Chapter 4 Pharmacological Assay Formats Binding

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

Chapter 4

Pharmacological Assay Formats: Binding

The yeoman work in any science . . . is done by the experimentalist who must keep the theoreticians
honest.
— Michio Kaku (1995)

4.1 The Structure of This Chapter 4.4 Experimental Prerequisites for 4.6 Derivations
4.2 Binding Theory and Experiment Correct Application of Binding References
4.3 Complex Binding Phenomena: Techniques
Agonist Affinity from 4.5 Chapter Summary and
Binding Curves Conclusions

4.1 THE STRUCTURE OF THIS CHAPTER amount of radioactivity detected. An essential part of this
process is the ability to separate the bound from the
This chapter discusses the application of binding techni- unbound molecule. This can be done by taking advantage
ques to the study of drug-receptor interaction. It will be of the size of the protein versus the soluble small mole-
seen that the theory of binding and the methods used to cule. The protein can be separated by centrifugation, equi-
quantify drug effect are discussed before the experimental librium dialysis, or filtration. Alternatively, the physical
prerequisites for good binding experiments are given. proximity of the molecule to the protein can be used. For
This may appear to be placing the cart before the horse in example, in scintillation proximity assays, the receptor
concept. However, the methods used to detect and rectify protein adheres to a bead containing scintillant, a chemi-
non-equilibrium experimental conditions utilize the very cal that produces light when close to radioactivity. Thus,
methods used to quantify drug effect. Therefore, they when radioactive molecules are bound to the receptor
must be understood before their application to optimize (and therefore are near the scintillant) a light signal is
experimental conditions can be discussed. This chapter produced, heralding the binding of the molecule. Other
first presents what the experiments strive to achieve, and methods of detecting molecules such as fluorescence are
then explores the possible pitfalls of experimental design increasingly being utilized in binding experiments. For
that may cause the execution to fall short of the intent. example, molecules that produce different qualities of
fluorescence, depending on their proximity to protein, can
be used to quantify binding. Similarly, in fluorescence
4.2 BINDING THEORY AND EXPERIMENT polarization experiments, fluorescent ligands (when not
bound to protein) reduce the degree of light polarization
A direct measure of the binding of a molecule to a protein of light passing through the medium through free rotation.
target can be made if there is some means to distinguish When these same ligands are bound, their rotation is
the bound molecule from the unbound, and a means to reduced, thereby concomitantly reducing the effect on
quantify the amount bound. Historically, the first widely polarization. Thus, binding can be quantified in terms of
used technique to do this was radioligand binding. the degree of light polarization in the medium.
Radioactive molecules can be detected by observation of In general, there are emerging technologies available to
radioactive decay, and their amount quantified through discern bound from unbound molecules, and many of these
calibration curves relating the amount of molecule to the can be applied to receptor studies. It will be assumed from

T. P. Kenakin: A Pharmacology Primer, Fourth edition. DOI: http://dx.doi.org/10.1016/B978-0-12-407663-1.00004-1


© 2014 Elsevier Inc. All rights reserved. 63
64 Chapter | 4 Pharmacological Assay Formats: Binding

this point that the technological problems associated with the tracer may bind to non-receptor sites (i.e., adsorption
determining bound species are not an experimental factor, sites, other non-specific proteins), and if a non-radioactive
and subsequent discussions will focus on the interpretation version of the same molecular structure is used to define
of the resulting binding data. Several excellent sources specific binding, it may protect those very same non-
of information on the technology and practical aspects of specific sites (which erroneously define specific binding).
binding are available [13]. A ligand with different chemical structure may not bind to
Binding experiments can be done in three modes: the same non-specific sites and thus lessen the potential of
saturation, displacement, and kinetic. Saturation binding defining nsb sites as biologically relevant receptors.
directly observes the binding of a tracer ligand (radioac- The non-specific binding of low concentrations of
tive, fluorescent, or otherwise detectable) to the receptor. biologically active ligands is essentially linear and non-
The method quantifies the maximal number of binding saturable within the ranges used in pharmacological binding
sites and the affinity of the ligand for the site (equilibrium experiments. For a traceable ligand (radioactive, fluores-
dissociation constant of the ligand-receptor complex). cent, and so on), non-specific binding is given as:
This is a direct measure of binding using the Langmuir
nsb 5 k  ½A  (4.1)
adsorption isotherm model. A major limitation of this
technique is the obvious need for the ligand to be trace- where k is a constant defining the concentration relationship
able (i.e., it can be done only for radioactive or fluores- for non-specific binding and [A ] is the concentration of
cent molecules). Displacement studies overcome this the traceable molecule. The specific binding is saturable
limitation by allowing measurement of the affinity of and defined by the Langmuir adsorption isotherm:
non-traceable ligands through their interference with the
½A 
binding of tracer ligands. Thus, molecules are used to dis- Specific binding 5 (4.2)
place or otherwise prevent the binding of tracer ligands ½A  1 Kd
and the reduction in signal is used to quantify the affinity where Kd is the equilibrium dissociation constant of the
of the displacing ligands. Finally, kinetic studies follow ligand-receptor complex. The total binding is the sum of
the binding of a tracer ligand with time. This can yield these and is given as:
first-order rate constants for the onset and offset of bind-
ing, which can be used to calculate equilibrium binding ½A   Bmax
Total binding 5 1 k  ½A  (4.3)
constants to assess the temporal approach to equilibrium ½A  1 Kd
or to determine binding reversibility or to detect allosteric
interactions. Each of these is considered separately. The The two experimentally derived variables are nsb and
first step is to discuss some methodological points com- total binding. These can be obtained by measuring the
mon to all these types of binding experiments. relationship between the ligand concentration and the
The aim of a binding experiment is to define and amount of ligand bound (total binding) and the amount
quantify the relationship between the concentration of bound in the presence of a protecting concentration of
ligand in the receptor compartment and the portion of the receptor-specific antagonist. This latter procedure defines
concentration that is bound to the receptor at any one the nsb. Theoretically, specific binding can be obtained by
instant. A first prerequisite is to know that the amount of subtracting these values for each concentration of ligand,
bound ligand that is measured is bound only to the receptor but a more powerful method is to fit the two data sets (total
and not to other sites in the sample tube or well (i.e., cell binding and nsb) to Equations 4.1 and 4.3 simultaneously.
membrane, wall of the vessel containing the experimental One reason that this is preferable is that more data points are
solution, and so on). The amount of ligand bound to these used to define specific binding. A second reason is that a
auxiliary sites but not specifically to the target is referred better estimate of the maximal binding (Bmax) can be made
to as non-specific binding (denoted nsb). The amount by simultaneously fitting two functions. Since Bmax is
bound only to the pharmacological target of interest is defined at theoretically infinite ligand concentrations, it is
termed the specific binding. The amount of specific bind- difficult to obtain data in this concentration region. When
ing is defined operationally as the bound ligand that can be there is a paucity of data points, non-linear fitting procedures
displaced by an excess concentration of a specific antago- tend to overestimate the maximal asymptote. The additional
nist for the receptor that is not radioactive (or otherwise experimental data (total plus non-specific binding) reduces
does not interfere with the signals). Therefore, another this effect and yields more accurate Bmax estimates.
prerequisite of binding experiments is the availability of a In binding, a good first experiment is to determine
non-tracer ligand (for the specific target defined as one the time required for the binding reaction to come to
that does not interfere with the signal, whether it be radio- equilibrium with the receptor. This is essential to know,
activity, fluorescence, or polarized light). Optimally, the since most binding reactions are made in stop-time mode,
chemical structure of the ligand used to define nsb should and real-time observation of the approach to equilibrium
be different from the binding tracer ligand. This is because is not possible (this is not true of more recent fluorescent
4.2 BINDING THEORY AND EXPERIMENT 65

B FIGURE 4.1 Time course for the onset of


1.1 a radioligand onto the receptor and the
reversal of radioligand binding upon addi-
0.9
Fractional binding

tion of a high concentration of a non-


0.7 A radioactive antagonist ligand. The object
of the experiment is to determine the times
0.5 required for steady-state receptor occupation
by the radioligand and confirmation of
0.3
reversibility of binding. The radioligand is
0.1 added at point A, and an excess competitive
antagonist of the receptor at point B.
−0.1
−50 0 50 100 150 200 250 300 350 400 450
Time (min.)

