Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143220

ABSTRACT

Guanidine is a chemically stable nitrogen compound that is excreted in human urine and is widely used in manufacturing of plastics, as a flame retardant and as a component of propellants, and is well known as a protein denaturant in biochemistry1-3. Guanidine occurs widely in nature and is used by several microorganisms as a nitrogen source, but microorganisms growing on guanidine as the only substrate have not yet been identified. Here we show that the complete ammonia oxidizer (comammox) Nitrospira inopinata and probably most other comammox microorganisms can grow on guanidine as the sole source of energy, reductant and nitrogen. Proteomics, enzyme kinetics and the crystal structure of a N. inopinata guanidinase homologue demonstrated that it is a bona fide guanidinase. Incubation experiments with comammox-containing agricultural soil and wastewater treatment plant microbiomes suggested that guanidine serves as substrate for nitrification in the environment. The identification of guanidine as a growth substrate for comammox shows an unexpected niche of these globally important nitrifiers and offers opportunities for their isolation.

2.
Cell ; 159(6): 1447-60, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25433700

ABSTRACT

The spectrin superfamily of proteins plays key roles in assembling the actin cytoskeleton in various cell types, crosslinks actin filaments, and acts as scaffolds for the assembly of large protein complexes involved in structural integrity and mechanosensation, as well as cell signaling. α-actinins in particular are the major actin crosslinkers in muscle Z-disks, focal adhesions, and actin stress fibers. We report a complete high-resolution structure of the 200 kDa α-actinin-2 dimer from striated muscle and explore its functional implications on the biochemical and cellular level. The structure provides insight into the phosphoinositide-based mechanism controlling its interaction with sarcomeric proteins such as titin, lays a foundation for studying the impact of pathogenic mutations at molecular resolution, and is likely to be broadly relevant for the regulation of spectrin-like proteins.


Subject(s)
Actinin/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Muscle, Skeletal/chemistry , Protein Structure, Tertiary , Scattering, Small Angle , Sequence Alignment , X-Ray Diffraction
3.
PLoS Biol ; 21(6): e3002187, 2023 06.
Article in English | MEDLINE | ID: mdl-37384774

ABSTRACT

In the past 2 decades, structural biology has transformed from a single technique used on single proteins to a multimodal integrative approach. Recently, protein structure prediction algorithms have opened new avenues to address challenging biological questions.


Subject(s)
Molecular Biology , Proteins , Algorithms , Computational Biology/methods
4.
J Cell Sci ; 136(20)2023 10 15.
Article in English | MEDLINE | ID: mdl-37737012

ABSTRACT

All endocytosis and exocytosis in the African trypanosome Trypanosoma brucei occurs at a single subdomain of the plasma membrane. This subdomain, the flagellar pocket, is a small vase-shaped invagination containing the root of the single flagellum of the cell. Several cytoskeleton-associated multiprotein complexes are coiled around the neck of the flagellar pocket on its cytoplasmic face. One of these, the hook complex, was proposed to affect macromolecule entry into the flagellar pocket lumen. In previous work, knockdown of T. brucei (Tb)MORN1, a hook complex component, resulted in larger cargo being unable to enter the flagellar pocket. In this study, the hook complex component TbSmee1 was characterised in bloodstream form T. brucei and found to be essential for cell viability. TbSmee1 knockdown resulted in flagellar pocket enlargement and impaired access to the flagellar pocket membrane by surface-bound cargo, similar to depletion of TbMORN1. Unexpectedly, inhibition of endocytosis by knockdown of clathrin phenocopied TbSmee1 knockdown, suggesting that endocytic activity itself is a prerequisite for the entry of surface-bound cargo into the flagellar pocket.


