Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Chemistry ; 29(23): e202203967, 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-36799129

ABSTRACT

The ephrin type-A receptor 2 (EPHA2) kinase belongs to the largest family of receptor tyrosine kinases. There are several indications of an involvement of EPHA2 in the development of infectious diseases and cancer. Despite pharmacological potential, EPHA2 is an under-examined target protein. In this study, we synthesized a series of derivatives of the inhibitor NVP-BHG712 and triazine-based compounds. These compounds were evaluated to determine their potential as kinase inhibitors of EPHA2, including elucidation of their binding mode (X-ray crystallography), affinity (microscale thermophoresis), and selectivity (Kinobeads assay). Eight inhibitors showed affinities in the low-nanomolar regime (KD <10 nM). Testing in up to seven colon cancer cell lines that express EPHA2 reveals that several derivatives feature promising effects for the control of human colon carcinoma. Thus, we have developed a set of powerful tool compounds for fundamental new research on the interplay of EPH receptors in a cellular context.


Subject(s)
Colorectal Neoplasms , Pyrazoles , Humans , Pyrazoles/chemistry , Pyrimidines/pharmacology , Pyrimidines/chemistry , Cell Line , Colorectal Neoplasms/drug therapy , Cell Line, Tumor
2.
Nat Commun ; 11(1): 5569, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33149120

ABSTRACT

Understanding the conformational sampling of translation-arrested ribosome nascent chain complexes is key to understand co-translational folding. Up to now, coupling of cysteine oxidation, disulfide bond formation and structure formation in nascent chains has remained elusive. Here, we investigate the eye-lens protein γB-crystallin in the ribosomal exit tunnel. Using mass spectrometry, theoretical simulations, dynamic nuclear polarization-enhanced solid-state nuclear magnetic resonance and cryo-electron microscopy, we show that thiol groups of cysteine residues undergo S-glutathionylation and S-nitrosylation and form non-native disulfide bonds. Thus, covalent modification chemistry occurs already prior to nascent chain release as the ribosome exit tunnel provides sufficient space even for disulfide bond formation which can guide protein folding.


Subject(s)
Cysteine/chemistry , Disulfides/chemistry , Protein Biosynthesis , Ribosomes/chemistry , Ribosomes/metabolism , gamma-Crystallins/chemistry , Cryoelectron Microscopy , Cysteine/metabolism , Glutathione/analogs & derivatives , Glutathione/chemistry , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Mutation , Oxidation-Reduction , Protein Conformation , Protein Folding , Ribosomes/genetics , S-Nitrosothiols/chemistry
3.
Mol Oncol ; 13(11): 2441-2459, 2019 11.
Article in English | MEDLINE | ID: mdl-31545551

ABSTRACT

Advanced colorectal carcinoma is currently incurable, and new therapies are urgently needed. We report that phosphotyrosine-dependent Eph receptor signaling sustains colorectal carcinoma cell survival, thereby uncovering a survival pathway active in colorectal carcinoma cells. We find that genetic and biochemical inhibition of Eph tyrosine kinase activity or depletion of the Eph ligand EphrinB2 reproducibly induces colorectal carcinoma cell death by autophagy. Spautin and 3-methyladenine, inhibitors of early steps in the autophagic pathway, significantly reduce autophagy-mediated cell death that follows inhibition of phosphotyrosine-dependent Eph signaling in colorectal cancer cells. A small-molecule inhibitor of the Eph kinase, NVP-BHG712 or its regioisomer NVP-Iso, reduces human colorectal cancer cell growth in vitro and tumor growth in mice. Colorectal cancers express the EphrinB ligand and its Eph receptors at significantly higher levels than numerous other cancer types, supporting Eph signaling inhibition as a potential new strategy for the broad treatment of colorectal carcinoma.


Subject(s)
Autophagy , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Molecular Targeted Therapy , Receptors, Eph Family/metabolism , Signal Transduction , Animals , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Ephrin-B2/metabolism , Female , Gene Silencing/drug effects , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Survival Analysis
4.
Nat Commun ; 10(1): 2915, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31266946

ABSTRACT

The bile acid-sensing transcription factor farnesoid X receptor (FXR) regulates multiple metabolic processes. Modulation of FXR is desired to overcome several metabolic pathologies but pharmacological administration of full FXR agonists has been plagued by mechanism-based side effects. We have developed a modulator that partially activates FXR in vitro and in mice. Here we report the elucidation of the molecular mechanism that drives partial FXR activation by crystallography- and NMR-based structural biology. Natural and synthetic FXR agonists stabilize formation of an extended helix α11 and the α11-α12 loop upon binding. This strengthens a network of hydrogen bonds, repositions helix α12 and enables co-activator recruitment. Partial agonism in contrast is conferred by a kink in helix α11 that destabilizes the α11-α12 loop, a critical determinant for helix α12 orientation. Thereby, the synthetic partial agonist induces conformational states, capable of recruiting both co-repressors and co-activators leading to an equilibrium of co-activator and co-repressor binding.


