Você está na página 1de 17

Diverse Biomolecular and Biomedical Applications of NMR-Review

Natural Product Communications

NMR Methods to Characterize Protein-


May 2019: 1–17
© The Author(s) 2019
Article reuse guidelines:
Ligand Interactions ​sagepub.​com/​journals-​permissions
DOI: 10.1177/1934578X19849296
​journals.​sagepub.​com/​home/​npx

Sanhita Maity1, Ravi Kumar Gundampati1, and


Thallapuranam Krishnaswamy Suresh Kumar1

Abstract
Structural information pertaining to the interactions between biological macromolecules and ligands is of potential signifi-
cance for understanding of molecular mechanisms in key biological processes. Recently, nuclear magnetic resonance (NMR)
spectroscopic techniques has come of age and has widened its scope to characterize binding interactions of small molecules
with biological macromolecules especially, proteins. NMR spectroscopy-based techniques are versatile due to their ability to
examine weak binding interactions and for rapid screening the binding affinities of ligands with proteins at atomic resolution.
In this review, we provide a broad overview of some of the important NMR approaches to investigate interactions of small
organic molecules with proteins.

Keywords
protein-small molecule interactions, NMR methods, binding constant (KD)

Received: November 8th, 2018; Accepted: January 21st, 2019.

Specific molecular interactions of biological macromole- investigate protein-ligand binding affinities in the nanomolar
cules with ligands of different sizes are central to the under- to millimolar regime.6-9 An additional advantage of NMR
standing of the regulation of key cellular processes.1 Proteins has been its ability to not only provide information on bind-
are the most important class of biological macromolecules as ing affinity between ligands and their target protein(s) but
they govern the crucial structural and functional properties also its strength to shed valuable insights into the protein
of cellular systems. A good understanding of the molecular conformational changes that plausibly occur upon binding to
mechanisms underlying important cellular processes will their ligand(s). Therefore, the aim of this review is to provide
obviously involve examination of structural interactions a comprehensive list of commonly used NMR-based tech-
forged by proteins with themselves or with other biological niques that can be used to investigate different aspects of
macromolecules such as, nucleic acids, polysaccharides, and protein interactions with small molecule organic ligands.
lipids. Owing to the central role of proteins as drivers of cel-
lular functions, specific interactions of small molecular
organic ligands with proteins are critical for a rational drug
1D-1H Line Broadening
design. Several biophysical and biochemical techniques, Ligand-observed 1D NMR methods are well established to
such as isothermal titration calorimetry, surface plasmon res- detect hits from chemical libraries because they prefer
onance, fluorescence resonance energy transfer, infrared ligands with KD values in the micromolar to millimolar range
spectroscopy, circular dichroism, confocal microscopy, and require lower data-acquisition times and reduced sample
enzyme-linked immunosorbent assay, have been success- requirements.10 The dissociation equilibrium constant for
fully used to characterize the binding constant (KD), kinetics, protein-ligand complex can be measured by a change in
dynamics, and thermodynamics characterizing protein-li-
gand interactions. In addition, computational studies have
also proved to be useful in the virtual high throughput 1
Department of Chemistry and Biochemistry, University of Arkansas,
screening of potential small molecule drug candidates Fayetteville, AR, USA
against protein targets.2 However, NMR spectroscopy has
Corresponding Author:
proven to be the most valuable tool to obtain an atomic-level Thallapuranam Krishnaswamy Suresh Kumar, Department of Chemistry
view of the ligand-protein interactions.3-5 NMR spectros- and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA.
copy evolved as a powerful tool over the past few decades to Email: ​sthalla@​uark.​edu

Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0
License (http://www.​creativecommons.​org/​licenses/​by-​nc/​4.​0/) which permits non-commercial use, reproduction and distribution of the work without
further permission provided the original work is attributed as specified on the SAGE and Open Access pages (https://​us.​sagepub.​com/​en-​us/​nam/​open-​
access-​at-​sage).
2 Natural Product Communications

(wB), and as a result, the increased linewidth causes a corre-


sponding decrease in the ligand peak height which is mea-
sured by the ratio of the NMR peak height (R). One can
obtain KD value from this equation (equation (2)) on the
basis of the relative changes in the ligand peak height.
Shortridge et al. rearranged equation-2 for measuring KD
(see equation (3)) values. The rearranged equation can also
be written in terms of fractional occupancy at a given protein
concentration (Rgiven) as,
[( [ ] ) ]
c P T [ ] [ ]
KD = Rgiven − c P − L T
(3)
‍ ‍

For proteins which possess multiple non-specific binding


sites, the nonspecific binding term n[P]T corresponds to a lin-
ear increase in fraction bound upon addition of protein,
accounts for the correction of the decrease in ligand signal at
Figure 1.  1D - 1H nuclear magnetic resonance (NMR) high protein concentration. After the addition of the n[P]T
spectra phenytoin (a) and naproxen (b) titrated with increasing term, equation (2) can be represented as
concentrations [(i) 0 µM, (ii) 0.4 µM, (iii) 1 µM, (iv) 2 µM, and (v) 4
µM] of HSA. The height of the NMR signal of the ligands decrease HB 1[ ]
[ ]
R= 1− HF = 1− +n P T
because of the shorter relaxation times of the ligand-human c P
1+ [L] +KT (4)
serum albumin complex.11 This figure has been reproduced from ‍ T D ‍
ref.11 with permission from the publisher.
However, the 1D -1H NMR spectra of small molecules
have extremely sharp peaks. However, upon binding to pro-
linewidth or chemical shift of the peaks corresponding to the tein(s), widening of the peak and subsequently decrease in
free ligand upon binding to a protein. Binding interactions the ligand’s NMR signal height are observed due to shorter
between a protein (MW >5000 Da) and a low molecular relaxation time of the ligand-protein complex. . Human
weight ligand (MW <500 Da) can be measured by monitoring serum albumin (HSA), which possesses multiple binding
the decrease in the NMR peak height of ligand upon addition sites, experiences 1D-1H line broadening effect upon binding
of ligands to a protein solution. Ligand that shows an observ- to small molecule drugs phenytoin and naproxen (Figure 1).11
able decrease in peak height is considered to be a binder. The observed increase in the linewidth of the ligand
Therefore, the KD values of a 1:1 protein (P)- ligand (L) depends on the dissociation equilibrium constant for the pro-
binding complex, can be represented by the equation, tein-ligand interaction, KD. In general, the observed change
in the ligand’s linewidth (wobs) for the fast exchange limit
‍ KD = [L]F [P]F /[PL]‍ (1) will follow the equation shown below
wherein [L]F is the concentration of the free ligand, [P]F ‍ wobs = wF + fB (wB − wF ) where fB ≈ [P]T /([L]T + KD )‍(5)
is the concentration of the free protein at equilibrium, and
[PL] is the concentration of the protein-ligand complex. wherein fB is the fraction of the bound protein-ligand
Shortridge et al., assuming that [L]T > [P]T (wherein [P]T is complex, wF is the free ligand NMR line-width, and wB is
total protein concentration and [L]T is the total ligand con- the line-width of the bound state of the ligand.
centration) transformed equation-1 using the Taylor series There are some assumptions considered here. If it is
expansion10 to calculate the NMR peak height ratio ‘R’ as, assumed that the linewidth of the protein-ligand complex is
higher than that for the free ligand, then the ratio of the
HB c[P]T
R= 1− = 1 − 1/(1 + [L]T +KD )‍ (2) ligand linewidth in the free state and in the bound state rep-
‍ HF
resent the remaining free ligand concentration which is
wherein‍c = wBwF − 1‍ expressed by equation (2). This assumption is valid for
HF and HB are the sum of NMR peak heights of ligand in ligand screening against a single protein target wherein the
the free state and the bound state, respectively. wF and wB are contribution of chemical and dynamic line-broadening of the
the NMR-linewidth(s) of the ligand in their free and bound ligand(s) may remain almost constant. As a matter of fact,
states, respectively. NMR line width ratio constant (c) rep- the lower contribution of linewidth from the exchange pro-
resents the change in the ligand linewidth upon binding of a cesses is not expected to change the ligand binding affinities.
ligand to a protein. In the bound state, the resonance of the This assumption is applicable for protein-ligand interaction
free ligand linewidth (wF) gains the linewidth of the protein for single binding-site of a protein. The assumption is not
Maity et al. 3

applicable for multisite binding or other factors such as observation and quantification of such effects needs well-sep-
allosteric interactions which would warrant a more complex arated signals.16 Another experimental approach was pro-
binding model.12,13 Upon binding to a protein target, the 1H posed that involves subtraction of the spectra responsible for
NMR resonances of the ligand would broaden significantly contributions stemming from binding ligands. However, this
or even disappear but background interference should be approach requires the inclusion of critical steps that are often
expected to increase with concentration and the size of pro- time-consuming.17 As binding induces relatively minor
tein accounting for the enhancement in false-negative rates. chemical shift perturbations (CSPs), as compared to the line-
Worley et al. reported a statistical method uncomplicated sta- width changes, the effects of relaxation rate are more effec-
tistical spectral remodeling (USSR) method to analyze tive and therefore preferred. A spin-labeled ligand is first
1D-1H NMR line-broadening spectra for removing interfer- selected that has a propensity to bind to the primary binding
ing protein baseline signals.14 It was reported that a chemical site of a protein. This ligand is used for screening for binding
library of 456 compounds identified 43 binders using BSA of other ligands to second binding site of protein(s).18 As a
but the background signals of BSA completely concealing consequence, T2 relaxation is enhanced on other ligand by
the ligand spectra.14 Identification of the protein baseline the presence of the spin labeled first ligand and this effect
from screening data requires highly reproducible sample depends on the distance between the spin label and the sec-
preparation, data collection and processing. The later of ond binding site on the protein. As a matter of fact, due to
these requirements can be achieved by the use of phase-scat- fast T2 relaxation, resonance signals of ligands that bind to a
ter correction (PSC) method.15 When protein signals are the second protein binding site is not detected in the T2-filtered
major problem, PSC methods yield the good results com- NMR spectrum. The binding of ligands with FKBP was
pared to the USSR method. Therefore, Worly et al. reported investigated by Jahnke et al. wherein spin labels (such as,
that the combined use of PSC and USSR method would pro- 2,2,6,6-tetramethylpiperidine-1-oxyl; TEMPO) were
vide the convenient analysis for 1H line-broadening screen- attached to the lysine side chains of FKBP, a FK506 binding
ing data in order to estimate the KD values.14 protein, to detect the binding ligands and these lysine resi-
dues were situated in the proximity of FK506 binding site
[Figure 2]. Therefore, any ligand that binds to this site would
Relaxation Time to Identify Ligand Binding experience increased T2 relaxation. This method was applied
The binding of ligand to the protein can be assessed by to a complex mixture of two p-hydroxybenzanilide and 4
line-broadening effect as described earlier. Line-broadening nonbinding aromatic compounds. Interestingly, the binding
effect also has limitation(s) for the detection of binding affinity of p-hydroxybenzanilide to FKBP was detected by
activity of one or more components in a complex mixture. using spin-labeled FKBP because p-hydroxybenzanilide
Therefore, in the investigation of complex mixtures, the experienced significant quenching in the presence of

Figure 2.  Relaxation experiments of a complex mixture of 2 p-hydroxybenzanilide and 4 nonbinding aromatic compounds without
FKBP (left), unlabeled FKBP (middle), and spin-labeled FKBP (right) with spin lock periods of 10 ms and 200 ms. Arrows indicate the
cancelation of signals generating from compounds with binding activity for FKBP.18 This figure has been reproduced from ref.18 with
permission from the publisher.
4 Natural Product Communications

spin-labeled FKBP. However, the method is limited when binding site exhibit significant 1H-15N CSP. Figure 3 represents
covalent modification of the protein is required because it the titration of new acidic fibroblast growth factor (n FGF-1)
affects the binding affinity.19 with sucrose octasulfate (SOS), and 2D 15N-HSQC spectra
were acquired at a nFGF-1 concentration of 1 mM. Sometimes
prominent 1H-15N CSP is also observed due to backbone con-
Chemical Shift Perturbations formational changes induced in the protein by the binding
Several parameters of protein and ligand change upon inter- ligand.25 In addition, 1H-15N CSP also furnishes information of
action of a protein with its ligand. (CSP) can occur due to the allosteric changes in the protein due to complexation to a
spatial proximity of aromatic rings or due to the non-cova- ligand. For the calculation of CSP two approaches are used.
lent interactions with ligands and solvent molecules. In this One of them is the quantum chemical approach using Gaussian
type of experiment, the chemical shift of the protein can be 98.26 In this method, CSP can be calculated from the shielding
assessed when ligand is titrated against protein into the solu- of nuclei from the external magnetic field by their electrons.
tion.20,21 Saturation of the protein binding site is observed The problem with this method is the accuracy and also it does
when the ligand is titrated into the protein in excess. The not provide significant information on the structural features
standard technique of 15N-heteronuclear single quantum cor- that generated the calculated shift. The second method is an
relation (HSQC) titration requires 15N labeled protein pro- empirical one wherein the chemical shifts are the summation of
duced in Escherichia coli with unlabeled ligand. The changes independent effects but any problem(s) in experimental data
in chemical shift during titration can be monitored by acquir- leads to errors in calculation such as, differences between cal-
ing 2D 15N-HSQC spectra within 30 minutes by using pro- culated and the experimentally observed values.27 This is pri-
tein concentrations in the range 200 µM.22 When interactions marily because the calculated values are based on crystal
occur, specific 1H-15N cross peaks in the 15N-HSQC spectra structures which are in variance with the ensemble of solution
are displaced from their original position. CSPs allows cal- structures which are calculated from the solution weighted
culation of equilibrium dissociation constant (KD) and also average of the chemical shift changes. In general, protein (P)
the ligand binding sites.23 It is essential to maintain some of binds to ligand (L) and forms a protein-ligand complex, P +
the physical parameters to remain constant during the course L↔PL, wherein the rate constant for forward and backward
of the experiment because small changes in sample pH, salt reactions are represented as kon and koff. At equilibrium, KD is
concentration, temperature and buffer composition will sig- equal to koff/ kon.28 In a HSQC spectrum representing weak
nificantly affect the 1H-15N CSP. interactions/fast exchange, the koff value is higher than the
When the ligand is added, some of the residues undergo chemical shift difference between the free and bound protein.
selective 1H-15N CSP.24 Generally, backbone amide protons of As a consequence, the protein-ligand complex is only stable for
residues in the protein located in close proximity to the ligand a short period of time. This aspect poses a challenge to acquire
reliable NMR data. Therefore, the observed chemical shift is
subjected to the weighted average of the chemical shifts of pro-
tein in free state and in the bound state. When koff is lower than
the chemical shift difference or for slow exchange (very strong
interactions), signals either for free protein or protein-ligand
binary complex are observed (Figure 4).
However, CSP of any protein signal measured at different
ligand concentrations can be used in a nonlinear least-square
fitting to predict the KD value using the following equation
reported by Becker et al.29 The equation can be expressed as
[ √
([P]T +[L]T +KD )
∆∂obs = ∆∂max 2[P]T − ([P]T + [L]T + KD )2 −
√ ] (6)
(4[P]T [L]T /2[P]T )
‍ ‍

wherein ‍∂ obs‍represents the change in the observed shift


relative to the free state, ‍∂ max ‍is the maximum shift change
when the protein is saturated with the ligand, [P]T is total
protein concentration and [L]T is total ligand concentration.

Figure 3.  Overlap of 1H–15N HSQC spectra of newt FGF-


1(red) in the presence of sucrose octasulfate (SOS) (cyan). The Paramagnetic Ligand Tagging
labeled residues are those in newt FGF-1 that undergo significant
chemical shift perturbations when the protein binds to SOS in Ligand-transferred paramagnetic NMR is a commonly used
a 1:1 ratio.25 This figure has been reproduced from ref.25 with sensitive technique for the identification of protein binding
permission from the publisher. site of low affinity drugs. Chemical shift of a protein is
Maity et al. 5

Figure 4.  The effect of exchange rate on peak shape in a 2D 1H-15N HSQC spectrum. Figure 4: depicts peaks shifts representing a fast
exchange: the peak shifts from free (blue) to bound (red) and for slow exchange: the intensity decreases of a peak in the free (blue) state
but increases (red) when the protein is bound to the ligand.

dependent on metal binding site. Paramagnetic effects are al. reported the complex between a lanthanide (Ln3+)-labeled
observed as a difference between NMR data that is acquired protein, the N-terminal domain of the subunit ε bound to the
in the presence of paramagnetic metal ion and a diamagnetic subunit ϴ of E. coli DNA polymerase III (ε186/ ϴ), and a
metal ion. The data acquired in the presence of the diamag- small ligand molecule, thymidine.31 Stoichiometric com-
netic metal ion serves as reference. However, paramagnetic plexes ε186/ ϴ /Ln3+ are easily obtained by the addition of
shifts are observed through bond interactions as well as equimolar amounts of LnCl3 [where Ln3+ are Dy3+, Tb3+, and
through space interactions. The pseudocontact shifts are sub- Er3+, (except La3+ and Lu3+ because they are used as diamag-
jected to through-space interactions when a paramagnetic netic references)]. Thymidine exchanges between the free
metal ion bound to a protein.30 This method is useful for the state and the ε186/ ϴ /Ln3+/thymidine complex state and the
detection of the paramagnetic properties of a metal ion bound PCS in the bound state are transferred to produce the aver-
to the target protein adjacent to the ligand binding site. Metal aged NMR signal in 15N-HSQC spectrum. Therefore, the
ions such as most of the lanthanide ions are paramagnetic transferred PCS and transferred paramagnetic line broaden-
(except La3+ and Lu3+) and possess suitable ionic radii to ings are useful to measure binding affinities and the structure
coordinate with the metal binding site of the target protein. of the protein-ligand complex.31 Paramagnetic tagging with
To determine the relative orientation of paramagnetic lantha- ligand also accounts for measuring binding affinities. The
nide ion in the area of binding site, pseudocontact shifts can lanthanide ion first recognizes the metal binding site of a
be expressed in terms of polar coordinates r, θ,‍ φ‍ of the protein, and then a lanthanide ion binding tag is covalently
nuclear spin with respect to the paramagnetic susceptibility anchored to a protein side chain. The transient complex of
tensor (ΔX) of the lanthanide ion. The pseudo-contact shift
lanthanide ion-ligand then interacts with the protein.
(δPCS) measured chemical shift of nuclear spin of the protein
Calmodulin-sevoflurane interaction was studied by para-
in paramagnetic and diamagnetic samples and can be repre-
magnetic tagging where the ligand sevoflurane was attached
sented as30
to DOTA.32 Brath et al. reported the interaction of complex
∂pcs = 1 [∆X (3 cos ∅ − 1) + 3/2∆X sin2 θ cos 2ϕ] of DOTA-bound sevoflurane La3+, Eu3+, Yb3+, or Dy3+ with
‍ 12πr3 ax rh
‍ calmodulin where the terminal fluorine of sevoflurane was
 (7)
substituted with oxygen because of similar electronic char-
wherein ΔXax and ΔXrh are the axial and rhombic compo- acter of oxygen and fluorine.33 Intramolecular ligand δPCS
nents of the magnetic susceptibility tensor (ΔX) of the lan- was measured for complexation Eu3+, Yb3+, or Dy3+ with
thanide ion. The angles θ and φ denote its orientation with DOTA-bound sevoflurane and the binding of the paramag-
respect to the protein coordination frame and r is the distance netically labeled drug to calmodulin binding site produced
between the lanthanoid and nuclear spin of protein. John et chemical shift changes due to the pseudocontact shift effect
6 Natural Product Communications

Figure 5.  (a) Overlay of 2D NMR 1H−13C HSQC spectra of calmodulin C-lobe titrated with the lanthanoid La3+/Dy3+ complexed
with sevoflurane analogs (DOTA-bound sevoflurane). 1H−13C HSQC spectra of 0.1 mM calmodulin C-lobe (black) titrated with 0.05
mM (blue), 0.1 mM (purple), and 0.2 mM (red) Dy3+ complexed sevoflurane analog. (b) represents δPCS of Calmodulin lobe induced by
Dy3+ labeled sevoflurane and control ligands. Amide proton δPCS for calmodulin lobes in the presence of sevoflurane ligand and control
compound complexed to Dy3+ are indicated as black and gray filled circles, respectively.33 This figure has been reproduced from 33Pwith
permission from the publisher. [https://pubs.acs.org/doi/10.1021/jacs.5b06220].

(Figure  5 (a,b)). The tagged sevoflurane was observed to provide information on the bound state of ligand that bind
possess KD of ∼4.0 mM for the N-lobe and ∼1.8 mM for the weakly (KD in µM to mM range) to the target. Transferred
C-lobe respectively. NOE measurements may increase the resolution and sig-
This method has some limitations. Selective attachment nal-to-noise ratio of intermolecular NOE cross peaks by
of the lanthanide ions to different side chain positions narrowing the linewidth in one of the two dimensions.
enhances the accuracy of the location of the binding moiety There are two important parameters for these measure-
and therefore protein site-directed mutagenesis is required. ments: (1) The mixing time should be short enough for the
Moreover, detection of the low affinity binding ligands negligible contribution of NOE signals of free ligands, but
sometimes may be challenging, because the observed para- long enough when bound to protein, (2) The molar ratio of
magnetic effect is small due to the low molar fraction of ligand molecules to the target protein. Therefore, screening
bound ligand. Modification of the structure of the ligand may of a library of compounds in the presence of a protein
influence its binding to protein, and therefore tag attachment receptor, the bound ligand molecules exhibit strong nega-
at several positions of the ligand may be necessary. tive NOEs, whereas non-binders will show weak positive
NOEs.35 The trNOE cross peaks arise due to intramolecular
interactions between protons in the bound ligand or inter-
The Transferred-NOESY, ILOE, and NOE molecular interactions between protons in the protein and
Pumping Experiment protons of the ligand. The trNOE approach has been used
The 1H-1H NOESY experiment attributes to the correlation for studying the conformation of small ligand-large pro-
signals caused by dipolar cross-relaxation between spa- teins complexes that are characterized by fast off-rates but
tially closed nuclei.34 Large molecules produce large nega- if dissociation constant (KD) or dissociation rates (koff) are
tive NOE peaks. For small molecules (MW <500 Da) and not known, a titration as well as the recording of trNOE
for medium-sized molecules (MW range of 500 to 1000 Da) spectra at each point is essential.36
the corresponding NOE peaks are positive and very small trNOESY measurement were first employed to examine
negative, respectively. Large molecules build-up NOE the binding affinity carbohydrates to lectin, Aleuria aurantia
quickly in contrast to smaller molecules and therefore for agglutinin (AAA).37 Lucas et al. demonstrated that
large molecules the maximum NOE is shifted to shorter exchange-transferred NOE (trNOE) on diffusion time scale
mixing times. Transferred NOE (trNOEs) spectrum yields a can provide valuable information on the multiple binding
negative NOE because in the bound state (protein-ligand orientations of HSA with dansylglycine and caprylate
complex), a small molecule (ligand) adopts subsequent wherein both the ligands exhibited primary binding affinity
NOE behavior of protein. Transferred NOE measurements for HSA’s drug site II (Figure 6(b)).38
Maity et al. 7

Figure 6.  (a) 1H BPP-STE (bipolar gradient pulse pair-stimulated echo) spectra for dansylglycine (DG) and caprylate (CAP) at a
different ligand to HSA ratio. (i) 25:1 CAP:HSA, (ii) 25:1 DG:HSA, (iii) 25:1:1 DG:CAP:HSA, and (iv) 25:25:1 DG:CAP:HSA. The small
peaks observed in (ii) and (iii) between 0 and 2.5 ppm are spectral subtraction artifacts. (b) represents NOESY spectrum for the 25:25:1
DG:CAP:HSA solution. All cross-peaks are the same phase as the diagonal. Intra-ligand cross-peaks (circled) arise from trNOE and
indicate binding but ilNOE cross-peaks (boxed) between the two ligands suggest that they are their binding sites on the protein are in
close spatial proximity.38 This figure has been reproduced from ref.38 with permission from the publisher.

