Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
J Biomol NMR ; 77(1-2): 39-53, 2023 Apr.
Article En | MEDLINE | ID: mdl-36512150

Fragment-based drug discovery (FBDD) and validation of small molecule binders using NMR spectroscopy is an established and widely used method in the early stages of drug discovery. Starting from a library of small compounds, ligand- or protein-observed NMR methods are employed to detect binders, typically weak, that become the starting points for structure-activity relationships (SAR) by NMR. Unlike the more frequently used ligand-observed 1D NMR techniques, protein-observed 2D 1H-15N or 1H-13C heteronuclear correlation (HSQC or HMQC) methods offer insights that include the mechanism of ligand engagement on the target and direct binding affinity measurements in addition to routine screening. We hereby present the development of a set of software tools within the MestReNova (Mnova) package for analyzing 2D NMR for FBDD and hit validation purposes. The package covers three main tasks: (1) unsupervised profiling of raw data to identify outlier data points to exclude in subsequent analyses; (2) batch processing of single-point spectra to identify and rank binders based on chemical shift perturbations or spectral peak intensity changes; and (3) batch processing of multiple titration series to derive binding affinities (KD) by tracing the changes in peak locations or measuring global spectral changes. Toward this end, we implemented and evaluated a set of algorithms for automated peak tracing, spectral binning, and variance analysis by PCA, and a new tool for spectral data intensity comparison using ECHOS. The accuracy and speed of the tools are demonstrated on 2D NMR binding data collected on ligands used in the development of potential inhibitors of the anti-apoptotic MCL-1 protein.


Algorithms , Magnetic Resonance Imaging , Ligands , Nuclear Magnetic Resonance, Biomolecular , Drug Discovery
2.
J Pharm Sci ; 110(12): 3819-3828, 2021 12.
Article En | MEDLINE | ID: mdl-34506864

The purpose of this investigation was to highlight the utility of nuclear magnetic resonance (NMR) as a multi-attribute method for the characterization of therapeutic antibodies. In this case study, we compared results from isothermal chemical denaturation (ICD) and NMR with standard methods to relate conformational states of a model monoclonal antibody (mAb1) with protein-protein interactions (PPI) that lead to self - association in concentrated solutions. The increase in aggregation rate and relative viscosity for mAb1 was found to be both concentration and pH dependent. The free energy of unfolding (∆G°) from ICD and thermal analysis in dilute solutions indicated that although the native state predominated between pH 4 - pH 7, it was disrupted at the CH2 and unfolded noncooperatively under acidic conditions. One-dimensional (1D) 1H NMR and two-dimensional (2D) 13C-1H NMR performed, in concentrated solutions, confirmed that PPI between pH 4-7 occurred while mAb1 was in the native state. NMR corroborated that mAb1 maintained a dominant native state at formulation-relevant conditions at the tested pH range, had increased global molecular tumbling dynamics at lower pH and confirmed increased PPI at higher pH conditions. This report aligns and compares typical characterization of an IgG1 with assessment of structure by NMR and provided a more precise assessment and deeper insight into the conformation of an IgG1 in concentrated solutions.


Antibodies, Monoclonal , Immunoglobulin G , Antibodies, Monoclonal/chemistry , Hydrogen-Ion Concentration , Immunoglobulin G/chemistry , Magnetic Resonance Spectroscopy , Protein Conformation , Protein Denaturation , Viscosity
3.
J Biomol NMR ; 73(12): 675-685, 2019 Dec.
Article En | MEDLINE | ID: mdl-31541395

Protein-based NMR spectroscopy has proven to be a very robust method for finding fragment leads to protein targets. However, one limitation of protein-based NMR is that the data acquisition and analysis can be time consuming. In order to streamline the scoring of protein-based NMR fragment screening data and the determination of ligand affinities using 2D NMR experiments we have developed a data analysis workflow based on principal component analysis (PCA) within the TREND NMR Pro software package. We illustrate this using four different proteins and sets of ligands which interact with these proteins over a range of affinities. Also, these PCA-based methods can be successfully applied even to systems where ligand binding to target proteins is in intermediate or even slow exchange on the NMR time scale. Finally, these methods will work for scoring of fragment binding data using protein spectra that are either highly overlapped or lower in resolution.


