You are on page 1of 24

2.

03 Receptor Theory and the Ligand–Macromolecule


Complex
J P V Heuvel, Penn State University, University Park, PA, USA
ª 2010 Elsevier Ltd. All rights reserved.

2.03.1 Introduction 28
2.03.2 Historical Perspective 28
2.03.2.1 Receptors and Chemical Selectivity 29
2.03.2.2 Law of Mass Action and Occupancy Theory 29
2.03.2.3 The Concept of Efficacy 30
2.03.2.4 The Concept of Spare Receptors 31
2.03.3 Quantitation of Receptor Occupancy 32
2.03.3.1 The Law of Mass Action 32
2.03.3.2 Assumptions Inherent to the Law of Mass Action 33
2.03.3.3 Assessment of Receptor Occupancy 33
2.03.3.3.1 Choice of label 33
2.03.3.3.2 The incubation 34
2.03.3.3.3 Separation of bound from free ligand 34
2.03.3.3.4 Nonspecific binding 36
2.03.3.4 Plotting Methods 37
2.03.3.4.1 Plotting saturation binding data 37
2.03.3.4.2 Scatchard plots 38
2.03.3.4.3 Double reciprocal plot 38
2.03.3.4.4 Hill plot 39
2.03.3.4.5 Linear regression and analysis error 40
2.03.3.4.6 Fitting a curve to determine Bmax and Kd 40
2.03.3.4.7 Evaluation of binding data: Have the assumptions been met? 40
2.03.3.5 Competitive Binding Experiments 41
2.03.3.5.1 Why use a competitive binding curve? 41
2.03.3.5.2 Performing the experiment 41
2.03.3.5.3 Analyzing competitive binding data 42
2.03.3.5.4 Calculating the KI from the IC50 43
2.03.3.6 Ligand Depletion 43
2.03.4 Quantification of Receptor Responses 43
2.03.4.1 Why Examine Receptors Based on Responses? 43
2.03.4.2 Assumptions for Determining Kd Values for Receptor–Ligand Interactions in Intact
Tissue Preparations 44
2.03.4.3 Full Agonists 44
2.03.4.4 Quantitation of Pharmacologic/Toxicologic Antagonism 46
2.03.4.4.1 Functional antagonism 47
2.03.4.4.2 Competitive antagonism 47
2.03.4.4.3 Noncompetitive antagonism 48
2.03.4.4.4 Irreversible and pseudoirreversible antagonists 49
2.03.4.4.5 Mixed antagonism 49
2.03.4.5 Partial Agonists 49
2.03.5 Conclusion 50
References 50

27
28 Basic Principles

Abbreviations FP fluorescence polarization


PEG polyethyleneglycol

2.03.1 Introduction historical account of the development of receptor


theory contains many references to drugs as opposed
A key concept in biological sciences including phar- to hormones, neurotransmitters, and xenobiotics.
macology and toxicology is that bioactive small The term ligand (L) is used interchangeably with
molecules such as drugs, hormones, and nutrients drug in this case, and simply denotes a chemical
must achieve adequate concentration at a target site that binds with affinity and specificity to a receptor.
in order to elicit a biological response. For many Drugs and certain xenobiotics presumably bind to
chemicals, the ultimate site of action is a cognate receptors designed for interaction with endogenous
protein or ‘receptor.’ The main criteria for the opera- hormones and neurotransmitters. By way of defini-
tional term ‘receptor’ are the functions of recognition tion, agonists are analogous to endogenous hormones
and transduction. By this definition, a receptor must and neurotransmitters, in the sense that they elicit a
recognize a distinct chemical entity and translate biological effect, although the effect elicited may be
information from that entity into a form that the cell stimulatory or inhibitory. In contrast, antagonists are
can interpret, and alter its state accordingly. This defined as agents that block receptor-mediated
altered state may be a change in permeability, activa- effects elicited by hormones, neurotransmitters, or
tion of a guanine nucleotide regulatory protein, or an agonist drugs by competing for receptor occupancy
alteration in the transcription of DNA. To differenti- or by interfering with agonist binding in other ways.
ate a receptor from an enzyme, the recognition unit Antagonists may not have an endogenous, physiolo-
should not chemically alter the small molecule and, to gically relevant counterpart in the strict sense of a
differentiate from a binding protein, a receptor must competitive inhibitor of receptor occupancy.
produce a biochemical change and transmit the signal. It is also worth noting that ligands, whether natu-
Often the receptor is a protein and a single compo- rally occurring, pharmaceutical, dietary, or
nent of a large complex of macromolecules that may environmental, have the same basic mechanism of
include other proteins, RNA, and DNA. interaction with receptors. The ligands as discussed
The quantification of the interaction between a herein are small chemicals that associate with the
macromolecule and a xenobiotic is often called recep- protein receptor, irrespective of the source of the
tor theory. As will be discussed later in the chapter, the molecule. We will discuss how this bimolecular
development of receptor theory predates many of the interaction is studied, the pertinent end points and
modern techniques of molecular biology and coin- approaches used to define the ligand–receptor com-
cides with developments in analytical biochemistry, plex. The interpretation of this data will vary based
basic enzymology, and pharmacology. The study of on whether the small molecule of interest is a drug or
structure–activity relationships and modifications of a pollutant, for example, but the steps taken to derive
chemicals to fit the active site of the macromolecule meaningful measurements of affinity are identical.
have become standard in the pharmaceutical indus-
try and are an important part of modern
pharmacology and toxicology. Also, the basic tools 2.03.2 Historical Perspective
of quantification and characterization of a bimolecu-
lar interaction are applicable to many disciplines The general treatises of ‘receptor theory’ have a long
including enzymology (Michaelis complex between history that date back at least 100 years and for all
enzyme and substrate), immunology (formation of intents and purposes mark the advent of contempor-
antibody–antigen complex), pharmacology (drug– ary pharmacology and toxicology. Modern scientists
receptor complex), and toxicology (toxicant– often take it for granted that drugs elicit their effects
receptor complex). via interaction with specific receptors. However, this
Much of the conceptual framework regarding concept was not always evident and evolved as a
receptor theory evolved from pharmacology and result of the remarkable insights of early scientists
the investigation of drug action. Consequently, the exploring a number of fundamental living processes.
Receptor Theory and the Ligand–Macromolecule Complex 29

In particular, pioneers such as Claude Bernard appropriate responsive cell. Erhlich conjectured
(1813–78), Paul Erhlich (1854–1915), John Langley that the normal function of cellular side chains
(1852–1926), and A. J. Clark (1885–1941) are respon- was the binding of cell nutrients, and that the
sible for much of our understanding of quantifiable affinity of toxic substances for these groups was
receptor responses. For an extensive review of this the fortuitous analogy between the structure of
historical perspective, the reader is directed to a book the exogenous toxic substance and the endogenous
of great value – Cell Surface Receptors: A Short Course on nutrient. All of his studies form the basis of quote
Theory and Methods by L. E. Limbird (1996). ‘‘Corpora non agunt nisi fixata’’ (agents cannot act
unless they are bound).
J. N. Langley studied the chemical basis for
2.03.2.1 Receptors and Chemical
autonomic transmission and neuromuscular commu-
Selectivity
nication and extended the work of Bernard. Langley
Although Claude Bernard did not talk about recep- demonstrated that nicotine could chemically stimu-
tors per se, he demonstrated that the effects of a drug late the frog gastrocnemius muscle even following
or toxin (in his case curare) depended on its ability to severing and degeneration of its motor nerves, and
access a particular site of action, what he called a locus that this action could be blocked by curare. The
of drug effect. Bernard concluded from a detailed set mutually antagonistic effects of curare and nicotine
of experiments with curare and frog gastrocnemius led Langley to conclude that nicotine and curare act
muscle contractions that this chemical did not affect on the same site within the nerve or muscle which he
the sensory nerves, but instead altered motor nerve called a receptive substance. He postulated that com-
function. Through isolating particular nerves, apply- plexes are formed between the receptive substance
ing curare followed by electrical stimulation, Bernard and the drugs according to some law by which the
was able to show a locus of effect whereby some sites relative concentration of the drugs and their affinity
were resistant to curare’s blockade of nerve transmis- for each other are critical factors. Thus, Langley first
sion. His experiments revealed the existence of a stated the concept of drug–receptor interactions
neuromuscular junction prior to the demonstration which predated the algebraic description of these
of the muscular endplate as a discrete anatomic struc- interactions, the mass action law, often attributed to
ture. Also, these studies showed that an unknown A. J. Clark (see Section 2.03.2.2).
component of particular cells made them more or
less sensitive to chemical insult.
The receptor concept is generally attributed to
2.03.2.2 Law of Mass Action
Paul Erhlich, who expanded the idea of chemical
and Occupancy Theory
‘selectivity.’ His most acclaimed studies, which
formed the basis of a Nobel Prize in Medicine in As described above, the concept of receptor-
1908, dealt with immunochemistry and antibody– mediated events dates back to the late 1800s and
antigen interactions. Erhlich demonstrated that helped explain, qualitatively, the selectivity and
antigen–antibody interactions could be described saturability of drug action. However, it was not
as direct chemical encounters. He postulated that until A. J. Clark (1885–1941) performed his research
mammalian cells possess entities or ‘side chains’ that the quantitative nature of receptor theory
that can associate with certain chemical groups on emerged. Based on studies of the antagonism
the antigen, and thus serve as the basis for attach- between acetylcholine and atropine, Clark postu-
ing the antigen to the cell. In addition to antigens, lated that drugs combine with their receptors at a
he also felt that small molecules possessed distinct rate dependent on the concentration of drug and
domains for binding to the target cell. His own receptor. Similarly, the resulting drug–receptor com-
studies established that the arsenoxide group of plex breaks down at a rate proportional to the
organic arsenicals was essential for the lethal number of complexes. These statements implied
effects of these agents. Target cells need to first that drug–receptor interactions obey the law of mass
‘bind’ the arsenical, and cells that were unable to action that was used to describe the absorption of
recognize certain constituents were insensitive to gases onto metal surfaces and other physical–chemi-
toxicity. Inherent in his ‘side chain theory’ was the cal binding isotherms. Based on these principles, a
concept of specific cell surface receptors as the mathematical expression can be provided to describe
basis for targeting bioactive agents to the drug–receptor interactions:
30 Basic Principles

Rate of combination ¼k1 ½L½R did not elicit a maximal response. These latter find-
Rate of dissociation ¼k – 1 ½LR ings suggested that even maximal saturating
occupancy of a receptor does not necessarily trans-
where k1¼rate constant for combination (also called late into a maximal physiological effect.
kon); k1¼rate constant for dissociation (also called k2 The lack of maximal response in the presence of
or koff); L¼concentration of drug; R¼concentration maximal occupancy proved to be troubling to early
of unoccupied receptor; and LR¼concentration of researchers and required the addition of a new con-
drug–receptor complex. At equilibrium (or apparent cept. Ariëns introduced the term intrinsic activity to
equilibrium, steady state), the rate of combination describe the ability of a drug to elicit its pharmaco-
equals rate of dissociation, or logical effect. He expressed the relationship between
k1 ½L½R ¼k – 1 ½LR the effect (EL) elicited by drug L and the concentra-
tion of drug–receptor complexes as follows.
which can be arranged to
EL ¼ ½LR
k – 1 ½L½R
¼ ¼ Kd ð1Þ He defined  as the ‘intrinsic activity’ of the
k1 ½LR
particular drug. The intrinsic activity of a drug was
The term Kd is defined as the equilibrium dissociation meant to be a constant that determines the effect
constant and is usually expressed as a concentration. elicited per unit of ligand–receptor complex formed.
(The equilibrium association constant, Ka, is equivalent This early definition of intrinsic activity addressed
to K1
d , but is of limited use.) Equation [1] relates the the observation that maximal receptor occupancy by
concentration of drug applied, L, to the proportion of some agonists did not elicit a maximal response.
receptors occupied. Fractional occupancy (Y) is the However, this conceptualization still could not
proportion of all receptors that are bound to ligand at explain dose–response relationships that were stee-
equilibrium. per than predicted by mass action law. Obviously, the
law of mass action and fractional occupancy were an
½LR ½LR
Fractional occupancy ¼ Y ¼ ¼ oversimplification of the biological response and
½RTotal ½R þ ½LR
would require refinement. An important concept
The above equation after some algebra (multiply the that developed from the work of Ariëns and Clark
numerator and denominator by [L] and divide by was that of efficacy, as described subsequently.
[LR]) can be described as follows.
½L
Y¼ ð2Þ 2.03.2.3 The Concept of Efficacy
½L þ Kd
It was Stephenson (1954) who introduced a major
It is important to note that the algebraic relationship conceptual advance in the understanding of the
describing fractional receptor occupancy as a func- quantitative relationship between receptor occu-
tion of drug concentration (eqn [2]) is analogous to pancy and receptor-elicited effects (1). Stephenson
the quantitative relationships between enzyme and argued that even A. J. Clark’s experimental findings
substrate introduced by Michaelis and Menton. were not in accord with a linear relationship between
Subsequently, Clark extended his hypothesis that occupancy and effect. Although Stephenson con-
drug–receptor interactions obeyed mass action law curred that eqn [2] is the probable relationship
by postulating that the fraction of receptors occupied, between the concentration of drug introduced and
Y, was directly proportional to the response of the the concentration of drug–receptor complexes
tissue. Some early data of Clark and others describing formed, he felt that there was no experimental justi-
concentration–response relationships in various fication for extending this relationship to the
model systems were consistent with this postulate. response of the tissue. He postulated three principles
However, certain data conflicted with this simple governing receptor-mediated functions that could
relationship between occupancy and effect. First, explain the previously anomalous observation that
the slope of the concentration–response relationships agonist–response curves were often steeper than the
reported was often steeper than predicted from eqn dose–response relationships that would be predicted
[2]. Second, a number of examples existed where the by simple mass action law. In addition, his postulates
application of extremely high concentrations of drug offered an explanation for the observed progressive
Receptor Theory and the Ligand–Macromolecule Complex 31

variation in the agonistic properties of a homologous 2


series of drugs: e[L]
S=
[L] + Kd
1. An agonist can produce a maximum effect while
occupying only a small proportion of the

Stimulus
receptors. e
1
2. The response is a function of receptor occupancy,
although not linearly proportional.
3. Different drugs may have varying capacities to
initiate a response and consequently occupy dif-
e=0
ferent proportions of the receptors when 0
producing equal responses. This property was
1 10 100 1000
referred to as the efficacy of the drug and is a
Log[L]
property of the L and the LR complex. A pure
antagonist would have an efficacy of zero. Figure 1 Dose–stimulus relationship with drugs of
increasing efficacy (e).

Stephenson followed these postulates with the fol-


lowing derivations: increase in the maximal stimulus without a major
change in the shape of the curve.
S¼stimulus to the tissue from the ligand, L.
S¼eY (the stimulus is proportional to fractional occu-
pancy), where e is efficacy as described above and Y is 2.03.2.4 The Concept of Spare Receptors
fractional receptor occupancy.
The law of mass action implies that the formation of a
e½L ligand–receptor complex is reversible. However, it
S¼ ðfrom eqn ½2 forY Þ ð3Þ
½L þ Kd was noted that certain antagonists did not follow this
R¼response of a tissue and R¼f(S). rule and were in fact irreversible, that is, they form
covalent bonds with a receptor. (The types of recep-
To test the validity of his postulates regarding tor antagonism will be discussed in detail in Section
various efficacies for different agonists, Stephenson 2.03.4.3.) This was shown when extensive washing of
studied a series of alkyl-trimethylammonium salts on a tissue preparation was unable to reduce the antago-
contraction of the guinea pig ileum. He noted that the nist’s effect. A receptor that is occupied by an
short alkyl chain lengths (e.g., butyl-trimethylammo- irreversible antagonist is permanently disabled; this
nium) behaved as an agonist like acetylcholine, property of irreversible antagonists was used to
whereas the longer chain length homologues acted examine a phenomenon known as spare receptors.
like atropine, an antagonist. He interpreted this antag- Research performed by Furchgott (1967) and others
onism as a property expected for a drug with low showed that there was a receptor reserve and that 100%
efficacy. Thus, the drug produces a response much occupancy was not required for maximal response.
less than the maximum even when occupying all or Consider a simplified model as shown in Figure 2 in
nearly all of the receptors. However, because a drug which drugs A, B, and C stimulate the same receptor.
with low efficacy can nonetheless occupy the recep- Although all three agonists can elicit a maximal
tors, it decreases the response elicited by a drug with response, each requires a different percentage of
high efficacy when added simultaneously. Stephenson receptors to be occupied (consider each drug–recep-
termed these low-efficacy drugs that possessed prop- tor complex to have a different intrinsic activity or
erties intermediate between agonists and antagonists efficacy). Upon irreversible inactivation of a popula-
partial agonists. tion of the receptors, it becomes possible to
The concept of efficacy may be best described by determine the fractional occupancy and rank the
examining a dose relationship for a hypothetical ser- efficacies for the three agonists. If 50% of the recep-
ies of drugs (see Figure 1) with efficacies ranging tors are inactivated, the maximal response of the
from 1 to 1000. Of course, an efficacy of zero would agonist A is decreased by 50%, whereas drugs B
result in no stimulus, and hence no response. The and C are unaffected. If 75% of the receptors are
response from efficacies greater than one would be a irreversibly inhibited, the response of A is decreased
function of the stimulus; an increase in e results in an by 75%, B’s effects are decreased by 50%, and C still
32 Basic Principles

Agonist 2.03.3 Quantitation of Receptor


A B C

Response
A Occupancy
B
2.03.3.1 The Law of Mass Action
C
Much of the discussion above showcases the diffi-
Log(Dose)
With irreversible antagonist culty in extrapolating receptor occupancy to
responses elicited, especially in light of the existence
A B of partial agonists, steep dose–response curves, and

Response
A
C spare receptors. However, the assessment of receptor
B
C occupation per se follows the law of mass action in
many instances. We will reexamine the law of mass
Log(Dose) action as outlined by Clark and others and subse-
Figure 2 Hypothetical model for examining the existence quently explain how these derivations may be used
of spare receptors. Three agonists (A, B, C) were examined in experimentally. Most analyses of radioligand-bind-
the absence (top) or presence (bottom) of an irreversible
ing experiments are based on this simple model:
antagonist. In the case of drug A, there is no spare receptor
and full occupancy is required for maximal induction. With k1
drugs B and C only 50% and 20% occupancy is required. !
LþR  LR
When one-half of the receptors are inhibited, only drug A’s k–1
effects are reduced. Greater than 80% inhibition would be
required to affect the maximal induction elicited by agonist C. where L is the ligand, R is the receptor, and LR is the
ligand–receptor complex.
The model is based on several simple ideas:
elicits a maximal response. Using this model system, 1. Binding occurs when the ligand and receptor
Nickerson (1956) demonstrated that occupancy of collide due to diffusion, and when the collision
only 1% of the histamine receptor population of has the correct orientation and sufficient energy.
guinea pig ileum was required to elicit maximal The rate of association (number of binding
contractile response, suggesting the existence of a events per unit of time) equals [L][R]k1, where
large receptor reserve for histamine receptors in k1 is the association rate constant in units of
this tissue. However, it is worth noting that receptor mol1l min1 .
reserves were not always so dramatic. For example, 2. Once binding has occurred, the ligand and recep-
Furchgott (1967) noted that for epinephrine and the tor remain bound together for a certain amount of
muscarinic receptor there was only a shift of a half time that is influenced by the affinity of the recep-
log unit, if anything, before a decrease in the max- tor and ligand for one another. The rate of
imum response was observed following exposure to dissociation (number of dissociation events per
the alkylating agent dibenamine. unit time) equals [L]k1, where k1 is the disso-
Goldstein (1974) offered a tenable teleological ciation rate constant expressed in units of min1.
explanation for such a phenomenon, that is, that 3. After dissociation, the ligand is the same as before
sensitivity to low drug concentrations and low affi- binding. As noted earlier, this is to differentiate a
nity is achieved by the spare receptor capacity. In receptor from an enzyme.
circumstances where the response desired is to be 4. Equilibrium is reached when the rate at which
rapid in onset and in termination, as in neurotrans- new LR complexes are formed equals the rate at
mission, a spare receptor capacity provides a which these complexes dissociate. Equilibrium
mechanism for obtaining a response at a very low may be more appropriately termed apparent equili-
concentration of an agonist that nonetheless has a brium or steady state, since the achievement of true
relatively low affinity for the receptor. As we shall equilibrium is often not possible in pharmacologic
discuss subsequently, the low affinity (i.e., high Kd) of systems.
the drug assures its more rapid rate of dissociation,
At equilibrium, LR complexes form at the same rate
since Kd¼k1/k1. If, alternatively, sensitivity to low
that they dissociate:
concentrations of agonist were achieved by a high
affinity of the drug for the receptor, then the rate of
½L½Rk1 ¼ ½LRk – 1
reversal of the effect would necessarily be slow.
Receptor Theory and the Ligand–Macromolecule Complex 33

which may be rearranged to define the equilibrium does not alter the binding to other unoccupied
dissociation constant Kd. sites (cooperative binding is not present).
2. The two reactants (receptor and ligand) are either free or
½L½R k – 1
¼ ¼ Kd bound to ligand. The model ignores any states of
½LR k1
partial binding. Also, the measured products do
The Kd, expressed in units of moles per liter or molar, not include degraded, metabolized, or other una-
is the concentration of ligand that occupies half of the vailable forms of drug or receptor. Importantly,
receptors at equilibrium (see below). A small Kd nonspecific binding must be accounted for such
means that the receptor has a high affinity for the that the concentration of the free or bound ligand
ligand whereas a large Kd means that the receptor has is measured accurately. Nonspecific binding
a low affinity for the ligand. Often Kd, the equilibrium includes any nonreceptor site for ligand binding
dissociation constant, is confused with k1, the dis- that would diminish the free concentration of the
sociation rate constant. However, they are obviously chemical being examined.
not the same, as denoted by the fact that their units 3. Binding is reversible. The association between a
are different. ligand and a receptor depends only on the inter-
The law of mass action predicts the fractional action of ligand and receptor and dissociation only
receptor occupancy at equilibrium as a function of on the breakdown of the ligand–receptor complex.
ligand concentration. Fractional occupancy (Y) is
The discussions that follow regarding the incubation
defined as the fraction of all receptors that are
plotting methods are dependent on the assumptions
bound to ligand:
above. If it is not possible to meet all of these criteria,
½LR ½LR ½L two choices exist. If the first assumption is not satisfied,
Y ¼ ¼ ¼ ðsee eqn ½2Þ there are more complicated model systems to describe
½RTOT ½R þ ½LR ½L þ Kd
ligand–receptor complexes that are cooperative in nat-
This equation predicts the following. When no ligand ure. The reader is directed to Limberg (1996), Cell
is available, the occupancy equals zero. When the Surface Receptors, for description of these models as
concentration of ligand is very high (many times they are beyond the scope of the present chapter. If
Kd), the fractional occupancy approaches (but never assumptions 2 and 3 are not met, the only option is to
reaches) 100%. When [L]¼Kd, the fractional occu- analyze the data in the usual way, but to interpret the
pancy is 50%. Equation [2] predicts that the results as an empirical description of the data without
approach to saturation as ligand concentration attributing rigorous thermodynamic meaning to the Kd
increases is quite slow. When the ligand concentra- values and rate constants. In other words, the data must
tion equals 4 times its Kd, it will only occupy 80% of be considered as highly suspect.
the receptors at equilibrium. The occupancy rises to
90% when the ligand concentration equals 9 times
the Kd. It takes a concentration equal to 99 times the 2.03.3.3 Assessment of Receptor
Kd to occupy 99% of the receptors at equilibrium. Occupancy
2.03.3.3.1 Choice of label
2.03.3.2 Assumptions Inherent to the Law In order to examine the binding of ligand and recep-
of Mass Action tor, one of these constituents must be easily
quantified. The most common means to quantify a
The law of mass action is a convenient and simple
ligand is to add an isotope to form a radioligand (often
model to describe a bimolecular interaction.
depicted as L). One of the most critical considera-
However, in order to coerce a complex biological
tions in the successful application of radioligand-
interaction into a simple model, several assumptions
binding techniques is the choice of an appropriate
or criteria must be met. Although termed a ‘law,’ the
isotope for radiolabeling the ligand. Each common
law of mass action is simply a model based on the
isotope has certain characteristics to be taken into
following assumptions:
consideration (see Table 1). For example, tritium is a
1. All receptor sites are equally accessible to ligand. In the popular choice because of its long half-life. However,
simplest model, all receptor sites are considered to tritium exchanges with water may require repurifica-
have equal affinity for ligand and to be indepen- tion. Carbon-14 (14C) has benefits of ease of
dent. That is, occupancy of some receptor sites incorporation into organic ligands, but suffers from
34 Basic Principles

Table 1 Considerations when choosing an isotope for labeling of drug in receptor binding assays

Specific activity
Isotope (Ci/mmol) Half-life Other considerations
3
H 29.4 12.3 years Bioactivity usually unchanged by tritiation; can introduce >1 mole 3H/mole
ligand to increase specific activity
125
I 2125 60.2 days Require tyrosine or unsaturated cyclic system in ligand structure to achieve
incorporation of 125I; high-specific radioactivity, especially useful when
receptor availability is limited
32
P 9760 14.2 days Short half-life makes 32P a difficult choice
35
S 4200 86.7 days Good sensitivity; used in metabolic labeling
14
C 0.064 5568 years Poor sensitivity

From Limbird, L. E. Cell Surface Receptors: A Short Course on Theory and Methods, 2nd ed.; Kluwer Academic Publishers: Boston, MA,
1996; pp 238.

low-specific activity. Iodine-125 (I125) is the most (femtomoles to picomoles per mg tissue). However,
common choice for radioligand products and affords in many instances the receptor is expressed in bac-
examination of low-affinity binding and low concen- teria, yeast, or insect cells, if possible. This allows for
tration of receptor. However, the incorporation of a much higher concentration of receptor than seen in
iodine into a drug may affect its affinity for a biolo- vivo, often at higher purity (>1mg ml1). However,
gical receptor. Taken together, the choice of isotope receptors that are multiprotein complexes or require
is dependent on sensitivity issues such as receptor posttranslational modifications are difficult to exam-
density, counting efficiency, and availability of tissue, ine using recombinant proteins.
as well as ease of synthesis. Another option that is
gaining popularity is to examine a drug or chemical’s
2.03.3.3.3 Separation of bound from free
fluorescence, either natural or added exogenously.
ligand
Whether this method can be performed depends on
To determine the quantity of LR accumulated, one
the ligand under question, but when available repre-
must have a method for determining the incubation
sents a sensitive and rapid means to examine
that permits the resolution of L from LR. When
receptor–ligand interactions.
receptors are embedded within membranes, the free
and bound forms of drug may be easily separated by
2.03.3.3.2 The incubation filtration or centrifugation. The speed of separation is
Critical components of the incubation include the an important consideration in determining which
labeled ligand, a source of receptor (crude membrane method to use to separate L from LR. If the rate of
preparation, recombinant protein, etc.) and a suitable dissociation is slow in comparison to the rate of
buffer. Also, the incubation must proceed for a suffi- separation, the amount of LR formed can be deter-
cient amount of time, at an appropriate temperature, mined accurately. This is the case for high affinity
until enough of the drug–receptor complex is formed ligands (low Kd). However, if the rate of ligand dis-
to be detectable. In general, increasing the concen- sociation is high, the rate of separation may become
tration of either or both of the reactants L and R can an important consideration for accurately measuring
increase the overall rate of association. These manip- the amount of drug–receptor complex.
ulations will also increase the amount of drug– Consider the following equation (for a derivation
receptor complex formed and thus lengthen the of eqn [4], see Limbird (1996)).
time required to reach equilibrium. Optimal incuba-
tion conditions are determined empirically for each
½L½R k – 1 6:9310 – 1 mol l – 1 s
system and are chosen to maximize complex forma- Kd ¼ ¼ ¼ ð4Þ
½LR k1 t1=2
tion while minimizing the degradation of L and R.
The source of receptor is dependent on the sys- where t1/2 is the half-life of the LR complex (amount
tem under study and may include crude extracts, of time required for LR to decrease by 50%). If a
membrane preparations, cytosol, or nuclear extracts. 10% loss of LR is considered acceptable during the
Typical concentrations of receptors in crude separation process, then 0.15 times the half-life is
preparations are 11012 to 1109mol l1 the allowable time required. (Note that these
Receptor Theory and the Ligand–Macromolecule Complex 35

Table 2 Relationship between equilibrium dissociation formed and reach equilibrium with the free drug.
constant Kd and allowable separation time of DR There are several variations of centrifugal separation
Kd Allowable separation time (0.15 t1/2) of drug–receptor complexes. If the receptor is mem-
brane bound, the drug–receptor complex may be
1012 1.2 days centrifuged and a tight pellet formed. The amount
1011 2.9 h
of radioactivity retained in the pellet represents LR
1010 17 min
109 1.7 min and the free drug is found in the supernatant. One
108 10 s minor variation is to layer the receptor-containing
107 0.1s solution on a high-density solution (sucrose or oil).
106 0.01 s This alteration results in less free drug being trapped
From Limbird, L. E. Cell Surface Receptors: A Short Course on in the pellet, but also makes the assay more cumber-
Theory and Methods, 2nd ed.; Kluwer Academic Publishers: some and increases the risk of LR dissociating during
Boston, MA, 1996; pp 238. the centrifugation.
In the case of soluble receptors, either the receptor
calculations are based on a k1 of 106mol1 l s1 and or the drug may be precipitated by centrifugation. In
that k1 is equivalent to ln2/t1/2.) As shown in the former situation, an antibody specific to the
Table 2, with low affinity ligand–receptor interac- receptor may be coupled to a large molecule such as
tions, the separation of L from LR must proceed sepharose. Upon centrifugation, the pellet will con-
exceedingly rapid. In the following section, the tain LR and the supernatant L, as described above for
basic separation techniques will be briefly discussed. membrane-bound receptors. Alternatively, agents
For a more detailed description of these methods, see such as polyethyleneglycol (PEG) or Sephadex
Levitzki (1984) or Limbird (1996). G-50 may be used to nonspecifically precipitate pro-
teins. Alternatively, the radioligand may be
2.03.3.3.3(i) Equilibrium dialysis The dialysis precipitated with the aid of dextran-coated charcoal,
chamber consists of two compartments separated by leaving the DR in the supernatant.
a membrane to which the receptor preparation is Typically, centrifugation can pellet the bulk of
added to one side and the radiolabeled ligand to the the particulate matter within 5 s, and as shown
other. After the solution reaches equilibrium, samples Table 2 is appropriate for receptors with a Kd of
from both chambers are taken and the radioactivity 10–8mol l1 and lower. This method is relatively
measured. The amount of drug–receptor complex fast and many replicates may be examined in one
formed is determined based on the assumption that assay. However, nonspecific binding and trapping of
the free drug is equal on the protein- and nonprotein- free drug is often a major limitation.
containing compartments.
Protein compartment LR þ L 2.03.3.3.3(iii) Vacuum filtration The incubation is
– Nonprotein compartment – L terminated by pouring the mixture through a filter
LR ðthe concentration under vacuum. The proteins (R and LR) are retained
Difference by the filter and the free radioactivity flows through.
of bound radioligandÞ
This procedure can handle a large number of samples
The advantages of equilibrium dialysis include and the filtration devices are commercially available.
the fact that equilibrium is not disturbed when sam- The filters are generally made from glass, nylon, or
ples are taken and therefore represents an accurate nitrocellulose in order to provide high affinity and
estimate of LR. Also, this method is useful for low- capacity for protein binding. The amount of LR
affinity receptor–drug interactions. Equilibrium dia- formed is determined by counting the amount of
lysis is no longer a frequently utilized method in radioactivity present on the filter after washing with
receptor-binding assays, predominantly due to the a suitable buffer. Filtration usually takes 2–5s per
long time required to reach equilibrium and second- wash, which means that the minimum ligand affinity
ary increases in the risk of protein or drug breakdown of the receptor that can be identified by this techni-
and decreased throughput. que is 108 moll1. As with other methods, the
washing of the drug–receptor complex is performed
2.03.3.3.3(ii)Centrifugation The following tech- with cold buffer to slow down the dissociation rate of
niques require that the drug–receptor complex be the drug. Nonspecific binding and trapping of
36 Basic Principles

radioligand to the filter are potential problems asso- 2.03.3.3.4 Nonspecific binding
ciated with vacuum filtration. In addition to ligand binding to receptors of physio-
logical interest, ligands often bind to nonreceptor
2.03.3.3.3(iv) Scintillation proximity assays A sites. Any interaction with a nonreceptor site would
newer type of assay is the scintillation proximity be considered nonspecific binding and is found at excess
assay, which does not require the free and the bound over true, specific interactions with the receptor.
drug to be separated. In this assay, the receptor is When performing radioligand-binding experiments,
attached to a synthetic bead which contains a scintillin a measure of both total and nonspecific binding is
(a material that emits photons when it absorbs radio- performed, and specific (receptor) binding is calcu-
active emissions). In most instances the receptor is lated as the difference (see Figure 3). The binding of
bacterially expressed, purified, and chemically radioligand is examined in the presence of excess
attached to the bead, although it is possible to adhere saturating amounts of unlabeled drug. This techni-
membrane-bound or crude receptor preparations. que assumes that a 100-fold excess of nonradioactive
When a radioactive ligand is added to the medium, ligand saturates the specific receptor binding but has
only the portion that is bound to the receptor will be no effect on the nonspecific sites. A useful rule of
close enough in proximity to the scintillin and thereby thumb is to use the unlabeled compound at a con-
produce a photon emission. Therefore, the LR is centration equal to 100 times its Kd for the receptor, if
assessed as the number of light units produced and is that is known from previous experiments or esti-
a true equilibrium-binding assay. Scintillation proxi- mated from preliminary analyses of the system. The
mity assays have become quite popular because of their drug used in excess may be the compound used as the
high-throughput capabilities. However, the production radioligand, but unlabeled, or it may be a chemical
of the synthetic beads containing the receptor of inter- that is known to bind to the same receptor. Ideally,
est may be an arduous task for many laboratories. the same results should be obtained when defining
nonspecific binding with a range of concentrations of
2.03.3.3.3(v) Fluorescence polarization several drugs. Nonspecific binding is generally pro-
assays Fluorescence polarization (FP) is a powerful portional to the concentration of radioligand (within
tool for studying molecular interactions by monitoring the range used) and will not plateau at high concen-
changes in the apparent size of fluorescently labeled trations. However, as shown in Figure 4 and
ligand. Similar to scintillation proximity assays, FP discussed subsequently in the chapter, the specific
enables the researcher to view molecular-binding binding is saturable and exhibits a sigmoidal shape
events in solution, allowing true equilibrium analysis when examined on a semilogarithmic plot.
into the low picomolar range. FP measurements do
not affect samples, so they can be treated and reana- 2.03.3.3.4(i) Additional criteria The binding of a
lyzed in order to ascertain the effect on binding by drug to a receptor is an in vitro experiment performed
such changes as pH, temperature, and salt concentra-
tion. In addition, because FP is a truly homogeneous [L][R ]tot
technique, it does not require the separation of bound [LR] =
K d + [L]
and free species. The theory of FP is based on the
observation that when a small fluorescent molecule is 100
excited with plane-polarized light, the emitted light is at ∞, y = Rtot = B max
[LR], bound ligand

largely depolarized because molecules tumble rapidly 75


in solution during its fluorescence lifetime. However,
if the tracer is bound by a larger molecule its effective B max [R]tot
50 or
molecular volume is increased. The fluorecent 2 2
ligand’s rotation is slowed so that the emitted light is
in the same plane as the excitation energy. The bound 25
and free states of the ligand each have an intrinsic
polarization value: a high value for the bound state and 0
a low value for the free state. The measured polariza- 0 20 40 60 80 100 120
tion is a weighted average of the two values, thus [Ligand]
providing a direct measure of the fraction of tracer Figure 3 The hyperbolic relationship of ligand binding
bound to receptor. isotherms.
Receptor Theory and the Ligand–Macromolecule Complex 37

150

125 Total

[LR], bound ligand


100 Specific
Specific
75

50

25 Nonspecific

0
0 20 40 60 80 100 0.1 1 10 100
[Ligand] [Ligand]
Figure 4 Hypothetical binding curves. The left figure shows total and nonspecific binding. The difference between total and
nonspecific binding is the specific binding and if this is plotted on a log-dose scale, it has a sigmoidal shape (right). If an
insufficient number of concentrations are used, often the plateau is not evident in such a representation.

under well-controlled situations. The desire is to methods must meet the following criteria: (1)
extend the data to a physiologically relevant situa- assumptions of the law of mass action, as described
tion, and therefore the following criteria may be used above, are met; (2) the incubation and separation
to assess whether the data generated can be extra- techniques are appropriate; and (3) nonspecific bind-
polated to a real-world (i.e., a real cell) situation. The ing has been adequately measured.
specific binding of L should be saturable, since a
finite number of receptors are expected in any 2.03.3.4.1 Plotting saturation binding
given biological preparation. The specificity of data
agents in competing with L for binding to R should Saturation binding experiments measure specific
parallel the specificity of physiological effect via the binding at equilibrium at various concentrations
putative receptor of interest. The use of competition (often 6–12) of the radioligand to determine receptor
binding will be discussed subsequently. Further, the number and affinity. Use of drug concentrations that
kinetics and LR of binding should be consistent with allow binding to approach saturation is crucial for
the time course of the biological effect elicited by L accurate examination of the interaction between the
and there should be consistency between Kd values drug and receptor. The next consideration is that of
obtained using steady-state incubations and those nonspecific binding. As mentioned above, a good rule
determined through kinetic experiments. of thumb is to use a concentration of unlabeled ligand
at 100 times the Kd. When examining binding in the
presence of 100Kd, the amount of [LR] detected is
2.03.3.4 Plotting Methods
considered nonspecific, and as shown in Figure 4 is
As shown in Figure 3, the relationship between linear with respect to free ligand concentration. Total
receptor occupancy ([LR]) and drug concentration binding is the amount of [LR] detected in parallel
is hyperbolic when a saturable receptor population samples without this competitor included in the
binds to ligand in a freely reversible, bimolecular incubation. The difference (total – nonspecific) is
interaction. Often the results are examined by linear the specific binding, and is the data that should conform
transformation of the experimental data. The advan- to the law of mass action. It is this binding that we
tage of such transformations is that Kd and receptor will examine in more detail.
density (Bmax or Rtot) can be easily identified. One of the criteria stated previously for a physio-
However, if the drug concentrations examined are logically relevant receptor is that the binding sites are
insufficient, as defined empirically, linear transfor- saturable. To assess saturability, the characteristics of
mations may give a distorted view of the binding binding as a function of increasing concentrations of
phenomena. In this section, both nonlinear and linear radioligand are determined. The saturation-binding
transformations will be discussed. All plotting curve can be described by eqn [2]
38 Basic Principles

½LR ½L concentration relative to amount


Y ¼ ¼ ðsee eqn ½2Þ
½Rtot ½L þ Kd of protein in the incubation (i.e.,
fmoles per mg protein).
which can be rearranged as follows
The reason for plotting B/F as the y-axis and B as the
½L½Rtot
½LR ¼ ð5Þ x-axis comes from rearranging eqn [5] (and substitut-
Kd þ ½L
ing B for LR, F for L, and Bmax for Rtot):
Equation [5] is the equation of a rectangular hyper-
ax ðF ÞðBmax Þ
bola y ¼ . An important consequence of this B¼ becomes ðBÞðKd Þ þ ðBÞðF Þ ¼ ðF ÞðBmax Þ
bþx Kd þ F
equation representing a rectangular hyperbola is
that the horizontal asymptote is Rtot or Bmax. Thus, Dividing by F, and rearranging results in eqn [6] in
Bmax can be obtained only at infinite concentrations the form of y¼mxþb:
of L. This will be of importance when linear trans-
ðB=F ÞðKd Þ þ B ¼ Bmax
formations are discussed. Other important pieces of
information can be obtained from eqn [5]. The term Bmax B
ðB=F Þ ¼ –
Kd is a measure of the affinity of the receptor for Kd Kd
ligand and is the concentration of ligand that occu- Bmax B
pies one-half of the maximal binding sites. This can ðB=F Þ ¼ –
Kd Kd
be easily derived from eqn [5] if [LR] is replaced by
½Rtot 1 Bmax
ðB=F Þ ¼ – ðBÞ þ ð6Þ
and solving for Kd (the result being Kd¼[L]). Kd Kd
2
The estimation of Bmax and Kd can be solved by either When making a Scatchard plot, you have to choose
linear transformation or by nonlinear regression, units for the y-axis. One choice is to express both free
which is described below. ligand and specific binding in counts per minute
(cpm), so the ratio of bound to free is a unitless
fraction. The advantage of this choice is that you
2.03.3.4.2 Scatchard plots can interpret y-values as the fraction of radioligand
The most common linear transformation of binding bound to receptors. If the highest y-value is large
data is the Scatchard plot. In this plot, the x-axis is (greater than 0.10), then the free concentration will
specific binding (usually labeled ‘bound’) and the be substantially less than the added concentration of
y-axis is the ratio of specific binding to concentration radioligand, and the standard analyses are not appro-
of free radioligand (usually labeled ‘bound or free’). priate. As such, one should either revise the
Some of the terms used in Scatchard plots are as experimental protocol or use special analysis meth-
follows: ods that deal with ligand depletion (see Section
B¼Bound (LR) Concentration of ligand in the incu- 2.03.3.6). The disadvantage is that the experimenter
bation that is specifically bound to cannot interpret the slope of the line without per-
the receptor at equilibrium. forming unit conversions. An alternative is to express
F¼Free (L) Concentration of free ligand present the y-axis as fmol ligand bound per mg protein per
in the incubation at equilibrium. As concentration (n mol l1). While these values are
discussed in Section 2.03.3.6, this is hard to interpret, they simplify calculation of the Kd
often estimated by the concentration which equals the reciprocal of the slope. The specific
of the drug added to the incubation. binding units cancel when the slope is calculated.
Kd Equilibrium dissociation constant. In The negative reciprocal of the slope is expressed in
the Scatchard plot, the slope of the units of concentration (n mol l1) which equals the Kd
line is equal to Kd1. Kd is expressed (see Figure 5).
in the same units as the drug con-
centration (i.e., molarity). 2.03.3.4.3 Double reciprocal plot
Bmax (Rtot) Maximum number of binding sites As the name implies, a double reciprocal plot is
in the incubation at equilibrium, or simply the reciprocal of the amount of specifically
total receptor concentration. Bmax is bound ligand versus the reciprocal of the free ligand
expressed in the same units as the concentration. The y-intercept yields the reciprocal
specific binding, usually as a of the value of maximal binding (1/Bmax) while the
Receptor Theory and the Ligand–Macromolecule Complex 39

20 4
Slope = n
x-intercept = Kd when n = 1
3
15 n=2
n=1

Log(B/Bmax–B)
1 2
Slope = −Ka = −
B/F

10 Kd
1
n = 0.5
5
0

0 –1
0 20 40 60 80 100 120 0.1 1 10 100 1000
B Log[L]
x-intercept = Bmax
Figure 7 Hill plot. When the log (specific binding/
Figure 5 Scatchard analysis. The slope of B/F versus B is
(maximum specific binding–specific binding)) is plotted
equal to –1/Kd whereas the x-intercept estimates the total
versus the log [Ligand] a straight line with a slope equal to 1
binding sites (Bmax or LRmax).
is obtained, if the law of mass action is being followed. If
positive cooperativity is observed, the slope is >1; in the
case of negative cooperativity the slope is <1.
0.030

0.025 The difference between eqns [5] and [7] is the inclu-
sion of n or the Hill coefficient. The Hill coefficent is
0.020
the maximum number of ligand molecules bound to
each molecular receptor and is also a measure of
1/LR

0.015
cooperativity between binding sites. When a ligand
0.010
binds to a single class of noncooperative receptors,
1/Bmax
this binding is described by the law of mass action
0.005 and is n¼1. If n is greater than 1, evidence supports
–1/Kd positive cooperativity, while n less than 1 indicates
negative cooperativity or multiple classes of binding
–1.00 –0.75 –0.50 –0.25 0.00 0.25 0.50 0.75 1.00 sites.
1/L Equation [7] can be rearranged to give the follow-
Figure 6 Double reciprocal plot. When the inverse of ing linear equation:
specific binding (1/LR) is plotted versus the inverse of ligand
concentration (1/L), a straight line is obtained. The ½LR
y-intercept is 1/Bmax and the x-interecept in 1/Kd. Log ¼ nLog½L – nLogðKd Þ ð8Þ
½LRmax – ½LR

x-intercept represents the negative reciprocal of the Therefore, a plot of log[[LR]/([LR]max–[LR])] as a


equilibrium dissociation constant (1/Kd). Note that function of log[L] yields a line whose slope is n and
this is identical to the Lineweaver–Burke plot (see whose intercept on the ordinate is –nlogKD (see
Figure 6). Figure 7).
In addition to the Hill plot, the cooperativity
2.03.3.4.4 Hill plot between binding sites may be examined by
The Hill plot was originally used to describe the Scatchard analysis. In this case the plot is nonlinear
cooperativity of binding of oxygen to hemoglobin and concave when positively cooperative and con-
and has been adapted to examine ligand–receptor vex when negatively cooperative (see Figure 8).
interactions. The general equation from which the Both the Hill plot and the Scatchard may be used
derivation is based is as follows: to determine if the experiments are fulfilling the
criteria listed in Section 2.03.3.2, in particular if the
½Ln  ½LRmax binding sites are of a single population and inde-
½LR ¼ ð7Þ
Kd þ ½Ln pendent of each other.
40 Basic Principles

40 2.03.3.4.6 Fitting a curve to determine


B max and K d
Equilibrium-specific binding at a particular radioli-
30 Positive cooperativity gand concentration equals fractional occupancy
Bound/free

times the total receptor number (Bmax or Rtot):


20
½L  Bmax
Specific binding ¼ ½LR ¼ See Eqn ½5
Kd þ ½L
10
This equation describes a rectangular hyperbola or a
binding isotherm. As before, [L] is the concentration
Negative cooperativity
0 of free radioligand and is plotted on the x-axis. Bmax is
50 60 70 80 90 100 the total number of receptors expressed in the same
Bound units as the y-values (i.e., cpm, sites per cell, or fmol
Figure 8 Nonlinear Scatchard plots. If the law of mass per mg protein) and Kd is the equilibrium dissociation
action is being observed, a plot of B/F versus B will be a constant (expressed in the same units as [L] usually
straight line. However, in the case of cooperative binding, n mol l1). Typical values might be a Bmax of
the Scatchard plot is nonlinear. 10–1000fmol binding sites per milligram of protein
and a Kd between 10 p mol l–1 and 100 n mol l1.
To determine the Bmax and Kd, the most accurate
2.03.3.4.5 Linear regression and analysis method is to fit the specific binding data to eqn [5]
error using nonlinear regression. There are currently many
While Scatchard, double reciprocal, and Hill plots resources available for performing nonlinear regres-
are useful plots for visualizing data, they may not sion and several statistics and plotting programs will
represent the most accurate way to analyze data. perform the analysis. An excellent source for a
Linear regression assumes that the data is normally detailed description of nonlinear regression may be
(Gaussian) distributed and that the standard devia- found at the GraphPad website.
tion is the same at every concentration of ligand. This
is not the case with data transformed in the Scatchard
plot. A second problem is that the value of X (bound) 2.03.3.4.7 Evaluation of binding data:
is used to calculate Y (bound/free). Since the Have the assumptions been met?
assumptions of linear regression are violated, the In order for the binding data to result in physiologi-
Bmax and Kd calculated by linear regression of cally relevant values of Kd and Bmax, the law of mass
Scatchard transformed data are likely to be further action must be followed. Also, the choice of ligand,
from their true values than the Bmax and Kd deter- separation procedures, and incubations must be
mined by nonlinear regression. appropriate. When examining one’s own data, or
With many receptor-binding experiments, there evaluating binding data within a manuscript, it is
are insufficient data points throughout the binding advantageous to evaluate the quality of studies.
Here are some guidelines that may be used for eva-
isotherm. This results in a clustering of data points
luation of studies on receptor–ligand interactions.
when the Scatchard analysis is performed. For
Has the law of mass action been observed? In general,
example, if there are few data points at the begin-
there are benchmarks that can be used to assess
ning of the curve and many in the plateau region,
whether the law of mass action is being followed.
the Scatchard transformation will assign undo
First, the Scatchard plot must be linear and the Hill
weight to the data point collected at the lowest
plot should have a slope equal to one. If a nonlinear
concentration of radioligand (the upper left points
Scatchard is observed, more than one population of
on the Scatchard plot). Although it is inappropriate
receptor exists, cooperativity is an issue, and Kd is not
to analyze data by performing linear regression on a a constant. Second, ligand depletion should not occur
Scatchard plot, it is often helpful to display data as a such that the amount of drug added to the incubation
Scatchard plot. As mentioned, visually interpreting is an appropriate approximation of free drug, or L.
data with Scatchard plots can be of diagnostic value, The amount of binding (total binding) in the incuba-
but it may not result in the most accurate values of tion should be less than 10% of the amount added.
Kd and Bmax. See Section 2.03.3.6 for details on what to do if this
Receptor Theory and the Ligand–Macromolecule Complex 41

value is exceeded. Third, the incubation must be at concentration of competitor should be 100 times
steady state, or pseudo-equilibrium. To decrease the the Kd of the drug–receptor complex.
time to steady state, one of the constituents should be
at considerably lower concentrations than the other.
Since the ligand is the constituent that is measured, 2.03.3.5 Competitive Binding Experiments
decreasing its concentration may affect sensitivity.
Therefore, the receptor concentration should be lim- 2.03.3.5.1 Why use a competitive binding
iting ([R]<<Kd; [R]<<[L]). Note, that this will also curve?
minimize the risk of ligand depletion. Competitive binding experiments measure the bind-
Is the binding physiologically relevant? The physiolo- ing of a single concentration of labeled ligand in the
gical relevance often cannot be addressed without presence of various concentrations of unlabeled
conducting studies beyond receptor binding. ligand. To quantify the potency of drugs in compet-
However, there are a few issues that can be exam- ing for the receptor, the IC50 (concentration that
ined. First, the formation of the ligand–receptor inhibits 50% of the specific radioligand binding)
complex must appear to be saturable. Note that value is determined for each competitor.
while 100% binding will never be achieved, com- Competitive binding experiments have several
plete saturation should be approached. If a plateau is advantages to direct binding assays and have been
not observed, either nonspecific binding has not been used for the following types of experiments:
properly accounted for, or a nonspecific interaction is
dominating the system. Second, the specificity of L
• Validate a direct binding assay. Competitive binding
experiments are an excellent way to examine the
for R should parallel the physiological effects, if physiological significance of a direct binding
known. That is, the Kd should be within an achievable assay. In this type of analysis the radioligand is
concentration at the receptor site. Examining a series competed with drugs whose potencies are known
of chemicals and measuring their affinity for a parti- from functional experiments. Demonstrating that
cular receptor may also address physiological these drugs bind with the expected potencies, or
relevance. Affinity and potency of effect should at least the expected order of potency, helps
have a similar rank order within this group of che- prove that the radioligand has identified the cor-
micals. This may be easier competition binding rect receptor.
experiments, as described subsequently.
Have the experiments been properly designed and exe-
• Determine whether a drug binds to the receptor.
Thousands of compounds can be screened to
cuted? Several important issues regarding the design identify drugs that bind to the receptor simply
and implementation of a ligand binding study was by determining if they can effectively compete
discussed in Section 2.03.3.3. The following questions with a known ligand. This can be faster and easier
should be asked when interpreting receptor charac- than other screening methods. In fact, this is the
terization studies. most common way the pharmaceutical companies
are identifying novel ligands for important recep-
1. Was the choice of radioligand and source of tor systems.
receptor appropriate? In particular, the source of
receptor is often an issue. Bacterial and yeast
• Investigate the interaction of low-affinity drugs with
receptors. Binding assays are only useful when the
expression systems are used frequently to produce radioligand has a high affinity (Kd<100 n mol l1).
soluble receptors. Often, these purification meth- A radioligand with low affinity generally has a fast
ods result in receptor preparations that dissociation rate constant, and will not stay bound
recapitulate the in vivo situation. However, if post- to the receptor while you wash the filters or pellet
transcriptional processing is required for receptor the complex.
function, these systems may not be appropriate.
2. Was the separation of bound and free drug per- 2.03.3.5.2 Performing the experiment
formed appropriately? In particular was the time The experiment is done with a single concentration
of separation adequate for the Kd of the receptor– of radioligand, usually equal to the Kd concentration
ligand interaction (see Table 1). of that ligand for the receptor as determined in pre-
3. Was nonspecific binding measured correctly? vious studies. A higher concentration will increase
Remember the rule of thumb that the the sensitivity of the assay and decrease counting
42 Basic Principles

error but will also increase nonspecific binding and 120


time to equilibrium. 81-fold range
As with direct binding experiments, the incuba- 100
tion should reach equilibrium. However, the

Specific binding
80
reaction is more complicated in the presence of
an inhibitor, often at high concentrations. To 60
ensure that a steady state has been reached, the
incubation proceeds for 4–5 times the half-life of 40
the radioligand for receptor dissociation as deter-
20
mined in an off-rate experiment. In order to have a
complete profile of the competition, typically 0
12–24 concentrations of unlabeled compound span- 0.1 1 10 100 1000
ning about six orders of magnitude are examined. Log(Competitor)
Figure 10 Analyzing assumptions of the law of mass
2.03.3.5.3 Analyzing competitive binding action. The law of mass action predicts that the
data concentration of competitor that results in 90% and 10%
Visual inspection of competition of radioligand bind- inhibition cover a 81-fold (roughly two orders of magnitude)
range.
ing reveals a hyperbolic curve, with the most potent
ligands inhibiting specific binding at lower concen- binding curve is determined by the law of mass
tration than less potent chemicals (Figure 9). The action. As shown in Figure 10, the curve descends
plateau at the top of the curve, the radioligand bind- from 90% specific binding to 10% specific binding
ing in the absence of the competing unlabeled drug, with an 81-fold increase in the concentration of the
represents total binding. Note that total binding is unlabeled drug. More simply, nearly the entire curve
not the same as Bmax since the receptor is not fully will cover two log units (100-fold change in concen-
saturated (if using a Kd concentration of radioligand). tration). This generalization can be determined in
The bottom of the curve is a plateau equal to non- terms of [L], Kd, and the fractional saturation, Y.
specific binding (NS) with the difference between the
top and bottom plateaus being specific binding. The ½L

IC50 is the concentration of unlabeled drug that Kd þ ½L
blocks half the specific binding.
If the labeled and unlabeled ligands compete for a The fractional occupancy at 90% and 10% satura-
single binding site, the steepness of the competitive tion will be
120 S0:9 S0:1
0:9 ¼ and 0:1 ¼
Kd þ S0:9 Kd þ S0:1
100 Total
Solving these equations simultaneously, and introdu-
Specific binding

80
cing the term cooperativity index (Rs), or the change
60 in drug concentration from 90% to 10%:
40 0:9
S0:9 0:1
Rs ¼ ¼ 0:1K ¼ 81
20 S0:1 0:9Kd
Nonspecific d
0
0.1 1 10 100 1000 When the interaction between competitor and
Log(Competitor) receptor does not proceed via the law of mass
Figure 9 Competition binding experiments. Total binding action, the value of Rs will not equal 81 (positive
of a single concentration radioligand is examined following cooperativity Rs<81; negative cooperativity
incubation with graded concentrations of competitor. Rs >81).
Shown are four competitors with different affinity for the
receptor. The plateau at the top of the curve represents total
Competitive binding curves are described by this
binding whereas the bottom plateau is nonspecific binding. equation:
The data may be analyzed using nonlinear regression to
Total – NS
Total – NS
calculate IC50 values. ½LRI ¼ NS þ . ½LRI ¼ NS þ ð9Þ
1 þ 10log½I – log IC50 1 þ 10log½I – log IC50
Receptor Theory and the Ligand–Macromolecule Complex 43

where [LR]I (y-axis) is the amount of binding mea- of ligand. All the analyses presented so far are based on
sured at each [I] (x-axis). Nonlinear regression to fit the assumption that a very small fraction of the ligand
your competitive binding curve to determine the binds to receptors (or to nonspecific sites), so you can
log(IC50). In order to determine the best-fit value of assume that the free concentration of ligand is approxi-
IC50, the nonlinear regression problem must deter- mately equal to the concentration added. In some
mine the 100% (total) and 0% (nonspecific) plateaus. experimental situations, the receptors are present in
Alternatively, values of IC50 can also be ascer- high concentration and have a high affinity for the
tained by logit-log plot (or pseudo-Hill). ligand, so that assumption is not valid. A large fraction
of the radioligand binds to receptors so the free con-
½LRI
log ¼ n log½I þ n log IC50 ð10Þ centration of radioligand is considerably lower than the
½LR – ½LRI
concentration added. If radioligand depletion occurs,
where [LR] is the amount of binding in the absence alternative analysis is required. For example, the free
of inhibitor and n is Hill coefficient. Plotting the log concentration of ligand can be measured in every tube.
([LR]I/[LR]–[LR]I) versus [I] will yield a straight Alternatively, the free concentration in each tube is
line with slope equal to n and an x-intercept equal to calculated by subtracting the number of cpm of total
IC50. binding from the cpm of added ligand. One problem
with this approach is that experimental error in deter-
2.03.3.5.4 Calculating the K I from the IC 50 mining specific binding also affects the calculated value
The IC50 value is not equivalent to Kd for the com- of free ligand concentration.
petitor (or Ki) and is dependent on the amount of
radioligand in the incubation. Therefore, the IC50
value can vary between studies whereas Ki is a con-
stant. The Ki can be calculated from IC50 using the
2.03.4 Quantification of Receptor
Cheng and Prusoff method:
Responses
2.03.4.1 Why Examine Receptors Based
IC50
Ki ¼ ½L 
ð11Þ on Responses?
1þ Kd
The interaction between the receptor and ligand is
where Ki is the equilibrium dissociation constant for the first step in the elicitation of a biological response.
the inhibitor and Kd is the equilibrium dissociation In fact, there are three components of a receptor
constant for the radioligand L. Several assumptions system: the ligand, the receptor, and the effector (E)
were made in the derivation of the Cheng and (see pathway below). The effector may be an
Prusoff equation that must be met. enzyme, an ion, or a transcription factor and it trans-
1. The radioligand and the inhibitor must interact mits the biophysical interaction of ligand and
with the receptor according to the law of mass receptor into a biochemical or molecular signal.
action. That is, the binding of both chemicals Physiological receptors are linked to the signal trans-
should be reversible and directed at a single popu- duction apparatus of the cell. Ultimately, the dose–
lation of R. Whether the law of mass action has response of a cell to a ligand is more complicated
been met can be determined from an indirect Hill than predicted by direct binding assays. While the
plot where the slope equals –1. ultimate response is proportional to the amount of
2. The concentration of L added should equal the ligand–receptor complex formed, direct proportion-
amount free. There should not be ligand depletion ality may not be observed. Therefore, it is often
([LR]<10%) and the concentration of receptor is difficult to determine Kd values for a ligand–receptor
much less than Kd. interaction based solely on the response of that cell.
3. The incubation must have reached equilibrium k1 k2
k3
! !
for radioligand and all concentrations of the LþR  LR þ E  LRE! Effect
k–1 k–2
competitor.
Beyond adding complexity, the effector system ampli-
fies the response. Therefore, detection of a receptor-
2.03.3.6 Ligand Depletion
mediated event is often easier examined than direct
The equations that describe the law of mass action binding per se. This is the major reason why receptors
include the variable [L] which is the free concentration are often characterized by their responses prior to the
44 Basic Principles

implementation of direct binding assays. Sensitive values. Both EC50 and IC50 are dependent on the
assays for detecting influx or efflux of Naþ, Kþ, nuances of the incubation conditions whereas Kd is
Ca2þ, or Cl, activation of adenylate cyclase, phos- only dependent on the receptor and its ligand.
phorylation of a receptor or effectors, and alterations However, there are many assumptions that must be
in mRNA expression have been developed. In addi- made in order to determine a Kd from a cellular
tion, binding of a chemical to a receptor says nothing response.
about the response elicited by the ligand. That is,
1. The response of the tissue or cell should be due solely to the
antagonists and agonists may bind with the same affi-
interaction of an agonist with one type of receptor. As
nity to a receptor (in fact antagonists are often more
such, the response on which Kd values are based
avidly associated) but are coupled to the effector
should not be a composite on effects of two or
molecular in a different manner. Therefore, receptor
more distinct receptor populations nor should it
responses are of more physiological relevance than
be the result of nonspecific or nonreceptor-
direct binding assays in isolation.
mediated effects of the agonist.
Prior to the advent of modern molecular biology,
2. The altered sensitivity to an agonist observed in the
which has allowed for cloning of receptors without
presence of an antagonist should be due to competition
knowledge of their function or ligands, the existence
for a shared recognition site. This holds true for
of a cognate receptor for a particular chemical was
pharmacological and toxicological antagonism of
based on cellular responses. Some of the criteria that
the competitive and irreversible variety (see
led investigators to believe that a given effect was in
Section 2.03.4.4).
fact receptor-mediated included:
3. The response obtained following addition of agonist
1. Specificity of response. It is often the specificity of a should be measured at a time when the maximal response
drug, hormone, or xenobiotic that persuades an is elicited. Biological preparations especially suited
investigator that the effects are receptor-mediated. for determination of Kd values for receptor–ligand
Rank order of potency for effect of a series of interactions are those that maintain a maximal
agonists should be characteristic of a particular response over a reasonable length of time.
receptor. 4. When agonists or competitive antagonists are added to
2. Existence of antagonists. Antagonists can selectively the tissue incubation, the concentration of ligand free in
block receptor-mediated events. The order of solution should be maintained at a known level. Tissue
potency of antagonists should be characteristic of uptake, degradation, and other losses must be pre-
a particular receptor. vented or overcome by continual re-addition of
3. Protection against irreversible receptor blockade. In the ligand.
presence of a ligand, the irreversible antagonist 5. The experimental design should contain proper controls
cannot reach its site of action. to permit measurement of, or correction for, any changes
4. Cross-desensitization. If you can downregulate the in tissue sensitivity over time. Internalization of a
receptor, ligands will have less of an effect. membrane-bound receptor, or proteolysis of solu-
Transgenic knockouts can be viewed as the ulti- ble receptors within the time frame of the
mate desensitizer. experiments may alter the sensitivity of the cell
to the ligand.
Examination of receptor responses offers a mechan-
ism by which the physiological relevance of a ligand–
receptor interaction can be confirmed. If the four
2.03.4.3 Full Agonists
criteria listed above can be fulfilled, the receptor
system is considered to be properly characterized The determination of a Kd for a full antagonist based
and the chemical(s) under question can be purported on responses is not as straightforward as Clark’s the-
to elicit effects via a cognate protein. ory of occupancy. Complicating factors such as the
existence of spare receptors and efficacy issues will
impact the calculations. Clark’s theory of occupancy
2.03.4.2 Assumptions for Determining Kd
may also be used for examining responses, if examin-
Values for Receptor–Ligand Interactions in
ing a full agonist (efficacy e is 100%) and the receptor
Intact Tissue Preparations
requires full occupancy for maximal response. The
As mentioned above in the context of IC50 values key feature of this formulation is that response is
versus Ki, Kd values are more useful than are EC50 proportional to the number of receptors occupied.
Receptor Theory and the Ligand–Macromolecule Complex 45

k1 efficacy (e) of the drug–receptor complex. There


! ke
LþR  LR! Effect
k–1
are several ways to irreversibly inactivate a receptor,
and for many receptors these agents have been
Thus, if ke (the rate constant for coupling, or of the described. If an irreversible antagonist has not been
effector system) is known, for any given [LR] the characterized, chemical cross-linking may be uti-
extent of the response can be predicted. lized. In this instance, a highly reactive side chain
  such as an –azido group may be added to the ligand.
½L½LRmax
Response ðÞ ¼ f ð½LRÞ ¼ f Upon binding to the receptor and activation with
Kd þ ½L
ultraviolet light, the ligand will covalently attach to
The maximal response max will occur at [LR]max the protein. With peptide ligands, bifunctional
 LR ½L reagents such as disuccinimidyl suberimidate may
¼ ¼ ð12Þ result in irreversible blockade. Nonspecific means
max LRmax Kd þ ½L
such as alkylation or proteolysis have been
As with receptor occupancy, the half-maximal employed. All means of irreversible inhibition must
response will occur at [L] equal to Kd. If it is assumed affect the receptor complex and not affect the effec-
that occupancy and response are proportional, the tor signaling pathways.
same graphical means to examine Kd used in receptor Substituting eqn [13] into that of [12] results in
binding may be employed. That is, a plot of fractional the following relationship:
response (/max) versus log[agonist] will result in  
a hyperbolic curve whereas log (/(max–)) ver-  ½L ½L
¼ fe ¼ f " LRmax  ð14Þ
sus log[agonist] represents a straight line. max Kd þ ½L Kd þ ½L
However, as mentioned in Section 2.03.2, this If the receptor preparation is treated with an irrever-
simple relationship between occupancy and effect sible antagonist, the concentration of total active
does not adequately predict many biological receptors is reduced to a fraction (q) of the original
responses. In particular, dose–response relationships LRmax. Since efficacy is equal to "LRmax, in the pre-
to drugs and other chemicals tend to have a steeper sence of irreversible blockade, e¼q"LRmax, and the
slope than predicted and the existence of spare following equation is derived:
receptors shows that maximal responses do not  
require full occupancy. 9 ½L9 ½L9
¼ fe ¼ f " qLRmax  ð15Þ
Stephenson and Furchgott formalized a nonlinear 9max Kd þ ½L9 Kd þ ½L9
relationship between receptor occupancy and biolo- where 9, 9max, [L9] represent values after irrever-
gical response: sible blockade of the receptor. When comparing
LR equal responses before and after inhibition, for exam-
Response ¼ f ðSÞ ¼ fe
LRmax ple EC50 values, it is assumed that the stimulus
Intrinsic efficacy ð"Þ : e ¼ "LRmax ð13Þ applied to the tissue is the same (S¼S9).
LR ½L ½L9
Response ¼ f ð"LRmax x Þ ¼ f ð"½LRÞ "LRmax ¼ "qLRmax
LRmax Kd þ ½L Kd þ ½L9
eliminating "LRmax ð16Þ
where e is efficacy, " is intrinsic efficacy, and S is the
½L ½L9
stimulus applied to the tissue or cell. The introduc- ¼q
Kd þ ½L Kd þ ½L9
tion of the term " effectively divided efficacy (e) into
two components: a ligand-dependent (e) and a tissue- The advantage of comparing equal responses is that
dependent (LRmax) term. the unknown LRmax is eliminated and the function f
In order to use this relationship to examine Kd for relating occupancy to response is no longer required.
a ligand–receptor interaction, the experiments are Equation [16] can be rearranged to the linear
similar to those used to determine the spare receptor equation:
reserve (discussed in Section 2.03.2.4). Dose–
½L9 1 ½L9ð1 – qÞ
response data for a particular agonist before and ¼ þ ð17Þ
½L q Kd  q
after irreversible receptor blockade is obtained. In
effect, what these experiments are doing is changing A plot of [L9]/[L] versus [L9] results in a straight
LRmax in a preparation, and hence altering the line with a y-intercept equal to 1/q and a slope
46 Basic Principles

1.0 12

0.8 Control 10
I1

8
0.6 I2
Δ/Δmax

1−q

L/L⬘
slope =
6 Kd •q
0.4 I3 y − intercept
Kd =
4 slope
0.2
I4 1
2 y − intercept =
q
0.0
1 10 100 1000 0 200 400 600 800 1000
L1 L2 L1⬘ L3 L2⬘ L3⬘
[L⬘]
[Agonist]
Figure 11 Determination of Kd for receptor–agonist interactions using irreversible receptor blockade.

ð1 – qÞ binds to the receptor at a site other than the


equal to . The Kd value can be calculated as
Kd q agonist-binding pocket, yet binding precludes
(y-intercept – 1)/slope (see Figure 11). the binding of the agonist. This is often caused
by the antagonist producing a conformational
change in the binding packet.
2.03.4.4 Quantitation of Pharmacologic/ 5. Mixed antagonism A combination of any of the
Toxicologic Antagonism types of antagonism listed above.

The use of antagonists to block receptor responses As stated above, antagonism is an important mechan-
represents a classical approach in pharmacology and ism by which drugs and other chemicals exert
a major mechanism by which chemicals cause toxi- beneficial responses and how certain toxicants produce
city. There are five basic types of antagonism: their hazardous effects. Therefore, it is appropriate to
use the methods described below to characterize the
1. Functional antagonism. This type of inhibition often chemical and determine which type of inhibition is
results from stimulation of two pathways with occurring. Knowing the mechanism of action may aid
opposite function (i.e., sympathetic versus para- in designing more potent drugs or in ameliorating
sympathetic nervous systems). Functional toxicity. However, antagonists are useful tools to char-
antagonism results from action via two distinct acterize the receptor as well. This will be the focus of
receptors and cannot be examined by the methods the present section. To characterize a receptor using
described herein. antagonists, such interaction must fulfill certain
2. Competitive antagonism. A competitive antagonist is
requirements:
binding to the receptor in roughly the same phy-
sical space as the agonist, precluding the latter 1. Blockade must be selective for the family of ago-
from interacting with the receptor to elicit a bio- nists in this receptor type.
logical response. A competitive antagonist is 2. Blockade of responses in different tissues by a
reversible, with a rate of dissociation that is rele- series of antagonists should result in a similar
vant to the time frame of the experiments. rank order of potency.
3. Irreversible antagonism. As with the competitive 3. Blockade should be sufficiently rapid to allow for
antagonist, the irreversible antagonist binds to precise quantitative measurement.
the same region of the receptor as the agonist. 4. An identical value for Ki in antagonizing different
However, in this instance, the rate of dissociation agonists that act at the same receptor should be
is extremely slow or practically nonexistent. obtained.
4. Noncompetitive antagonism. Receptors are compli- 5. Values for the dissociation constants Ki derived from
cated macromolecules that may interact with studies assessing tissue response should be identical
small molecules in a variety of ways. A noncom- to antagonist dissociation constants obtained in stu-
petitive antagonist is defined as a chemical that dies of binding to a particular type of receptor.
Receptor Theory and the Ligand–Macromolecule Complex 47

2.03.4.4.1 Functional antagonism ½L


 LR ½LR Kd
Functional antagonism is defined as antagonism of ¼ ¼ ¼ ð18Þ
max LRmax ½R þ ½LR þ ½IR 1 þ ½L þ ½I
tissue response that is unrelated to blockade at Kd KI

drug receptors but instead represents blockade of This can be rearranged (divide by [L]/Kd):
tissue response at a site distal to receptors. This
precludes its precise quantitation using the meth-  1
¼   ð19Þ
ods described. It may affect second messengers or max 1 þ Kd 1 þ ½I
L Ki
depress cellular excitability and/or energy status.
Alternatively, it may produce an opposing action As you can see, the extent of antagonism depends on
through a different receptor. the agonist and antagonist concentration, as well as
their dissociation constants, Kd and Ki. In the presence
2.03.4.4.2 Competitive antagonism of competitive inhibitor I, fractional occupation
Competitive antagonists are ligands that compete [LR]/[LR]max decreases as a consequence of an
with agonists, usually for a common binding site in increase in the apparent equilibrium dissociation
a receptor. The interaction of agonist (L), competi- constant of the agonist L. This decrease is from a
tive antagonist (I) with receptor (R), is described value of Kd in the absence of inhibitor to a value of KD
using the following scheme. (1þ[I]/Ki) in the presence of I. The increase in
apparent dissociation constant yields an equation
I with the same shape dose–response curve, only
þ K!
d Ke
shifted to the right with no effect on maximal
Lþ  LR! Effect response (see Figure 12).
R
Since the competitive antagonist acts by excluding
Ki "#
the agonist from binding, it does not change the rela-
IR tionship between occupation and response. At equal
fractional occupancy, equal responses are observed.
Note that the inhibitor–receptor complex is ineffec- When /max in the absence of competitor equals
tual, and cannot couple occupancy with a biological 9/max of the presence of competitor I, then
response. The formation of the ligand–receptor and
½L ½L9
the inhibitor–receptor complex may be described by ¼   ð20Þ
½L þ Kd ½L9 þ Kd 1 þ ½I
the Clark equations and have equilibrium dissocia- K i
tion constants of Kd and Ki respectively. As a result of
IR formation, there are fewer receptors available for Where L and L9 are the concentration required to
LR formation. generate equivalent responses /max and

(a) (b)
Raw data Schild plot
1.0 10
No inhibitor
I = 50
I = 500
0.8 I = 1000
8
Log(x−1)
Δ/Δmax

0.6 6

0.4 4

0.2 2
−LogKI

0.0 0
0.1 1 L 10 L⬘ 100 1000 10000 0 2 4 6 8 10
Dose agonist −Log inhibitor

Figure 12 Determination of the equilibrium dissociation constant (KI) for a competitive antagonist. (a) The fractional response
was calculated using eqn [18] with a Kd of 10, KI of 10 and varying the inhibitor concentration from 0 to 1000. (b) Schild plot
with log(dose ratio1) versus concentration of inhibitor. The KI may be calculated from either the x- or y-intercept.
48 Basic Principles

9/max in the absence and presence of I. This 2.03.4.4.3 Noncompetitive antagonism


equation can be simplified: A noncompetitive antagonist interacts with the
receptor, but this interaction takes place at a site
½L9 ½I
–1 ¼ different than that of the agonist. Upon binding to
½L Ki
the noncompetitive antagonist, the affinity of the
And by taking the logarithms of this equation and receptor for the agonist is altered, possible the result
substituting x for [L9]/[L], the Schild equation is of a conformational change in the protein structure.
derived: The interaction of agonist (L), noncompetitive
antagonist (I) with receptor (R), is described using
logðx – 1Þ ¼ log½I – log Ki ð21Þ
the following scheme.
Note that the Kd is removed from the equation and
I I
hence is not required to calculate Ki. The term dose þ Kd þ Ke
ratio (x) is applied to the expression [L]/[L9]. Dose L þ R $ LR ! Effect
ratios depend on the [I] and Ki. (This approach Ki "# Ki "#
assumes that the dose ratio is independent of the IR $ LIR
agonist used and that a single receptor type is being An antagonist of this sort produces quantitative
examined.) The determination of dose-ratios is iden- changes in the agonist dose–response curves: the max-
tical to that described above for full agonists (Section imal response is decreased while the EC50 of the
2.03.4.3), in which concentrations in the presence and
agonist does not change (Figure 14). The antagonist
absence of inhibitor result in equivalent responses.
does not alter the concentration of bound agonist
The Schild equation does not assume there is a linear
(LRþLRI) but the LRI complex is nonfunctional.
relationship between occupancy and effect. However,
The response of the agonist may be derived as follows:
the Lineweaver–Burke plot does. Therefore it is not
appropriate to calculate a KI, but is of diagnostic value.  ke ½LR ke ½LR
¼ ¼
As shown in Figure 13, double reciprocal plots in the max ½RTotal ½R þ ½LR þ ½IR þ ½ILR
presence of increasing amounts of competitive inhibi- Which can be rearranged to : ð22Þ
tor intersect at 1/Bmax. Non-competitive inhibitors
 ke ½L
will result in a plot with the intersection occurring at ¼  
max ð½L þ Kd Þ 1 þ ½I
–1/Kd, as discussed below. KI

Competitive Noncompetitive

100

80

60
1/Δ/Δmax

40

20

−0.25 0.00 0.25 0.50 0.75 1.00 1.25 1.50 1.75 −0.25 0.00 0.25 0.50 0.75 1.00 1.25 1.50 1.75
1/[Ligand] 1/[Ligand]
Figure 13 Comparison of double reciprocal plots for competitive and noncompetitive antagonists.
Receptor Theory and the Ligand–Macromolecule Complex 49

1.0 I=0
I=1
0.8

I=5
Δ/Δmax

0.6
KI = IC50
I = 10
0.4

0.2
I = 50
I = 100
0.0
0.1 1 10 100 1000 0.1 1 10 100 1000 10000
Log agonist Log antagonist
Figure 14 Determination of the equilibrium dissociation constant (KI) for a noncompetitive antagonist.

The response is decreased by a factor whose magni- 2.03.4.5 Partial Agonists


tude is increased by [I] and low values of Ki (i.e., high
A partial agonist has affinity, but less than maximal
affinity). For noncompetitive antagonists Ki¼IC50,
efficacy. The only way to determine the Kd for a
the concentration of antagonist that inhibits the
partial agonist (designated Kdp) is to compare it to
response by 50%.
a full agonist (Figure 15). However, many assump-
tions will be made to simplify the comparisons.
2.03.4.4.4 Irreversible and First, it is assumed that the concentration of full
pseudoirreversible antagonists agonist (A) resulting in a response is small relative
Some types of antagonists produce irreversible to Kda. We can therefore simplify eqn [2], for a full
antagonism of agonist-mediated responses. This is agonist to
most commonly observed when an antagonist pro-
duces a covalent modification in a receptor. ½A
YA ¼
Irreversible antagonists will yield curves that are Kda
identical to noncompetitive antagonist if the
response is proportional to the number of receptors For a partial agonist, eqn [2] cannot be simplified and
occupied. (However, remember spare receptors.) remains
Note that if the k1 is sufficiently slow, competitive ½P
antagonists may appear to be irreversible. Yp ¼
½P þ Kdp
Noncompetitive, irreversible, and pseudoirreversible
antagonism share a common property in that they are
nonsurmountable antagonists, that is, increased ago- To determine the Kdp of a partial agonist, a dose–
nist does not cause maximal response. response curve for the full agonist A and the partial
agonist P is obtained. As is the case in the previous
examples, comparisons of [A] and [P] that elicit equal
2.03.4.4.5 Mixed antagonism responses are used.
Mixed antagonism is observed when antagonists have
multiple types of interaction with receptor systems. S ¼ eA Ya
For example, the antagonist may initially block S ¼ eP Y p
receptors competitively but over time may become
At equal response
irreversible. Two other possibilities exist if the LRI
complex is able to produce a response or if the KI for ½A ½P
eA ¼ eP
R does not equal that of LR. Kda ½P þ Kdp
50 Basic Principles

1.0 12

0.8 Full agonist 10


slope
Kdp =
y − intercept

1/[Full agonist]
8
0.6
Δ/Δmax

Partial agonist
6
0.4

4
0.2
2
0.0
1 10 100 1000 0 200 400 600 800 1000
A1 A2 P3 P2
[Agonist] 1/[Partial agonist]
Figure 15 Determination of the equilibrium dissociation constant (Kdp) for a partial agonist.

which can be arranged to the following linear disciplines including enzymology (Michaelis com-
equation plex between enzyme and substrate), immunology
  (formation of antibody–antigen complex), pharma-
1 eA Kdp 1 eA
¼ þ ð23Þ cology (drug–receptor complex), and toxicology
½A eP Kda ½P eP Kda
(xenobiotic–receptor complex). The shape of the
Thus, a plot of 1/[A] versus 1/[P] should be linear dose–response curve, the most important relation-
eA Kdp ship examined in toxicology, is dictated by receptor
with the slope equal to and a y-intercept of
eP Kda theory. Therefore, by applying the equations
eA described herein, taking into account all assumptions
. The Kdp can be determined by dividing the
eP Kda and limitations, the estimation of risk posed by che-
slope by the y-intercept. It is important to stress that mical exposure may be more accurately determined.
an assumption was made regarding the Kda versus the
concentration of a full agonist that results in a
response. If this is not the case, the mathematics References
become much more difficult (see Limbird 1996).
Limbird, L. E. Cell Surface Receptors: A Short Course on Theory
and Methods, 2nd ed.; Kluwer Academic Publishers:
Boston, MA, 1996; pp 238.
2.03.5 Conclusion Levitzki, A. Receptors: A Quantitative Approach, Benjamin/
Cummings Pub. Co.: Menlo Park, CA, 1984; pp xv, 142.
The quantification of the interaction between a Taylor, P.; Insel, P. A. In Principles of Drug Action: The Basis of
Pharmacology; Pratt, W. B., Taylor, P., Goldstein, A., Ed.,
macromolecule (receptor) and a xenobiotic (ligand) Churchill Livingstone: New York, 1990; pp 103–200.
is called receptor theory and is often overlooked in
modern molecular toxicology. The proper use of
receptor theory can be used to examine structure–
activity relationships and modifications of chemicals Relevant Websites
to fit the active site of the macromolecule. Also, the
basic tools of quantification and characterization of a http://www.graphpad.com – Introduction to nonlinear
bimolecular interaction are applicable to many regression, Copyright 1999 by GraphPad Software, Inc.

You might also like