In general, NOESY experiments are recorded in the pres- show (Bipolar gradient pulse pair-stimulated echo) BPP-
ence of two ligands that bind simultaneously to adjacent STE pulse sequence and diffusion-assisted NOE pumping
binding pockets of the protein forming a ternary complex, to pulse sequence, respectively.42 Diffusion-assisted NOE-
yield intermolecular ligand-ligand NOEs (ILOEs).39 ILOEs pumping experiments were used for interactions of HSA
were successfully used to examine the complexation of with a binding ligand (salicylic acid) and nonbinding ligands
NAD+ and lycolate to lactate dehydrogenase.40 Intermolecular (l-ascorbic acid and glucose).42,43 1D- 1H NMR spectrum of
trNOEs are important for the analysis of protein-ligand inter- salicylic acid, L-ascorbic acid, glucose, and HSA in D2O
actions. There are two effects such as spin diffusion and with and without BPP-STE is shown in Figure 8a and stack
magnetic leakage to the protein can obscure the analysis of plot of 1D 1H spectra of HSA with three ligands is shown in
the bound conformations of low-molecular weight ligands. (Figure 8 (b)).42 A standard set-up of BPP-STE diffusion-fil-
As spin diffusion and leakage of magnetism to the protein are tered experiment (Figure  8 (b)) is unable to distinguish a
the results of intermolecular NOE between protein and mixture of binding ligand (Salicylic acid) and non-binding
ligand. Therefore, in case of spin diffusion, false confirma- ligands (L-ascorbic acid and glucose) with HSA because of
tions of the complex may be obtained. Magnetic leakage to insignificant differences in diffusion co-efficient for 3 ligands
the protein, which occurs due to use of longer mixing times, whereas in NOE-pumping experiment, sufficient gradient is
an artifactual change in the conformation of the ligand-pro- applied to discriminate the binding ligand signal from the
tein complex may be observed.19 Several techniques have unbinding ligands signals, therefore binding interaction of
been developed to diminish these two effects.41 salicylic acid with HSA is easily detected. NOE-pumping
Diffusion-assisted NOE-pumping technique relies on experiment can successfully identify the binding ligands
NOE to transfer the signal from the target protein to the whereas the affinity NMR method fails to discriminate
bound ligands wherein signal of the non-binding ligands can between a binding and non-binding ligands.
be suppressed through diffusion filter.42 The non-binding A reverse of the previous experimental set-up was also
ligands are separated by the diffusion experiment at the reported wherein for the first set-up involves intermolecular
beginning of the NOE experiment and at the end of the NOE trNOEs between ligand protons and protein protons are sup-
experiment, only bound ligand signal is identified because pressed. In the second reference set-up, the T2 filter is applied
the magnetization is transferred from protein to the bound after the mixing period so that during the mixing time inter-
ligand only. After ligand dissociation from the protein, the molecular trNOEs build up and lead to a decrease of the
delivered magnetization is preserved by the relatively long ligand signal intensities.44 As a result, the difference of those
T1 of the ligands in the free state. As a consequence, ligand experiments provide signals of the ligands which are specif-
signals can still be observed at shorter T2. Figure  7 (a,b) ically bound to the protein.
8 Natural Product Communications

Figure 7.  (a and b) represent pulse sequence for the BPP-STE (bipolar gradient pulse pair-stimulated echo) diffusion-assisted NOE
pumping experiments.42 This figure is reproduced from ref.42 with permission from the publisher.

Interligand NOE for Pharmacophore competitively but not simultaneously to the same target pro-
Mapping (INPHARMA) tein, tubulin.45
In general, during the mixing time in NOESY experiment,
INPHARMA peaks are observed when two ligands can bind
ligand LA binds to the receptor protein P and magnetization
competitively to the same binding pocket of a target protein.
INPHARMA method is observed through spin diffusion is transferred from its proton HLA to the receptor proton HP
mediated (via protons of protein), transferred NOEs between and ligand LA dissociates from the receptor protein P and
two ligands that bind competitively to the same target mole- then ligand LB binds into the same binding site of the target
cule. This method was first successfully used for two ligands protein. Subsequently, magnetization transfers from the
such as epothilone A and baccatin III which can bind receptor proton HP to a proton in the ligand LB (HLB)

Figure 8.  (a) (Top panel) 1D- 1H NMR spectrum and (Bottom panel) 1D- 1H NMR spectrum with BPP-STE of 10 mM salicylic acid
(S), 10 mM L-ascorbic acid (A), 10 mM glucose (G), and 100 µM HSA in D2O. (b) stack plot of 1D- 1H spectra of HSA (at 300 K) in the
presence of three ligands as the mixing time tm is increased from 5 ms to 1.2 s in intervals of 0.05 s.42 This figure is reproduced from
ref.42 with permission from the publisher.
Maity et al. 9

Figure 9.  (a), Schematic presentation of the INPHARMA method, two competitive binding ligands LA and LB can bind to the same
receptor (P) consecutively but not simultaneously. (b) represents the schematic representation of LOGSY-titration method, for reversibly
binding ligands magnetization received from water during the WaterLOGSY experiment is a combination of cross-relaxation obtained
from bulk water and cross-relaxation delivered via protein-bound water molecule as proposed in ref.46

resulting in the observation of an intermolecular transferred Therefore, difference in the spectrum (SSTD=S0-SSAT) only
NOE between HLA and HLB through spin diffusion-mediated measures the signal of the ligand that received saturation
effect via a proton of the protein. Figure  9a shows two transfer from the protein as compared to non-binding ligands.
ligands, LA and LB, bind to a common target consecutively However, in the presence of large excess of ligand, the satu-
and not simultaneously. This method is particularly useful to ration of free ligands in solution gets amplified because the
optimize the ligands from chemical library as it detects rela- relaxation of small molecules is slower than the saturation
tive orientation of different compounds in the same target transfer. Viegas et al. analyzed STD effect in terms of ampli-
binding site. fication factor (ASTD) which can be expressed in the follow-
ing form,49
Saturation Transfer Difference (STD) NMR S0 −SSAT
[ ]
L T SSTD
[ ]
L T
ASTD = × [ ] = × [ ] (8)
Spectroscopy ‍ S0 P S0 P ‍

The STD-NMR screening technique is useful to identify


wherein ASTD is the amplification factor which is defined
ligand moieties that are important for binding to target pro-
as the average number of ligand molecules saturated per
teins. The STD-NMR experiment is observed on the ligand-
molecule of receptor. In equation (8), SSTD/S0 denotes the
based resonance signals and the technique is based on the
relative STD effect of ligand concentration[L]T which is
principle of nuclear Overhauser effect (NOE). The target
present excess of the protein [P] at a molar ratio of [L]T/[P].
protein is irradiated with a radiofrequency field which selec-
Viegas et al. rearranged equation (8) for measuring KD value
tively hits resonances of the protein generally known as
and which can be represented as
on-resonance spectrum. Therefore, in the fast exchange
between ligand in the free and in the bound state, the satura- [ ]
aSTD L
tion is delivered to the bound ligand via the protein and that ASTD = [ ]
KD + L ‍ (9)
‍
saturation is carried over to the free ligand. As a conse-
quence, the second spectrum has to be collected wherein sat- ‍aSTD ‍is the maximum amplification factor and this expres-
uration takes place off-resonance. Therefore, the difference sion is valid if [L] = [L]T. It was predicted that observed KD
measured in STD spectrum represents resonance which orig- value changes with saturation time and accurate KD values
inates selectively from the saturated ligand in ligand-protein could only be obtained for experiments set up at saturation
complex.47 STD NMR was reported for the interaction of times that were close to zero. The deviation of KD was sub-
Alisertib (MLN8237), an orally administered inhibitor of jected to the rebinding of partially saturated ligands to the
Aurora A kinase, with HSA.48 A schematic representation of protein during the protein irradiation period. Moreover,
STD method is shown in Figure 10a. STD measures the dif- STD-NMR experiment does not distinguish the specific and
ference of saturated peak of protein (on-resonance spectrum) non-specific binding of ligand(s).
with signal intensities SSAT, from without protein saturation A novel method was introduced based on STD-NMR for
(off-resonance spectrum), with signal intensities S0. the structural determination of ligands complexed to target
10 Natural Product Communications

Figure 10.  (a) Schematic representation of the STD-NMR experiment. (b) STD-NMR spectra of 2-(3’-pyridyl)-benzimidazole in the
presence of unlabeled FKBP, perdeuterated, Ile-protonated FKBP, perdeuterated,Val-protonated FKBP, perdeuterated, Leu-protonated
FKBP, perdeuterated, Met-protonated FKBP, and perdeuterated FKBP. The resonances corresponding to the ligand are highlighted. (c)
STD-NMR spectra of uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) in the presence of unlabeled MurA, perdeuterated, His-
protonated MurA, perdeuterated, Trp-protonated MurA, and perdeuterated, Phe protonated MurA. The resonances corresponding to
UDP-GlcNAc are highlighted.50 This figure has been reproduced from ref.50 with permission from the publisher.

1
protein independent of size and composition called SOS- H of bulk water is excited and magnetization is transferred
NMR.50 This method is focused on a ligand complexed to a from 1H of bound water to 1H of bound ligand via indirect
series of proteins (deuterated except for specific amino acids) spin diffusion.53 In this experiment, compounds that can
and the amino acid composition of the ligand-binding site interact with the target protein usually yield positive reso-
can be identified, thus leading to the definition of the 3-D nances (that have the same sign as protein resonances), and
structure of the protein-ligand complex. This method is use- compounds that do not bind to the target usually show-up as
ful when XRD and conventional NMR methods fail to pro- negative resonances. Water-LOGSY experiments provide
duce the required information on the ligand binding site. information on the bound ligand population. This method
This method was successfully applied to 2 systems, FKBP- can be applied for the characterization of ligand-protein
2-(3’-pyridyl)-benzimidazole complex and MurA-uridine interactions in relatively large molecular weight systems.54
diphosphate N-acetylglucosamine complex, respectively The KD value was successfully measured for HSA-L-
(Figure 10 (b and c)).50 This method has some limitations. tryptophan complex using Water-LOGSY method. The KD
For example, for the application of this method, moderately values calculated are accurate and are consistent with those
soluble ligand is required in fast exchange NMR time scale calculated using other methods.17,55,56 Another reports
(typically KD >1 µM). Therefore in the case of “lead-like” showed that KD values became higher for titration experi-
compounds, the potency will be small or moderate.51 Thus, ment of varying concentrations of bovine serum albumin
this method may be useful in preliminary stages for ligand (BSA) with L-tryptophan.57 The deviations of KD value was
screening in the drug discovery process. Another novel also observed for binding of 3-fluorophenylboronic acid to
approach to STD-NMR was proposed by Jayalakshmi et al.52 α-chymotrypsin at higher concentrations of α-chymotrypsin
This method relies on the optimization of intensity-restrained and long mixing time. However, accurate KD value may be
procedure to predict the structures of ligand in the bound obtained at lower concentrations of protein.58 The use of
protein. This is achieved by inclusion of a structure refine- Water-LOGSY method is restricted in fragment screening
ment method involving CORCEMA (complete relaxation where determination of KD is the needful step.58 Water-
and conformational exchange matrix) calculation. This LOGSY method also is limited due to its inability to differ-
method was used for screening of carbohydrate and peptide entiate between buried and surface-accessible area of ligands.
ligands. A novel technique was emerged called LOGSY titration to
discriminate between buried and solvent-accessible area of
ligands and to map protein-bound water molecules in close
Water-LOGSY Method proximity to the ligand.59 The scheme of LOGSY titration is
Water-LOGSY is one of the powerful techniques used to shown in Figure 9 (b). The method was applied to determine
screen ligand binding to target proteins. In this experiment, buried and solvent-accessible epitopes for binding of two
Maity et al. 11

Figure 11.  LOGSY titration of 6-(3,4-Dimethoxyphenyl)−3-methyl1,2,4 triazolo[4,3-b]-pyridazine (a) and 1-Methyl-5-phenoxy-


1H-pyrazole-4-carboxylic Acid Ethyl Ester (b)with perdeuterated bromodomain 1 present in protein 4 (Brd4-BD1) ligands. 100%
corresponds to the signal with the strongest slope. X-ray crystal structures of the complexes Brd4-BD1 with ligand are shown here. H2O
molecules resolved in the electron density map and within a distance of 6 Å from the ligand are shown as blue spheres.59 This figure has
been reproduced from ref.59 with permission from the publisher.[https://pubs.acs.org/doi/abs/10.1021/acs.jmedchem.7b00845

synthetic molecules 6-(3,4-Dimethoxyphenyl)-3-methyl1,2,4 It is observed that mapping of binding site of 15-nucleo-


triazolo[4,3-b]-pyridazine and 1-Methyl-5-phenoxy-1H- tide RNA on the RNA-binding protein, Nova1 KH3,
pyrazole-4-carboxylic acid ethyl ester with bromodomain 1 obtained from cross-saturation 1H/15N-HSQC experiments
present in protein 4 (Brd4-BD1), a prominent cancer target was almost similar to the binding site predicted from the
(Figure 11 (a and b).59 The merit of this method as compared CSP method and X-ray crystallography.60 Therefore, it can
to conventional ligand-based methods is its independence of be suggested that combined use of CSP and cross-saturation
the molecular mass of the protein. data will be helpful to identify the interaction sites with high
accuracy. Ferrage et al. proposed a novel approach to the
direct determination of the likely pose of a peptide from the
Cross Saturation and Transferred Cross proline-enriched tyrosine phosphatase, onto a protein Src
Saturation homology 3 (SH3) domain of the C-terminal Src kinase
The characterization of protein-large molecules interfaces is (CSK) by utilizing frequency-selective cross-saturation
a key factor to understanding of biological mechanisms at a using a low stringency isotopic labeling method(s) and side-
molecular level. Cross-saturation experiment provides accu- chain protons as probes of the interface (Figure  12(a and
rate information on the interface of larger protein com- b).61 It is demonstrated that the advantages of the methods
plexes.46 The cross-saturation mapping of protein–large related to the enhancement of overall efficiency of satura-
molecule interfaces is based on the steady-state NOE- tion along with better coverage of interaction interface
difference experiment wherein 1H resonance signals of a could lead to the increase in accuracy of the overall results.
non-deuterated target components (protein II) are saturated The biggest advantage of this approach involves significant
and accounts for observation of magnetization of NH pro- variations of the saturation transfer between experiments
tons of a second deuterated and 15N-labeled protein (protein could be associated to the local chemical shift distribution in
I) (Figure 13(a)). However, in cross-saturation method resi- the protein to achieve site-specific restriction on both ele-
due-specific information can be helpful to provide accurate ments which will be helpful for molecular docking stud-
information about protein-large molecule interfaces but the ies.62,63 This method will be very applicable for confirmation
cost of sample preparation is high. of most occupied contacts in the particular cases wherein
12 Natural Product Communications

Figure 12.  Saturation of the signals from 13C bonded aromatic (a) and aliphatic (b) protons in side chains in SH3 by the sequence
employed for the selective saturation transfer. The ratio of intensities in HSQC with and without selective saturation is plotted along the
y axis.61 This Figure has been reproduced from ref.61 with permission from the publisher.

substantial motion is present in a complex which may lead the interaction interface, and the contact residues of protein
to design of potential ligands. However, cross-saturation I in the complex are identified by observing the decrease of
method is difficult to be applied to the protein complexes the peak intensities on a 1H–15N shift correlation spectrum
with a molecular mass greater than 150 kDa. The molecular (Figure  13(b)). The information on the contact residues is
weight limitation is overcome by introducing an extended transferred on the ligand proteins in the complex, from the
version of cross-saturation method called transferred ligand proteins in the bound state to those in the free state,
cross-saturation (TCS) method.64 In this method, saturation under a fast exchange process (kex>0.1 s).65 The TCS method
in target protein (protein II) in the complex is transferred has a higher sensitivity toward low-affinity transient interac-
significantly to the residues of the bound uniformly isotopi- tions. It is demonstrated that this method is effective for the
cally labeled (with 2H and 15N) ligand protein (protein I) at investigation of very large systems including

Figure 13.  Schematic representation of cross-saturation (a) and transferred cross-saturation (b) techniques.
Maity et al. 13

membrane-embedded protein interactions, liposomes, and In a mixture of ligands, multiple pairs of diffusion coeffi-
insoluble biomolecules.66 cients for each ligands arise from a pair of DOSY spectra in
the presence and absence of protein. Separation of diffusion
data characterizing ligands along two dimensions would
Diffusion-Ordered Spectroscopy (DOSY) allow for more efficient clustering to distinguish between
The translational diffusion coefficient of a protein is an use- ligands. Snyder et al. demonstrated the use of CoLD-CoP
ful parameter to obtain information on a protein’s hydrody- (Clustering of Ligand Diffusion Coefficient Pairs) technique
namic state in solution as well as to study conformational to identify specific ligands that bind to target protein(s).71
changes upon addition of ligands.67 In the DOSY method, it Thus CoLD-CoP technique was useful to discriminate the
is possible to measure translational diffusion in solution by binders such as, N-acetylglucosamine (GlcNAc) and trypt-
utilizing pulse-field gradients (PFGs) with higher accuracy.68 amine with lysozyme and tartrate with wheat germ acid
This method is a valuable approach for the detection of bind- phosphatase (WGAP) from non-binders. This method is not
ing of low-molecular-weight compounds to large molecular applicable when the protein binding is weak and when pro-
weight protein receptors, ie specific for molecules with weak tein concentration is limited. However, this method is partic-
binding affinity (KD>1 mM). Small molecules bound to pro- ularly useful for fragment-based ligand screening.71 Didenko
tein diffuse at the slower rate of the protein than at the typical et al. reported that 3D DOSY–heteronuclear multiple quan-
rate for small molecules. Therefore, it is possible to distin- tum coherence (HMQC) pulse sequence that uses the TROSY
guish compounds in the presence or absence of protein from (transverse relaxation-optimized spectroscopy) effect of the
a mixture of compounds according to their diffusion proper- HMQC sequence on 13C methyl-labeled proteins and is
ties.69 Ferrage et al. reported a novel pulsed field gradi- highly suitable for measuring the diffusion coefficient of
ent(PFG) NMR method for measuring the translational large proteins.72 3D DOSY–HMQC experiment is suitable
diffusion coefficient of an integral membrane protein/surfac- for a detailed analysis of the diffusion coefficients of com-
tant complex.70 The novel pulsed field gradient NMR method plex mixtures of small organic molecules.73
has advantage compared to the conventional NMR methods
with pulse field gradient because this method can be applied
to macromolecular assemblies (>25 kDa) in aqueous solu- SAR (Surface Activity Relationship) by
tion at room temperature. This new method was successfully
applied to a water-soluble complex of tOmpA, the hydropho- NMR
bic transmembrane domain of bacterial outer membrane pro- SAR by NMR-based approach is introduced for discovering
tein A, with the detergent octyltetraoxyethylene (C8E4; total high-affinity ligand binding to the individual binding sites of
mass of complex ~ 45 kDa) (Figure 14(b)). Another novel protein.74 Binding of small molecules to proximal sites in a
method was proposed for considering multiple diffusion protein are primarily identified and optimized by this method.
measurements associated with each molecule in a mixture. Small molecules are screened by 15N-1H- chemical shift

Figure 14.  (a) Pulse sequence for the measurement of diffusion coefficients using longitudinal magnetization of a heteronucleus such
as nitrogen-15 with stimulated echoes (X-STE). Narrow and wide pulses represent 90° and 180° pulses, respectively. (b), Nitrogen-15-
filtered proton spectra of [15N]-tOmpA in C8E4, pH 8) at 23°C. Arrows indicate the limits of the spectral region (7.6-8.9 ppm) over
which signals were integrated. The free induction decays were multiplied by exponentially decaying functions corresponding to 100 Hz
line broadening.70 This Figure has been reproduced from ref.70 with permission from the publisher.
14 Natural Product Communications

Figure 15.  (a), Schematic representation of SAR by NMR. (b), list of compounds with different substituents tested in 1H/15N-HSQC
binding experiments to FKBP.74 The equilibrium binding constants(KD) are given in the table for different compounds. This Figure has been
reproduced from ref.74 with permission from the publisher.

changes monitored by 1H-15N HSQC using 15N labeled pro- Among all other compounds, trimethoxyphenyl pipecolinic
teins. The subsequent step requires the optimization of the acid derivative (1) showed the highest binding affinity for
second weakly interacting ligand to an adjacent binding site FKBP (KD = 2.0 µM). The binding site was predicted for (1)
and the binding is identified via the observed chemical shift was the same as for FK506. In the presence of saturating
changes. Final step involves, structural orientation of the amount of FKBP and compound 1 binary complex, screen-
bound ligands in order to guide their linkage and maintain ing of benzanilide derivative (compound 2) was examined
this structural orientation and information to obtain high which bound to the adjacent binding site with a KD value of
binding specificity to the target compound (Figure 15(a)). about 100 µM. Self-linked fragment approach was applied to
First a 15 N-1H HSQC spectrum of the labeled protein is produce a higher-affinity ligand by covalently conjugating
collected as a reference spectrum and if the resonance signal the two fragments 1 and 2 through a variety of linkers. By
is shifted in presence of one or many other molecules with using linkers, Compound 9 is the most optimal ligand as it
respect to reference spectrum it is marked as a sign of bind- shows the highest binding affinity (KD = 19 µM for FKBP,
ing (Figure 16). The SAR by NMR method effectively leads Figure 15, Panel-b).
to the development of highly potent compounds and this
method has been emerging as one of most powerful tech-
niques for fragment-based lead discovery (FBLD) as it pro- Conclusion
duced a large number of high-affinity ligands for target Characterization of protein-ligand interface is key to ratio-
macromolecules.75 The SAR by NMR approach has been nal drug discovery. Detailed structural analysis of
successfully used to screen ligands that bind to the FK506- ligand-protein interface will likely provide valuable clues
binding protein, FKBP.74 A number of compounds from a for the design of specific and potent small molecule antag-
designated chemical library were first screened for binding. onists against key cellular processes associated with
Maity et al. 15

Figure 16.  An overlay of 1H/15N-HSQC spectra for FKBP in the absence (magenta contours) and presence (black contours) of
benzanilide derivative (compound 2) with saturating amounts of trimethoxyphenyl pipecolinic acid derivative (Compound 1)(2 mM).
1 15
H/ N CSP observed for resonance signals of the residues are indicated.74 This figure is reproduced from ref.74 with permission from
the publisher.

disease(s). NMR spectroscopy has come of age. Recent References


advancements in instrumentation technology in conjunc-
1. Du X, Li Y, Xia Y-L, et al. Insights into Protein–Ligand inter-
tion with the development of new methods has significantly
actions: mechanisms, models, and methods. Int J Mol Sci.
increased the versatility of NMR spectroscopy as the
2016;17(2):144.
method of choice in modern drug discovery. In addition,
2. Sousa SF, Ribeiro AJM, Coimbra JTS, et  al. Protein-ligand
advent of cryoprobes has significantly decreased the data
docking in the new millennium--a retrospective of 10 years in
acquisition times and sample requirements. Further, with
the field. Curr Med Chem. 2013;20(18):2296-2314.
availability of the microprobe technology, multidimen-
sional NMR data can now be acquired even under high salt 3. Diercks T, Coles M, Kessler H. Applications of NMR in drug
concentrations. Most importantly, NMR spectroscopy is discovery. Curr Opin Chem Biol. 2001;5(3):285-291.
now amenable for high throughput ligand binding studies. 4. Kumar TKS, Yu C. Monitoring protein folding at atomic reso-
The applications of NMR spectroscopy are fast expanding lution. Acc Chem Res. 2004;37(12):929-936.
and in this context the recently emerging in-cell NMR 5. Aguirre C, Cala O, Krimm I. Overview of probing protein-li-
spectroscopy offers a new avenue(s) for investigating the gand interactions using NMR. Curr Protoc Protein Sci.
effects of small molecule drug candidates under ex-vivo 2015;81:1-24.
condition. 6. Gounarides JS, Chen A, Shapiro MJ. Nuclear magnetic
resonance chromatography: applications of pulse field
Declaration of Conflicting Interests gradient diffusion NMR to mixture analysis and ligand-re-
The author(s) declared no potential conflicts of interest with respect ceptor interactions. J Chromatogr B Biomed Sci Appl.
to the research, authorship, and/or publication of this article. 1999;725(1):79-90.
7. Shapiro MJ, Wareing JR. High resolution NMR for screen-
Funding ing ligand/protein binding. Curr Opin Drug Discov Devel.
The author(s) disclosed receipt of the following financial support 1999;2(4):396-400.
for the research, authorship, and/or publication of this article: This 8. Carlomagno T. NMR in natural products: understanding con-
work was supported by the Department of Energy (grant number formation, configuration and receptor interactions. Nat Prod
DE-FG02-01ER15161), the National Institutes of Health/National Rep. 2012;29(5):536-554.
Cancer Institute (NIH/NCI) (1 RO1 CA 172631) and the NIH 9. Harner MJ, Frank AO, Fesik SW. Fragment-based drug
through the COBRE program (P30 GM103450), and the Arkansas discovery using NMR spectroscopy. J Biomol NMR.
Biosciences Institute (ABI-TKSK-2016-17). 2013;56(2):65-75.
16 Natural Product Communications

10. Peng JW, Moore J, Abdul-Manan N. NMR experiments for 28. Fersht AR. Structure and Mechanism in Protein Science. New
lead generation in drug discovery. Prog Nucl Magn Reson York: Freeman WH; 1999.
Spectrosc. 2004;44(3-4):225-256. 29. Becker W, Bhattiprolu KC, Gubensäk N, Zangger K. Investi-
11. Shortridge MD, Hage DS, Harbison GS, Powers R. Estimating gating protein-ligand interactions by solution nuclear magnetic
protein-ligand binding affinity using high-throughput screen- resonance spectroscopy. Chemphyschem. 2018;19(8):895-906.
ing by NMR. J Comb Chem. 2008;10(6):948-958. 30. Otting G. Protein NMR using paramagnetic ions. Annu Rev
12. Chen J, Ohnmacht C, Hage DS. Studies of phenytoin binding Biophys. 2010;39(1):387-405.
to human serum albumin by high-performance affinity chroma- 31. John M, Pintacuda G, Park AY, Dixon NE, Otting G.
tography. J Chromatogr B. 2004;809(1):137-145. Structure determination of protein-ligand complexes
13. Levitt MH. Spin Dynamics, Basics of Nuclear Magnetic Reso- by transferred paramagnetic shifts. J Am Chem Soc.
nance; John Wiley & Sons. UK: Chichester; 2001:520-537. 2006;128(39):12910-12916.
14. Worley B, Sisco NJ, Powers R. Statistical removal of back- 32. Graham B, Loh CT, Swarbrick JD, et al. DOTA-amide lantha-
ground signals from high-throughput (1)H NMR line-broadening nide tag for reliable generation of pseudocontact shifts in pro-
ligand-affinity screens
. J Biomol NMR. 2015;63(1):53-58. tein NMR spectra. Bioconjug Chem. 2011;22(10):2118-2125.
15. Worley B, Powers R. Simultaneous phase and scatter cor- 33. Brath U, Swamy SI, Veiga AX, Tung C-C, Van Petegem F,
rection for NMR datasets. Chemometr Intell Lab Syst. Erdélyi M. Paramagnetic ligand tagging to identify protein
2014b;131:1-6. binding sites. J Am Chem Soc. 2015;137(35):11391-11398.
16. Moore JM. NMR techniques for characterization of ligand 34. Neuhaus D, Williamson MP. The Nuclear Overhauser Effect
binding: utility for lead generation and optimization in drug in Structural and Conformational Analysis, Wiley-VCH. 2nd
discovery. Biopolymers. 1999;51(3):221-243. edition; 2000.
17. Hajduk PJ, Sheppard G, Nettesheim DG, et  al. Discovery of 35. Post CB. Exchange-transferred NOE spectroscopy and
potent nonpeptide inhibitors of stromelysin using SAR by bound ligand structure determination. Curr Opin Struct Biol.
NMR. J Am Chem Soc. 1997;119(25):5818-5827. 2003;13(5):581-588.
18. Jahnke W, Perez LB, Paris CG, Strauss A, Fendrich G, Nalin 36. del Carmen Fernández-Alonso M, Díaz D, Berbis Manuel
CM. Second-site NMR screening with a spin-labeled first Álvaro, Marcelo F, Cañada J, Jiménez-Barbero J. Pro-
ligand. J Am Chem Soc. 2000;122(30):7394-7395. tein-carbohydrate interactions studied by NMR: from molec-
19. Jahnke W, Rüdisser S, Zurini M. Spin label enhanced NMR ular recognition to drug design. Curr Protein Pept Sci.
screening. J Am Chem Soc. 2001;123(13):3149-3150. 2012;13(8):816-830.
20. Kathir KM, Kumar TKS, Yu C. Understanding the mecha- 37. Meyer B, Weimar T, Peters T. Screening mixtures for biologi-
nism of the antimitogenic activity of suramin. Biochemistry. cal activity by NMR. Eur J Biochem. 1997;246(3):705-709.
2006;45(3):899-906. 38. Lucas LH, Price KE, Larive CK. Epitope mapping and com-
21. Jayaraman G, Krishnaswamy T, Kumar S, Yu C. Binding of petitive binding of HSA drug site II ligands by NMR diffusion
nucleotide triphosphates to cardiotoxin analogue II from the measurements. J Am Chem Soc. 2004;126(43):14258-14266.
Taiwan cobra venom (Naja naja atra). Elucidation of the struc- 39. Cala O, Guillière F, Krimm I. NMR-based analysis of protein-li-
tural interactions in the dATP-cardiotoxin analogue ii complex. gand interactions. Anal Bioanal Chem. 2014;406(4):943-956.
J Biol Chem. 1999;274(25):17869-17875. 40. Li D, DeRose EF, London RE. The inter-ligand Overhauser
22. Zuiderweg ERP. Mapping protein-protein interactions in solu- effect: a powerful new NMR approach for mapping struc-
tion by NMR spectroscopy. Biochemistry. 2002;41(1):1-7. tural relationships of macromolecular ligands. J Biomol NMR.
23. McCoy MA, Wyss DF. Spatial localization of ligand bind- 1999;15(1):71-76.
ing sites from electron current density surfaces calculated 41. Krishna NR, Moseley HNBKrishna NR, Berliner LJ, eds..
from NMR chemical shift perturbations. J Am Chem Soc. Structural Computation and Dynamics in Protein NMR. . New
2002;124(39):11758-11763. York: Kluwer Academic; 1999:17. 223-307.
24. Chi Y-H, Kumar TKS, Chiu I-M, Yu C. Identification of 42. Chen A, Shapiro MJ. NOE pumping: a novel NMR technique
rare partially unfolded states in equilibrium with the native for identification of compounds with binding affinity to macro-
conformation in an all beta-barrel protein. J Biol Chem. molecules. J Am Chem Soc. 1998;120(39):10258-10259.
2002;277(38):34941-34948. 43. Brown KF, Crooks MJ. Displacement of tolbutamide, gliben-
25. Arunkumar AI, Srisailam S, Kumar TKS, et al. Structure and calmide and chlorpropamide from serum albumin by anionic
stability of an acidic fibroblast growth factor from Notophthal- drugs. Biochem Pharmacol. 1976;25(10):1175-1178.
mus viridescens. J Biol Chem. 2002;277(48):46424-46432. 44. Chen A, Shapiro MJ. NOE pumping. 2. a high-throughput
26. Frisch MJ, Trucks GW, Schlegel HB, et al. Gaussian98. Pitts- method to determine compounds with binding affinity to mac-
burgh, PA; 1998. romolecules by NMR. J Am Chem Soc. 2000;122(2):414-415.
27. Kohlhoff KJ, Robustelli P, Cavalli A, Salvatella X, Ven- 45. Sánchez-Pedregal VM, Reese M, Meiler J, Blommers MJJ,
druscolo M. Fast and accurate predictions of protein NMR Griesinger C, Carlomagno T. The INPHARMA method: pro-
chemical shifts from interatomic distances. J Am Chem Soc. tein-mediated interligand NOEs for pharmacophore mapping.
2009;131(39):13894-13895. Angew Chem Int Ed Engl. 2005;44(27):4172-4175.
Maity et al. 17

46. Shimada I. NMR techniques for identifying the interface 61. Ferrage F, Dutta K, Cowburn D. Identification of hydro-
of a larger protein-protein complex: cross-saturation and phobic interfaces in protein-ligand complexes by selec-
transferred cross-saturation experiments. Meth Enzymol. tive saturation transfer NMR spectroscopy. Molecules.
2005;394:483-506. 2015;20(12):21992-21999.
47. Mayer M, Meyer B. Group epitope mapping by saturation 62. Igarashi S, Osawa M, Takeuchi K, Ozawa S-ichiro, Shimada I.
transfer difference NMR to identify segments of a ligand Amino acid selective cross-saturation method for identification
in direct contact with a protein receptor. J Am Chem Soc. of proximal residue pairs in a protein-protein complex. J Am
2001;123(25):6108-6117. Chem Soc. 2008;130(36):12168-12176.
48. Yang H, Liu J, Huang Y, et  al. Domain-specific interactions 63. Kanamori E, Igarashi S, Osawa M, Fukunishi Y, Shimada
between MLN8237 and human serum albumin estimated by I, Nakamura H. Structure determination of a protein assem-
STD and WaterLOGSY NMR, ITC, spectroscopic, and dock- bly by amino acid selective cross-saturation. Proteins.
ing techniques. Sci Rep. 2017;7(1):45514.
2011;79(1):179-190.
49. Viegas A, Manso João, Nobrega FL, Cabrita EJ. Satura-
64. Nakanishi T, Miyazawa M, Sakakura M, Terasawa H, Taka-
tion-transfer difference (STD) NMR: a simple and fast method
hashi H, Shimada I. Determination of the interface of a large
for ligand screening and characterization of protein binding. J
protein complex by transferred cross-saturation measurements.
Chem Educ. 2011;88(7):990-994.
J Mol Biol. 2002;318(2):245-249.
50. Hajduk PJ, Mack JC, Olejniczak ET, Park C, Dandliker PJ,
65. Ueda T, Takeuchi K, Nishida N, et  al. Cross-saturation and
Beutel BA. SOS-NMR: A saturation transfer NMR-based
method for determining the structures of protein-ligand com- transferred cross-saturation experiments. Q Rev Biophys.
plexes. J Am Chem Soc. 2004;126(8):2390-2398. 2014;47(2):143-187.
51. Oprea TI, Davis AM, Teague SJ, Leeson PD. Is there a dif- 66. Yokogawa M, Takeuchi K, Shimada I. Bead-linked pro-
ference between leads and drugs? A historical perspective. J teoliposomes: a reconstitution method for NMR analyses
Chem Inf Comput Sci. 2001;41(5):1308-1315. of membrane protein-ligand interactions. J Am Chem Soc.
52. Jayalakshmi V, Rama Krishna N. CORCEMA refinement of the 2005;127(34):12021-12027.
bound ligand conformation within the protein binding pocket 67. Serdyuk IN, Zaccai NR, Zaccai J, Zaccai G. Methods in molec-
in reversibly forming weak complexes using STD-NMR inten- ular biophysics: structure, dynamics, function. Cambridge
sities. J Magn Reson. 2004;168(1):36-45. University Press; 2007.
53. Dalvit C, Fogliatto G, Stewart A, Veronesi M, Stockman B. 68. Johnson CS. Diffusion ordered nuclear magnetic resonance
WaterLOGSY as a method for primary NMR screening: spectroscopy: principles and applications. Prog Nucl Magn
practical aspects and range of applicability. J Biomol NMR. Reson Spectrosc. 1999;34(3-4):203-256.
2001;21(4):349-359. 69. Cohen Y, Avram L, Frish L. Diffusion NMR spectroscopy in
54. Antanasijevic A, Ramirez B, Caffrey M. Comparison of the supramolecular and combinatorial chemistry: an old parame-
sensitivities of WaterLOGSY and saturation transfer difference ter?New insights. Angew Chem Int Ed. 2005;44(4):520-554.
NMR experiments. J Biomol NMR. 2014;60(1):37-44. 70. Ferrage F, Zoonens M, Warschawski DE, Popot J-L, Bod-
55. McMenamy RH, Oncley JL. The specific binding of L-trypto- enhausen G. Slow diffusion of macromolecular assemblies
phan to serum albumin. J Biol Chem. 1958;233(6):1436-1447. by a new pulsed field gradient NMR method. J Am Chem Soc.
56. Bertuzzi A, Mingrone G, Gandolfi A, Greco AV, Ringoir S, 2003;125(9):2541-2545.
Vanholder R. Binding of indole-3-acetic acid to human serum
71. Snyder DA, Chantova M, Chaudhry S. Analysis of ligand-pro-
albumin and competition with L-tryptophan. Clinica Chimica
tein exchange by clustering of ligand diffusion coefficient pairs
Acta. 1997;265(2):183-192.
(CoLD-CoP. J Magn Reson. 2015;255:44-50.
57. Fielding L, Rutherford S, Fletcher D. Determination of
72. Didenko T, Boelens R, Rudiger SGD. 3D DOSY-TROSY to
protein-ligand binding affinity by NMR: observations
determine the translational diffusion coefficient of large protein
from serum albumin model systems. Magn Reson Chem.
complexes. Protein Eng Des Select. 2011;24(1-2):99-103.
2005;43(6):463-470.
73. Barjat H, Morris GA, Swanson AG. A three-dimensional
58. Huang R, Bonnichon A, Claridge TDW, Leung IKH. Pro-
tein-ligand binding affinity determination by the waterLOGSY DOSY–HMQC experiment for the high-resolution analysis of
method: an optimised approach considering ligand rebinding. complex mixtures. J Magn Reson. 1998;131(1):131-138.
Sci Rep. 2017;7(1):43727. 74. Shuker SB, Hajduk PJ, Meadows RP, Fesik SW. Discover-
59. Geist L, Mayer M, Cockcroft X-L, et al. Direct NMR probing ing high-affinity ligands for proteins: SAR by NMR. Science.
of hydration shells of protein ligand interfaces and its applica- 1996;274(5292):1531-1534.
tion to drug design. J Med Chem. 2017;60(21):8708-8715. 75. Dias DM, Ciulli A. NMR approaches in structure-based lead
60. Ramos A, Kelly G, Hollingworth D, Pastore A, Frenkiel T. Map- discovery: recent developments and new frontiers for tar-
ping the interfaces of Protein−Nucleic acid complexes using geting multi-protein complexes. Prog Biophys Mol Biol.
cross-saturation. J Am Chem Soc. 2000;122(46):11311-11314. 2014;116(2-3):101-112.

Você também pode gostar