Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
1.
Cell ; 165(3): 690-703, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27062925

ABSTRACT

Pili are proteinaceous polymers of linked pilins that protrude from the cell surface of many bacteria and often mediate adherence and virulence. We investigated a set of 20 Bacteroidia pilins from the human microbiome whose structures and mechanism of assembly were unknown. Crystal structures and biochemical data revealed a diverse protein superfamily with a common Greek-key ß sandwich fold with two transthyretin-like repeats that polymerize into a pilus through a strand-exchange mechanism. The assembly mechanism of the central, structural pilins involves proteinase-assisted removal of their N-terminal ß strand, creating an extended hydrophobic groove that binds the C-terminal donor strands of the incoming pilin. Accessory pilins at the tip and base have unique structural features specific to their location, allowing initiation or termination of the assembly. The Bacteroidia pilus, therefore, has a biogenesis mechanism that is distinct from other known pili and likely represents a different type of bacterial pilus.


Subject(s)
Fimbriae Proteins/chemistry , Fimbriae, Bacterial , Gastrointestinal Microbiome , Amino Acid Sequence , Crystallography, X-Ray , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Humans , Lipoproteins/chemistry , Lipoproteins/metabolism , Models, Molecular , Molecular Sequence Data , Sequence Alignment
2.
Int J Mol Sci ; 24(23)2023 Nov 23.
Article in English | MEDLINE | ID: mdl-38068959

ABSTRACT

The ability to quickly discover reliable hits from screening and rapidly convert them into lead compounds, which can be verified in functional assays, is central to drug discovery. The expedited validation of novel targets and the identification of modulators to advance to preclinical studies can significantly increase drug development success. Our SaXPyTM ("SAR by X-ray Poses Quickly") platform, which is applicable to any X-ray crystallography-enabled drug target, couples the established methods of protein X-ray crystallography and fragment-based drug discovery (FBDD) with advanced computational and medicinal chemistry to deliver small molecule modulators or targeted protein degradation ligands in a short timeframe. Our approach, especially for elusive or "undruggable" targets, allows for (i) hit generation; (ii) the mapping of protein-ligand interactions; (iii) the assessment of target ligandability; (iv) the discovery of novel and potential allosteric binding sites; and (v) hit-to-lead execution. These advances inform chemical tractability and downstream biology and generate novel intellectual property. We describe here the application of SaXPy in the discovery and development of DNA damage response inhibitors against DNA polymerase eta (Pol η or POLH) and apurinic/apyrimidinic endonuclease 1 (APE1 or APEX1). Notably, our SaXPy platform allowed us to solve the first crystal structures of these proteins bound to small molecules and to discover novel binding sites for each target.


Subject(s)
DNA-Directed DNA Polymerase , Drug Discovery , DNA-Directed DNA Polymerase/metabolism , Binding Sites , Endonucleases/metabolism , Crystallography, X-Ray , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism
3.
Proc Natl Acad Sci U S A ; 112(15): 4666-71, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25825768

ABSTRACT

NANOG (from Irish mythology Tír na nÓg) transcription factor plays a central role in maintaining pluripotency, cooperating with OCT4 (also known as POU5F1 or OCT3/4), SOX2, and other pluripotency factors. Although the physiological roles of the NANOG protein have been extensively explored, biochemical and biophysical properties in relation to its structural analysis are poorly understood. Here we determined the crystal structure of the human NANOG homeodomain (hNANOG HD) bound to an OCT4 promoter DNA, which revealed amino acid residues involved in DNA recognition that are likely to be functionally important. We generated a series of hNANOG HD alanine substitution mutants based on the protein-DNA interaction and evolutionary conservation and determined their biological activities. Some mutant proteins were less stable, resulting in loss or decreased affinity for DNA binding. Overexpression of the orthologous mouse NANOG (mNANOG) mutants failed to maintain self-renewal of mouse embryonic stem cells without leukemia inhibitory factor. These results suggest that these residues are critical for NANOG transcriptional activity. Interestingly, one mutant, hNANOG L122A, conversely enhanced protein stability and DNA-binding affinity. The mNANOG L122A, when overexpressed in mouse embryonic stem cells, maintained their expression of self-renewal markers even when retinoic acid was added to forcibly drive differentiation. When overexpressed in epiblast stem cells or human induced pluripotent stem cells, the L122A mutants enhanced reprogramming into ground-state pluripotency. These findings demonstrate that structural and biophysical information on key transcriptional factors provides insights into the manipulation of stem cell behaviors and a framework for rational protein engineering.


