Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Phys Chem B ; 127(39): 8344-8357, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37751332

ABSTRACT

Monoclonal antibodies (mAbs) make up a major class of biotherapeutics with a wide range of clinical applications. Their physical stability can be affected by various environmental factors. For instance, an acidic pH can be encountered during different stages of the mAb manufacturing process, including purification and storage. Therefore, understanding the behavior of flexible mAb molecules in acidic solution environments will benefit the development of stable mAb products. This study used small-angle X-ray scattering (SAXS) and complementary biophysical characterization techniques to investigate the conformational flexibility and protein-protein interactions (PPI) of a model mAb molecule under near-neutral and acidic conditions. The study also characterized the interactions between Fab and Fc fragments under the same buffer conditions to identify domain-domain interactions. The results suggest that solution pH significantly influences mAb flexibility and thus could help mAbs remain physically stable by maximizing local electrostatic repulsions when mAbs become crowded in solution. Under acidic buffer conditions, both Fab and Fc contribute to the repulsive PPI observed among the full mAb at a low ionic strength. However, as ionic strength increases, hydrophobic interactions lead to the self-association of Fc fragments and, subsequently, could affect the aggregation state of the mAb.


Subject(s)
Antibodies, Monoclonal , Immunoglobulin G , Antibodies, Monoclonal/chemistry , Scattering, Small Angle , Immunoglobulin G/chemistry , X-Ray Diffraction , Sodium Chloride , Acids , Immunoglobulin Fc Fragments/chemistry , Hydrogen-Ion Concentration
2.
J Mol Biol ; 434(2): 167391, 2022 01 30.
Article in English | MEDLINE | ID: mdl-34890647

ABSTRACT

Previous reports present different models for the stabilization of the Fc-FcγRI immune complex. Although accord exists on the importance of L235 in IgG1 and some hydrophobic contacts for complex stabilization, discord exists regarding the existence of stabilizing glycoprotein contacts between glycans of IgG1 and a conserved FG-loop (171MGKHRY176) of FcγRIa. Complexes formed from the FcγRIa receptor and IgG1s containing biantennary glycans with N-acetylglucosamine, galactose, and α2,6-N-acetylneuraminic terminations were measured by hydrogen-deuterium exchange mass spectrometry (HDX-MS), classified for dissimilarity with Welch's ANOVA and Games-Howell post hoc procedures, and modeled with molecular dynamics (MD) simulations. For each glycoform of the IgG1-FcγRIa complex peptic peptides of Fab, Fc and FcγRIa report distinct H/D exchange rates. MD simulations corroborate the differences in the peptide deuterium content through calculation of the percent of time that transient glycan-peptide bonds exist. These results indicate that stability of IgG1-FcγRIa complexes correlate with the presence of intermolecular glycoprotein interactions between the IgG1 glycans and the 173KHR175 motif within the FG-loop of FcγRIa. The results also indicate that intramolecular glycan-protein bonds stabilize the Fc region in isolated and complexed IgG1. Moreover, HDX-MS data evince that the Fab domain has glycan-protein binding contacts within the IgG1-FcγRI complex.


Subject(s)
Antigen-Antibody Complex/chemistry , Glycoproteins/chemistry , Hydrogen Deuterium Exchange-Mass Spectrometry/methods , Immunoglobulin G/chemistry , Molecular Dynamics Simulation , Receptors, IgG/chemistry , Antibodies, Monoclonal/chemistry , Antigen-Antibody Complex/metabolism , Galactose , Glycoproteins/metabolism , Membrane Proteins/chemistry , Peptides/chemistry , Peptides/metabolism , Polysaccharides , Protein Binding
3.
J Pharm Sci ; 110(5): 1948-1957, 2021 05.
Article in English | MEDLINE | ID: mdl-33453208

