Ligand binding assay

Last updated

A ligand binding assay (LBA) is an assay, or an analytic procedure, which relies on the binding of ligand molecules to receptors, antibodies or other macromolecules. [1] A detection method is used to determine the presence and extent of the ligand-receptor complexes formed, and this is usually determined electrochemically or through a fluorescence detection method. [2] This type of analytic test can be used to test for the presence of target molecules in a sample that are known to bind to the receptor. [3]

Contents

There are numerous types of ligand binding assays, both radioactive and non-radioactive. [4] [5] [6] As such, ligand binding assays are a superset of radiobinding assays, which are the conceptual inverse of radioimmunoassays (RIA). Some newer types are called "mix-and-measure" assays because they do not require separation of bound from unbound ligand. [5]

Ligand binding assays are used primarily in pharmacology for various demands. Specifically, despite the human body's endogenous receptors, hormones, and other neurotransmitters, pharmacologists utilize assays in order to create drugs that are selective, or mimic, the endogenously found cellular components. On the other hand, such techniques are also available to create receptor antagonists in order to prevent further cascades. [7] Such advances provide researchers with the ability not only to quantify hormones and hormone receptors, but also to contribute important pharmacological information in drug development and treatment plans. [8]

History

Historically, ligand binding assay techniques were used extensively to quantify hormone or hormone receptor concentrations in plasma or in tissue. The ligand-binding assay methodology quantified the concentration of the hormone in the test material by comparing the effects of the test sample to the results of varying amounts of known protein (ligand).

The foundations for which ligand binding assay have been built are a result of Karl Landsteiner, in 1945, and his work on immunization of animals through the production of antibodies for certain proteins. [9] Landsteiner's work demonstrated that immunoassay technology allowed researchers to analyze at the molecular level. The first successful ligand binding assay was reported in 1960 by Rosalyn Sussman Yalow and Solomon Berson. [9] They investigated the binding interaction for insulin and an insulin-specific antibody, in addition to developing the first radioimmunoassay (RIA) for insulin. These discoveries provided precious information regarding both the sensitivity and specificity of protein hormones found within blood-based fluids. [9] Yalow and Berson received the Nobel Prize in Medicine as a result of their advancements. Through the development of RIA technology, researchers have been able to move beyond the use of radioactivity, and instead, use liquid- and solid-phase, competitive, and immunoradiometric assays. [9] As a direct result of these monumental findings, researchers have continued the advancement of ligand binding assays in many facets in the fields of biology, chemistry, and the like. For instance, the Lois lab at Caltech is using engineered artificial ligands and receptors on neurons to trace information flow in the brain. [10] They are specifically using ligand-induced intramembrane proteolysis to unravel the wiring of the brain in Drosophila and other models. [11] When the artificial ligand on one neuron binds to the receptor on another, GFP expression is activated in the acceptor neuron demonstrating the usefulness of ligand binding assays in neuroscience and biology. [12]

Applications

Ligand binding assays provide a measure of the interactions that occur between two molecules, such as protein-bindings, as well as the degree of affinity (weak, strong, or no connection) for which the reactants bind together. [13] Essential aspects of binding assays include, but are not limited to, the concentration level of reactants or products (see radioactive section), maintaining the equilibrium constant of reactants throughout the assay, and the reliability and validity of linked reactions. [13] Although binding assays are simple, they fail to provide information on whether or not the compound being tested affects the target's function. [14]

Radioligand assays

Radioligands are used to measure the ligand binding to receptors and should ideally have high affinity, low non-specific binding, high specific activity to detect low receptor densities, and receptor specificity. [7]

Levels of radioactivity for a radioligand (per mole) are referred to as the specific activity (SA), which is measured in Ci/mmol. [15] The actual concentration of a radioligand is determined by the specific stock mix for which the radioligand originated (from the manufactures.) [15] The following equation determines the actual concentration:

[15]

Saturation binding

Saturation analysis is used in various types of tissues, such as fractions of partially purified plasma from tissue homogenates, cells transfected with cloned receptors, and cells that are either in culture or isolated prior to analysis. [7] Saturation binding analysis can determine receptor affinity and density. It requires that the concentration chosen must be determined empirically for a new ligand.

