Receptor-Ligand Binding Assays
Receptor-Ligand Binding Assays
Konstantin Yakimchuk (Konstantin dot Yakimchuk at ki dot se)
Karolinska Institutet, Stockholm, Sweden
DOI
//dx.doi.org/10.13070/mm.en.1.199
Date
last modified : 2022-10-25; original version : 2011-01-19
Cite as
MATER METHODS 2011;1:199
Abstract

A comprehensive review of research methods used for studying the binding of a protein receptor and its ligand.

Protein Receptor-ligand Interaction/Binding Assays

Physiological processes are regulated by molecular mechanisms involving protein-protein and protein receptor-ligand interactions. Quantification of ligand binding to specific receptors is a key concept of both theoretical studies and drug development research. The main aspects of ligand-receptor binding interactions include binding affinity and kinetics, conformations of targets, binding thermodynamics and ligand efficiency. Every step of ligand-target interactions can be analyzed by different ligand-binding assays. Ligand-binding assays include labeled, label-free, structure-based, thermodynamic, and whole cell ligand-binding assays (Table 1).

Group of assaysAssayMechanismAdvantagesDisadvantages
Labeled ligand-binding assaysFluorescent ligand binding assaysFluorescent labeled ligand binding assay is applied to detect its binding to a target.These assays have a broad spectrum of wavelengths, therefore it can apply multiple colors.The assays are susceptible to different fluorescence interferences. Also, the labeling of a ligand, can lead to undesirable alterations in the binding characteristics of the ligand. In addition, it is difficult to obtain good fluorescent ligands for many receptors.
Radioligand binding assaysRadioactively labeled ligand binding assay is applied to detect its binding to a target.The most popular assay for membrane-bound targets. Good robustness, precise determination of receptor density and distribution, ligand binding sites and affinity.High cost and hazards of handling high levels of radioactivity.
Bioluminescent binding assays using nanoluciferaseBioluminescent reporter nanoluciferase (NanoLuc) is used to study interactions of protein hormones with their receptors.NanoLuc-based assays have several advantages: high sensitivity, high stability and reproducibility.Since NanoLuc reporter is larger than radioisotopes, it may potentially influence the receptor binding.
Label-free ligand binding assaysSurface plasmon resonance (SPR)SPR applies light-excited surface plasmon polaritons to track the binding of ligands to the receptors bound to a gold surface.SPR is the most popular label-free ligand binding assay. This assay can be used to determine binding kinetics.This assay has low sensitivity to binding-induced conformational change.
Plasmon-waveguide resonance (PWR)PWR applies a continuous polarized wave laser exciting electromagnetic waves in a resonator made of a thin silver film with a layer of SiO2 and a glass prism. Ligand binding changes amplitude, position and width of reflected lights.PWR examines anisotropic optical contents of receptor-ligand complexes, thus differentiating mass density changes from conformational changes.PWR has lower sensitivity than SPR with regard to refractive index, thickness and mass parameters.
SPR imaging for affinity-based biosensorsThis assay measures the binding kinetics and is related to intensity modulation and analyzes the reflectivity of monochromatic incident p-polarized light detected at a fixed angle.This assay can be used to monitor binding events in real time.This assay characterizes different compounds for a specific target, it still provides low throughput.
Nanofluidic Fluorescence Microscopy (NFM)NFM combines nanofluidic-based biosensors and fluorescence microscopy to analyze the protein binding kinetics.Protein binding kinetics can be analyzed in one single experiment. The time of analysis by NFM is shorter compared to other microfluidic assays. Statistical errors are reduced and there are no limitations on the size of the analyzed molecules.Limitations of fluorescence microscopy.
Whispering gallery microresonator (WGM)WGM is based on wave sensing binding of molecules to the surface of the cavity induces changes of the resonant wavelength changes. The resonant changes of light permit multiple analyses of molecules.WGM is a highly sensitive assay for molecule detection. The possibility to study drug-target interactions under non-equilibrium conditions using this assay is still unclear.
Resonant waveguide grating (RWG)RWG uses a nanograting to couple light into the waveguide via diffraction. The light illuminates the biosensors in microplate at a nominally normal incident angle. The drug binding of the immobilized receptors results in a shift in the resonant wavelength.RWG assay is effective for affinity screening.This assay is not applicable to study cells due to the large size of cells and limited penetration ability of an evanescent wave.
Biolayer Interferometry Biosensor (BIB)This assay uses a spectrometer to detect interference patterns formed by light reflected from an optical layer and a biolayer containing proteins of interest.Effective assay to study binding kinetics.This assay has been validated mostly for small molecule detection.
Structure-based ligand binding assaysNuclear magnetic resonance (NMR)NMR analyzes the magnetic characteristics of certain atomic nuclei, which absorb electromagnetic radiation in the magnetic field.NMR can be applied to analyze the structure of proteins and the molecular details of protein-ligand interactions and to assist structure-based drug design. Besides, NMR can be used for any class of compounds, for almost all soluble proteins, natively unstructured proteins or membrane proteins.The high cost of the assay, time-consuming, requires a long time to analyze obtained spectra. The resolving power of NMR is lower than X-ray crystallography.
X-ray crystallographyThis assay measures the changes of the diffracted X-ray beams in order to produce a three-dimensional image of electron density within the molecule of interest. Obtained image of electron density is used to determine the structure of the molecules. The solvent effect can be examined by X-ray crystallography. The whole 3D structure can be obtained by the systematic analysis of a crystallized material.Solutions and behavior of molecules in solution cannot be examined. Direct determination of secondary structures including domain movements cannot be performed.
Thermodynamic binding assaysThermal denaturation assays (TDA)TDA detect the thermal denaturation of proteins by differential scanning fluorimetry, which applies a prob fluorophore to monitor thermal denaturation process of proteins in the presence and ligands.These methods have found applications in chemical profiling of different protein families, identifying novel ligands, and investigating the stability of proteins in buffers in solutionThese methods may not be suitable for analysis of very small proteins that do not aggregate upon denaturation.
Isothermal titration calorimetry (ITC)ITC measures the binding enthalpy variation by sensing the heat caused by the binding reactionThis method is useful for detecting highly potent and entropy-driven ligands, compared to others.This method shows low throughput and sensitivity, and requires large sample volumes.
Whole cell ligand-binding assaysSurface acoustic wave (SAW) biosensorThe surface of the biosensor is excited to oscillate with a high frequency. The phase of the generated acoustic wave is shifted upon mass changes. Conformational characteristics are indicated by a change in amplitude.Using this assay, the sequential binding of several distinct ligands, targeting different cell surface molecules, to the whole cells can be detected directly in real time.SAW biosensors for analysis of small molecules using living cells have not developed.
RWG biosensorRWG biosensor can transform drug-receptor interactions in cells into cell phenotypic signatures named dynamic mass redistribution.This assay measures the consequence of binding, and the stimulus-response interpretation of the initial interaction of drug and receptor. Also, RWG can analyze the functional consequences of ligand binding and dissociation.RWG biosensor may not be used to detect ligand binding directly.
Table 1. Ligand binding assays: mechanisms, advantages and disadvantages of the methods.
Labeled Ligand-Binding Assays
Fluorescent ligand binding assays

