Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Cell ; 177(2): 272-285.e16, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30853216

ABSTRACT

Proper brain function requires high-precision neuronal expansion and wiring, processes controlled by the transmembrane Roundabout (Robo) receptor family and their Slit ligands. Despite their great importance, the molecular mechanism by which Robos' switch from "off" to "on" states remains unclear. Here, we report a 3.6 Å crystal structure of the intact human Robo2 ectodomain (domains D1-8). We demonstrate that Robo cis dimerization via D4 is conserved through hRobo1, 2, and 3 and the C. elegans homolog SAX-3 and is essential for SAX-3 function in vivo. The structure reveals two levels of auto-inhibition that prevent premature activation: (1) cis blocking of the D4 dimerization interface and (2) trans interactions between opposing Robo receptors that fasten the D4-blocked conformation. Complementary experiments in mouse primary neurons and C. elegans support the auto-inhibition model. These results suggest that Slit stimulation primarily drives the release of Robo auto-inhibition required for dimerization and activation.


Subject(s)
Receptors, Immunologic/metabolism , Receptors, Immunologic/ultrastructure , Animals , Axons/metabolism , COS Cells , Caenorhabditis elegans/metabolism , Carrier Proteins , Chlorocebus aethiops , HEK293 Cells , Humans , Mice , Mice, Inbred ICR , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Primary Cell Culture , Signal Transduction , Roundabout Proteins
2.
EMBO Rep ; 24(12): e56920, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37988244

ABSTRACT

Ufmylation plays a crucial role in various cellular processes including DNA damage response, protein translation, and ER homeostasis. To date, little is known about how the enzymes responsible for ufmylation coordinate their action. Here, we study the details of UFL1 (E3) activity, its binding to UFC1 (E2), and its relation to UBA5 (E1), using a combination of structural modeling, X-ray crystallography, NMR, and biochemical assays. Guided by Alphafold2 models, we generate an active UFL1 fusion construct that includes its partner DDRGK1 and solve the crystal structure of this critical interaction. This fusion construct also unveiled the importance of the UFL1 N-terminal helix for binding to UFC1. The binding site suggested by our UFL1-UFC1 model reveals a conserved interface, and competition between UFL1 and UBA5 for binding to UFC1. This competition changes in the favor of UFL1 following UFM1 charging of UFC1. Altogether, our study reveals a novel, terminal helix-mediated regulatory mechanism, which coordinates the cascade of E1-E2-E3-mediated transfer of UFM1 to its substrate and provides new leads to target this modification.


Subject(s)
Binding Sites , Crystallography, X-Ray
3.
Nucleic Acids Res ; 51(15): 8237-8254, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37378428

ABSTRACT

Specificity in protein-DNA recognition arises from the synergy of several factors that stem from the structural and chemical signatures encoded within the targeted DNA molecule. Here, we deciphered the nature of the interactions driving DNA recognition and binding by the bacterial transcription factor PdxR, a member of the MocR family responsible for the regulation of pyridoxal 5'-phosphate (PLP) biosynthesis. Single particle cryo-EM performed on the PLP-PdxR bound to its target DNA enabled the isolation of three conformers of the complex, which may be considered as snapshots of the binding process. Moreover, the resolution of an apo-PdxR crystallographic structure provided a detailed description of the transition of the effector domain to the holo-PdxR form triggered by the binding of the PLP effector molecule. Binding analyses of mutated DNA sequences using both wild type and PdxR variants revealed a central role of electrostatic interactions and of the intrinsic asymmetric bending of the DNA in allosterically guiding the holo-PdxR-DNA recognition process, from the first encounter through the fully bound state. Our results detail the structure and dynamics of the PdxR-DNA complex, clarifying the mechanism governing the DNA-binding mode of the holo-PdxR and the regulation features of the MocR family of transcription factors.


Subject(s)
Bacterial Proteins , Transcription Factors , Bacteria/genetics , Bacterial Proteins/metabolism , DNA/metabolism , Protein Binding , Pyridoxal Phosphate/metabolism , Transcription Factors/metabolism , Bacillus clausii/genetics
4.
J Biol Chem ; 299(4): 103033, 2023 04.
Article in English | MEDLINE | ID: mdl-36806680

ABSTRACT

N-acetyl-d-glucosamine (GlcNAc) is a major component of bacterial cell walls. Many organisms recycle GlcNAc from the cell wall or metabolize environmental GlcNAc. The first step in GlcNAc metabolism is phosphorylation to GlcNAc-6-phosphate. In bacteria, the ROK family kinase N-acetylglucosamine kinase (NagK) performs this activity. Although ROK kinases have been studied extensively, no ternary complex showing the two substrates has yet been observed. Here, we solved the structure of NagK from the human pathogen Plesiomonas shigelloides in complex with GlcNAc and the ATP analog AMP-PNP. Surprisingly, PsNagK showed distinct conformational changes associated with the binding of each substrate. Consistent with this, the enzyme showed a sequential random enzyme mechanism. This indicates that the enzyme acts as a coordinated unit responding to each interaction. Our molecular dynamics modeling of catalytic ion binding confirmed the location of the essential catalytic metal. Additionally, site-directed mutagenesis confirmed the catalytic base and that the metal-coordinating residue is essential. Together, this study provides the most comprehensive insight into the activity of a ROK kinase.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor) , Plesiomonas , Humans , Acetylglucosamine/metabolism , Glucosamine , Metals , Phosphotransferases (Alcohol Group Acceptor)/metabolism , rho-Associated Kinases , Plesiomonas/enzymology
5.
J Biol Chem ; 298(5): 101903, 2022 05.
Article in English | MEDLINE | ID: mdl-35398092

ABSTRACT

The sugars streptose and dihydrohydroxystreptose (DHHS) are unique to the bacteria Streptomyces griseus and Coxiella burnetii, respectively. Streptose forms the central moiety of the antibiotic streptomycin, while DHHS is found in the O-antigen of the zoonotic pathogen C. burnetii. Biosynthesis of these sugars has been proposed to follow a similar path to that of TDP-rhamnose, catalyzed by the enzymes RmlA, RmlB, RmlC, and RmlD, but the exact mechanism is unclear. Streptose and DHHS biosynthesis unusually requires a ring contraction step that could be performed by orthologs of RmlC or RmlD. Genome sequencing of S. griseus and C. burnetii has identified StrM and CBU1838 proteins as RmlC orthologs in these respective species. Here, we demonstrate that both enzymes can perform the RmlC 3'',5'' double epimerization activity necessary to support TDP-rhamnose biosynthesis in vivo. This is consistent with the ring contraction step being performed on a double epimerized substrate. We further demonstrate that proton exchange is faster at the 3''-position than the 5''-position, in contrast to a previously studied ortholog. We additionally solved the crystal structures of CBU1838 and StrM in complex with TDP and show that they form an active site highly similar to those of the previously characterized enzymes RmlC, EvaD, and ChmJ. These results support the hypothesis that streptose and DHHS are biosynthesized using the TDP pathway and that an RmlD paralog most likely performs ring contraction following double epimerization. This work will support the elucidation of the full pathways for biosynthesis of these unique sugars.


Subject(s)
Antigens, Bacterial/biosynthesis , Carbohydrate Epimerases , Coxiella burnetii/enzymology , Streptomyces griseus/enzymology , Carbohydrate Epimerases/genetics , Nucleoside Diphosphate Sugars/biosynthesis , Thymine Nucleotides/biosynthesis
6.
Faraday Discuss ; 240(0): 303-311, 2022 11 08.
Article in English | MEDLINE | ID: mdl-35929538

ABSTRACT

Helical reconstruction is the method of choice for obtaining 3D structures of filaments from electron cryo-microscopy (cryoEM) projections. This approach relies on applying helical symmetry parameters deduced from Fourier-Bessel or real space analysis, such as sub-tomogram averaging. While helical reconstruction continues to provide invaluable structural insights into filaments, its inherent dependence on imposing a pre-defined helical symmetry can also introduce bias. The applied helical symmetry produces structures that are infinitely straight along the filament's axis and can average out biologically important heterogeneities. Here, we describe a simple workflow aimed at overcoming these drawbacks in order to provide truer representations of filamentous structures.


Subject(s)
Image Processing, Computer-Assisted , Cryoelectron Microscopy/methods , Image Processing, Computer-Assisted/methods
7.
Proc Natl Acad Sci U S A ; 116(35): 17251-17260, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31395737

ABSTRACT

Microsomal triglyceride transfer protein (MTP) plays an essential role in lipid metabolism, especially in the biogenesis of very low-density lipoproteins and chylomicrons via the transfer of neutral lipids and the assembly of apoB-containing lipoproteins. Our understanding of the molecular mechanisms of MTP has been hindered by a lack of structural information of this heterodimeric complex comprising an MTPα subunit and a protein disulfide isomerase (PDI) ß-subunit. The structure of MTP presented here gives important insights into the potential mechanisms of action of this essential lipid transfer molecule, structure-based rationale for previously reported disease-causing mutations, and a means for rational drug design against cardiovascular disease and obesity. In contrast to the previously reported structure of lipovitellin, which has a funnel-like lipid-binding cavity, the lipid-binding site is encompassed in a ß-sandwich formed by 2 ß-sheets from the C-terminal domain of MTPα. The lipid-binding cavity of MTPα is large enough to accommodate a single lipid. PDI independently has a major role in oxidative protein folding in the endoplasmic reticulum. Comparison of the mechanism of MTPα binding by PDI with previously published structures gives insights into large protein substrate binding by PDI and suggests that the previous structures of human PDI represent the "substrate-bound" and "free" states rather than differences arising from redox state.


Subject(s)
Carrier Proteins/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Protein Conformation, beta-Strand
8.
Plant J ; 103(3): 1215-1232, 2020 08.
Article in English | MEDLINE | ID: mdl-32369638

ABSTRACT

The Arabidopsis thaliana BON1 gene product is a member of the evolutionary conserved eukaryotic calcium-dependent membrane-binding protein family. The copine protein is composed of two C2 domains (C2A and C2B) followed by a vWA domain. The BON1 protein is localized on the plasma membrane, and is known to suppress the expression of immune receptor genes and to positively regulate stomatal closure. The first structure of this protein family has been determined to 2.5-Å resolution and shows the structural features of the three conserved domains C2A, C2B and vWA. The structure reveals the third Ca2+ -binding region in C2A domain is longer than classical C2 domains and a novel Ca2+ binding site in the vWA domain. The structure of BON1 bound to Mn2+ is also presented. The binding of the C2 domains to phospholipid (PSF) has been modeled and provides an insight into the lipid-binding mechanism of the copine proteins. Furthermore, the selectivity of the separate C2A and C2B domains and intact BON1 to bind to different phospholipids has been investigated, and we demonstrated that BON1 could mediate aggregation of liposomes in response to Ca2+ . These studies have formed the basis of further investigations into the important role that the copine proteins play in vivo.


Subject(s)
Arabidopsis Proteins/chemistry , Calcium-Binding Proteins/chemistry , Carrier Proteins/metabolism , Membrane Proteins/chemistry , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Calcium/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Membrane/metabolism , Liposomes/metabolism , Manganese/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Conformation , Sequence Alignment
9.
J Struct Biol ; 212(3): 107657, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33148525

ABSTRACT

The removal of carbon dioxide from the waste streams of industrial processes is a major challenge for creation of a sustainable circular economy. This makes the synthesis of formate from CO2 by NAD+ dependent formate dehydrogenases (FDHs) an attractive process for this purpose. The efficiency of this reaction is however low and to achieve a viable industrial process an optimised engineered enzyme needs to be developed. In order to understand the detailed enzymatic mechanism of catalysis structures of different cofactor and substrate complexes of the FDH from the thermophilic filamentous fungus, Chaetomium thermophilum have been determined to 1.2-1.3 Å resolution. The substrate formate is shown to be held by four hydrogen bonds in the FDH catalytic site within the ternary complex with substrate and NAD+and a secondary formate binding site is observed in crystals soaked with substrate. Water molecules are excluded from the FDH catalytic site when the substrate is bound. The angle between the plane of the NAD+ cofactor pyridine ring and the plane of the formate molecule is around 27°. Additionally, structures of a FDH mutant enzyme, N120C, in complex with the reduced form of the cofactor have also been determined both in the presence and absence of formate bound at the secondary site. These structures provide further understanding of the catalytic mechanism of this fungal enzyme.