Subject(s)
Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/chemistry , Animals , Cell Line , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Humans , Hydrogen Bonding , Ligands , Liver/metabolism , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Protein Binding , Protein Conformation, alpha-Helical , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism
5.
ChemMedChem ; 13(18): 1988-1996, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30058283

ABSTRACT

A ligand-binding study is presented focusing on thermodynamics of fragment expansion. The binding of four compounds with increasing molecular weight to protein kinase A (PKA) was analyzed. The ligands display affinities between low-micromolar to nanomolar potency despite their low molecular weight. Binding free energies were measured by isothermal titration calorimetry, revealing a trend toward more entropic and less enthalpic binding with increase in molecular weight. All protein-ligand complexes were analyzed by crystallography and solution NMR spectroscopy. Crystal structures and solution NMR data are highly consistent, and no major differences in complex dynamics across the series are observed that would explain the differences in the thermodynamic profiles. Instead, the thermodynamic trends result either from differences in the solvation patterns of the conformationally more flexible ligand in aqueous solution prior to protein binding as molecular dynamics simulations suggest, or from local shifts of the water structure in the ligand-bound state. Our data thus provide evidence that changes in the solvation pattern constitute an important parameter for the understanding of thermodynamic data in protein-ligand complex formation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , Sulfonamides/chemistry , Thermodynamics , Water/chemistry , Animals , CHO Cells , Cricetulus , Crystallography, X-Ray , Cyclic AMP-Dependent Protein Kinases/isolation & purification , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Hydrophobic and Hydrophilic Interactions , Ligands , Models, Molecular , Molecular Structure , Molecular Weight , Structure-Activity Relationship
6.
ACS Chem Biol ; 13(8): 1921-1931, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29927572

ABSTRACT

The Polo-like kinases (Plks) are an evolutionary conserved family of Ser/Thr protein kinases that possess, in addition to the classical kinase domain at the N-terminus, a C-terminal polo-box domain (PBD) that binds to phosphorylated proteins and modulates the kinase activity and its localization. Plk1, which regulates the formation of the mitotic spindle, has emerged as a validated drug target for the treatment of cancer, because it is required for numerous types of cancer cells but not for the cell division in noncancer cells. Here, we employed chemical biology methods to investigate the allosteric communication between the PBD and the catalytic domain of Plk1. We identified small compounds that bind to the catalytic domain and inhibit or enhance the interaction of Plk1 with the phosphorylated peptide PoloBoxtide in vitro. In cells, two new allosteric Plk1 inhibitors affected the proliferation of cancer cells in culture and the cell cycle but had distinct phenotypic effects on spindle formation. Both compounds inhibited Plk1 signaling, indicating that they specifically act on Plk1 in cultured cells.


Subject(s)
Cell Cycle Proteins/agonists , Cell Cycle Proteins/antagonists & inhibitors , Enzyme Activators/chemistry , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/agonists , Proto-Oncogene Proteins/antagonists & inhibitors , Small Molecule Libraries/chemistry , Allosteric Regulation/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Catalytic Domain , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Centrosome/metabolism , Enzyme Activators/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , HeLa Cells , Humans , Kinetochores/metabolism , Oligopeptides/chemistry , Phosphopeptides/chemistry , Phosphopeptides/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Small Molecule Libraries/pharmacology , Spodoptera/chemistry , Polo-Like Kinase 1
7.
ChemMedChem ; 13(16): 1629-1633, 2018 08 20.
Article in English | MEDLINE | ID: mdl-29928781

ABSTRACT

Erythropoietin-producing hepatocellular (EPH) receptors are transmembrane receptor tyrosine kinases. Their extracellular domains bind specifically to ephrin A/B ligands, and this binding modulates intracellular kinase activity. EPHs are key players in bidirectional intercellular signaling, controlling cell morphology, adhesion, and migration. They are increasingly recognized as cancer drug targets. We analyzed the binding of NVP-BHG712 (NVP) to EPHA2 and EPHB4. Unexpectedly, all tested commercially available NVP samples turned out to be a regioisomer (NVPiso) of the inhibitor, initially described in a Novartis patent application. They only differ by the localization of a single methyl group on either one of two adjacent nitrogen atoms. The two compounds of identical mass revealed different binding modes. Furthermore, both in vitro and in vivo experiments showed that the isomers differ in their kinase affinity and selectivity.