There are two common strategies that are adopted for this type of experiment: [7] Increasing the amount of radioligand added while maintaining both the constant specific activity and constant concentration of radioligand, or decreasing the specific activity of the radioligand due to the addition of an unlabeled ligand. [7]

Scatchard plot

An example of a Scatchard plot Scatchardthree.gif
An example of a Scatchard plot

A Scatchard plot (Rosenthal plot) can be used to show radioligand affinity. In this type of plot, the ratio of Bound/Free radioligand is plotted against the Bound radioligand. The slope of the line is equal to the negative reciprocal of the affinity constant (K). The intercept of the line with the X axis is an estimate of Bmax. [7] The Scatchard plot can be standardized against an appropriate reference so that there can be a direct comparison of receptor density in different studies and tissues. [7] This sample plot indicates that the radioligand binds with a single affinity. If the ligand were to have bound to multiple sites that have differing radioligand affinities, then the Scatchard plot would have shown a concave line instead. [7]

Nonlinear curve fitting

Nonlinear curve-fitting programs, such as Equilibrium Binding Data Analysis (EBDA) and LIGAND, are used to calculate estimates of binding parameters from saturation and competition-binding experiments. [16] EBDA performs the initial analysis, which converts measured radioactivity into molar concentrations and creates Hill slopes and Scatchard transformations from the data. The analysis made by EBDA can then be used by LIGAND to estimate a specified model for the binding. [16]

Competition binding

Competition binding is used to determine the presence of selectivity for a particular ligand for receptor sub-types, which allows the determination of the density and proportion of each sub-type in the tissue. [7] Competition curves are obtained by plotting specific binding, which is the percentage of the total binding, against the log concentration of the competing ligand. [7] A steep competition curve is usually indicative of binding to a single population of receptors, whereas a shallow curve, or a curve with clear inflection points, is indicative of multiple populations of binding sites. [16]

Non-radioactive binding assays

Despite the different techniques used for non-radioactive assays, they require that ligands exhibit similar binding characteristics to its radioactive equivalent. Thus, results in both non-radioactive and radioactive assays will remain consistent. [5] One of the largest differences between radioactive and non-radioactive ligand assays are in regards of dangers to human health. Radioactive assays are harmful in that they produce radioactive waste; whereas, non-radioactive ligand assays utilize a different method to avoid producing toxic waste. These methods include, but are not limited to, fluorescence polarization (FP), fluorescence resonance energy transfer (FRET), and surface plasmon resonance (SPR). In order to measure process of ligand-receptor binding, most non-radioactive methods require that labeling avoids interfering with molecular interactions. [5]

Fluorescence polarization

Fluorescence polarization (FP) is synonymous with fluorescence anisotropy. This method measures the change in the rotational speed of a fluorescent-labeled ligand once it is bound to the receptor. [5] Polarized light is used in order to excite the ligand, and the amount of light emitted is measured. [5] Depolarization of the emitted light depends on ligand being bound (e.g., to receptor). If ligand is unbound, it will have a large depolarization (ligand is free to spin rapidly, rotating the light). If the ligand is bound, the combined larger size results in slower rotation and therefore, reduced depolarization. [5] An advantage of this method is that it requires only one labeling step. However, this method is less precise at low nanomolar concentrations. [5]

Kinetic exclusion assay

Kinetic exclusion assay (KinExA) measures free (unbound) ligand or free receptor present in a mixture of ligand, receptor, and ligand-receptor complex. The measurements allow quantitation of the active ligand concentration and the binding constants (equilibrium, on and off rates) of the interaction. [17]

Fluorescence resonance energy transfer

Jablonski diagram of FRET FRET-Jablonski2 English.png
Jablonski diagram of FRET

Fluorescence Resonance Energy Transfer (FRET) utilizes energy transferred between the donor and the acceptor molecules that are in close proximity. [5] FRET uses a fluorescently labeled ligand, as with FP. [5] Energy transfer within FRET begins by exciting the donor. [5] The dipole–dipole interaction between the donor and the acceptor molecule transfers the energy from the donor to the acceptor molecule. [5] If the ligand is bound to the receptor-antibody complex, then the acceptor will emit light. [5] When using FRET, it is critical that there is a distance smaller than 10 nm between the acceptor and donor, in addition to an overlapping absorption spectrum between acceptor and donor, and that the antibody does not interfere or block the ligand binding site. [5]

Surface plasmon resonance

Surface Plasmon Resonance (SPR) configuration Surface Plasmon Resonance (SPR).jpg
Surface Plasmon Resonance (SPR) configuration

Surface Plasmon Resonance (SPR) does not require labeling of the ligand. [5] Instead, it works by measuring the change in the angle at which the polarized light is reflected from a surface (refractive index). [5] The angle is related to the change in mass or layer of thickness, such as immobilization of a ligand changing the resonance angle, which increases the reflected light. [5] The device for which SPR is derived includes a sensor chip, a flow cell, a light source, a prism, and a fixed angle position detector. [5]

Liquid-phase binding assays

Immunoprecipitation

The liquid-phase ligand binding assay of immunoprecipitation (IP) is a method that is used to purify or enrich a specific protein, or a group of proteins, using an antibody from a complex mixture. The extract of disrupted tissue or cells is mixed with an antibody against the antigen of interest, which produces the antigen-antibody complex. [18] When antigen concentration is low, the antigen-antibody complex precipitation can take hours or even days and becomes hard to isolate the small amount of precipitate formed. [18]

The enzyme-linked immunosorbent assay (ELISA) or Western blotting are two different ways that the purified antigen (or multiple antigens) can be obtained and analyzed. This method involves purifying an antigen through the aid of an attached antibody on a solid (beaded) support, such as agarose resin. [19] The immobilized protein complex can be accomplished either in a single step or successively. [19]

IP can also be used in conjunction with biosynthetic radioisotope labeling. Using this technique combination, one can determine if a specific antigen is synthesized by a tissue or by a cell. [18]

Solid-phase binding assays

Multiwell plate

Multiwell plates-set Multiwell cell culture plates-set.jpg
Multiwell plates-set

Multiwell plates are multiple petri dishes incorporated into one container, with the number of individual wells ranging from 6 to over 1536. Multiwell Plate Assays are convenient for handling necessary dosages and replicates. [20] There are a wide range of plate types that have a standardized footprint, supporting equipment, and measurement systems. [20] Electrodes can be integrated into the bottom of the plates to capture information as a result of the binding assays. [9] The binding reagents become immobilized on the electrode surface and then can be analyzed. [9]

The multiwell plates are manufactured to allow researchers to create and manipulate different types of assays (i.e., bioassays, immunoassays, etc.) within each multiwell plate. [20] Due to the variability in multiwell plate formatting, it is not uncommon for artifacts to arise. Artifacts are due to the different environments found within the different wells on the plate, especially near the edges and center of the wells. Such effects are known as well effects, edge effects, and plate effects. Thus, emphasizing the necessity to position assay designs in the correct manner both within, and between, each plate. [20]

The use of multiwell plates are common when measuring in vitro biological assay activity, or measuring immunoreactivity through immunoassays. [20] Artifacts can be avoided by maintaining plate uniformity by applying the same dose of the specific medium in each well, in addition to maintaining atmospheric pressure and temperature rates in order to reduce humidity. [20]

On-bead binding

On-Bead Ligand Binding assays are isolation methods for basic proteins, DNA/RNA or other biomolecules located in undefined suspensions and can be used in multiple biochromatographic applications. Bioaffine ligands are covalently bound to silica beads with terminal negatively charged silanol groups or polystyrene beads and are used for isolation and purification of basic proteins or adsorption of biomolecules. After binding the separation is performed by centrifugation (density separation) or by magnetic field attraction (for magnetic particles only). The beads can be washed to provide purity of the isolated molecule before dissolving it by ion exchange methods. Direct analyzation methods based on enzymatic/fluorescent detection (e.g. HRP, fluorescent dye) can be used for on-bead determination or quantification of bound biomolecules. [21] [22] [23]

On-column binding

Filter

Filter assays are a solid-phase ligand binding assay that use filters to measure the affinity between two molecules. In a filter binding assay, the filters are used to trap cell membranes by sucking the medium through them. [8] This rapid method occurs at a fast speed in which filtration and a recovery can be achieved for the found fraction. [24] Washing filters with a buffer removes residual unbound ligands and any other ligands present that are capable of being washed away from the binding sites. [8] The receptor-ligand complexes present while the filter is being washed will not dissociate significantly because they will be completely trapped by the filters. [8] Characteristics of the filter are important for each job being done. A thicker filter is useful to get a more complete recovery of small membrane pieces, but may require a longer wash time. [8] It is recommended to pretreat the filters to help trap negatively charged membrane pieces. [8] Soaking the filter in a solution that would give the filter a positive surface charge would attract the negatively charged membrane fragments. [8]

Real-time cell-binding

In this type of assay the binding of a ligand to cells is followed over time. The obtained signal is proportional to the number of ligands bound to a target structure, often a receptor, on the cell surface. Information about the ligand-target interaction is obtained from the signal change over time and kinetic parameters such as the association rate constant ka, the dissociation rate constant kd and the affinity KD can be calculated. [25] By measuring the interaction directly on cells, no isolation of the target protein is needed, which can otherwise be challenging, especially for some membrane proteins. [26] To ensure that the interaction with the intended target structure is measured appropriate biological controls, such as cells not expressing the target structure, are recommended.