Subject(s)
Chaetomium/chemistry , Formate Dehydrogenases/chemistry , Formates/chemistry , NAD/chemistry , Binding Sites/physiology , Carbon Dioxide/chemistry , Carbon Dioxide/metabolism , Catalysis , Catalytic Domain/physiology , Chaetomium/metabolism , Formate Dehydrogenases/metabolism , Formates/metabolism , Hydrogen Bonding , NAD/metabolism , Protein Domains/physiology , Protein Engineering/methods
10.
Proc Natl Acad Sci U S A ; 114(52): E11151-E11160, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29229824

ABSTRACT

Microbial pathogens employ sophisticated virulence strategies to cause infections in humans. The intracellular pathogen Legionella pneumophila encodes RidL to hijack the host scaffold protein VPS29, a component of retromer and retriever complexes critical for endosomal cargo recycling. Here, we determined the crystal structure of L. pneumophila RidL in complex with the human VPS29-VPS35 retromer subcomplex. A hairpin loop protruding from RidL inserts into a conserved pocket on VPS29 that is also used by cellular ligands, such as Tre-2/Bub2/Cdc16 domain family member 5 (TBC1D5) and VPS9-ankyrin repeat protein for VPS29 binding. Consistent with the idea of molecular mimicry in protein interactions, RidL outcompeted TBC1D5 for binding to VPS29. Furthermore, the interaction of RidL with retromer did not interfere with retromer dimerization but was essential for association of RidL with retromer-coated vacuolar and tubular endosomes. Our work thus provides structural and mechanistic evidence into how RidL is targeted to endosomal membranes.


Subject(s)
Bacterial Proteins/chemistry , Legionella pneumophila/chemistry , Protein Multimerization , Virulence Factors/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Legionella pneumophila/genetics , Legionella pneumophila/metabolism , Legionella pneumophila/pathogenicity , Protein Domains , Protein Structure, Quaternary , Protein Structure, Secondary , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
11.
J Biol Chem ; 293(50): 19429-19440, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30337369

ABSTRACT

Toxin-antitoxin (TA) systems are present in many bacteria and play important roles in bacterial growth, physiology, and pathogenicity. Those that are best studied are the type II TA systems, in which both toxins and antitoxins are proteins. The HicAB system is one of the prototypic TA systems, found in many bacterial species. Complex interactions between the protein toxin (HicA), the protein antitoxin (HicB), and the DNA upstream of the encoding genes regulate the activity of this system, but few structural details are available about how HicA destabilizes the HicB-DNA complex. Here, we determined the X-ray structures of HicB and the HicAB complex to 1.8 and 2.5 Å resolution, respectively, and characterized their DNA interactions. This revealed that HicB forms a tetramer and HicA and HicB form a heterooctameric complex that involves structural reorganization of the C-terminal (DNA-binding) region of HicB. Our observations indicated that HicA has a profound impact on binding of HicB to DNA sequences upstream of hicAB in a stoichiometric-dependent way. At low ratios of HicA:HicB, there was no effect on DNA binding, but at higher ratios, the affinity for DNA declined cooperatively, driving dissociation of the HicA:HicB:DNA complex. These results reveal the structural mechanisms by which HicA de-represses the HicB-DNA complex.


Subject(s)
Antitoxins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , DNA/metabolism , Toxins, Biological/chemistry , Toxins, Biological/metabolism , Antitoxins/chemistry , Bacterial Proteins/genetics , Burkholderia pseudomallei , Models, Molecular , Operon/genetics , Protein Binding , Protein Conformation , Toxins, Biological/genetics
12.
Mol Biol Evol ; 34(6): 1463-1478, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28333212

ABSTRACT

In the development of the human brain, human-specific genes are considered to play key roles, conferring its unique advantages and vulnerabilities. At the time of Homo lineage divergence from Australopithecus, SRGAP2C gradually emerged through a process of serial duplications and mutagenesis from ancestral SRGAP2A (3.4-2.4 Ma). Remarkably, ectopic expression of SRGAP2C endows cultured mouse brain cells, with human-like characteristics, specifically, increased dendritic spine length and density. To understand the molecular mechanisms underlying this change in neuronal morphology, we determined the structure of SRGAP2A and studied the interplay between SRGAP2A and SRGAP2C. We found that: 1) SRGAP2A homo-dimerizes through a large interface that includes an F-BAR domain, a newly identified F-BAR extension (Fx), and RhoGAP-SH3 domains. 2) SRGAP2A has an unusual inverse geometry, enabling associations with lamellipodia and dendritic spine heads in vivo, and scaffolding of membrane protrusions in cell culture. 3) As a result of the initial partial duplication event (∼3.4 Ma), SRGAP2C carries a defective Fx-domain that severely compromises its solubility and membrane-scaffolding ability. Consistently, SRGAP2A:SRAGP2C hetero-dimers form, but are insoluble, inhibiting SRGAP2A activity. 4) Inactivation of SRGAP2A is sensitive to the level of hetero-dimerization with SRGAP2C. 5) The primal form of SRGAP2C (P-SRGAP2C, existing between ∼3.4 and 2.4 Ma) is less effective in hetero-dimerizing with SRGAP2A than the modern SRGAP2C, which carries several substitutions (from ∼2.4 Ma). Thus, the genetic mutagenesis phase contributed to modulation of SRGAP2A's inhibition of neuronal expansion, by introducing and improving the formation of inactive SRGAP2A:SRGAP2C hetero-dimers, indicating a stepwise involvement of SRGAP2C in human evolutionary history.


Subject(s)
GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , Brain/growth & development , Brain/physiology , Cell Movement/genetics , Cell Movement/physiology , Crystallography, X-Ray/methods , Dendritic Spines , Evolution, Molecular , GTPase-Activating Proteins/metabolism , Humans , Neurons/metabolism , Protein Structure, Tertiary/genetics , Pseudopodia , Structure-Activity Relationship
13.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 11): 2344-53, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26527149

ABSTRACT

The three-dimensional structures of the native enzyme and the FMN complex of the overexpressed form of the oxygenating component of the type II Baeyer-Villiger 3,6-diketocamphane monooxygenase have been determined to 1.9 Å resolution. The structure of this dimeric FMN-dependent enzyme, which is encoded on the large CAM plasmid of Pseudomonas putida, has been solved by a combination of multiple anomalous dispersion from a bromine crystal soak and molecular replacement using a bacterial luciferase model. The orientation of the isoalloxazine ring of the FMN cofactor in the active site of this TIM-barrel fold enzyme differs significantly from that previously observed in enzymes of the bacterial luciferase-like superfamily. The Ala77 residue is in a cis conformation and forms a ß-bulge at the C-terminus of ß-strand 3, which is a feature observed in many proteins of this superfamily.


Subject(s)
Bacterial Proteins/chemistry , Oxygenases/chemistry , Pseudomonas putida/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , FMN Reductase/metabolism , Flavin Mononucleotide/metabolism , Models, Molecular , Molecular Sequence Data , Oxygenases/genetics , Oxygenases/metabolism , Plasmids/genetics , Protein Conformation , Protein Folding , Pseudomonas putida/genetics , Pseudomonas putida/metabolism , Sequence Alignment
14.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 9): 2430-43, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25195756

ABSTRACT

The presence of pseudo-symmetry in a macromolecular crystal and its interplay with twinning may lead to an incorrect space-group (SG) assignment. Moreover, if the pseudo-symmetry is very close to an exact crystallographic symmetry, the structure can be solved and partially refined in the wrong SG. Typically, in such incorrectly determined structures all or some of the pseudo-symmetry operations are, in effect, taken for crystallographic symmetry operations and vice versa. A mistake only becomes apparent when the R(free) ceases to decrease below 0.39 and further model rebuilding and refinement cannot improve the refinement statistics. If pseudo-symmetry includes pseudo-translation, the uncertainty in SG assignment may be associated with an incorrect choice of origin, as demonstrated by the series of examples provided here. The program Zanuda presented in this article was developed for the automation of SG validation. Zanuda runs a series of refinements in SGs compatible with the observed unit-cell parameters and chooses the model with the highest symmetry SG from a subset of models that have the best refinement statistics.


Subject(s)
Macromolecular Substances/chemistry , Automation , Humans , Lectins, C-Type/chemistry , Molecular Structure , Protein Conformation , Receptors, Cell Surface/chemistry
15.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 10): 2607-18, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25286845

ABSTRACT

Carbonic anhydrase enzymes catalyse the reversible hydration of carbon dioxide to bicarbonate. A thermophilic Thermovibrio ammonificans α-carbonic anhydrase (TaCA) has been expressed in Escherichia coli and structurally and biochemically characterized. The crystal structure of TaCA has been determined in its native form and in two complexes with bound inhibitors. The tetrameric enzyme is stabilized by a unique core in the centre of the molecule formed by two intersubunit disulfides and a single lysine residue from each monomer that is involved in intersubunit ionic interactions. The structure of this core protects the intersubunit disulfides from reduction, whereas the conserved intrasubunit disulfides are not formed in the reducing environment of the E. coli host cytosol. When oxidized to mimic the environment of the periplasmic space, TaCA has increased thermostability, retaining 90% activity after incubation at 70°C for 1 h, making it a good candidate for industrial carbon-dioxide capture. The reduction of all TaCA cysteines resulted in dissociation of the tetrameric molecule into monomers with lower activity and reduced thermostability. Unlike other characterized α-carbonic anhydrases, TaCA does not display esterase activity towards p-nitrophenyl acetate, which appears to result from the increased rigidity of its protein scaffold.