Subject(s)
Pyrazoles/metabolism , Pyrimidines/metabolism , Receptor, EphA2/metabolism , Receptor, EphB4/metabolism , Crystallography, X-Ray , Humans , Isomerism , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Receptor, EphA2/chemistry , Receptor, EphB4/chemistry
8.
J Med Chem ; 61(14): 5922-5933, 2018 Jul 26.
Article in English | MEDLINE | ID: mdl-29909615

ABSTRACT

Biophysical parameters can accelerate drug development; e.g., rigid ligands may reduce entropic penalty and improve binding affinity. We studied systematically the impact of ligand rigidification on thermodynamics using a series of fasudil derivatives inhibiting protein kinase A by crystallography, isothermal titration calorimetry, nuclear magnetic resonance, and molecular dynamics simulations. The ligands varied in their internal degrees of freedom but conserve the number of heteroatoms. Counterintuitively, the most flexible ligand displays the entropically most favored binding. As experiment shows, this cannot be explained by higher residual flexibility of ligand, protein, or formed complex nor by a deviating or increased release of water molecules upon complex formation. NMR and crystal structures show no differences in flexibility and water release, although strong ligand-induced adaptations are observed. Instead, the flexible ligand entraps more efficiently water molecules in solution prior to protein binding, and by release of these waters, the favored entropic binding is observed.


Subject(s)
Entropy , Protein Kinases/metabolism , Solvents/chemistry , Drug Design , Ligands , Models, Molecular , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry , Water/chemistry
9.
Chembiochem ; 18(23): 2340-2350, 2017 12 05.
Article in English | MEDLINE | ID: mdl-28950050

ABSTRACT

The impact of the incorporation of a non-natural amino acid (NNAA) on protein structure, dynamics, and ligand binding has not been studied rigorously so far. NNAAs are regularly used to modify proteins post-translationally in vivo and in vitro through click chemistry. Herein, structural characterisation of the impact of the incorporation of azidohomoalanine (AZH) into the model protein domain PDZ3 is examined by means of NMR spectroscopy and X-ray crystallography. The structure and dynamics of the apo state of AZH-modified PDZ3 remain mostly unperturbed. Furthermore, the binding of two PDZ3 binding peptides are unchanged upon incorporation of AZH. The interface of the AZH-modified PDZ3 and an azulene-linked peptide for vibrational energy transfer studies has been mapped by means of chemical shift perturbations and NOEs between the unlabelled azulene-linked peptide and the isotopically labelled protein. Co-crystallisation and soaking failed for the peptide-bound holo complex. NMR spectroscopy, however, allowed determination of the protein-ligand interface. Although the incorporation of AZH was minimally invasive for PDZ3, structural analysis of NNAA-modified proteins through the methodology presented herein should be performed to ensure structural integrity of the studied target.


Subject(s)
Alanine/analogs & derivatives , Disks Large Homolog 4 Protein/chemistry , Ligands , Alanine/chemistry , Amino Acid Sequence , Circular Dichroism , Crystallography, X-Ray , Disks Large Homolog 4 Protein/genetics , Disks Large Homolog 4 Protein/metabolism , Isotope Labeling , Magnetic Resonance Spectroscopy , Mutagenesis , PDZ Domains/genetics , PDZ Domains/physiology , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry
10.
ChemMedChem ; 12(12): 999-1011, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28544567

ABSTRACT

The receptor tyrosine kinase EPHA2 has gained attention as a therapeutic drug target for cancer and infectious diseases. However, EPHA2 research and EPHA2-based therapies have been hampered by the lack of selective small-molecule inhibitors. Herein we report the synthesis and evaluation of dedicated EPHA2 inhibitors based on the clinical BCR-ABL/SRC inhibitor dasatinib as a lead structure. We designed hybrid structures of dasatinib and the previously known EPHA2 binders CHEMBL249097, PD-173955, and a known EPHB4 inhibitor in order to exploit both the ATP pocket entrance as well as the ribose pocket as binding epitopes in the kinase EPHA2. Medicinal chemistry and inhibitor design were guided by a chemical proteomics approach, allowing early selectivity profiling of the newly synthesized inhibitor candidates. Concomitant protein crystallography of 17 inhibitor co-crystals delivered detailed insight into the atomic interactions that underlie the structure-affinity relationship. Finally, the anti-proliferative effect of the inhibitor candidates was confirmed in the glioblastoma cell line SF-268. In this work, we thus discovered a novel EPHA2 inhibitor candidate that features an improved selectivity profile while maintaining potency against EPHA2 and anticancer activity in SF-268 cells.


Subject(s)
Chemistry, Pharmaceutical , Drug Discovery , Protein Kinase Inhibitors/pharmacology , Proteomics , Receptor, EphA2/antagonists & inhibitors , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Receptor, EphA2/metabolism , Structure-Activity Relationship
11.
ACS Chem Biol ; 11(12): 3400-3411, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27768280

ABSTRACT

The receptor tyrosine kinase EPHA2 (Ephrin type-A receptor 2) plays important roles in oncogenesis, metastasis, and treatment resistance, yet therapeutic targeting, drug discovery, or investigation of EPHA2 biology is hampered by the lack of appropriate inhibitors and structural information. Here, we used chemical proteomics to survey 235 clinical kinase inhibitors for their kinase selectivity and identified 24 drugs with submicromolar affinities for EPHA2. NMR-based conformational dynamics together with nine new cocrystal structures delineated drug-EPHA2 interactions in full detail. The combination of selectivity profiling, structure determination, and kinome wide sequence alignment allowed the development of a classification system in which amino acids in the drug binding site of EPHA2 are categorized into key, scaffold, potency, and selectivity residues. This scheme should be generally applicable in kinase drug discovery, and we anticipate that the provided information will greatly facilitate the development of selective EPHA2 inhibitors in particular and the repurposing of clinical kinase inhibitors in general.


Subject(s)
Drug Discovery/methods , Protein Kinase Inhibitors/pharmacology , Proteomics/methods , Receptor, EphA2/antagonists & inhibitors , Receptor, EphA2/metabolism , Amino Acid Sequence , Amino Acids/chemistry , Amino Acids/metabolism , Cell Line, Tumor , Clinical Trials as Topic , Humans , Ligands , Models, Molecular , Protein Binding , Protein Kinase Inhibitors/chemistry , Receptor, EphA2/chemistry
12.
Chembiochem ; 17(23): 2257-2263, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27685543

ABSTRACT

The receptor tyrosine kinase EPHA2 is overexpressed in several cancers (breast, head and neck, non-small-cell lung cancer). Small-molecule-based inhibition of the EPHA2 kinase domain (KD) is seen as an important strategy for therapeutic intervention. However, obtaining structural information by crystallography or NMR spectroscopy for drug discovery is severely hampered by the lack of pure, homogeneous protein. Here, different fragments of the EPHA2 KD were expressed and purified from both bacterial (Escherichia coli, BL21(DE3) cells) and insect cells (Spodoptera frugiperda, Sf9 cells).1 H,15 N HSQC was used to determine the proper folding and homogeneity of all the constructs. Protein from E. coli was well-folded but unstable, and it did not crystallize. However, a construct (D596-G900) produced in Sf9 cells yielded homogenous, well-folded protein that crystallized readily, thereby resulting in eleven new EPHA2-ligand crystal structures. We have also established a strategy for selective and uniform 15 N-amino acid labeling of EPHA2 KD in Sf9 cells for investigating dynamics and EPHA2-drug interactions by NMR.


Subject(s)
Chemical Fractionation , Nuclear Magnetic Resonance, Biomolecular , Protein Domains , Receptor, EphA2/chemistry , Animals , Crystallography, X-Ray , Escherichia coli/cytology , Escherichia coli/metabolism , Humans , Models, Molecular , Receptor, EphA2/biosynthesis , Receptor, EphA2/isolation & purification , Spodoptera/cytology , Spodoptera/metabolism
14.
ACS Chem Biol ; 10(11): 2570-9, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26279064

ABSTRACT

Polo-like kinase 1 (Plk1) is a central regulator of mitosis and has been validated as a target for antitumor therapy. The polo-box domain (PBD) of Plk1 regulates its kinase activity and mediates the subcellular localization of Plk1 and its interactions with a subset of its substrates. Functional inhibition of the Plk1 PBD by low-molecular weight inhibitors has been shown to represent a viable strategy by which to inhibit the enzyme, while avoiding selectivity issues caused by the conserved nature of the ATP binding site. Here, we report structure-activity relationships and mechanistic analysis for the first reported Plk1 PBD inhibitor, Poloxin. We present the identification of the optimized analog Poloxin-2, displaying significantly improved potency and selectivity over Poloxin. Poloxin-2 induces mitotic arrest and apoptosis in cultured human tumor cells at low micromolar concentrations, highlighting it as a valuable tool compound for exploring the function of the Plk1 PBD in living cells.


Subject(s)
Apoptosis/drug effects , Benzoates/chemistry , Benzoates/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Mitosis/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Quinones/chemistry , Quinones/pharmacology , Cell Cycle Proteins/classification , Cell Line, Tumor , Fluorescence , HeLa Cells , Humans , Inhibitory Concentration 50 , Molecular Structure , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/classification , Protein Structure, Tertiary , Proto-Oncogene Proteins/classification , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Polo-Like Kinase 1
15.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 8): 1088-93, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26249705

ABSTRACT

Protein kinases (PKs) are dynamic regulators of numerous cellular processes. Their phosphorylation activity is determined by the conserved kinase core structure, which is maintained by the interaction and dynamics with associated domains or interacting proteins. The prototype enzyme for investigations to understand the activity and regulation of PKs is the catalytic subunit of cAMP-dependent protein kinase (PKAc). Major effects of functional regulation and ligand binding are driven by only minor structural modulations in protein-protein interactions. In order to resolve such minor structural differences, very high resolution structures are required. Here, the high-resolution X-ray structure of PKAc from Cricetulus griseus is reported.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP/chemistry , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Animals , Base Sequence , Catalytic Domain , Cloning, Molecular , Cricetulus , Crystallization , Crystallography, X-Ray , Cyclic AMP-Dependent Protein Kinases/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Sequence Alignment
16.
J Biol Chem ; 290(26): 16415-30, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25979334

ABSTRACT

Low levels of reactive oxygen species (ROS) act as important signaling molecules, but in excess they can damage biomolecules. ROS regulation is therefore of key importance. Several polyphenols in general and flavonoids in particular have the potential to generate hydroxyl radicals, the most hazardous among all ROS. However, the generation of a hydroxyl radical and subsequent ROS formation can be prevented by methylation of the hydroxyl group of the flavonoids. O-Methylation is performed by O-methyltransferases, members of the S-adenosyl-l-methionine (SAM)-dependent O-methyltransferase superfamily involved in the secondary metabolism of many species across all kingdoms. In the filamentous fungus Podospora anserina, a well established aging model, the O-methyltransferase (PaMTH1) was reported to accumulate in total and mitochondrial protein extracts during aging. In vitro functional studies revealed flavonoids and in particular myricetin as its potential substrate. The molecular architecture of PaMTH1 and the mechanism of the methyl transfer reaction remain unknown. Here, we report the crystal structures of PaMTH1 apoenzyme, PaMTH1-SAM (co-factor), and PaMTH1-S-adenosyl homocysteine (by-product) co-complexes refined to 2.0, 1.9, and 1.9 Å, respectively. PaMTH1 forms a tight dimer through swapping of the N termini. Each monomer adopts the Rossmann fold typical for many SAM-binding methyltransferases. Structural comparisons between different O-methyltransferases reveal a strikingly similar co-factor binding pocket but differences in the substrate binding pocket, indicating specific molecular determinants required for substrate selection. Furthermore, using NMR, mass spectrometry, and site-directed active site mutagenesis, we show that PaMTH1 catalyzes the transfer of the methyl group from SAM to one hydroxyl group of the myricetin in a cation-dependent manner.


Subject(s)
Fungal Proteins/chemistry , Fungal Proteins/metabolism , Methyltransferases/chemistry , Methyltransferases/metabolism , Podospora/enzymology , S-Adenosylmethionine/metabolism , Biophysics , Crystallography, X-Ray , Flavonoids/chemistry , Flavonoids/metabolism , Fungal Proteins/genetics , Methyltransferases/genetics , Oxidative Stress , Podospora/chemistry , Podospora/genetics , Podospora/growth & development
17.
Proteins ; 81(11): 2007-22, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23852655

ABSTRACT

Eukaryotic proteins with important biological function can be partially unstructured, conformational flexible, or heterogenic. Crystallization trials often fail for such proteins. In NMR spectroscopy, parts of the polypeptide chain undergoing dynamics in unfavorable time regimes cannot be observed. De novo NMR structure determination is seriously hampered when missing signals lead to an incomplete chemical shift assignment resulting in an information content of the NOE data insufficient to determine the structure ab initio. We developed a new protein structure determination strategy for such cases based on a novel NOE assignment strategy utilizing a number of model structures but no explicit reference structure as it is used for bootstrapping like algorithms. The software distinguishes in detail between consistent and mutually exclusive pairs of possible NOE assignments on the basis of different precision levels of measured chemical shifts searching for a set of maximum number of consistent NOE assignments in agreement with 3D space. Validation of the method using the structure of the low molecular-weight-protein tyrosine phosphatase A (MptpA) showed robust results utilizing protein structures with 30-45% sequence identity and 70% of the chemical shift assignments. About 60% of the resonance assignments are sufficient to identify those structural models with highest conformational similarity to the real structure. The software was benchmarked by de novo solution structures of fibroblast growth factor 21 (FGF21) and the extracellular fibroblast growth factor receptor domain FGFR4 D2, which both failed in crystallization trials and in classical NMR structure determination.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Proteins/chemistry , Software , Algorithms , Receptors, Fibroblast Growth Factor/chemistry
18.
Cancer Cell ; 23(4): 489-501, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23597563

ABSTRACT

The fibroblast growth factor (FGF)/fibroblast growth factor receptor (FGFR) signaling network plays an important role in cell growth, survival, differentiation, and angiogenesis. Deregulation of FGFR signaling can lead to cancer development. Here, we report an FGFR inhibitor, SSR128129E (SSR), that binds to the extracellular part of the receptor. SSR does not compete with FGF for binding to FGFR but inhibits FGF-induced signaling linked to FGFR internalization in an allosteric manner, as shown by crystallography studies, nuclear magnetic resonance, Fourier transform infrared spectroscopy, molecular dynamics simulations, free energy calculations, structure-activity relationship analysis, and FGFR mutagenesis. Overall, SSR is a small molecule allosteric inhibitor of FGF/FGFR signaling, acting via binding to the extracellular part of the FGFR.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Allosteric Regulation/drug effects , Binding, Competitive , Cell Growth Processes/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Conformation/drug effects , Protein Structure, Tertiary , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
19.
Biol Chem ; 393(10): 1131-40, 2012 Sep 08.
Article in English | MEDLINE | ID: mdl-23096351

ABSTRACT

Splicing of pre-mRNA requires the activity of at least eight different DEAD/H-box proteins that are involved in distinct steps of the splicing process. These proteins are driving the spliceosomal machinery by ATP-dependent unwinding of dsRNA and/or disrupting protein-RNA complexes. The spliceosomal DEAH-box proteins Prp2, Prp16, Prp22 and Prp43 share homologous C-terminal domains (CTD). We have determined the crystal structure of the CTD of human Prp22 by means of MAD. The fold of the human Prp22-CTD closely resembles that of the yeast Prp43-CTD. The similarity of these helicase-associated CTDs to the winged-helix and ratchet domains of the DNA helicase Hel308 suggests an analogous function in dsRNA binding and unwinding. Here, we also demonstrate that the CTD has a significant impact on the ATPase activity of yPrp22 in vitro. Homology modeling of the CTDs of hPrp2, hPrp16 and hPrp43 suggests that the CTDs of spliceosomal helicases contain conserved positively charged patches on their surfaces representing a common RNA-binding surface as well as divergent regions most likely mediating specific interactions with different proteins of the spliceosome.


Subject(s)
DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Enzyme Stability , Humans , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , RNA/metabolism , RNA Splicing Factors , Saccharomyces cerevisiae/enzymology , Sequence Homology, Amino Acid
20.
Protein Expr Purif ; 85(1): 142-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22841618

ABSTRACT

The TATA binding protein (TBP) is the central core protein of the transcription factor II D that binds directly to the TATA box and therefore plays an integral part in eukaryotic transcription. This pivotal position of TBP is underlined by the vast number of interaction partners involved. Expression and purification of human TATA binding protein (hTBP) has remained a challenge due to protein instability and the protein loss during expression and purification involved. Here, we present a novel approach for high yield expression and purification of human TBP core (hTBPc) protein. Protein fold and activity are verified by nuclear magnetic resonance (NMR) spectroscopy and microscale thermophoresis (MST).


Subject(s)
Cloning, Molecular , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/isolation & purification , Amino Acid Sequence , Escherichia coli/genetics , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Folding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...