Drug Discovery/methods , Nuclear Magnetic Resonance, Biomolecular/methods , Principal Component Analysis/methods , Ligands , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding
4.
J Mol Biol ; 428(3): 579-589, 2016 Feb 13.
Article En | MEDLINE | ID: mdl-26774853

STAT3 is a member of STAT (signal transducer and activator of transcription) transcription activators. Aberration in STAT3 activity due to constitutive activation or mutations leads to diseases such as cancer and hyper-immunoglobulin E syndrome (HIES). STAT3 contains several structured domains including the Src homology 2 domain (SH2), linker domain (LD), DNA-binding domain (DBD) and the coiled-coil domain. Here we report the discovery of inter-domain allosteric communications in STAT3 from studies using nuclear magnetic resonance (NMR) and other methods. We found that pTyr-peptide interactions with SH2 cause structural and dynamics changes in LD and DBD. The inter-domain allosteric effect is likely mediated by the flexibility in the hydrophobic core. In addition, a mutation in LD found in HIES (I568F) induces NMR chemical shift perturbation in SH2, DBD and the coiled-coil domain, suggesting conformational changes in these domains. Consistent with conformational changes in SH2, the I568F mutant reduces SH2's binding affinity to a pTyr-containing peptide. This study provides an example of dynamics-dependent allosteric effects, and due to the structural conservation of the STAT family of proteins, the inter-domain allosteric communication observed in STAT3 likely occurs in other STATs.


STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/metabolism , Allosteric Regulation , Binding Sites , Humans , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , STAT3 Transcription Factor/genetics , src Homology Domains
5.
Cancer Res ; 76(3): 652-63, 2016 Feb 01.
Article En | MEDLINE | ID: mdl-26088127

STAT3 offers an attractive target for cancer therapy, but small-molecule inhibitors with appealing pharmacologic properties have been elusive. Here, we report hydroxamic acid-based and benzoic acid-based inhibitors (SH5-07 and SH4-54, respectively) with robust bioactivity. Both inhibitors blocked STAT3 DNA-binding activity in vitro and in human glioma, breast, and prostate cancer cells and in v-Src-transformed murine fibroblasts. STAT3-dependent gene transcription was blocked along with Bcl-2, Bcl-xL, Mcl-1, cyclin D1, c-Myc, and survivin expression. Nuclear magnetic resonance analysis of STAT3-inhibitor complexes defined interactions with the SH2 and DNA-binding domains of STAT3. Ectopic expression of the SH2 domain in cells was sufficient to counter the STAT3-inhibitory effects of SH4-54. Neither compound appreciably affected STAT1 or STAT5 DNA-binding activities, STAT3-independent gene transcription, or activation of a panel of oncogenic kinases in malignant cells. Each compound decreased the proliferation and viability of glioma, breast, and prostate cancer cells and v-Src-transformed murine fibroblasts harboring constitutively active STAT3. Further, in mouse xenograft models of glioma and breast cancer, administration of SH5-07 or SH4-54 effectively inhibited tumor growth. Our results offer preclinical proof of concept for SH5-07 and SH4-54 as candidates for further development as cancer therapeutics.


Benzoates/pharmacology , Brain Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Glioma/drug therapy , Hydroxamic Acids/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Glioma/metabolism , Glioma/pathology , Humans , MCF-7 Cells , Male , Mice , Mice, Nude , NIH 3T3 Cells , STAT3 Transcription Factor/metabolism , Signal Transduction , Transfection
6.
Nat Commun ; 5: 4968, 2014 Sep 29.
Article En | MEDLINE | ID: mdl-25263960

Ubiquitin-like (Ubl) modifications regulate nearly all cellular functions in eukaryotes with the largest superfamily of Ubl-specific proteases being Cys proteases. SENP1 is a model for this protease family and responsible for processing SUMO. Here using nuclear magnetic resonance relaxation measurements, chemical shift perturbation and enzyme kinetic analysis, we provide structural insights into the mechanism of substrate recognition coupled enzymatic activation within SENP1. We find that residues in the catalytic channel of SENP1, including the 'lid' residue Trp465, exhibit dynamics over a range of timescales, both in the presence and absence of bound substrates. The ß-grasp domain of SUMO1 alone induces structural changes at ~20 Å away in the active site of SENP1, revealing the importance of this domain in activating the enzyme. These findings likely represent general properties of the mechanism of substrate recognition and processing by SENPs and other Ubl-specific proteases, and illuminate how adaptive substrate binding can allosterically enhance enzyme activity.


Endopeptidases/chemistry , Allosteric Site , Catalytic Domain , Cysteine Endopeptidases , Escherichia coli/metabolism , Humans , Kinetics , Magnetic Resonance Spectroscopy , Neoplasms/metabolism , Peptides/chemistry , Permeability , Protein Binding , Protein Interaction Mapping , Protein Structure, Secondary , Substrate Specificity , Ubiquitin/chemistry
7.
ACS Chem Biol ; 8(7): 1435-41, 2013 Jul 19.
Article En | MEDLINE | ID: mdl-23614497

Enzymes called SENPs catalyze both the maturation of small ubiquitin-like modifier (SUMO) precursors and removal of SUMO modifications, which regulate essential cellular functions such as cell cycle progression, DNA damage response, and intracellular trafficking. Some members, such as SENP1, are potential targets for developing cancer therapeutics. We searched for small molecule inhibitors of SENPs using in silico screening in conjunction with biochemical assays and identified a new chemotype of small molecule inhibitors that noncovalently inhibit SENPs. The inhibitors confer the noncompetitive inhibitory mechanism, as shown by nuclear magnetic resonance (NMR) and quantitative enzyme kinetic analysis. The NMR data also provided evidence for substrate-assisted inhibitor binding, which indicates the need for caution in using artificial substrates for compound screening, as the inhibitory effects could be significantly different from using the physiological substrates. This finding also suggests the possibility of designing inhibitors for this class of enzymes that are tuned for substrate-specificity.


Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Catalytic Domain , Cysteine Endopeptidases , Electrophoresis, Polyacrylamide Gel , Endopeptidases/chemistry , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , HeLa Cells , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Models, Chemical , Small Molecule Libraries/chemistry
8.
Proc Natl Acad Sci U S A ; 109(24): 9623-8, 2012 Jun 12.
Article En | MEDLINE | ID: mdl-22623533

Computer-aided lead optimization derives a unique, orally bioavailable inhibitor of the signal transducer and activator of transcription (Stat)3 Src homology 2 domain. BP-1-102 binds Stat3 with an affinity (K(D)) of 504 nM, blocks Stat3-phospho-tyrosine (pTyr) peptide interactions and Stat3 activation at 4-6.8 µM, and selectively inhibits growth, survival, migration, and invasion of Stat3-dependent tumor cells. BP-1-102-mediated inhibition of aberrantly active Stat3 in tumor cells suppresses the expression of c-Myc, Cyclin D1, Bcl-xL, Survivin, VEGF, and Krüppel-like factor 8, which is identified as a Stat3 target gene that promotes Stat3-mediated breast tumor cell migration and invasion. Treatment of breast cancer cells with BP-1-102 further blocks Stat3-NF-κB cross-talk, the release of granulocyte colony-stimulating factor, soluble intercellular adhesion molecule 1, macrophage migration-inhibitory factor/glycosylation-inhibiting factor, interleukin 1 receptor antagonist, and serine protease inhibitor protein 1, and the phosphorylation of focal adhesion kinase and paxillin, while enhancing E-cadherin expression. Intravenous or oral gavage delivery of BP-1-102 furnishes micromolar or microgram levels in tumor tissues and inhibits growth of human breast and lung tumor xenografts.


Breast Neoplasms/therapy , Lung Neoplasms/therapy , STAT3 Transcription Factor/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Cell Line , Cell Line, Tumor , Female , Humans , Mice , STAT3 Transcription Factor/administration & dosage , Xenograft Model Antitumor Assays
9.
J Biomol NMR ; 53(2): 113-24, 2012 Jun.
Article En | MEDLINE | ID: mdl-22532128

The development of methyl-TROSY approaches and specific (13)C-(1)H labeling of Ile, Leu and Val methyl groups in highly deuterated proteins has made it possible to study high molecular weight proteins, either alone or in complexes, using solution nuclear magnetic resonance (NMR) spectroscopy. Here we present 2-dimensional (2D) and 3-dimensional (3D) NMR experiments designed to achieve complete separation of the methyl resonances of Val and Leu, labeled using the same precursor, α-ketoisovalerate or acetolactate. The 2D experiment can further select the methyl resonances of Val or Leu based on the C(α) or C(ß) chemical shift values of Val or Leu, respectively. In the 3D spectrum, the methyl cross peaks of Val and Leu residues have opposite signs; thus, not only can the residue types be easily distinguished, but the methyl pairs from the same residue can also be identified. The feasibility of this approach, implemented in both 2D and 3D experiments, has been demonstrated on an 82 kDa protein, malate synthase G. The methods developed in this study will reduce resonance overlaps and also facilitate structure-guided resonance assignments.


Leucine/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Proteins/chemistry , Valine/chemistry , Carbon Isotopes , Escherichia coli Proteins/chemistry , Malate Synthase/chemistry , Models, Molecular , Molecular Weight
10.
J Biol Chem ; 287(5): 3231-40, 2012 Jan 27.
Article En | MEDLINE | ID: mdl-22147707

The small ubiquitin-like modifiers (SUMOs) regulate many essential cellular functions. Only one type of SUMO-interacting motif (SIM) has been identified that can extend the ß-sheet of SUMO as either a parallel or an antiparallel strand. The molecular determinants of the bound orientation and paralogue specificity of a SIM are unclear. To address this question, we have conducted structural studies of SUMO1 in complex with a SUMO1-specific SIM that binds to SUMO1 with high affinity without post-translational modifications using nuclear magnetic resonance methods. In addition, the SIM sequence requirements have been investigated by peptide arrays in comparison with another high affinity SIM that binds in the opposing orientation. We found that antiparallel binding SIMs tolerate more diverse sequences, whereas the parallel binding SIMs prefer the more strict sequences consisting of (I/V)DLT that have a preference in high affinity SUMO2 and -3 binding. Comparison of two high affinity SUMO1-binding SIMs that bind in opposing orientations has revealed common SUMO1-specific interactions needed for high affinity binding. This study has significantly advanced our understanding of the molecular determinants underlining SUMO-SIM recognition.


SUMO-1 Protein/chemistry , Small Ubiquitin-Related Modifier Proteins/chemistry , Ubiquitins/chemistry , Amino Acid Motifs , Cell Line, Tumor , Humans , Nuclear Magnetic Resonance, Biomolecular , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism
11.
Proc Natl Acad Sci U S A ; 108(30): 12289-94, 2011 Jul 26.
Article En | MEDLINE | ID: mdl-21746900

Pin1 is a modular enzyme that accelerates the cis-trans isomerization of phosphorylated-Ser/Thr-Pro (pS/T-P) motifs found in numerous signaling proteins regulating cell growth and neuronal survival. We have used NMR to investigate the interaction of Pin1 with three related ligands that include a pS-P substrate peptide, and two pS-P substrate analogue inhibitors locked in the cis and trans conformations. Specifically, we compared the ligand binding modes and binding-induced changes in Pin1 side-chain flexibility. The cis and trans binding modes differ, and produce different mobility in Pin1. The cis-locked inhibitor and substrate produced a loss of side-chain flexibility along an internal conduit of conserved hydrophobic residues, connecting the domain interface with the isomerase active site. The trans-locked inhibitor produces a weaker conduit response. Thus, the conduit response is stereoselective. We further show interactions between the peptidyl-prolyl isomerase and Trp-Trp (WW) domains amplify the conduit response, and alter binding properties at the remote peptidyl-prolyl isomerase active site. These results suggest that specific input conformations can gate dynamic changes that support intraprotein communication. Such gating may help control the propagation of chemical signals by Pin1, and other modular signaling proteins.


Peptidylprolyl Isomerase/chemistry , Peptidylprolyl Isomerase/metabolism , Amino Acid Motifs , Binding, Competitive , Biophysical Phenomena , Catalytic Domain , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Hydrophobic and Hydrophilic Interactions , In Vitro Techniques , Models, Molecular , NIMA-Interacting Peptidylprolyl Isomerase , Nuclear Magnetic Resonance, Biomolecular , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/genetics , Phosphorylation , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Static Electricity , Stereoisomerism , Substrate Specificity
12.
J Am Chem Soc ; 132(16): 5607-9, 2010 Apr 28.
Article En | MEDLINE | ID: mdl-20356313

Drug design involves iterative ligand modifications. For flexible ligands, these modifications often entail restricting conformational flexibility. However, defining optimal restriction strategies can be challenging if the relationship between ligand flexibility and biological activity is unclear. Here, we describe an approach for ligand flexibility-activity studies using Nuclear Magnetic Resonance (NMR) spin relaxation. Specifically, we use (13)C relaxation dispersion measurements to compare site-specific changes in ligand flexibility for a series of related ligands that bind a common macromolecular receptor. The flexibility changes reflect conformational reorganization resulting from formation of the receptor-ligand complex. We demonstrate this approach on three structurally similar but flexibly differentiated ligands of human Pin1, a peptidyl-prolyl isomerase. The approach is able to map the ligand dynamics relevant for activity and expose changes in those dynamics caused by conformational locking. Thus, NMR flexibility-activity studies can provide information to guide strategic ligand rigidification. As such, they help establish an experimental basis for developing flexibility-activity relationships (FAR) to complement traditional structure-activity relationships (SAR) in molecular design.