Fluorescence has a wide spectrum of wavelengths, therefore multiple colors can be applied for the detection of a specific target. Multiple techniques such as fluorescence anisotropy, fluorescence correlation spectroscopy, time-resolved fluorescence, fluorescence polarization, fluorescence and bioluminescence resonance energy transfer are used to measure fluorescence intensity. All of them have been applied in ligand binding assays. Fluorescence anisotropy can be applied to measure unequal intensities of the light emitted from a fluorophore along different axes of polarization [4]. An example is the recent study on the ligand binding dynamics of G protein-coupled receptors (GPCRs) [5]. Fluorescence correlation spectroscopy analyzes fluctuations of the fluorescence intensity, measuring the average number of fluorescent particles and diffusion time, detecting the passage of the particles through space [6]. Time-resolved fluorescence applies convolution integral to measure the lifetime from fluorescence decay after excitation [7]. Fluorescence polarization method detects the difference between the polarization of the polarized light used to excite the fluorophore and light emitted from a fluorophore [8].

Resonance energy transfer methods apply labels which are attached to reactive amino acid side chains. These labels include fluorescent proteins and light-emitting enzymes. Fluorescence and bioluminescence resonance energy transfer (FRET and BRET) methods are used to monitor the distances between two labels. These methods analyze conformational changes of single protein molecules or protein-protein interactions. Furthermore, FRET method detects the non-radiative transfer of energy from one donor chromophore to an acceptor chromophore (Figure 1) [1]. The method was recently used to decipher the interactions between the estrogen alpha receptor and its co-activator [9], to screen GCPR ligand libraries [10] and to detect receptor-ligand binding in situ [11], leading the way to visualizing such interaction in their native environment. Zhao LH et al detected FRET to measure the binding kinetics between GFP-tagged parathyroid hormone receptor-1 and tetramethylrhodamine-labeled ligand in live HEK-293 cells [12].

Receptor-Ligand Binding Assays figure 1
Figure 1. The principle of inter- (A) and intra-molecular (B) fluorescence resonance energy transfer (FRET). Proteins (gray) labeled with the fluorophores CFP (cyan) and YFP (yellow). If the fluorophores are close to each other (<10 nm), the excited CFP transfers energy to YFP and yellow emission results. A small change in the distance between CFP and YFP may result from the approximation of the two proteins in A or a conformational change of the protein in B and result in marked changes in FRET. [1].

Recent studies applied a combination of multi-channel fluorescence imaging with microarrays to study molecular interactions. Protein microarrays such as kinase and G protein-coupled receptor (GPCR) arrays have been designed and applied to analyze ligand binding [13]. Analysis of protein kinase A and leukocyte-specific protein tyrosine kinase with 80 kinase inhibitor library confirmed previously identified inhibitors for these two kinases [14].

Multiple fluorescent ligands have been developed for the studies of their specific receptors. Among the most recent are ligands for the GPCR receptors [15], bradykinin B1 receptor [16], sigma-1 receptor [17], μ-opioid receptor [18], the 5-HT3 receptor [19], and the oxytocin receptor [20].

Recent improvements in the technique allowed for the monitoring of protein kinetics in real time (e.g., by using nanofluidic fluorescence microscopy [21] ), and the quantitative analysis of protein-ligand interactions [22].

Radioligand binding assays

A radioactively labeled ligand is applied in this assay to detect its binding to a target. Radioligand binding assays are mainly used for membrane-bound molecules, such as G protein-coupled receptors like angiotensin II type 1 receptor [23] or apelin receptor [24]. Radioligand binding assays can determine the type, anatomical distribution and density of the receptors and ligand affinity and binding sites [25]. There are three experimental types of radioligand binding assays: saturation assay, competitive assay and kinetic assay [26]. Saturation assays analyze the equilibrium binding of radioactively labeled ligand to the receptor, using a fixed receptor level and increasing concentrations of the ligand. The assay measures the tissue / cell-specific affinity and the density of the analyzed receptor. A competitive assay investigates equilibrium binding at a fixed concentration of radioligand and in the presence of different concentrations of an unlabeled competitor, for example, [12, 23, 24, 27]. Binding data analysis provides the affinity of the receptor for the competitor molecule. Kinetic assays are used to determine receptor / ligand pair specific dissociation and association constants. Multiple receptor specific radioligand binding assays protocols have been developed [28]. Zhao LH et al measured the binding affinity of wild-type and mutant parathyroid hormone receptor-1 against its ligand in HEK-293T cells through radioiodine-labeled parathyroid hormone in the presence of varying concentrations of unlabelled ligand [12]. The binding can be measured through a scintillation proximity assay [27, 29] or a polyethylene glycol precipitation assay [29].

Bioluminescent binding assays using nanoluciferase

Nanoluciferase (NanoLuc)-based methods have several important advantages, including high sensitivity and reproducibility, and were applied to the BRET analysis [30], in vivo imaging [31], and membrane internalization assessment [32]. Recently, NanoLuc has been effectively used in binding assays for various protein hormones and their receptors. Several studies reported applications of NanoLuc-based assays for the detection of relaxin [33], ghrelin [34] and erythropoietin [35]. In addition, NanoLuc was recently used in the development of the BRET-based assessment of the G Protein-Coupled Receptors (GPCR) ligand binding [15]. Both chemical conjugation and genetic fusion may be applied for the development of bioluminescent binding assays. With regard to the chemical conjugation technique, the selected protein hormones are specifically conjugated with a NanoLuc reporter. The NanoLuc-bound proteins can be isolated by chromatography and analyzed by bioluminescence techniques.In contrast to chemical conjugation, the genetic approach applies fusion of the bioluminescent reporter with a chosen protein domain, followed by the overexpression of the generated complex in the bacterial or mammalian cells. In particular, genetic fusion was reported to be effective for the analysis of interactions of erythropoietin with its membrane receptor [35].

Others

Selenium labeling of peptide ligands, coupled with inductively coupled plasma mass spectrometry, has been evaluated for measuring the receptor-ligand interaction [36].

Label-free Ligand Binding Assays
Surface plasmon resonance (SPR)

Surface plasmon resonance is based on the surface plasmon polaritons (SPP) which are electromagnetic waves generated when light interacts with surface charges in the gold surface (Figure 2). The main application of SPR is to analyze the binding of the ligands to the receptors linked to a gold surface. Therefore, SPR is used to study binding kinetics [37]. The analyzed interactions between a drug and its target include the formation of a ligand-receptor complex and dissociation of this complex followed by the ligand removal. SPR is applied to follow the association and dissociation mechanisms in real time by creating a sensorgram which detects the changes in wavelength. Importantly, SPR is a quantitative method, since these changes are related to the number of bound ligand molecules. However, the sensitivity of SPR is low in the detection of binding-induced conformational change [38]. SPR assay can be used for different soluble proteins including kinases and proteases, as well as for membrane protein [39]. In particular, SPR assay has recently been used to analyze the binding of extracelluar domains of membrane proteins [40], the interaction of orthosteric and allosteric ligands with chemokine receptor, CCR5, lipid-protein interactions [41], protein-carotenoid interactions [42], protein-protein interactions [43], toxin binding [44] to give just a few examples.