Subject(s)
Cell Proliferation/genetics , Cellular Reprogramming/genetics , Homeodomain Proteins/genetics , Mutation , Pluripotent Stem Cells/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cells, Cultured , Crystallography, X-Ray , DNA/chemistry , DNA/genetics , DNA/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Germ Layers/cytology , Germ Layers/metabolism , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Nanog Homeobox Protein , Nucleic Acid Conformation , Pluripotent Stem Cells/cytology , Promoter Regions, Genetic/genetics , Protein Binding , Protein Structure, Tertiary , Transfection
4.
J Biol Chem ; 291(18): 9482-91, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-26940874

ABSTRACT

Clan CD cysteine peptidases, a structurally related group of peptidases that include mammalian caspases, exhibit a wide range of important functions, along with a variety of specificities and activation mechanisms. However, for the clostripain family (denoted C11), little is currently known. Here, we describe the first crystal structure of a C11 protein from the human gut bacterium, Parabacteroides merdae (PmC11), determined to 1.7-Å resolution. PmC11 is a monomeric cysteine peptidase that comprises an extended caspase-like α/ß/α sandwich and an unusual C-terminal domain. It shares core structural elements with clan CD cysteine peptidases but otherwise structurally differs from the other families in the clan. These studies also revealed a well ordered break in the polypeptide chain at Lys(147), resulting in a large conformational rearrangement close to the active site. Biochemical and kinetic analysis revealed Lys(147) to be an intramolecular processing site at which cleavage is required for full activation of the enzyme, suggesting an autoinhibitory mechanism for self-preservation. PmC11 has an acidic binding pocket and a preference for basic substrates, and accepts substrates with Arg and Lys in P1 and does not require Ca(2+) for activity. Collectively, these data provide insights into the mechanism and activity of PmC11 and a detailed framework for studies on C11 peptidases from other phylogenetic kingdoms.


Subject(s)
Bacterial Proteins/chemistry , Bacteroidaceae/enzymology , Cysteine Proteases/chemistry , Gastrointestinal Microbiome , Crystallography, X-Ray , Humans , Protein Structure, Secondary , Protein Structure, Tertiary
5.
PLoS Biol ; 12(10): e1001979, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25349992

ABSTRACT

One of the simplest organisms to divide asymmetrically is the bacterium Caulobacter crescentus. The DivL pseudo-histidine kinase, positioned at one cell pole, regulates cell-fate by controlling the activation of the global transcription factor CtrA via an interaction with the response regulator (RR) DivK. DivL uniquely contains a tyrosine at the histidine phosphorylation site, and can achieve these regulatory functions in vivo without kinase activity. Determination of the DivL crystal structure and biochemical analysis of wild-type and site-specific DivL mutants revealed that the DivL PAS domains regulate binding specificity for DivK∼P over DivK, which is modulated by an allosteric intramolecular interaction between adjacent domains. We discovered that DivL's catalytic domains have been repurposed as a phosphospecific RR input sensor, thereby reversing the flow of information observed in conventional histidine kinase (HK)-RR systems and coupling a complex network of signaling proteins for cell-fate regulation.


Subject(s)
Bacterial Proteins/metabolism , Caulobacter crescentus/metabolism , Protein Kinases/metabolism , Cell Cycle , Dimerization , Histidine Kinase , Protein Binding , Protein Structure, Tertiary
6.
Proc Natl Acad Sci U S A ; 111(42): 15054-9, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25288771

ABSTRACT

The signaling phosphatidylinositol lipids PI(4,5)P2 (PIP2) and PI(3,4,5)P3 (PIP3) bind nuclear receptor 5A family (NR5As), but their regulatory mechanisms remain unknown. Here, the crystal structures of human NR5A1 (steroidogenic factor-1, SF-1) ligand binding domain (LBD) bound to PIP2 and PIP3 show the lipid hydrophobic tails sequestered in the hormone pocket, as predicted. However, unlike classic nuclear receptor hormones, the phosphoinositide head groups are fully solvent-exposed and complete the LBD fold by organizing the receptor architecture at the hormone pocket entrance. The highest affinity phosphoinositide ligand PIP3 stabilizes the coactivator binding groove and increases coactivator peptide recruitment. This receptor-ligand topology defines a previously unidentified regulatory protein-lipid surface on SF-1 with the phosphoinositide head group at its nexus and poised to interact with other proteins. This surface on SF-1 coincides with the predicted binding site of the corepressor DAX-1 (dosage-sensitive sex reversal, adrenal hypoplasia critical region on chromosome X), and importantly harbors missense mutations associated with human endocrine disorders. Our data provide the structural basis for this poorly understood cluster of human SF-1 mutations and demonstrates how signaling phosphoinositides function as regulatory ligands for NR5As.


Subject(s)
Phosphatidylinositols/chemistry , Steroidogenic Factor 1/chemistry , Amino Acids/chemistry , Animals , Biological Transport , Cell Nucleus/metabolism , Chromatography , Computer Simulation , Crystallography, X-Ray , Electrons , Humans , Ligands , Lipids/chemistry , Mice , Models, Molecular , Molecular Conformation , Mutation , Mutation, Missense , Peptides/chemistry , Signal Transduction , Solvents/chemistry , Surface Plasmon Resonance , Surface Properties , Temperature , Water/chemistry
7.
J Struct Biol ; 192(3): 342-348, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26416531

ABSTRACT

The nuclear receptor LRH-1 (Liver Receptor Homolog-1, NR5A2) is a transcription factor that regulates gene expression programs critical for many aspects of metabolism and reproduction. Although LRH-1 is able to bind phospholipids, it is still considered an orphan nuclear receptor (NR) with an unknown regulatory hormone. Our prior cellular and structural studies demonstrated that the signaling phosphatidylinositols PI(4,5)P2 (PIP2) and PI(3,4,5)P3 (PIP3) bind and regulate SF-1 (Steroidogenic Factor-1, NR5A1), a close homolog of LRH-1. Here, we describe the crystal structure of human LRH-1 ligand binding domain (LBD) bound by PIP3 - the first phospholipid with a head group endogenous to mammals. We show that the phospholipid hormone binds LRH-1 with high affinity, stabilizing the receptor LBD. While the hydrophobic PIP3 tails (C16/C16) are buried inside the LRH-1 ligand binding pocket, the negatively charged PIP3 head group is presented on the receptor surface, similar to the phosphatidylinositol binding mode observed in the PIP3-SF-1 structure. Thus, data presented in this work reinforce our earlier findings demonstrating that signaling phosphatidylinositols regulate the NR5A receptors LRH-1 and SF-1.


Subject(s)
Phosphatidylinositols/chemistry , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/ultrastructure , Steroidogenic Factor 1/ultrastructure , Binding Sites/physiology , Crystallography, X-Ray , DAX-1 Orphan Nuclear Receptor/chemistry , Humans , Models, Molecular , Protein Binding/physiology , Protein Structure, Tertiary , Steroidogenic Factor 1/chemistry
8.
J Biol Chem ; 288(23): 16789-16799, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23572527

ABSTRACT

DUF2233, a domain of unknown function (DUF), is present in many bacterial and several viral proteins and was also identified in the mammalian transmembrane glycoprotein N-acetylglucosamine-1-phosphodiester α-N-acetylglucosaminidase ("uncovering enzyme" (UCE)). We report the crystal structure of BACOVA_00430, a 315-residue protein from the human gut bacterium Bacteroides ovatus that is the first structural representative of the DUF2233 protein family. A notable feature of this structure is the presence of a surface cavity that is populated by residues that are highly conserved across the entire family. The crystal structure was used to model the luminal portion of human UCE (hUCE), which is involved in targeting of lysosomal enzymes. Mutational analysis of several residues in a highly conserved surface cavity of hUCE revealed that they are essential for function. The bacterial enzyme (BACOVA_00430) has ∼1% of the catalytic activity of hUCE toward the substrate GlcNAc-P-mannose, the precursor of the Man-6-P lysosomal targeting signal. GlcNAc-1-P is a poor substrate for both enzymes. We conclude that, for at least a subset of proteins in this family, DUF2233 functions as a phosphodiester glycosidase.


