Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Lipid Res ; 61(8): 1192-1202, 2020 08.
Article in English | MEDLINE | ID: mdl-32482718

ABSTRACT

Lysosomal acid lipase (LAL) is a serine hydrolase that hydrolyzes cholesteryl ester (CE) and TGs delivered to the lysosomes into free cholesterol and fatty acids. LAL deficiency due to mutations in the LAL gene (LIPA) results in accumulation of TGs and cholesterol esters in various tissues of the body leading to pathological conditions such as Wolman's disease and CE storage disease (CESD). Here, we present the first crystal structure of recombinant human LAL (HLAL) to 2.6 Å resolution in its closed form. The crystal structure was enabled by mutating three of the six potential glycosylation sites. The overall structure of HLAL closely resembles that of the evolutionarily related human gastric lipase (HGL). It consists of a core domain belonging to the classical α/ß hydrolase-fold family with a classical catalytic triad (Ser-153, His-353, Asp-324), an oxyanion hole, and a "cap" domain, which regulates substrate entry to the catalytic site. Most significant structural differences between HLAL and HGL exist at the lid region. Deletion of the short helix, 238NLCFLLC244, at the lid region implied a possible role in regulating the highly hydrophobic substrate binding site from self-oligomerization during interfacial activation. We also performed molecular dynamic simulations of dog gastric lipase (lid-open form) and HLAL to gain insights and speculated a possible role of the human mutant, H274Y, leading to CESD.


Subject(s)
Cholesterol Ester Storage Disease/enzymology , Sterol Esterase/chemistry , Sterol Esterase/metabolism , Cholesterol Ester Storage Disease/genetics , Crystallography, X-Ray , Glycosylation , Humans , Models, Molecular , Mutation , Protein Domains , Sterol Esterase/genetics
2.
Org Lett ; 20(3): 812-815, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29327935

ABSTRACT

The design, synthesis, and application of [4-(acetylamino)phenyl]imidodisulfuryl difluoride (AISF), a shelf-stable, crystalline reagent for the synthesis of sulfur(VI) fluorides, is described. The utility of AISF is demonstrated in the synthesis of a diverse array of aryl fluorosulfates and sulfamoyl fluorides under mild conditions. Additionally, a single-step preparation of AISF was developed that installed the bis(fluorosulfonyl)imide group on acetanilide utilizing an oxidative C-H functionalization protocol.

3.
Nat Commun ; 8(1): 1908, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29199275

ABSTRACT

Lysosomal integral membrane protein-2 (LIMP-2/SCARB2) contributes to endosomal and lysosomal function. LIMP-2 deficiency is associated with neurological abnormalities and kidney failure and, as an acid glucocerebrosidase receptor, impacts Gaucher and Parkinson's diseases. Here we report a crystal structure of a LIMP-2 luminal domain dimer with bound cholesterol and phosphatidylcholine. Binding of these lipids alters LIMP-2 from functioning as a glucocerebrosidase-binding monomer toward a dimeric state that preferentially binds anionic phosphatidylserine over neutral phosphatidylcholine. In cellular uptake experiments, LIMP-2 facilitates transport of phospholipids into murine fibroblasts, with a strong substrate preference for phosphatidylserine. Taken together, these biophysical and cellular studies define the structural basis and functional importance of a form of LIMP-2 for lipid trafficking. We propose a model whereby switching between monomeric and dimeric forms allows LIMP-2 to engage distinct binding partners, a mechanism that may be shared by SR-BI and CD36, scavenger receptor proteins highly homologous to LIMP-2.