Oligopeptides/chemistry , Oligopeptides/metabolism , Peptidylprolyl Isomerase/metabolism , Amino Acid Sequence , Humans , Ligands , Magnetic Resonance Spectroscopy , NIMA-Interacting Peptidylprolyl Isomerase , Structure-Activity Relationship , Temperature
13.
J Biomol NMR ; 45(1-2): 171-83, 2009 Sep.
Article En | MEDLINE | ID: mdl-19639385

Flexible ligands pose challenges to standard structure-activity studies since they frequently reorganize their conformations upon protein binding and catalysis. Here, we demonstrate the utility of side chain (13)C relaxation dispersion measurements to identify and quantify the conformational dynamics that drive this reorganization. The dispersion measurements probe methylene (13)CH(2) and methyl (13)CH(3) groups; the latter are highly prevalent side chain moieties in known drugs. Combining these side chain studies with existing backbone dispersion studies enables a comprehensive investigation of mus-ms conformational dynamics related to binding and catalysis. We perform these measurements at natural (13)C abundance, in congruence with common pharmaceutical research settings. We illustrate these methods through a study of the interaction of a phosphopeptide ligand with the peptidyl-prolyl isomerase, Pin1. The results illuminate the side-chain moieties that undergo conformational readjustments upon complex formation. In particular, we find evidence that multiple exchange processes influence the side chain dispersion profiles. Collectively, our studies illustrate how side-chain relaxation dispersion can shed light on ligand conformational transitions required for activity, and thereby suggest strategies for its optimization.


Carbon Isotopes/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Proteins/chemistry , Amino Acid Motifs , Binding Sites , Humans , Ligands , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/chemistry , Phosphopeptides/chemistry , Protein Binding , Protein Conformation
14.
Nat Struct Mol Biol ; 14(4): 325-31, 2007 Apr.
Article En | MEDLINE | ID: mdl-17334375

The current canon attributes the binding specificity of protein-recognition motifs to distinctive chemical moieties in their constituent amino acid sequences. However, we show for a WW domain that the sequence crucial for specificity is an intrinsically flexible loop that partially rigidifies upon ligand docking. A single-residue deletion in this loop simultaneously reduces loop flexibility and ligand binding affinity. These results suggest that sequences of recognition motifs may reflect natural selection of not only chemical properties but also dynamic modes that augment specificity.


Peptidylprolyl Isomerase/chemistry , Peptidylprolyl Isomerase/metabolism , Amino Acid Sequence , Apoproteins/chemistry , Humans , Magnetic Resonance Spectroscopy , Mutant Proteins/metabolism , NIMA-Interacting Peptidylprolyl Isomerase , Nitrogen Isotopes , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship , Substrate Specificity , Temperature , Thermodynamics , Titrimetry , ras-GRF1/metabolism
15.
Structure ; 15(3): 313-27, 2007 Mar.
Article En | MEDLINE | ID: mdl-17355867

Pin1 is a peptidyl-prolyl isomerase consisting of a WW domain and a catalytic isomerase (PPIase) domain connected by a flexible linker. Pin1 recognizes phospho-Ser/Thr-Pro motifs in cell-signaling proteins, and is both a cancer and an Alzheimer's disease target. Here, we provide novel insight into the functional motions underlying Pin1 substrate interaction using nuclear magnetic resonance deuterium ((2)D) and carbon ((13)C) spin relaxation. Specifically, we compare Pin1 side-chain motions in the presence and absence of a known phosphopeptide substrate derived from the mitotic phosphatase Cdc25. Substrate interaction alters Pin1 side-chain motions on both the microsecond-millisecond (mus-ms) and picosecond-nanosecond (ps-ns) timescales. Alterations include loss of ps-ns flexibility along an internal conduit of hydrophobic residues connecting the catalytic site with the interdomain interface. These residues are conserved among Pin1 homologs; hence, their dynamics are likely important for the Pin1 mechanism.


Amino Acids/chemistry , Amino Acids/metabolism , Conserved Sequence , Hydrophobic and Hydrophilic Interactions , Peptidylprolyl Isomerase/metabolism , Amino Acid Sequence , Amino Acids/genetics , Crystallography, X-Ray , Humans , Molecular Sequence Data , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/chemistry , Peptidylprolyl Isomerase/genetics , Substrate Specificity , Thermodynamics
...