Real-time measurements using label-free or label-based approaches have been used to analyze biomolecular interactions on fixated or on living cells. [27] [28]

The advantage of measuring ligand-receptor interactions in real-time, is that binding equilibrium does not need to be reached for accurate determination of the affinity. [29]

Binding specificity

The effects of a drug are a result of their binding selectivity with macromolecule properties of an organism, or the affinity with which different ligands bind to a substrate. [30] More specifically, the specificity and selectivity of a ligand to its respective receptor provides researchers the opportunity to isolate and produce specific drug effects through the manipulation of ligand concentrations and receptor densities. [30] Hormones and neurotransmitters are essential endogenous regulatory ligands that affect physiological receptors within an organism. [30] Drugs that act upon these receptors are incredibly selective in order to produce required responses from signaling molecules. [30]

Specific binding refers to the binding of a ligand to a receptor, and it is possible that there is more than one specific binding site for one ligand. [31] Non specific binding refers to the binding of a ligand to something other than its designated receptor such as various other receptors, or different types of transporters in the cell membrane. [31] For example, various antagonists can bind to multiple types receptors. In the case of muscarinic antagonists, they can also bind to histamine receptors. [31] Such binding patterns are technically considered specific, as the destination of the ligand is specific to multiple receptors. However, researchers may not be focused on such behaviors compared to other binding factors. [31] Nevertheless, nonspecific binding behavior is very important information to acquire. These estimates are measured by examining how a ligand binds to a receptor while simultaneously reacting to a substitute agent (antagonist) that will prevent specific binding to occur. [31]

Specific binding types to ligand and receptor interactions: [30]

Mimics Endogenous EffectsInhibits Endogenous Effects
Agonist Antagonist
Partial Agonist Negative Antagonists (see: Inverse agonist)

Technological advances

Technologies for ligand binding assay continue to advance related to the increasing the speed and to keeping cost-effective procedures while maintaining and increasing the accuracy and sensitivity. [9] Some technological advances include new binding reagents as alternatives to antibodies, [9] alternative dye solutions and micro plate systems, and the development of a method to skip the filtration step, which is required in many ligand binding assay processes. [16]

A prominent signaling molecule in cells is Calcium, (Ca2+), which can be detected with a Fluo-4 acetoxymethyl dye. It binds to free Ca2+ ions, which in turn slightly increase fluorescence of the Fluo-4 AM. [16] The drawback of the Fluo-4 dye formulation is that a washing step is required to remove extracellular dye, which may provide unwanted background signals. For instance, washing puts additional stress on the cells, as well as consumes time, which prevents a timely analysis. [16] Recently, an alternative dye solution and microplate system has been developed called FLIPR® (fluorometric imaging plate reader), which uses a Calcium 3 assay reagent that does not require a washing step. As a result, change in dye fluorescence can be viewed in real time with no delay using an excitatory laser and a charge-coupled device. [16]

Many ligand binding assays require a filtration step to separate bound and unbound ligands before screening. A method called Scintillation proximity assay (SPA) has been recently developed, which eliminates this otherwise crucial step. It works through crystal lattice beads, which are coated with ligand coupling molecules and filled with cerium ions. These give off bursts of light when stimulated by an isotope, which can easily be measured. Ligands are radiolabeled using either 3H or 125I, and released into the assay. Since only the radioligands that directly bind to the beads initiate a signal, free-ligands do not interfere during the screening process. [16]

Limitations

Whole-body PET scan using F-FDG PET-MIPS-anim.gif
Whole-body PET scan using F-FDG

By nature, assays must be carried out in a controlled environment in vitro, so this method does not provide information about receptor binding in vivo. The results obtained can only verify that a specific ligand fits a receptor, but assays provide no way of knowing the distribution of ligand-binding receptors in an organism.

In vivo ligand binding and receptor distribution can be studied using Positron Emission Tomography (PET), which works by induction of a radionuclide into a ligand, which is then released into the body of a studied organism. The radiolabeled ligands are spatially located by a PET scanner to reveal areas in the organism with high concentrations of receptors. [16]

See also

Related Research Articles

<span class="mw-page-title-main">ELISA</span> Method to detect an antigen using an antibody and enzyme

The enzyme-linked immunosorbent assay (ELISA) is a commonly used analytical biochemistry assay, first described by Eva Engvall and Peter Perlmann in 1971. The assay is a solid-phase type of enzyme immunoassay (EIA) to detect the presence of a ligand in a liquid sample using antibodies directed against the protein to be measured. ELISA has been used as a diagnostic tool in medicine, plant pathology, and biotechnology, as well as a quality control check in various industries.

<span class="mw-page-title-main">Immunostaining</span> Biochemical technique

In biochemistry, immunostaining is any use of an antibody-based method to detect a specific protein in a sample. The term "immunostaining" was originally used to refer to the immunohistochemical staining of tissue sections, as first described by Albert Coons in 1941. However, immunostaining now encompasses a broad range of techniques used in histology, cell biology, and molecular biology that use antibody-based staining methods.

<span class="mw-page-title-main">Immunofluorescence</span> Technique used for light microscopy

Immunofluorescence(IF) is a light microscopy-based technique that allows detection and localization of a wide variety of target biomolecules within a cell or tissue at a quantitative level. The technique utilizes the binding specificity of antibodies and antigens. The specific region an antibody recognizes on an antigen is called an epitope. Several antibodies can recognize the same epitope but differ in their binding affinity. The antibody with the higher affinity for a specific epitope will surpass antibodies with a lower affinity for the same epitope.

<span class="mw-page-title-main">Radioimmunoassay</span> Immunoassay that uses radiolabeled molecules

A radioimmunoassay (RIA) is an immunoassay that uses radiolabeled molecules in a stepwise formation of immune complexes. A RIA is a very sensitive in vitro assay technique used to measure concentrations of substances, usually measuring antigen concentrations by use of antibodies.

<span class="mw-page-title-main">Immunoassay</span> Biochemical test for a protein or other molecule using an antibody

An immunoassay (IA) is a biochemical test that measures the presence or concentration of a macromolecule or a small molecule in a solution through the use of an antibody (usually) or an antigen (sometimes). The molecule detected by the immunoassay is often referred to as an "analyte" and is in many cases a protein, although it may be other kinds of molecules, of different sizes and types, as long as the proper antibodies that have the required properties for the assay are developed. Analytes in biological liquids such as serum or urine are frequently measured using immunoassays for medical and research purposes.

Affinity chromatography is a method of separating a biomolecule from a mixture, based on a highly specific macromolecular binding interaction between the biomolecule and another substance. The specific type of binding interaction depends on the biomolecule of interest; antigen and antibody, enzyme and substrate, receptor and ligand, or protein and nucleic acid binding interactions are frequently exploited for isolation of various biomolecules. Affinity chromatography is useful for its high selectivity and resolution of separation, compared to other chromatographic methods.

A protein microarray is a high-throughput method used to track the interactions and activities of proteins, and to determine their function, and determining function on a large scale. Its main advantage lies in the fact that large numbers of proteins can be tracked in parallel. The chip consists of a support surface such as a glass slide, nitrocellulose membrane, bead, or microtitre plate, to which an array of capture proteins is bound. Probe molecules, typically labeled with a fluorescent dye, are added to the array. Any reaction between the probe and the immobilised protein emits a fluorescent signal that is read by a laser scanner. Protein microarrays are rapid, automated, economical, and highly sensitive, consuming small quantities of samples and reagents. The concept and methodology of protein microarrays was first introduced and illustrated in antibody microarrays in 1983 in a scientific publication and a series of patents. The high-throughput technology behind the protein microarray was relatively easy to develop since it is based on the technology developed for DNA microarrays, which have become the most widely used microarrays.

<span class="mw-page-title-main">T-cell receptor</span> Protein complex on the surface of T cells that recognises antigens

The T-cell receptor (TCR) is a protein complex found on the surface of T cells, or T lymphocytes, that is responsible for recognizing fragments of antigen as peptides bound to major histocompatibility complex (MHC) molecules. The binding between TCR and antigen peptides is of relatively low affinity and is degenerate: that is, many TCRs recognize the same antigen peptide and many antigen peptides are recognized by the same TCR.

<span class="mw-page-title-main">Ligand (biochemistry)</span> Substance that forms a complex with a biomolecule

In biochemistry and pharmacology, a ligand is a substance that forms a complex with a biomolecule to serve a biological purpose. The etymology stems from Latin ligare, which means 'to bind'. In protein-ligand binding, the ligand is usually a molecule which produces a signal by binding to a site on a target protein. The binding typically results in a change of conformational isomerism (conformation) of the target protein. In DNA-ligand binding studies, the ligand can be a small molecule, ion, or protein which binds to the DNA double helix. The relationship between ligand and binding partner is a function of charge, hydrophobicity, and molecular structure.

<span class="mw-page-title-main">Digoxigenin</span> Chemical compound

Digoxigenin (DIG) is a steroid found exclusively in the flowers and leaves of the plants Digitalis purpurea, Digitalis orientalis and Digitalis lanata (foxgloves), where it is attached to sugars, to form the glycosides.

In biochemistry, avidity refers to the accumulated strength of multiple affinities of individual non-covalent binding interactions, such as between a protein receptor and its ligand, and is commonly referred to as functional affinity. Avidity differs from affinity, which describes the strength of a single interaction. However, because individual binding events increase the likelihood of occurrence of other interactions, avidity should not be thought of as the mere sum of its constituent affinities but as the combined effect of all affinities participating in the biomolecular interaction. A particular important aspect relates to the phenomenon of 'avidity entropy'. Biomolecules often form heterogenous complexes or homogeneous oligomers and multimers or polymers. If clustered proteins form an organized matrix, such as the clathrin-coat, the interaction is described as a matricity.

<span class="mw-page-title-main">Schild equation</span>

In pharmacology, Schild regression analysis, based upon the Schild equation, both named for Heinz Otto Schild, are tools for studying the effects of agonists and antagonists on the response caused by the receptor or on ligand-receptor binding.

There are many methods to investigate protein–protein interactions which are the physical contacts of high specificity established between two or more protein molecules involving electrostatic forces and hydrophobic effects. Each of the approaches has its own strengths and weaknesses, especially with regard to the sensitivity and specificity of the method. A high sensitivity means that many of the interactions that occur are detected by the screen. A high specificity indicates that most of the interactions detected by the screen are occurring in reality.

A thermal shift assay (TSA) measures changes in the thermal denaturation temperature and hence stability of a protein under varying conditions such as variations in drug concentration, buffer pH or ionic strength, redox potential, or sequence mutation. The most common method for measuring protein thermal shifts is differential scanning fluorimetry (DSF) or thermofluor, which utilizes specialized fluorogenic dyes.

Antigen-antibody interaction, or antigen-antibody reaction, is a specific chemical interaction between antibodies produced by B cells of the white blood cells and antigens during immune reaction. The antigens and antibodies combine by a process called agglutination. It is the fundamental reaction in the body by which the body is protected from complex foreign molecules, such as pathogens and their chemical toxins. In the blood, the antigens are specifically and with high affinity bound by antibodies to form an antigen-antibody complex. The immune complex is then transported to cellular systems where it can be destroyed or deactivated.

<span class="mw-page-title-main">Fluorescence polarization immunoassay</span> Class of invitro biochemical test

Fluorescence polarization immunoassay (FPIA) is a class of in vitro biochemical test used for rapid detection of antibody or antigen in sample. FPIA is a competitive homogenous assay, that consists of a simple prepare and read method, without the requirement of separation or washing steps.

<span class="mw-page-title-main">Protein–ligand complex</span>

A protein–ligand complex is a complex of a protein bound with a ligand that is formed following molecular recognition between proteins that interact with each other or with other molecules. Formation of a protein-ligand complex is based on molecular recognition between biological macromolecules and ligands, where ligand means any molecule that binds the protein with high affinity and specificity. Molecular recognition is not a process by itself since it is part of a functionally important mechanism involving the essential elements of life like in self-replication, metabolism, and information processing. For example DNA-replication depends on recognition and binding of DNA double helix by helicase, DNA single strand by DNA-polymerase and DNA segments by ligase. Molecular recognition depends on affinity and specificity. Specificity means that proteins distinguish the highly specific binding partner from less specific partners and affinity allows the specific partner with high affinity to remain bound even if there are high concentrations of less specific partners with lower affinity.

<span class="mw-page-title-main">Single colour reflectometry</span>

Single colour reflectometry (SCORE), formerly known as imaging Reflectometric Interferometry (iRIf) and 1-lambda Reflectometry, is a physical method based on interference of monochromatic light at thin films, which is used to investigate (bio-)molecular interactions. The obtained binding curves using SCORE provide detailed information on kinetics and thermodynamics of the observed interaction(s) as well as on concentrations of the used analytes. These data can be relevant for pharmaceutical screening and drug design, biosensors and other biomedical applications, diagnostics, and cell-based assays.

<span class="mw-page-title-main">Kinetic exclusion assay</span>

A kinetic exclusion assay (KinExA) is a type of bioassay in which a solution containing receptor, ligand, and receptor-ligand complex is briefly exposed to additional ligand immobilized on a solid phase.

References

  1. Luckey JA, Drossman H, Kostichka T, Smith LM (1993). "High-speed DNA sequencing by capillary gel electrophoresis". Recombinant DNA Part I. Methods in Enzymology. Vol. 218. pp. 154–72. doi:10.1016/0076-6879(93)18014-4. ISBN   9780121821197. PMID   8510530.
  2. Ninfa AJ, Ballou DP, Benore M, eds. (2010). Fundamental Laboratory Approaches for Biochemistry and Biotechnology. Dearborn, MI: University of Michigan.
  3. Mullis KB, Faloona FA (1987). "Specific synthesis of DNA in vitro via a polymerase-catalyzed chain reaction". Recombinant DNA Part F. Methods in Enzymology. Vol. 155. pp. 335–50. doi:10.1016/0076-6879(87)55023-6. ISBN   9780121820565. PMID   3431465.
  4. Sittampalam GS, Kahl SD, Janzen WP (October 1997). "High-throughput screening: advances in assay technologies". Current Opinion in Chemical Biology. 1 (3): 384–91. doi:10.1016/S1367-5931(97)80078-6. PMID   9667878.
  5. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 de Jong LA, Uges DR, Franke JP, Bischoff R (December 2005). "Receptor-ligand binding assays: technologies and applications". Journal of Chromatography. B, Analytical Technologies in the Biomedical and Life Sciences. 829 (1–2): 1–25. doi:10.1016/j.jchromb.2005.10.002. PMID   16253574.
  6. Joseph R. Lakowicz. (1991) Topics in Fluorescence Spectroscopy: Biochemical applications.
  7. 1 2 3 4 5 6 7 8 9 10 Davenport AP, Russel FD (1996). "Radioligand Binding Assays: Theory and Practice". In Stephen J. Mather (ed.). Current directions in radiopharmaceutical research and development. Springer Netherlands. pp. 169–179. ISBN   978-94-010-7289-2.
  8. 1 2 3 4 5 6 7 Hulme EC, Trevethick MA (November 2010). "Ligand binding assays at equilibrium: validation and interpretation". British Journal of Pharmacology. 161 (6): 1219–37. doi:10.1111/j.1476-5381.2009.00604.x. PMC   3000649 . PMID   20132208.
  9. 1 2 3 4 5 6 7 8 Khan MN, Findlay JW, eds. (2009). Ligand-binding assays development, validation, and implementation in the drug development arena. Hoboken, N.J.: John Wiley & Sons. ISBN   978-0470541494.
  10. "LOIS LAB". LOIS LAB. Retrieved 2024-01-30.
  11. Huang, Ting-Hao; Niesman, Peter; Arasu, Deepshika; Lee, Donghyung; De La Cruz, Aubrie L.; Callejas, Antuca; Hong, Elizabeth J.; Lois, Carlos (2017-12-12). "Tracing neuronal circuits in transgenic animals by transneuronal control of transcription (TRACT)". eLife. 6: e32027. doi: 10.7554/eLife.32027 . ISSN   2050-084X. PMC   5777821 . PMID   29231171.
  12. Huang, Ting-Hao; Niesman, Peter; Arasu, Deepshika; Lee, Donghyung; De La Cruz, Aubrie L.; Callejas, Antuca; Hong, Elizabeth J.; Lois, Carlos (2017-12-12). "Tracing neuronal circuits in transgenic animals by transneuronal control of transcription (TRACT)". eLife. 6: e32027. doi: 10.7554/eLife.32027 . ISSN   2050-084X. PMC   5777821 . PMID   29231171.
  13. 1 2 Pollard TD (December 2010). "A guide to simple and informative binding assays". Molecular Biology of the Cell. 21 (23): 4061–7. doi:10.1091/mbc.e10-08-0683. PMC   2993736 . PMID   21115850.
  14. Offermanns S, Walter Rosenthal, eds. (2008). Encyclopedia of molecular pharmacology (2nd ed.). Berlin: Springer. p. 585. ISBN   9783540389163.
  15. 1 2 3 Kahl SD, Sittampalam GS, Weidner J (May 2012). "Calculations and Instrumentation used for Radioligand Binding Assays". Assay Guidance Manual: 1–21. PMID   22553868.
  16. 1 2 3 4 5 6 7 8 9 Davenport AP (2005). Receptor Binding Techniques. Humana Press. pp. 18–19, 101–102, 121–122, 203–204. ISBN   978-1-58829-420-3.
  17. Darling, Ryan J; Brault, Pierre-Alexandre (2004). "Kinetic Exclusion Assay Technology: Characterization of Molecular Interactions". ASSAY and Drug Development Technology. 2 (6): 647–657.
  18. 1 2 3 Goldsby RA (2003). Immunology (5e éd. ed.). New York: W. H. Freeman. p.  152. ISBN   978-0716749479.
  19. 1 2 "Immunoprecipitation (IP) technical guide and protocols" (PDF). Thermo Fisher Scientific Inc. Archived from the original (PDF) on 24 March 2014. Retrieved 20 March 2014.
  20. 1 2 3 4 5 6 Robinson CJ, Sadick M, Deming SN, Estdale S, Bergelson S, Little L (January 2014). "Assay Acceptance Criteria for Multiwell-Plate–Based Biological Potency Assays". BioProcess International. 12 (1): 30–41.
  21. "SIMAG Basic : Magnetic nano and micro particles".
  22. "SIMAG Affinity : Magnetic nano and micro particles by chemicell".
  23. "Home - Accelero® Bioanalytics GMBH".
  24. Moss T, ed. (2001). "Filter-Binding Assays". DNA-protein interactions : principles and protocols (2nd ed.). Totowa, NJ: Humana Press. pp. 1–12. ISBN   9780896036710.
  25. Bondza S, Foy E, Brooks J, Andersson K, Robinson J, Richalet P, Buijs J (2017). "Real-time Characterization of Antibody Binding to Receptors on Living Immune Cells". Frontiers in Immunology. 8: 455. doi: 10.3389/fimmu.2017.00455 . PMC   5401896 . PMID   28484455.
  26. Smith SM (2011). "Strategies for the Purification of Membrane Proteins". Protein Chromatography. Methods in Molecular Biology. Vol. 681. Humana Press. pp. 485–96. doi:10.1007/978-1-60761-913-0_29. hdl:2262/72470. ISBN   9781607619123. PMID   20978985.
  27. Wang W, Yin L, Gonzalez-Malerva L, Wang S, Yu X, Eaton S, et al. (October 2014). "In situ drug-receptor binding kinetics in single cells: a quantitative label-free study of anti-tumor drug resistance". Scientific Reports. 4 (1): 6609. Bibcode:2014NatSR...4E6609W. doi:10.1038/srep06609. PMC   4196117 . PMID   25312029.
  28. Björkelund H, Gedda L, Barta P, Malmqvist M, Andersson K (2011-09-12). "Gefitinib induces epidermal growth factor receptor dimers which alters the interaction characteristics with ¹²⁵I-EGF". PLOS ONE. 6 (9): e24739. Bibcode:2011PLoSO...624739B. doi: 10.1371/journal.pone.0024739 . PMC   3171474 . PMID   21931838.
  29. Xu B, Varasteh Z, Orlova A, Andersson K, Larhammar D, Björkelund H (November 2013). "Detecting ligand interactions with G protein-coupled receptors in real-time on living cells". Biochemical and Biophysical Research Communications. 441 (4): 820–4. doi:10.1016/j.bbrc.2013.10.149. PMID   24211197.
  30. 1 2 3 4 5 Goodman & Gilman's The Pharmacological Basis of Therapeutics. The McGraw Hill Companies Inc. 1996. pp. 29–37. ISBN   978-0-07-026266-9.
  31. 1 2 3 4 5 Haylett DG (2003). "Direct Measurement of Drug Binding to Receptors". In Foreman JC, Johansen T (eds.). Textbook of Receptor Pharmacology (Second ed.). Boca Raton, Florida: CRC L.L.C. pp. 153–180. ISBN   978-0849310294.