techniques where visualization of binding in real time Alternatively, another method of linearizing the data
can be achieved). A useful experiment is to observe the points is by using:
approach to equilibrium of a given concentration of tracer
1 1 Kd 1
ligand and then to observe reversal of binding by addition 
5   1 (4.6)
½A R ½A  Bmax Bmax
of a competitive antagonist of the receptor. An example
of this experiment is shown in Figure 4.1. Valuable data This is referred to as a double reciprocal or lineweaver
are obtained with this approach, since it indicates the Burk plot. From this linear plot, Kd 5 slope/intercept
time needed to reach equilibrium, and confirms the fact and the 1/intercept 5 Bmax. Finally, a linear plot can be
that the binding is reversible. Reversibility is essential to achieved with:
the attainment of steady states and equilibria (i.e., irre-
½A  ½A  Kd
versible binding reactions do not come to equilibrium). 
5 1 (4.7)
½A R Bmax Bmax
This is referred to as a Hanes, HildebrandBenesi, or
Scott plot. From this linear plot, Kd 5 intercept/slope and
4.2.1 Saturation Binding 1/slope 5 Bmax.
A saturation binding experiment consists of the equilibra- Examples of these are shown for the saturation data
tion of the receptor with a range of concentrations of in Figure 4.2. At first glance, these transformations may
traceable ligand in the absence (total binding) and pres- seem like ideal methods for analyzing saturation data.
ence of a high concentration (approximately 100 3 Kd) of However, transformation of binding data is not generally
antagonist to protect the receptors (and thus determine recommended. This is because transformed plots can
the nsb). Simultaneous fitting of the total binding curve distort experimental uncertainty, produce compression of
(Equation 4.3) and nsb line (Equation 4.1) yields the specific data, and cause large differences in data placement. Also,
binding with parameters of maximal number of binding these transformations violate the assumptions of linear
sites (Bmax) and equilibrium dissociation constant of regression and can be curvilinear simply because of statis-
the ligand-receptor complex (Kd) (see Equation 4.2). An tical factors (for example, Scatchard plots combine
example of this procedure for the human calcitonin receptor dependent and independent variables). These transforma-
is shown in Figure 4.2. Before the widespread use of non- tions are valid only for ideal data and are extremely
linear fitting programs, the Langmuir equation was linear- sensitive to different types of experimental errors. They
ized for ease of fitting graphically. Thus, specific binding should not be used for estimation of binding parameters.
([A R]) according to mass action, represented as: Scatchard plots compress data to the point where a linear
plot can be obtained. Figure 4.3 shows a curve with
½A R ½A  an estimate of Bmax that falls far short of being able to
5  (4.4)
Bmax ½A  1 Kd furnish an experimental estimate of the Bmax, yet the
Scatchard plot is linear with an apparently valid estimate
yields a straight line with the transforms: from the abscissal intercept.
½A R Bmax ½A R In general, non-linear fitting of the data is essential
5 2 (4.5) for parameter estimation. Linear transformations, how-
½A  Kd Kd
ever, are useful for visualization of trends in data.
referred to alternatively as a Scatchard, Eadie, or Variances from a straight edge are more discernible to the
EadieHofstee plot. From this linear plot, Kd 5 21/slope human eye than are differences from curvilinear shapes,
and the x intercept equals Bmax. so linear transforms can be a useful diagnostic tool.
66 Chapter | 4 Pharmacological Assay Formats: Binding

0.20 Scatchard
0.16

[A*R] / [A*]
0.12

0.06

0.04

0.00
0 1 2 3 4 5 6 7 8
16 [A*R] : pM
14
14 Double recip.
12
12

(1/ [A*R]) x 1011


[A*R] : pM

10 10
8 8
6 6
4
4
2
2
0
0 0 5 10 15 20 25 30
0 50 100 150 200 250 300
1/ [A*] x 1010
[A*] : pM
35 Hanes plot
30
25
[A*] / [A*R]

20
15
10
5
0
0 50 100 150 200 250 300
[A*] : pM
FIGURE 4.2 Saturation binding. Left panel: Curves showing total binding (filled circles), non-specific binding
(filled squares), and specific binding (open circles) of the calcitonin receptor antagonist radiolabel 125I AC512
(Bmax 5 6.63 pM; Kd 5 26.8 pM). Data redrawn from [1]. Panels to the right show linear variants of the specific binding
curve: Scatchard (Equation 4.5), double reciprocal (Equation 4.6), and Hanes plots (Equation 4.7) cause distortion and
compression of data. Non-linear curve-fitting techniques are preferred.

FIGURE 4.3 Erroneous estima- 1.4 0.9


tion of maximal binding with 1.2 0.8
Scatchard plots. The saturation 0.7
Fraction max.

1.0
[A*R] / [A*]

binding curve shown to the left has 0.6


no data points available to estimate 0.8 0.5
the true Bmax. The Scatchard trans- 0.6 0.4
formation to the right linearizes the 0.4 0.3
0.2
existing points, allowing an esti- 0.2 0.1
mate of the maximum to be made 0.0 0.0
from the x-axis intercept. However, −4 −2 0 2 4 0 0.2 0.4 0.6 0.8 1 1.2
this intercept in no way estimates
Log ([A*] / Kd) [A*R] / Kd
the true Bmax since there are no
data to define this parameter.

An example of where the Scatchard transformation shows levels of binding, a common finding if slightly too much
significant deviation from a rectangular hyperbola is protein is used in the binding assay (see Section 4.4.1).
shown in Figure 4.4. The direct presentation of the While this difference is nearly undetectable when the
data shows little deviation from the saturation binding data are presented as a direct binding curve, it does
curve as defined by the Langmuir adsorption isotherm. produce a deviation from linearity in the Scatchard curve
The data at 10 and 30 nM yield slightly underestimated (see Figure 4.4B).
4.2 BINDING THEORY AND EXPERIMENT 67

30
40
25

[A*R] / [A*] x 104


Bo (pmol/mg) 20 30

15
20
10
10
5

0 0
−9 −8 −7 −6 −5 −4 0 5 10 15 20 25
(A) Log [A*] (B) [A*R] : pM
FIGURE 4.4 Saturation binding expressed directly and with a Scatchard plot. (A) Direct representation of a
saturation binding plot (Bmax 5 25 pmol/mg, Kd 5 50 nM). Data points are slightly deviated from ideal behavior
(lower two concentrations yield slightly lower values for binding, as is common when slightly too much receptor
protein is used in the assay, vide infra). (B) Scatchard plot of the data shown in panel A. It can be seen that the
slight deviations in the data lead to considerable deviations from linearity on the Scatchard plot.

0.18 FIGURE 4.5 Saturation binding


of the radioligand human 125I-
[Bound] : nM

human calcitonin to human calcito-


0.12 nin receptors in a recombinant cell
system in human embryonic kid-
0.06 ney cells. Left-hand panel shows
total binding (open circles), non-
specific binding (open squares), and
0.00 specific receptor binding (open tri-
−11 −10 −9 −8 −7 −6 angles). The specific binding
125I-hCal] appears to reach a maximal asymp-
Log [
125I-hCal totic value. The specific binding is
80 plotted on a semi-logarithmic scale
(shown in the right-hand panel). The
[Bound] : pM

60 solid line on this curve indicates an


estimate of the maximal receptor
40 binding. The data points (open cir-
cles) on this curve show that the
20 data define less than half the
computer-estimated total saturation
0 curve. Data redrawn from [4].
0 4 6 8 10
nM

Estimating the Bmax value is technically difficult a saturation binding curve. An example of how to
since it basically is an exercise in estimating an effect at measure the affinity of a radioligand and obtain an esti-
infinite drug concentration. Therefore, the accuracy of mate of Bmax (maximal number of binding sites for that
the estimate of Bmax is proportional to the maximal radioligand) is given in Section 13.1.1.
levels of radioligand that can be used in the experiment.
The attainment of saturation binding can be deceiving
when the ordinates are plotted on a linear scale, as they
are in Figure 4.2. Figure 4.5 shows a saturation curve
4.2.2 Displacement Binding
for calcitonin binding that appears to reach a maximal In practice, there will be a limited number of ligands
asymptote on a linear scale. However, replotting the available that are chemically traceable (i.e., radioactive,
graph on a semi-logarithmic scale illustrates the illusion fluorescent). Therefore, the bulk of radioligand experiments
of maximal binding on the linear scale and, in this case, designed to quantify ligand affinity are done in a displace-
how far short of true maxima a linear scale can present ment mode whereby a ligand is used to displace or otherwise
68 Chapter | 4 Pharmacological Assay Formats: Binding

affect the binding of a traceable ligand. In general, an indicates that the allosteric displacing ligand produces a
inverse sigmoidal curve is obtained with reduction in radioli- tenfold decrease in the affinity of the receptor for the
gand binding upon addition of non-radioactive antagonist. tracer ligand).
An example of how to measure the affinity of a displacing As noted previously, in all cases these various functions
ligand is given in Section 13.1.2. describe an inverse sigmoidal curve between the displacing
The equations describing the amount of bound radioli- ligand and the signal. Therefore, the mechanism of interac-
gand observed in the presence of a range of concentrations tion cannot be determined from a single displacement curve.
of non-traceable ligand vary with the model used for the However, observation of a pattern of such curves obtained
molecular antagonism. These are provided in material at different tracer ligand concentrations (range of [A ]
following, with brief descriptions. More detailed discussions values) may indicate whether the displacements are due to a
of these mechanisms can be found in Chapter 6. If the competitive, non-competitive, or allosteric mechanism.
binding is competitive (both ligands compete for the same Competitive displacement for a range of [A ] values
binding domain on the receptor), the amount of tracer (Equation 4.8) yields the pattern of curves shown in
ligand-receptor complex (ρ ) is given as (see Section 4.6.1): Figure 4.6A. A useful way to quantify the displacement is
to determine the concentration of displacing ligand that
½A =Kd
ρ 5 (4.8) produces a diminution of the signal to 50% of the original
½A =K d 1 ½B=KB 1 1 value. This concentration of displacing ligand will be
where the concentration of tracer ligand is [A ], the referred to as the IC50 (inhibitory concentration for 50%
non-traceable displacing ligand is [B], and Kd and KB are decrease). For competitive antagonists, it can be shown
respective equilibrium dissociation constants. If the binding that the IC50 is related to the concentration of tracer
is non-competitive (binding of the antagonist precludes the ligand [A ] by (see Section 4.6.4):
binding of the tracer ligand), the signal is given by (see IC50 5 KB  ð½A =Kd 1 1Þ (4.11)
Section 4.6.2):
This is a linear relation often referred to as the
 ½A =Kd ChengPrusoff relationship [5]. It is characteristic of
ρ 5  (4.9)
½A =Kd ð½B=KB 1 1Þ 1 ½B=KB 1 1 competitive ligand-receptor interactions. An example is
shown in Figure 4.6B.
If the ligand allosterically affects the affinity of the
In most conventional biochemical binding studies, the
receptor (antagonist binds to a site separate from that for the
concentration of receptor protein is well below that of the
tracer ligand) to produce a change in receptor conformation
ligands and thus the binding process does not signifi-
to affect the affinity of the tracer (vide infra) for the tracer
cantly deplete the ligands. However, there are certain pro-
ligand (see Chapter 7 for more detail), the displacement
cedures such as fluorescent binding assays which require
curve is given by (see Section 4.6.3):
high concentrations of receptor to maximize the window
½A =Kd ð1 1 α½B=KB Þ for observing a response. Under these circumstances the
ρ 5 (4.10) fluorescent probe concentration is kept below the Kd
½A =Kd ð1 1 α½B=KB Þ 1 ½B=KB 1 1
value (where Kd is the equilibrium dissociation constant
where α is the multiple factor by which the non-tracer of the fluorescent probe-receptor complex) and the recep-
ligand affects the affinity of the tracer ligand (i.e., α 5 0.1 tor concentration is maximized (above the Kd value) [6].

