Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Protein Sci ; 33(6): e5009, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38747379

ABSTRACT

PHPT1 is a histidine phosphatase that modulates signaling in eukaryotes through its catalytic activity. Here, we present an analysis of the structure and dynamics of PHPT1 through a combination of solution NMR, molecular dynamics, and biochemical experiments. We identify a salt bridge formed between the R78 guanidinium moiety and the C-terminal carboxyl group on Y125 that is critical for ligand binding. Disruption of the salt bridge by appending a glycine residue at the C-terminus (G126) leads to a decrease in catalytic activity and binding affinity for the pseudo substrate, para-nitrophenylphosphate (pNPP), as well as the active site inhibitor, phenylphosphonic acid (PPA). We show through NMR chemical shift, 15N relaxation measurements, and analysis of molecular dynamics trajectories, that removal of this salt bridge results in an active site that is altered both structurally and dynamically thereby significantly impacting enzymatic function and confirming the importance of this electrostatic interaction.


Subject(s)
Catalytic Domain , Molecular Dynamics Simulation , Humans , Nuclear Magnetic Resonance, Biomolecular , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Substrate Specificity
2.
J Am Chem Soc ; 141(32): 12634-12647, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31339043

ABSTRACT

Active-site loops are integral to the function of numerous enzymes. They enable substrate and product binding and release, sequester reaction intermediates, and recruit catalytic groups. Here, we examine the catalytic loop in the enzyme protein tyrosine phosphatase 1B (PTP1B). PTP1B has a mobile so-called WPD loop (named for its three N-terminal residues) that initiates the dephosphorylation of phosphor-tyrosine substrates upon loop closure. We have combined X-ray crystallography, solution NMR, and pre-steady-state kinetics experiments on wild-type and five WPD loop mutants to identify the relationships between the loop structure, dynamics, and function. The motions of the WPD loop are modulated by the formation of weak molecular interactions, where perturbations of these interactions modulate the conformational equilibrium landscape. The point mutants in the WPD loop alter the loop equilibrium position from a predominantly open state (P185A) to 50:50 (F182A), 35:65 (P188A), and predominantly closed states (T177A and P188A). Surprisingly, there is no correlation between the observed catalytic rates in the loop mutants and changes to the WPD loop equilibrium position. Rather, we observe a strong correlation between the rate of dephosphorylation of the phosphocysteine enzyme intermediate and uniform millisecond motions, not only within the loop but also in the adjacent α-helical domain of PTP1B. Thus, the control of loop motion and thereby catalytic activity is dispersed and resides within not only the loop sequence but also the surrounding protein architecture. This has broad implications for the general mechanistic understanding of enzyme reactions and the role that flexible loops play in the catalytic cycle.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Biocatalysis , Catalytic Domain/genetics , Crystallography, X-Ray , Humans , Kinetics , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics
3.
J Mol Biol ; 429(15): 2360-2372, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28625849

ABSTRACT

Drug-like molecules targeting allosteric sites in proteins are of great therapeutic interest; however, identification of potential sites is not trivial. A straightforward approach to identify hidden allosteric sites is demonstrated in protein tyrosine phosphatases (PTP) by creation of single alanine mutations in the catalytic acid loop of PTP1B and VHR. This approach relies on the reciprocal interactions between an allosteric site and its coupled orthosteric site. The resulting NMR chemical shift perturbations (CSPs) of each mutant reveal clusters of distal residues affected by acid loop mutation. In PTP1B and VHR, two new allosteric clusters were identified in each enzyme. Mutations in these allosteric clusters altered phosphatase activity with changes in kcat/KM ranging from 30% to nearly 100-fold. This work outlines a simple method for identification of new allosteric sites in PTP, and given the basis of this method in thermodynamics, it is expected to be generally useful in other systems.


Subject(s)
Allosteric Site , Dual Specificity Phosphatase 3/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Amino Acid Substitution , DNA Mutational Analysis , Dual Specificity Phosphatase 3/genetics , Dual Specificity Phosphatase 3/metabolism , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
4.
Biochem Mol Biol Educ ; 45(5): 403-410, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28294503

ABSTRACT

Here, we present a 10-week project-oriented laboratory module designed to provide a course-based undergraduate research experience in biochemistry that emphasizes the importance of biomolecular structure and dynamics in enzyme function. This module explores the impact of mutagenesis on an important active site loop for a biomedically-relevant human enzyme, protein tyrosine phosphatase 1B (PTP1B). Over the course of the semester students guide their own mutant of PTP1B from conception to characterization in a cost-effective manner and gain exposure to fundamental techniques in biochemistry, including site-directed DNA mutagenesis, bacterial recombinant protein expression, affinity column purification, protein quantitation, SDS-PAGE, and enzyme kinetics. This project-based approach allows an instructor to simulate a research setting and prepare students for productive research beyond the classroom. Potential modifications to expand or contract this module are also provided. © 2017 by The International Union of Biochemistry and Molecular Biology, 45(5):403-410, 2017.


Subject(s)
Biochemistry/education , Laboratories , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Research/education , Humans , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/isolation & purification , Students
5.
Biochemistry ; 56(1): 96-106, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27959494

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) is a known regulator of the insulin and leptin signaling pathways and is an active target for the design of inhibitors for the treatment of type II diabetes and obesity. Recently, cichoric acid (CHA) and chlorogenic acid (CGA) were predicted by docking methods to be allosteric inhibitors that bind distal to the active site. However, using a combination of steady-state inhibition kinetics, solution nuclear magnetic resonance experiments, and molecular dynamics simulations, we show that CHA is a competitive inhibitor that binds in the active site of PTP1B. CGA, while a noncompetitive inhibitor, binds in the second aryl phosphate binding site, rather than the predicted benzfuran binding pocket. The molecular dynamics simulations of the apo enzyme and cysteine-phosphoryl intermediate states with and without bound CGA suggest CGA binding inhibits PTP1B by altering hydrogen bonding patterns at the active site. This study provides a mechanistic understanding of the allosteric inhibition of PTP1B.


Subject(s)
Caffeic Acids/pharmacology , Chlorogenic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Succinates/pharmacology , Algorithms , Allosteric Regulation , Binding Sites , Binding, Competitive , Caffeic Acids/chemistry , Caffeic Acids/metabolism , Catalytic Domain , Chlorogenic Acid/chemistry , Chlorogenic Acid/metabolism , Enzyme Inhibitors/metabolism , Humans , Hydrogen Bonding , Kinetics , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Protein Binding , Protein Domains , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Succinates/chemistry , Succinates/metabolism
6.
Biochem Mol Biol Educ ; 44(6): 550-554, 2016 Nov 12.
Article in English | MEDLINE | ID: mdl-27123933

ABSTRACT

Fundamental concepts in biochemistry important for drug design often lack connection to the macroscopic world and can be difficult for students to grasp, particularly those in introductory science courses at the high school and college level. Educational research has shown that multisensory teaching facilitates learning, but teaching at the high school and college level is almost exclusively limited to the visual and auditory senses. This approach neglects the lifetime of experience our students bring to the classroom in the form of taste perception and makes our teaching less supportive of those with sensory impairment. In this article, we outline a novel guided-inquiry activity that utilizes taste perception for a series of natural and artificial sweetener solutions to introduce the concepts of substrate affinity and selectivity in the context of drug design. The findings from this study demonstrate clear gains in student knowledge, as well as an increase in enthusiasm for the fields of biochemistry and drug design. © 2016 by The International Union of Biochemistry and Molecular Biology, 44(6):550-554, 2016.


Subject(s)
Biochemistry/education , Biomedical Research/education , Drug Design , Problem-Based Learning/methods , Students/psychology , Sweetening Agents/analysis , Curriculum , Educational Measurement , Humans , Laboratories/standards , Models, Biological , Taste/physiology
7.
Structure ; 18(12): 1596-607, 2010 Dec 08.
Article in English | MEDLINE | ID: mdl-21134639

ABSTRACT

Imidazole glycerol phosphate synthase (IGPS) is a V-type allosteric enzyme, which is catalytically inactive for glutamine hydrolysis until the allosteric effector, N'-[(5'-phosphoribulosyl)formimino]-5-aminoimidazole-4-carboxamide-ribonucleotide (PRFAR) binds 30 Å away. In the apo state, NMR relaxation dispersion experiments indicate the absence of millisecond (ms) timescale motions. Binding of the PRFAR to form the active ternary complex is endothermic with a large positive entropy change. In addition, there is a protein wide enhancement of conformational motions in the ternary complex, which connect the two active sites. NMR chemical shift changes and acrylamide quenching experiments suggest that little in the way of structural changes accompany these motions. The data indicate that enzyme activation in the ternary complex is primarily due to an enhancement of ms motions that allows formation of a population of enzymatically active conformers.


Subject(s)
Allosteric Site/physiology , Aminohydrolases/chemistry , Aminohydrolases/metabolism , Motion , Aminohydrolases/physiology , Binding Sites , Entropy , Imidazoles/chemistry , Imidazoles/metabolism , Kinetics , Ligands , Magnetic Resonance Imaging , Models, Biological , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Ribonucleotides/chemistry , Ribonucleotides/metabolism , Time Factors
8.
J Biomol NMR ; 45(1-2): 73-84, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19565337

ABSTRACT

IGPS is a 51 kDa heterodimeric enzyme comprised of two proteins, HisH and HisF, that catalyze the hydrolysis of glutamine to produce NH(3) in the HisH active site and the cyclization of ammonia with N'-[(5'-phosphoribulosyl)formimino]-5-aminoimidazole-4-carboxamide-ribonucleotide (PRFAR) in HisF to produce imidazole glycerol phosphate (IGP) and 5-aminoimidazole-4-carboxamide ribotide (AICAR). Binding of PRFAR and IGP stimulates glutaminase activity in the HisH enzyme over 5,000 and 100-fold, respectively, despite the active sites being >25 A apart. The details of this long-range protein communication process were investigated by solution NMR spectroscopy and CPMG relaxation dispersion experiments. Formation of the heterodimer enzyme results in a reduction in millisecond motions in HisF that extend throughout the protein. Binding of lGP results in an increase in protein-wide millisecond dynamics evidenced as severe NMR line broadening and elevated R (ex) values. Together, these data demonstrate a grouping of flexible residues that link the HisF active site with the protein interface to which HisH binds and provide a model for the path of communication between the IGPS active sites.


Subject(s)
Aminohydrolases/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Thermotoga maritima/enzymology , Allosteric Regulation , Catalytic Domain , Models, Biological , Models, Molecular , Protein Conformation
9.
Methods Mol Biol ; 490: 115-34, 2009.
Article in English | MEDLINE | ID: mdl-19157081

ABSTRACT

The ability of proteins to interact with small molecules or other proteins is essential in all aspects of biology. In many cases these interactions cause detectable changes in NMR chemical shifts, lineshapes, and relaxation rates and therefore provide a means by which to study these biologically important phenomena. Here we review the theory upon which this analysis is based, provide several illustrative examples, and highlight potential problems in the study of binding interactions by solution NMR.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Proteins/chemistry , Protein Binding , Protein Conformation
10.
Biomol NMR Assign ; 2(2): 219-21, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19636909

ABSTRACT

HisF comprises one half of the heterodimeric protein complex imidazole glycerol phosphate (IGP) synthase responsible for the fifth step of histidine biosynthesis. Here we report backbone and side chain assignments necessary for characterization of protein dynamics involved in the allosteric mechanism of IGP synthase.


Subject(s)
Aminohydrolases/chemistry , Magnetic Resonance Spectroscopy/methods , Thermotoga maritima/enzymology , Amino Acid Sequence , Carbon Isotopes/chemistry , Molecular Sequence Data , Molecular Weight , Nitrogen Isotopes/chemistry , Protein Structure, Tertiary , Protons
11.
J Med Chem ; 46(10): 1940-7, 2003 May 08.
Article in English | MEDLINE | ID: mdl-12723956

ABSTRACT

The energies and physical descriptors for the binding of 20 novel 1-(2,6-difluorobenzyl)-2-(2,6-difluorophenyl)benzimidazole analogues (BPBIs) to HIV-1 reverse transcriptase (RT) have been determined using Monte Carlo (MC) simulations. The crystallographic structure of the lead compound, 1-(2,6-difluorobenzyl)-2-(2,6-difluorophenyl)-4-methylbenzimidazole, was used as a starting point to model the inhibitors in both the bound and the unbound states. The energy terms and physical descriptors obtained from the calculations were correlated with their respective experimental EC(50) values, resulting in an r(2) value of 0.70 and a root-mean-square deviation (rms) of 0.53 kcal/mol. The terms in the correlation include the change in total Coulombic energy and solvent-accessible surface area. Structural analysis of the data files from the BPBI calculations reveals that all of the analogues with good biological activity show the formation of a hydrogen bond between the ligand and the backbone nitrogen atom of lysine 103. By use of the structural results, two novel BPBI inhibitors have been designed and calculations have been carried out. The results show the formation of the desired hydrogen bonds, and the DeltaG(binding) values predict the compounds to be excellent RT inhibitors. Subsequent synthesis and biological activity testing of these analogues have shown the validity of the predictive calculations. If the BPBIs are modeled in a site constructed from the crystal coordinates of a member of another class of nonnucleoside inhibitors (the 4,5,6,7-tetrahydroimidazo[4,5,1-jk][1,4]benzodiazepine-2(1H)-thione and -one (TIBO) compounds), the correlation with the same terms drops slightly, giving an r(2) value of 0.61 with an associated root-mean-square value of 0.53 kcal/mol. Conversely, if the TIBO compounds are modeled in a site constructed from the BPBI complex crystal coordinates, a correlation can be obtained using the drug-protein interaction energy and change in the total number of hydrogen bonds, giving an r(2) value of 0.63. These are the same descriptors that were used for the TIBO compounds modeled in their own sites, where the r(2) value was 0.72. These data suggest that it may be possible, in some cases, to design novel inhibitors utilizing structural data from related, but not identical, inhibitors.


Subject(s)
Benzimidazoles/chemistry , HIV Reverse Transcriptase/antagonists & inhibitors , Reverse Transcriptase Inhibitors/chemistry , Benzodiazepines/chemistry , Binding Sites , Crystallography, X-Ray , HIV Reverse Transcriptase/chemistry , Models, Molecular , Monte Carlo Method , Protein Binding , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...