exact Series Solution and Approximate Analytical Solutions" (http://match.pmf.kg.ac.rs/electronic_
versions/Match63/n2/match63n2_283-318.pdf) (PDF). MATCH Communications in Mathematical
and in Computer Chemistry. 63: 283.
21. Jones ME (December 1992). "Analysis of algebraic weighted least-squares estimators for enzyme
parameters" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1132043). The Biochemical Journal.
288 (Pt 2): 533–8. doi:10.1042/bj2880533 (https://doi.org/10.1042%2Fbj2880533). PMC 1132043
(https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1132043). PMID 1463456 (https://pubmed.ncbi.nl
m.nih.gov/1463456).
22. Tseng SJ, Hsu JP (August 1990). "A comparison of the parameter estimating procedures for the
Michaelis-Menten model". Journal of Theoretical Biology. 145 (4): 457–64.
Bibcode:1990JThBi.145..457T (https://ui.adsabs.harvard.edu/abs/1990JThBi.145..457T).
doi:10.1016/S0022-5193(05)80481-3 (https://doi.org/10.1016%2FS0022-5193%2805%2980481-
3). PMID 2246896 (https://pubmed.ncbi.nlm.nih.gov/2246896).

https://en.wikipedia.org/wiki/Enzyme_kinetics 17/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

23. Bravo IG, Busto F, De Arriaga D, Ferrero MA, Rodríguez-Aparicio LB, Martínez-Blanco H, Reglero
A (September 2001). "A normalized plot as a novel and time-saving tool in complex enzyme
kinetic analysis" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1222113). The Biochemical
Journal. 358 (Pt 3): 573–83. doi:10.1042/bj3580573 (https://doi.org/10.1042%2Fbj3580573).
PMC 1222113 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1222113). PMID 11577687 (https://
pubmed.ncbi.nlm.nih.gov/11577687).
24. Almaas E, Kovács B, Vicsek T, Oltvai ZN, Barabási AL (February 2004). "Global organization of
metabolic fluxes in the bacterium Escherichia coli". Nature. 427 (6977): 839–43. arXiv:q-
bio/0403001 (https://arxiv.org/abs/q-bio/0403001). Bibcode:2004Natur.427..839A (https://ui.adsab
s.harvard.edu/abs/2004Natur.427..839A). doi:10.1038/nature02289 (https://doi.org/10.1038%2Fn
ature02289). PMID 14985762 (https://pubmed.ncbi.nlm.nih.gov/14985762). S2CID 715721 (http
s://api.semanticscholar.org/CorpusID:715721).
25. Reed JL, Vo TD, Schilling CH, Palsson BO (2003). "An expanded genome-scale model of
Escherichia coli K-12 (iJR904 GSM/GPR)" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC19365
4). Genome Biology. 4 (9): R54. doi:10.1186/gb-2003-4-9-r54 (https://doi.org/10.1186%2Fgb-2003
-4-9-r54). PMC 193654 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC193654). PMID 12952533
(https://pubmed.ncbi.nlm.nih.gov/12952533).
26. for a complete derivation, see here
27. Dirr H, Reinemer P, Huber R (March 1994). "X-ray crystal structures of cytosolic glutathione S-
transferases. Implications for protein architecture, substrate recognition and catalytic function" (htt
ps://doi.org/10.1111%2Fj.1432-1033.1994.tb18666.x). European Journal of Biochemistry. 220 (3):
645–61. doi:10.1111/j.1432-1033.1994.tb18666.x (https://doi.org/10.1111%2Fj.1432-1033.1994.tb
18666.x). PMID 8143720 (https://pubmed.ncbi.nlm.nih.gov/8143720).
28. Stone SR, Morrison JF (July 1988). "Dihydrofolate reductase from Escherichia coli: the kinetic
mechanism with NADPH and reduced acetylpyridine adenine dinucleotide phosphate as
substrates". Biochemistry. 27 (15): 5493–9. doi:10.1021/bi00415a016 (https://doi.org/10.1021%2F
bi00415a016). PMID 3052577 (https://pubmed.ncbi.nlm.nih.gov/3052577).
29. Fisher PA (1994). Enzymologic mechanism of replicative DNA polymerases in higher eukaryotes
(https://archive.org/details/progressinnuclei0000unse/page/371). Progress in Nucleic Acid
Research and Molecular Biology. 47. pp. 371–97 (https://archive.org/details/progressinnuclei0000
unse/page/371). doi:10.1016/S0079-6603(08)60257-3 (https://doi.org/10.1016%2FS0079-6603%
2808%2960257-3). ISBN 978-0-12-540047-3. PMID 8016325 (https://pubmed.ncbi.nlm.nih.gov/80
16325).
30. Akerman SE, Müller S (August 2003). "2-Cys peroxiredoxin PfTrx-Px1 is involved in the
antioxidant defence of Plasmodium falciparum". Molecular and Biochemical Parasitology. 130 (2):
75–81. doi:10.1016/S0166-6851(03)00161-0 (https://doi.org/10.1016%2FS0166-6851%2803%29
00161-0). PMID 12946843 (https://pubmed.ncbi.nlm.nih.gov/12946843).
31. Bravo IG, Barrallo S, Ferrero MA, Rodríguez-Aparicio LB, Martínez-Blanco H, Reglero A
(September 2001). "Kinetic properties of the acylneuraminate cytidylyltransferase from
Pasteurella haemolytica A2" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1222114). The
Biochemical Journal. 358 (Pt 3): 585–98. doi:10.1042/bj3580585 (https://doi.org/10.1042%2Fbj35
80585). PMC 1222114 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1222114). PMID 11577688
(https://pubmed.ncbi.nlm.nih.gov/11577688).
32. Kraut J (1977). "Serine proteases: structure and mechanism of catalysis". Annual Review of
Biochemistry. 46: 331–58. doi:10.1146/annurev.bi.46.070177.001555 (https://doi.org/10.1146%2F
annurev.bi.46.070177.001555). PMID 332063 (https://pubmed.ncbi.nlm.nih.gov/332063).
33. Cornish-Bowden A (2012). Fundamentals of Enzyme Kinetics. Wiley-Blackwell. "Some enzymes
are much more effective catalysts for one direction than the other. As a striking example, the
limiting rates of the forward reaction catalyzed by methionine adenosyltransferase is about 105
greater than that for the reverse direction, even though the equilibrium constant is close to unity
(page 59)."

https://en.wikipedia.org/wiki/Enzyme_kinetics 18/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

34. Srinivasan, Bharath (2021). "Explicit Treatment of Non Michaelis-Menten and Atypical Kinetics in
Early Drug Discovery". ChemMedChem. 16 (6): 899–918. doi:10.1002/cmdc.202000791 (https://d
oi.org/10.1002%2Fcmdc.202000791). PMID 33231926 (https://pubmed.ncbi.nlm.nih.gov/3323192
6). S2CID 227157473 (https://api.semanticscholar.org/CorpusID:227157473).
35. Srinivasan, Bharath; Forouhar, Farhad; Shukla, Arpit; Sampangi, Chethana; Kulkarni, Sonia;
Abashidze, Mariam; Seetharaman, Jayaraman; Lew, Scott; Mao, Lei; Acton, Thomas B.; Xiao,
Rong (March 2014). "Allosteric regulation and substrate activation in cytosolic nucleotidase II from
Legionella pneumophila" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3982195). FEBS
Journal. 281 (6): 1613–1628. doi:10.1111/febs.12727 (https://doi.org/10.1111%2Ffebs.12727).
PMC 3982195 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3982195). PMID 24456211 (https://
pubmed.ncbi.nlm.nih.gov/24456211).
36. Ricard J, Cornish-Bowden A (July 1987). "Co-operative and allosteric enzymes: 20 years on" (http
s://doi.org/10.1111%2Fj.1432-1033.1987.tb13510.x). European Journal of Biochemistry. 166 (2):
255–72. doi:10.1111/j.1432-1033.1987.tb13510.x (https://doi.org/10.1111%2Fj.1432-1033.1987.tb
13510.x). PMID 3301336 (https://pubmed.ncbi.nlm.nih.gov/3301336).
37. Ward WH, Fersht AR (July 1988). "Tyrosyl-tRNA synthetase acts as an asymmetric dimer in
charging tRNA. A rationale for half-of-the-sites activity". Biochemistry. 27 (15): 5525–30.
doi:10.1021/bi00415a021 (https://doi.org/10.1021%2Fbi00415a021). PMID 3179266 (https://pubm
ed.ncbi.nlm.nih.gov/3179266).
38. Helmstaedt K, Krappmann S, Braus GH (September 2001). "Allosteric regulation of catalytic
activity: Escherichia coli aspartate transcarbamoylase versus yeast chorismate mutase" (https://w
ww.ncbi.nlm.nih.gov/pmc/articles/PMC99034). Microbiology and Molecular Biology Reviews. 65
(3): 404–21, table of contents. doi:10.1128/MMBR.65.3.404-421.2001 (https://doi.org/10.1128%2F
MMBR.65.3.404-421.2001). PMC 99034 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC99034).
PMID 11528003 (https://pubmed.ncbi.nlm.nih.gov/11528003).
39. Schirmer T, Evans PR (January 1990). "Structural basis of the allosteric behaviour of
phosphofructokinase". Nature. 343 (6254): 140–5. Bibcode:1990Natur.343..140S (https://ui.adsab
s.harvard.edu/abs/1990Natur.343..140S). doi:10.1038/343140a0 (https://doi.org/10.1038%2F343
140a0). PMID 2136935 (https://pubmed.ncbi.nlm.nih.gov/2136935). S2CID 4272821 (https://api.s
emanticscholar.org/CorpusID:4272821).
40. Hill AV (1910). "The possible effects of the aggregation of the molecules of haemoglobin on its
dissociation curves". J. Physiol. 40: iv–vii.
41. Hartley BS, Kilby BA (February 1954). "The reaction of p-nitrophenyl esters with chymotrypsin and
insulin" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1269615). The Biochemical Journal. 56
(2): 288–97. doi:10.1042/bj0560288 (https://doi.org/10.1042%2Fbj0560288). PMC 1269615 (http
s://www.ncbi.nlm.nih.gov/pmc/articles/PMC1269615). PMID 13140189 (https://pubmed.ncbi.nlm.ni
h.gov/13140189).
42. Fersht, Alan (1999). Structure and mechanism in protein science: a guide to enzyme catalysis and
protein folding. San Francisco: W.H. Freeman. ISBN 978-0-7167-3268-6.
43. Bender ML, Begué-Cantón ML, Blakeley RL, Brubacher LJ, Feder J, Gunter CR, Kézdy FJ,
Killheffer JV, Marshall TH, Miller CG, Roeske RW, Stoops JK (December 1966). "The
determination of the concentration of hydrolytic enzyme solutions: alpha-chymotrypsin, trypsin,
papain, elastase, subtilisin, and acetylcholinesterase". Journal of the American Chemical Society.
88 (24): 5890–913. doi:10.1021/ja00976a034 (https://doi.org/10.1021%2Fja00976a034).
PMID 5980876 (https://pubmed.ncbi.nlm.nih.gov/5980876).
44. Cleland WW (January 2005). "The use of isotope effects to determine enzyme mechanisms".
Archives of Biochemistry and Biophysics. 433 (1): 2–12. doi:10.1016/j.abb.2004.08.027 (https://do
i.org/10.1016%2Fj.abb.2004.08.027). PMID 15581561 (https://pubmed.ncbi.nlm.nih.gov/1558156
1).

https://en.wikipedia.org/wiki/Enzyme_kinetics 19/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

45. Northrop DB (1981). "The expression of isotope effects on enzyme-catalyzed reactions". Annual
Review of Biochemistry. 50: 103–31. doi:10.1146/annurev.bi.50.070181.000535 (https://doi.org/1
0.1146%2Fannurev.bi.50.070181.000535). PMID 7023356 (https://pubmed.ncbi.nlm.nih.gov/7023
356).
46. Baillie TA, Rettenmeier AW (1986). "Drug biotransformation: mechanistic studies with stable
isotopes". Journal of Clinical Pharmacology. 26 (6): 448–51. doi:10.1002/j.1552-
4604.1986.tb03556.x (https://doi.org/10.1002%2Fj.1552-4604.1986.tb03556.x). PMID 3734135 (h
ttps://pubmed.ncbi.nlm.nih.gov/3734135). S2CID 39193680 (https://api.semanticscholar.org/Corp
usID:39193680).
47. Cleland WW (1982). "Use of isotope effects to elucidate enzyme mechanisms". CRC Critical
Reviews in Biochemistry. 13 (4): 385–428. doi:10.3109/10409238209108715 (https://doi.org/10.3
109%2F10409238209108715). PMID 6759038 (https://pubmed.ncbi.nlm.nih.gov/6759038).
48. Christianson DW, Cox JD (1999). "Catalysis by metal-activated hydroxide in zinc and manganese
metalloenzymes". Annual Review of Biochemistry. 68: 33–57.
doi:10.1146/annurev.biochem.68.1.33 (https://doi.org/10.1146%2Fannurev.biochem.68.1.33).
PMID 10872443 (https://pubmed.ncbi.nlm.nih.gov/10872443).
49. Kraut DA, Carroll KS, Herschlag D (2003). "Challenges in enzyme mechanism and energetics".
Annual Review of Biochemistry. 72: 517–71. doi:10.1146/annurev.biochem.72.121801.161617 (htt
ps://doi.org/10.1146%2Fannurev.biochem.72.121801.161617). PMID 12704087 (https://pubmed.n
cbi.nlm.nih.gov/12704087).
50. Walsh R, Martin E, Darvesh S (December 2011). "Limitations of conventional inhibitor
classifications". Integrative Biology. 3 (12): 1197–201. doi:10.1039/c1ib00053e (https://doi.org/10.
1039%2Fc1ib00053e). PMID 22038120 (https://pubmed.ncbi.nlm.nih.gov/22038120).
51. Cleland WW (February 1963). "The kinetics of enzyme-catalyzed reactions with two or more
substrates or products. III. Prediction of initial velocity and inhibition patterns by inspection".
Biochimica et Biophysica Acta. 67: 188–96. doi:10.1016/0006-3002(63)91816-x (https://doi.org/1
0.1016%2F0006-3002%2863%2991816-x). PMID 14021669 (https://pubmed.ncbi.nlm.nih.gov/14
021669).
52. Walsh R, Martin E, Darvesh S (May 2007). "A versatile equation to describe reversible enzyme
inhibition and activation kinetics: modeling beta-galactosidase and butyrylcholinesterase".
Biochimica et Biophysica Acta (BBA) - General Subjects. 1770 (5): 733–46.
doi:10.1016/j.bbagen.2007.01.001 (https://doi.org/10.1016%2Fj.bbagen.2007.01.001).
PMID 17307293 (https://pubmed.ncbi.nlm.nih.gov/17307293).
53. Srinivasan B, Skolnick J (May 2015). "Insights into the slow-onset tight-binding inhibition of
Escherichia coli dihydrofolate reductase: detailed mechanistic characterization of pyrrolo [3,2-f]
quinazoline-1,3-diamine and its derivatives as novel tight-binding inhibitors" (https://www.ncbi.nlm.
nih.gov/pmc/articles/PMC4445455). The FEBS Journal. 282 (10): 1922–38.
doi:10.1111/febs.13244 (https://doi.org/10.1111%2Ffebs.13244). PMC 4445455 (https://www.ncbi.
nlm.nih.gov/pmc/articles/PMC4445455). PMID 25703118 (https://pubmed.ncbi.nlm.nih.gov/25703
118).
54. Srinivasan B, Tonddast-Navaei S, Skolnick J (October 2015). "Ligand binding studies, preliminary
structure-activity relationship and detailed mechanistic characterization of 1-phenyl-6,6-dimethyl-
1,3,5-triazine-2,4-diamine derivatives as inhibitors of Escherichia coli dihydrofolate reductase" (htt
ps://www.ncbi.nlm.nih.gov/pmc/articles/PMC4610388). European Journal of Medicinal Chemistry.
103: 600–14. doi:10.1016/j.ejmech.2015.08.021 (https://doi.org/10.1016%2Fj.ejmech.2015.08.02
1). PMC 4610388 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4610388). PMID 26414808 (htt
ps://pubmed.ncbi.nlm.nih.gov/26414808).

https://en.wikipedia.org/wiki/Enzyme_kinetics 20/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

55. Srinivasan B, Rodrigues JV, Tonddast-Navaei S, Shakhnovich E, Skolnick J (July 2017). "Rational
Design of Novel Allosteric Dihydrofolate Reductase Inhibitors Showing Antibacterial Effects on
Drug-Resistant Escherichia coli Escape Variants" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC
5819740). ACS Chemical Biology. 12 (7): 1848–1857. doi:10.1021/acschembio.7b00175 (https://d
oi.org/10.1021%2Facschembio.7b00175). PMC 5819740 (https://www.ncbi.nlm.nih.gov/pmc/articl
es/PMC5819740). PMID 28525268 (https://pubmed.ncbi.nlm.nih.gov/28525268).
56. Koshland DE (February 1958). "Application of a Theory of Enzyme Specificity to Protein
Synthesis" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC335371). Proceedings of the National
Academy of Sciences of the United States of America. 44 (2): 98–104.
Bibcode:1958PNAS...44...98K (https://ui.adsabs.harvard.edu/abs/1958PNAS...44...98K).
doi:10.1073/pnas.44.2.98 (https://doi.org/10.1073%2Fpnas.44.2.98). PMC 335371 (https://www.n
cbi.nlm.nih.gov/pmc/articles/PMC335371). PMID 16590179 (https://pubmed.ncbi.nlm.nih.gov/165
90179).
57. Hammes GG (July 2002). "Multiple conformational changes in enzyme catalysis". Biochemistry.
41 (26): 8221–8. doi:10.1021/bi0260839 (https://doi.org/10.1021%2Fbi0260839). PMID 12081470
(https://pubmed.ncbi.nlm.nih.gov/12081470).
58. Sutcliffe MJ, Scrutton NS (July 2002). "A new conceptual framework for enzyme catalysis.
Hydrogen tunnelling coupled to enzyme dynamics in flavoprotein and quinoprotein enzymes" (htt
p://content.febsjournal.org/cgi/content/full/269/13/3096). European Journal of Biochemistry. 269
(13): 3096–102. doi:10.1046/j.1432-1033.2002.03020.x (https://doi.org/10.1046%2Fj.1432-1033.2
002.03020.x). PMID 12084049 (https://pubmed.ncbi.nlm.nih.gov/12084049).
59. Henri V (1902). "Theorie generale de l'action de quelques diastases". Compt. Rend. Acad. Sci.
Paris. 135: 916–9.
60. Sørensen PL (1909). "Enzymstudien {II}. Über die Messung und Bedeutung der
Wasserstoffionenkonzentration bei enzymatischen Prozessen" [Enzyme studies III: About the
measurement and significance of the hydrogen ion concentration in enzymatic processes].
Biochem. Z. (in German). 21: 131–304.
61. Michaelis L, Menten M (1913). "Die Kinetik der Invertinwirkung" [The Kinetics of Invertase Action].
Biochem. Z. (in German). 49: 333–369.; Michaelis L, Menten ML, Johnson KA, Goody RS
(October 2011). "The original Michaelis constant: translation of the 1913 Michaelis-Menten paper"
(https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3381512). Biochemistry. 50 (39): 8264–9.
doi:10.1021/bi201284u (https://doi.org/10.1021%2Fbi201284u). PMC 3381512 (https://www.ncbi.
nlm.nih.gov/pmc/articles/PMC3381512). PMID 21888353 (https://pubmed.ncbi.nlm.nih.gov/21888
353).
62. Briggs GE, Haldane JB (1925). "A Note on the Kinetics of Enzyme Action" (https://www.ncbi.nlm.n
ih.gov/pmc/articles/PMC1259181). The Biochemical Journal. 19 (2): 338–9.
doi:10.1042/bj0190338 (https://doi.org/10.1042%2Fbj0190338). PMC 1259181 (https://www.ncbi.
nlm.nih.gov/pmc/articles/PMC1259181). PMID 16743508 (https://pubmed.ncbi.nlm.nih.gov/16743
508).
63. Flomenbom O, Velonia K, Loos D, Masuo S, Cotlet M, Engelborghs Y, Hofkens J, Rowan AE,
Nolte RJ, Van der Auweraer M, de Schryver FC, Klafter J (February 2005). "Stretched exponential
decay and correlations in the catalytic activity of fluctuating single lipase molecules" (https://www.
ncbi.nlm.nih.gov/pmc/articles/PMC548972). Proceedings of the National Academy of Sciences of
the United States of America. 102 (7): 2368–72. Bibcode:2005PNAS..102.2368F (https://ui.adsab
s.harvard.edu/abs/2005PNAS..102.2368F). doi:10.1073/pnas.0409039102 (https://doi.org/10.107
3%2Fpnas.0409039102). PMC 548972 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC548972).
PMID 15695587 (https://pubmed.ncbi.nlm.nih.gov/15695587).
64. English BP, Min W, van Oijen AM, Lee KT, Luo G, Sun H, Cherayil BJ, Kou SC, Xie XS (February
2006). "Ever-fluctuating single enzyme molecules: Michaelis-Menten equation revisited". Nature
Chemical Biology. 2 (2): 87–94. doi:10.1038/nchembio759 (https://doi.org/10.1038%2Fnchembio7
59). PMID 16415859 (https://pubmed.ncbi.nlm.nih.gov/16415859). S2CID 2201882 (https://api.se
manticscholar.org/CorpusID:2201882).
https://en.wikipedia.org/wiki/Enzyme_kinetics 21/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

65. Lu HP, Xun L, Xie XS (December 1998). "Single-molecule enzymatic dynamics". Science. 282
(5395): 1877–82. Bibcode:1998Sci...282.1877P (https://ui.adsabs.harvard.edu/abs/1998Sci...282.
1877P). doi:10.1126/science.282.5395.1877 (https://doi.org/10.1126%2Fscience.282.5395.1877).
PMID 9836635 (https://pubmed.ncbi.nlm.nih.gov/9836635).
66. Xue X, Liu F, Ou-Yang ZC (September 2006). "Single molecule Michaelis-Menten equation
beyond quasistatic disorder". Physical Review E. 74 (3 Pt 1): 030902. arXiv:cond-mat/0604364 (ht
tps://arxiv.org/abs/cond-mat/0604364). Bibcode:2006PhRvE..74c0902X (https://ui.adsabs.harvar
d.edu/abs/2006PhRvE..74c0902X). doi:10.1103/PhysRevE.74.030902 (https://doi.org/10.1103%2
FPhysRevE.74.030902). PMID 17025584 (https://pubmed.ncbi.nlm.nih.gov/17025584).
S2CID 41674948 (https://api.semanticscholar.org/CorpusID:41674948).
67. Bevc S, Konc J, Stojan J, Hodošček M, Penca M, Praprotnik M, Janežič D (2011). "ENZO: a web
tool for derivation and evaluation of kinetic models of enzyme catalyzed reactions" (https://www.nc
bi.nlm.nih.gov/pmc/articles/PMC3139599). PLOS ONE. 6 (7): e22265.
Bibcode:2011PLoSO...622265B (https://ui.adsabs.harvard.edu/abs/2011PLoSO...622265B).
doi:10.1371/journal.pone.0022265 (https://doi.org/10.1371%2Fjournal.pone.0022265).
PMC 3139599 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3139599). PMID 21818304 (https://
pubmed.ncbi.nlm.nih.gov/21818304). ENZO server (http://enzo.cmm.ki.si/)

Further reading
Introductory

Cornish-Bowden, Athel (2012). Fundamentals of enzyme kinetics (4th ed.). Weinheim: Wiley-


Blackwell. ISBN 978-3-527-33074-4.
Stevens L, Price NC (1999). Fundamentals of enzymology: the cell and molecular biology of
catalytic proteins (https://archive.org/details/fundamentalsofen0000pric_h0c7). Oxford
[Oxfordshire]: Oxford University Press. ISBN 978-0-19-850229-6.
Bugg, Tim (2004). Introduction to Enzyme and Coenzyme Chemistry. Cambridge, MA: Blackwell
Publishers. ISBN 978-1-4051-1452-3.
Segel, Irwin H. (1993). Enzyme kinetics: behavior and analysis of rapid equilibrium and steady
state enzyme systems. New York: Wiley. ISBN 978-0-471-30309-1.

Advanced

Fersht, Alan (1999). Structure and mechanism in protein science: a guide to enzyme catalysis and
protein folding. San Francisco: W.H. Freeman. ISBN 978-0-7167-3268-6.
Schnell S, Maini PK (2004). "A century of enzyme kinetics: Reliability of the KM and vmax
estimates" (https://citeseerx.ist.psu.edu/viewdoc/download;jsessionid=DA30EF5E01F31EA2CE72
C32310D20791?doi=10.1.1.493.7178&rep=rep1&type=pdf). Comments on Theoretical Biology. 8
(2–3): 169–87. CiteSeerX 10.1.1.493.7178 (https://citeseerx.ist.psu.edu/viewdoc/summary?doi=1
0.1.1.493.7178). doi:10.1080/08948550302453 (https://doi.org/10.1080%2F08948550302453).
Retrieved 22 September 2020.
Walsh, Christopher (1979). Enzymatic reaction mechanisms. San Francisco: W. H. Freeman.
ISBN 978-0-7167-0070-8.
Cleland WW, Cook P (2007). Enzyme kinetics and mechanism. New York: Garland Science.
ISBN 978-0-8153-4140-6.

External links
https://en.wikipedia.org/wiki/Enzyme_kinetics 22/23
9/13/21, 10:38 PM Enzyme kinetics - Wikipedia

Animation of an enzyme assay (http://www.kscience.co.uk/animations/model.swf) — Shows


effects of manipulating assay conditions
MACiE (http://www.ebi.ac.uk/thornton-srv/databases/MACiE/) — A database of enzyme reaction
mechanisms
ENZYME (https://web.archive.org/web/20061006064937/http://us.expasy.org/enzyme/) — Expasy
enzyme nomenclature database
ENZO (http://enzo.cmm.ki.si/) — Web application for easy construction and quick testing of kinetic
models of enzyme catalyzed reactions.
ExCatDB (http://ezcatdb.cbrc.jp/EzCatDB/) — A database of enzyme catalytic mechanisms
BRENDA (http://www.brenda-enzymes.info/) — Comprehensive enzyme database, giving
substrates, inhibitors and reaction diagrams
SABIO-RK (http://sabio.h-its.org/) — A database of reaction kinetics
Joseph Kraut's Research Group, University of California San Diego (https://web.archive.org/web/2
0060820062258/http://chem-faculty.ucsd.edu/kraut/dhfr.html) — Animations of several enzyme
reaction mechanisms
Symbolism and Terminology in Enzyme Kinetics (https://web.archive.org/web/20060712051732/ht
tp://www.chem.qmul.ac.uk/iubmb/kinetics/) — A comprehensive explanation of concepts and
terminology in enzyme kinetics
An introduction to enzyme kinetics (https://web.archive.org/web/20040612065857/http://orion1.pai
sley.ac.uk/kinetics/contents.html) — An accessible set of on-line tutorials on enzyme kinetics
Enzyme kinetics animated tutorial (http://www.wiley.com/college/pratt/0471393878/student/animati
ons/enzyme_kinetics/index.html) — An animated tutorial with audio

Retrieved from "https://en.wikipedia.org/w/index.php?title=Enzyme_kinetics&oldid=1042599196"

This page was last edited on 5 September 2021, at 20:25 (UTC).

Text is available under the Creative Commons Attribution-ShareAlike License;


additional terms may apply. By using this
site, you agree to the Terms of Use and Privacy Policy. Wikipedia® is a registered trademark of the Wikimedia
Foundation, Inc., a non-profit organization.

https://en.wikipedia.org/wiki/Enzyme_kinetics 23/23

You might also like