ABSTRACT

Protein aggregation can affect the quality of protein-based therapeutics. Attempting to unravel factors influencing protein aggregation involves systematic studies. These studies often include sodium azide or similar preservatives in the aggregation buffer. This work shows effects of azide on aggregation of two highly purified reference proteins, both a bovine serum albumin (BSA) as well as a monoclonal antibody (NISTmAb). The proteins were aggregated by thermomechanical stress, consisting of simultaneous heating of the solution with gentle agitation. Protein aggregates were characterized by asymmetric flow field flow fractionation (AF4) with light scattering measurements along with quantification by UV spectroscopy, revealing strong time-dependent generation of aggregated protein and an increase in aggregate molar mass. Gel electrophoresis was used to probe the reversibility of the aggregation and demonstrated complete reversibility for the NISTmAb, but not so for the BSA. Kinetic fitting to a commonly implemented nucleated polymerization model was also employed to provide mechanistic details into the kinetic process. The model suggests that the aggregation of the NISTmAb proceeds via nucleated growth and aggregate-aggregate condensation in a way that is dependent on the concentration (and presence) of the azide anion. This work overall implicates azide preservatives as having demonstrable effects on thermomechanical stress and aggregation of proteins undergoing systematic aggregation and stability studies.


Subject(s)
Azides , Fractionation, Field Flow , Kinetics , Serum Albumin, Bovine , Spectrum Analysis
5.
Sci Rep ; 10(1): 18531, 2020 10 28.
Article in English | MEDLINE | ID: mdl-33116203

ABSTRACT

Inhibition of human Monoacylglycerol Lipase (hMGL) offers a novel approach for treating neurological diseases. The design of inhibitors, targeting active-inactive conformational transitions of the enzyme, can be aided by understanding the interplay between structure and dynamics. Here, we report the effects of mutations within the catalytic triad on structure, conformational gating and dynamics of hMGL by combining kinetics, NMR, and HDX-MS data with metadynamics simulations. We found that point mutations alter delicate conformational equilibria between active and inactive states. HDX-MS reveals regions of the hMGL that become substantially more dynamic upon substitution of catalytic acid Asp-239 by alanine. These regions, located far from the catalytic triad, include not only loops but also rigid α-helixes and ß-strands, suggesting their involvement in allosteric regulation as channels for long-range signal transmission. The results identify the existence of a preorganized global communication network comprising of tertiary (residue-residue contacts) and quaternary (rigid-body contacts) networks that mediate robust, rapid intraprotein signal transmission. Catalytic Asp-239 controls hMGL allosteric communications and may be considered as an essential residue for the integration and transmission of information to enzymes' remote regions, in addition to its well-known role to facilitate Ser-122 activation. Our findings may assist in the identification of new druggable sites in hMGL.


Subject(s)
Monoacylglycerol Lipases/genetics , Monoacylglycerol Lipases/metabolism , Monoacylglycerol Lipases/physiology , Allosteric Regulation , Catalysis , Humans , Kinetics , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Molecular Dynamics Simulation , Mutation , Mutation, Missense , Protein Conformation , Structure-Activity Relationship
6.
Mol Cell Proteomics ; 18(10): 2089-2098, 2019 10.
Article in English | MEDLINE | ID: mdl-31409669

ABSTRACT

The N-terminal regions (tails) of histone proteins are dynamic elements that protrude from the nucleosome and are involved in many aspects of chromatin organization. Their epigenetic role is well-established, and post-translational modifications present on these regions contribute to transcriptional regulation. Considering their biological significance, relatively few structural details have been established for histone tails, mainly because of their inherently disordered nature. Although hydrogen/deuterium exchange mass spectrometry (HX-MS) is well-suited for the analysis of dynamic structures, it has seldom been employed in this context, presumably because of the poor N-terminal coverage provided by pepsin. Inspired from histone-clipping events, we profiled the activity of cathepsin-L under HX-MS quench conditions and characterized its specificity employing the four core histones (H2A, H2B, H3 and H4). Cathepsin-L demonstrated cleavage patterns that were substrate- and pH-dependent. Cathepsin-L generated overlapping N-terminal peptides about 20 amino acids long for H2A, H3, and H4 proving its suitability for the analysis of histone tails dynamics. We developed a comprehensive HX-MS method in combination with pepsin and obtained full sequence coverage for all histones. We employed our method to analyze histones H3 and H4. We observe rapid deuterium exchange of the N-terminal tails and cooperative unfolding (EX1 kinetics) in the histone-fold domains of histone monomers in-solution. Overall, this novel strategy opens new avenues for investigating the dynamic properties of histones that are not apparent from the crystal structures, providing insights into the structural basis of the histone code.


Subject(s)
Cathepsin L/metabolism , Histones/chemistry , Histone Code , Humans , Hydrogen Deuterium Exchange-Mass Spectrometry , Hydrogen-Ion Concentration , Models, Molecular , Protein Conformation , Protein Folding , Proteolysis , Substrate Specificity
7.
Anal Chem ; 91(11): 7336-7345, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31045344

ABSTRACT

Hydrogen-deuterium exchange mass spectrometry (HDX-MS) is an established, powerful tool for investigating protein-ligand interactions, protein folding, and protein dynamics. However, HDX-MS is still an emergent tool for quality control of biopharmaceuticals and for establishing dynamic similarity between a biosimilar and an innovator therapeutic. Because industry will conduct quality control and similarity measurements over a product lifetime and in multiple locations, an understanding of HDX-MS reproducibility is critical. To determine the reproducibility of continuous-labeling, bottom-up HDX-MS measurements, the present interlaboratory comparison project evaluated deuterium uptake data from the Fab fragment of NISTmAb reference material (PDB: 5K8A ) from 15 laboratories. Laboratories reported ∼89 800 centroid measurements for 430 proteolytic peptide sequences of the Fab fragment (∼78 900 centroids), giving ∼100% coverage, and ∼10 900 centroid measurements for 77 peptide sequences of the Fc fragment. Nearly half of peptide sequences are unique to the reporting laboratory, and only two sequences are reported by all laboratories. The majority of the laboratories (87%) exhibited centroid mass laboratory repeatability precisions of ⟨ sLab⟩ ≤ (0.15 ± 0.01) Da (1σx̅). All laboratories achieved ⟨sLab⟩ ≤ 0.4 Da. For immersions of protein at THDX = (3.6 to 25) °C and for D2O exchange times of tHDX = (30 s to 4 h) the reproducibility of back-exchange corrected, deuterium uptake measurements for the 15 laboratories is σreproducibility15 Laboratories( tHDX) = (9.0 ± 0.9) % (1σ). A nine laboratory cohort that immersed samples at THDX = 25 °C exhibited reproducibility of σreproducibility25C cohort( tHDX) = (6.5 ± 0.6) % for back-exchange corrected, deuterium uptake measurements.


Subject(s)
Antibodies, Monoclonal/chemistry , Hydrogen Deuterium Exchange-Mass Spectrometry , Immunoglobulin Fab Fragments/analysis
9.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 9): 524-529, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30198883

ABSTRACT

As the link between antigen binding and immune activation, the antibody Fc region has received extensive structural study. In this report, the structure of the Fc fragment of the NIST IgG1 mAb (reference material 8671) is described at 2.1 Šresolution in space group P212121, with approximate unit-cell parameters a = 50, b = 80, c = 138 Å. Prior Fc structures with a wide variety of modifications are also surveyed, focusing on those in the same crystal form. To facilitate the analysis of conformations, a reference frame and a two-parameter metric are proposed, considering the CH2 domains as mobile with respect to a fixed dimeric CH3 core. Over several human Fc structures, a significant variation in Fc elbow conformations is observed, which may serve to facilitate the regulation of Fc effector signaling.


Subject(s)
Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Protein Subunits/chemistry , Receptors, Fc/chemistry , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Subunits/genetics , Protein Subunits/immunology , Receptors, Fc/genetics , Receptors, Fc/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Thermodynamics
10.
Sci Rep ; 8(1): 1719, 2018 01 29.
Article in English | MEDLINE | ID: mdl-29379013

ABSTRACT

An understanding of how conformational dynamics modulates function and catalysis of human monoacylglycerol lipase (hMGL), an important pharmaceutical target, can facilitate the development of novel ligands with potential therapeutic value. Here, we report the discovery and characterization of an allosteric, regulatory hMGL site comprised of residues Trp-289 and Leu-232 that reside over 18 Å away from the catalytic triad. These residues were identified as critical mediators of long-range communication and as important contributors to the integrity of the hMGL structure. Nonconservative replacements of Trp-289 or Leu-232 triggered concerted motions of structurally distinct regions with a significant conformational shift toward inactive states and dramatic loss in catalytic efficiency of the enzyme. Using a multimethod approach, we show that the dynamically relevant Trp-289 and Leu-232 residues serve as communication hubs within an allosteric protein network that controls signal propagation to the active site, and thus, regulates active-inactive interconversion of hMGL. Our findings provide new insights into the mechanism of allosteric regulation of lipase activity, in general, and may provide alternative drug design possibilities.


Subject(s)
Monoacylglycerol Lipases/genetics , Monoacylglycerol Lipases/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Allosteric Regulation , Amino Acid Substitution , DNA Mutational Analysis , Humans , Models, Molecular , Monoacylglycerol Lipases/chemistry , Mutant Proteins/chemistry , Protein Conformation
11.
Biologicals ; 50: 27-34, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28965821

ABSTRACT

Monoclonal antibody pharmaceuticals are the fastest-growing class of therapeutics, with a wide range of clinical applications. To assure their safety, these protein drugs must demonstrate highly consistent purity and stability. Key to these objectives is higher order structure measurements validated by calibration to reference materials. We describe preparation, characterization, and crystal structure of the Fab fragment prepared from the NIST Reference Antibody RM 8671 (NISTmAb). NISTmAb is a humanized IgG1κ antibody, produced in murine cell culture and purified by standard biopharmaceutical production methods, developed at the National Institute of Standards and Technology (NIST) to serve as a reference material. The Fab fragment was derived from NISTmAb through papain cleavage followed by protein A based purification. The purified Fab fragment was characterized by SDS-PAGE, capillary gel electrophoresis, multi-angle light scattering, size exclusion chromatography, mass spectrometry, and x-ray crystallography. The crystal structure at 0.2 nm resolution includes four independent Fab molecules with complete light chains and heavy chains through Cys 223, enabling assessment of conformational variability and providing a well-characterized reference structure for research and engineering applications. This nonproprietary, publically available reference material of known higher-order structure can support metrology in biopharmaceutical applications, and it is a suitable platform for validation of molecular modeling studies.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin G/chemistry , Protein Domains , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/isolation & purification , Biophysical Phenomena , Chromatography, Gel , Crystallography, X-Ray , Electrophoresis, Capillary , Electrophoresis, Polyacrylamide Gel , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/isolation & purification , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Mass Spectrometry , Mice , Models, Molecular , Protein Binding/immunology , Reference Standards
12.
Anal Biochem ; 536: 90-95, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28822686

ABSTRACT

Human monoacylglycerol lipase (hMAGL) plays a key role in homeostatic tuning of the endocannabinoid signaling system and supports aggressive tumorogenesis, making this enzyme a promising therapeutic target. hMAGL features a membrane-associated lid domain that regulates entry of endocannabinoid lipid substrates into the hydrophobic channel accessing the active site, likely from the membrane bilayer. The present work applied simultaneous surface plasmon resonance and electrochemical impedance spectroscopy measurements to show that, in absence of the substrate, hMAGL can remove phospholipid molecules from the membrane and, thereby, disintegrate pre-formed, intact, tethered phospholipid bilayer membrane mimetics (tBLMs) composed of unsaturated phosphatidylcholines. To probe the mechanism of hMAGL-induced on tBLMs compromise, we investigated the effect of wild type and mutant hMAGLs and hMAGL rendered catalytically inactive, as a function of concentration and in the presence of chemically distinct active-site inhibitors. Our data show that hMAGL's lid domain and hydrophobic substrate-binding pocket play important roles in hMAGL-induced bilayer lipid mobilization, whereas hydrolytic activity of the enzyme does not appear to be a factor.


Subject(s)
Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Monoacylglycerol Lipases/chemistry , Monoacylglycerol Lipases/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Binding Sites , Dielectric Spectroscopy , Humans , Monoacylglycerol Lipases/genetics , Mutation , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Surface Plasmon Resonance
13.
Biointerphases ; 12(2): 02D408, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28511544

ABSTRACT

The structural characterization of peripheral membrane proteins represents a tremendous challenge in structural biology due to their transient interaction with the membrane and the potential multitude of protein conformations during this interaction. Neutron reflectometry is uniquely suited to address this problem because of its ability to structurally characterize biological model systems nondestructively and under biomimetic conditions that retain full protein functionality. Being sensitive to only the membrane-bound fraction of a water-soluble peripheral protein, neutron reflectometry obtains a low-resolution average structure of the protein-membrane complex that is further refined using integrative modeling strategies. Here, the authors review the current technological state of biological neutron reflectometry exemplified by a detailed report on the structure determination of the myristoylated human immunodeficiency virus-1 (HIV-1) Gag matrix associated with phosphoserine-containing model membranes. The authors found that the HIV-1 Gag matrix is able to adopt different configurations at the membrane in a pH-dependent manner and that the myristate group orients the protein in a way that is conducive to PIP2-binding.


Subject(s)
Cell Membrane/chemistry , HIV-1/chemistry , Lipoylation , Membranes, Artificial , Neutron Diffraction , gag Gene Products, Human Immunodeficiency Virus/chemistry , Humans , Phosphoserine/chemistry
14.
J Virol ; 90(9): 4544-4555, 2016 May.
Article in English | MEDLINE | ID: mdl-26912608

ABSTRACT

UNLABELLED: By assembling in a protein lattice on the host's plasma membrane, the retroviral Gag polyprotein triggers formation of the viral protein/membrane shell. The MA domain of Gag employs multiple signals--electrostatic, hydrophobic, and lipid-specific-to bring the protein to the plasma membrane, thereby complementing protein-protein interactions, located in full-length Gag, in lattice formation. We report the interaction of myristoylated and unmyristoylated HIV-1 Gag MA domains with bilayers composed of purified lipid components to dissect these complex membrane signals and quantify their contributions to the overall interaction. Surface plasmon resonance on well-defined planar membrane models is used to quantify binding affinities and amounts of protein and yields free binding energy contributions, ΔG, of the various signals. Charge-charge interactions in the absence of the phosphatidylinositide PI(4,5)P2 attract the protein to acidic membrane surfaces, and myristoylation increases the affinity by a factor of 10; thus, our data do not provide evidence for a PI(4,5)P2 trigger of myristate exposure. Lipid-specific interactions with PI(4,5)P2, the major signal lipid in the inner plasma membrane, increase membrane attraction at a level similar to that of protein lipidation. While cholesterol does not directly engage in interactions, it augments protein affinity strongly by facilitating efficient myristate insertion and PI(4,5)P2 binding. We thus observe that the isolated MA protein, in the absence of protein-protein interaction conferred by the full-length Gag, binds the membrane with submicromolar affinities. IMPORTANCE: Like other retroviral species, the Gag polyprotein of HIV-1 contains three major domains: the N-terminal, myristoylated MA domain that targets the protein to the plasma membrane of the host; a central capsid-forming domain; and the C-terminal, genome-binding nucleocapsid domain. These domains act in concert to condense Gag into a membrane-bounded protein lattice that recruits genomic RNA into the virus and forms the shell of a budding immature viral capsid. In binding studies of HIV-1 Gag MA to model membranes with well-controlled lipid composition, we dissect the multiple interactions of the MA domain with its target membrane. This results in a detailed understanding of the thermodynamic aspects that determine membrane association, preferential lipid recruitment to the viral shell, and those aspects of Gag assembly into the membrane-bound protein lattice that are determined by MA.


Subject(s)
Cell Membrane/metabolism , HIV Antigens/metabolism , HIV-1/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , Cell Membrane/chemistry , Cholesterol/chemistry , Cholesterol/metabolism , Humans , Kinetics , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Lipid-Linked Proteins/metabolism , Lipids/chemistry , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Binding , gag Gene Products, Human Immunodeficiency Virus/chemistry
15.
J Biol Chem ; 291(6): 2556-65, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26555264

ABSTRACT

The serine hydrolase monoacylglycerol lipase (MGL) functions as the main metabolizing enzyme of 2-arachidonoyl glycerol, an endocannabinoid signaling lipid whose elevation through genetic or pharmacological MGL ablation exerts therapeutic effects in various preclinical disease models. To inform structure-based MGL inhibitor design, we report the direct NMR detection of a reversible equilibrium between active and inactive states of human MGL (hMGL) that is slow on the NMR time scale and can be modulated in a controlled manner by pH, temperature, and select point mutations. Kinetic measurements revealed that hMGL substrate turnover is rate-limited across this equilibrium. We identify a network of aromatic interactions and hydrogen bonds that regulates hMGL active-inactive state interconversion. The data highlight specific inter-residue interactions within hMGL modulating the enzymes function and implicate transitions between active (open) and inactive (closed) states of the hMGL lid domain in controlling substrate access to the enzymes active site.


Subject(s)
Monoacylglycerol Lipases/chemistry , Humans , Hydrogen-Ion Concentration , Kinetics , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary , Structure-Activity Relationship
16.
Hum Genomics ; 9: 12, 2015 Jun 20.
Article in English | MEDLINE | ID: mdl-26092435

ABSTRACT

Cancer, like many common disorders, has a complex etiology, often with a strong genetic component and with multiple environmental factors contributing to susceptibility. A considerable number of genomic variants have been previously reported to be causative of, or associated with, an increased risk for various types of cancer. Here, we adopted a next-generation sequencing approach in 11 members of two families of Greek descent to identify all genomic variants with the potential to predispose family members to cancer. Cross-comparison with data from the Human Gene Mutation Database identified a total of 571 variants, from which 47 % were disease-associated polymorphisms, 26 % disease-associated polymorphisms with additional supporting functional evidence, 19 % functional polymorphisms with in vitro/laboratory or in vivo supporting evidence but no known disease association, 4 % putative disease-causing mutations but with some residual doubt as to their pathological significance, and 3 % disease-causing mutations. Subsequent analysis, focused on the latter variant class most likely to be involved in cancer predisposition, revealed two variants of prime interest, namely MSH2 c.2732T>A (p.L911R) and BRCA1 c.2955delC, the first of which is novel. KMT2D c.13895delC and c.1940C>A variants are additionally reported as incidental findings. The next-generation sequencing-based family genomics approach described herein has the potential to be applied to other types of complex genetic disorder in order to identify variants of potential pathological significance.


Subject(s)
Genetic Predisposition to Disease , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Neoplasms/genetics , BRCA1 Protein/genetics , DNA-Binding Proteins/genetics , Humans , MutS Homolog 2 Protein/genetics , Mutation , Neoplasm Proteins/genetics , Neoplasms/pathology , Pedigree , Polymorphism, Single Nucleotide
17.
FEBS J ; 282(5): 864-73, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25546421

ABSTRACT

ß2 glycoprotein I (ß2GPI) is the most common antigen for autoimmune antibodies in antiphospholipid syndrome (APS). Thrombosis is a clinical feature of APS. We created a molecule (A1-A1) that consists of two identical ß2GPI-binding modules from ApoE receptor 2 (ApoER2). A1-A1 binds to ß2GPI/antibody complexes, preventing their association with ApoER2 and anionic phospholipids, and reducing thrombus size in the mouse model of APS. Here, we describe a mutant of A1-A1 (mA1-A1ND) with improved affinity for ß2GPI. mA1-A1ND inhibits the binding of ß2GPI to cardiolipin in the presence of anti-ß2GPI antibodies, and inhibits the binding to phospholipids in plasma samples of APS patients, affecting the clotting time. Reduction of the clotting time demonstrates the presence of soluble ß2GPI/antibody complexes in patients' plasma. These complexes either already exist in patients' plasma or form rapidly in the proximity to phospholipids. All members of the low-density lipoprotein receptor family bind ß2GPI. Modeling studies of A1 in a complex with domain V of ß2GPI (ß2GPI-DV) revealed two possible modes of interaction of a ligand-binding module from lipoprotein receptors with ß2GPI-DV. In both orientations, the ligand-binding module interferes with binding of ß2GPI to anionic phospholipids; however, it interacts with two different but overlapping sets of lysine residues in ß2GPI-DV, depending on the orientation.


Subject(s)
Antiphospholipid Syndrome/immunology , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Recombinant Proteins/pharmacology , beta 2-Glycoprotein I/chemistry , beta 2-Glycoprotein I/immunology , beta 2-Glycoprotein I/metabolism , Animals , Antiphospholipid Syndrome/blood , Calcium/metabolism , Cardiolipins/analysis , Cardiolipins/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Ligands , Mice, Inbred BALB C , Models, Molecular , Molecular Docking Simulation , Point Mutation , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Whole Blood Coagulation Time , beta 2-Glycoprotein I/antagonists & inhibitors
18.
Biochemistry ; 52(29): 5016-26, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23795559

ABSTRACT

Human monoacylglycerol lipase (hMGL) regulates endocannabinoid signaling primarily by deactivating the lipid messenger 2-arachidonoylglycerol. Agents that carbamylate hMGLs catalytic Ser(122) constitute a leading class of therapeutically promising hMGL inhibitors. We have applied peptide-level hydrogen/deuterium exchange mass spectrometry to characterize hMGL's conformational responses to two potent carbamylating inhibitors, AM6580 (irreversible) and AM6701 (slowly reversible). A dynamic, solvent-exposed lid domain is characteristic of hMGL's solution conformation. Both hMGL inhibitors restricted backbone enzyme motility in the active-site region and increased substrate binding-pocket solvent exposure. Covalent reaction of AM6580 with hMGL generates a bulkier carbamylated Ser(122) residue as compared to the more discrete Ser(122) modification by AM6701, a difference reflected in AM6580's more pronounced effect upon hMGL conformation. We demonstrate that structurally distinct carbamylating hMGL inhibitors generate particular conformational ensembles characterized by region-specific hMGL dynamics. By demonstrating the distinctive influences of two hMGL inhibitors on enzyme conformation, this study furthers our understanding at the molecular level of the dynamic features of hMGL interaction with small-molecule ligands.


Subject(s)
Enzyme Inhibitors/chemistry , Mass Spectrometry/methods , Monoacylglycerol Lipases/antagonists & inhibitors , Catalytic Domain , Humans , Hydrogen/chemistry , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry
19.
ACS Chem Neurosci ; 3(5): 393-9, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22860208

ABSTRACT

In the mammalian central nervous system, monoacylglycerol lipase (MGL) is principally responsible for inactivating the endocannabinoid signaling lipid 2-arachidonoylglycerol (2-AG) and modulates cannabinoid-1 receptor (CB1R) desensitization and signal intensity. MGL is also a drug target for diseases in which CB1R stimulation may be therapeutic. To inform the design of human MGL (hMGL) inhibitors, we have engineered a Leu(Leu(169);Leu(176))-to-Ser(Ser(169);Ser(176)) double hMGL mutant (sol-hMGL) which exhibited enhanced solubility properties, and we further mutated this variant by substituting its catalytic-triad Ser(122) with Cys (sol-S-hMGL). The hMGL variants hydrolyzed both 2-AG and a fluorogenic reporter substrate with comparable affinities. Our results suggest that the hMGL cysteine mutant maintains the same overall architecture as wild-type hMGL. The results also underscore the superior nucleophilic nature of the reactive catalytic Ser(122) residue as compared to that of Cys(122) in the sol-S-hMGL mutant and suggest that the nucleophilic character of the Cys(122) residue is not commensurately enhanced within the three dimensional architecture of hMGL. The interaction of the sol-hMGL variants with the irreversible inhibitors AM6580 and N-arachidonylmaleimide (NAM) and the reversible inhibitor AM10212 was profiled. LC/MS analysis of tryptic digests from sol-S-hMGL directly demonstrate covalent modification of this variant by NAM and AM6580, consistent with enzyme thiol alkylation and carbamoylation, respectively. These data provide insight into hMGL catalysis, the key role of the nucleophilic character of Ser(122), and the mechanisms underlying hMGL inhibition by different classes of small molecules.


Subject(s)
Endocannabinoids/metabolism , Monoacylglycerol Lipases/antagonists & inhibitors , Monoacylglycerol Lipases/metabolism , Protein Engineering/methods , Sulfhydryl Compounds/metabolism , Amino Acid Substitution/genetics , Endocannabinoids/chemistry , Endocannabinoids/genetics , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Monoacylglycerol Lipases/genetics , Mutation , Solubility , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacology
20.
Mol Biosyst ; 6(8): 1381-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20464001

ABSTRACT

Intramolecular hydrogen bonding is an important determinant of enzyme structure, catalysis, and inhibitor action. Monoacylglycerol lipase (MGL) modulates cannabinergic signaling as the main enzyme responsible for deactivating 2-arachidonoylglycerol (2-AG), a primary endocannabinoid lipid messenger. By enhancing tissue-protective 2-AG tone, targeted MGL inhibitors hold therapeutic promise for managing pain and treating inflammatory and neurodegenerative diseases. We report study of purified, solubilized human MGL (hMGL) to explore the details of hMGL catalysis by using two known covalent hMGL inhibitors, the carbamoyl tetrazole AM6701 and N-arachidonoylmaleimide (NAM), that act through distinct mechanisms. Using proton nuclear magnetic resonance spectroscopy (NMR) with purified wild-type and mutant hMGLs, we have directly observed a strong hydrogen-bond network involving Asp239 and His269 of the catalytic triad and neighboring Leu241 and Cys242 residues. hMGL inhibition by AM6701 alters this hydrogen-bonding pattern through subtle active-site structural rearrangements without influencing hydrogen-bond occupancies. Rapid carbamoylation of hMGL Ser122 by AM6701 and elimination of the leaving group is followed by a slow hydrolysis of the carbamate group, ultimately regenerating catalytically competent hMGL. In contrast, hMGL titration with NAM, which leads to cysteine alkylation, stoichiometrically decreases the population of the active-site hydrogen bonds. NAM prevents reformation of this network, and in this manner inhibits hMGL irreversibly. These data provide detailed molecular insight into the distinctive mechanisms of two covalent hMGL inhibitors and implicate a hydrogen-bond network as a structural feature of hMGL catalytic function.


Subject(s)
Enzyme Inhibitors/pharmacokinetics , Magnetic Resonance Spectroscopy/methods , Monoacylglycerol Lipases/antagonists & inhibitors , Monoacylglycerol Lipases/chemistry , Monoacylglycerol Lipases/metabolism , Amino Acid Substitution/genetics , Catalysis/drug effects , Catalytic Domain/drug effects , Catalytic Domain/genetics , Catalytic Domain/physiology , Enzyme Activation/drug effects , Enzyme Inhibitors/metabolism , Humans , Hydrogen Bonding/drug effects , In Vitro Techniques , Kinetics , Monoacylglycerol Lipases/genetics , Mutagenesis, Site-Directed , Tetrazoles/metabolism , Tetrazoles/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...