Subject(s)
Bacterial Proteins/chemistry , Bacteroides/enzymology , Phosphoric Diester Hydrolases/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Humans , Mutagenesis , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Structural Homology, Protein
9.
Proteins ; 82(1): 164-70, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23852666

ABSTRACT

PF10014 is a novel family of 2-oxyglutarate-Fe(2+) -dependent dioxygenases that are involved in biosynthesis of antibiotics and regulation of biofilm formation, likely by catalyzing hydroxylation of free amino acids or other related ligands. The crystal structure of a PF10014 member from Methylibium petroleiphilum at 1.9 Å resolution shows strong structural similarity to cupin dioxygenases in overall fold and active site, despite very remote homology. However, one of the ß-strands of the cupin catalytic core is replaced by a loop that displays conformational isomerism that likely regulates the active site.


Subject(s)
Catalytic Domain/genetics , Comamonadaceae/enzymology , Conserved Sequence/genetics , Dioxygenases/chemistry , Models, Molecular , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Crystallization , DNA Primers/genetics , Dioxygenases/genetics , Molecular Sequence Data , Sequence Analysis, DNA
10.
Proteins ; 82(6): 1086-92, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24174223

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen commonly found in humans and other organisms and is an important cause of infection especially in patients with compromised immune defense mechanisms. The PA3611 gene of P. aeruginosa PAO1 encodes a secreted protein of unknown function, which has been recently classified into a small Pseudomonas-specific protein family called DUF4146. As part of our effort to extend structural coverage of novel protein space and provide a structure-based functional insight into new protein families, we report the crystal structure of PA3611, the first structural representative of the DUF4146 protein family.


Subject(s)
Bacterial Proteins/chemistry , Pseudomonas aeruginosa , Amino Acid Sequence , Conserved Sequence , Crystallography, X-Ray , Models, Molecular , Protein Structure, Secondary , Quorum Sensing
11.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 10): 2640-51, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25286848

ABSTRACT

The crystal structure of arabinose-5-phosphate isomerase (API) from Bacteroides fragilis (bfAPI) was determined at 1.7 Šresolution and was found to be a tetramer of a single-domain sugar isomerase (SIS) with an endogenous ligand, CMP-Kdo (cytidine 5'-monophosphate-3-deoxy-D-manno-oct-2-ulosonate), bound at the active site. API catalyzes the reversible isomerization of D-ribulose 5-phosphate to D-arabinose 5-phosphate in the first step of the Kdo biosynthetic pathway. Interestingly, the bound CMP-Kdo is neither the substrate nor the product of the reaction catalyzed by API, but corresponds to the end product in the Kdo biosynthetic pathway and presumably acts as a feedback inhibitor for bfAPI. The active site of each monomer is located in a surface cleft at the tetramer interface between three monomers and consists of His79 and His186 from two different adjacent monomers and a Ser/Thr-rich region, all of which are highly conserved across APIs. Structure and sequence analyses indicate that His79 and His186 may play important catalytic roles in the isomerization reaction. CMP-Kdo mimetics could therefore serve as potent and specific inhibitors of API and provide broad protection against many different bacterial infections.


Subject(s)
Aldose-Ketose Isomerases/chemistry , Aldose-Ketose Isomerases/metabolism , Bacteroides fragilis/chemistry , Aldose-Ketose Isomerases/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Cytidine Monophosphate/analogs & derivatives , Cytidine Monophosphate/chemistry , Histidine/chemistry , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid , Sugar Acids/chemistry
12.
J Bacteriol ; 195(24): 5555-66, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24123814

ABSTRACT

Approximately 50% of cell wall peptidoglycan in Gram-negative bacteria is recycled with each generation. The primary substrates used for peptidoglycan biosynthesis and recycling in the cytoplasm are GlcNAc-MurNAc(anhydro)-tetrapeptide and its degradation product, the free tetrapeptide. This complex process involves ∼15 proteins, among which the cytoplasmic enzyme ld-carboxypeptidase A (LdcA) catabolizes the bond between the last two l- and d-amino acid residues in the tetrapeptide to form the tripeptide, which is then utilized as a substrate by murein peptide ligase (Mpl). LdcA has been proposed as an antibacterial target. The crystal structure of Novosphingobium aromaticivorans DSM 12444 LdcA (NaLdcA) was determined at 1.89-Šresolution. The enzyme was biochemically characterized and its interactions with the substrate modeled, identifying residues potentially involved in substrate binding. Unaccounted electron density at the dimer interface in the crystal suggested a potential site for disrupting protein-protein interactions should a dimer be required to perform its function in bacteria. Our analysis extends the identification of functional residues to several other homologs, which include enzymes from bacteria that are involved in hydrocarbon degradation and destruction of coral reefs. The NaLdcA crystal structure provides an alternate system for investigating the structure-function relationships of LdcA and increases the structural coverage of the protagonists in bacterial cell wall recycling.


Subject(s)
Carboxypeptidases/chemistry , Carboxypeptidases/metabolism , Peptidoglycan/metabolism , Sphingomonadaceae/enzymology , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Multimerization
13.
Mol Microbiol ; 83(4): 712-27, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22211578

ABSTRACT

The tad (tight adherence) locus encodes a protein translocation system that produces a novel variant of type IV pili. The pilus assembly protein TadZ (called CpaE in Caulobacter crescentus) is ubiquitous in tad loci, but is absent in other type IV pilus biogenesis systems. The crystal structure of TadZ from Eubacterium rectale (ErTadZ), in complex with ATP and Mg(2+) , was determined to 2.1 Å resolution. ErTadZ contains an atypical ATPase domain with a variant of a deviant Walker-A motif that retains ATP binding capacity while displaying only low intrinsic ATPase activity. The bound ATP plays an important role in dimerization of ErTadZ. The N-terminal atypical receiver domain resembles the canonical receiver domain of response regulators, but has a degenerate, stripped-down 'active site'. Homology modelling of the N-terminal atypical receiver domain of CpaE indicates that it has a conserved protein-protein binding surface similar to that of the polar localization module of the social mobility protein FrzS, suggesting a similar function. Our structural results also suggest that TadZ localizes to the pole through the atypical receiver domain during an early stage of pili biogenesis, and functions as a hub for recruiting other pili components, thus providing insights into the Tad pilus assembly process.


Subject(s)
Bacterial Proteins/chemistry , Eubacterium/genetics , Membrane Transport Proteins/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Bacterial Proteins/metabolism , Crystallography, X-Ray , Fimbriae, Bacterial/metabolism , Magnesium/chemistry , Magnesium/metabolism , Membrane Transport Proteins/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Protein Multimerization , Protein Structure, Tertiary
14.
J Bacteriol ; 194(11): 2987-99, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22467785

ABSTRACT

MtfA of Escherichia coli (formerly YeeI) was previously identified as a regulator of the phosphoenolpyruvate (PEP)-dependent:glucose phosphotransferase system. MtfA homolog proteins are highly conserved, especially among beta- and gammaproteobacteria. We determined the crystal structures of the full-length MtfA apoenzyme from Klebsiella pneumoniae and its complex with zinc (holoenzyme) at 2.2 and 1.95 Å, respectively. MtfA contains a conserved H(149)E(150)XXH(153)+E(212)+Y(205) metallopeptidase motif. The presence of zinc in the active site induces significant conformational changes in the region around Tyr205 compared to the conformation of the apoenzyme. Additionally, the zinc-bound MtfA structure is in a self-inhibitory conformation where a region that was disordered in the unliganded structure is now observed in the active site and a nonproductive state of the enzyme is formed. MtfA is related to the catalytic domain of the anthrax lethal factor and the Mop protein involved in the virulence of Vibrio cholerae, with conservation in both overall structure and in the residues around the active site. These results clearly provide support for MtfA as a prototypical zinc metallopeptidase (gluzincin clan).


Subject(s)
Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Klebsiella pneumoniae/enzymology , Metalloproteases/chemistry , Metalloproteases/metabolism , Zinc/metabolism , Amino Acid Motifs , Amino Acid Sequence , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Catalytic Domain , Crystallography, X-Ray , Klebsiella pneumoniae/chemistry , Klebsiella pneumoniae/genetics , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Metalloproteases/genetics , Models, Molecular , Molecular Sequence Data , Protein Binding , Sequence Alignment
15.
J Biol Chem ; 286(14): 12450-60, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21247902

ABSTRACT

Considerable attention has recently been paid to the N-Myc downstream-regulated gene (NDRG) family because of its potential as a tumor suppressor in many human cancers. Primary amino acid sequence information suggests that the NDRG family proteins may belong to the α/ß-hydrolase (ABH) superfamily; however, their functional role has not yet been determined. Here, we present the crystal structures of the human and mouse NDRG2 proteins determined at 2.0 and 1.7 Å resolution, respectively. Both NDRG2 proteins show remarkable structural similarity to the ABH superfamily, despite limited sequence similarity. Structural analysis suggests that NDRG2 is a nonenzymatic member of the ABH superfamily, because it lacks the catalytic signature residues and has an occluded substrate-binding site. Several conserved structural features suggest NDRG may be involved in molecular interactions. Mutagenesis data based on the structural analysis support a crucial role for helix α6 in the suppression of TCF/ß-catenin signaling in the tumorigenesis of human colorectal cancer, via a molecular interaction.