Subject(s)
Trypanosoma brucei brucei , Trypanosoma , Trypanosoma/metabolism , Endocytosis/physiology , Trypanosoma brucei brucei/metabolism , Cell Membrane/metabolism , Cilia/metabolism , Flagella/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
5.
PLoS Biol ; 19(4): e3001148, 2021 04.
Article in English | MEDLINE | ID: mdl-33844684

ABSTRACT

Sarcomeres, the basic contractile units of striated muscle cells, contain arrays of thin (actin) and thick (myosin) filaments that slide past each other during contraction. The Ig-like domain-containing protein myotilin provides structural integrity to Z-discs-the boundaries between adjacent sarcomeres. Myotilin binds to Z-disc components, including F-actin and α-actinin-2, but the molecular mechanism of binding and implications of these interactions on Z-disc integrity are still elusive. To illuminate them, we used a combination of small-angle X-ray scattering, cross-linking mass spectrometry, and biochemical and molecular biophysics approaches. We discovered that myotilin displays conformational ensembles in solution. We generated a structural model of the F-actin:myotilin complex that revealed how myotilin interacts with and stabilizes F-actin via its Ig-like domains and flanking regions. Mutant myotilin designed with impaired F-actin binding showed increased dynamics in cells. Structural analyses and competition assays uncovered that myotilin displaces tropomyosin from F-actin. Our findings suggest a novel role of myotilin as a co-organizer of Z-disc assembly and advance our mechanistic understanding of myotilin's structural role in Z-discs.


Subject(s)
Actins/metabolism , Protein Multimerization , Sarcomeres/metabolism , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actins/chemistry , Actins/genetics , Animals , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Humans , Mice , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Contraction/genetics , Muscle, Skeletal/metabolism , Protein Binding/genetics , Protein Interaction Domains and Motifs/genetics , Protein Multimerization/genetics , Sarcomeres/genetics , Tropomyosin/chemistry , Tropomyosin/genetics , Tropomyosin/metabolism
6.
Proc Natl Acad Sci U S A ; 117(1): 317-327, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31852826

ABSTRACT

Host-pathogen interactions are central to understanding microbial pathogenesis. The staphylococcal pore-forming cytotoxins hijack important immune molecules but little is known about the underlying molecular mechanisms of cytotoxin-receptor interaction and host specificity. Here we report the structures of a staphylococcal pore-forming cytotoxin, leukocidin GH (LukGH), in complex with its receptor (the α-I domain of complement receptor 3, CD11b-I), both for the human and murine homologs. We observe 2 binding interfaces, on the LukG and the LukH protomers, and show that human CD11b-I induces LukGH oligomerization in solution. LukGH binds murine CD11b-I weakly and is inactive toward murine neutrophils. Using a LukGH variant engineered to bind mouse CD11b-I, we demonstrate that cytolytic activity does not only require binding but also receptor-dependent oligomerization. Our studies provide an unprecedented insight into bicomponent leukocidin-host receptor interaction, enabling the development of antitoxin approaches and improved animal models to explore these approaches.


Subject(s)
Bacterial Proteins/metabolism , CD11b Antigen/metabolism , Leukocidins/metabolism , Macrophage-1 Antigen/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Bacterial Proteins/immunology , Bacterial Proteins/ultrastructure , CD11b Antigen/immunology , CD11b Antigen/ultrastructure , Cell Line , Cell Membrane/metabolism , Crystallography, X-Ray , Humans , Leukocidins/immunology , Macrophage-1 Antigen/immunology , Macrophage-1 Antigen/ultrastructure , Mice , Models, Molecular , Neutrophils/cytology , Neutrophils/immunology , Neutrophils/metabolism , Protein Domains/immunology , Protein Multimerization/immunology , Rabbits , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Species Specificity , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity
7.
Proc Natl Acad Sci U S A ; 117(26): 14926-14935, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32554495

ABSTRACT

Molecular ON-switches in which a chemical compound induces protein-protein interactions can allow cellular function to be controlled with small molecules. ON-switches based on clinically applicable compounds and human proteins would greatly facilitate their therapeutic use. Here, we developed an ON-switch system in which the human retinol binding protein 4 (hRBP4) of the lipocalin family interacts with engineered hRBP4 binders in a small molecule-dependent manner. Two different protein scaffolds were engineered to bind to hRBP4 when loaded with the orally available small molecule A1120. The crystal structure of an assembled ON-switch shows that the engineered binder specifically recognizes the conformational changes induced by A1120 in two loop regions of hRBP4. We demonstrate that this conformation-specific ON-switch is highly dependent on the presence of A1120, as demonstrated by an ∼500-fold increase in affinity upon addition of the small molecule drug. Furthermore, the ON-switch successfully regulated the activity of primary human CAR T cells in vitro. We anticipate that lipocalin-based ON-switches have the potential to be broadly applied for the safe pharmacological control of cellular therapeutics.


Subject(s)
Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Cell Line , Cytokines/immunology , Humans , Lipocalins/genetics , Lipocalins/immunology , Molecular Conformation , Piperidines/chemistry , Piperidines/pharmacology , Receptors, Chimeric Antigen/genetics , Retinol-Binding Proteins, Plasma/genetics , Retinol-Binding Proteins, Plasma/immunology , T-Lymphocytes/drug effects
8.
Proc Natl Acad Sci U S A ; 117(36): 22101-22112, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32848067

ABSTRACT

The actin cytoskeleton, a dynamic network of actin filaments and associated F-actin-binding proteins, is fundamentally important in eukaryotes. α-Actinins are major F-actin bundlers that are inhibited by Ca2+ in nonmuscle cells. Here we report the mechanism of Ca2+-mediated regulation of Entamoeba histolytica α-actinin-2 (EhActn2) with features expected for the common ancestor of Entamoeba and higher eukaryotic α-actinins. Crystal structures of Ca2+-free and Ca2+-bound EhActn2 reveal a calmodulin-like domain (CaMD) uniquely inserted within the rod domain. Integrative studies reveal an exceptionally high affinity of the EhActn2 CaMD for Ca2+, binding of which can only be regulated in the presence of physiological concentrations of Mg2+ Ca2+ binding triggers an increase in protein multidomain rigidity, reducing conformational flexibility of F-actin-binding domains via interdomain cross-talk and consequently inhibiting F-actin bundling. In vivo studies uncover that EhActn2 plays an important role in phagocytic cup formation and might constitute a new drug target for amoebic dysentery.


Subject(s)
Actinin/metabolism , Calcium/pharmacology , Entamoeba histolytica/metabolism , Actinin/chemistry , Actinin/genetics , Catalytic Domain , Entamoeba histolytica/genetics , Gene Expression Regulation , Models, Molecular , Protein Conformation , Protein Domains
9.
Biochemistry ; 60(8): 621-634, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33586945

ABSTRACT

Chlorite dismutases (Clds) are heme b-containing oxidoreductases that can decompose chlorite to chloride and molecular oxygen. They are divided in two clades that differ in oligomerization, subunit architecture, and the hydrogen-bonding network of the distal catalytic arginine, which is proposed to switch between two conformations during turnover. To understand the impact of the conformational dynamics of this basic amino acid on heme coordination, structure, and catalysis, Cld from Cyanothece sp. PCC7425 was used as a model enzyme. As typical for a clade 2 Cld, its distal arginine 127 is hydrogen-bonded to glutamine 74. The latter has been exchanged with either glutamate (Q74E) to arrest R127 in a salt bridge or valine (Q74V) that mirrors the setting in clade 1 Clds. We present the X-ray crystal structures of Q74V and Q74E and demonstrate the pH-induced changes in the environment and coordination of the heme iron by ultraviolet-visible, circular dichroism, and electron paramagnetic resonance spectroscopies as well as differential scanning calorimetry. The conformational dynamics of R127 is shown to have a significant role in heme coordination during the alkaline transition and in the thermal stability of the heme cavity, whereas its impact on the catalytic efficiency of chlorite degradation is relatively small. The findings are discussed with respect to (i) the flexible loop connecting the N-terminal and C-terminal ferredoxin-like domains, which differs in clade 1 and clade 2 Clds and carries Q74 in clade 2 proteins, and (ii) the proposed role(s) of the arginine in catalysis.


Subject(s)
Arginine/metabolism , Chlorides/metabolism , Cyanothece/enzymology , Heme/metabolism , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Temperature , Arginine/chemistry , Catalysis , Enzyme Stability , Heme/chemistry , Hydrogen Bonding , Kinetics , Models, Molecular
10.
J Biol Chem ; 295(39): 13488-13501, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32723869

ABSTRACT

Since the advent of protein crystallography, atomic-level macromolecular structures have provided a basis to understand biological function. Enzymologists use detailed structural insights on ligand coordination, interatomic distances, and positioning of catalytic amino acids to rationalize the underlying electronic reaction mechanisms. Often the proteins in question catalyze redox reactions using metal cofactors that are explicitly intertwined with their function. In these cases, the exact nature of the coordination sphere and the oxidation state of the metal is of utmost importance. Unfortunately, the redox-active nature of metal cofactors makes them especially susceptible to photoreduction, meaning that information obtained by photoreducing X-ray sources about the environment of the cofactor is the least trustworthy part of the structure. In this work we directly compare the kinetics of photoreduction of six different heme protein crystal species by X-ray radiation. We show that a dose of ∼40 kilograys already yields 50% ferrous iron in a heme protein crystal. We also demonstrate that the kinetics of photoreduction are completely independent from variables unique to the different samples tested. The photoreduction-induced structural rearrangements around the metal cofactors have to be considered when biochemical data of ferric proteins are rationalized by constraints derived from crystal structures of reduced enzymes.


Subject(s)
Ferric Compounds/chemistry , Heme/chemistry , Metalloproteins/chemistry , Metmyoglobin/chemistry , Peroxidase/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Horses , Kinetics , Klebsiella pneumoniae/enzymology , Models, Molecular , Oxidation-Reduction , Peroxidase/metabolism , Photochemical Processes , X-Rays
11.
Proc Natl Acad Sci U S A ; 114(5): 1015-1020, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096424

ABSTRACT

Stable anchoring of titin within the muscle Z-disk is essential for preserving muscle integrity during passive stretching. One of the main candidates for anchoring titin in the Z-disk is the actin cross-linker α-actinin. The calmodulin-like domain of α-actinin binds to the Z-repeats of titin. However, the mechanical and kinetic properties of this important interaction are still unknown. Here, we use a dual-beam optical tweezers assay to study the mechanics of this interaction at the single-molecule level. A single interaction of α-actinin and titin turns out to be surprisingly weak if force is applied. Depending on the direction of force application, the unbinding forces can more than triple. Our results suggest a model where multiple α-actinin/Z-repeat interactions cooperate to ensure long-term stable titin anchoring while allowing the individual components to exchange dynamically.


Subject(s)
Actinin/metabolism , Connectin/metabolism , Actinin/chemistry , Amino Acid Sequence , Animals , Connectin/chemistry , Cysteine/chemistry , Cystine/chemistry , Humans , Mutagenesis, Site-Directed , Optical Tweezers , Protein Domains , Protein Interaction Mapping , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Repetitive Sequences, Amino Acid , Sarcomeres/chemistry , Sarcomeres/ultrastructure , Stress, Mechanical
12.
J Biol Chem ; 293(4): 1330-1345, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29242189

ABSTRACT

Oxidation of halides and thiocyanate by heme peroxidases to antimicrobial oxidants is an important cornerstone in the innate immune system of mammals. Interestingly, phylogenetic and physiological studies suggest that homologous peroxidases are already present in mycetozoan eukaryotes such as Dictyostelium discoideum This social amoeba kills bacteria via phagocytosis for nutrient acquisition at its single-cell stage and for antibacterial defense at its multicellular stages. Here, we demonstrate that peroxidase A from D. discoideum (DdPoxA) is a stable, monomeric, glycosylated, and secreted heme peroxidase with homology to mammalian peroxidases. The first crystal structure (2.5 Å resolution) of a mycetozoan peroxidase of this superfamily shows the presence of a post-translationally-modified heme with one single covalent ester bond between the 1-methyl heme substituent and Glu-236. The metalloprotein follows the halogenation cycle, whereby compound I oxidizes iodide and thiocyanate at high rates (>108 m-1 s-1) and bromide at very low rates. It is demonstrated that DdPoxA is up-regulated and likely secreted at late multicellular development stages of D. discoideum when migrating slugs differentiate into fruiting bodies that contain persistent spores on top of a cellular stalk. Expression of DdPoxA is shown to restrict bacterial contamination of fruiting bodies. Structure and function of DdPoxA are compared with evolutionary-related mammalian peroxidases in the context of non-specific immune defense.


Subject(s)
Dictyostelium/enzymology , Heme Oxygenase (Decyclizing)/chemistry , Heme Oxygenase (Decyclizing)/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Catalysis , Dictyostelium/genetics , Heme Oxygenase (Decyclizing)/genetics , Oxidation-Reduction , Protozoan Proteins/genetics , Structure-Activity Relationship
13.
J Biol Chem ; 293(38): 14823-14838, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30072383

ABSTRACT

Dye-decolorizing peroxidases (DyPs) represent the most recently classified hydrogen peroxide-dependent heme peroxidase family. Although widely distributed with more than 5000 annotated genes and hailed for their biotechnological potential, detailed biochemical characterization of their reaction mechanism remains limited. Here, we present the high-resolution crystal structures of WT B-class DyP from the pathogenic bacterium Klebsiella pneumoniae (KpDyP) (1.6 Å) and the variants D143A (1.3 Å), R232A (1.9 Å), and D143A/R232A (1.1 Å). We demonstrate the impact of elimination of the DyP-typical, distal residues Asp-143 and Arg-232 on (i) the spectral and redox properties, (ii) the kinetics of heterolytic cleavage of hydrogen peroxide, (iii) the formation of the low-spin cyanide complex, and (iv) the stability and reactivity of an oxoiron(IV)porphyrin π-cation radical (Compound I). Structural and functional studies reveal that the distal aspartate is responsible for deprotonation of H2O2 and for the poor oxidation capacity of Compound I. Elimination of the distal arginine promotes a collapse of the distal heme cavity, including blocking of one access channel and a conformational change of the catalytic aspartate. We also provide evidence of formation of an oxoiron(IV)-type Compound II in KpDyP with absorbance maxima at 418, 527, and 553 nm. In summary, a reaction mechanism of the peroxidase cycle of B-class DyPs is proposed. Our observations challenge the idea that peroxidase activity toward conventional aromatic substrates is related to the physiological roles of B-class DyPs.


Subject(s)
Arginine/metabolism , Aspartic Acid/metabolism , Coloring Agents/metabolism , Hydrogen Peroxide/metabolism , Peroxidases/metabolism , Amino Acid Substitution , Catalysis , Catalytic Domain , Circular Dichroism , Color , Crystallography, X-Ray , Dimerization , Enzyme Stability , Heme/chemistry , Hydrogen-Ion Concentration , Hydrolysis , Klebsiella pneumoniae/metabolism , Peroxidases/chemistry , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrophotometry, Ultraviolet
14.
Mol Biol Rep ; 46(3): 3101-3112, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30977085

