Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Virol ; 97(12): e0105223, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38032197

ABSTRACT

IMPORTANCE: Human metapneumovirus (hMPV) is a common pathogen causing lower respiratory tract infections worldwide and can develop severe symptoms in high-risk populations such as infants, the elderly, and immunocompromised patients. There are no approved hMPV vaccines or neutralizing antibodies available for therapeutic or prophylactic use. The trimeric hMPV fusion F protein is the major target of neutralizing antibodies in human sera. Understanding the immune recognition of antibodies to hMPV-F antigen will provide critical insights into developing efficacious hMPV monoclonal antibodies and vaccines.


Subject(s)
Metapneumovirus , Paramyxoviridae Infections , Aged , Humans , Antibodies, Neutralizing , Antibodies, Viral , Epitopes , Metapneumovirus/physiology , Paramyxoviridae Infections/immunology , Viral Fusion Proteins , Viral Vaccines/immunology
2.
Protein Expr Purif ; 179: 105796, 2021 03.
Article in English | MEDLINE | ID: mdl-33221505

ABSTRACT

TREM2 has been identified by genomic analysis as a potential and novel target for the treatment of Alzheimer's disease. To enable structure-based screening of potential small molecule therapeutics, we sought to develop a robust crystallization platform for the TREM2 Ig-like domain. A systematic set of constructs containing the structural chaperone, maltose binding protein (MBP), fused to the Ig domain of TREM2, were evaluated in parallel expression and purification, followed by crystallization studies. Using protein crystallization and high-resolution diffraction as a readout, a MBP-TREM2 Ig fusion construct was identified that generates reproducible protein crystals diffracting at 2.0 Å, which makes it suitable for soaking of potential ligands. Importantly, analysis of crystal packing interfaces indicates that most of the surface of the TREM2 Ig domain is available for small molecule binding. A proof of concept co-crystallization study with a small library of fragments validated potential utility of this system for the discovery of new TREM2 therapeutics.


Subject(s)
Crystallization/methods , Membrane Glycoproteins , Molecular Chaperones , Receptors, Immunologic , Recombinant Fusion Proteins , Humans , Maltose-Binding Proteins/chemistry , Maltose-Binding Proteins/genetics , Maltose-Binding Proteins/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
3.
Nat Commun ; 10(1): 4153, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31515478

ABSTRACT

Respiratory syncytial virus (RSV) infection is the leading cause of hospitalization and infant mortality under six months of age worldwide; therefore, the prevention of RSV infection in all infants represents a significant unmet medical need. Here we report the isolation of a potent and broadly neutralizing RSV monoclonal antibody derived from a human memory B-cell. This antibody, RB1, is equipotent on RSV A and B subtypes, potently neutralizes a diverse panel of clinical isolates in vitro and demonstrates in vivo protection. It binds to a highly conserved epitope in antigenic site IV of the RSV fusion glycoprotein. RB1 is the parental antibody to MK-1654 which is currently in clinical development for the prevention of RSV infection in infants.


Subject(s)
Antibodies, Viral/immunology , Broadly Neutralizing Antibodies/immunology , Conserved Sequence , Glycoproteins/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/immunology , Animals , Antibodies, Monoclonal/isolation & purification , B-Lymphocytes/immunology , Binding Sites , Disease Models, Animal , Epitopes/immunology , Female , Humans , Immunologic Memory , Models, Molecular , Protein Binding , Sigmodontinae
4.
Bioorg Med Chem Lett ; 28(6): 1122-1126, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29534798

ABSTRACT

An internal HTS effort identified a novel PDE2 inhibitor series that was subsequently optimized for improved PDE2 activity and off-target selectivity. The optimized lead, compound 4, improved cognitive performance in a rodent novel object recognition task as well as a non-human primate object retrieval task. In addition, co-crystallization studies of close analog of 4 in the PDE2 active site revealed unique binding interactions influencing the high PDE isoform selectivity.


Subject(s)
Acetic Acid/pharmacology , Cognitive Dysfunction/drug therapy , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Indoles/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Acetic Acid/chemical synthesis , Acetic Acid/chemistry , Animals , Catalytic Domain/drug effects , Cognitive Dysfunction/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Dose-Response Relationship, Drug , Indoles/chemical synthesis , Indoles/chemistry , Molecular Structure , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/chemistry , Rats , Structure-Activity Relationship
5.
J Biol Chem ; 290(33): 20360-73, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26134571

ABSTRACT

G-protein-coupled receptor (GPCR) kinases (GRKs) bind to and phosphorylate GPCRs, initiating the process of GPCR desensitization and internalization. GRK4 is implicated in the regulation of blood pressure, and three GRK4 polymorphisms (R65L, A142V, and A486V) are associated with hypertension. Here, we describe the 2.6 Å structure of human GRK4α A486V crystallized in the presence of 5'-adenylyl ß,γ-imidodiphosphate. The structure of GRK4α is similar to other GRKs, although slight differences exist within the RGS homology (RH) bundle subdomain, substrate-binding site, and kinase C-tail. The RH bundle subdomain and kinase C-terminal lobe form a strikingly acidic surface, whereas the kinase N-terminal lobe and RH terminal subdomain surfaces are much more basic. In this respect, GRK4α is more similar to GRK2 than GRK6. A fully ordered kinase C-tail reveals interactions linking the C-tail with important determinants of kinase activity, including the αB helix, αD helix, and the P-loop. Autophosphorylation of wild-type GRK4α is required for full kinase activity, as indicated by a lag in phosphorylation of a peptide from the dopamine D1 receptor without ATP preincubation. In contrast, this lag is not observed in GRK4α A486V. Phosphopeptide mapping by mass spectrometry indicates an increased rate of autophosphorylation of a number of residues in GRK4α A486V relative to wild-type GRK4α, including Ser-485 in the kinase C-tail.


Subject(s)
G-Protein-Coupled Receptor Kinase 4/chemistry , G-Protein-Coupled Receptor Kinase 4/metabolism , Hypertension/genetics , Amino Acid Sequence , Crystallography, X-Ray , G-Protein-Coupled Receptor Kinase 4/genetics , Humans , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Conformation , Sequence Homology, Amino Acid , Substrate Specificity
6.
J Biol Chem ; 288(47): 34073-34080, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24108127

ABSTRACT

The emergence of antibiotic-resistant strains of pathogenic bacteria is an increasing threat to global health that underscores an urgent need for an expanded antibacterial armamentarium. Gram-negative bacteria, such as Escherichia coli, have become increasingly important clinical pathogens with limited treatment options. This is due in part to their lipopolysaccharide (LPS) outer membrane components, which dually serve as endotoxins while also protecting Gram-negative bacteria from antibiotic entry. The LpxC enzyme catalyzes the committed step of LPS biosynthesis, making LpxC a promising target for new antibacterials. Here, we present the first structure of an LpxC enzyme in complex with the deacetylation reaction product, UDP-(3-O-(R-3-hydroxymyristoyl))-glucosamine. These studies provide valuable insight into recognition of substrates and products by LpxC and a platform for structure-guided drug discovery of broad spectrum Gram-negative antibiotics.


Subject(s)
Amidohydrolases/chemistry , Escherichia coli/enzymology , Myristic Acids/chemistry , Protons , Uridine Diphosphate N-Acetylglucosamine/analogs & derivatives , Amidohydrolases/metabolism , Crystallography, X-Ray , Lipopolysaccharides/biosynthesis , Lipopolysaccharides/chemistry , Myristic Acids/metabolism , Protein Structure, Tertiary , Uridine Diphosphate N-Acetylglucosamine/chemistry , Uridine Diphosphate N-Acetylglucosamine/metabolism
7.
J Am Chem Soc ; 134(30): 12342-5, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22793495

ABSTRACT

The cooperative assembly of FtsZ, the prokaryotic homologue of tubulin, plays an essential role in cell division. FtsZ is a potential drug target, as illustrated by the small-molecule cell-cycle inhibitor and antibacterial agent PC190723 that targets FtsZ. We demonstrate that PC190723 negatively modulates Staphylococcus aureus FtsZ polymerization cooperativity as reflected in polymerization at lower concentrations without a defined critical concentration. The crystal structure of the S. aureus FtsZ-PC190723 complex shows a domain movement that would stabilize the FtsZ protofilament over the monomeric state, with the conformational change mediated from the GTP-binding site to the C-terminal domain via helix 7. Together, the results reveal the molecular mechanism of FtsZ modulation by PC190723 and a conformational switch to the high-affinity state that enables polymer assembly.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Cytoskeletal Proteins/metabolism , Protein Conformation/drug effects , Pyridines/pharmacology , Staphylococcus aureus/drug effects , Thiazoles/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/chemistry , Humans , Molecular Docking Simulation , Protein Structure, Tertiary/drug effects , Staphylococcal Infections/drug therapy , Staphylococcus aureus/chemistry , Staphylococcus aureus/metabolism
8.
Sci Transl Med ; 4(126): 126ra35, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22440737

ABSTRACT

Despite the need for new antibiotics to treat drug-resistant bacteria, current clinical combinations are largely restricted to ß-lactam antibiotics paired with ß-lactamase inhibitors. We have adapted a Staphylococcus aureus antisense knockdown strategy to genetically identify the cell division Z ring components-FtsA, FtsZ, and FtsW-as ß-lactam susceptibility determinants of methicillin-resistant S. aureus (MRSA). We demonstrate that the FtsZ-specific inhibitor PC190723 acts synergistically with ß-lactam antibiotics in vitro and in vivo and that this combination is efficacious in a murine model of MRSA infection. Fluorescence microscopy localization studies reveal that synergy between these agents is likely to be elicited by the concomitant delocalization of their cognate drug targets (FtsZ and PBP2) in MRSA treated with PC190723. A 2.0 Å crystal structure of S. aureus FtsZ in complex with PC190723 identifies the compound binding site, which corresponds to the predominant location of mutations conferring resistance to PC190723 (PC190723(R)). Although structural studies suggested that these drug resistance mutations may be difficult to combat through chemical modification of PC190723, combining PC190723 with the ß-lactam antibiotic imipenem markedly reduced the spontaneous frequency of PC190723(R) mutants. Multiple MRSA PC190723(R) FtsZ mutants also displayed attenuated virulence and restored susceptibility to ß-lactam antibiotics in vitro and in a mouse model of imipenem efficacy. Collectively, these data support a target-based approach to rationally develop synergistic combination agents that mitigate drug resistance and effectively treat MRSA infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , beta-Lactams/pharmacology , Animals , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Division/drug effects , Crystallography, X-Ray , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Drug Resistance, Bacterial/drug effects , Drug Synergism , Gene Regulatory Networks/genetics , Guanosine Diphosphate , Imipenem/pharmacology , Methicillin-Resistant Staphylococcus aureus/cytology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Microbial Sensitivity Tests , Mutation/genetics , Protein Structure, Secondary , Protein Transport/drug effects , Pyridines/chemistry , Pyridines/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Thiazoles/chemistry , Thiazoles/pharmacology , Virulence/drug effects , beta-Lactams/therapeutic use
9.
J Biol Chem ; 286(13): 11218-25, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21247903

ABSTRACT

The receptor tyrosine kinase c-Met is implicated in oncogenesis and is the target for several small molecule and biologic agents in clinical trials for the treatment of cancer. Binding of the hepatocyte growth factor to the cell surface receptor of c-Met induces activation via autophosphorylation of the kinase domain. Here we describe the structural basis of c-Met activation upon autophosphorylation and the selective small molecule inhibiton of autophosphorylated c-Met. MK-2461 is a potent c-Met inhibitor that is selective for the phosphorylated state of the enzyme. Compound 1 is an MK-2461 analog with a 20-fold enthalpy-driven preference for the autophosphorylated over unphosphorylated c-Met kinase domain. The crystal structure of the unbound kinase domain phosphorylated at Tyr-1234 and Tyr-1235 shows that activation loop phosphorylation leads to the ejection and disorder of the activation loop and rearrangement of helix αC and the G loop to generate a viable active site. Helix αC adopts a orientation different from that seen in activation loop mutants. The crystal structure of the complex formed by the autophosphorylated c-Met kinase domain and compound 1 reveals a significant induced fit conformational change of the G loop and ordering of the activation loop, explaining the selectivity of compound 1 for the autophosphorylated state. The results highlight the role of structural plasticity within the kinase domain in imparting the specificity of ligand binding and provide the framework for structure-guided design of activated c-Met inhibitors.


Subject(s)
Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/chemistry , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/chemistry , Animals , Cell Line , Crystallography, X-Ray , Drug Design , Humans , Phosphorylation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Spodoptera , Structure-Activity Relationship , c-Mer Tyrosine Kinase
10.
BMC Struct Biol ; 10: 16, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20540760

ABSTRACT

BACKGROUND: The unique S28 family of proteases is comprised of the carboxypeptidase PRCP and the aminopeptidase DPP7. The structural basis of the different substrate specificities of the two enzymes is not understood nor has the structure of the S28 fold been described. RESULTS: The experimentally phased 2.8 A crystal structure is presented for human PRCP. PRCP contains an alpha/beta hydrolase domain harboring the catalytic Asp-His-Ser triad and a novel helical structural domain that caps the active site. Structural comparisons with prolylendopeptidase and DPP4 identify the S1 proline binding site of PRCP. A structure-based alignment with the previously undescribed structure of DPP7 illuminates the mechanism of orthogonal substrate specificity of PRCP and DPP7. PRCP has an extended active-site cleft that can accommodate proline substrates with multiple N-terminal residues. In contrast, the substrate binding groove of DPP7 is occluded by a short amino-acid insertion unique to DPP7 that creates a truncated active site selective for dipeptidyl proteolysis of N-terminal substrates. CONCLUSION: The results define the structure of the S28 family of proteases, provide the structural basis of PRCP and DPP7 substrate specificity and enable the rational design of selective PRCP modulators.


Subject(s)
Carboxypeptidases/chemistry , Amino Acid Sequence , Binding Sites , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Catalytic Domain , Crystallography, X-Ray , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
11.
Article in English | MEDLINE | ID: mdl-20516604

ABSTRACT

Prolylcarboxypeptidase (PrCP) is a lysosomal serine carboxypeptidase that cleaves a variety of C-terminal amino acids adjacent to proline and has been implicated in diseases such as hypertension and obesity. Here, the robust production, purification and crystallization of glycosylated human PrCP from stably transformed CHO cells is described. Purified PrCP yielded crystals belonging to space group R32, with unit-cell parameters a = b = 181.14, c = 240.13 A, that diffracted to better than 2.8 A resolution.


Subject(s)
Carboxypeptidases/chemistry , Animals , CHO Cells , Carboxypeptidases/genetics , Carboxypeptidases/isolation & purification , Cricetinae , Cricetulus , Crystallization , Crystallography, X-Ray , Gene Expression , Glycosylation , Humans
12.
J Virol ; 84(15): 7625-33, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20484498

ABSTRACT

HIV/AIDS continues to be a menace to public health. Several drugs currently on the market have successfully improved the ability to manage the viral burden in infected patients. However, new drugs are needed to combat the rapid emergence of mutated forms of the virus that are resistant to existing therapies. Currently, approved drugs target three of the four major enzyme activities encoded by the virus that are critical to the HIV life cycle. Although a number of inhibitors of HIV RNase H activity have been reported, few inhibit by directly engaging the RNase H active site. Here, we describe structures of naphthyridinone-containing inhibitors bound to the RNase H active site. This class of compounds binds to the active site via two metal ions that are coordinated by catalytic site residues, D443, E478, D498, and D549. The directionality of the naphthyridinone pharmacophore is restricted by the ordering of D549 and H539 in the RNase H domain. In addition, one of the naphthyridinone-based compounds was found to bind at a second site close to the polymerase active site and non-nucleoside/nucleotide inhibitor sites in a metal-independent manner. Further characterization, using fluorescence-based thermal denaturation and a crystal structure of the isolated RNase H domain reveals that this compound can also bind the RNase H site and retains the metal-dependent binding mode of this class of molecules. These structures provide a means for structurally guided design of novel RNase H inhibitors.


Subject(s)
Enzyme Inhibitors/metabolism , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/chemistry , HIV-1/drug effects , Naphthyridines/metabolism , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Binding Sites , Catalytic Domain , Cations/metabolism , Crystallography, X-Ray , HIV , HIV Reverse Transcriptase/metabolism , HIV-1/chemistry , Humans , Metals/metabolism , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Ribonuclease H, Human Immunodeficiency Virus/metabolism
13.
J Biol Chem ; 283(50): 34864-72, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-18922802

ABSTRACT

Prostasin (also called channel activating protease-1 (CAP1)) is an extracellular serine protease implicated in the modulation of fluid and electrolyte regulation via proteolysis of the epithelial sodium channel. Several disease states, particularly hypertension, can be affected by modulation of epithelial sodium channel activity. Thus, understanding the biochemical function of prostasin and developing specific agents to inhibit its activity could have a significant impact on a widespread disease. We report the expression of the prostasin proenzyme in Escherichia coli as insoluble inclusion bodies, refolding and activating via proteolytic removal of the N-terminal propeptide. The refolded and activated enzyme was shown to be pure and monomeric, with kinetic characteristics very similar to prostasin expressed from eukaryotic systems. Active prostasin was crystallized, and the structure was determined to 1.45 A resolution. These apoprotein crystals were soaked with nafamostat, allowing the structure of the inhibited acyl-enzyme intermediate structure to be determined to 2.0 A resolution. Comparison of the inhibited and apoprotein forms of prostasin suggest a mechanism of regulation through stabilization of a loop which interferes with substrate recognition.


Subject(s)
Hypertension/metabolism , Serine Endopeptidases/chemistry , Amino Acid Sequence , Apoproteins/chemistry , Benzamidines , Crystallography, X-Ray/methods , Escherichia coli/metabolism , Guanidines/chemistry , Humans , Molecular Conformation , Molecular Sequence Data , Protein Conformation , Protein Folding , Protein Renaturation , Sequence Homology, Amino Acid , Serine Endopeptidases/genetics , Substrate Specificity
14.
J Med Chem ; 51(20): 6503-11, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18826204

ABSTRACT

Non-nucleoside reverse transcriptase inhibitors (NNRTIs) have been shown to be a key component of highly active antiretroviral therapy (HAART). The use of NNRTIs has become part of standard combination antiviral therapies producing clinical outcomes with efficacy comparable to other antiviral regimens. There is, however, a critical issue with the emergence of clinical resistance, and a need has arisen for novel NNRTIs with a broad spectrum of activity against key HIV-1 RT mutations. Using a combination of traditional medicinal chemistry/SAR analyses, crystallography, and molecular modeling, we have designed and synthesized a series of novel, highly potent NNRTIs that possess broad spectrum antiviral activity and good pharmacokinetic profiles. Further refinement of key compounds in this series to optimize physical properties and pharmacokinetics has resulted in the identification of 8e (MK-4965), which has high levels of potency against wild-type and key mutant viruses, excellent oral bioavailability and overall pharmacokinetics, and a clean ancillary profile.


Subject(s)
HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , HIV-1/enzymology , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/pharmacology , Administration, Oral , Animals , Bromine Compounds/chemical synthesis , Bromine Compounds/chemistry , Crystallography, X-Ray , Drug Evaluation, Preclinical , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , HIV-1/genetics , Models, Molecular , Molecular Structure , Mutation/genetics , Nucleosides/chemistry , Nucleosides/pharmacology , Pyrazoles/chemistry , Pyridines/chemistry , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Inhibitors/chemistry , Structure-Activity Relationship
15.
Protein Sci ; 16(12): 2626-35, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17965187

ABSTRACT

The p90 ribosomal S6 kinases (RSKs) also known as MAPKAP-Ks are serine/threonine protein kinases that are activated by ERK or PDK1 and act as downstream effectors of mitogen-activated protein kinase (MAPK). RSK1, a member of the RSK family, contains two distinct kinase domains in a single polypeptide chain, the regulatory C-terminal kinase domain (CTKD) and the catalytic N-terminal kinase domain (NTKD). Autophosphorylation of the CTKD leads to activation of the NTKD that subsequently phosphorylates downstream substrates. Here we report the crystal structures of the unactivated RSK1 NTKD bound to different ligands at 2.0 A resolution. The activation loop and helix alphaC, key regulatory elements of kinase function, are disordered. The DFG motif of the inactive RSK1 adopts an "active-like" conformation. The beta-PO(4) group in the AMP-PCP complex adopts a unique conformation that may contribute to inactivity of the enzyme. Structures of RSK1 ligand complexes offer insights into the design of novel anticancer agents and into the regulation of the catalytic activity of RSKs.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Purines/chemistry , Ribosomal Protein S6 Kinases, 90-kDa/chemistry , Staurosporine/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Humans , Hydrophobic and Hydrophilic Interactions , Ligands , Models, Molecular , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Purines/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Staurosporine/metabolism
16.
Biochemistry ; 44(27): 9430-40, 2005 Jul 12.
Article in English | MEDLINE | ID: mdl-15996097

ABSTRACT

The type 1 insulin-like growth factor receptor (IGF-1R) is often overexpressed on tumor cells and is believed to play an important role in anchorage-independent proliferation. Additionally, cell culture studies have indicated that IGF-1R confers increased resistance to apoptosis caused by radiation or chemotherapeutic agents. Thus, inhibitors of the intracellular kinase domain of this receptor may have utility for the clinical treatment of cancer. As part of an effort to develop clinically useful inhibitors of IGF-1R kinase, a novel class of pyrrole-5-carboxaldehyde compounds was investigated. The compounds exhibited selectivity against the closely related insulin receptor kinase intrinsically and in cell-based assays. The inhibitors formed a reversible, covalent adduct at the kinase active site, and treatment of such adducts with sodium borohydride irreversibly inactivated the enzyme. Analysis of a tryptic digest of a covalently modified IGF-1R kinase fragment revealed that the active site Lys1003 had been reductively alkylated with the aldehyde inhibitor. Reductive alkylation of the insulin receptor kinase with one of these inhibitors led to a similarly inactivated enzyme which was examined by X-ray crystallography. The crystal structure confirmed the modification of the active site lysine side chain and revealed details of the key interactions between the inhibitor and enzyme.


Subject(s)
Aldehydes/chemistry , Protein Kinase Inhibitors/chemistry , Pyrroles/chemistry , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, Insulin/antagonists & inhibitors , Receptor, Insulin/chemistry , Aldehydes/metabolism , Amino Acid Sequence , Binding Sites , Borohydrides/chemistry , Cell Line , Crystallography, X-Ray , Enzyme Activation , Humans , Molecular Sequence Data , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Protein Kinase Inhibitors/metabolism , Protein Structure, Tertiary , Pyrroles/metabolism , Receptor, Insulin/metabolism , Schiff Bases/chemistry , Structure-Activity Relationship
17.
J Biol Chem ; 277(41): 38797-802, 2002 Oct 11.
Article in English | MEDLINE | ID: mdl-12138114

ABSTRACT

The x-ray structure of the unactivated kinase domain of insulin-like growth factor-1 receptor (IGFRK-0P) is reported here at 2.7 A resolution. IGFRK-0P is composed of two lobes connected by a hinge region. The N-terminal lobe of the kinase is a twisted beta-sheet flanked by a single helix, and the C-terminal lobe comprises eight alpha-helices and four short beta-strands. The ATP binding pocket and the catalytic center reside at the interface of the two lobes. Despite the overall similarity to other receptor tyrosine kinases, three notable conformational modifications are observed: 1) this kinase adopts a more closed structure, with its two lobes rotated further toward each other; 2) the conformation of the proximal end of the activation loop (residues 1121-1129) is different; 3) the orientation of the nucleotide-binding loop is altered. Collectively, these alterations lead to a different ATP-binding pocket that might impact on inhibitor designs for IGFRK-0P. Two molecules of IGFRK-0P are seen in the asymmetric unit; they are associated as a dimer with their ATP binding clefts facing each other. The ordered N terminus of one monomer approaches the active site of the other, suggesting that the juxtamembrane region of one molecule could come into close proximity to the active site of the other.


Subject(s)
Protein Structure, Tertiary , Receptor, IGF Type 1/chemistry , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Crystallography, X-Ray , Dimerization , Insecta/enzymology , Models, Molecular , Protein Structure, Quaternary , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...