Receptor-Ligand Binding Assays figure 2
Figure 2. Surface plasmon resonance (SPR) determines binding kinetics. A. A microfluidic device is applied in SPR to detect the association and dissociation of drug compounds binding to the target immobilized on the golden surface. B. The resonance angle shifts when drug compounds bind to the target. [2].
Nanofluidic Fluorescence Microscopy (NFM)

Nanofluidic Fluorescence Microscopy (NFM) applies a combination of nanofluidic-based biosensors and fluorescence microscopy. Nanofluidic immunosensor is effectively used for real-time kinetic assessment of protein/ligand interactions. In contrast to other immunoassays, NFM allows the measurement of binding kinetics in one simplified step and reduces the length of analysis [21]. In particular, this method may be used to analyze antigen-antibody binding by the direct assessment with a fluorescence microscope [45].

Plasmon-waveguide resonance

Similarly to SPR, plasmon-waveguide resonance method also applies surface-excited plasmon. PWR uses a polarized continuous wave laser exciting electromagnetic waves in a resonator made of a thin silver film with a layer of SiO2 and a glass prism (Figure 3). In this assay, receptors are immobilized on the outer SiO2 surface. Ligand binding changes amplitude, position and width of reflected lights. In contrast to SPR, PWR examines anisotropic optical contents of receptor-ligand complexes and differentiates mass density changes from conformational changes [46]. Thus, PWR can be applied to analyze the changes in receptor conformation and local mass density [47]. This assay shows that distinct ligands including full agonists, partial agonists and antagonists cause distinct conformational changes in receptors. PWR has been reported to be a useful tool to investigate receptor-mediated processes in real biological membranes, since it has been used to study ligand binding to integral membrane proteins obtained from bacterial and mammalian cell membranes [48].

Receptor-Ligand Binding Assays figure 3
Figure 3. Plasmon-waveguide resonance (PWR) determines both binding kinetics and conformational changes. A. A polarized continuous wave laser excites electromagnetic waves in a resonator made of a thin silver film with a layer of SiO2 and a glass prism. B. The resonance angle shifts when drug compounds bind to the immobilized target. [2].
SPR imaging for affinity-based biosensors

SPR imaging for affinity-based biosensors measures the binding kinetics and is related to intensity modulation and analyzes the reflectivity of monochromatic incident p-polarized light detected at a fixed angle [49]. This method studies binding events across the entire sensor surface. Besides, SPR imaging studies are often combined with microarrays to measure specific microspots [50]. SPR imaging analyzes different chemical compounds for a target of interest. In addition, this method has been used for biomarker identification, for detecting microRNA and for analysis of interactions of proteins, DNA and RNA and whole cells.

Whispering gallery microresonator (WGM)

Whispering gallery microresonator (WGM) is based on wave sensing and applies different microresonators or resonant cavities having high-quality factors to limit light, which generates very sharp resonances for the light [51]. Binding of molecules to the surface of the cavity induces changes of the resonant wavelength changes. The resonant changes of light permit multiple analyses of molecules. In contrast to other label-free ligand binding assays, backscattering interferometry (BSI) has an advantage as homogenous solution-based biosensing. BSI has been used to detect the equilibrium dissociation constants for different types of receptor-ligand binding, including antigen-antibody interactions, and host-pathogen interactions [52].

Several improvements to the technique already enable enhanced device performance [53], allowing their use in complex environments [54]. Xu et al [55] recently demonstrated that microresonators might be soon biodegradable and based on biocompatible proteins. More information on the newest device in the area of biosensing and perspectives of further developments can be found in a recent review (2016) [56].

Resonant waveguide grating (RWG)

Resonant waveguide grating (RWG) applies the electromagnetic wave bound to the surface to analyze biomolecular interactions [57]. In this method, light is coupled into the thin film by diffraction leading to the generation of an evanescent wave (Figure 4). RWG uses the surface-linked electromagnetic wave to analyze interactions between ligand and receptor. In the RWG method the maximum in coupling efficiency is obtained at the resonance angle or wavelength which is a function of the local refractive index at the sensor surface. In addition, RWG is usually performed in microplate formats and is designed for affinity screening [58].

Receptor-Ligand Binding Assays figure 4
Figure 4. Microplate-based resonant waveguide grating (RWG) determines binding affinity screening. A. Light is coupled into the waveguide via diffraction. B. The intensity of the reflected light is measured. The compound binding of the immobilized receptors causes a shift in the resonant wavelength. [2].
Biolayer interferometry biosensor (BIB)

Biolayer interferometry biosensor applies a spectrometer to detect interference patterns formed by light reflected from an optical layer and a biolayer containing proteins of interest (Figure 5). These two types of light interact with each other and create a specific interference pattern. When ligands bind to the receptors, the inference pattern is altered. These changes of the interference pattern represented by specific peaks and troughs can be detected by a spectrometer. This method helps to study binding kinetics [59]. Moreover, a subtype of BIB, biolayer interferometry (BLI), can be effectively used to measure ligand binding [60] or protein-drug interaction [61]. Pastushok L et al identified a positive antibody fragment phage display clone against RAD51 protein, studied the binding of the resultant antibody fragment with RAD51, and the inhibitory effect of the antibody fragment against the binding between RAD51 and single-stranded DNA with an Octet RED384 instrument from ForteBio [62]. Silva MC et al used ForteBio Octet Red384 instrument to measure the biophysical interaction between small molecules and biotinylated tau proteins with streptavidin sensors [63]. Rice HC et al measured the binding between the GABABR1a Sushi-1 or -2 domain and sAPP peptides with a ForteBio BLItz instrument [64]. E Verschueren et al validated the interaction between AXL and IL1RL1 via BIB using an Octet Red system [40].

Receptor-Ligand Binding Assays figure 5
Figure 5. Biolayer interferometry biosensor (BIB). A. A broadband light resource is used to illuminate the interfaces. Reflected light waves originate from the immobilized receptor surface and the optical layer, interacting with each other and creating interference patterns. B. Compound binding affects the inference pattern. [2].
Other methods

Mass spectrometry (MS) is emerging as a useful tool for the receptor-ligand binding interactions. The sensitivity of MS equipment is increasing progressively, it is possible now to detect low ligand quantities that are usually found in ligand binding assays. Some examples of its applications, are the MS binding assays developed for human dopamine [65, 66], norepinephrine, and serotonin transporters [65]. MS-based assays are often the method of choice for high-throughput drug screening and detection strategies designed to reduce the number of false positive interactions [67]. Rice HC et al purfied sAPP-binding proteins with sAPP-Fc baits from rat brain homogenates and identified these proteins through mass spectrometry [64].

Structure-based Ligand Binding Assays
Nuclear magnetic resonance (NMR)

Nuclear magnetic resonance (NMR) methods are well-known techniques applied to study protein–ligand complexes [68]. They include protein-observed and ligand-observed NMR methods. Protein-observed NMR methods detect the chemical shift changes of the protein resonances and therefore can be used to identify the ligand binding site. There are several ligand-based NMR methods depending on intermolecular magnetization transfer, such as saturation transfer difference spectroscopy, transferred-nuclear Overhauser effects (NOEs) spectroscopy and water–ligand observed via gradient spectroscopy experiments [3].

Ligand-observed NMR methods, which rely on intermolecular magnetization transfer, include: transferred-NOE spectroscopy (NOESY), saturation transfer difference (STD) spectroscopy and water–ligand observed via gradient spectroscopy (WaterLOGSY) [3]. In particular, transferred NOESY method analyzes spectrum on a sample of low molecular weight molecules in the presence of a substoichiometric amount of a protein. In a mixture of molecules in the presence of a protein, the molecules binding to the protein will show strong negative NOEs, whereas non-binding molecules will demonstrate very weak positive NOEs. If two ligands bind simultaneously with similar residence times on the protein surface, forming a complex. In this situation, strong negative ligand–ligand NOEs can be observed (Figure 6). In addition, when two ligand compounds bind competitively to the same binding pocket, NOE can be detected. Such NOESY cross-peaks are referred to as INPHARMA NOEs (Figure 6). INPHARMA methods were used for the detection of concentration-dependent binding of ligands to membranes containing GCPR40 [69]. Two recent reviews by Aguirre et al (2015) and Oxenoid and Chou (2016) provide great overviews of the use of both protein-observed NMR experiments and ligand-observed NMR experiments [70], as well as on the use of NMR methods in the study of ligand-induced membrane protein conformational changes [71]. Moreover, recent advances in hydrogen/deuterium exchange mass spectrometry (HDX-MS) allows for the use of such technique to identify the binding sites and the structural changes associated with protein-protein or protein-ligand interactions [72]. Rice HC et al used both protein-based and ligand-based NMR on a Bruker Avance III HD 800 MHz spectrometer equipped with a TCI cryoprobe to obtain the binding of sAPP 9mer peptide with Sushi1 domain of GABABR1a [64].

Receptor-Ligand Binding Assays figure 6
Figure 6. The interligand nuclear Overhasuer effect (ILOE) versus the ILOE for pharmacophore mapping (INPHARMA) experiment. Hp, Hp’, H1, H1’, H1” and H2, H2’ represent protons of the protein (grey), the reference molecule (1, pink) and the fragment (2, green or violet). The magnetization transfer between two protons is represented by arrows. Schematic representations of nuclear Overhasuer effect spectroscopy (NOESY) spectra are also shown. Nuclear Overhasuer effects (NOEs) are represented by black dots and white dots represent intermolecular NOEs are represented by white dots. [3].
X-ray crystallography

X-ray crystallography (and increasingly cryo-EM) provides a three-dimensional structure of receptor-ligand complex and is important for the target-based approach. There are several public databases such as ChEMBL, PubChem and DrugBank, which provide valuable information regarding the structures of biologically active chemicals. This information can be effectively used for structure-based ligand design. In addition, a combination of medicinal chemistry with crystallographic data on protein-ligand complexes became a powerful method. Moreover, many in silico methods such as cheminformatics [73] and virtual screening [74] are very useful for drug development.

X-ray-based methods have been used to identify the structure of GCPRs [75-77], such as histamine H1 receptor [78], sphingosine 1-phosphate S1P1 receptor [79], muscarinic M3 acetylcholine receptor [80] and other receptors [81-83]. In addition, X-ray-based assays were used to find new chemical ligands with different affinities for adenosine A2A receptor [84, 85], D3 dopamine receptors [86] and for CXCR4 receptor [87].

Thermodynamic Binding Assays
Thermal denaturation assays (TDA)

Thermal denaturation assays (TDA) detect the thermal denaturation of proteins by differential scanning fluorimetry (DSF) with, for example, a Quant Studio 6 real-time PCR system from Thermo Fisher [23]. In DSF, probe fluorophores are used to detect changes in the thermal denaturation patterns of proteins under different conditions, for instance in the absence or presence of ligands. Upon interaction with their specific ligands, receptors undergo structural and dynamic changes that tend to increase their thermal stability and their melting point (Tm). Such changes are detected by TDAs. So far, TDA has been used for chemical profiling of protein families and identification of novel ligands [88] or to screen proteins against a large number of putative ligand compounds in a time-effective manner and with low false-positive rates [88]. For example, Alam MM et al measured the thermostability of Plasmodium falciparum protein kinase PfCLK3 with different ligands through differential scanning fluorimetry assays using Applied Biosystems QuantStudio 6 and Protein Thermal shift software [89]. A program called Meltdown [90] can be used to ease the analysis of the thermal melt curve data obtained from DSF experiments. The program performs control checks, curve normalization, outlier rejection, and Tm estimation.

A thorough description of the DSF technique is done in the recent review of DeSantis and Reinking [91]. Also, a methodological workflow for small-scale, high-throughput thermal denaturation of recombinant proteins in the presence of SYPRO Orange dye was published recently [92].

Isothermal titration calorimetry (ITC)

Isothermal titration calorimetry (ITC) evaluates the binding enthalpy variation by sensing the heat caused by the binding reaction. ITC experiments allow the distinction between enthalpic and entropic contributions to binding modes. Moreover, the ITC method can be used to compare ligands with similar binding affinities by detecting differences in binding modes. In particular, ITC is very reliable in analyzing entropy-driven ligands [96]. Moreover, an enthalpic array has been recently developed and can be used for the analysis of label-free interactions. ITC has been used to study the functioning of mammalian and bacterial membrane proteins [97]. In addition, ITC was applied to investigate ligand binding to functional domains of GPCR, including chemokine receptors, C5a receptor and parathyroid receptor N-terminal domains [97]. Bhogaraju S et al measured the binding affinities of Legionella SidJ protein with either apo-calmodulin or calmodulin in ITC experiments with a MicroCal ITC200 from Malvern [98].

ITC represents the gold-standard method for understanding the thermodynamics of biomolecular binding processes. It allows for the reliable determination of the dissociation constant (KD) of only one ligand, and the detection range is limited to 1nM-100 μM. Lower KDs can only be determined by performing competition or displacement assays, if a competitive ligand is available. Krainer and Keller (2015) introduced an ITC displacement assay that allows for the simultaneous characterization of two ligands of with different affinities for the same binding site [99].

SupplierInstrumentsNumSample references
MicroCalITC2004the binding of XEG1 or XLP1 with GIP1 [100], and of SGN3 extracellular domain with CIF2 [101], of RALF23 peptides with FER ectodomain [102], of sAPP fragments with GABABR1a Sushi-1 or -2 domain [64]
VP-ITC1the binding of SGN3 extracellular domain with CIF2 at pH 5.8 [101]
TA InstrumentsAffinity1the binding of Reck and Wnt7 peptides [103]
Table 2. Commonly used ITC instruments among the formal publications Labome has surveyed. Num: number of publications.
Whole Cell Ligand-Binding assays
Surface acoustic wave (SAW) biosensor

Surface acoustic wave (SAW) biosensor detects and analyzes the amplitudes of mechanical acoustic waves. This method is based on a sensor, in which the surface, made from a mono-crystalline piezo-material, is excited to oscillate by acoustic waves. The phase of the acoustic waves is shifted when masses of the molecules change. The conformational changes of the proteins result in changes in the amplitude of acoustic waves, which can be detected by SAW [104]. It is established that signal induced by ligand binding is related to mass changes on the surface. SAW biosensors were shown to be effective for analysis of cell/substrate interactions using class I Major Histocompatibility Complex molecule HLA-A2 and anti-HLA monoclonal antibodies [105]. Also, SAW biosensors were applied for functional assays of olfactory receptors [106].

RWG biosensor

RWG biosensor is used for biomolecular interaction analysis. Moreover, the RWG biosensor can transform drug-receptor interactions in cells into cell phenotypic signatures named dynamic mass redistribution [107, 108]. Moreover, RWG biosensor can detect cellular response formed by many cellular events which change local mass density at the surface of the sensor. After detection, the sensor analyzes the consequences of the binding reaction. Moreover, recently developed RWG imager analyzes the consequences of ligand binding using a microfluidic perfusion scheme to control the duration of exposure to the ligand [109]. The RWG imager detects the perfusion flow in real time in order to measure the effect of ligand binding (Figure 6). In particular, RWG biosensor can be used to control the duration of agonist exposure [109]. In addition, the microfluidics can detect the duration of the functional recovery of the receptor. The RWG sensor can be used to analyze the impact of protease-activated receptor PAR1 agonists on the receptor desensitization and resensitization [110]. These studies demonstrated that the RWG sensor could be used to study the functional effects of ligand-receptor interactions in living cells.

Conclusion

Ligand binding assays are effectively applied for both studies aiming to understand the regulation of biological functions and the research focused on developing new drugs which are strongly based on the determination of binding affinity and efficacy. It is assumed that data of in vitro binding affinity of a ligand may provide important information about in vivo efficacy of this ligand. However, many additional aspects of ligand binding should be considered. Since ligand molecules can induce conformational changes in the target proteins, structure-based methods such as X-ray and NMR should help to identify conformational changes and new binding sites. In addition, binding kinetics should be optimized in order to maximize the therapeutic efficacy of compounds. SPR, PWR and RWG methods are effectively applied for the studies of binding kinetics [109, 110]. Thermodynamics of ligand-protein interactions can be analyzed by thermodynamic binding assays. Moreover, ligand binding should be analyzed not only using purified protein molecules but also in living cells by applying the whole cell ligand-binding methods. The detailed understanding of the complexity of ligand-receptor interactions may be achieved by applying different ligand-binding assays such as labeled, label-free, thermodynamic, structure-based and whole cell ligand-binding methods.