ABSTRACT

Chronic overuse of common pharmaceuticals, e.g. acetaminophen (paracetamol), often leads to the development of acute liver failure (ALF). This study aimed to elucidate the effect of cultured mesenchymal stem cells (MSCs) proteome on the onset of liver damage and regeneration dynamics in animals with ALF induced by acetaminophen, to test the liver protective efficacy of MSCs proteome depending on the oxygen tension in cell culture, and to blueprint protein components responsible for the effect. Protein compositions prepared from MSCs cultured in mild hypoxic (5% and 10% O2) and normal (21% O2) conditions were used to treat ALF induced in mice by injection of acetaminophen. To test the effect of reduced oxygen tension in cell culture on resulting MSCs proteome content we applied a combination of high performance liquid chromatography and mass-spectrometry (LC-MS/MS) for the identification of proteins in lysates of MSCs cultured at different O2 levels. The treatment of acetaminophen-administered animals with proteins released from cultured MSCs resulted in the inhibition of inflammatory reactions in damaged liver; the area of hepatocyte necrosis being reduced in the first 24 h. Compositions obtained from MSCs cultured at lower O2 level were shown to be more potent than a composition prepared from normoxic cells. A comparative characterization of protein pattern and identification of individual components done by a cytokine assay and proteomics analysis of protein compositions revealed that even moderate hypoxia produces discrete changes in the expression of various subsets of proteins responsible for intracellular respiration and cell signaling. The application of proteins prepared from MSCs grown in vitro at reduced oxygen tension significantly accelerates healing process in damaged liver tissue. The proteomics data obtained for different preparations offer new information about the potential candidates in the MSCs protein repertoire sensitive to oxygen tension in culture medium, which can be involved in the generalized mechanisms the cells use to respond to acute liver failure.


Subject(s)
Acetaminophen/adverse effects , Chemical and Drug Induced Liver Injury/therapy , Culture Media, Conditioned/metabolism , Liver Failure, Acute/etiology , Liver Failure, Acute/therapy , Mesenchymal Stem Cells/metabolism , Proteome , Animals , Biomarkers , Biopsy , Cells, Cultured , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Disease Models, Animal , Hypoxia/metabolism , Liver Failure, Acute/pathology , Male , Mass Spectrometry , Mesenchymal Stem Cells/cytology , Mice , Oxygen Consumption , Protective Agents/metabolism , Protective Agents/pharmacology , Proteomics/methods
15.
Nucleic Acids Res ; 45(16): 9741-9759, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28934471

ABSTRACT

Poly(ADP-ribose) glycohydrolase (PARG) regulates cellular poly(ADP-ribose) (PAR) levels by rapidly cleaving glycosidic bonds between ADP-ribose units. PARG interacts with proliferating cell nuclear antigen (PCNA) and is strongly recruited to DNA damage sites in a PAR- and PCNA-dependent fashion. Here we identified PARG acetylation site K409 that is essential for its interaction with PCNA, its localization within replication foci and its recruitment to DNA damage sites. We found K409 to be part of a non-canonical PIP-box within the PARG disordered regulatory region. The previously identified putative N-terminal PIP-box does not bind PCNA directly but contributes to PARG localization within replication foci. X-ray structure and MD simulations reveal that the PARG non-canonical PIP-box binds PCNA in a manner similar to other canonical PIP-boxes and may represent a new type of PIP-box. While the binding of previously described PIP-boxes is based on hydrophobic interactions, PARG PIP-box binds PCNA via both stabilizing hydrophobic and fine-tuning electrostatic interactions. Our data explain the mechanism of PARG-PCNA interaction through a new PARG PIP-box that exhibits non-canonical sequence properties but a canonical mode of PCNA binding.


Subject(s)
Glycoside Hydrolases/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Acetylation , Calorimetry/methods , Chromatin/metabolism , Crystallography, X-Ray , DNA Damage , Fluorescence Resonance Energy Transfer , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/genetics , HeLa Cells , Humans , Immunoprecipitation , Lasers , Lysine/genetics , Lysine/metabolism , Molecular Dynamics Simulation , Proliferating Cell Nuclear Antigen/chemistry , Protein Conformation , S Phase/genetics , Static Electricity
16.
J Biol Chem ; 292(20): 8244-8261, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28348079

ABSTRACT

Myeloperoxidase (MPO) is synthesized by neutrophil and monocyte precursor cells and contributes to host defense by mediating microbial killing. Although several steps in MPO biosynthesis and processing have been elucidated, many questions remained, such as the structure-function relationship of monomeric unprocessed proMPO versus the mature dimeric MPO and the functional role of the propeptide. Here we have presented the first and high resolution (at 1.25 Å) crystal structure of proMPO and its solution structure obtained by small-angle X-ray scattering. Promyeloperoxidase hosts five occupied glycosylation sites and six intrachain cystine bridges with Cys-158 of the very flexible N-terminal propeptide being covalently linked to Cys-319 and thereby hindering homodimerization. Furthermore, the structure revealed (i) the binding site of proMPO-processing proconvertase, (ii) the structural motif for subsequent cleavage to the heavy and light chains of mature MPO protomers, and (iii) three covalent bonds between heme and the protein. Studies of the mutants C158A, C319A, and C158A/C319A demonstrated significant differences from the wild-type protein, including diminished enzymatic activity and prevention of export to the Golgi due to prolonged association with the chaperone calnexin. These structural and functional findings provide novel insights into MPO biosynthesis and processing.


Subject(s)
Enzyme Precursors , Peroxidase , Amino Acid Substitution , Calnexin/chemistry , Calnexin/genetics , Calnexin/metabolism , Crystallography, X-Ray , Enzyme Activation/physiology , Enzyme Precursors/biosynthesis , Enzyme Precursors/chemistry , Enzyme Precursors/genetics , Golgi Apparatus/enzymology , Golgi Apparatus/genetics , HEK293 Cells , Humans , K562 Cells , Mutation, Missense , Peroxidase/biosynthesis , Peroxidase/chemistry , Peroxidase/genetics , Protein Domains
17.
PLoS Pathog ; 12(12): e1006079, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27973613

ABSTRACT

Vaccinia virus interferes with early events of the activation pathway of the transcriptional factor NF-kB by binding to numerous host TIR-domain containing adaptor proteins. We have previously determined the X-ray structure of the A46 C-terminal domain; however, the structure and function of the A46 N-terminal domain and its relationship to the C-terminal domain have remained unclear. Here, we biophysically characterize residues 1-83 of the N-terminal domain of A46 and present the X-ray structure at 1.55 Å. Crystallographic phases were obtained by a recently developed ab initio method entitled ARCIMBOLDO_BORGES that employs tertiary structure libraries extracted from the Protein Data Bank; data analysis revealed an all ß-sheet structure. This is the first such structure solved by this method which should be applicable to any protein composed entirely of ß-sheets. The A46(1-83) structure itself is a ß-sandwich containing a co-purified molecule of myristic acid inside a hydrophobic pocket and represents a previously unknown lipid-binding fold. Mass spectrometry analysis confirmed the presence of long-chain fatty acids in both N-terminal and full-length A46; mutation of the hydrophobic pocket reduced the lipid content. Using a combination of high resolution X-ray structures of the N- and C-terminal domains and SAXS analysis of full-length protein A46(1-240), we present here a structural model of A46 in a tetrameric assembly. Integrating affinity measurements and structural data, we propose how A46 simultaneously interferes with several TIR-domain containing proteins to inhibit NF-κB activation and postulate that A46 employs a bipartite binding arrangement to sequester the host immune adaptors TRAM and MyD88.


Subject(s)
Vaccinia virus/chemistry , Vaccinia virus/metabolism , Viral Proteins/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Escherichia coli , HEK293 Cells , Humans , Mass Spectrometry , Models, Molecular , Protein Conformation, beta-Strand , Protein Structure, Quaternary , Scattering, Small Angle , Spectrometry, Mass, Electrospray Ionization , Viral Proteins/metabolism
18.
Cell Mol Life Sci ; 74(13): 2413-2438, 2017 07.
Article in English | MEDLINE | ID: mdl-28243699

ABSTRACT

The BAR domain is the eponymous domain of the "BAR-domain protein superfamily", a large and diverse set of mostly multi-domain proteins that play eminent roles at the membrane cytoskeleton interface. BAR domain homodimers are the functional units that peripherally associate with lipid membranes and are involved in membrane sculpting activities. Differences in their intrinsic curvatures and lipid-binding properties account for a large variety in membrane modulating properties. Membrane activities of BAR domains are further modified and regulated by intramolecular or inter-subunit domains, by intermolecular protein interactions, and by posttranslational modifications. Rather than providing detailed cell biological information on single members of this superfamily, this review focuses on biochemical, biophysical, and structural aspects and on recent findings that paradigmatically promote our understanding of processes driven and modulated by BAR domains.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Animals , Humans , Phosphorylation , Protein Binding , Protein Domains , Protein Interaction Mapping
19.
J Gen Virol ; 98(3): 471-485, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27959783

ABSTRACT

Protein phosphorylation has important regulatory functions in cell homeostasis and is tightly regulated by kinases and phosphatases. The tegument of human cytomegalovirus (CMV) contains not only several proteins reported to be extensively phosphorylated but also cellular protein phosphatases (PP1 and PP2A). To investigate this apparent inconsistency, we evaluated the phosphorylation status of the tegument proteins pUL32 and pp65 by enzymatic dephosphorylation and MS. Enzymatic dephosphorylation with bacterial λ phosphatase, but not with PP1, shifted the pUL32-specific signal on reducing SDS-PAGE from ~150 to ~148 kDa, a mass still much larger than the ~118 kDa obtained from our diffusion studies and from the calculated protein mass of ~113 kDa. Remarkably, inhibition of phosphatases through treatment with the phosphatase inhibitors calyculin A and okadaic acid resulted in a shift to ~190 or ~180 kDa, respectively, indicating that a considerable number of potential phosphorylated residues on pUL32 are not phosphorylated under normal conditions. MS revealed a general state of hypophosphorylation of CMV phosphoproteins with only 17 phosphorylated residues detected on pUL32 and 19 on pp65, respectively. Moreover, bioinformatics analysis shows that the C-terminal two-thirds of pUL32 are intrinsically disordered and that most phosphorylations map to this region. In conclusion, we show that important CMV tegument proteins are indeed phosphorylated, though to a lesser extent than previously reported, and the difference in mobility on SDS-PAGE and calculated mass of pUL32 may not be attributed to phosphorylation but more likely due to the partially intrinsically disordered nature of pUL32.


Subject(s)
Cytomegalovirus/metabolism , Phosphoproteins/metabolism , Viral Matrix Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Humans , Marine Toxins , Okadaic Acid/pharmacology , Oxazoles/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylation , Protein Processing, Post-Translational
20.
Nucleic Acids Res ; 43(1): 661-73, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25510494

ABSTRACT

In Gram-negative bacteria, the multi-domain protein S1 is essential for translation initiation, as it recruits the mRNA and facilitates its localization in the decoding centre. In sharp contrast to its functional importance, S1 is still lacking from the high-resolution structures available for Escherichia coli and Thermus thermophilus ribosomes and thus the molecular mechanism governing the S1-ribosome interaction has still remained elusive. Here, we present the structure of the N-terminal S1 domain D1 when bound to the ribosome at atomic resolution by using a combination of NMR, X-ray crystallography and cryo-electron microscopy. Together with biochemical assays, the structure reveals that S1 is anchored to the ribosome primarily via a stabilizing π-stacking interaction within the short but conserved N-terminal segment that is flexibly connected to domain D1. This interaction is further stabilized by salt bridges involving the zinc binding pocket of protein S2. Overall, this work provides one hitherto enigmatic piece in the 'ribosome puzzle', namely the detailed molecular insight into the topology of the S1-ribosome interface. Moreover, our data suggest novel mechanisms that have the potential to modulate protein synthesis in response to environmental cues by changing the affinity of S1 for the ribosome.


Subject(s)
Escherichia coli Proteins/chemistry , Ribosomal Proteins/chemistry , Ribosomes/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Models, Molecular , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , Ribosomal Proteins/metabolism , Ribosome Subunits, Small, Bacterial/metabolism , Ribosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL