Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
ACS Chem Biol ; 17(12): 3527-3534, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36417668

ABSTRACT

Glycans attached to glycoproteins can contribute to stability, mediate interactions with other proteins, and initiate signal transduction. Glycan conformation, which is critical to these processes, is highly variable and often depicted as sampling a multitude of conformers. These conformers can be generated by molecular dynamics simulations, and more inclusively by accelerated molecular dynamics, as well as other extended sampling methods. However, experimental assessments of the contribution that various conformers make to a native ensemble are rare. Here, we use long-range pseudo-contact shifts (PCSs) of NMR resonances from an isotopically labeled glycoprotein to identify preferred conformations of its glycans. The N-terminal domain from human Carcinoembryonic Antigen Cell Adhesion Molecule 1, hCEACAM1-Ig1, was used as the model glycoprotein in this study. It has been engineered to include a lanthanide-ion-binding loop that generates PCSs, as well as a homogeneous set of three 13C-labeled N-glycans. Analysis of the PCSs indicates that preferred glycan conformers have extensive contacts with the protein surface. Factors leading to this preference appear to include interactions between N-acetyl methyls of GlcNAc residues and hydrophobic surface pockets on the protein surface.


Subject(s)
Cell Adhesion Molecules , Polysaccharides , Humans , Antigens, CD/chemistry , Antigens, CD/metabolism , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Glycoproteins/chemistry , Polysaccharides/chemistry , Polysaccharides/metabolism , Protein Conformation
2.
J Magn Reson ; 345: 107336, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36442299

ABSTRACT

Not all proteins are amenable to uniform isotopic labeling with 13C and 15N, something needed for the widely used, and largely deductive, triple resonance assignment process. Among them are proteins expressed in mammalian cell culture where native glycosylation can be maintained, and proper formation of disulfide bonds facilitated. Uniform labeling in mammalian cells is prohibitively expensive, but sparse labeling with one or a few isotopically enriched amino acid types is an option for these proteins. However, assignment then relies on accessing the best match between a variety of measured NMR parameters and predictions based on 3D structure, often from X-ray crystallography. Finding this match is a challenging process that has benefitted from many computational tools, including trained neural nets for chemical shift prediction, genetic algorithms for searches through a myriad of assignment possibilities, and now AI-based prediction of high-quality structures for protein targets. AssignSLP_GUI, a new version of a software package for assignment of resonances from sparsely-labeled proteins, uses many of these tools. These tools and new additions to the package are highlighted in an application to a sparsely-labeled domain from a glycoprotein, CEACAM1.

3.
J Biol Chem ; 298(6): 102047, 2022 06.
Article in English | MEDLINE | ID: mdl-35597280

ABSTRACT

Thrombospondin type-1 repeats (TSRs) are small protein motifs containing six conserved cysteines forming three disulfide bonds that can be modified with an O-linked fucose. Protein O-fucosyltransferase 2 (POFUT2) catalyzes the addition of O-fucose to TSRs containing the appropriate consensus sequence, and the O-fucose modification can be elongated to a Glucose-Fucose disaccharide with the addition of glucose by ß3-glucosyltransferase (B3GLCT). Elimination of Pofut2 in mice results in embryonic lethality in mice, highlighting the biological significance of O-fucose modification on TSRs. Knockout of POFUT2 in HEK293T cells has been shown to cause complete or partial loss of secretion of many proteins containing O-fucosylated TSRs. In addition, POFUT2 is localized to the endoplasmic reticulum (ER) and only modifies folded TSRs, stabilizing their structures. These observations suggest that POFUT2 is involved in an ER quality control mechanism for TSR folding and that B3GLCT also participates in quality control by providing additional stabilization to TSRs. However, the mechanisms by which addition of these sugars result in stabilization are poorly understood. Here, we conducted molecular dynamics (MD) simulations and provide crystallographic and NMR evidence that the Glucose-Fucose disaccharide interacts with specific amino acids in the TSR3 domain in thrombospondin-1 that are within proximity to the O-fucosylation modification site resulting in protection of a nearby disulfide bond. We also show that mutation of these amino acids reduces the stabilizing effect of the sugars in vitro. These data provide mechanistic details regarding the importance of O-fucosylation and how it participates in quality control mechanisms inside the ER.


Subject(s)
Fucose , Fucosyltransferases , Thrombospondin 1 , Animals , Disaccharides , Disulfides , Endoplasmic Reticulum/metabolism , Fucose/metabolism , Fucosyltransferases/metabolism , Galactosyltransferases , Glucose , Glucosyltransferases/metabolism , HEK293 Cells , Humans , Mice , Molecular Dynamics Simulation , Thrombospondin 1/chemistry
4.
Commun Biol ; 5(1): 452, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35551273

ABSTRACT

High resolution hydroxyl radical protein footprinting (HR-HRPF) is a mass spectrometry-based method that measures the solvent exposure of multiple amino acids in a single experiment, offering constraints for experimentally informed computational modeling. HR-HRPF-based modeling has previously been used to accurately model the structure of proteins of known structure, but the technique has never been used to determine the structure of a protein of unknown structure. Here, we present the use of HR-HRPF-based modeling to determine the structure of the Ig-like domain of NRG1, a protein with no close homolog of known structure. Independent determination of the protein structure by both HR-HRPF-based modeling and heteronuclear NMR was carried out, with results compared only after both processes were complete. The HR-HRPF-based model was highly similar to the lowest energy NMR model, with a backbone RMSD of 1.6 Å. To our knowledge, this is the first use of HR-HRPF-based modeling to determine a previously uncharacterized protein structure.


Subject(s)
Protein Footprinting , Proteins , Computer Simulation , Hydroxyl Radical/chemistry , Immunoglobulin Domains , Mass Spectrometry , Protein Footprinting/methods , Proteins/chemistry
5.
Biochemistry ; 59(15): 1527-1536, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32227851

ABSTRACT

Skp1 is an adapter that links F-box proteins to cullin-1 in the Skp1/cullin-1/F-box (SCF) protein family of E3 ubiquitin ligases that targets specific proteins for polyubiquitination and subsequent protein degradation. Skp1 from the amoebozoan Dictyostelium forms a stable homodimer in vitro with a Kd of 2.5 µM as determined by sedimentation velocity studies yet is monomeric in crystal complexes with F-box proteins. To investigate the molecular basis for the difference, we determined the solution NMR structure of a doubly truncated Skp1 homodimer (Skp1ΔΔ). The solution structure of the Skp1ΔΔ dimer reveals a 2-fold symmetry with an interface that buries ∼750 Å2 of predominantly hydrophobic surface. The dimer interface overlaps with subsite 1 of the F-box interaction area, explaining why only the Skp1 monomer binds F-box proteins (FBPs). To confirm the model, Rosetta was used to predict amino acid substitutions that might disrupt the dimer interface, and the F97E substitution was chosen to potentially minimize interference with F-box interactions. A nearly full-length version of Skp1 with this substitution (Skp1ΔF97E) behaved as a stable monomer at concentrations of ≤500 µM and actively bound a model FBP, mammalian Fbs1, which suggests that the dimeric state is not required for Skp1 to carry out a basic biochemical function. Finally, Skp1ΔF97E is expected to serve as a monomer model for high-resolution NMR studies previously hindered by dimerization.


Subject(s)
F-Box Proteins/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Binding Sites , Dimerization , F-Box Proteins/chemistry , Humans , Models, Molecular , S-Phase Kinase-Associated Proteins/chemistry
6.
J Mol Biol ; 431(12): 2369-2382, 2019 05 31.
Article in English | MEDLINE | ID: mdl-31034888

ABSTRACT

Characterization of proteins using NMR methods begins with assignment of resonances to specific residues. This is usually accomplished using sequential connectivities between nuclear pairs in proteins uniformly labeled with NMR active isotopes. This becomes impractical for larger proteins, and especially for proteins that are best expressed in mammalian cells, including glycoproteins. Here an alternate protocol for the assignment of NMR resonances of sparsely labeled proteins, namely, the ones labeled with a single amino acid type, or a limited subset of types, isotopically enriched with 15N or 13C, is described. The protocol is based on comparison of data collected using extensions of simple two-dimensional NMR experiments (correlated chemical shifts, nuclear Overhauser effects, residual dipolar couplings) to predictions from molecular dynamics trajectories that begin with known protein structures. Optimal pairing of predicted and experimental values is facilitated by a software package that employs a genetic algorithm, ASSIGN_SLP_MD. The approach is applied to the 36-kDa luminal domain of the sialyltransferase, rST6Gal1, in which all phenylalanines are labeled with 15N, and the results are validated by elimination of resonances via single-point mutations of selected phenylalanines to tyrosines. Assignment allows the use of previously published paramagnetic relaxation enhancements to evaluate placement of a substrate analog in the active site of this protein. The protocol will open the way to structural characterization of the many glycosylated and other proteins that are best expressed in mammalian cells.


Subject(s)
Glycoproteins/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Algorithms , Animals , HEK293 Cells , Humans , Models, Molecular , Protein Conformation , Rats , Sialyltransferases/chemistry , Software
7.
J Biol Chem ; 294(11): 4027-4044, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30655293

ABSTRACT

As complications associated with antibiotic resistance have intensified, copper (Cu) is attracting attention as an antimicrobial agent. Recent studies have shown that copper surfaces decrease microbial burden, and host macrophages use Cu to increase bacterial killing. Not surprisingly, microbes have evolved mechanisms to tightly control intracellular Cu pools and protect against Cu toxicity. Here, we identified two genes (copB and copL) encoded within the Staphylococcus aureus arginine-catabolic mobile element (ACME) that we hypothesized function in Cu homeostasis. Supporting this hypothesis, mutational inactivation of copB or copL increased copper sensitivity. We found that copBL are co-transcribed and that their transcription is increased during copper stress and in a strain in which csoR, encoding a Cu-responsive transcriptional repressor, was mutated. Moreover, copB displayed genetic synergy with copA, suggesting that CopB functions in Cu export. We further observed that CopL functions independently of CopB or CopA in Cu toxicity protection and that CopL from the S. aureus clone USA300 is a membrane-bound and surface-exposed lipoprotein that binds up to four Cu+ ions. Solution NMR structures of the homologous Bacillus subtilis CopL, together with phylogenetic analysis and chemical-shift perturbation experiments, identified conserved residues potentially involved in Cu+ coordination. The solution NMR structure also revealed a novel Cu-binding architecture. Of note, a CopL variant with defective Cu+ binding did not protect against Cu toxicity in vivo Taken together, these findings indicate that the ACME-encoded CopB and CopL proteins are additional factors utilized by the highly successful S. aureus USA300 clone to suppress copper toxicity.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Cell Membrane/metabolism , Copper/toxicity , Operon/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Bacterial Proteins/genetics , Carrier Proteins/genetics , Cell Membrane/drug effects , Copper/metabolism , Staphylococcus aureus/metabolism
8.
ACS Chem Biol ; 13(9): 2560-2567, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30063822

ABSTRACT

An enzyme- and click chemistry-mediated methodology for the site-specific nitroxide spin labeling of glycoproteins has been developed and applied. The procedure relies on the presence of single N-glycosylation sites that are present natively in proteins or that can be engineered into glycoproteins by mutational elimination of all but one glycosylation site. Recombinantly expressing glycoproteins in HEK293S (GnT1-) cells results in N-glycans with high-mannose structures that can be processed to leave a single GlcNAc residue. This can in turn be modified by enzymatic addition of a GalNAz residue that is subject to reaction with an alkyne-carrying TEMPO moiety using copper(I)-catalyzed click chemistry. To illustrate the procedure, we have made an application to a two-domain construct of Robo1, a protein that carries a single N-glycosylation site in its N-terminal domains. The construct has also been labeled with 15N at amide nitrogens of lysine residues to provide a set of sites that are used to derive an effective location of the paramagnetic nitroxide moiety of the TEMPO group. This, in turn, allowed measurements of paramagnetic perturbations to the spectra of a new high affinity heparan sulfate ligand. Calculation of distance constraints from these data facilitated determination of an atomic level model for the docked complex.


Subject(s)
Glycoproteins/metabolism , Heparitin Sulfate/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Click Chemistry , Cyclic N-Oxides/analysis , Cyclic N-Oxides/metabolism , Electron Spin Resonance Spectroscopy , Glycoproteins/chemistry , Glycosylation , HEK293 Cells , Heparitin Sulfate/chemistry , Humans , Ligands , Molecular Docking Simulation , Nerve Tissue Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Receptors, Immunologic/chemistry , Roundabout Proteins
9.
Biochemistry ; 57(5): 511-515, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29251491

ABSTRACT

Cullin-ring-ligases mediate protein polyubiquitination, a signal for degradation in the 26S proteasome. The CRL1 class consists of Skp1/cullin-1/F-box protein/Rbx1 (SCF) complexes that cyclically associate with ubiquitin-E2 to build the polyubiquitin chain. Within the SCF complex, the 162-amino acid DdSkp1 from Dictyostelium bridges cullin-1 with an F-box protein (FBP), the specificity factor for substrate selection. The hydroxylation-dependent glycosylation of Pro143 of DdSkp1 by a pentasaccharide forms the basis of a novel O2-sensing mechanism in the social amoeba Dictyostelium and other protists. Previous evidence indicated that glycosylation promotes increased α-helical content correlating with enhanced interaction with three F-box proteins. To localize these differences, we used nuclear magnetic resonance (NMR) methods to compare nonglycosylated DdSkp1 and a glycoform with a single GlcNAc sugar (Gn-DdSkp1). We report NMR assignments of backbone 1HN, 15N, 13Cα, and 13CO nuclei as well as side-chain 13Cß and methyl 13C/1H nuclei of Ile(δ1), Leu, and Val in both unmodified DdSkp1 and Gn-DdSkp1. The random coil index and 15N{1H} HNOE indicate that the C-terminal region, which forms a helix-loop-helix motif centered on Pro143 at the crystallographically defined binding interface with F-box domains, remains dynamic in both DdSkp1 and Gn-DdSkp1. Chemical shifts indicate that the variation of conformation in Gn-DdSkp1, relative to DdSkp1, is limited to this region and characterized by increased helical fold. Extension of the glycan chain results in further changes, also limited to this region. Thus, glycosylation may control F-box protein interactions via a local effect on DdSkp1 conformation, by a mechanism that may be general to many unicellular eukaryotes.


Subject(s)
Dictyostelium/metabolism , F-Box Motifs , F-Box Proteins/metabolism , Protozoan Proteins/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Amino Acid Sequence , Dictyostelium/chemistry , F-Box Proteins/chemistry , Glycosylation , Humans , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Maps , Protozoan Proteins/chemistry , S-Phase Kinase-Associated Proteins/chemistry , SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/metabolism , Sequence Alignment
10.
Nature ; 538(7625): 329-335, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27626386

ABSTRACT

Naturally occurring, pharmacologically active peptides constrained with covalent crosslinks generally have shapes that have evolved to fit precisely into binding pockets on their targets. Such peptides can have excellent pharmaceutical properties, combining the stability and tissue penetration of small-molecule drugs with the specificity of much larger protein therapeutics. The ability to design constrained peptides with precisely specified tertiary structures would enable the design of shape-complementary inhibitors of arbitrary targets. Here we describe the development of computational methods for accurate de novo design of conformationally restricted peptides, and the use of these methods to design 18-47 residue, disulfide-crosslinked peptides, a subset of which are heterochiral and/or N-C backbone-cyclized. Both genetically encodable and non-canonical peptides are exceptionally stable to thermal and chemical denaturation, and 12 experimentally determined X-ray and NMR structures are nearly identical to the computational design models. The computational design methods and stable scaffolds presented here provide the basis for development of a new generation of peptide-based drugs.


Subject(s)
Computer-Aided Design , Drug Design , Peptides/chemistry , Peptides/chemical synthesis , Protein Stability , Amino Acid Motifs , Crystallography, X-Ray , Cyclization , Disulfides/chemistry , Hot Temperature , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Peptides/genetics , Peptides, Cyclic/chemistry , Peptides, Cyclic/genetics , Protein Denaturation , Protein Structure, Secondary , Protein Structure, Tertiary , Stereoisomerism
11.
Protein Sci ; 25(1): 30-45, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26293815

ABSTRACT

We have developed an online NMR / X-ray Structure Pair Data Repository. The NIGMS Protein Structure Initiative (PSI) has provided many valuable reagents, 3D structures, and technologies for structural biology. The Northeast Structural Genomics Consortium was one of several PSI centers. NESG used both X-ray crystallography and NMR spectroscopy for protein structure determination. A key goal of the PSI was to provide experimental structures for at least one representative of each of hundreds of targeted protein domain families. In some cases, structures for identical (or nearly identical) constructs were determined by both NMR and X-ray crystallography. NMR spectroscopy and X-ray diffraction data for 41 of these "NMR / X-ray" structure pairs determined using conventional triple-resonance NMR methods with extensive sidechain resonance assignments have been organized in an online NMR / X-ray Structure Pair Data Repository. In addition, several NMR data sets for perdeuterated, methyl-protonated protein samples are included in this repository. As an example of the utility of this repository, these data were used to revisit questions about the precision and accuracy of protein NMR structures first outlined by Levy and coworkers several years ago (Andrec et al., Proteins 2007;69:449-465). These results demonstrate that the agreement between NMR and X-ray crystal structures is improved using modern methods of protein NMR spectroscopy. The NMR / X-ray Structure Pair Data Repository will provide a valuable resource for new computational NMR methods development.


Subject(s)
Crystallography, X-Ray , Databases, Protein , Nuclear Magnetic Resonance, Biomolecular , Models, Molecular , Protein Conformation , Proteins/chemistry
12.
J Am Chem Soc ; 137(35): 11242-5, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-26293598

ABSTRACT

NMR residual dipolar couplings (RDCs) are exquisite probes of protein structure and dynamics. A new solution NMR experiment named 2D SE2 J-TROSY is presented to measure N-H RDCs for proteins and supramolecular complexes in excess of 200 kDa. This enables validation and refinement of their X-ray crystal and solution NMR structures and the characterization of structural and dynamic changes occurring upon complex formation. Accurate N-H RDCs were measured at 750 MHz (1)H resonance frequency for 11-mer 93 kDa (2)H,(15)N-labeled Trp RNA-binding attenuator protein tumbling with a correlation time τc of 120 ns. This is about twice as long as that for the most slowly tumbling system, for which N-H RDCs could be measured, so far, and corresponds to molecular weights of ∼200 kDa at 25 °C. Furthermore, due to the robustness of SE2 J-TROSY with respect to residual (1)H density from exchangeable protons, increased sensitivity at (1)H resonance frequencies around 1 GHz promises to enable N-H RDC measurement for even larger systems.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Proteins/chemistry , Bacterial Proteins/chemistry , Models, Molecular , Molecular Weight , Protein Conformation , RNA-Binding Proteins/chemistry , Solutions , Transcription Factors/chemistry
13.
Biomol NMR Assign ; 9(1): 135-138, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24722902

ABSTRACT

The 500 kDa protein plectin is essential for the cytoskeletal organization of most mammalian cells and it is up-regulated in some types of cancer. Here, we report nearly complete sequence-specific polypeptide backbone, (13)C(ß) and methyl group resonance assignments for 24 kDa human plectin(4403-4606) containing the C-terminal plectin repeat domain 6.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemistry , Plectin/chemistry , Humans , Protein Structure, Tertiary
14.
PLoS One ; 9(7): e101787, 2014.
Article in English | MEDLINE | ID: mdl-25010333

ABSTRACT

Bacterial species in the Enterobacteriaceae typically contain multiple paralogues of a small domain of unknown function (DUF1471) from a family of conserved proteins also known as YhcN or BhsA/McbA. Proteins containing DUF1471 may have a single or three copies of this domain. Representatives of this family have been demonstrated to play roles in several cellular processes including stress response, biofilm formation, and pathogenesis. We have conducted NMR and X-ray crystallographic studies of four DUF1471 domains from Salmonella representing three different paralogous DUF1471 subfamilies: SrfN, YahO, and SssB/YdgH (two of its three DUF1471 domains: the N-terminal domain I (residues 21-91), and the C-terminal domain III (residues 244-314)). Notably, SrfN has been shown to have a role in intracellular infection by Salmonella Typhimurium. These domains share less than 35% pairwise sequence identity. Structures of all four domains show a mixed α+ß fold that is most similar to that of bacterial lipoprotein RcsF. However, all four DUF1471 sequences lack the redox sensitive cysteine residues essential for RcsF activity in a phospho-relay pathway, suggesting that DUF1471 domains perform a different function(s). SrfN forms a dimer in contrast to YahO and SssB domains I and III, which are monomers in solution. A putative binding site for oxyanions such as phosphate and sulfate was identified in SrfN, and an interaction between the SrfN dimer and sulfated polysaccharides was demonstrated, suggesting a direct role for this DUF1471 domain at the host-pathogen interface.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Salmonella typhimurium , Amino Acid Sequence , Ligands , Models, Molecular , Molecular Sequence Data , Polysaccharides/chemistry , Polysaccharides/metabolism , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Sulfates/chemistry
15.
J Struct Funct Genomics ; 15(4): 201-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24941917

ABSTRACT

High-quality solution NMR structures of three homeodomains from human proteins ALX4, ZHX1 and CASP8AP2 were solved. These domains were chosen as targets of a biomedical theme project pursued by the Northeast Structural Genomics Consortium. This project focuses on increasing the structural coverage of human proteins associated with cancer.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Calcium-Binding Proteins/chemistry , DNA-Binding Proteins/chemistry , Homeodomain Proteins/chemistry , Neoplasm Proteins/chemistry , Neoplasms/chemistry , Transcription Factors/chemistry , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary
16.
ChemMedChem ; 9(2): 282-5, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24403054

ABSTRACT

The bacterial genus Corynebacteria contains several pathogenic species that cause diseases such as diphtheria in humans and "cheesy gland" in goats and sheep. Thus, identifying new therapeutic targets to treat Corynebacteria infections is both medically and economically important. CG2496, a functionally uncharacterized protein from Corynebacterium glutamicum, was evaluated using an NMR ligand-affinity screen. A total of 11 compounds from a library of 460 biologically active compounds were shown to selectively bind CG2496 in a highly conserved region of the protein. The best binder was identified to be methiothepin (KD =54 ± 19 µM), an FDA-approved serotonin receptor antagonist. Methiothepin was also shown to inhibit the growth of C. glutamicum, but not bacteria that lack CG2496 homologs. Our results suggest that CG2496 is a novel therapeutic target and methiothepin is a potential lead compound or structural scaffold for developing new antibiotics specifically targeting Corynebacteria.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Corynebacterium Infections/drug therapy , Corynebacterium Infections/veterinary , Corynebacterium/drug effects , Animals , Bacterial Proteins/chemistry , Corynebacterium/chemistry , Corynebacterium/growth & development , Corynebacterium/metabolism , Drug Discovery , Goats , Humans , Ligands , Magnetic Resonance Spectroscopy , Methiothepin/chemistry , Methiothepin/pharmacology , Models, Molecular , Sheep , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
17.
J Struct Funct Genomics ; 14(4): 155-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24048810

ABSTRACT

A high-quality structure of the 68-residue protein CD1104B from Clostridium difficile strain 630 exhibits a distinct all α-helical fold. The structure presented here is the first representative of bacterial protein domain family PF14203 (currently 180 members) of unknown function (DUF4319) and reveals that the side-chains of the only two strictly conserved residues (Glu 8 and Lys 48) form a salt bridge. Moreover, these two residues are located in the vicinity of the largest surface cleft which is predicted to contribute to a surface area involved in protein-protein interactions. This, along with its coding in transposon CTn4, suggests that CD1104B (and very likely all members of Pfam 14203) functions by interacting with other proteins required for the transfer of transposons between different bacterial species.


Subject(s)
Bacterial Proteins/chemistry , Clostridioides difficile/chemistry , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary , Amino Acid Sequence , Molecular Sequence Data , Sequence Alignment , Solutions
18.
J Struct Funct Genomics ; 14(3): 119-26, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23963952

ABSTRACT

High-quality NMR structures of the C-terminal domain comprising residues 484-537 of the 537-residue protein Bacterial chlorophyll subunit B (BchB) from Chlorobium tepidum and residues 9-61 of 61-residue Asr4154 from Nostoc sp. (strain PCC 7120) exhibit a mixed α/ß fold comprised of three α-helices and a small ß-sheet packed against second α-helix. These two proteins share 29% sequence similarity and their structures are globally quite similar. The structures of BchB(484-537) and Asr4154(9-61) are the first representative structures for the large protein family (Pfam) PF08369, a family of unknown function currently containing 610 members in bacteria and eukaryotes. Furthermore, BchB(484-537) complements the structural coverage of the dark-operating protochlorophyllide oxidoreductase.


Subject(s)
Chlorophyll Binding Proteins/ultrastructure , Nuclear Magnetic Resonance, Biomolecular/methods , Oxidoreductases/ultrastructure , Bacterial Proteins/chemistry , Bacterial Proteins/ultrastructure , Chlorobium/chemistry , Chlorophyll Binding Proteins/chemistry , Nostoc/chemistry , Oxidoreductases/chemistry , Protochlorophyllide/metabolism
19.
J Am Chem Soc ; 134(51): 20589-92, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23215000

ABSTRACT

A new method is presented for measuring kinetic isotope effects (KIEs) by (1)H-detected 2D [(13)C,(1)H]-heteronuclear single quantum coherence (HSQC) NMR spectroscopy. The high accuracy of this approach was exemplified for the reaction catalyzed by glucose-6-phosphate dehydrogenase by comparing the 1-(13)C KIE with the published value obtained using isotope ratio mass spectrometry. High precision was demonstrated for the reaction catalyzed by 1-deoxy-D-xylulose-5-phosphate reductoisomerase from Mycobacterium tuberculosis. 2-, 3-, and 4-(13)C KIEs were found to be 1.0031(4), 1.0303(12), and 1.0148(2), respectively. These KIEs provide evidence for a cleanly rate-limiting retroaldol step during isomerization. The high intrinsic sensitivity and signal dispersion of 2D [(13)C,(1)H]-HSQC offer new avenues to study challenging systems where low substrate concentration and/or signal overlap impedes 1D (13)C NMR data acquisition. Moreover, this approach can take advantage of highest-field spectrometers, which are commonly equipped for (1)H detection with cryogenic probes.

20.
Biochemistry ; 51(43): 8475-7, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23062074

ABSTRACT

The yeast mitochondrial protein Sdh5 is required for the covalent attachment of flavin adenine dinucleotide (FAD) to protein Sdh1, a subunit of the heterotetrameric enzyme succinate dehydrogenase. The NMR structure of Sdh5 represents the first eukaryotic structure of Pfam family PF03937 and reveals a conserved surface region, which likely represents a putative Sdh1-Sdh5 interaction interface. Point mutations in this region result in the loss of covalent flavinylation of Sdh1. Moreover, chemical shift perturbation measurements showed that Sdh5 does not bind FAD in vitro, indicating that it is not a simple cofactor transporter in vivo.


Subject(s)
Flavin-Adenine Dinucleotide/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism , Succinate Dehydrogenase/metabolism , Amino Acid Sequence , Binding Sites , Mitochondrial Proteins/genetics , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Point Mutation , Protein Binding , Protein Conformation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...