Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
J Med Chem ; 66(8): 5859-5872, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37071570

ABSTRACT

The development of low-affinity fragment hits into higher-affinity leads is a major hurdle in fragment-based drug design. Here, we demonstrate the Rapid Elaboration of Fragments into Leads (REFiL) by applying an integrated workflow that provides a systematic approach to generate higher-affinity binders without the need for structural information. The workflow involves the selection of commercial analogues of fragment hits to generate preliminary structure-activity relationships. This is followed by parallel microscale chemistry using chemoinformatically designed reagent libraries to rapidly explore chemical diversity. After a fragment screen against bromodomain-3 extra-terminal (BRD3-ET) domain, we applied the REFiL workflow, which allowed us to develop a series of ligands that bind to BRD3-ET. With REFiL, we were able to rapidly improve binding affinity > 30-fold. REFiL can be applied readily to a broad range of proteins without the need for a structure, allowing the efficient evolution of low-affinity fragments into higher-affinity leads and chemical probes.


Subject(s)
Drug Design , Proteins , Proteins/metabolism , Structure-Activity Relationship , Protein Domains , Ligands
2.
Molecules ; 28(7)2023 Mar 30.
Article in English | MEDLINE | ID: mdl-37049868

ABSTRACT

Human immunodeficiency virus type I (HIV-1) is a retrovirus that infects cells of the host's immune system leading to acquired immunodeficiency syndrome and potentially death. Although treatments are available to prevent its progression, HIV-1 remains a major burden on health resources worldwide. Continued emergence of drug-resistance mutations drives the need for novel drugs that can inhibit HIV-1 replication through new pathways. The viral protein reverse transcriptase (RT) plays a fundamental role in the HIV-1 replication cycle, and multiple approved medications target this enzyme. In this study, fragment-based drug discovery was used to optimize a previously identified hit fragment (compound B-1), which bound RT at a novel site. Three series of compounds were synthesized and evaluated for their HIV-1 RT binding and inhibition. These series were designed to investigate different vectors around the initial hit in an attempt to improve inhibitory activity against RT. Our results show that the 4-position of the core scaffold is important for binding of the fragment to RT, and a lead compound with a cyclopropyl substitution was selected and further investigated. Requirements for binding to the NNRTI-binding pocket (NNIBP) and a novel adjacent site were investigated, with lead compound 27-a minimal but efficient NNRTI-offering a starting site for the development of novel dual NNIBP-Adjacent site inhibitors.


Subject(s)
Acquired Immunodeficiency Syndrome , Anti-HIV Agents , HIV-1 , Humans , Reverse Transcriptase Inhibitors/chemistry , HIV Reverse Transcriptase , Acquired Immunodeficiency Syndrome/drug therapy , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use
3.
Cell Rep ; 28(8): 2023-2036.e4, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31433980

ABSTRACT

Lymphatic vascular development establishes embryonic and adult tissue fluid balance and is integral in disease. In diverse vertebrate organs, lymphatic vessels display organotypic function and develop in an organ-specific manner. In all settings, developmental lymphangiogenesis is considered driven by vascular endothelial growth factor (VEGF) receptor-3 (VEGFR3), whereas a role for VEGFR2 remains to be fully explored. Here, we define the zebrafish Vegf/Vegfr code in receptor binding studies. We find that while Vegfd directs craniofacial lymphangiogenesis, it binds Kdr (a VEGFR2 homolog) but surprisingly, unlike in mammals, does not bind Flt4 (VEGFR3). Epistatic analyses and characterization of a kdr mutant confirm receptor-binding analyses, demonstrating that Kdr is indispensible for rostral craniofacial lymphangiogenesis, but not caudal trunk lymphangiogenesis, in which Flt4 is central. We further demonstrate an unexpected yet essential role for Kdr in inducing lymphatic endothelial cell fate. This work reveals evolutionary divergence in the Vegf/Vegfr code that uncovers spatially restricted mechanisms of developmental lymphangiogenesis.


Subject(s)
Endothelial Cells/metabolism , Evolution, Molecular , Lymphangiogenesis , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/metabolism , Amino Acid Sequence , Animals , HEK293 Cells , Humans , Ligands , Mice , Protein Binding , Proteolysis , Reproducibility of Results , Vascular Endothelial Growth Factor C/chemistry , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism
4.
J Biol Chem ; 294(10): 3720-3734, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30598509

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) is a transcriptional regulator of lipid metabolism. GW7647 is a potent PPARα agonist that must reach the nucleus to activate this receptor. In cells expressing human fatty acid-binding protein 1 (FABP1), GW7647 treatment increases FABP1's nuclear localization and potentiates GW7647-mediated PPARα activation; GW7647 is less effective in cells that do not express FABP1. To elucidate the underlying mechanism, here we substituted residues in FABP1 known to dictate lipid signaling by other intracellular lipid-binding proteins. Substitutions of Lys-20 and Lys-31 to Ala in the FABP1 helical cap affected neither its nuclear localization nor PPARα activation. In contrast, Ala substitution of Lys-57, Glu-77, and Lys-96, located in the loops adjacent to the ligand-binding portal region, abolished both FABP1 nuclear localization and GW7647-induced PPARα activation but had little effect on GW7647-FABP1 binding affinity. Using solution NMR spectroscopy, we determined the WT FABP1 structure and analyzed the dynamics in the apo and GW7647-bound structures of both the WT and the K57A/E77A/K96A triple mutant. We found that GW7647 binding causes little change in the FABP1 backbone, but solvent exposes several residues in the loops around the portal region, including Lys-57, Glu-77, and Lys-96. These residues also become more solvent-exposed upon binding of FABP1 with the endogenous PPARα agonist oleic acid. Together with previous observations, our findings suggest that GW7647 binding stabilizes a FABP1 conformation that promotes its interaction with PPARα. We conclude that full PPARα agonist activity of GW7647 requires FABP1-dependent transport and nuclear localization processes.


Subject(s)
Butyrates/pharmacology , Fatty Acid-Binding Proteins/chemistry , Fatty Acid-Binding Proteins/metabolism , PPAR alpha/agonists , Phenylurea Compounds/pharmacology , Butyrates/metabolism , Fatty Acid-Binding Proteins/genetics , Humans , Ligands , Models, Molecular , Mutation , Phenylurea Compounds/metabolism , Protein Conformation/drug effects
5.
J Biol Chem ; 293(43): 16559-16571, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30181210

ABSTRACT

The worldwide incidence of neisserial infections, particularly gonococcal infections, is increasingly associated with antibiotic-resistant strains. In particular, extensively drug-resistant Neisseria gonorrhoeae strains that are resistant to third-generation cephalosporins are a major public health concern. There is a pressing clinical need to identify new targets for the development of antibiotics effective against Neisseria-specific processes. In this study, we report that the bacterial disulfide reductase DsbD is highly prevalent and conserved among Neisseria spp. and that this enzyme is essential for survival of N. gonorrhoeae DsbD is a membrane-bound protein that consists of two periplasmic domains, n-DsbD and c-DsbD, which flank the transmembrane domain t-DsbD. In this work, we show that the two functionally essential periplasmic domains of Neisseria DsbD catalyze electron transfer reactions through unidirectional interdomain interactions, from reduced c-DsbD to oxidized n-DsbD, and that this process is not dictated by their redox potentials. Structural characterization of the Neisseria n- and c-DsbD domains in both redox states provides evidence that steric hindrance reduces interactions between the two periplasmic domains when n-DsbD is reduced, thereby preventing a futile redox cycle. Finally, we propose a conserved mechanism of electron transfer for DsbD and define the residues involved in domain-domain recognition. Inhibitors of the interaction of the two DsbD domains have the potential to be developed as anti-neisserial agents.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Disulfides/metabolism , Neisseria gonorrhoeae/enzymology , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Protein Conformation , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Disulfides/chemistry , Models, Molecular , Oxidation-Reduction , Protein Domains
6.
ChemMedChem ; 13(15): 1555-1565, 2018 08 10.
Article in English | MEDLINE | ID: mdl-29862651

ABSTRACT

The p75 splice variant of lens epithelium-derived growth factor (LEDGF) is a 75 kDa protein, which is recruited by the human immunodeficiency virus (HIV) to tether the pre-integration complex to the host chromatin and promote integration of proviral DNA into the host genome. We designed a series of small cyclic peptides that are structural mimics of the LEDGF binding domain, which interact with integrase as potential binding inhibitors. Herein we present the X-ray crystal structures, NMR studies, SPR analysis, and conformational studies of four cyclic peptides bound to the HIV-1 integrase core domain. Although the X-ray studies show that the peptides closely mimic the LEDGF binding loop, the measured affinities of the peptides are in the low millimolar range. Computational analysis using conformational searching and free energy calculations suggest that the low affinity of the peptides is due to mismatch between the low-energy solution and bound conformations.


Subject(s)
HIV Integrase/chemistry , Intercellular Signaling Peptides and Proteins/chemistry , Molecular Mimicry , Peptides, Cyclic/chemistry , Crystallography, X-Ray , HIV-1/enzymology , Protein Conformation , Spectrum Analysis/methods
7.
Antibiotics (Basel) ; 7(1)2018 Feb 22.
Article in English | MEDLINE | ID: mdl-29470422

ABSTRACT

In bacteria, the DnaG primase is responsible for synthesis of short RNA primers used to initiate chain extension by replicative DNA polymerase(s) during chromosomal replication. Among the proteins with which Escherichia coli DnaG interacts is the single-stranded DNA-binding protein, SSB. The C-terminal hexapeptide motif of SSB (DDDIPF; SSB-Ct) is highly conserved and is known to engage in essential interactions with many proteins in nucleic acid metabolism, including primase. Here, fragment-based screening by saturation-transfer difference nuclear magnetic resonance (STD-NMR) and surface plasmon resonance assays identified inhibitors of the primase/SSB-Ct interaction. Hits were shown to bind to the SSB-Ct-binding site using 15N-¹H HSQC spectra. STD-NMR was used to demonstrate binding of one hit to other SSB-Ct binding partners, confirming the possibility of simultaneous inhibition of multiple protein/SSB interactions. The fragment molecules represent promising scaffolds on which to build to discover new antibacterial compounds.

8.
Angew Chem Int Ed Engl ; 56(29): 8495-8499, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28513074

ABSTRACT

Δ-Myrtoxin-Mp1a (Mp1a), a 49-residue heterodimeric peptide from the venom of Myrmecia pilosula, comprises a 26-mer A chain and a 23-mer B chain connected by two disulfide bonds in an antiparallel arrangement. Combination of the individual synthetic chains through aerial oxidation remarkably resulted in the self-assembly of Mp1a as a homogenous product without the need for directed disulfide-bond formation. NMR analysis revealed a well-defined, unique structure containing an antiparallel α-helix pair. Dual polarization interferometry (DPI) analysis showed strong interaction with supported lipid bilayers and insertion within the bilayers. Mp1a caused non-specific Ca2+ influx in SH-SY5Y cells with a half maximal effective concentration (EC50 ) of 4.3 µm. Mp1a also displayed broad-spectrum antimicrobial activity, with the highest potency against Gram-negative Acinetobacter baumannii (MIC 25 nm). Intraplantar injection (10 µm) in mice elicited spontaneous pain and mechanical allodynia. Single- and two-chain mimetics of Mp1a revealed functional selectivity.


Subject(s)
Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , Hyperalgesia/drug therapy , Pain/drug therapy , Peptides/pharmacology , Venoms/chemistry , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemistry , Ants , Calcium/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Mice , Microbial Sensitivity Tests , Models, Molecular , Peptides/administration & dosage , Peptides/chemistry
9.
PLoS One ; 12(3): e0173436, 2017.
Article in English | MEDLINE | ID: mdl-28346540

ABSTRACT

At a time when the antibiotic drug discovery pipeline has stalled, antibiotic resistance is accelerating with catastrophic implications for our ability to treat bacterial infections. Globally we face the prospect of a future when common infections can once again kill. Anti-virulence approaches that target the capacity of the bacterium to cause disease rather than the growth or survival of the bacterium itself offer a tantalizing prospect of novel antimicrobials. They may also reduce the propensity to induce resistance by removing the strong selection pressure imparted by bactericidal or bacteriostatic agents. In the human pathogen Pseudomonas aeruginosa, disulfide bond protein A (PaDsbA1) plays a central role in the oxidative folding of virulence factors and is therefore an attractive target for the development of new anti-virulence antimicrobials. Using a fragment-based approach we have identified small molecules that bind to PaDsbA1. The fragment hits show selective binding to PaDsbA1 over the DsbA protein from Escherichia coli, suggesting that developing species-specific narrow-spectrum inhibitors of DsbA enzymes may be feasible. Structures of a co-complex of PaDsbA1 with the highest affinity fragment identified in the screen reveal that the fragment binds on the non-catalytic surface of the protein at a domain interface. This biophysical and structural data represent a starting point in the development of higher affinity compounds, which will be assessed for their potential as selective PaDsbA1 inhibitors.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Protein Disulfide-Isomerases/antagonists & inhibitors , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Crystallography, X-Ray , Drug Discovery , Humans , Molecular Docking Simulation , Protein Binding , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/metabolism , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Virulence Factors/metabolism
10.
Mol Pharm ; 14(3): 566-579, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28099023

ABSTRACT

Improved models of the gastrointestinal environment have great potential to assist the complex process of drug formulation. Molecular dynamics (MD) is a powerful method for investigating phase behavior at a molecular level. In this study we use multiple MD simulations to calculate phase diagrams for bile before and after digestion. In these computational models, undigested bile is represented by mixtures of palmitoyl-oleoylphosphatidylcholine (POPC), sodium glycodeoxycholate (GDX), and water. Digested bile is modeled using a 1:1 mixture of oleic acid and palmitoylphosphatidylcholine (lysophosphatidylcholine, LPC), GDX, and water. The computational phase diagrams of undigested and digested bile are compared, and we describe the typical intermolecular interactions that occur between phospholipids and bile salts. The diffusion coefficients measured from MD simulation are compared to experimental diffusion data measured by DOSY-NMR, where we observe good qualitative agreement. In an additional set of simulations, the effect of different ionization states of oleic acid on micelle formation is investigated.


Subject(s)
Body Fluids/chemistry , Digestion/physiology , Gastrointestinal Tract/chemistry , Bile/chemistry , Bile Acids and Salts/chemistry , Chemistry, Pharmaceutical/methods , Computer Simulation , Glycodeoxycholic Acid/chemistry , Magnetic Resonance Spectroscopy/methods , Micelles , Molecular Dynamics Simulation , Oleic Acid/chemistry , Phosphatidylcholines/chemistry , Phospholipids/chemistry , Water/chemistry
11.
Biochimie ; 122: 227-34, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26403495

ABSTRACT

Proteolysis has a critical role in transmitting information within a biological system and therefore an important element of biology is to determine the subset of proteins amenable to proteolysis. Until recently, it has been thought that proteases cleave native protein substrates only within solvent exposed loops, but recent evidence indicates that cleavage sites located within α-helices can also be cleaved by proteases, despite the conformation of this secondary structure being generally incompatible with binding into an active site of a protease. In this study, we address the mechanism by which a serine endopeptidase, thrombin, recognizes and cleaves a target sequence located within an α-helix. Thrombin was able to cleave a model substrate, protein G, within its α-helix when a suitable cleavage sequence for the enzyme was introduced into this region. However, structural data for the complex revealed that thrombin was not perturbing the structure of the α-helix, thus it was not destabilizing the helix in order to allow it to fit within its active site. This indicated that thrombin was only cleaving within the α-helix when it was in an unfolded state. In support of this, the introduction of destabilizing mutations within the protein increased the efficiency of cleavage by the enzyme. Our data suggest that a folded α-helix cannot be proteolytically cleaved by thrombin, but the species targeted are the unfolded conformations of the native state ensemble.


Subject(s)
Bacterial Proteins/metabolism , Protein Structure, Secondary , Protein Unfolding , Serine Proteases/metabolism , Thrombin/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Electrophoresis, Polyacrylamide Gel , Humans , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Mutation , Proteolysis , Sequence Homology, Amino Acid , Substrate Specificity
12.
Sci Rep ; 5: 12905, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26243377

ABSTRACT

The biogenesis of membranes from constituent proteins and lipids is a fundamental aspect of cell biology. In the case of proteins assembled into bacterial outer membranes, an overarching question concerns how the energy required for protein insertion and folding is accessed at this remote location of the cell. The translocation and assembly module (TAM) is a nanomachine that functions in outer membrane biogenesis and virulence in diverse bacterial pathogens. Here we demonstrate the interactions through which TamA and TamB subunits dock to bridge the periplasm, and unite the outer membrane aspects to the inner membrane of the bacterial cell. We show that specific functional features in TamA have been conserved through evolution, including residues surrounding the lateral gate and an extensive surface of the POTRA domains. Analysis by nuclear magnetic resonance spectroscopy and small angle X-ray scattering document the characteristic structural features of these POTRA domains and demonstrate rigidity in solution. Quartz crystal microbalance measurements pinpoint which POTRA domain specifically docks the TamB subunit of the nanomachine. We speculate that the POTRA domain of TamA functions as a lever arm in order to drive the activity of the TAM, assembling proteins into bacterial outer membranes.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Amino Acid Sequence , Bacterial Outer Membrane Proteins/physiology , Conserved Sequence , Evolution, Molecular , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Protein Structure, Secondary
13.
Proc Natl Acad Sci U S A ; 112(22): 6979-84, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-26038551

ABSTRACT

Fragment-based screening methods can be used to discover novel active site or allosteric inhibitors for therapeutic intervention. Using saturation transfer difference (STD) NMR and in vitro activity assays, we have identified fragment-sized inhibitors of HIV-1 reverse transcriptase (RT) with distinct chemical scaffolds and mechanisms compared to nonnucleoside RT inhibitors (NNRTIs) and nucleoside/nucleotide RT inhibitors (NRTIs). Three compounds were found to inhibit RNA- and DNA-dependent DNA polymerase activity of HIV-1 RT in the micromolar range while retaining potency against RT variants carrying one of three major NNRTI resistance mutations: K103N, Y181C, or G190A. These compounds also inhibit Moloney murine leukemia virus RT but not the Klenow fragment of Escherichia coli DNA polymerase I. Steady-state kinetic analyses demonstrate that one of these fragments is a competitive inhibitor of HIV-1 RT with respect to deoxyribonucleoside triphosphate (dNTP) substrate, whereas a second compound is a competitive inhibitor of RT polymerase activity with respect to the DNA template/primer (T/P), and consequently also inhibits RNase H activity. The dNTP competing RT inhibitor retains activity against the NRTI-resistant mutants K65R and M184V, demonstrating a drug resistance profile distinct from the nucleotide competing RT inhibitors indolopyridone-1 (INDOPY-1) and 4-dimethylamino-6-vinylpyrimidine-1 (DAVP-1). In antiviral assays, the T/P competing compound inhibits HIV-1 replication at a step consistent with an RT inhibitor. Screening of additional structurally related compounds to the three fragments led to the discovery of molecules with improved potency against HIV-1 RT. These fragment inhibitors represent previously unidentified scaffolds for development of novel drugs for HIV-1 prevention or treatment.


Subject(s)
Drug Discovery/methods , HIV-1/enzymology , Prodrugs/isolation & purification , Reverse Transcriptase Inhibitors/isolation & purification , Reverse Transcriptase Inhibitors/pharmacology , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , Magnetic Resonance Spectroscopy , Prodrugs/analysis , Reverse Transcriptase Inhibitors/analysis , Ribonuclease H/antagonists & inhibitors , Small Molecule Libraries , Virus Replication/drug effects
14.
J Mol Recognit ; 28(5): 316-24, 2015 May.
Article in English | MEDLINE | ID: mdl-25720550

ABSTRACT

The transfer of antibiotic resistance between bacteria is mediated by mobile genetic elements such as plasmids and transposons. TnpX is a member of the large serine recombinase subgroup of site-specific recombinases and is responsible for the excision and insertion of mobile genetic elements that encode chloramphenicol resistance in the pathogens Clostridium perfringens and Clostridium difficile. TnpX consists of three structural domains: domain I contains the catalytic site, whereas domains II and III contain DNA-binding motifs. We have solved the solution structure of residues 1-120 of the catalytic domain I of TnpX. The TnpX catalytic domain shares the same overall fold as other serine recombinases; however, differences are evident in the identity of the proposed hydrogen donor and in the size, amino acid composition, conformation, and dynamics of the TnpX active site loops. To obtain the interaction surface of TnpX1-120 , we titrated a DNA oligonucleotide containing the circular intermediate joint attCI recombination site into (15) N-labeled TnpX1-120 and observed progressive nuclear magnetic resonance chemical shift perturbations using (15) N HSQC spectra. Perturbations were largely confined to a region surrounding the catalytic serine and encompassed residues of the active site loops. Utilizing the perturbation map and the data-driven docking program, HADDOCK, we have generated a model of the DNA interaction complex for the TnpX catalytic domain.


Subject(s)
Bacterial Proteins/chemistry , DNA, Bacterial/chemistry , Recombinases/chemistry , Amino Acid Sequence , Catalytic Domain , Clostridium perfringens/enzymology , Molecular Docking Simulation , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Conformation , Protein Binding
15.
Angew Chem Int Ed Engl ; 54(7): 2179-84, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25556635

ABSTRACT

The thiol-disulfide oxidoreductase enzyme DsbA catalyzes the formation of disulfide bonds in the periplasm of Gram-negative bacteria. DsbA substrates include proteins involved in bacterial virulence. In the absence of DsbA, many of these proteins do not fold correctly, which renders the bacteria avirulent. Thus DsbA is a critical mediator of virulence and inhibitors may act as antivirulence agents. Biophysical screening has been employed to identify fragments that bind to DsbA from Escherichia coli. Elaboration of one of these fragments produced compounds that inhibit DsbA activity in vitro. In cell-based assays, the compounds inhibit bacterial motility, but have no effect on growth in liquid culture, which is consistent with selective inhibition of DsbA. Crystal structures of inhibitors bound to DsbA indicate that they bind adjacent to the active site. Together, the data suggest that DsbA may be amenable to the development of novel antibacterial compounds that act by inhibiting bacterial virulence.


Subject(s)
Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Protein Disulfide-Isomerases/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Humans , Molecular Docking Simulation , Protein Disulfide-Isomerases/metabolism
16.
ACS Chem Biol ; 9(11): 2526-34, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25144524

ABSTRACT

The aqueous cytoplasm of cells poses a potentially significant barrier for many lipophilic drugs to reach their sites of action. Fatty acid binding proteins (FABPs) bind to poorly water-soluble fatty acids (FAs) and lipophilic compounds and facilitate their intracellular transport. Several structures of FA in complex with FABPs have been described, but data describing the binding sites of other lipophilic ligands including drugs are limited. Here the environmentally sensitive fluorophores, 1-anilinonapthalene 8-sulfonic acid (ANS), and 11-dansylamino undecanoic acid (DAUDA) were used to investigate drug binding to human intestinal FABP (hIFABP). Most drugs that bound hIFABP were able to displace both ANS and DAUDA. A notable exception was ketorolac, a non-steroidal anti-inflammatory drug that bound to hIFABP and displaced DAUDA but failed to displace ANS. Isothermal titration calorimetry revealed that for the majority of ligands including FA, ANS, and DAUDA, binding to hIFABP was exothermic. In contrast, ketorolac binding to hIFABP was endothermic and entropy-driven. The X-ray crystal structure of DAUDA-hIFABP revealed a FA-like binding mode where the carboxylate of DAUDA formed a network of hydrogen bonds with residues at the bottom of the binding cavity and the dansyl group interacted with residues in the portal region. In contrast, NMR chemical shift perturbation (CSP) data suggested that ANS bound only toward the bottom of the hIFABP cavity, whereas ketorolac occupied only the portal region. The CSP data further suggested that ANS and ketorolac were able to bind simultaneously to hIFABP, consistent with the lack of displacement of ANS observed by fluorescence and supported by a model of the ternary complex. The NMR solution structure of the ketorolac-hIFABP complex therefore describes a newly characterized, hydrophobic ligand binding site in the portal region of hIFABP.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Fluorescent Dyes , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Spectrometry, Fluorescence
17.
J Biol Chem ; 289(12): 8240-51, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24488493

ABSTRACT

In mammals, the αßT cell receptor (TCR) signaling complex is composed of a TCRαß heterodimer that is noncovalently coupled to three dimeric signaling molecules, CD3εδ, CD3εγ, and CD3ζζ. The nature of the TCR signaling complex and subunit arrangement in different species remains unclear however. Here we present a structural and biochemical analysis of the more primitive ancestral form of the TCR signaling complex found in chickens. In contrast to mammals, chickens do not express separate CD3δ and CD3γ chains but instead encode a single hybrid chain, termed CD3δ/γ, that is capable of pairing with CD3ε. The NMR structure of the chicken CD3εδ/γ heterodimer revealed a unique dimer interface that results in a heterodimer with considerable deviation from the distinct side-by-side architecture found in human and murine CD3εδ and CD3εγ. The chicken CD3εδ/γ heterodimer also contains a unique molecular surface, with the vast majority of surface-exposed, nonconserved residues being clustered to a single face of the heterodimer. Using an in vitro biochemical assay, we demonstrate that CD3εδ/γ can assemble with both chicken TCRα and TCRß via conserved polar transmembrane sites. Moreover, analogous to the human TCR signaling complex, the presence of two copies of CD3εδ/γ is required for ζζ assembly. These data provide insight into the evolution of this critical receptor signaling apparatus.


Subject(s)
Avian Proteins/chemistry , CD3 Complex/chemistry , Chickens/metabolism , Receptor-CD3 Complex, Antigen, T-Cell/chemistry , Amino Acid Sequence , Animals , CD3 Complex/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Multimerization , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Sequence Alignment
18.
J Biol Chem ; 288(52): 36796-809, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24187131

ABSTRACT

We have determined the structure of the human integrin α1I domain bound to a triple-helical collagen peptide. The structure of the α1I-peptide complex was investigated using data from NMR, small angle x-ray scattering, and size exclusion chromatography that were used to generate and validate a model of the complex using the data-driven docking program, HADDOCK (High Ambiguity Driven Biomolecular Docking). The structure revealed that the α1I domain undergoes a major conformational change upon binding of the collagen peptide. This involves a large movement in the C-terminal helix of the αI domain that has been suggested to be the mechanism by which signals are propagated in the intact integrin receptor. The structure suggests a basis for the different binding selectivity observed for the α1I and α2I domains. Mutational data identify residues that contribute to the conformational change observed. Furthermore, small angle x-ray scattering data suggest that at low collagen peptide concentrations the complex exists in equilibrium between a 1:1 and 2:1 α1I-peptide complex.


Subject(s)
Collagen/chemistry , Integrin alpha1/chemistry , Peptides/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Collagen/genetics , Collagen/metabolism , Humans , Integrin alpha1/metabolism , Molecular Docking Simulation , Peptides/genetics , Peptides/metabolism , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Scattering, Small Angle , X-Ray Diffraction
19.
RNA Biol ; 10(4): 579-89, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23603827

ABSTRACT

The RNA-binding protein TIAR is an mRNA-binding protein that acts as a translational repressor, particularly important under conditions of cellular stress. It binds to target mRNA and DNA via its RNA recognition motif (RRM) domains and is involved in both splicing regulation and translational repression via the formation of "stress granules." TIAR has also been shown to bind ssDNA and play a role in the regulation of transcription. Here we show, using surface plasmon resonance and nuclear magnetic resonance spectroscopy, specific roles of individual TIAR domains for high-affinity binding to RNA and DNA targets. We confirm that RRM2 of TIAR is the major RNA- and DNA-binding domain. However, the strong nanomolar affinity binding to U-rich RNA and T-rich DNA depends on the presence of the six amino acid residues found in the linker region C-terminal to RRM2. On its own, RRM1 shows preferred binding to DNA over RNA. We further characterize the interaction between RRM2 with the C-terminal extension and an AU-rich target RNA sequence using NMR spectroscopy to identify the amino acid residues involved in binding. We demonstrate that TIAR RRM2, together with its C-terminal extension, is the major contributor for the high-affinity (nM) interactions of TIAR with target RNA sequences.


Subject(s)
Amino Acid Motifs , DNA-Binding Proteins/metabolism , DNA/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Binding Sites , DNA/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Binding , RNA Splicing , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Sequence Alignment , Surface Plasmon Resonance
20.
J Biomol Screen ; 18(2): 147-59, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23139382

ABSTRACT

Fragment screening is becoming widely accepted as a technique to identify hit compounds for the development of novel lead compounds. In neighboring laboratories, we have recently, and independently, performed a fragment screening campaign on the HIV-1 integrase core domain (IN) using similar commercially purchased fragment libraries. The two campaigns used different screening methods for the preliminary identification of fragment hits; one used saturation transfer difference nuclear magnetic resonance spectroscopy (STD-NMR), and the other used surface plasmon resonance (SPR) spectroscopy. Both initial screens were followed by X-ray crystallography. Using the STD-NMR/X-ray approach, 15 IN/fragment complexes were identified, whereas the SPR/X-ray approach found 6 complexes. In this article, we compare the approaches that were taken by each group and the results obtained, and we look at what factors could potentially influence the final results. We find that despite using different approaches with little overlap of initial hits, both approaches identified binding sites on IN that provided a basis for fragment-based lead discovery and further lead development. Comparison of hits identified in the two studies highlights a key role for both the conditions under which fragment binding is measured and the criteria selected to classify hits.


Subject(s)
HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , Magnetic Resonance Spectroscopy/methods , Small Molecule Libraries , Surface Plasmon Resonance/methods , Crystallography, X-Ray , Drug Evaluation, Preclinical , HIV Integrase/chemistry , HIV Integrase/genetics , HIV Integrase Inhibitors/chemistry , Humans , Protein Binding/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...