References
  1. Lohse M, Nuber S, Hoffmann C. Fluorescence/bioluminescence resonance energy transfer techniques to study G-protein-coupled receptor activation and signaling. Pharmacol Rev. 2012;64:299-336 pubmed publisher
  2. Fang Y. Ligand-receptor interaction platforms and their applications for drug discovery. Expert Opin Drug Discov. 2012;7:969-88 pubmed
  3. Cala O, Guillière F, Krimm I. NMR-based analysis of protein-ligand interactions. Anal Bioanal Chem. 2014;406:943-56 pubmed publisher
  4. Owicki J. Fluorescence polarization and anisotropy in high throughput screening: perspectives and primer. J Biomol Screen. 2000;5:297-306 pubmed
  5. Veiksina S, Kopanchuk S, Mazina O, Link R, Lille A, Rinken A. Homogeneous fluorescence anisotropy-based assay for characterization of ligand binding dynamics to GPCRs in budded baculoviruses: the case of Cy3B-NDP-?-MSH binding to MC4 receptors. Methods Mol Biol. 2015;1272:37-50 pubmed publisher
  6. Lieto A, Cush R, Thompson N. Ligand-receptor kinetics measured by total internal reflection with fluorescence correlation spectroscopy. Biophys J. 2003;85:3294-302 pubmed
  7. Handl H, Gillies R. Lanthanide-based luminescent assays for ligand-receptor interactions. Life Sci. 2005;77:361-71 pubmed
  8. Rossi A, Taylor C. Analysis of protein-ligand interactions by fluorescence polarization. Nat Protoc. 2011;6:365-87 pubmed publisher
  9. Moore T, Gunther J, Katzenellenbogen J. Estrogen receptor alpha/co-activator interaction assay: TR-FRET. Methods Mol Biol. 2015;1278:545-53 pubmed publisher
  10. Oueslati N, Hounsou C, Belhocine A, Rodriguez T, Dupuis E, Zwier J, et al. Time-resolved FRET strategy to screen GPCR ligand library. Methods Mol Biol. 2015;1272:23-36 pubmed publisher
  11. Axmann M, Schütz G, Huppa J. Measuring TCR-pMHC Binding In Situ using a FRET-based Microscopy Assay. J Vis Exp. 2015;:e53157 pubmed publisher
  12. Zhao L, Ma S, Sutkeviciute I, Shen D, Zhou X, de Waal P, et al. Structure and dynamics of the active human parathyroid hormone receptor-1. Science. 2019;364:148-153 pubmed publisher
  13. Labaer J, Ramachandran N. Protein microarrays as tools for functional proteomics. Curr Opin Chem Biol. 2005;9:14-9 pubmed
  14. Li T, Liu D, Wang Z. Screening kinase inhibitors with a microarray-based fluorescent and resonance light scattering assay. Anal Chem. 2010;82:3067-72 pubmed publisher
  15. Stoddart L, White C, Nguyen K, Hill S, Pfleger K. Fluorescence- and bioluminescence-based approaches to study GPCR ligand binding. Br J Pharmacol. 2016;173:3028-37 pubmed publisher
  16. Charest Morin X, Marceau F. Biotechnological Fluorescent Ligands of the Bradykinin B1 Receptor: Protein Ligands for a Peptide Receptor. PLoS ONE. 2016;11:e0148246 pubmed publisher
  17. Abate C, Riganti C, Pati M, Ghigo D, Berardi F, Mavlyutov T, et al. Development of sigma-1 (σ1) receptor fluorescent ligands as versatile tools to study σ1 receptors. Eur J Med Chem. 2016;108:577-585 pubmed publisher
  18. Schembri L, Stoddart L, Briddon S, Kellam B, Canals M, Graham B, et al. Synthesis, Biological Evaluation, and Utility of Fluorescent Ligands Targeting the ?-Opioid Receptor. J Med Chem. 2015;58:9754-67 pubmed publisher
  19. Jack T, Simonin J, Ruepp M, Thompson A, Gertsch J, Lochner M. Characterizing new fluorescent tools for studying 5-HT₃ receptor pharmacology. Neuropharmacology. 2015;90:63-73 pubmed publisher
  20. Karpenko I, Margathe J, Rodriguez T, Pflimlin E, Dupuis E, Hibert M, et al. Selective nonpeptidic fluorescent ligands for oxytocin receptor: design, synthesis, and application to time-resolved FRET binding assay. J Med Chem. 2015;58:2547-52 pubmed publisher
  21. Teerapanich P, Pugniere M, Henriquet C, Lin Y, Chou C, Leïchlé T. Nanofluidic Fluorescence Microscopy (NFM) for real-time monitoring of protein binding kinetics and affinity studies. Biosens Bioelectron. 2017;88:25-33 pubmed publisher
  22. Breen C, Raverdeau M, Voorheis H. Development of a quantitative fluorescence-based ligand-binding assay. Sci Rep. 2016;6:25769 pubmed publisher
  23. Wingler L, Skiba M, McMahon C, Staus D, Kleinhenz A, Suomivuori C, et al. Angiotensin and biased analogs induce structurally distinct active conformations within a GPCR. Science. 2020;367:888-892 pubmed publisher
  24. Ma Y, Ding Y, Song X, Ma X, Li X, Zhang N, et al. Structure-guided discovery of a single-domain antibody agonist against human apelin receptor. Sci Adv. 2020;6:eaax7379 pubmed publisher
  25. Glickman J, Schmid A, Ferrand S. Scintillation proximity assays in high-throughput screening. Assay Drug Dev Technol. 2008;6:433-55 pubmed publisher
  26. Hulme E, Trevethick M. Ligand binding assays at equilibrium: validation and interpretation. Br J Pharmacol. 2010;161:1219-37 pubmed publisher
  27. Staus D, Hu H, Robertson M, Kleinhenz A, Wingler L, Capel W, et al. Structure of the M2 muscarinic receptor-β-arrestin complex in a lipid nanodisc. Nature. 2020;579:297-302 pubmed publisher
  28. Dong C, Liu Z, Wang F. Radioligand saturation binding for quantitative analysis of ligand-receptor interactions. Biophys Rep. 2015;1:148-155 pubmed
  29. Nielsen J, Brandt J, Boesen T, Hummelshøj T, Slaaby R, Schluckebier G, et al. Structural Investigations of Full-Length Insulin Receptor Dynamics and Signalling. J Mol Biol. 2022;434:167458 pubmed publisher
  30. Robers M, Dart M, Woodroofe C, Zimprich C, Kirkland T, Machleidt T, et al. Target engagement and drug residence time can be observed in living cells with BRET. Nat Commun. 2015;6:10091 pubmed publisher
  31. Karlsson E, Meliopoulos V, Savage C, Livingston B, Mehle A, Schultz Cherry S. Visualizing real-time influenza virus infection, transmission and protection in ferrets. Nat Commun. 2015;6:6378 pubmed publisher
  32. Liu Y, Song G, Shao X, Liu Y, Guo Z. Quantitative measurement of cell membrane receptor internalization by the nanoluciferase reporter: Using the G protein-coupled receptor RXFP3 as a model. Biochim Biophys Acta. 2015;1848:688-94 pubmed publisher
  33. Wu Q, Zhang L, Shao X, Wang J, Gao Y, Xu Z, et al. Application of the novel bioluminescent ligand-receptor binding assay to relaxin-RXFP1 system for interaction studies. Amino Acids. 2016;48:1099-107 pubmed publisher
  34. Liu Y, Shao X, Zhang L, Song G, Liu Y, Xu Z, et al. Novel bioluminescent receptor-binding assays for peptide hormones: using ghrelin as a model. Amino Acids. 2015;47:2237-43 pubmed publisher
  35. Song G, Wu Q, Xu T, Liu Y, Xu Z, Zhang S, et al. Quick preparation of nanoluciferase-based tracers for novel bioluminescent receptor-binding assays of protein hormones: Using erythropoietin as a model. J Photochem Photobiol B. 2015;153:311-6 pubmed publisher
  36. Cheignon C, Cordeau E, Prache N, Cantel S, Martinez J, Subra G, et al. Receptor-Ligand Interaction Measured by Inductively Coupled Plasma Mass Spectrometry and Selenium Labeling. J Med Chem. 2018;61:10173-10184 pubmed publisher