Subject(s)
Carbonic Anhydrases/chemistry , Carbonic Anhydrases/metabolism , Acetazolamide/chemistry , Bacteria/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carbon Dioxide/metabolism , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/metabolism , Carbonic Anhydrases/genetics , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Disulfides/chemistry , Enzyme Stability , Kinetics , Models, Molecular , Nitrophenols/metabolism , Protein Conformation , Sulfanilamide , Sulfanilamides/chemistry , Temperature
16.
Nat Commun ; 15(1): 5050, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877033

ABSTRACT

Amongst the major types of archaeal filaments, several have been shown to closely resemble bacterial homologues of the Type IV pili (T4P). Within Sulfolobales, member species encode for three types of T4P, namely the archaellum, the UV-inducible pilus system (Ups) and the archaeal adhesive pilus (Aap). Whereas the archaellum functions primarily in swimming motility, and the Ups in UV-induced cell aggregation and DNA-exchange, the Aap plays an important role in adhesion and twitching motility. Here, we present a cryoEM structure of the Aap of the archaeal model organism Sulfolobus acidocaldarius. We identify the component subunit as AapB and find that while its structure follows the canonical T4P blueprint, it adopts three distinct conformations within the pilus. The tri-conformer Aap structure that we describe challenges our current understanding of pilus structure and sheds new light on the principles of twitching motility.


Subject(s)
Cryoelectron Microscopy , Sulfolobus acidocaldarius , Sulfolobus acidocaldarius/metabolism , Sulfolobus acidocaldarius/genetics , Sulfolobus acidocaldarius/physiology , Archaeal Proteins/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Fimbriae, Bacterial/ultrastructure , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/physiology , Fimbriae, Bacterial/chemistry , Fimbriae Proteins/metabolism , Fimbriae Proteins/chemistry , Fimbriae Proteins/genetics , Models, Molecular
17.
Elife ; 132024 Jan 22.
Article in English | MEDLINE | ID: mdl-38251732

ABSTRACT

Surface layers (S-layers) are resilient two-dimensional protein lattices that encapsulate many bacteria and most archaea. In archaea, S-layers usually form the only structural component of the cell wall and thus act as the final frontier between the cell and its environment. Therefore, S-layers are crucial for supporting microbial life. Notwithstanding their importance, little is known about archaeal S-layers at the atomic level. Here, we combined single-particle cryo electron microscopy, cryo electron tomography, and Alphafold2 predictions to generate an atomic model of the two-component S-layer of Sulfolobus acidocaldarius. The outer component of this S-layer (SlaA) is a flexible, highly glycosylated, and stable protein. Together with the inner and membrane-bound component (SlaB), they assemble into a porous and interwoven lattice. We hypothesise that jackknife-like conformational changes in SlaA play important roles in S-layer assembly.


Subject(s)
Sulfolobus acidocaldarius , Sulfolobus acidocaldarius/metabolism , Archaea , Bacteria , Cell Wall
18.
Biochim Biophys Acta ; 1824(3): 422-32, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22197591