Subject(s)
CD36 Antigens/metabolism , Cholesterol/metabolism , Lysosomal Membrane Proteins/metabolism , Phosphatidylcholines/metabolism , Phosphatidylserines/metabolism , Receptors, Scavenger/metabolism , Animals , Crystallography, X-Ray , Fibroblasts/metabolism , HEK293 Cells , Humans , Mice , Phospholipids/metabolism
4.
ACS Chem Biol ; 12(8): 2015-2020, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28718624

ABSTRACT

Irreversible enzyme inhibitors and covalent chemical biology probes often utilize the reaction of a protein cysteine residue with an appropriately positioned electrophile (e.g., acrylamide) on the ligand template. However, cysteine residues are not always available for site-specific protein labeling, and therefore new approaches are needed to expand the toolkit of appropriate electrophiles ("warheads") that target alternative amino acids. We previously described the rational targeting of tyrosine residues in the active site of a protein (the mRNA decapping scavenger enzyme, DcpS) using inhibitors armed with a sulfonyl fluoride electrophile. These inhibitors subsequently enabled the development of clickable probe technology to measure drug-target occupancy in live cells. Here we describe a fluorosulfate-containing inhibitor (aryl fluorosulfate probe (FS-p1)) with excellent chemical and metabolic stability that reacts selectively with a noncatalytic serine residue in the same active site of DcpS as confirmed by peptide mapping experiments. Our results suggest that noncatalytic serine targeting using fluorosulfate electrophilic warheads could be a suitable strategy for the development of covalent inhibitor drugs and chemical probes.


Subject(s)
Enzyme Inhibitors/chemistry , Fluorides/chemistry , Serine/chemistry , Sulfuric Acids/chemistry , Animals , Catalytic Domain , Cell Line , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Enzyme Stability , Humans
5.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 11): 840-845, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27827355

ABSTRACT

Crystals of phosphorylated JAK1 kinase domain were initially generated in complex with nucleotide (ADP) and magnesium. The tightly bound Mg2+-ADP at the ATP-binding site proved recalcitrant to ligand displacement. Addition of a molar excess of EDTA helped to dislodge the divalent metal ion, promoting the release of ADP and allowing facile exchange with ATP-competitive small-molecule ligands. Many kinases require the presence of a stabilizing ligand in the ATP site for crystallization. This procedure could be useful for developing co-crystallization systems with an exchangeable ligand to enable structure-based drug design of other protein kinases.


Subject(s)
Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Crystallization/methods , Edetic Acid/chemistry , Janus Kinase 1/chemistry , Magnesium/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Binding Sites , Cations, Divalent , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , Humans , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Magnesium/metabolism , Models, Molecular , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sf9 Cells , Spodoptera
6.
Sci Rep ; 6: 30859, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27527709

ABSTRACT

Interleukin-17A (IL-17A) is a principal driver of multiple inflammatory and immune disorders. Antibodies that neutralize IL-17A or its receptor (IL-17RA) deliver efficacy in autoimmune diseases, but no small-molecule IL-17A antagonists have yet progressed into clinical trials. Investigation of a series of linear peptide ligands to IL-17A and characterization of their binding site has enabled the design of novel macrocyclic ligands that are themselves potent IL-17A antagonists.


Subject(s)
Interleukin-17/antagonists & inhibitors , Interleukin-17/chemistry , Peptides, Cyclic/pharmacology , Small Molecule Libraries/pharmacology , Algorithms , Binding Sites , Cells, Cultured , Drug Design , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Molecular Dynamics Simulation , Peptides, Cyclic/chemistry , Protein Binding , Small Molecule Libraries/chemistry , Structure-Activity Relationship
7.
Biochem J ; 473(5): 581-92, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26635351

ABSTRACT

AMP-activated protein kinase (AMPK) is a serine/threonine protein kinase that serves as a pleotropic regulator of whole body energy homoeostasis. AMPK exists as a heterotrimeric complex, composed of a catalytic subunit (α) and two regulatory subunits (ß and γ), each present as multiple isoforms. In the present study, we compared the enzyme kinetics and allosteric modulation of six recombinant AMPK isoforms, α1ß1γ1, α1ß2γ1, α1ß2γ3, α2ß1γ1, α2ß2γ1 and α2ß2γ3 using known activators, A769662 and AMP. The α1-containing complexes exhibited higher specific activities and lower Km values for a widely used peptide substrate (SAMS) compared with α2-complexes. Surface plasmon resonance (SPR)-based direct binding measurements revealed biphasic binding modes with two distinct equilibrium binding constants for AMP, ADP and ATP across all isoforms tested. The α2-complexes were ∼25-fold more sensitive than α1-complexes to dephosphorylation of a critical threonine on their activation loop (pThr(172/174)). However, α2-complexes were more readily activated by AMP than α1-complexes. Compared with ß1-containing heterotrimers, ß2-containing AMPK isoforms are less sensitive to activation by A769662, a synthetic activator. These data demonstrate that ligand induced activation of AMPK isoforms may vary significantly based on their AMPK subunit composition. Our studies provide insights for the design of isoform-selective AMPK activators for the treatment of metabolic diseases.


Subject(s)
AMP-Activated Protein Kinases/chemistry , Adenosine Monophosphate/chemistry , Allosteric Regulation , Biphenyl Compounds , Enzyme Activation , Enzyme Activators/chemistry , Enzyme Assays , Humans , Isoenzymes/chemistry , Kinetics , Protein Structure, Tertiary , Protein Subunits/chemistry , Pyrones/chemistry , Recombinant Proteins/chemistry , Thiophenes/chemistry
8.
Protein Expr Purif ; 110: 22-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25620107

ABSTRACT

Lysosomal acid lipase (LAL) is a serine hydrolase which hydrolyzes cholesteryl ester and triglycerides delivered to the lysosomes into free cholesterol and free fatty acids. Mutations in the LAL gene (LIPA) result in accumulation of triglycerides and cholesterol esters in various tissues of the body, leading to pathological conditions such as Wolman's disease (WD) and cholesteryl ester storage disease (CESD). CESD patients homozygous for His295Tyr (H295Y) mutation have less than 5% of normal LAL activity. To shed light on the molecular basis for this loss-of-function phenotype, we have generated the recombinant H295Y enzyme and studied its biophysical and biochemical properties. No significant differences were observed in the expression levels or glycosylation patterns between the mutant and the wild type LAL. However, the H295Y mutant displayed only residual enzymatic activity (<5%) compared to the wild type. While wild type LAL is mostly a monomer at pH 5.0, the vast majority H295Y exists as a high molecular soluble aggregate. Besides, the H295Y mutant has a 20°C lower melting temperature compared to the wild type. Transient expression studies in WD fibroblasts showed that mutation of His295 to other amino acids resulted in a significant loss of enzymatic activity. A homology model of LAL revealed that His295 is located on an α-helix of the cap domain and could be important for tethering it to its core domain. The observed loss-of-function phenotype in CESD patients might arise from a combination of protein destabilization and the shift to a non-functional soluble aggregate.


Subject(s)
Lysosomes/enzymology , Sterol Esterase/genetics , Wolman Disease/enzymology , Amino Acid Sequence , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Cholesterol Esters/chemistry , Cholesterol Esters/metabolism , Cloning, Molecular , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression , Glycosylation , Humans , Kinetics , Lipid Metabolism , Lysosomes/pathology , Models, Molecular , Molecular Sequence Data , Mutation , Plasmids/chemistry , Plasmids/metabolism , Protein Aggregates , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Sf9 Cells , Spodoptera , Sterol Esterase/isolation & purification , Sterol Esterase/metabolism , Wolman Disease/genetics , Wolman Disease/pathology
9.
Structure ; 22(8): 1161-1172, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25066137

ABSTRACT

AMP-activated protein kinase (AMPK) is a principal metabolic regulator affecting growth and response to cellular stress. Comprised of catalytic and regulatory subunits, each present in multiple forms, AMPK is best described as a family of related enzymes. In recent years, AMPK has emerged as a desirable target for modulation of numerous diseases, yet clinical therapies remain elusive. Challenges result, in part, from an incomplete understanding of the structure and function of full-length heterotrimeric complexes. In this work, we provide the full-length structure of the widely expressed α1ß1γ1 isoform of mammalian AMPK, along with detailed kinetic and biophysical characterization. We characterize binding of the broadly studied synthetic activator A769662 and its analogs. Our studies follow on the heels of the recent disclosure of the α2ß1γ1 structure and provide insight into the distinct molecular mechanisms of AMPK regulation by AMP and A769662.


Subject(s)
AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/physiology , Enzyme Activation/physiology , Models, Molecular , AMP-Activated Protein Kinases/metabolism , Adenosine Monophosphate/metabolism , Allosteric Site/genetics , Biphenyl Compounds , Drug Delivery Systems , Humans , Kinetics , Ligands , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Isoforms/physiology , Pyrones/metabolism , Structure-Activity Relationship , Surface Plasmon Resonance , Thiophenes/metabolism
10.
Nat Chem Biol ; 10(8): 629-31, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24997604

ABSTRACT

We report that 4-(3-(benzyloxy)phenyl)-2-ethylsulfinyl-6-(trifluoromethyl)pyrimidine (BETP), which behaves as a positive allosteric modulator at the glucagon-like peptide-1 receptor (GLP-1R), covalently modifies cysteines 347 and 438 in GLP-1R. C347, located in intracellular loop 3 of GLP-1R, is critical to the activity of BETP and a structurally distinct GLP-1R ago-allosteric modulator, N-(tert-butyl)-6,7-dichloro-3-(methylsulfonyl)quinoxalin-2-amine. We further show that substitution of cysteine for phenylalanine 345 in the glucagon receptor is sufficient to confer sensitivity to BETP.


Subject(s)
Pyrimidines/chemistry , Receptors, Glucagon/metabolism , Animals , CHO Cells , Cricetulus , Cysteine/chemistry , Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide-1 Receptor , Humans , Ligands , Pyrimidines/metabolism , Receptors, Glucagon/chemistry
11.
Structure ; 21(11): 1942-53, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24076403

ABSTRACT

AMP-activated protein kinase (AMPK) monitors cellular energy, regulates genes involved in ATP synthesis and consumption, and is allosterically activated by nucleotides and synthetic ligands. Analysis of the intact enzyme with hydrogen/deuterium exchange mass spectrometry reveals conformational perturbations of AMPK in response to binding of nucleotides, cyclodextrin, and a synthetic small molecule activator, A769662. Results from this analysis clearly show that binding of AMP leads to conformational changes primarily in the γ subunit of AMPK and subtle changes in the α and ß subunits. In contrast, A769662 causes profound conformational changes in the glycogen binding module of the ß subunit and in the kinase domain of the α subunit, suggesting that the molecular binding site of the latter resides between the α and ß subunits. The distinct short- and long-range perturbations induced upon binding of AMP and A769662 suggest fundamentally different molecular mechanisms for activation of AMPK by these two ligands.


Subject(s)
AMP-Activated Protein Kinases/chemistry , Allosteric Regulation , Biphenyl Compounds , Catalytic Domain , Deuterium Exchange Measurement , Enzyme Activation , Enzyme Activators/chemistry , Humans , Models, Molecular , Protein Binding , Protein Structure, Secondary , Pyrones/chemistry , Thiophenes/chemistry
12.
Nat Commun ; 4: 1888, 2013.
Article in English | MEDLINE | ID: mdl-23695682

ABSTRACT

The constituent polypeptides of the interleukin-17 family form six different homodimeric cytokines (IL-17A-F) and the heterodimeric IL-17A/F. Their interactions with IL-17 receptors A-E (IL-17RA-E) mediate host defenses while also contributing to inflammatory and autoimmune responses. IL-17A and IL-17F both preferentially engage a receptor complex containing one molecule of IL-17RA and one molecule of IL-17RC. More generally, IL-17RA appears to be a shared receptor that pairs with other members of its family to allow signaling of different IL-17 cytokines. Here we report crystal structures of homodimeric IL-17A and its complex with IL-17RA. Binding to IL-17RA at one side of the IL-17A molecule induces a conformational change in the second, symmetry-related receptor site of IL-17A. This change favors, and is sufficient to account for, the selection of a different receptor polypeptide to complete the cytokine-receptor complex. The structural results are supported by biophysical studies with IL-17A variants produced by site-directed mutagenesis.


Subject(s)
Interleukin-17/chemistry , Receptors, Interleukin-17/chemistry , Allosteric Regulation , Amino Acid Sequence , Conserved Sequence , Crystallization , Crystallography, X-Ray , HEK293 Cells , Humans , Interleukin-17/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Interleukin-17/metabolism , Surface Plasmon Resonance
13.
Protein Expr Purif ; 87(1): 27-34, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23069765

ABSTRACT

The T helper cell-derived cytokine interleukin-17A (IL-17A) is a variably glycosylated disulfide-linked homodimer of 34-38 kDa. Its polypeptide monomer contains one canonical N-glycosylation site at Asn68, and human recombinant IL-17A was partly N-glycosylated when expressed in human kidney (HEK293) cells as a fusion protein with a melittin signal sequence and an N-terminal hexahistidine tag. Orbitrap mass analyses of the tryptic N-glycopeptide 63-69 indicated that the N-glycosylation was of the GalNAc-terminated type characteristic of cultured kidney cells. The mass spectrum of IL-17A monomer also included peaks shifted by +948 Da from the respective masses of unglycosylated and N-glycosylated polypeptides. These were caused by unpredicted partial O-glycosylation of Thr26 with the mucin-like structure -GalNAc(-NeuNAc)-Gal-NeuNAc. Identical O-glycosylation occurred in commercially sourced recombinant IL-17A also expressed in HEK293 cells but with a different N-terminal sequence. Therefore, the kidney host cell line not only imposed its characteristic pattern of N-glycosylation on recombinant IL-17A but additionally created an O-glycosylation not known to be present in the T cell-derived cytokine. Mammalian host cell lines for recombinant protein expression generally impose their characteristic patterns of N-glycosylation on the product, but this work exemplifies how a host may also unpredictably O-glycosylate a protein that is probably not normally O-glycosylated.


Subject(s)
Interleukin-17/biosynthesis , Protein Processing, Post-Translational , Recombinant Fusion Proteins/biosynthesis , Amino Acid Sequence , CD4-Positive T-Lymphocytes/metabolism , Carbohydrate Conformation , Carbohydrate Sequence , Glycosylation , HEK293 Cells , Humans , Insect Proteins/biosynthesis , Insect Proteins/chemistry , Interleukin-17/chemistry , Melitten/biosynthesis , Melitten/chemistry , Molecular Sequence Data , Molecular Weight , Peptide Mapping , Protein Sorting Signals , Recombinant Fusion Proteins/chemistry , Tandem Mass Spectrometry
14.
Protein Expr Purif ; 73(2): 189-97, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20451617

ABSTRACT

AMP-activated protein kinase (AMPK) is an energy-sensing serine/threonine protein kinase that plays a central role in whole-body energy homeostasis. AMPK is a heterotrimeric enzyme with a catalytic (alpha) subunit and two regulatory (beta and gamma) subunits. The muscle-specific AMPK heterotrimeric complex (alpha2beta2gamma3) is involved in glucose and fat metabolism in skeletal muscle and therefore has emerged as an attractive target for drug development for diabetes and metabolic syndrome. To date, expression of recombinant full-length human AMPK alpha2beta2gamma3 has not been reported. Here we describe the expression, purification and biochemical characterization of functional full-length AMPK alpha2beta2gamma3 heterotrimeric complex using an Escherichia coli expression system. All three subunits of AMPK alpha2beta2gamma3 were transcribed as a single tricistronic transcript driven by the T7 RNA polymerase promoter, allowing spontaneous formation of the heterotrimeric complex in the bacterial cytosol. The self-assembled trimeric complex was purified from the cell lysate by nickel-ion chromatography using the hexahistidine tag fused exclusively at the N-terminus of the alpha 2 domain. The un-assembled beta 2 and gamma 3 domains were removed by extensive washing of the column. Further purification of the heterotrimer was performed using size exclusion chromatography. The final yield of the recombinant AMPK alpha2beta2gamma3 complex was 1.1mg/L culture in shaker flasks. The E. coli expressed enzyme was catalytically inactive after purification, but was activated in vitro by upstream kinases such as CaMKKbeta and LKB1. The kinase activity of activated AMPK alpha2beta2gamma3 complex was significantly enhanced by AMP (an allosteric activator) but not by thienopyridone A-769662, a known small molecule activator of AMPK. Mass spectrometric characterization of recombinant AMPK alpha2beta2gamma3 showed significant heterogeneity before and after activation that could potentially hamper crystallographic studies of this complex.


Subject(s)
AMP-Activated Protein Kinases/physiology , Escherichia coli/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adenosine Monophosphate/metabolism , Biphenyl Compounds , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/physiology , Catalytic Domain , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit/genetics , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit/metabolism , Enzyme Activation/drug effects , Escherichia coli/genetics , Homeostasis , Humans , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Protein Subunits/genetics , Protein Subunits/metabolism , Pyrones/pharmacology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Thiophenes/pharmacology
15.
Biochemistry ; 48(13): 2941-9, 2009 Apr 07.
Article in English | MEDLINE | ID: mdl-19222187

ABSTRACT

Proprotein convertase subtilisin-kexin type 9 (PCSK9) binds to the low-density lipoprotein receptor (LDLR) on target cells and lowers the level of receptor by impeding its recycling. PCSK9 is self-processed to a complex of its prodomain and catalytic domain like a typical protein convertase, but it does not develop normal proteolytic activity. Instead, its propeptide remains complexed with the catalytic domain, and the C-terminal Gln152 of the prodomain occupies the active site like a substrate for peptide synthesis. To probe its latent catalytic activity, PCSK9 and its complex with the soluble LDLR extracellular domain were separately transferred into H218O, and time point samples were analyzed by peptide mapping with mass spectrometry to measure the rate and extent of incorporation of 18O into the Gln152 carboxylate. In free wild-type or D374Y mutant PCSK9, the t1/2 for exchange of 18O for both oxygens was near 5 min. This slow process progressed to completion, with the distribution of oxygen isotopes in the Gln152 carboxylate finally matching that in solvent. In contrast, exchange reached its final state in <30 s in LDLR-complexed D374Y mutant PCSK9, but approximately 40% of the molecules gave data indicating the presence of only one 18O atom in Gln152. With support from further experiments, this was attributed to hydrolysis of acylenzyme in H216O during preparations for digestion and indicated that PCSK9 complexed with LDLR contains approximately 40% intramolecular acylenzyme at equilibrium. The synthetic EGF-A domain of LDLR induced similar effects as the full-length receptor. The data suggest the existence of distinct conformational states in free and receptor-bound PCSK9.


Subject(s)
Biocatalysis , Receptors, LDL/metabolism , Serine Endopeptidases/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Glutamine/metabolism , Humans , Isotopes , Mass Spectrometry , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Peptides/chemistry , Peptides/metabolism , Proprotein Convertase 9 , Proprotein Convertases , Serine Endopeptidases/chemistry , Substrate Cycling
16.
Bioconjug Chem ; 19(8): 1604-13, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18646836

ABSTRACT

Cholesteryl ester transfer protein (CETP) transfers neutral lipids between different types of plasma lipoprotein. Inhibitors of CETP elevate the fraction of plasma cholesterol associated with high-density lipoproteins and are being developed as new agents for the prevention and treatment of cardiovascular disease. The molecular basis of their function is not yet fully understood. To aid in the study of inhibitor interactions with CETP, a torcetrapib-related compound was coupled to different biotin-terminated spacer groups, and the binding of CETP to the streptavidin-bound conjugates was monitored on agarose beads and in a surface plasmon resonance biosensor. CETP binding was poor with a 2.0 nm spacer arm, but efficient with polyethyleneglycol spacers of 3.5 or 4.6 nm. The conjugate based on a 4.6 nm spacer was used for further biosensor experiments. Soluble inhibitor blocked the binding of CETP to the immobilized drug, as did preincubation with a disulfide-containing covalent inhibitor. To provide a first estimate of the binding site for torcetrapib-like inhibitors, CETP was modified with a disulfide-containing agent that modifies Cys-13 of CETP. Mass spectrometry of the modified protein indicated that a single half-molecule of the disulfide was covalently bound to CETP, and peptide mapping after digestion with pepsin confirmed previous reports based on mutagenesis that Cys-13 was the site of modification. Modified CETP was unable to bind to the biosensor-mounted torcetrapib analog, indicating that the binding site on CETP for torcetrapib is in the lipid-binding pocket near the N-terminus of the protein. The crystal structure of CETP shows that the sulfhydryl group of Cys-13 resides at the bottom of this pocket.


Subject(s)
Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cholesterol Ester Transfer Proteins/metabolism , Surface Plasmon Resonance/methods , Affinity Labels/chemistry , Affinity Labels/metabolism , Binding Sites , Binding, Competitive , Biotin/metabolism , Cholesterol Ester Transfer Proteins/chemistry , Cholesterol Ester Transfer Proteins/genetics , Ligands , Mutagenesis , Protein Binding , Quinolines/chemistry , Quinolines/metabolism
17.
Nat Struct Mol Biol ; 14(5): 413-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17435765

ABSTRACT

Proprotein convertase subtilisin kexin type 9 (PCSK9) lowers the abundance of surface low-density lipoprotein (LDL) receptor through an undefined mechanism. The structure of human PCSK9 shows the subtilisin-like catalytic site blocked by the prodomain in a noncovalent complex and inaccessible to exogenous ligands, and that the C-terminal domain has a novel fold. Biosensor studies show that PCSK9 binds the extracellular domain of LDL receptor with K(d) = 170 nM at the neutral pH of plasma, but with a K(d) as low as 1 nM at the acidic pH of endosomes. The D374Y gain-of-function mutant, associated with hypercholesterolemia and early-onset cardiovascular disease, binds the receptor 25 times more tightly than wild-type PCSK9 at neutral pH and remains exclusively in a high-affinity complex at the acidic pH. PCSK9 may diminish LDL receptors by a mechanism that requires direct binding but not necessarily receptor proteolysis.


Subject(s)
Hypercholesterolemia/genetics , Mutation, Missense/physiology , Serine Endopeptidases/metabolism , Binding Sites , Humans , Hydrogen-Ion Concentration , Hypercholesterolemia/etiology , Proprotein Convertase 9 , Proprotein Convertases , Protein Binding/genetics , Protein Conformation , Receptors, LDL/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics
18.
Protein Sci ; 16(5): 897-905, 2007 May.
Article in English | MEDLINE | ID: mdl-17456742

ABSTRACT

Lasofoxifene is a new and potent selective estrogen receptor modulator (SERM). The structural basis of its interaction with the estrogen receptor has been investigated by crystallographic analysis of its complex with the ligand-binding domain of estrogen receptor alpha at a resolution of 2.0 A. As with other SERMs, lasofoxifene diverts the receptor from its agonist-bound conformation by displacing the C-terminal AF-2 helix into the site at which the LXXLL motif of coactivator proteins would otherwise be able to bind. Lasofoxifene achieves this effect by occupying the space normally filled by residue Leu 540, as well as by modulating the conformation of residues of helix 11 (His 524, Leu 525). A well-defined salt bridge between lasofoxifene and Asp 351 suggests that charge neutralization in this region of the receptor may explain the some of the antiestrogenic effects of lasofoxifene. The results suggest general features of ERalpha/SERM recognition, and add a new dimension to efforts to rationalize differences between the biological activity profiles exhibited by these important pharmacological agents.


Subject(s)
Estrogen Receptor alpha/chemistry , Pyrrolidines/chemistry , Tetrahydronaphthalenes/chemistry , Crystallography, X-Ray , Estrogen Receptor alpha/metabolism , Hydrogen Bonding , Molecular Structure , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Pyrrolidines/metabolism , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/metabolism , Tetrahydronaphthalenes/metabolism
19.
Nat Struct Mol Biol ; 14(2): 106-13, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17237796

ABSTRACT

Cholesteryl ester transfer protein (CETP) shuttles various lipids between lipoproteins, resulting in the net transfer of cholesteryl esters from atheroprotective, high-density lipoproteins (HDL) to atherogenic, lower-density species. Inhibition of CETP raises HDL cholesterol and may potentially be used to treat cardiovascular disease. Here we describe the structure of CETP at 2.2-A resolution, revealing a 60-A-long tunnel filled with two hydrophobic cholesteryl esters and plugged by an amphiphilic phosphatidylcholine at each end. The two tunnel openings are large enough to allow lipid access, which is aided by a flexible helix and possibly also by a mobile flap. The curvature of the concave surface of CETP matches the radius of curvature of HDL particles, and potential conformational changes may occur to accommodate larger lipoprotein particles. Point mutations blocking the middle of the tunnel abolish lipid-transfer activities, suggesting that neutral lipids pass through this continuous tunnel.


Subject(s)
Cholesterol Ester Transfer Proteins/chemistry , Cholesterol Esters/chemistry , Models, Molecular , Phosphatidylcholines/chemistry , Triglycerides/chemistry , Animals , Binding Sites , CHO Cells , Cholesterol Ester Transfer Proteins/genetics , Cricetinae , Cricetulus , Crystallography, X-Ray , Humans , Hydrophobic and Hydrophilic Interactions , Ligands , Point Mutation , Protein Binding , Protein Conformation
20.
Protein Expr Purif ; 52(2): 313-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17169570

ABSTRACT

The membrane-anchored metalloproteinase ADAM17 (TNF-alpha converting enzyme; TACE; EC 3.4.24.86) continues to be an attractive drug target in inflammatory diseases and cancer. Cocrystallization of its catalytic domain with a lead compound was complicated by the tenacious retention of the prodomain that has been shown to be enhanced if ADAM17 is expressed without the disintegrin/cysteine-rich domain that normally follows the N-terminal metalloproteinase. When a truncated form of ADAM17 composed of the signal peptide with the pro- and catalytic domains was expressed in baculovirus-infected insect cells, the major secreted product was a ternary complex of two prodomain fragments with the catalytic domain. The component polypeptides of the ternary complex were characterized by N-terminal analysis and mass spectrometry. Internal cleavage of the propeptide occurred following Arg-58, and a carboxypeptidase variably removed up to three basic residues from the newly created C-terminus. Cleavage at the C-terminus of the propeptide occurred after Arg-214. To prepare ADAM17 for crystal growth, a drug-like inhibitor was used to displace the propeptide and the complex of the catalytic domain with the inhibitor was isolated by size-exclusion chromatography and crystallized.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Catalytic Domain , Hydroxamic Acids/pharmacology , ADAM Proteins/chemistry , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM17 Protein , Chromatography, Gel , Chromatography, High Pressure Liquid , Crystallization , Enzyme Inhibitors/pharmacology , Gene Expression , Humans , Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...