Subject(s)
Crystallography, X-Ray/methods , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line, Tumor , HEK293 Cells , Humans , Immunoprecipitation , Molecular Sequence Data , Protein Structure, Secondary , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
16.
Proteins ; 80(6): 1545-59, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22411095

ABSTRACT

TM0077 from Thermotoga maritima is a member of the carbohydrate esterase family 7 and is active on a variety of acetylated compounds, including cephalosporin C. TM0077 esterase activity is confined to short-chain acyl esters (C2-C3), and is optimal around 100°C and pH 7.5. The positional specificity of TM0077 was investigated using 4-nitrophenyl-ß-D-xylopyranoside monoacetates as substrates in a ß-xylosidase-coupled assay. TM0077 hydrolyzes acetate at positions 2, 3, and 4 with equal efficiency. No activity was detected on xylan or acetylated xylan, which implies that TM0077 is an acetyl esterase and not an acetyl xylan esterase as currently annotated. Selenomethionine-substituted and native structures of TM0077 were determined at 2.1 and 2.5 Å resolution, respectively, revealing a classic α/ß-hydrolase fold. TM0077 assembles into a doughnut-shaped hexamer with small tunnels on either side leading to an inner cavity, which contains the six catalytic centers. Structures of TM0077 with covalently bound phenylmethylsulfonyl fluoride and paraoxon were determined to 2.4 and 2.1 Å, respectively, and confirmed that both inhibitors bind covalently to the catalytic serine (Ser188). Upon binding of inhibitor, the catalytic serine adopts an altered conformation, as observed in other esterase and lipases, and supports a previously proposed catalytic mechanism in which Ser hydroxyl rotation prevents reversal of the reaction and allows access of a water molecule for completion of the reaction.


Subject(s)
Acetylesterase/chemistry , Thermotoga maritima/enzymology , Acetylesterase/antagonists & inhibitors , Acetylesterase/metabolism , Catalytic Domain , Computer Simulation , Crystallography, X-Ray , Enzyme Stability , Hot Temperature , Hydrogen-Ion Concentration , Models, Molecular , Protein Conformation , Reproducibility of Results , Serine/chemistry , Serine/metabolism
17.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 4): 391-403, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22505259

ABSTRACT

Phasing by molecular replacement remains difficult for targets that are far from the search model or in situations where the crystal diffracts only weakly or to low resolution. Here, the process of determining and refining the structure of Cgl1109, a putative succinyl-diaminopimelate desuccinylase from Corynebacterium glutamicum, at ∼3 Šresolution is described using a combination of homology modeling with MODELLER, molecular-replacement phasing with Phaser, deformable elastic network (DEN) refinement and automated model building using AutoBuild in a semi-automated fashion, followed by final refinement cycles with phenix.refine and Coot. This difficult molecular-replacement case illustrates the power of including DEN restraints derived from a starting model to guide the movements of the model during refinement. The resulting improved model phases provide better starting points for automated model building and produce more significant difference peaks in anomalous difference Fourier maps to locate anomalous scatterers than does standard refinement. This example also illustrates a current limitation of automated procedures that require manual adjustment of local sequence misalignments between the homology model and the target sequence.


Subject(s)
Amidohydrolases/analysis , Automation, Laboratory/methods , Corynebacterium glutamicum/enzymology , Crystallography, X-Ray/methods , Software , Amidohydrolases/chemistry , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Structural Homology, Protein
18.
PLoS Biol ; 7(9): e1000205, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19787035

ABSTRACT

The genome projects have unearthed an enormous diversity of genes of unknown function that are still awaiting biological and biochemical characterization. These genes, as most others, can be grouped into families based on sequence similarity. The PFAM database currently contains over 2,200 such families, referred to as domains of unknown function (DUF). In a coordinated effort, the four large-scale centers of the NIH Protein Structure Initiative have determined the first three-dimensional structures for more than 250 of these DUF families. Analysis of the first 248 reveals that about two thirds of the DUF families likely represent very divergent branches of already known and well-characterized families, which allows hypotheses to be formulated about their biological function. The remainder can be formally categorized as new folds, although about one third of these show significant substructure similarity to previously characterized folds. These results infer that, despite the enormous increase in the number and the diversity of new genes being uncovered, the fold space of the proteins they encode is gradually becoming saturated. The previously unexplored sectors of the protein universe appear to be primarily shaped by extreme diversification of known protein families, which then enables organisms to evolve new functions and adapt to particular niches and habitats. Notwithstanding, these DUF families still constitute the richest source for discovery of the remaining protein folds and topologies.


Subject(s)
Proteins/chemistry , Animals , Databases, Protein , Humans , Models, Molecular , Multigene Family , Protein Structure, Secondary , Protein Structure, Tertiary , Structural Homology, Protein , Time Factors
19.
Proteins ; 79(7): 2146-60, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21538547

ABSTRACT

The crystal structures of an unliganded and adenosine 5'-monophosphate (AMP) bound, metal-dependent phosphoesterase (YP_910028.1) from Bifidobacterium adolescentis are reported at 2.4 and 1.94 Å, respectively. Functional characterization of this enzyme was guided by computational analysis and then confirmed by experiment. The structure consists of a polymerase and histidinol phosphatase (PHP, Pfam: PF02811) domain with a second domain (residues 105-178) inserted in the middle of the PHP sequence. The insert domain functions in binding AMP, but the precise function and substrate specificity of this domain are unknown. Initial bioinformatics analyses yielded multiple potential functional leads, with most of them suggesting DNA polymerase or DNA replication activity. Phylogenetic analysis indicated a potential DNA polymerase function that was somewhat supported by global structural comparisons identifying the closest structural match to the alpha subunit of DNA polymerase III. However, several other functional predictions, including phosphoesterase, could not be excluded. Theoretical microscopic anomalous titration curve shapes, a computational method for the prediction of active sites from protein 3D structures, identified potential reactive residues in YP_910028.1. Further analysis of the predicted active site and local comparison with its closest structure matches strongly suggested phosphoesterase activity, which was confirmed experimentally. Primer extension assays on both normal and mismatched DNA show neither extension nor degradation and provide evidence that YP_910028.1 has neither DNA polymerase activity nor DNA-proofreading activity. These results suggest that many of the sequence neighbors previously annotated as having DNA polymerase activity may actually be misannotated.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bifidobacterium/enzymology , Esterases/chemistry , Esterases/metabolism , 4-Nitrophenylphosphatase/chemistry , 4-Nitrophenylphosphatase/metabolism , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Amino Acid Sequence , Binding Sites , Catalytic Domain , Computer Simulation , Crystallography , DNA Polymerase III/chemistry , DNA Polymerase III/metabolism , Histidinol-Phosphatase/chemistry , Histidinol-Phosphatase/metabolism , Models, Molecular , Molecular Sequence Data , Phylogeny , Reproducibility of Results , Structure-Activity Relationship
20.
Acta Crystallogr D Biol Crystallogr ; 67(Pt 4): 368-75, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21460455

ABSTRACT

The Joint Center for Structural Genomics (JCSG), one of four large-scale structure-determination centers funded by the US Protein Structure Initiative (PSI) through the National Institute for General Medical Sciences, has been operating an automated distributed structure-solution pipeline, Xsolve, for well over half a decade. During PSI-2, Xsolve solved, traced and partially refined 90% of the JCSG's nearly 770 MAD/SAD structures at an average resolution of about 2 Šwithout human intervention. Xsolve executes many well established publicly available crystallography software programs in parallel on a commodity Linux cluster, resulting in multiple traces for any given target. Additional software programs have been developed and integrated into Xsolve to further minimize human effort in structure refinement. Consensus-Modeler exploits complementarities in traces from Xsolve to compute a single optimal model for manual refinement. Xpleo is a powerful robotics-inspired algorithm to build missing fragments and qFit automatically identifies and fits alternate conformations.


Subject(s)
Crystallography, X-Ray/methods , Proteins/analysis , Software Design , Algorithms , Automation, Laboratory , Models, Molecular , Protein Structure, Tertiary , Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL