Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 17(4): e1009477, 2021 04.
Article in English | MEDLINE | ID: mdl-33857257

ABSTRACT

The lack of efficient methods to control the major diseases of crops most important to agriculture leads to huge economic losses and seriously threatens global food security. Many of the most important microbial plant pathogens, including bacteria, fungi, and oomycetes, secrete necrosis- and ethylene-inducing peptide 1 (Nep1)-like proteins (NLPs), which critically contribute to the virulence and spread of the disease. NLPs are cytotoxic to eudicot plants, as they disturb the plant plasma membrane by binding to specific plant membrane sphingolipid receptors. Their pivotal role in plant infection and broad taxonomic distribution makes NLPs a promising target for the development of novel phytopharmaceutical compounds. To identify compounds that bind to NLPs from the oomycetes Pythium aphanidermatum and Phytophthora parasitica, a library of 587 small molecules, most of which are commercially unavailable, was screened by surface plasmon resonance. Importantly, compounds that exhibited the highest affinity to NLPs were also found to inhibit NLP-mediated necrosis in tobacco leaves and Phytophthora infestans growth on potato leaves. Saturation transfer difference-nuclear magnetic resonance and molecular modelling of the most promising compound, anthranilic acid derivative, confirmed stable binding to the NLP protein, which resulted in decreased necrotic activity and reduced ion leakage from tobacco leaves. We, therefore, confirmed that NLPs are an appealing target for the development of novel phytopharmaceutical agents and strategies, which aim to directly interfere with the function of these major microbial virulence factors. The compounds identified in this study represent lead structures for further optimization and antimicrobial product development.


Subject(s)
Phytophthora/pathogenicity , Plant Diseases/prevention & control , Pythium/pathogenicity , Solanum tuberosum/genetics , Molecular Dynamics Simulation , Necrosis , Phytophthora/genetics , Plant Diseases/parasitology , Plant Leaves/genetics , Plant Leaves/parasitology , Pythium/genetics , Solanum tuberosum/parasitology , Surface Plasmon Resonance , Nicotiana/genetics , Nicotiana/parasitology
2.
Sci Rep ; 11(1): 6572, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33753805

ABSTRACT

Aegerolysins are proteins produced by bacteria, fungi, plants and protozoa. The most studied fungal aegerolysins share a common property of interacting with membranes enriched with cholesterol in combination with either sphingomyelin or ceramide phosphorylethanolamine (CPE), major sphingolipids in the cell membranes of vertebrates and invertebrates, respectively. However, genome analyses show a particularly high frequency of aegerolysin genes in bacteria, including the pathogenic genera Pseudomonas and Vibrio; these are human pathogens of high clinical relevance and can thrive in a variety of other species. The knowledge on bacterial aegerolysin-lipid interactions is scarce. We show that Pseudomonas aeruginosa aegerolysin RahU interacts with CPE, but not with sphingomyelin-enriched artificial membranes, and that RahU interacts with the insect cell line producing CPE. We report crystal structures of RahU alone and in complex with tris(hydroxymethyl)aminomethane (Tris), which, like the phosphorylethanolamine head group of CPE, contains a primary amine. The RahU structures reveal that the two loops proximal to the amino terminus form a cavity that accommodates Tris, and that the flexibility of these two loops is important for this interaction. We show that Tris interferes with CPE-enriched membranes for binding to RahU, implying on the importance of the ligand cavity between the loops and its proximity in RahU membrane interaction. We further support this by studying the interaction of single amino acid substitution mutants of RahU with the CPE-enriched membranes. Our results thus represent a starting point for a better understanding of the role of P. aeruginosa RahU, and possibly other bacterial aegerolysins, in bacterial interactions with other organisms.


Subject(s)
Bacterial Proteins/chemistry , Ethanolamines/chemistry , Fungal Proteins/chemistry , Hemolysin Proteins/chemistry , Models, Molecular , Protein Conformation , Pseudomonas aeruginosa , Animals , Bacterial Proteins/metabolism , Ethanolamines/metabolism , Fungal Proteins/metabolism , Hemolysin Proteins/metabolism , Humans , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Pseudomonas aeruginosa/metabolism , Sf9 Cells , Structure-Activity Relationship
3.
FASEB J ; 34(8): 11068-11086, 2020 08.
Article in English | MEDLINE | ID: mdl-32648626

ABSTRACT

NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome is a multiprotein complex which forms within cells in response to various microbial and self-derived triggers. Mutations in the gene encoding NLRP3 cause rare cryopyrin-associated periodic syndromes (CAPS) and growing evidence links NLRP3 inflammasome to common diseases such as Alzheimer´s disease. In order to modulate different stages of NLRP3 inflammasome assembly nine peptides whose sequences correspond to segments of inflammasome components NLRP3 and apoptosis-associated speck-like protein containing a CARD (ASC) were selected. Five peptides inhibited IL-1ß release, caspase-1 activation and ASC oligomerization in response to soluble and particulate NLRP3 triggers. Modulatory peptides also attenuated IL-1ß maturation induced by constitutive CAPS-associated NLRP3 mutants. Peptide corresponding to H2-H3 segment of ASC pyrin domain selectively inhibited NLRP3 inflammasome by binding to NLRP3 pyrin domain in the micromolar range. The peptide had no effect on AIM2 and NLRC4 inflammasomes as well as NF-κB pathway. The peptide effectively dampened neutrophil infiltration in the silica-induced peritonitis and when equipped with Antennapedia or Angiopep-2 motifs crossed the blood-brain barrier in a mouse model. Our study demonstrates that peptides represent an important tool for targeting multiprotein inflammatory complexes and can serve as the basis for the development of novel anti-inflammatory strategies for neurodegeneration.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Peptides/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Blood-Brain Barrier/metabolism , Caspase 1/metabolism , Cells, Cultured , Cryopyrin-Associated Periodic Syndromes/metabolism , DNA-Binding Proteins/metabolism , Female , Inflammation/metabolism , Interleukin-1beta/metabolism , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Neutrophil Infiltration/physiology , Peritonitis/metabolism
4.
J Infect Dis ; 222(9): 1505-1516, 2020 10 01.
Article in English | MEDLINE | ID: mdl-31970394

ABSTRACT

BACKGROUND: Bacterial toxins disrupt plasma membrane integrity with multitudinous effects on host cells. The secreted pore-forming toxin listeriolysin O (LLO) of the intracellular pathogen Listeria monocytogenes promotes egress of the bacteria from vacuolar compartments into the host cytosol often without overt destruction of the infected cell. Intracellular LLO activity is tightly controlled by host factors including compartmental pH, redox, proteolytic, and proteostatic factors, and inhibited by cholesterol. METHODS: Combining infection studies of L. monocytogenes wild type and isogenic mutants together with biochemical studies with purified phospholipases, we investigate the effect of their enzymatic activities on LLO. RESULTS: Here, we show that phosphocholine (ChoP), a reaction product of the phosphatidylcholine-specific phospholipase C (PC-PLC) of L. monocytogenes, is a potent inhibitor of intra- and extracellular LLO activities. Binding of ChoP to LLO is redox-independent and leads to the inhibition of LLO-dependent induction of calcium flux, mitochondrial damage, and apoptosis. ChoP also inhibits the hemolytic activities of the related cholesterol-dependent cytolysins (CDC), pneumolysin and streptolysin. CONCLUSIONS: Our study uncovers a strategy used by L. monocytogenes to modulate cytotoxic LLO activity through the enzymatic activity of its PC-PLC. This mechanism appears to be widespread and also used by other CDC pore-forming toxin-producing bacteria.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Heat-Shock Proteins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Listeria monocytogenes/drug effects , Phosphorylcholine/pharmacology , Apoptosis , Calcium/metabolism , Caspase 3/metabolism , HeLa Cells , Humans , Listeria monocytogenes/enzymology , Listeria monocytogenes/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
5.
PLoS Pathog ; 15(9): e1007951, 2019 09.
Article in English | MEDLINE | ID: mdl-31479498

ABSTRACT

Necrosis and ethylene-inducing peptide 1 (Nep1)-like proteins (NLPs) are secreted by several phytopathogenic microorganisms. They trigger necrosis in various eudicot plants upon binding to plant sphingolipid glycosylinositol phosphorylceramides (GIPC). Interestingly, HaNLP3 from the obligate biotroph oomycete Hyaloperonospora arabidopsidis does not induce necrosis. We determined the crystal structure of HaNLP3 and showed that it adopts the NLP fold. However, the conformations of the loops surrounding the GIPC headgroup-binding cavity differ from those of cytotoxic Pythium aphanidermatum NLPPya. Essential dynamics extracted from µs-long molecular dynamics (MD) simulations reveals a limited conformational plasticity of the GIPC-binding cavity in HaNLP3 relative to toxic NLPs. This likely precludes HaNLP3 binding to GIPCs, which is the underlying reason for the lack of toxicity. This study reveals that mutations at key protein regions cause a switch between non-toxic and toxic phenotypes within the same protein scaffold. Altogether, these data provide evidence that protein flexibility is a distinguishing trait of toxic NLPs and highlight structural determinants for a potential functional diversification of non-toxic NLPs utilized by biotrophic plant pathogens.


Subject(s)
Oomycetes/genetics , Oomycetes/metabolism , Plant Diseases/parasitology , Amino Acid Sequence , Ethylenes/metabolism , Necrosis/metabolism , Peptides/metabolism , Peronospora/genetics , Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
6.
Sci Rep ; 9(1): 12243, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31439922

ABSTRACT

Cholera is a life-threatening diarrhoeal disease caused by the human pathogen Vibrio cholerae. Infection occurs after ingestion of the bacteria, which colonize the human small intestine and secrete their major virulence factor - the cholera toxin (CT). The GM1 ganglioside is considered the primary receptor of the CT, but recent studies suggest that also fucosylated receptors such as histo-blood group antigens are important for cellular uptake and toxicity. Recently, a special focus has been on the histo-blood group antigen Lewisx (Lex), however, where and how the CT binds to Lex remains unclear. Here we report the high-resolution crystal structure (1.5 Å) of the receptor-binding B-subunits of the CT bound to the Lex trisaccharide, and complementary quantitative binding data for CT holotoxins. Lex, and also L-fucose alone, bind to the secondary binding site of the toxin, distinct from the GM1 binding site. In contrast, fucosyl-GM1 mainly binds to the primary binding site due to high-affinity interactions of its GM1 core. Lex is the first histo-blood group antigen of non-secretor phenotype structurally investigated in complex with CT. Together with the quantitative binding data, this allows unique insight into why individuals with non-secretor phenotype are more prone to severe cholera than so-called 'secretors'.


Subject(s)
Blood Group Antigens/metabolism , Cholera Toxin/chemistry , Cholera/metabolism , G(M1) Ganglioside/analogs & derivatives , Vibrio cholerae/metabolism , Binding Sites , Blood Group Antigens/chemistry , Cholera/microbiology , Cholera Toxin/metabolism , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/metabolism , Glycosylation , Humans , Protein Binding , Vibrio cholerae/chemistry , Vibrio cholerae/genetics
7.
Mol Microbiol ; 112(4): 1253-1269, 2019 10.
Article in English | MEDLINE | ID: mdl-31376198

ABSTRACT

Proteins of the aegerolysin family have a high abundance in Fungi. Due to their specific binding to membrane lipids, and their membrane-permeabilization potential in concert with protein partner(s) belonging to a membrane-attack-complex/perforin (MACPF) superfamily, they were proposed as useful tools in different biotechnological and biomedical applications. In this work, we performed functional studies on expression of the genes encoding aegerolysin and MACPF-like proteins in Aspergillus niger. Our results suggest the sporulation process being crucial for strong induction of the expression of all these genes. However, deletion of either of the aegerolysin genes did not influence the growth, development, sporulation efficiency and phenotype of the mutants, indicating that aegerolysins are not key factors in the sporulation process. In all our expression studies we noticed a strong correlation in the expression of one aegerolysin and MACPF-like gene. Aegerolysins were confirmed to be secreted from the fungus. We also showed the specific interaction of a recombinant A. niger aegerolysin with an invertebrate-specific membrane sphingolipid. Moreover, using this protein labelled with mCherry we successfully stained insect cells membranes containing this particular sphingolipid. Our combined results suggest, that aegerolysins in this species, and probably also in other aspergilli, could be involved in defence against predators.


Subject(s)
Complement Membrane Attack Complex/metabolism , Fungal Proteins/metabolism , Hemolysin Proteins/metabolism , Perforin/metabolism , Aspergillus niger/genetics , Aspergillus niger/metabolism , Complement Membrane Attack Complex/genetics , Fungal Proteins/physiology , Gene Expression Regulation, Fungal/genetics , Hemolysin Proteins/physiology , Membrane Proteins/metabolism , Perforin/genetics , Sphingolipids/metabolism , Spores, Fungal/genetics , Spores, Fungal/metabolism
8.
Methods Mol Biol ; 2003: 53-70, 2019.
Article in English | MEDLINE | ID: mdl-31218613

ABSTRACT

Surface plasmon resonance (SPR) is an established method for studying molecular interactions in real time. It allows obtaining qualitative and quantitative data on interactions of proteins with lipids or lipid membranes. In most of the approaches a lipid membrane or a membrane-mimetic surface is prepared on the surface of Biacore (GE Healthcare) sensor chips HPA or L1, and the studied protein is then injected across the surface. Here we provide an overview of SPR in protein-lipid and protein-membrane interactions, different approaches described in the literature and a general protocol for conducting an SPR experiment including lipid membranes, together with some experimental considerations.


Subject(s)
Membrane Lipids/metabolism , Membranes/metabolism , Proteins/metabolism , Protein Binding/physiology , Surface Plasmon Resonance/methods
9.
Structure ; 27(7): 1094-1102.e4, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31056420

ABSTRACT

Bacteria identify and respond to DNA damage using the SOS response. LexA, a central repressor in the response, has been implicated in the regulation of lysogeny in various temperate bacteriophages. During infection of Bacillus thuringiensis with GIL01 bacteriophage, LexA represses the SOS response and the phage lytic cycle by binding DNA, an interaction further stabilized upon binding of a viral protein, gp7. Here we report the crystallographic structure of phage-borne gp7 at 1.7-Å resolution, and characterize the 4:2 stoichiometry and potential interaction with LexA using surface plasmon resonance, static light scattering, and small-angle X-ray scattering. These data suggest that gp7 stabilizes LexA binding to operator DNA via coordination of the N- and C-terminal domains of LexA. Furthermore, we have found that gp7 can interact with LexA from Staphylococcus aureus, a significant human pathogen. Our results provide structural evidence as to how phage factors can directly associate with LexA to modulate the SOS response.


Subject(s)
Bacillus Phages/genetics , Bacillus thuringiensis/genetics , Bacterial Proteins/chemistry , DNA, Bacterial/chemistry , Serine Endopeptidases/chemistry , Staphylococcus aureus/genetics , Viral Regulatory and Accessory Proteins/chemistry , Amino Acid Sequence , Bacillus Phages/metabolism , Bacillus thuringiensis/metabolism , Bacillus thuringiensis/virology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hydrogen Bonding , Lysogeny/genetics , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , SOS Response, Genetics , Sequence Alignment , Sequence Homology, Amino Acid , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Staphylococcus aureus/metabolism , Staphylococcus aureus/virology , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism
10.
DNA Repair (Amst) ; 79: 50-54, 2019 07.
Article in English | MEDLINE | ID: mdl-31129429

ABSTRACT

Cells employ specific and nonspecific mechanisms to protect their genome integrity against exogenous and endogenous factors. The clbS gene is part of the polyketide synthase machinery (pks genomic island) encoding colibactin, a genotoxin implicated in promoting colorectal cancer. The pks is found among the Enterobacteriaceae, in particular Escherichia coli strains of the B2 phylogenetic group. Several resistance mechanisms protect toxin producers against toxicity of their products. ClbS, a cyclopropane hydrolase, was shown to confer colibactin resistance by opening its electrophilic cyclopropane ring. Here we report that ClbS sustained viability and enabled growth also of E. coli expressing another genotoxin, the Usp nuclease. The recA::gfp reporter system showed that ClbS protects against Usp induced DNA damage. To elucidate the mechanism of ClbS mediated protection, we studied the DNA binding ability of the ClbS protein. We show that ClbS directly interacts with single-stranded DNA (ssDNA) and double-stranded DNA (dsDNA), whereas ssDNA seems to be the preferred substrate. Thus, the ClbS DNA-binding characteristics may serve bacteria to protect their genomes against DNA degradation.


Subject(s)
DNA Damage , DNA, Bacterial/metabolism , DNA-Binding Proteins/metabolism , Escherichia coli Proteins/metabolism , Peptides/metabolism , Polyketides/metabolism , DNA, Bacterial/genetics , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Escherichia coli , Escherichia coli Proteins/genetics , Protein Binding
11.
Sci Rep ; 9(1): 5073, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30911026

ABSTRACT

Aegerolysins ostreolysin A (OlyA) and pleurotolysin A (PlyA), and pleurotolysin B (PlyB) with the membrane-attack-complex/perforin domain are proteins from the mushroom genus Pleurotus. Upon binding to sphingomyelin/cholesterol-enriched membranes, OlyA and PlyA can recruit PlyB to form multimeric bi-component transmembrane pores. Recently, Pleurotus aegerolysins OlyA, PlyA2 and erylysin A (EryA) were demonstrated to preferentially bind to artificial lipid membranes containing 50 mol% ceramide phosphoethanolamine (CPE), the main sphingolipid in invertebrate cell membranes. In this study, we demonstrate that OlyA6, PlyA2 and EryA bind to insect cells and to artificial lipid membranes with physiologically relevant CPE concentrations. Moreover, these aegerolysins permeabilize these membranes when combined with PlyB. These aegerolysin/PlyB complexes show selective toxicity toward western corn rootworm larvae and adults and Colorado potato beetle larvae. These data strongly suggest that these aegerolysin/PlyB complexes recognize CPE as their receptor molecule in the insect midgut. This mode of binding is different from those described for similar aegerolysin-based bacterial complexes, or other Bacillus thuringiensis Cry toxins, which have protein receptors. Targeting of Pleurotus aegerolysins to CPE and formation of transmembrane pores in concert with PlyB suggest the use of aegerolysin/PlyB complexes as novel biopesticides for the control of western corn rootworm and Colorado potato beetle.


Subject(s)
Hemolysin Proteins/metabolism , Insecticides/chemistry , Insecticides/pharmacology , Pleurotus/chemistry , Sphingomyelins/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Coleoptera , Dogs , Fungal Proteins/metabolism , Larva/drug effects , Madin Darby Canine Kidney Cells , Sf9 Cells , Surface Plasmon Resonance , Unilamellar Liposomes/metabolism
12.
Int J Mol Sci ; 20(3)2019 Feb 06.
Article in English | MEDLINE | ID: mdl-30736336

ABSTRACT

Diarrhea caused by enterotoxigenic Escherichia coli (ETEC) is one of the leading causes of mortality in children under five years of age and is a great burden on developing countries. The major virulence factor of the bacterium is the heat-labile enterotoxin (LT), a close homologue of the cholera toxin. The toxins bind to carbohydrate receptors in the gastrointestinal tract, leading to toxin uptake and, ultimately, to severe diarrhea. Previously, LT from human- and porcine-infecting ETEC (hLT and pLT, respectively) were shown to have different carbohydrate-binding specificities, in particular with respect to N-acetyllactosamine-terminating glycosphingolipids. Here, we probed 11 single-residue variants of the heat-labile enterotoxin with surface plasmon resonance spectroscopy and compared the data to the parent toxins. In addition we present a 1.45 Å crystal structure of pLTB in complex with branched lacto-N-neohexaose (Galß4GlcNAcß6[Galß4GlcNAcß3]Galß4Glc). The largest difference in binding specificity is caused by mutation of residue 94, which links the primary and secondary binding sites of the toxins. Residue 95 (and to a smaller extent also residues 7 and 18) also contribute, whereas residue 4 shows no effect on monovalent binding of the ligand and may rather be important for multivalent binding and avidity.


Subject(s)
Enterotoxigenic Escherichia coli/genetics , Enterotoxins/chemistry , Enterotoxins/genetics , Models, Molecular , Mutagenesis, Site-Directed , Binding Sites , Carbohydrates/chemistry , Crystallography, X-Ray , Humans , Molecular Conformation , Protein Binding , Structure-Activity Relationship , Surface Plasmon Resonance
13.
FEBS J ; 286(3): 536-554, 2019 02.
Article in English | MEDLINE | ID: mdl-30570212

ABSTRACT

The rules of the genetic code are established by aminoacyl-tRNA synthetases (aaRSs) enzymes, which covalently link tRNA with the cognate amino acid. Many aaRSs are involved in diverse cellular processes beyond translation, acting alone, or in complex with other proteins. However, studies of aaRS noncanonical assembly and functions in plants are scarce, as are structural studies of plant aaRSs. Here, we have solved the crystal structure of Arabidopsis thaliana cytosolic seryl-tRNA synthetase (SerRS), which is the first crystallographic structure of a plant aaRS. Arabidopsis SerRS displays structural features typical of canonical SerRSs, except for a unique intrasubunit disulfide bridge. In a yeast two-hybrid screen, we identified BEN1, a protein involved in the metabolism of plant brassinosteroid hormones, as a protein interactor of Arabidopsis SerRS. The SerRS:BEN1 complex is one of the first protein complexes of plant aaRSs discovered so far, and is a rare example of an aaRS interacting with an enzyme involved in primary or secondary metabolism. To pinpoint regions responsible for this interaction, we created truncated variants of SerRS and BEN1, and identified that the interaction interface involves the SerRS globular catalytic domain and the N-terminal extension of BEN1 protein. BEN1 does not have a strong impact on SerRS aminoacylation activity, indicating that the primary function of the complex is not the modification of SerRS canonical activity. Perhaps SerRS performs as yet unknown noncanonical functions mediated by BEN1. These findings indicate that - via SerRS and BEN1 - a link exists between the protein translation and steroid metabolic pathways of the plant cell. DATABASE: Structural data are available in the PDB under the accession number PDB ID 6GIR.


Subject(s)
Alcohol Oxidoreductases/chemistry , Arabidopsis Proteins/chemistry , Arabidopsis/chemistry , Serine-tRNA Ligase/chemistry , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Amino Acid Sequence , Arabidopsis/enzymology , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Binding Sites , Brassinosteroids/biosynthesis , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Kinetics , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Serine-tRNA Ligase/genetics , Serine-tRNA Ligase/metabolism , Substrate Specificity , Two-Hybrid System Techniques
14.
J Enzyme Inhib Med Chem ; 33(1): 1239-1247, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30141354

ABSTRACT

Autolysin E (AtlE) is a cell wall degrading enzyme that catalyzes the hydrolysis of the ß-1,4-glycosidic bond between the N-acetylglucosamine and N-acetylmuramic acid units of the bacterial peptidoglycan. Using our recently determined crystal structure of AtlE from Staphylococcus aureus and a combination of pharmacophore modeling, similarity search, and molecular docking, a series of (Phenylureido)piperidinyl benzamides were identified as potential binders and surface plasmon resonance (SPR) and saturation-transfer difference (STD) NMR experiments revealed that discovered compounds bind to AtlE in a lower micromolar range. (phenylureido)piperidinyl benzamides are the first reported non-substrate-like compounds that interact with this enzyme and enable further study of the interaction of small molecules with bacterial AtlE as potential inhibitors of this target.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , N-Acetylmuramoyl-L-alanine Amidase/antagonists & inhibitors , Piperidines/pharmacology , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , N-Acetylmuramoyl-L-alanine Amidase/chemistry , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Piperidines/chemical synthesis , Piperidines/chemistry , Staphylococcus aureus/enzymology , Structure-Activity Relationship
15.
Langmuir ; 34(30): 8983-8993, 2018 07 31.
Article in English | MEDLINE | ID: mdl-29983071

ABSTRACT

Understanding of the interactions between proteins and natural and artificially prepared lipid membrane surfaces and embedded nonpolar cores is important in studies of physiological processes and their pathologies and is applicable to nanotechnologies. In particular, rapidly growing interest in cellular droplets defines the need for simplified biomimetic lipid model systems to overcome in vivo complexity and variability. We present a protocol for the preparation of kinetically stable nanoemulsions with nanodroplets composed of sphingomyelin (SM) and cholesterol (Chol), as amphiphilic surfactants, and trioleoylglycerol (TOG), at various molar ratios. To prepare stable SM/Chol-coated monodisperse lipid nanodroplets, we modified a reverse phase evaporation method and combined it with ultrasonication. Lipid composition, ζ-potential, gyration and hydrodynamic radius, shape, and temporal stability of the lipid nanodroplets were characterized and compared to extruded SM/Chol large unilamellar vesicles. Lipid nanodroplets and large unilamellar vesicles with theoretical SM/Chol/TOG molar ratios of 1/1/4.7 and 4/1/11.7 were further investigated for the orientational order of their interfacial water molecules using a second harmonic scattering technique, and for interactions with the SM-binding and Chol-binding pore-forming toxins equinatoxin II and perfringolysin O, respectively. The surface characteristics (ζ-potential, orientational order of interfacial water molecules) and binding of these proteins to the nanodroplet SM/Chol monolayers were similar to those for the SM/Chol bilayers of the large unilamellar vesicles and SM/Chol Langmuir monolayers, in terms of their surface structures. We propose that such SM/Chol/TOG nanoparticles with the required lipid compositions can serve as experimental models for monolayer membrane to provide a system that imitates the natural lipid droplets.


Subject(s)
Cholesterol/chemistry , Lipids/chemistry , Nanostructures/chemistry , Proteins/metabolism , Sphingomyelins/chemistry , Protein Binding , Proteins/chemistry , Triolein/chemistry , Unilamellar Liposomes/chemistry , Water/chemistry
16.
Bioconjug Chem ; 29(8): 2763-2775, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30024737

ABSTRACT

Affinity chromatography based on bacterial immunoglobulin (Ig)-binding proteins represents the cornerstone of therapeutic antibody downstream processing. However, there is a pressing need for more robust affinity ligands that would withstand the harsh column sanitization conditions, while still displaying high selectivity for antibodies. Here, we report the development of linear peptide IgG ligands, identified from combinatorial phage-display library screens. The lead peptide was shown to compete with staphylococcal protein A for the IgG Fc region. Trimming analysis and alanine scanning revealed the minimal structural requirements of the peptide for Fc binding, and the minimized peptide GSYWYQVWF recognized all human IgG subtypes. Mutation of glutamine located at the nonessential position 6 to aspartate led to the optimized peptide GSYWYDVWF with 18-fold higher affinity ( KD app. 0.6 µM) compared to the parent peptide. When coupled to paramagnetic beads or a chromatographic matrix, the optimized ligand was shown to selectively enrich antibodies from complex protein mixtures.


Subject(s)
Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Peptides/metabolism , Amino Acid Sequence , Bacteriophages/genetics , Chromatography, Affinity/methods , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Ligands , Peptides/chemistry , Staphylococcal Protein A/metabolism , Surface Plasmon Resonance
17.
Nucleic Acids Res ; 46(18): 9432-9443, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30053203

ABSTRACT

The GIL01 bacteriophage is a temperate phage that infects the insect pathogen Bacillus thuringiensis. During the lytic cycle, phage gene transcription is initiated from three promoters: P1 and P2, which control the expression of the early phage genes involved in genome replication and P3, which controls the expression of the late genes responsible for virion maturation and host lysis. Unlike most temperate phages, GIL01 lysogeny is not maintained by a dedicated phage repressor but rather by the host's regulator of the SOS response, LexA. Previously we showed that the lytic cycle was induced by DNA damage and that LexA, in conjunction with phage-encoded protein gp7, repressed P1. Here we examine the lytic/lysogenic switch in more detail and show that P3 is also repressed by a LexA-gp7 complex, binding to tandem LexA boxes within the promoter. We also demonstrate that expression from P3 is considerably delayed after DNA damage, requiring the phage-encoded DNA binding protein, gp6. Surprisingly, gp6 is homologous to LexA itself and, thus, is a rare example of a LexA homologue directly activating transcription. We propose that the interplay between these two LexA family members, with opposing functions, ensures the timely expression of GIL01 phage late genes.


Subject(s)
Bacterial Proteins/genetics , Bacteriophages/genetics , Lysogeny/genetics , Serine Endopeptidases/genetics , Transcription, Genetic/genetics , Viral Proteins/physiology , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacillus thuringiensis/genetics , Bacillus thuringiensis/metabolism , Bacterial Proteins/metabolism , Bacteriophages/metabolism , Base Sequence , Cytotoxins/genetics , Cytotoxins/metabolism , Gene Expression Regulation, Viral , Promoter Regions, Genetic , Sequence Homology , Serine Endopeptidases/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
18.
Sci Rep ; 8(1): 6894, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29720597