ABSTRACT

D-Serine dehydratase from Escherichia coli is a member of the ß-family (fold-type II) of the pyridoxal 5'-phosphate-dependent enzymes, catalyzing the conversion of D-serine to pyruvate and ammonia. The crystal structure of monomeric D-serine dehydratase has been solved to 1.97Å-resolution for an orthorhombic data set by molecular replacement. In addition, the structure was refined in a monoclinic data set to 1.55Å resolution. The structure of DSD reveals a larger pyridoxal 5'-phosphate-binding domain and a smaller domain. The active site of DSD is very similar to those of the other members of the ß-family. Lys118 forms the Schiff base to PLP, the cofactor phosphate group is liganded to a tetraglycine cluster Gly279-Gly283, and the 3-hydroxyl group of PLP is liganded to Asn170 and N1 to Thr424, respectively. In the closed conformation the movement of the small domain blocks the entrance to active site of DSD. The domain movement plays an important role in the formation of the substrate recognition site and the catalysis of the enzyme. Modeling of D-serine into the active site of DSD suggests that the hydroxyl group of D-serine is coordinated to the carboxyl group of Asp238. The carboxyl oxygen of D-serine is coordinated to the hydroxyl group of Ser167 and the amide group of Leu171 (O1), whereas the O2 of the carboxyl group of D-serine is hydrogen-bonded to the hydroxyl group of Ser167 and the amide group of Thr168. A catalytic mechanism very similar to that proposed for L-serine dehydratase is discussed.


Subject(s)
Bacterial Proteins/chemistry , Escherichia coli/chemistry , Hydro-Lyases/chemistry , Pyridoxal Phosphate/chemistry , Amino Acid Sequence , Amino Acids , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Escherichia coli/enzymology , Hydro-Lyases/isolation & purification , Hydro-Lyases/metabolism , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Pyridoxal Phosphate/metabolism
19.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 4): 564-76, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23519665

ABSTRACT

The crystal structures and inhibitor complexes of two industrially important ω-aminotransferase enzymes from Pseudomonas aeruginosa and Chromobacterium violaceum have been determined in order to understand the differences in their substrate specificity. The two enzymes share 30% sequence identity and use the same amino acceptor, pyruvate; however, the Pseudomonas enzyme shows activity towards the amino donor ß-alanine, whilst the Chromobacterium enzyme does not. Both enzymes show activity towards S-α-methylbenzylamine (MBA), with the Chromobacterium enzyme having a broader substrate range. The crystal structure of the P. aeruginosa enzyme has been solved in the holo form and with the inhibitor gabaculine bound. The C. violaceum enzyme has been solved in the apo and holo forms and with gabaculine bound. The structures of the holo forms of both enzymes are quite similar. There is little conformational difference observed between the inhibitor complex and the holoenzyme for the P. aeruginosa aminotransferase. In comparison, the crystal structure of the C. violaceum gabaculine complex shows significant structural rearrangements from the structures of both the apo and holo forms of the enzyme. It appears that the different rigidity of the protein scaffold contributes to the substrate specificity observed for the two ω-aminotransferases.


Subject(s)
Chromobacterium/enzymology , Pseudomonas aeruginosa/enzymology , Transaminases/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Crystallography, X-Ray , Cyclohexanecarboxylic Acids/chemistry , Cyclohexanecarboxylic Acids/metabolism , Holoenzymes/chemistry , Holoenzymes/metabolism , Substrate Specificity , Transaminases/antagonists & inhibitors , Transaminases/metabolism
20.
Front Microbiol ; 14: 1267570, 2023.
Article in English | MEDLINE | ID: mdl-38045033

ABSTRACT

The enzyme cyclic di-phosphoglycerate synthetase that is involved in the production of the osmolyte cyclic 2,3-diphosphoglycerate has been studied both biochemically and structurally. Cyclic 2,3-diphosphoglycerate is found exclusively in the hyperthermophilic archaeal methanogens, such as Methanothermus fervidus, Methanopyrus kandleri, and Methanothermobacter thermoautotrophicus. Its presence increases the thermostability of archaeal proteins and protects the DNA against oxidative damage caused by hydroxyl radicals. The cyclic 2,3-diphosphoglycerate synthetase enzyme has been crystallized and its structure solved to 1.7 Šresolution by experimental phasing. It has also been crystallized in complex with its substrate 2,3 diphosphoglycerate and the co-factor ADP and this structure has been solved to 2.2 Šresolution. The enzyme structure has two domains, the core domain shares some structural similarity with other NTP-dependent enzymes. A significant proportion of the structure, including a 127 amino acid N-terminal domain, has no structural similarity to other known enzyme structures. The structure of the complex shows a large conformational change that occurs in the enzyme during catalytic turnover. The reaction involves the transfer of the γ-phosphate group from ATP to the substrate 2,3 -diphosphoglycerate and the subsequent SN2 attack to form a phosphoanhydride. This results in the production of the unusual extremolyte cyclic 2,3 -diphosphoglycerate which has important industrial applications.

SELECTION OF CITATIONS
SEARCH DETAIL