FIGURE 4.6 Displacement of a radioli- 1.2 12


gand by a competitive non-radioactive
ligand. (A) Displacement of radioactivity 1.0 10
(ordinate scale) as curves shown for a range 300
of concentrations of displacing ligand 100
Fraction max.

0.8 8
30
[IC50] / KB

(abscissae as log scale). Curves shown for a


range of radioligand concentrations denoted 10
0.6 6
on the graph in units of [A ]/Kd. Curved line
shows the path of the IC50 for the displace- 1
0.4 4
ment curves along the antagonist concentra-
[A*] / Kd = 0.03
tion axis. (B) Multiple values of the Ki for
0.2 2
the competitive displacing ligand (ordinate
scale) as a function of the concentration of
0.0 0
radioligand being displaced (abscissae as lin-
ear scale). Linear relationship shows the −3 −1 1 3 5 0 2 4 6 8 10
increase in observed IC50 of the antagonist (A) Log ([B] / KB) (B) [A*] / Kd
with increasing concentrations of radioligand
to be displaced (according to Equation 4.11).
4.2 BINDING THEORY AND EXPERIMENT 69

In these types of assays, the standard correction for IC50 constant of the non-competitive antagonist-receptor
to Ki values is not valid and a revised procedure utilizing complex (see Section 4.6.2).
the following equation must be used [6]: Allosteric antagonist effects can be an amalgam of
competitive and non-competitive profiles in terms of the
½I50
Ki 5 (4.12) relationship between IC50 and [A ]. This relates to the
½A  50 =Kd 1 ½R0 =Kd 1 1 magnitude of the term α, specifically the multiple ratio of
where [I]50 is the free antagonist concentration at 50% the affinity of the receptor for [A ] imposed by the binding
inhibition, Kd is the equilibrium dissociation constant of of the allosteric antagonist. A hallmark of allosteric inhibi-
the fluorescent probe-receptor complex, [A ]50 is the free tion is that it is saturable (i.e., the antagonism maximizes
concentration of fluorescent probe at 50% inhibition and upon saturation of the allosteric binding site). Therefore, if
[R]0 is the free concentration of receptor at 0% inhibition. a given antagonist has a value of α of 0.1, this means that
The practical application of this equation is discussed in the saturation binding curve will shift to the right by a fac-
detail in Section 4.6.4. tor of ten in the presence of an infinite concentration of
The displacement of a tracer ligand, for a range of allosteric antagonist. Depending on the initial concentration
tracer ligand concentrations, by a non-competitive antago- of radioligand, this may cause the displacement binding
nist is shown in Figure 4.7. In contrast to the pattern curve to fail to reach nsb levels. This effect is illustrated in
shown for competitive antagonists, the IC50 for inhibition Figure 4.8. Therefore, in contrast to competitive antago-
of tracer binding does not change with increasing tracer nists, where displacement curves all take binding of the
ligand concentrations. In fact, it can be shown that the radioligand to nsb values, an allosteric ligand will displace
IC50 for inhibition is equal to the equilibrium dissociation only to a maximum value determined by the initial concen-
tration of radioligand and the value of α for the allosteric
antagonist. In fact, if a displacement curve is observed
1.2 where the radioligand binding is not displaced to nsb
1.0 values, this is presumptive evidence that the antagonist is
operating through an allosteric mechanism. The maximum
Fraction max.

0.8
displacement of a given concentration of radioligand [A ]
0.6 by an allosteric antagonist with given values of α is (see
0.4 Section 4.6.5):
0.2 ½A Kd 1 1
Maximal Fractional Inhibition 5 (4.13)
0.0 ½A =Kd 1 1=α
−3 −1 1 3 5 where Kd is the equilibrium dissociation constant of the
Log ([B] / KB) radioligand-receptor complex (obtained from saturation
FIGURE 4.7 Displacement curves for a non-competitive binding studies). The observed displacement for a range
antagonist. Displacement curve according to Equation 4.9 for values of allosteric antagonists for two concentrations of radio-
of radioligand [A ]/Kd 5 0.3 (curve with lowest ordinate scale beginning ligands is shown in Figure 4.9. The effects shown in
at 0.25), 1, 3, 10, 30, and 100. While the ordinate scale on these curves
Figure 4.9 indicate a practical test for the detection of
increases with increasing [A ]/Kd values, the location parameter along
the x-axis does not change. allosteric versus competitive antagonism in displacement

1.2 α = 0.01 1.2 α = 0.1


1.0 1.0 [A*] / Kd = 100
Fraction max.

Fraction max.

0.8 0.8 30

0.6 [A*] / Kd = 100 10


0.6
30 3
10 1
0.4 3 0.4
1
0.3 0.3
0.2 0.2
0.0 0.0
−3 −1 1 3 5 −3 −1 1 3 5
(A) Log ([B] / KB) (B) Log ([B] / KB)
FIGURE 4.8 Displacement curves according to Equation 4.10 for an allosteric antagonist with different cooperativity
factors (panel A, α 5 0.01; panel B, α 5 0.1). Curves shown for varying values of radioligand ([A ]/Kd). It can be seen
that the curves do not reach nsb values for high values of radioligand and that this effect occurs at lower concentrations
of radioligand for antagonists of higher values of α.
70 Chapter | 4 Pharmacological Assay Formats: Binding

0.35 1.0
0.30
0.8
0.25 α
0.20 0.6 0.5
Bo

Bo
0.15 α 0.3
0.4
0.5
0.10
0.3 0.1
0.2
0.05 0.1 0.03
0.03 0.01
0.00 0.0
0.001 0.01 0.1 1 10 100 100 0.001 0.01 0.1 1 10 100 100
(A) [B] / KB : Log Scale (B) [B] / KB : Log Scale

FIGURE 4.9 Displacement curves for allosteric antagonists with varying values of α (shown on figure). Ordinates:
bound radioligand. (A) Concentration of radioligand [A ]/Kd 5 0.1. (B) Displacement of higher concentration of radioli-
gand [A ]/Kd 5 3.

100

80 70
60 α = 0.003
60 50
%Bo

[IC50] / KB

40 0.01
40
30
20 0.03
20
10 0.1

0 0
−11 −9 −7 −5 0 10 20 30 40 50 60 70
Log [antagonist] [A*] / Kd
FIGURE 4.10 Displacement of bound 125I-MIP-1α from chemokine C FIGURE 4.11 Relationship between the observed IC50 for allosteric
receptors type 1 (CCR1) by MIP-1α (filled circles) and the allosteric antagonists and the amount of radioligand present in the assay according
ligand UCB35625 (open circles). Note how the displacement by the allo- to Equation 4.14. Dotted line shows relationship for a competitive
steric ligand is incomplete. Data redrawn from [7]. antagonist.

binding studies. If the value of the maximal displace- an allosteric antagonist, the relationship is hyperbolic and
ment varies with different concentrations of radioligand, given by (see Section 4.6.6):
this would suggest that an allosteric mechanism is ð1 1 ð½A =Kd ÞÞ
operative. Figure 4.10 shows the displacement of the IC50 5 KB (4.14)
ð1 1 αð½A =Kd Þ
radioactive peptide ligand 125I-MIP-1α from chemokine
CCR1 receptors by non-radioactive peptide MIP-1α and It can be seen from this equation that the maximum
by the allosteric small molecule modulator UCB35625. of the hyperbola defined by a given antagonist (with
Clearly, the non-peptide ligand does not reduce binding ordinate values expressed as the ratio of IC50 to KB) will
to nsb levels, indicating an allosteric mechanism for this have a maximum asymptote of 1/α. Therefore, observa-
effect [7]. tion of a range of IC50 values needed to block a range of
Another, more rigorous, method to detect allosteric radioligand concentrations can be used to estimate the
mechanisms (and one that may furnish a value of α for the value of α for a given allosteric antagonist. Figure 4.11
antagonist) is to formally observe the relationship between shows the relationship between the IC50 for allosteric
the concentration of radioligand and the observed antago- antagonism and the concentration of radioligand used in
nism by displacement with the IC50 of the antagonist. the assay, as a function of α. It can be seen that, unlike
As shown with Equation 4.11, for a competitive antagonist the linear relationship predicted by Equation 4.11 (see
this relationship is linear (ChengPrusoff correction). For Figure 4.6B), the curves are hyperbolic in nature. This is
4.2 BINDING THEORY AND EXPERIMENT 71

250
[3H]Atropine binding
TABLE 4.1 Differential Effects of the Allosteric
Modulator Alcuronium on Various Probes for
200
the m2 Muscarinic Receptor

150 Agonistsa (1/α)


% basal

Arecoline 1.7
100 Acetylcholine 10
Bethanechol 10
50 [3H]methyl-QNB binding
Carbachol 9.5

0 Furmethide 8.4
−9 −8 −7 −6 −5 −4 −3 Methylfurmethide 7.3
Log [alcuronium]
Antagonists
FIGURE 4.12 Effect of alcuronium on the binding of [3H]-methyl-
QNB (filled circles) and [3H]-atropine (open circles) on muscarinic Atropineb 0.26
receptors. Ordinates are percentage of initial radioligand binding. b
Methyl-N-piperidinyl benzilate 0.54
Alcuronium decreases the binding of [3H]-methyl-QNB and increases
c
the binding of [3H]-atropine. Data redrawn from [8]. Methyl-N-quinuclidinyl benzilate 63
Methyl-N-scopolamine 0.24
a
From [10].
another hallmark of allosteric versus simple competitive b
From [11].
antagonist behavior.
c
From [8].

An allosteric ligand changes the shape of the receptor,


and in so doing will necessarily alter the rate of association
and dissociation of some trace ligands. This means that
allosterism is tracer dependent (i.e., an allosteric change of the tracer ligand. This is an initial assumption for this
detected by one radioligand may not be detected in experimental approach. Under these circumstances, the rate
the same way, or even detected at all, by another). For of dissociation of the tracer ligand (ρA t) in the presence of
example, Figure 4.12 shows the displacement binding of the allosteric ligand is given by [12,13]:
two radioligand antagonists, [3H]-methyl-QNB and [3H]-
atropine, on muscarinic receptors by the allosteric ligand
ρA t 5 ρA  e2koff2obs t (4.15)
alcuronium. It can be seen that quite different effects are
observed. In the case of [3H]-methyl-QNB, the allosteric
where ρA is the tracer ligand receptor occupancy at equi-
ligand displaces the radioligand and reduces binding to the
librium and koff-obs is given by:
nsb level. In the case of [3H]-atropine, the allosteric ligand
actually enhances binding of the radioligand [8]. There are
α½Bkoff2A B =KB 1 koff2A
numerous cases of probe dependence for allosteric effects. koff2obs 5 (4.16)
For example, the allosteric ligand strychnine has little 1 1 α½B=KB
effect on the affinity of the agonist methylfurmethide
(twofold enhanced binding) but a much greater effect on Therefore, the rate of offset of the tracer ligand in the
the agonist bethanechol (49-fold enhancement of binding presence of various concentrations of allosteric ligand can
[9]). An example of the striking variation of allosteric be used to detect allosterism (change in rates with alloste-
effects on different probes by the allosteric modulator ric ligand presence) and to quantify both the affinity
alcuronium is shown in Table 4.1 [8,10,11]. (1/KB) and α value for the allosteric ligand. Allosteric
modulators (antagonists) will generally decrease the rate
of association and/or increase the rate of dissociation of
the tracer ligand. Figure 4.13 shows the effect of the allo-
4.2.3 Kinetic Binding Studies steric ligand 5-(N-ethyl-N-isopropyl)-amyloride (EPA) on
A more sensitive and rigorous method of detecting and the kinetics of binding (rate of offset) of the tracer ligand
quantifying allosteric effects is through observation of the [3H]-yohimbine to α2-adrenoceptors. It can be seen from
kinetics of binding. this figure that EPA produces a concentration-dependent
In general, the kinetics of most allosteric modulators increase in the rate of offset of the tracer ligand, thereby
have been shown to be faster than the kinetics of binding indicating an allosteric effect on the receptor.
72 Chapter | 4 Pharmacological Assay Formats: Binding

0.0 0.40
−0.2
−0.4 0.30

Log (Bt / Bo)


−0.6

Koffobs
0.20
−0.8
−1.0 0.10
−1.2
−1.4 0.00
0 10 20 30 40 −2 −1 0 1
(A) Time (min.) (B) Log [EPA]
FIGURE 4.13 Effect of the allosteric modulator 5-(N-ethyl-N-isopropyl)-amyloride (EPA) on the kinetics
dissociation of [3H] yohimbine from α2-adrenoceptors. (A) Receptor occupancy of [3H] yohimbine with
time in the absence (filled circles) and presence (open circles) of EPA 0.03 mM, 0.1 mM (filled triangles),
0.3 mM (open squares), 1 mM (filled squares), and 3 mM (open triangles). (B) Regression of observed rate
constant for offset of concentration of [3H] yohimbine in the presence of various concentrations of EPA on
concentrations of EPA (abscissae in mM on a logarithmic scale). Data redrawn from [13].

4.3 COMPLEX BINDING PHENOMENA: the G-protein subunits and subsequent activation of
AGONIST AFFINITY FROM BINDING effectors (see Chapter 2). For the purposes of binding, this
process can lead to an aberration in the binding reaction
CURVES
as perceived in experimental binding studies. Specifically,
The foregoing discussion has been restricted to the simple the activation of the receptor with subsequent binding of
Langmuirian system of the binding of a ligand to a that receptor to another protein (to form a ternary complex
receptor. The assumption is that this process produces no of receptor, ligand, and G-protein) can lead to the apparent
change in the receptor (i.e., analogous to Langmuir’s observation of a “high-affinity” site  a ghost site that
binding of molecules to an inert surface). The conclusions has no physical counterpart but appears to be a separate
drawn from a system where the binding of the ligand binding site on the receptor. This is caused by two-stage
changes the receptor are different. One such process is binding reactions, represented as:
agonist binding, in which, due to the molecular property Ka Kg
of efficacy, the agonist produces a change in the receptor A 1 R 

! AR 1 ½G 

! ARG (4.19)
upon binding to elicit a response. Under these circum-
In the absence of two-stage binding, the relative quanti-
stances, the simple schemes for binding discussed for
ties of [AR] and [R] are controlled by the magnitude of Ka
antagonists may not apply. Specifically, if the binding of
in the presence of ligand [A]. This, in turn, defines the
the ligand changes the receptor (produces an isomeriza-
affinity of the ligand for R (affinity 5 [AR]/([A] [R])).
tion to another form) the system can be described as:
Therefore, if an outside influence alters the quantity of
Ka χ [AR], the observed affinity of the ligand for the receptor R
A 1 R 

! AR 

! AR

(4.17) will change. If a ligand predisposes the receptor to bind to
σ
G-protein, then the presence of G-protein will drive the
Under these circumstances, the observed affinity of binding reaction to the right (i.e., [AR] complex will be
the ligand for the receptor will not be described by KA removed from the equilibrium defined by Ka). Under these
(where KA 5 1/Ka) but rather by that microaffinity modi- circumstances, more [AR] complex will be produced than
fied by a term describing the avidity of the isomerization that governed by Ka. The observed affinity will be higher
reaction. The observed affinity will be given by (see than it would be in the absence of G-protein. Therefore,
Section 4.6.7): the property of the ligand that causes the formation of the
KA  χ=σ ternary ligand/receptor/G-protein complex (in this case,
Kobs 5 (4.18) efficacy) will cause the ligand to have a higher affinity
1 1 χ=σ
than it would have if the receptor were present in isolation
One target type for which the molecular mechanism (no G-protein present). Figure 4.14 shows the effect of
of efficacy has been partly elucidated is the G-protein- adding a G-protein to a receptor system on the affinity of
coupled receptor (GPCR). It is known that activation an agonist. As shown in this figure, the muscarinic agonist
of GPCRs leads to an interaction of the receptor with oxotremorine has a receptor equilibrium dissociation
separate membrane G-proteins to cause dissociation of constant of 6 μM in a reconstituted phospholipid vesicle
4.3 COMPLEX BINDING PHENOMENA: AGONIST AFFINITY FROM BINDING CURVES 73

devoid of G-proteins. However, upon addition of G0 pro- value of KG, the equilibrium dissociation constant for the
tein, the affinity increases by a factor of 600 (10 nM). ternary complex of ligand/receptor/G-protein). The effects
This effect can actually be used to estimate the of various concentrations of G-protein on the binding
efficacy of an agonist (i.e., the propensity of a ligand to saturation curve to an agonist ligand are shown in
demonstrate high affinity in the presence of G-protein, Figure 4.15A. It can be seen from this figure that increas-
vide infra). The observed affinity of such a ligand is ing concentrations of G-protein in this system cause a
given by (see Section 4.6.8): progressive shift to the left of the saturation dose-response
curve. Similarly, the same effect is observed in displace-
KA ment experiments. Figure 4.15B shows the effect of differ-
Kobs 5 (4.20)
1 1 ½G=KG ent concentrations of G-protein on the displacement of a
radioligand by a non-radioactive agonist.
where KG is the equilibrium dissociation constant of the
The previous discussion assumes that there is no
receptor/G-protein complex. A low value for KG indicates
limitation on the stoichiometry relating receptors and
tight binding between receptors and G-proteins (i.e., high
G-proteins. In recombinant systems, where receptors
efficacy). It can be seen that the observed affinity of the
are expressed in surrogate cells (often in large quanti-
ligand will be increased (decrease in the equilibrium disso-
ties), it is possible that there may be limited quantities
ciation constant of the ligand-receptor complex) with
of G-protein available for complexation with receptors.
increasing quantities of G-protein [G] and/or very efficient
Under these circumstances, complex saturation and/or
binding of the ligand-bound receptor to the G-protein (low
displacement curves can be observed in binding studies.
Figure 4.16A shows the effect of different submaximal
effects of G-protein on the saturation binding curve
120
to an agonist radioligand. It can be seen that clear two-
100 phase curves can be obtained. Similarly, two-phase dis-
placement curves also can be seen with agonist ligands
80 displacing a radioligand in binding experiments with
% Bo

60
subsaturating quantities of G-protein (Figure 4.16B).
Figure 4.17 shows an experimental displacement curve
40 of the antagonist radioligand for human calcitonin recep-
tors [125I]-AC512 by the agonist amylin in a recombi-
20
nant system where the number of receptors exceeds the
0 amount of G-protein available for complexation to
−11 −9 −7 −5 −3 the ternary complex state. It can be seen that the dis-
Log [oxotremorine] placement curve has two distinct phases: a high-affinity
FIGURE 4.14 Effects of G-protein on the displacement of the mus- (presumably due to coupling to G-protein) binding
carinic antagonist radioligand [3H]-L-quinuclidinyl benzylate by the process followed by a lower-affinity binding (no benefit
agonist oxotremorine. Displacement in reconstituted phospholipid of G-protein coupling).
vesicles (devoid of G-protein sububits) shown in open circles. Addition
While high-affinity binding due to ternary complex for-
of G-protein (G0 5.9 nM βγ-subunit/3.4 nM α0-IDP subunit) shifts the
displacement curve to the left (higher affinity; see filled circles) by a mation (ligand binding to the receptor followed by binding
factor of 600. Data redrawn from [14]. to a G-protein) can be observed in isolated systems where

120 120 FIGURE 4.15 Complex binding


curves for agonists in G-protein
100 100 unlimited receptor systems.
(A) Saturation binding curves for
80 80 an agonist where there is high-
0 = [G]/[KG] 0 = [G]/[KG] affinity binding due to G-protein
% Bo

% Bo

60 60 complexation. Numbers next to


100 100 curves refer to the amount of
40 30 40 30 G-protein in the system. (B)
1 1 Displacement of antagonist radio-
20 10 20 10 ligand by same agonist in
G-protein unlimited system.
0 0
−5 −3 −1 1 3 −5 −3 −1 1 3
(A) Log ([A*] / Kd) (B) Log ([B] / KB)
74 Chapter | 4 Pharmacological Assay Formats: Binding

120 120

100 100

80 80 0
% Bo [G]/[Rtot] = 1 0.15

% Bo
60 0.75 60 0.50
0.50 0.75
40 40
0.15 [G]/[Rtot] = 1
20 0 20

0 0
−5 −3 −1 1 3 −5 −3 −1 1 3
(A) Log ([A*] / Kd) (B) Log ([B] / KB)

FIGURE 4.16 Complex binding curves for agonists in G-protein limited receptor systems. (A) Saturation
binding curves for an agonist where the high-affinity binding due to G-protein complexation 5 100 3 Kd
(i.e., Kobs 5 Kd/100). Numbers next to curves refer to ratio of G-protein to receptor. (B) Displacement of
antagonist radioligand by same agonist in G-protein limited system.

10
9
100 8
7
80
6
60
% Bo

pKI

5
40 4
20 3
2
0
−12 −11 −10 −9 −8 −7 −6 −5 1

Log [amylin] 0
1 2 3 4 5 6
FIGURE 4.17 Displacement of antagonist radioligand 125I-AC512 FIGURE 4.18 Affinity of adenosine receptor agonists in whole cells
by the agonist amylin. Ordinates: percentage of initial binding value (red bars) and membranes (blue bars, high-affinity binding
for AC512. Abscissae: logarithms of molar concentrations of rat amylin. site). Data shown for (1) 2-phenylaminoadenosine, (2) 2-chloroadenosine,
Open circles are data points, solid line fit to two-site model for binding. (3) 5’-N-ethylcarboxamidoadenosine, (4) N6-cyclohexyladenosine, (5)
Dotted line indicates a single phase displacement binding curve with a (-)-(R)-N6-phenylisopropyladenosine, and (6) N6-cyclopentyladenosine.
slope of unity. Data redrawn from [4]. Data redrawn from [15].

the ternary complex can accumulate and be quantified, this concentration of GTP can be depleted and thus the
effect is cancelled in systems where the ternary complex is two-stage binding reaction is observed (i.e., the ternary
not allowed to accumulate. Specifically, in the presence of complex accumulates). However, in whole-cell experi-
high concentrations of GTP (or a chemically stable analog ments, the intracellular concentration of GTP is high and
of GTP such as GTPγS), the formation of the ternary the ternary complex [ARG] species does not accumulate.
complex [ARG] is followed immediately by hydrolysis of Under these circumstances, the high-affinity binding of
GTP and the G-protein and dissociation of the G-protein agonists is not observed, only the so-called “low-affinity”
into α- and γβ-subunits (see Chapter 2 for further details). state of agonist binding to the receptor. Figure 4.18 shows
This causes subsequent dissolution of the ternary complex. the binding (by displacement experiments) of a series of
Under these conditions, the G-protein complex does not adenosine receptor agonists to a broken-cell membrane
accumulate, and the coupling reaction promoted by ago- preparation (where high-affinity binding can be observed)
nists is essentially nullified (with respect to the observable and the same agonists in a whole-cell preparation (where
radioactive species in the binding reaction). When this the results of G-protein coupling are not observed). It can
occurs, the high-affinity state is not observed in the binding be seen from this figure that a phase shift for the affinity
experiment. This has a practical consequence in binding of the agonists under these two binding experiment condi-
experiments. In broken-cell preparations for binding, the tions is observed. The broken-cell preparation reveals the
4.4 EXPERIMENTAL PREREQUISITES FOR CORRECT APPLICATION OF BINDING TECHNIQUES 75

180
160 TABLE 4.2 Criteria for Binding Experiments
140
120 Minimal criteria and optimal conditions for binding
KL / KH

100 experiments: The means of making the ligand chemically


80 detectable (i.e., addition of radioisotope label, fluorescent
60 probe) does not significantly alter the receptor biology of the
40 molecule.
20 G The binding is saturable.
0 G The binding is reversible and able to be displaced by other
0.0 0.2 0.4 0.6 0.8 1.0 1.2 ligands.
Intrinsic activity G There is a ligand available to determine non-specific

FIGURE 4.19 Correlation of the GTP shift for β-adrenoceptor agonists binding.
G There is sufficient biological binding material to yield a
in turkey erythrocytes (ordinates) and intrinsic activity of the agonists in
functional studies (abscissae). Data redrawn from [16]. good signal-to-noise ratio but not too much so as to cause
depletion of the tracer ligand.
G For optimum binding experiments, the following conditions
should be met: There is a high degree of specific binding
and a concomitantly low degree of non-specific binding.
effects of the ability of the agonists to promote G-protein
G Agonist and antagonist tracer ligands are available.
coupling of the receptor. This latter property, in effect, is G The kinetics of binding are rapid.
the efficacy of the agonist. Thus, ligands that have a high G The ligand used for determination of non-specific binding
observed affinity in broken-cell systems often have a high has a different molecular structure from the tracer ligand.
efficacy. A measure of this efficacy can be obtained by
observing the magnitude of the phase shift of the affinities
measured in broken-cell and whole-cell systems.
A more controlled experiment to measure the ability of
agonists to induce the high-affinity state, in effect
a measure of efficacy, can be done in broken-cell prepara- 4.4 EXPERIMENTAL PREREQUISITES
tions in the presence and absence of saturating concentra- FOR CORRECT APPLICATION OF
tions of GTP (or GTPγS). Thus, the ratio of the affinity in BINDING TECHNIQUES
the absence and presence of GTP (ratio of the high-
affinity and low-affinity states) yields an estimate of 4.4.1 The Effect of Protein Concentration
the efficacy of the agonist. This type of experiment is
on Binding Curves
termed the “GTP shift” after the shift to the right of
the displacement curve for agonist ligands after cancella- In the quest for optimal conditions for binding experi-
tion of G-protein coupling. Figure 4.19 shows the effects ments, there are two mutually exclusive factors with regard
of saturating concentrations of GTPγS on the affinity to the amount of receptor used for the binding reaction.
of β-adrenoceptor agonists in turkey erythrocytes. As can On the one hand, increasing receptor (Bmax) also increases
be seen from this figure, a correlation of the magnitude of the signal strength and usually the signal-to-noise ratio.
GTP shifts for a series of agonists and their intrinsic activ- This is a useful variable to manipulate. On the other hand,
ities as measured in functional studies (a more direct mea- a very important prerequisite to the use of the Langmuirian
sure of agonist efficacy; see Chapter 5). The GTP shift type kinetics for binding curves is that the binding reaction
experiment is a method to estimate the efficacy of an ago- does not change the concentration of tracer ligand being
nist in binding studies. bound. If this is violated (i.e., if the binding is high enough
The previous discussions indicate how binding experi- to deplete the ligand), then distortion of the binding curves
ments can be useful in characterizing and quantifying the will result. The amount of tracer ligand-receptor complex
activity of drugs (provided the effects are detectable as as a function of the amount of receptor protein present is
changes in ligand affinity). As for any experimental pro- given as (see Section 4.6.9):
cedure, there are certain prerequisite conditions that must
be attained for the correct application of this technique to 1   
½A R 5 AT 1 Kd 1 Bmax
the study of drugs and receptors. A short list of required 2
qffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi (4.21)
and optimal experimental conditions for successful  
binding experiments is given in Table 4.2. Some special 2 ð½AT 1Kd 1Bmax Þ2 2 4½AT Bmax g
experimental procedures for determining equilibrium
conditions involve the adjustment of biological material where the radioligand-receptor complex is [A R] and
(i.e., membrane or cells) for maximal signal-to-noise ½AT   is the total concentration of radioligand. Ideally,
ratios and/or temporal approach to equilibrium. These are the amount of receptor (magnitude of Bmax) should not
outlined in the material following. limit the amount of [A R] complex formed and there
76 Chapter | 4 Pharmacological Assay Formats: Binding

4 should be a linear relationship between [A R] and Bmax.


However, Equation 4.21 indicates that the amount of [A R]
complex formed for a given [A ] indeed can be limited by
Bmax (mol/L) x 109

3
the amount of receptor present (magnitude of Bmax) as Bmax
2 values exceed Kd. A graph of [A R] for a concentration
of [A ] 5 3 3 Kd as a function of Bmax is shown in
1 Figure 4.20. It can be seen that as Bmax increases, the rela-
tionship changes from linear to curvilinear as the receptor
0
begins to deplete the tracer ligand. The degree of curvature
0 1 2 3 4 5 6 varies with the initial amount of [A ] present. Lower
[AR] nM concentrations are affected at lower Bmax values than are
FIGURE 4.20 Effect of increasing protein concentration on the bind- higher concentrations. The relationship between [AR] and
ing of a tracer ligand present at a concentration of 3 3 Kd. Ordinates: Bmax for a range of concentrations of [A ] is shown in
[A R] in moles/L calculated with Equation 4.21. Abscissae: Bmax in Figure 4.21A. When Bmax levels are exceeded (beyond the
moles/L 3 109. Values of Bmax greater than the vertical solid line indi- linear range), saturation curves shift to the right and do not
cate region where the relationship between Bmax and [A R] begins to be
come to an observable maximal asymptotic value. The
non-linear and where aberrations in the binding curves will be expected
to occur. effect of excess receptor concentrations on a saturation
curve is shown in Figure 4.21B.

140 Bmax = 130 pM 60 Bmax = 60 nM

120 50
100
40
[A*R] : nM
[A*R] : pM

80
30
60
20
40
20 10

0 0
−12 −11 −10 −9 −8 −6 −12 −11 −10 −9 −8 −6
Log [A*] Log [A*]

5 100

4 Bmax = 130 pM
Log [A*R] : pM

3
% max.

50
2 60 nM

0 0
1 2 3 4 5 6 −11 −10 −9 −8 −7 −6
(A) Log (Bmax) : pM (B) Log [A*]
FIGURE 4.21 Effects of excess protein on saturation curves. (A) Bound ligand for a range of concentrations of radioligand,
as a function of pM of receptor (Figure 4.20 is one example of these types of curves). The binding of the range of concentrations of
radioligands are taken at two values of Bmax (shown by the dotted lines; namely, 130 pM and 60 nM) and plotted as saturation
curves for both Bmax values on the top panels (note the difference in the ordinate scales). (B) The saturation curves shown on the
top panels are replotted as a percentage of the maximal binding for each level of Bmax. These comparable scales allow comparison
of the saturation curves and show the dextral displacement of the curves with increasing protein concentration.
4.4 EXPERIMENTAL PREREQUISITES FOR CORRECT APPLICATION OF BINDING TECHNIQUES 77

100 Radioligand alone


1

Fraction radioligand occupancy


0.8
% Bo

50
−Log Bmax 9 8 7 6
0.6

0.4
0 Radioligand + Competitor
−3 −1 1 3 5 0.2
Log [antagonist]
FIGURE 4.22 Effect of excess protein concentration on displace- 0
ment curves (as predicted by Equation 4.22). As the Bmax increases 50 100 150 200
(2 log Bmax values shown next to curves) the displacement curves shift Time (min)
to the right.
FIGURE 4.23 Binding kinetics of a radioligand with: k1 5 3.0 3
105 min21 mol21, k2 5 3.0 3 1023 min21, [A]/KA 5 3.0 in the absence
For displacement curves, a similar error occurs (solid line) and presence (dotted line) of a competitor for receptor bind-
with excess protein concentrations. The concentration of ing (k3 5 106 min21 mol21, k4 5 0.03 min21, [B]/KB 5 10 ).
[A R] in the presence of a non-tracer-displacing ligand [B]
as a function of Bmax is given by (see Section 4.6.10): k3
B 1 R 

! BR (4.24)
1 k4
½A R5 ½AT  1Kd ð11 ½B=KB Þ1Bmax

2
qffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi where [B] is the competitor and k3 and k4 the respective
2 ð½AT  1Kd ð11½B=KB Þ1Bmax Þ2 24½AT  Bmax g rates of onset and offset from the receptor. As described by
Motulsky and Mahan [18], the following differential equa-
(4.22) tions describe the binding of the radioligand and competitor
with time:
where the concentration of the displacing ligand is [B] and
KB is the equilibrium dissociation constant of the displacing d½A R=dt 5 ½A ½Rk1 2 ½ARk2 (4.25)
ligand-receptor complex. A shift to the right of displacement
curves, with a resulting error in the IC50 values, occurs with d½BR=dt 5 ½B½Rk3 2 ½BRk4 (4.26)
excess protein concentration (see Figure 4.22).
The solution to the differential equations leads to an
expression that describes the amount of radioligand bound
4.4.2 The Importance of Equilibration Time to receptors with time in the presence of the competitor:
for Equilibrium between Two Ligands  
k1 ½A  k4 ðΩ 2 ΨÞ ðk4 2ΩÞ 2Ωt ðk4 2 ΨÞ 2Ψt
In terms of ensuring that adequate time is allowed for the ρA t 5 1 e 2 e
attainment of equilibrium between a single ligand and Ω2ψ ΩΨ Ω Ψ
receptors, the experiment shown in Figure 4.1 is useful. (4.27)
However, in displacement experiments there are two
ligands (tracer and non-traceable ligand) present and they where:
must compete for the receptor. This competition can take
considerably longer than the time required for just a sin- Ω 5 1/2½k3 ½B 1 k4 1 k1 ½A  1 k2
gle ligand. This is because the free ligands can bind only 1 ððk3 ½B1k4 2k1 ½A 2k2 Þ2 1 4k3 k1 ½A ½BÞ1=2 
to free unbound receptors (except in the case of allosteric
mechanisms, vide infra). Therefore, the likelihood of a and:
receptor being free to accept a ligand depends on the
reversibility of the other ligand, and vice versa. Assuming Ψ 5 1/2½k3 ½B 1 k4 1 k1 ½A  1 k2
mass action kinetics describe the binding of the radioli-
2 ððk3 ½B1k4 2k1 ½A 2k2 Þ2 14k3 k1 ½A ½BÞ1=2 
gand [A ] and competitive antagonist [B]:
k1 Figure 4.23 shows the kinetics of binding of a radioli-
A 1 R 

! 
A R (4.23) gand in the absence and presence of a competitor with
k2
comparatively rapid binding kinetics; it can be seen that it
where [A] is the radioligand and k1 and k2 the respective takes longer to reach equilibrium for the radioligand in
rates of onset and offset from the receptor. the presence of the competitor. This should be considered
78 Chapter | 4 Pharmacological Assay Formats: Binding

0.6 0.6
[B] / KB = 0

0.4 0.4
Fract. Bo

Fract. Bo
10 240 min 30 min
0.2 0.2
30

0.0 0.0
−10 40 90 140 190 240 −10 −9 −8 −7 −6 −5
(A) Time (min.) (B) Log [antagonist]
FIGURE 4.24 Time course for equilibration of two ligands for a single receptor. (A) Time course for dis-
placement of a radioligand present at a concentration of [A ]/Kd 5 1. Kinetic parameter for the radioligand
k1 5 105 s21 mol21, k2 5 0.05 s21. Equilibrium is attained within 30 minutes in the absence of a second ligand
([B]/KB 5 0). Addition of an antagonist (kinetic parameters 5 k1 5 106 s21 mol21, k2 5 0.001 s21) at concentra-
tions of [B]/KB 5 10 and 30, as shown in panel A. (B) Displacement of radioligand [A ] by the antagonist B mea-
sured at 30 minutes and at 240 minutes. It can be seen that a tenfold error in the potency of the displacing ligand
[B] is introduced into the experiment by inadequate equilibration time.

O H H presence of a receptor antagonist (at two concentrations


N
N N

O
[B]/KB 5 10 and 30), a clearly biphasic receptor occupancy
S

O
0.06 Ki20227 O pattern by the radioligand can be observed, in which
O N the radioligand binds to free receptors quickly (before
occupancy by the slower acting antagonist) and then a
0.04
FRET

re-equilibration occurs as the radioligand and antagonist


Sunitinib redistribute according to the rate constants for receptor
H occupancy of each. The equilibrium for the two ligands
0.02 N H

F
N N
does not occur until . 240 minutes. Figure 4.23B shows
O
N
H
O
the difference in the measured affinity of the antagonist at
times of 30 and 240 minutes. Figure 4.25 shows this effect
0 250 500 750 1000 1250 1500 1750 with Fluorescence Resonance Energy Transfer (FRET)
Time (seconds)
binding where the tracer and antagonist are added simulta-
neously and the FRET signal is monitored in real time. This
FIGURE 4.25 Fluorescence Resonance Energy Transfer (FRET) signal
for a labeled antibody for colony-stimulating factor 1 receptors and small
figure shows that a biphasic binding curve is seen for the
molecule kinase inhibitor tracer conjugated to a label. Antibody, tracer slowly dissociating antagonist Ki20227 and not for the rap-
and antagonist were added simultaneously and the FRET signal moni- idly dissociating antagonist sunitinib [20]. It can also be
tored in real time; data shown for Ki20227 (316 nM, t1/2 5 330 min) and seen from these data that the times thought adequate from
sunitinib (316 nM, t1/2 5 1 min). Data redrawn from [20]. the observation of a single ligand to the receptor (as that
shown in Figure 4.1) may be quite inadequate compared
to the time needed for two ligands to come to temporal
when designing binding experiments, i.e., measurement
equilibrium with the receptor. Therefore, in the case of
of radioligand kinetics to determine when the experiment
displacement experiments utilizing more than one ligand,
should be terminated and measurements taken by obser-
temporal experiments should be carried out to ensure that
vation of radioligand binding alone may underestimate
adequate times are allowed for complete equilibrium to be
the time needed for attainment of equilibrium.
achieved for two ligands.
Radioligand binding experiments are usually initiated
by addition of the membrane to a premade mixture of radio-
active and non-radioactive ligand. After a period of time
thought adequate to achieve equilibrium (guided by experi- 4.5 CHAPTER SUMMARY AND
ments like that shown in Figure 4.1), the binding reaction is CONCLUSIONS
halted and the amount of bound radioligand is quantified.
Figure 4.24 shows the potential hazard of using kinetics G If there is a means to detect (i.e., radioactivity,
observed for a single ligand (i.e., the radioligand) as being fluorescence) and differentiate between protein-
indicative of a two-ligand system. In the absence of another bound and free ligand in solution, then binding can
ligand, Figure 4.23A shows that the radioligand comes directly quantify the interaction between ligands
to equilibrium binding within 30 minutes. However, in the and receptors.
4.6 DERIVATIONS 79

G Binding experiments are done in three general 4.6.1 Displacement Binding: Competitive
modes: saturation, displacement, and kinetic binding. Interaction
G Saturation binding requires a traceable ligand but
directly measures the interaction between a ligand The effect of a non-radioactive ligand [B] displacing a
and a receptor. radioligand [A ] by a competitive interaction is shown
G Displacement binding can be done with any mole- schematically as:
cule and measures the interference of the molecule
with a bound tracer.
G Displacement experiments yield an inverse sigmoi-
dal curve for nearly all modes of antagonism. ð4:28Þ
Competitive, non-competitive, and allosteric antago-
nism can be discerned from the pattern of multiple
displacement curves.
G Allosteric antagonism is characterized by the fact
that it attains a maximal value. A sensitive method where Ka and Kb are the respective ligand-receptor associ-
for the detection of allosteric effects is through ation constants for radioligand and non-radioactive ligand.
studying the kinetics of binding. The following equilibrium constants are defined:
G Kinetic experiments are also useful to determine
the time needed for attainment of equilibria and to ½A R
½R 5 (4.29)
confirm reversibility of binding. ½A Ka
G Agonists can produce complex binding profiles due
to the formation of different protein species (i.e., Kb ½B½A R
ternary complexes with G-proteins). The extent of ½BR 5 Kb ½B½R 5 (4.30)
½A Ka
this phenomenon is related to the magnitude of ago-
nist efficacy and can be used to quantify efficacy. Total receptor concentration ½Rtot  5 ½R 1 ½A R 1 ½BR
G While the signal-to-noise ratio can be improved with
(4.31)
increasing the amount of membrane used in binding
studies, too much membrane can lead to depletion of
radioligand with a concomitant introduction of errors This leads to the expression for the radioactive species
in the estimates of ligand affinity. [A R]/[Rtot] (denoted as ρ ):
G The time to reach equilibrium for two ligands and
a receptor can be much greater than that required ½A Ka
ρ 5 (4.32)
for a single receptor and a single ligand. ½A K a 1 ½BKb 1 1

Converting to equilibrium dissociation constants (i.e.,


4.6 DERIVATIONS Kd 5 1/Ka) leads to the equation:

G Displacement binding: competitive interaction (4.6.1). ½A =Kd


ρ 5 (4.33)
G Displacement binding: non-competitive interaction ½A =Kd 1 ½B=KB 1 1
(4.6.2).
G Displacement of a radioligand by an allosteric
antagonist (4.6.3).
G Relationship between IC50 and KI for competitive 4.6.2 Displacement Binding: Non-
antagonists (4.6.4). competitive Interaction
G Maximal inhibition of binding by an allosteric
antagonist (4.6.5). It is assumed that mass action defines the binding of the
G Relationship between IC50 and KI for allosteric radioligand to the receptor and that the non-radioactive
antagonists (4.6.6). ligand precludes binding of the radioligand [A ] to receptor.
G Two-stage binding reactions (4.6.7). There is no interaction between the radioligand and displa-
G Effect of G-protein coupling on observed agonist cing ligand. Therefore, the receptor occupancy by the
affinity (4.6.8). radioligand is defined by mass action times the fraction q of
G Effect of excess receptor in binding experiments: receptor not occupied by non-competitive antagonist:
saturation binding curve (4.6.9).
½A =Kd
G Effect of excess receptor in binding experiments: ρ 5 q (4.34)
displacement experiments (4.6.10). ½A =Kd 1 1
80 Chapter | 4 Pharmacological Assay Formats: Binding

where Kd is the equilibrium dissociation constant of αKa 5 ½A RB=½BR½A  (4.41)


the radioligand-receptor complex. The fraction of receptor
bound by the non-competitive antagonist is given as (1 2 q). αKb 5 ½A RB=½A R½B (4.42)
This yields the following expression for q: Solving for the radioligand-bound receptor species [A R]
and [A RB] as a function of the total receptor species:
q 5 ð11½B=KB Þ21 (4.35)
ð½Rtot  5 ½R 1 ½A R 1 ½BR 1 ½A RBÞ yields
Combining Equations 4.34 and 4.35 and rearranging
yield the following expression for radioligand bound in
the presence of a non-competititve antagonist: ½A R 1 ½A RB
½Rtot 
½A =Kd
ρ 5 (4.36) ðð1=α½BKb Þ 1 1Þ
½A =Kd ð½B=KB 1 1Þ 1 ½B=KB 1 1 5
ðð1=α½BKb Þ 1 ð1=αKa Þ 1 ð1=α½A Ka Kb Þ 1 1Þ
The concentration that reduces binding by 50% is (4.43)
denoted as the IC50. The following relation can be defined:

½A =Kd 0:5½A =Kd Simplifying and changing association to dissociation


5 constants (i.e., Kd 5 1/Ka) yield (as defined by Ehlert, [19]):
½A =Kd ðIC50 =KB 1 1Þ 1 IC50 =KB 1 1 ½A =Kd 1 1
(4.37) ½A =Kd ð1 1 α½B=KB Þ
ρ 5 (4.44)
½A =Kd ð1 1 α½B=KB Þ 1 ½B=KB
 11
It can be seen that the equality defined in Equation
4.37 is true only when IC50 5 KB (i.e., the concentration
of a non-competitive antagonist that reduces the binding
of a tracer ligand by 50% is equal to the equilibrium dis-
sociation constant of the antagonist-receptor complex). 4.6.4 Relationship between IC50 and KI
for Competitive Antagonists
4.6.3 Displacement of a Radioligand A concentration of displacing ligand that produces a
50% decrease in ρ is defined as the IC50. The following
by an Allosteric Antagonist relation can be defined:
It is assumed that the radioligand [A ] binds to a site sep-
arate from the one binding an allosteric antagonist [B]. ½A =Kd 0:5½A =Kd
Both ligands have equilibrium association constants for 
5  (4.45)
½A =Kd 1 1 ½A =Kd 1 IC50 =KB 1 1
receptor complexes of Ka and Kb, respectively. The bind-
ing of either ligand to the receptor modifies the affinity of
From this, the relationship between the IC50 and the
the receptor for the other ligand by a factor α. There can
amount of tracer ligand [A ] is defined as [2]:
be three ligand-bound receptor species; namely, [A R],
[BR], and [BA R]:
IC50 5 KB  ð½A =Kd 1 1Þ (4.46)

If it cannot be assumed that the free concentration of


binding probe molecule (in most cases a fluorescent) and/
or competing ligand does not change with receptor bind-
ð4:38Þ ing, then the calculation of Ki values from IC50 values
requires a different procedure [6]. The base equation for
the conversion is:

½I50
Ki 5 (4.47)
½A  50 =kd 1 ½R0 =Kd 1 1
The resulting equilibrium equations are:
where [I]50 is the free antagonist concentration at 50%
  inhibition, Kd is the equilibrium dissociation constant of
Ka 5 ½A R=½A ½R (4.39)
the fluorescent probe-receptor complex, [A ]50 is the free
concentration of fluorescent probe at 50% inhibition and
Kb 5 ½BR=½B½R (4.40) [R]0 is the free concentration of receptor at 0% inhibition.
4.6 DERIVATIONS 81

The value for [R]0 is obtained from calculating the positive 4.6.5 Maximal Inhibition of Binding by
root of: an Allosteric Antagonist
½R0 2 1 ½R0 ðKd 1 ½A T Þ  ½RT 5 0 (4.48) From Equation 4.39, the ratio of bound radioligand [A ]
in the absence and presence of an allosteric antagonist
where [A ]T and [R]T are the total concentration of fluo- [B], denoted by ρA /ρA B, is given by:
rescent probe and receptor respectively. The positive root
of Equation 4.48 is: ρA  B ½A =Kd ð1 1 α½B=KB Þ 1 ½B=KB 1 1
5 (4.56)
qffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi  ρA  ð½A =Kd 1 1Þ  ð1 1 α½B=KB Þ
½R0 5 0:5 ðKd 1½A T Þ2 14½RT 2Kd 2½A  (4.49) The fractional inhibition is the reciprocal; namely,
T
ρA /ρA B. The maximal fractional inhibition occurs as
The conservation equation for total receptor in the [B]/KB-N. Under these circumstances, maximal inhibi-
absence of antagonist [B] is: tion is given by:

½RT 5 ½R0 1 ½A R0 (4.50) ½A =Kd 1 1


Maximal Inhibition 5 (4.57)
 ½A =Kd 1 1=α
A value for [A R]0 can be calculated. For the total
fluorescent probe concentration, the following relation
holds:
½A T 5 ½A 0 1½A R0 (4.51) 4.6.6 Relationship between IC50 and KI
for Allosteric Antagonists
From Equation 4.51 and obtaining [A R]0 from
Equation 4.50, a value for [A ]0 is obtained. A value of The concentration of allosteric antagonist [B] that reduces
[A R]50 is then defined as the concentration of tracer-recep- a signal from a bound amount [A ] of radioligand by
tor complex present at 50% inhibition of binding 50% is defined as the IC50:
([A R]50 5 [A R]0/2). By analogy to Equation 4.51,
[A ]50 5 [A R]T[A R]50 5 [A ]T[A R]0 /2. The conser- ð1 1 ½A =Kd Þ
5 0:5 (4.58)
vation equation for receptor in the presence of a concentra- ½A =K d ð1 1 αIC50 =KB Þ 1 IC50 =KB 1 1
tion of antagonist that produces 50% reduction in binding is:
This equation reduces to:
½RT 5½R50 1½A R50 1½BR50 (4.52)
ð1 1 ð½A =Kd ÞÞ
The value for free receptor at the 50% inhibition point IC50 5 KB (4.59)
(defined as [R]50) is given by the mass action equation ð1 1 αð½A =Kd ÞÞ
for free tracer ligand concentration at 50% inhibition:

Kd 5 ½R50 ½A 50 =½A 50 (4.53)
4.6.7 Two-Stage Binding Reactions
By analogy to Equation 4.51:
Assume that the ligand [A] binds to receptor [R] to pro-
I50 5IC50 2 ½BR50 (4.54) duce a complex [AR], and by that reaction changes the
where IC50 is the concentration of antagonist found to receptor from [R] to [R ]:
reduce the binding by 50% under experimental condi- Ka χ
tions. Substituting for [BR]50 from Equation 4.52 with A 1 R 

! AR 

! AR

(4.60)
σ
[A R]50 as [A R]0/2 from Equation 4.53 yields:
 The equilibrium equations are:
½I50 5IC50 2 ½RT 1 Kd ½A R50 =½A 50 1½A R50
Ka 5 ½A½R=½AR (4.61)
(4.55)

χ=σ 5 ½AR=½AR  (4.62)
Thus the procedure begins with the determination of
[R]0 (Equation 4.49), then [A R]0 (Equation 4.50), and The receptor conservation equation is:
obtaining [A ]0 (Equation 4.51). This is followed by divid-
½Rtot  5 ½R 1 ½AR 1 ½AR  (4.63)
ing [A R]0 by 2 to yield [A R]50, calculating [R]50
(Equation 4.53) which then allows calculation of [BR]50 Therefore, the quantity of end product [AR ] formed
(Equation 4.52). The I50 value then is calculated (Equation for various concentrations of [A] is given as:
4.54) Substituting for I50, [A ]50 and [R]0 into Equation 4.47
allows calculation of the true Ki value for the antagonist. ½AR  ½A=KA
5 (4.64)
½Rtot  ½A=KA ð1 1 χ=σÞ 1 χ=σ
82 Chapter | 4 Pharmacological Assay Formats: Binding

where KA 5 1/Ka. The observed equilibrium dissociation One solution to Equation 4.73 is:
constant (Kobs) of the complete two-stage process is given as:
1  
½A R 5 AT 1 Kd 1 Bmax
KA  χ=σ 2
Kobs 5 (4.65) qffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi
1 1 χ=σ  
2 ð½AT 1Kd 1Bmax Þ2 2 4½AT Bmax
It can be seen that for non-zero positive values of χ/σ;
(binding promotes formation of R ), Kobs , KA. (4.74)

4.6.8 Effect of G-protein Coupling on


Observed Agonist Affinity 4.6.10 Effect of Excess Receptor in Binding
Receptor [R] binds to agonist [A] and goes on to form a Experiments: Displacement Experiments
ternary complex with G-protein [G]: The equation for the displacement of a radioligand [A ]
Ka Kg by a non-radioactive ligand [B] can be rewritten in terms
A 1 R 

! AR 1 ½G 

! ARG (4.66) of one where binding does deplete the amount of radioli-
gand in the medium (no change in ½A free ):
The equilibrium equations are:


Ka 5 ½A½R=½AR (4.67)  ð½AT  2 ½A RÞBmax


½A R 5  (4.75)
½AT  2 ½A R 1 Kd 1 ½B=KB
Kg 5 ½AR½G=½ARG (4.68)
where Bmax reflects the maximal formation of
The receptor conservation equation is: radioligand-receptor complex. Under these circumstances,
½Rtot  5 ½R 1 ½AR 1 ½ARG (4.69) the concentration of radioligand bound in the presence of
a non-radioactive ligand displacement is:
Converting association to dissociation constants (i.e.,

1/Ka 5 KA): ½A R2 2½A RðBmax 1½AT 1 Kd ð1 1½B=KBÞÞ
 (4.76)
½ARG ð½A=KA Þð½G=KG Þ 1 ½AT Bmax 5 0:
5 (4.70)
½Rtot  ½A=KA ð1 1 ½G=KG Þ 1 1
One solution to Equation 4.76 is:
The observed affinity according to Equation 4.70 is:
1 
KA ½A R 5 ½AT  1 Kd ð1 1 ½B=KB Þ 1 Bmax
Kobs 5 (4.71) 2
1 1 ð½G=KG Þ qffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi
 
2 ð½AT 1Kd ð11½BKB Þ1Bmax Þ2 2 4½AT Bmax
4.6.9 Effect of Excess Receptor in Binding
Experiments: Saturation Binding Curve (4.77)

The Langmuir adsorption isotherm for radioligand bind-


ing [A ] to a receptor to form a radioligand-receptor com-
plex [A R] can be rewritten in terms of one where it is REFERENCES
not assumed that receptor binding produces a negligible
[1] Hulme EC. Receptor biochemistry: A practical approach. Oxford:
effect on the free concentration of ligand
Oxford University Press; 1990.

ð½AT  2 ½A RÞBmax [2] Klotz IM. Ligand-receptor energetics: A guide for the perplexed.
½A R 5  (4.72) New York: John Wiley and Sons; 1997.
½AT  2 ½A R 1 Kd [3] Limbird LE. Cell surface receptors: A short course on theory and
methods. Boston: Martinus Nihjoff; 1995.
where Bmax reflects the maximal binding (in this case,
[4] Chen W-J, Armour S, Way J, Chen G, Watson C, Irving P, et al.
the maximal amount of radioligand-receptor complex). Expression cloning and receptor pharmacology of human calcitonin
Under these circumstances, analogous to the derivation receptors from MCF-7 cells and their relationship to amylin recep-
shown in Section 2.11.4, the concentration of radioli- tors. Mol Pharmacol 1997;52:116475.
gand bound is: [5] Cheng YC, Prusoff WH. Relationship between the inhibition
constant (Ki) and the concentration of inhibitor which causes 50 per-
½A R2 2 ½A RðBmax 1 ½AT   1 Kd Þ 1 ½AT  Bmax 5 0 cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol
(4.73) 1973;22:3099108.
REFERENCES 83

[6] Nikolovska-Coleska Z, Wang R, X. fang, Pan H, Tomita Y, Li P, G protein-coupled receptors: interactions of strychnine and acetylcho-
et al. Development and optimization of a binding assay for the line at muscarinic receptors. Mol Pharmacol 1995;48:36278.
XIAP BIR3 domain using fluorescence polarization. Analyt [14] Leppick RA, Lazareno S, Mynett A, Birdsall NJ. Characterization of
Biochem 2004;332:26173. the allosteric interactions between antagonists and amiloride at the
[7] Sabroe I, Peck MJ, Van Keulen BJ, Jorritsma A, Simmons G, human α2A-adrenergic receptor. Mol Pharmacol 1998;53:91625.
Clapham PR, et al. A small molecule antagonist of chemokine [15] Florio VA, Sternweis PC. Mechanism of muscarinic receptor action
receptors CCR1 and CCR3. J Biol Chem 2000;275:2598592. on Go in reconstituted phospholipid vesicles. J Biol Chem
[8] Hejnova L, Tucek S, El-Fakahany EE. Positive and negative 1989;264:390915.
allosteric interactions on muscarinic receptors. Eur J Pharmacol [16] Gerwins P, Nordstedt C, Fredholm BB. Characterization of adeno-
1995;291:42730. sine A1 receptors in intact DDT1 MF-2 smooth muscle cells. Mol
[9] Jakubic J, El-Fakahany EE. Positive cooperativity of acetylcholine Pharmacol 1990;38:6606.
and other agonists with allosteric ligands on muscarinic acetylcho- [17] Lefkowitz RJ, Caron MG, Michel T, Stadel JM. Mechanisms of
line receptors. Mol Pharmacol 1997;52:1727. hormone-effector coupling: the β-adrenergic receptor and adenylate
[10] Jakubic J, Bacakova L, El-Fakahany EE, Tucek S. Positive coopera- cyclase. Fed Proc 1982;41:266470.
tivity of acetylcholine and other agonists with allosteric ligands on [18] Motulsky HJ, Mahan LC. The kinetics of competitive radioligand
muscarinic acetylcholine receptors. Mol Pharmacol 1997;52:1729. binding predicted by the law of mass action. Mol Pharmacol
[11] Proska J, Tucek S. Mechanisms of steric and cooperative interac- 1984;25:19.
tions of alcuronium on cardiac muscarinic acetylcholine receptors. [19] Ehlert FJ. The relationship between muscarinic receptor occupancy
Mol Pharmacol 1994;45:70917. and adenylate cyclase inhibition in the rabbit myocardium. Mol
[12] Christopoulos A. Quantification of allosteric interactions at Pharmacol 1985;28:41021.
G-protein coupled receptors using radioligand assays. In: Enna SJ, [20] Uitdhaag CM, Sunnen CM, van Doornmalen AM, de Rouw N,
editor. Current protocol in pharmacology. New York: Wiley and Oubrie A, Azevedo R, et al. Multidimensional profiling of CSF1R
Sons; 2000. p. 1.22.211.22.40. screening hits and inhibitors: assessing cellular activity, target resi-
[13] Lazareno S, Birdsall NJM. Detection, quantitation, and verification of dence time, and selectivity in a higher throughput way. J Biomolec
allosteric interactions of agents with labeled and unlabeled ligands at Screen 2011;16:100717.

You might also like