ABSTRACT

Listeria monocytogenes is a mammalian pathogen that causes gastroenteritis, miscarriages and infections of the central nervous system in immunocompromised individuals. Its main virulence factor is listeriolysin O (LLO), a pore-forming cholesterol-dependent cytolysin (CDC), which enables bacterial escape from the phagolysosome and contributes to bacterial pathogenicity. Details of cholesterol (Chol) recognition and membrane binding mechanisms by LLO are still not known. Here we used 19F-NMR spectroscopy in order to assess LLO-Chol interactions in solution and in a Chol-rich membrane environment. LLO has six tryptophan residues located in the region of the molecule that is first in contact with lipid membranes. 19F-LLO, which contained 5-fluoro-tryptophans, was prepared by using isotopic labelling in an E. coli expression system. Signals in the 19F-NMR spectrum of 19F-LLO were unambiguously assigned by using a series of single Trp → Phe point mutations. The results employing various cholesterol preparations in solution indicate that tryptophan residues are not directly involved in Chol binding in solution. However, significant chemical shift changes were observed upon LLO binding to Chol-rich membranes, highlighting the role of tryptophan residues in membrane interactions (W512) and oligomerisation (W189 and W489).


Subject(s)
Bacterial Toxins/chemistry , Cell Membrane/chemistry , Fluorine-19 Magnetic Resonance Imaging , Heat-Shock Proteins/chemistry , Hemolysin Proteins/chemistry , Membrane Lipids/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Cell Membrane/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Membrane Lipids/metabolism , Models, Molecular , Molecular Conformation , Mutation , Protein Binding , Structure-Activity Relationship
19.
Science ; 358(6369): 1431-1434, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29242345

ABSTRACT

Necrosis and ethylene-inducing peptide 1-like (NLP) proteins constitute a superfamily of proteins produced by plant pathogenic bacteria, fungi, and oomycetes. Many NLPs are cytotoxins that facilitate microbial infection of eudicot, but not of monocot plants. Here, we report glycosylinositol phosphorylceramide (GIPC) sphingolipids as NLP toxin receptors. Plant mutants with altered GIPC composition were more resistant to NLP toxins. Binding studies and x-ray crystallography showed that NLPs form complexes with terminal monomeric hexose moieties of GIPCs that result in conformational changes within the toxin. Insensitivity to NLP cytolysins of monocot plants may be explained by the length of the GIPC head group and the architecture of the NLP sugar-binding site. We unveil early steps in NLP cytolysin action that determine plant clade-specific toxin selectivity.


Subject(s)
Arabidopsis/parasitology , Cytotoxins/metabolism , Host Specificity , Phytophthora/metabolism , Plant Diseases/parasitology , Pythium/metabolism , Sphingolipids/metabolism , Toxins, Biological/metabolism , Binding Sites , Crystallography, X-Ray , Cytotoxins/chemistry , Ethylenes/metabolism , Sphingolipids/chemistry
20.
Drug Metab Dispos ; 45(8): 974-976, 2017 08.
Article in English | MEDLINE | ID: mdl-28536098

ABSTRACT

Statins are well known lipid lowering agents that inhibit the enzyme 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase. They also activate drug metabolism but their exact receptor-mediated action has not been proven so far. We tested whether atorvastatin and rosuvastatin are direct ligands of human constitutive androstane receptor (CAR). We measured binding activities of atorvastatin and rosuvastatin to the human constitutive androstane receptor/retinoid X receptor α ligand-binding domain (CAR/RXRα-LBD) heterodimer with surface plasmon resonance (SPR). Additionally, three-dimensional models of CAR/RXRα-LBD were constructed by ligand-based and structure-based in silico modeling. Experiments and computational modeling show that atorvastatin and rosuvastatin bind to the human CAR/RXRα-LBD heterodimer, suggesting both can modulate the activity of CAR through direct interaction with the LBD of this receptor. We confirm that atorvastatin and rosuvastatin are direct ligands of CAR. The clinical consequences of CAR activation by statins are in their potential drug-drug interactions, and changes in glucose and energy metabolism.


Subject(s)
Atorvastatin/metabolism , Computer Simulation , Receptors, Cytoplasmic and Nuclear/metabolism , Retinoid X Receptor alpha/metabolism , Rosuvastatin Calcium/metabolism , Anticholesteremic Agents/metabolism , Constitutive Androstane Receptor , Hep G2 Cells , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Ligands , Protein Binding/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...