  37. Daghestani H, Day B. Theory and applications of surface plasmon resonance, resonant mirror, resonant waveguide grating, and dual polarization interferometry biosensors. Sensors (Basel). 2010;10:9630-46 pubmed publisher
  38. Rich R, Myszka D. Survey of the 2009 commercial optical biosensor literature. J Mol Recognit. 2011;24:892-914 pubmed publisher
  39. Aristotelous T, Hopkins A, Navratilova I. Surface plasmon resonance analysis of seven-transmembrane receptors. Methods Enzymol. 2015;556:499-525 pubmed publisher
  40. Verschueren E, Husain B, Yuen K, Sun Y, Paduchuri S, Senbabaoglu Y, et al. The Immunoglobulin Superfamily Receptome Defines Cancer-Relevant Networks Associated with Clinical Outcome. Cell. 2020;182:329-344.e19 pubmed publisher
  41. Del Vecchio K, Stahelin R. Using Surface Plasmon Resonance to Quantitatively Assess Lipid-Protein Interactions. Methods Mol Biol. 2016;1376:141-53 pubmed publisher
  42. Vachali P, Li B, Bartschi A, Bernstein P. Surface plasmon resonance (SPR)-based biosensor technology for the quantitative characterization of protein-carotenoid interactions. Arch Biochem Biophys. 2015;572:66-72 pubmed publisher
  43. Nikolovska Coleska Z. Studying protein-protein interactions using surface plasmon resonance. Methods Mol Biol. 2015;1278:109-38 pubmed publisher
  44. Martin Eauclaire M, Ferracci G, Bosmans F, Bougis P. A surface plasmon resonance approach to monitor toxin interactions with an isolated voltage-gated sodium channel paddle motif. J Gen Physiol. 2015;145:155-62 pubmed publisher
  45. Lin Y, Huang Y, Teerapanich P, Leïchlé T, Chou C. Multiplexed immunosensing and kinetics monitoring in nanofluidic devices with highly enhanced target capture efficiency. Biomicrofluidics. 2016;10:034114 pubmed publisher
  46. Tollin G, Salamon Z, Hruby V. Techniques: plasmon-waveguide resonance (PWR) spectroscopy as a tool to study ligand-GPCR interactions. Trends Pharmacol Sci. 2003;24:655-9 pubmed
  47. Alves I, Cowell S, Salamon Z, Devanathan S, Tollin G, Hruby V. Different structural states of the proteolipid membrane are produced by ligand binding to the human delta-opioid receptor as shown by plasmon-waveguide resonance spectroscopy. Mol Pharmacol. 2004;65:1248-57 pubmed
  48. Salamon Z, Fitch J, Cai M, Tumati S, Navratilova E, Tollin G. Plasmon-waveguide resonance studies of ligand binding to integral proteins in membrane fragments derived from bacterial and mammalian cells. Anal Biochem. 2009;387:95-101 pubmed publisher
  49. Scarano S, Mascini M, Turner A, Minunni M. Surface plasmon resonance imaging for affinity-based biosensors. Biosens Bioelectron. 2010;25:957-66 pubmed publisher
  50. Lausted C, Hu Z, Hood L. Quantitative serum proteomics from surface plasmon resonance imaging. Mol Cell Proteomics. 2008;7:2464-74 pubmed publisher
  51. Vollmer F, Arnold S, Braun D, Teraoka I, Libchaber A. Multiplexed DNA quantification by spectroscopic shift of two microsphere cavities. Biophys J. 2003;85:1974-9 pubmed
  52. Saetear P, Perrin A, Bartholdson S, Wanaguru M, Kussrow A, Bornhop D, et al. Quantification of Plasmodium-host protein interactions on intact, unmodified erythrocytes by back-scattering interferometry. Malar J. 2015;14:88 pubmed publisher
  53. Francois A, Reynolds T, Monro T. A fiber-tip label-free biological sensing platform: a practical approach toward in-vivo sensing. Sensors (Basel). 2015;15:1168-81 pubmed publisher
  54. Wang F, Anderson M, Bernards M, Hunt H. PEG Functionalization of Whispering Gallery Mode Optical Microresonator Biosensors to Minimize Non-Specific Adsorption during Targeted, Label-Free Sensing. Sensors (Basel). 2015;15:18040-60 pubmed publisher
  55. Xu L, Jiang X, Zhao G, Ma D, Tao H, Liu Z, et al. High-Q silk fibroin whispering gallery microresonator. Opt Express. 2016;24:20825-30 pubmed publisher
  56. Righini G, Soria S. Biosensing by WGM Microspherical Resonators. Sensors (Basel). 2016;16: pubmed publisher
  57. Young J, Garvey J, Huang P. Glial immunoreactivity for metallothionein in the rat brain. Glia. 1991;4:602-10 pubmed
  58. Heeres J, Hergenrother P. High-throughput screening for modulators of protein-protein interactions: use of photonic crystal biosensors and complementary technologies. Chem Soc Rev. 2011;40:4398-410 pubmed publisher
  59. Concepcion J, Witte K, Wartchow C, Choo S, Yao D, Persson H, et al. Label-free detection of biomolecular interactions using BioLayer interferometry for kinetic characterization. Comb Chem High Throughput Screen. 2009;12:791-800 pubmed
  60. Wartchow C, Podlaski F, Li S, Rowan K, Zhang X, Mark D, et al. Biosensor-based small molecule fragment screening with biolayer interferometry. J Comput Aided Mol Des. 2011;25:669-76 pubmed publisher
  61. Bellail A, Jin H, Lo H, Jung S, Hamdouchi C, Kim D, et al. Ubiquitination and degradation of SUMO1 by small-molecule degraders extends survival of mice with patient-derived tumors. Sci Transl Med. 2021;13:eabh1486 pubmed publisher
  62. Pastushok L, Fu Y, Lin L, Luo Y, DeCoteau J, Lee K, et al. A Novel Cell-Penetrating Antibody Fragment Inhibits the DNA Repair Protein RAD51. Sci Rep. 2019;9:11227 pubmed publisher
  63. Silva M, Ferguson F, Cai Q, Donovan K, Nandi G, Patnaik D, et al. Targeted degradation of aberrant tau in frontotemporal dementia patient-derived neuronal cell models. elife. 2019;8: pubmed publisher
  64. Rice H, de Malmazet D, Schreurs A, Frere S, Van Molle I, Volkov A, et al. Secreted amyloid-β precursor protein functions as a GABABR1a ligand to modulate synaptic transmission. Science. 2019;363: pubmed publisher
  65. Grimm S, Höfner G, Wanner K. MS Binding Assays for the Three Monoamine Transporters Using the Triple Reuptake Inhibitor (1R,3S)-Indatraline as Native Marker. ChemMedChem. 2015;10:1027-39 pubmed publisher
  66. Neiens P, Höfner G, Wanner K. MS Binding Assays for D1 and D5 Dopamine Receptors. ChemMedChem. 2015;10:1924-31 pubmed publisher
  67. Imaduwage K, Go E, Zhu Z, Desaire H. HAMS: High-Affinity Mass Spectrometry Screening. A High-Throughput Screening Method for Identifying the Tightest-Binding Lead Compounds for Target Proteins with No False Positive Identifications. J Am Soc Mass Spectrom. 2016;27:1870-1877 pubmed
  68. Pellecchia M, Bertini I, Cowburn D, Dalvit C, Giralt E, Jahnke W, et al. Perspectives on NMR in drug discovery: a technique comes of age. Nat Rev Drug Discov. 2008;7:738-45 pubmed publisher
  69. Bartoschek S, Klabunde T, Defossa E, Dietrich V, Stengelin S, Griesinger C, et al. Drug design for G-protein-coupled receptors by a ligand-based NMR method. Angew Chem Int Ed Engl. 2010;49:1426-9 pubmed publisher
  70. Aguirre C, Cala O, Krimm I. Overview of Probing Protein-Ligand Interactions Using NMR. Curr Protoc Protein Sci. 2015;81:17.18.1-24 pubmed publisher
  71. Oxenoid K, Chou J. A functional NMR for membrane proteins: dynamics, ligand binding, and allosteric modulation. Protein Sci. 2016;25:959-73 pubmed publisher
  72. Lee J, Park Y, Lee K. Hydrogen-deuterium exchange mass spectrometry for determining protein structural changes in drug discovery. Arch Pharm Res. 2015;38:1737-45 pubmed publisher
  73. Irwin J, Shoichet B. ZINC--a free database of commercially available compounds for virtual screening. J Chem Inf Model. 2005;45:177-82 pubmed
  74. Lyne P. Structure-based virtual screening: an overview. Drug Discov Today. 2002;7:1047-55 pubmed
  75. Lee S, Booe J, Pioszak A. Structural insights into ligand recognition and selectivity for classes A, B, and C GPCRs. Eur J Pharmacol. 2015;763:196-205 pubmed publisher
  76. Shonberg J, Kling R, Gmeiner P, Löber S. GPCR crystal structures: Medicinal chemistry in the pocket. Bioorg Med Chem. 2015;23:3880-906 pubmed publisher
  77. Jazayeri A, Dias J, Marshall F. From G Protein-coupled Receptor Structure Resolution to Rational Drug Design. J Biol Chem. 2015;290:19489-95 pubmed publisher
  78. Shimamura T, Shiroishi M, Weyand S, Tsujimoto H, Winter G, KATRITCH V, et al. Structure of the human histamine H1 receptor complex with doxepin. Nature. 2011;475:65-70 pubmed publisher
  79. Hanson M, Roth C, Jo E, Griffith M, Scott F, Reinhart G, et al. Crystal structure of a lipid G protein-coupled receptor. Science. 2012;335:851-5 pubmed publisher
  80. Kruse A, Hu J, Pan A, Arlow D, Rosenbaum D, Rosemond E, et al. Structure and dynamics of the M3 muscarinic acetylcholine receptor. Nature. 2012;482:552-6 pubmed publisher
  81. Grimes L, Young M. Purinergic P2X receptors: structural and functional features depicted by X-ray and molecular modelling studies. Curr Med Chem. 2015;22:783-98 pubmed
  82. Habermacher C, Dunning K, Chataigneau T, Grutter T. Molecular structure and function of P2X receptors. Neuropharmacology. 2016;104:18-30 pubmed publisher
  83. Tatulian S. Structural Dynamics of Insulin Receptor and Transmembrane Signaling. Biochemistry. 2015;54:5523-32 pubmed publisher
  84. Carlsson J, Yoo L, Gao Z, Irwin J, Shoichet B, Jacobson K. Structure-based discovery of A2A adenosine receptor ligands. J Med Chem. 2010;53:3748-55 pubmed publisher
  85. Yuan G, Gedeon N, Jankins T, Jones G. Novel approaches for targeting the adenosine A2A receptor. Expert Opin Drug Discov. 2015;10:63-80 pubmed publisher
  86. Carlsson J, Coleman R, Setola V, Irwin J, Fan H, Schlessinger A, et al. Ligand discovery from a dopamine D3 receptor homology model and crystal structure. Nat Chem Biol. 2011;7:769-78 pubmed publisher
  87. Mysinger M, Weiss D, Ziarek J, Gravel S, Doak A, Karpiak J, et al. Structure-based ligand discovery for the protein-protein interface of chemokine receptor CXCR4. Proc Natl Acad Sci U S A. 2012;109:5517-22 pubmed publisher
  88. Senisterra G, Chau I, Vedadi M. Thermal denaturation assays in chemical biology. Assay Drug Dev Technol. 2012;10:128-36 pubmed publisher
  89. Alam M, Sánchez Azqueta A, Janha O, Flannery E, Mahindra A, Mapesa K, et al. Validation of the protein kinase PfCLK3 as a multistage cross-species malarial drug target. Science. 2019;365: pubmed publisher
  90. Rosa N, Ristic M, Seabrook S, Lovell D, Lucent D, Newman J. Meltdown: A Tool to Help in the Interpretation of Thermal Melt Curves Acquired by Differential Scanning Fluorimetry. J Biomol Screen. 2015;20:898-905 pubmed publisher
  91. Desantis K, Reinking J. Use of Differential Scanning Fluorimetry to Identify Nuclear Receptor Ligands. Methods Mol Biol. 2016;1443:21-30 pubmed publisher
  92. Huynh K, Partch C. Analysis of protein stability and ligand interactions by thermal shift assay. Curr Protoc Protein Sci. 2015;79:28.9.1-14 pubmed publisher
  93. Martinez Molina D, Jafari R, Ignatushchenko M, Seki T, Larsson E, Dan C, et al. Monitoring drug target engagement in cells and tissues using the cellular thermal shift assay. Science. 2013;341:84-7 pubmed publisher
  94. Seashore Ludlow B, Lundbäck T. Early Perspective. J Biomol Screen. 2016;21:1019-1033 pubmed publisher
  95. Ross P, Weihofen W, Siu F, Xie A, Katakia H, Wright S, et al. Isothermal chemical denaturation to determine binding affinity of small molecules to G-protein coupled receptors. Anal Biochem. 2015;473:41-5 pubmed publisher
  96. Torres F, Recht M, Coyle J, Bruce R, Williams G. Higher throughput calorimetry: opportunities, approaches and challenges. Curr Opin Struct Biol. 2010;20:598-605 pubmed publisher
  97. Rajarathnam K, ROSGEN J. Isothermal titration calorimetry of membrane proteins - progress and challenges. Biochim Biophys Acta. 2014;1838:69-77 pubmed publisher
  98. Bhogaraju S, Bonn F, Mukherjee R, Adams M, Pfleiderer M, Galej W, et al. Inhibition of bacterial ubiquitin ligases by SidJ-calmodulin catalysed glutamylation. Nature. 2019;572:382-386 pubmed publisher
  99. Krainer G, Keller S. Single-experiment displacement assay for quantifying high-affinity binding by isothermal titration calorimetry. Methods. 2015;76:116-23 pubmed publisher
  100. Ma Z, Zhu L, Song T, Wang Y, Zhang Q, Xia Y, et al. A paralogous decoy protects Phytophthora sojae apoplastic effector PsXEG1 from a host inhibitor. Science. 2017;355:710-714 pubmed publisher
  101. Doblas V, Smakowska Luzan E, Fujita S, Alassimone J, Barberon M, Madalinski M, et al. Root diffusion barrier control by a vasculature-derived peptide binding to the SGN3 receptor. Science. 2017;355:280-284 pubmed publisher
  102. Stegmann M, Monaghan J, Smakowska Luzan E, Rovenich H, Lehner A, Holton N, et al. The receptor kinase FER is a RALF-regulated scaffold controlling plant immune signaling. Science. 2017;355:287-289 pubmed publisher
  103. Eubelen M, Bostaille N, Cabochette P, Gauquier A, Tebabi P, Dumitru A, et al. A molecular mechanism for Wnt ligand-specific signaling. Science. 2018;361: pubmed publisher
  104. Rocha Gaso M, March Iborra C, Montoya Baides A, Arnau Vives A. Surface generated acoustic wave biosensors for the detection of pathogens: a review. Sensors (Basel). 2009;9:5740-69 pubmed publisher
  105. Saitakis M, Tsortos A, Gizeli E. Probing the interaction of a membrane receptor with a surface-attached ligand using whole cells on acoustic biosensors. Biosens Bioelectron. 2010;25:1688-93 pubmed publisher
  106. Wu C, Du L, Wang D, Wang L, Zhao L, Wang P. A novel surface acoustic wave-based biosensor for highly sensitive functional assays of olfactory receptors. Biochem Biophys Res Commun. 2011;407:18-22 pubmed publisher
  107. Fang Y, Ferrie A. Label-free optical biosensor for ligand-directed functional selectivity acting on beta(2) adrenoceptor in living cells. FEBS Lett. 2008;582:558-64 pubmed publisher
  108. Fang Y. Label-Free Receptor Assays. Drug Discov Today Technol. 2011;7:e5-e11 pubmed
  109. Goral V, Jin Y, Sun H, Ferrie A, Wu Q, Fang Y. Agonist-directed desensitization of the β2-adrenergic receptor. PLoS ONE. 2011;6:e19282 pubmed publisher
  110. Goral V, Wu Q, Sun H, Fang Y. Label-free optical biosensor with microfluidics for sensing ligand-directed functional selectivity on trafficking of thrombin receptor. FEBS Lett. 2011;585:1054-60 pubmed publisher
ISSN : 2329-5139