Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 608
Filter
1.
Commun Biol ; 7(1): 655, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806706

ABSTRACT

The gut microbiota influences human health and the development of chronic diseases. However, our understanding of potentially protective or harmful microbe-host interactions at the molecular level is still in its infancy. To gain further insights into the hidden gut metabolome and its impact, we identified a cryptic non-ribosomal peptide BGC in the genome of Bacillus cereus DSM 28590 from the mouse intestine ( www.dsmz.de/miBC ), which was predicted to encode a thiazol(in)e substructure. Cloning and heterologous expression of this BGC revealed that it produces bacillamide D. In-depth functional evaluation showed potent cytotoxicity and inhibition of cell migration using the human cell lines HCT116 and HEK293, which was validated using primary mouse organoids. This work establishes the bacillamides as selective cytotoxins from a bacterial gut isolate that affect mammalian cells. Our targeted structure-function-predictive approach is demonstrated to be a streamlined method to discover deleterious gut microbial metabolites with potential effects on human health.


Subject(s)
Bacillus cereus , Gastrointestinal Microbiome , Bacillus cereus/metabolism , Bacillus cereus/genetics , Animals , Mice , Humans , HEK293 Cells , Cytotoxins/metabolism , Cytotoxins/genetics , HCT116 Cells , Intestines/microbiology , Cell Movement , Organoids/metabolism
2.
Microbiology (Reading) ; 169(9)2023 09.
Article in English | MEDLINE | ID: mdl-37702594

ABSTRACT

Cholesterol-dependent cytolysins (CDCs) are a large family of pore-forming toxins, produced by numerous Gram-positive pathogens. CDCs depend on host membrane cholesterol for pore formation; some CDCs also require surface-associated human CD59 (hCD59) for binding, conferring specificity for human cells. We purified a recombinant version of a putative CDC encoded in the genome of Streptococcus oralis subsp. tigurinus, tigurilysin (TGY), and used CRISPR/Cas9 to construct hCD59 knockout (KO) HeLa and JEG-3 cell lines. Cell viability assays with TGY on wild-type and hCD59 KO cells showed that TGY is a hCD59-dependent CDC. Two variants of TGY exist among S. oralis subsp. tigurinus genomes, only one of which is functional. We discovered that a single amino acid change between these two TGY variants determines its activity. Flow cytometry and oligomerization Western blots revealed that the single amino acid difference between the two TGY isoforms disrupts host cell binding and oligomerization. Furthermore, experiments with hCD59 KO cells and cholesterol-depleted cells demonstrated that TGY is fully dependent on both hCD59 and cholesterol for activity, unlike other known hCD59-dependent CDCs. Using full-length CDCs and toxin constructs differing only in the binding domain, we determined that having hCD59 dependence leads to increased lysis efficiency, conferring a potential advantage to organisms producing hCD59-dependent CDCs.


Subject(s)
Cytotoxins , Host Specificity , Humans , Cell Line, Tumor , Cytotoxins/genetics , Cholesterol , Amino Acids , CD59 Antigens/genetics
3.
Biosens Bioelectron ; 222: 114939, 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36459819

ABSTRACT

Developing rapid and non-invasive diagnostics for Helicobacter pylori (HP) is imperative to prevent associated diseases such as stomach gastritis, ulcers, and cancers. Owing to HP strain heterogeneity, not all HP-infected individuals incur side effects. Cytotoxin-associated gene A (CagA), and vacuolating cytotoxin A (VacA) genes predominantly drive HP pathogenicity. Therefore, diagnosing CagA and VacA genotypes could alert active infection and decide suitable therapeutics. We report an enhanced LbCas12a trans-cleavage activity with extended reporters and reductants (CEXTRAR) for early detection of HP. We demonstrate that extended ssDNA reporter acts as an excellent signal amplifier, making it a potential alternative substrate for LbCas12a collateral activity. Through a systematic investigation of various buffer components, we demonstrate that reductants improve LbCas12a trans-cleavage activity. Overall, our novel reporter and optimal buffer increased the trans-cleavage activity to an order of 16-fold, achieving picomolar sensitivity (171 pM) without target pre-amplification. Integrated with loop-mediated isothermal amplification (LAMP), CEXTRAR successfully attained attomolar sensitivity for HP detection using real-time fluorescence (43 and 96 aM), in-tube fluorescence readouts (430 and 960 aM), and lateral flow (4.3 and 9.6 aM) for CagA and VacA, respectively. We also demonstrate a rapid 2-min Triton X-100 lysis for clinical sample analysis, which could provide clinicians with actionable information for rapid diagnosis. CEXTRAR could potentially spot the 13C urea breath test false-negatives. For the first time, our study unveils an experimental outlook to manipulate reporters and reconsider precise cysteine substitution via protein engineering for Cas variants with enhanced catalytic activities for use in diagnostics and genetic engineering.


Subject(s)
Biosensing Techniques , Helicobacter Infections , Helicobacter pylori , Peptic Ulcer , Stomach Neoplasms , Humans , Antigens, Bacterial/metabolism , Bacterial Proteins/genetics , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Reducing Agents , CRISPR-Cas Systems , Early Detection of Cancer , Peptic Ulcer/diagnosis , Peptic Ulcer/genetics , Genotype , Cytotoxins/genetics , Helicobacter Infections/diagnosis , Helicobacter Infections/genetics , Helicobacter Infections/metabolism
4.
Biochem Biophys Res Commun ; 637: 240-246, 2022 12 31.
Article in English | MEDLINE | ID: mdl-36410272

ABSTRACT

Cholesterol-dependent cytolysin (CDC) is a bacterial toxin that binds to eukaryotic cholesterol-containing membranes, forms oligomeric complexes, and is inserted into the bilayer to create large aqueous pores. Recently, we reported a species-specific duplication of the hemolysin gene in group III Clostridium botulinum. The duplicated genes (bly1 and bly2) encoded two separate CDC proteins (botulinolysins; BLY1 and BLY2). Here, we aimed to investigate whether BLY1 and BLY2 exert differential cytotoxicity. We isolated two bly genes from C. botulinum and evaluated the cytotoxicity of two recombinant BLY proteins (rBLY1 and rBLY2) in HeLa, IEC-6, and NRK cells. rBLYs were cytotoxic to equine erythrocytes. rBLY1 showed higher hemolytic activity than rBLY2. rBLY2 showed no or very weak cytotoxicity to the HeLa, IEC-6, and NRK cells, whereas rBLY1 showed high cytotoxicity to these cells. The comparison of the amino acid sequence of BLYs with those of other CDCs revealed that the already-known amino acid residues involved in cholesterol-containing membrane recognition, oligomerization, and insertion into membranes are well conserved in both BLYs. However, several amino acid substitutions were observed in the conserved regions, particularly in L2 and L3 regions involved in cell binding. These findings suggest that gene duplication in group III C. botulinum evolved distinct functional specializations, and differential cytotoxicity of BLY1 and BLY2 could be due to the amino acid substitution in the conserved regions. However, the structural and functional comparisons of the two BLYs are essential to gain insights into the function of the CDCs.


Subject(s)
Clostridium botulinum , Gene Duplication , Animals , Horses , Cytotoxins/genetics , Clostridium , Cholesterol
5.
J Nat Prod ; 85(11): 2610-2619, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36331369

ABSTRACT

Myxobacteria have proven to be a rich source of natural products, but their biosynthetic potential seems to be underexplored given the high number of biosynthetic gene clusters present in their genomes. In this study, a truncated ajudazol biosynthetic gene cluster in Cystobacter sp. SBCb004 was identified using mutagenesis and metabolomics analyses and a set of novel ajudazols (named ajudazols C-J, 3-10, respectively) were detected and subsequently isolated. Their structures were elucidated using comprehensive HR-MS and NMR spectroscopy. Unlike the known ajudazols A (1) and B (2), which utilize acetyl-CoA as the biosynthetic starter unit, these novel ajudazols were proposed to incorporate 3,3-dimethylacrylyl CoA as the starter. Ajudazols C-J (3-10, respectively) are characterized by varying degrees of hydroxylation, desaturation, and different glycosylation patterns. Two P450-dependent enzymes and one glycosyltransferase are shown to be responsible for the hydroxylation at C-8, the desaturation at C-15 and C-33, and the transfer of a d-ß-glucopyranose, respectively, based on mutagenesis results. One of the cytochrome P450-dependent enzymes and the glycosyltransferase were found to be encoded by genes located outside the biosynthetic gene cluster. Ajudazols C-H (3-8, respectively) exhibit cytotoxicity against various cancer cell lines.


Subject(s)
Cytotoxins , Myxococcales , Cytotoxins/biosynthesis , Cytotoxins/genetics , Glycosyltransferases , Multigene Family , Mutagenesis , Myxococcales/genetics , Myxococcales/metabolism , Genome, Bacterial
6.
J Biol Chem ; 298(10): 102441, 2022 10.
Article in English | MEDLINE | ID: mdl-36055404

ABSTRACT

Vibrio cholerae cytolysin (VCC) is a potent membrane-damaging ß-barrel pore-forming toxin. Upon binding to the target membranes, VCC monomers first assemble into oligomeric prepore intermediates and subsequently transform into transmembrane ß-barrel pores. VCC harbors a designated pore-forming motif, which, during oligomeric pore formation, inserts into the membrane and generates a transmembrane ß-barrel scaffold. It remains an enigma how the molecular architecture of the pore-forming motif regulates the VCC pore-formation mechanism. Here, we show that a specific pore-forming motif residue, E289, plays crucial regulatory roles in the pore-formation mechanism of VCC. We find that the mutation of E289A drastically compromises pore-forming activity, without affecting the structural integrity and membrane-binding potential of the toxin monomers. Although our single-particle cryo-EM analysis reveals WT-like oligomeric ß-barrel pore formation by E289A-VCC in the membrane, we demonstrate that the mutant shows severely delayed kinetics in terms of pore-forming ability that can be rescued with elevated temperature conditions. We find that the pore-formation efficacy of E289A-VCC appears to be more profoundly dependent on temperature than that of the WT toxin. Our results suggest that the E289A mutation traps membrane-bound toxin molecules in the prepore-like intermediate state that is hindered from converting into the functional ß-barrel pores by a large energy barrier, thus highlighting the importance of this residue for the pore-formation mechanism of VCC.


Subject(s)
Bacterial Proteins , Cytotoxins , Pore Forming Cytotoxic Proteins , Vibrio cholerae , Virulence Factors , Cell Membrane/metabolism , Cytotoxins/chemistry , Cytotoxins/genetics , Vibrio cholerae/pathogenicity , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Virulence Factors/chemistry , Virulence Factors/genetics , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/genetics , Amino Acid Motifs , Mutation , Glutamic Acid/chemistry , Glutamic Acid/genetics
7.
J Bacteriol ; 204(5): e0055521, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35435721

ABSTRACT

Alpha-pore-forming toxins (α-PFTs) are secreted by many species of bacteria, including Escherichia coli, Aeromonas hydrophila, and Bacillus thuringiensis, as part of their arsenal of virulence factors, and are often cytotoxic. In particular, for α-PFTs, the membrane-spanning channel they form is composed of hydrophobic α-helices. These toxins oligomerize at the surface of target cells and transition from a soluble to a protomer state in which they expose their hydrophobic regions and insert into the membrane to form a pore. The pores may be composed of homooligomers of one component or heterooligomers with two or three components, resulting in bi- or tripartite toxins. The multicomponent α-PFTs are often expressed from a single operon. Recently, motility-associated killing factor A (MakA), an α-PFT, was discovered in Vibrio cholerae. We report that makA is found on the V. cholerae GI-10 genomic island within an operon containing genes for two other potential α-PFTs, MakB and MakE. We determined the X-ray crystal structures for MakA, MakB, and MakE and demonstrated that all three are structurally related to the α-PFT family in the soluble state, and we modeled their protomer state based on the α-PFT AhlB from A. hydrophila. We found that MakA alone is cytotoxic at micromolar concentrations. However, combining MakA with MakB and MakE is cytotoxic at nanomolar concentrations, with specificity for J774 macrophage cells. Our data suggest that MakA, -B, and -E are α-PFTs that potentially act as a tripartite pore-forming toxin with specificity for phagocytic cells. IMPORTANCE The bacterium Vibrio cholerae causes gastrointestinal, wound, and skin infections. The motility-associated killing factor A (MakA) was recently shown to be cytotoxic against colon, prostate, and other cancer cells. However, at the outset of this study, the capacity of MakA to damage cells in combination with other Mak proteins encoded in the same operon had not been elucidated. We determined the structures of three Mak proteins and established that they are structurally related to the α-PFTs. Compared to MakA alone, the combination of all three toxins was more potent specifically in mouse macrophages. This study highlights the idea that the Mak toxins are selectively cytotoxic and thus may function as a tripartite toxin with cell type specificity.


Subject(s)
Vibrio cholerae , Animals , Cytotoxins/genetics , Cytotoxins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Genomic Islands , Mice , Pore Forming Cytotoxic Proteins , Protein Subunits/metabolism , Vibrio cholerae/metabolism , Virulence Factors/metabolism
8.
J Microbiol Methods ; 191: 106358, 2021 12.
Article in English | MEDLINE | ID: mdl-34743930

ABSTRACT

L-asparaginase from endophytic Fusarium proliferatum (isolate CCH, GenBank accession no. MK685139) isolated from the medicinal plant Cymbopogon citratus (Lemon grass), was optimized for its L-asparaginase production and its subsequent cytotoxicity towards Jurkat E6 cell line. The following factors were optimized; carbon source and concentration, nitrogen source and concentration, incubation period, temperature, pH and agitation rate. Optimization of L-asparaginase production was performed using One-Factor-At-A-Time (OFAT) and Response surface methodology (RSM) model. The cytotoxicity of the crude enzyme from isolate CCH was tested on leukemic Jurkat E6 cell line. The optimization exercise revealed that glucose concentration, nitrogen source, L-asparagine concentration and temperature influenced the L-asparaginase production of CCH. The optimum condition suggested using OFAT and RSM results were consistent. As such, the recommended conditions were 0.20% of glucose, 0.99% of L-asparagine and 5.34 days incubation at 30.50 °C. The L-asparaginase production of CCH increased from 16.75 ± 0.76 IU/mL to 22.42 ± 0.20 IU/mL after optimization. The cytotoxicity of the crude enzyme on leukemic Jurkat cell line recorded IC50 value at 33.89 ± 2.63% v/v. To conclude, the enzyme extract produced from Fusarium proliferatum under optimized conditions is a potential alternative resource for L-asparaginase.


Subject(s)
Asparaginase/biosynthesis , Cytotoxins/biosynthesis , Endophytes/metabolism , Fusarium/metabolism , Antineoplastic Agents , Asparaginase/genetics , Asparaginase/isolation & purification , Carbon , Culture Media/chemistry , Cytotoxins/genetics , Databases, Nucleic Acid , Endophytes/enzymology , Endophytes/genetics , Fusarium/enzymology , Fusarium/genetics , Hydrogen-Ion Concentration , Microbiological Techniques/methods , Nitrogen , Plants, Medicinal , Temperature
9.
PLoS Negl Trop Dis ; 15(10): e0009841, 2021 10.
Article in English | MEDLINE | ID: mdl-34634067

ABSTRACT

Development of a rapid, on-site detection tool for snakebite is highly sought after, owing to its clinically and forensically relevant medicolegal significance. Polyvalent antivenom therapy in the management of such envenomation cases is finite due to its poor venom neutralization capabilities as well as diagnostic ramifications manifested as untoward immunological reactions. For precise molecular diagnosis of elapid venoms of the big four snakes, we have developed a lateral flow kit using a monoclonal antibody (AB1; IgG1 - κ chain; Kd: 31 nM) generated against recombinant cytotoxin-7 (rCTX-7; 7.7 kDa) protein of the elapid venom. The monoclonal antibody specifically detected the venoms of Naja naja (p < 0.0001) and Bungarus caeruleus (p<0.0001), without showing any immunoreactivity against the viperidae snakes in big four venomous snakes. The kit developed attained the limit of quantitation of 170 pg/µL and 2.1 ng/µL in spiked buffer samples and 28.7 ng/µL and 110 ng/µL in spiked serum samples for detection of N. naja and B. caeruleus venoms, respectively. This kit holds enormous potential in identification of elapid venom of the big four snakes for effective prognosis of an envenomation; as per the existing medical guidelines.


Subject(s)
Colorimetry/methods , Cytotoxins/analysis , Elapidae/immunology , Immunoassay/methods , Immunotoxins/analysis , Snake Venoms/analysis , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Bungarus/genetics , Bungarus/physiology , Cytotoxins/genetics , Cytotoxins/immunology , Elapid Venoms/analysis , Elapid Venoms/genetics , Elapid Venoms/immunology , Elapidae/physiology , Immunotoxins/genetics , Immunotoxins/immunology , Naja naja/immunology , Naja naja/physiology , Snake Venoms/immunology , Viperidae/immunology , Viperidae/physiology
10.
Toxins (Basel) ; 13(9)2021 08 24.
Article in English | MEDLINE | ID: mdl-34564597

ABSTRACT

BACKGROUND: Helicobacter pylori (Hp) colonizes the human stomach and can induce gastric cancer and mucosa-associated lymphoid tissue (MALT) lymphoma. Clinical observations suggest a role for the Hp virulence factor cytotoxin-associated gene A (CagA) in pathogenesis. The pathogenic activity of CagA is partly regulated by tyrosine phosphorylation of C-terminal Glu-Pro-Ile-Tyr-Ala (EPIYA) motifs in host cells. However, CagA differs considerably in EPIYA motifs, whose functions have been well characterized in epithelial cells. Since CagA is fragmented in immune cells, different CagA variants may exhibit undetected functions in B cells. METHODS: B cells were infected with Hp isolates and isogenic mutants expressing different CagA EPIYA variants. CagA translocation and tyrosine phosphorylation were investigated by Western blotting. Apoptosis was analyzed by flow cytometry and metabolic activity was detected by an MTT assay. RESULTS: Isogenic CagA EPIYA variants are equally well translocated into B cells, followed by tyrosine phosphorylation and cleavage. B cell apoptosis was induced in a CagA-independent manner. However, variants containing at least one EPIYA-C motif affected metabolic activity independently of phosphorylation or multiplication of EPIYA-C motifs. CONCLUSIONS: The diverse structure of CagA regulates B cell physiology, whereas B cell survival is independent of CagA.


Subject(s)
Helicobacter Infections/metabolism , Helicobacter Infections/physiopathology , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Lymphoma, B-Cell, Marginal Zone/metabolism , Lymphoma, B-Cell, Marginal Zone/physiopathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/physiopathology , Cytotoxins/genetics , Cytotoxins/metabolism , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Neoplastic , Genetic Variation , Genotype , Host-Pathogen Interactions/genetics , Humans
11.
PLoS Genet ; 17(2): e1009387, 2021 02.
Article in English | MEDLINE | ID: mdl-33577603

ABSTRACT

Cholesterol-dependent cytolysins (CDCs), of which intermedilysin (ILY) is an archetypal member, are a group of pore-forming toxins secreted by a large variety of pathogenic bacteria. These toxins, secreted as soluble monomers, oligomerize upon interaction with cholesterol in the target membrane and transect it as pores of diameters of up to 100 to 300 Å. These pores disrupt cell membranes and result in cell lysis. The immune receptor CD59 is a well-established cellular factor required for intermedilysin pore formation. In this study, we applied genome-wide CRISPR-Cas9 knock-out screening to reveal additional cellular co-factors essential for ILY-mediated cell lysis. We discovered a plethora of genes previously not associated with ILY, many of which are important for membrane constitution. We show that heparan sulfates facilitate ILY activity, which can be inhibited by heparin. Furthermore, we identified hits in both protein and lipid glycosylation pathways and show a role for glucosylceramide, demonstrating that membrane organization is important for ILY activity. We also cross-validated identified genes with vaginolysin and pneumolysin and found that pneumolysin's cytolytic activity strongly depends on the asymmetric distribution of membrane phospholipids. This study shows that membrane-targeting toxins combined with genetic screening can identify genes involved in biological membrane composition and metabolism.


Subject(s)
Bacterial Proteins/metabolism , Bacteriocins/metabolism , Cell Membrane/metabolism , Cholesterol/metabolism , Cytotoxins/metabolism , Heparitin Sulfate/metabolism , Bacterial Proteins/genetics , Bacteriocins/genetics , CD59 Antigens/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Cytotoxins/genetics , HEK293 Cells , Humans , Porosity
12.
Nat Commun ; 11(1): 5818, 2020 11 16.
Article in English | MEDLINE | ID: mdl-33199689

ABSTRACT

Cholesterol-dependent cytolysins (CDCs) are pore-forming proteins that serve as major virulence factors for pathogenic bacteria. They target eukaryotic cells using different mechanisms, but all require the presence of cholesterol to pierce lipid bilayers. How CDCs use cholesterol to selectively lyse cells is essential for understanding virulence strategies of several pathogenic bacteria, and for repurposing CDCs to kill new cellular targets. Here we address that question by trapping an early state of pore formation for the CDC intermedilysin, bound to the human immune receptor CD59 in a nanodisc model membrane. Our cryo electron microscopy map reveals structural transitions required for oligomerization, which include the lateral movement of a key amphipathic helix. We demonstrate that the charge of this helix is crucial for tuning lytic activity of CDCs. Furthermore, we discover modifications that overcome the requirement of cholesterol for membrane rupture, which may facilitate engineering the target-cell specificity of pore-forming proteins.


Subject(s)
Cell Membrane/metabolism , Cytotoxins/chemistry , Cytotoxins/metabolism , CD59 Antigens/metabolism , Cell Membrane/ultrastructure , Cryoelectron Microscopy , Cytotoxins/genetics , Humans , Models, Biological , Models, Molecular , Mutation/genetics , Protein Structure, Secondary , Structure-Activity Relationship
13.
Int J Mol Sci ; 21(19)2020 Sep 28.
Article in English | MEDLINE | ID: mdl-32998313

ABSTRACT

The edible mushroom Agrocybe aegerita produces a ribotoxin-like protein known as Ageritin. In this work, the gene encoding Ageritin was characterized by sequence analysis. It contains several typical features of fungal genes such as three short introns (60, 55 and 69 bp) located at the 5' region of the coding sequence and typical splice junctions. This sequence codes for a precursor of 156 amino acids (~17-kDa) containing an additional N-terminal peptide of 21 amino acid residues, absent in the purified toxin (135 amino acid residues; ~15-kDa). The presence of 17-kDa and 15-kDa forms was investigated by Western blot in specific parts of fruiting body and in mycelia of A. aegerita. Data show that the 15-kDa Ageritin is the only form retrieved in the fruiting body and the principal form in mycelium. The immunolocalization by confocal laser scanning microscopy and transmission electron microscopy proves that Ageritin has vacuolar localization in hyphae. Coupling these data with a bioinformatics approach, we suggest that the N-terminal peptide of Ageritin (not found in the purified toxin) is a new signal peptide in fungi involved in intracellular routing from endoplasmic reticulum to vacuole, necessary for self-defense of A. aegerita ribosomes from Ageritin toxicity.


Subject(s)
Agrocybe/genetics , Cytotoxins/genetics , Fruiting Bodies, Fungal/metabolism , Fungal Proteins/genetics , Mycelium/metabolism , Ribonucleases/genetics , Agrocybe/metabolism , Agrocybe/ultrastructure , Amino Acid Sequence , Computational Biology , Cytotoxins/biosynthesis , Cytotoxins/isolation & purification , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Exons , Fruiting Bodies, Fungal/ultrastructure , Fungal Proteins/biosynthesis , Fungal Proteins/isolation & purification , Gene Expression , Introns , Mycelium/ultrastructure , Open Reading Frames , Protein Sorting Signals/genetics , Protein Transport , Ribonucleases/biosynthesis , Ribonucleases/isolation & purification , Ribosomes/genetics , Ribosomes/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Vacuoles/metabolism , Vacuoles/ultrastructure
14.
mBio ; 11(5)2020 09 29.
Article in English | MEDLINE | ID: mdl-32994330

ABSTRACT

The cholesterol-dependent cytolysins (CDCs) are bacterial, ß-barrel, pore-forming toxins. A central enigma of the pore-forming mechanism is how completion of the prepore is sensed to initiate its conversion to the pore. We identified a motif that is conserved between the CDCs and a diverse family of nearly 300 uncharacterized proteins present in over 220 species that span at least 10 bacterial and 2 eukaryotic phyla. Except for this motif, these proteins exhibit little similarity to the CDCs at the primary structure level. Studies herein show this motif is a critical component of the sensor that initiates the prepore-to-pore transition in the CDCs. We further show by crystallography, single particle analysis, and biochemical studies of one of these CDC-like (CDCL) proteins from Elizabethkingia anophelis, a commensal of the malarial mosquito midgut, that a high degree of structural similarity exists between the CDC and CDCL monomer structures and both form large oligomeric pore complexes. Furthermore, the conserved motif in the E. anophelis CDCL crystal structure occupies a nearly identical position and makes similar contacts to those observed in the structure of the archetype CDC, perfringolysin O (PFO). This suggests a common function in the CDCs and CDCLs and may explain why only this motif is conserved in the CDCLs. Hence, these studies identify a critical component of the sensor involved in initiating the prepore-to-pore transition in the CDCs, which is conserved in a large and diverse group of distant relatives of the CDCs.IMPORTANCE The cholesterol-dependent cytolysins' pore-forming mechanism relies on the ability to sense the completion of the oligomeric prepore structure and initiate the insertion of the ß-barrel pore from the assembled prepore structure. These studies show that a conserved motif is an important component of the sensor that triggers the prepore-to-pore transition and that it is conserved in a large family of previously unidentified CDC-like proteins, the genes for which are present in a vast array of microbial species that span most terrestrial environments, as well as most animal and human microbiomes. These studies establish the foundation for future investigations that will probe the contribution of this large family of CDC-like proteins to microbial survival and human disease.


Subject(s)
Amino Acid Motifs , Cholesterol/metabolism , Cytotoxins/chemistry , Flavobacteriaceae/chemistry , Animals , Cell Membrane/metabolism , Crystallography, X-Ray , Culicidae/microbiology , Cytotoxins/genetics , Flavobacteriaceae/genetics , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/genetics
15.
Infect Genet Evol ; 85: 104483, 2020 11.
Article in English | MEDLINE | ID: mdl-32731044

ABSTRACT

Streptococcus mitis strain Nm-65 secretes an atypical 5-domain-type cholesterol-dependent cytolysin (CDC) called S. mitis-derived human platelet aggregation factor (Sm-hPAF) originally described as a platelet aggregation factor. Sm-hPAF belongs to Group III CDC that recognize both membrane cholesterol and human CD59 as the receptors, and shows preferential activity towards human cells. Draft genome analyses have shown that the Nm-65 strain also harbors a gene encoding another CDC called mitilysin (MLY). This CDC belongs to Group I CDC that recognizes only membrane cholesterol as a receptor, and it is a homolog of the pneumococcal CDC, pneumolysin. The genes encoding each CDC are located about 20 kb apart on the Nm-65 genome. Analysis of the genomic locus of these CDC-encoding genes in silico showed that the gene encoding Sm-hPAF and the region including the gene encoding MLY were both inserted into a specific locus of the S. mitis genome. The results obtained using deletion mutants of the gene(s) encoding CDC in Nm-65 indicated that each CDC contributes to both hemolysis and cytotoxicity, and that MLY is the major hemolysin/cytolysin in Nm-65. The present study aimed to determine the potential pathogenicity of an S. mitis strain that produces two CDC with different receptor recognition properties and secretion modes.


Subject(s)
Bacterial Toxins/genetics , Cytotoxins/genetics , Cytotoxins/toxicity , Hemolysin Proteins/genetics , Hemolysin Proteins/toxicity , Streptococcus mitis/genetics , Amino Acid Sequence , CD59 Antigens/isolation & purification , Cholesterol , Genetic Variation , Genotype , Humans , Mutation , Platelet Aggregation Inhibitors/isolation & purification , Streptococcus mitis/chemistry
16.
Food Res Int ; 129: 108788, 2020 03.
Article in English | MEDLINE | ID: mdl-32036912

ABSTRACT

Probiotics are naturally occurring microorganisms that confer health benefits by altering host commensal microbiota, modulating immunity, enhancing intestinal barrier function, or altering pain perception. Enterococci are human and animal intestinal commensals that are used as probiotics and in food production. These microorganisms, however, express many virulence traits including cytolysin, proteases, aggregation substance, capsular polysaccharide, enterococcal surface protein, biofilm formation, extracellular superoxide, intestinal translocation, and resistance to innate immunity that can lead to serious hospital-acquired infections. In addition, enterococci are facile in acquiring antibiotic resistance genes to many clinically important antibiotics encoded on a wide variety of conjugative plasmids, transposons, and bacteriophages. The pathogenicity and disease burden caused by enterococci render them poor choices as probiotics. No large, randomized, placebo-controlled clinical trials have demonstrated the safety and efficacy of any enterococcal probiotic. As a result, no enterococcal probiotic has been approved by the United States Food and Drug Administration for the treatment, cure, or amelioration of human disease. In 2007, the European Food Safety Authority concluded that enterococci do not meet the standard for "Qualified Presumption of Safety". Enterococcal strains used or proposed for use as probiotics should be carefully screened for efficacy and safety.


Subject(s)
Enterococcus/metabolism , Food Contamination/analysis , Probiotics/adverse effects , Cytotoxins/genetics , Cytotoxins/metabolism , Drug Resistance, Microbial/genetics , Enterococcus/genetics , Enterococcus/isolation & purification , Enterococcus faecalis/isolation & purification , Enterococcus faecalis/metabolism , Food Microbiology , Food Safety , Genetic Loci , Immunity, Innate , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Perforin/genetics , Perforin/metabolism , Risk Factors , United States , United States Food and Drug Administration , Virulence Factors/genetics
17.
Methods Mol Biol ; 2033: 1-14, 2019.
Article in English | MEDLINE | ID: mdl-31332743

ABSTRACT

Antibody-drug conjugates (ADCs) have been proven to be a successful therapeutic concept, allowing targeted delivery of highly potent active pharmaceutical ingredients (HPAPIs) selectively to tumor tissue. So far, HPAPIs have been mainly attached to the antibody via a chemical reaction of the payload with lysine or cysteine side chains of the antibody backbone. However, these conventional conjugation technologies result in formation of rather heterogeneous products with undesired properties. To overcome the limitations of heterogeneous ADC mixtures, several site-specific conjugation technologies have been developed over the last years. Originally pioneered by scientist from Genentech with their work on THIOMABs, several engineered cysteine mAb ADCs (ECM-ADCs) are now investigated in clinical trials. Here, we describe in detail how to engineer additional cysteines into antibodies and efficiently use them as highly site-specific conjugation sites for HPAPIs.


Subject(s)
Antibodies, Monoclonal, Humanized/genetics , Cytotoxins/genetics , Immunoconjugates/genetics , Protein Engineering , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological , Cell Proliferation/drug effects , Cysteine/chemistry , Cysteine/genetics , Cytotoxins/chemistry , Cytotoxins/immunology , Cytotoxins/pharmacology , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunoconjugates/pharmacology , Mice , Mutagenesis, Site-Directed , Sulfhydryl Compounds/chemistry , Trastuzumab/chemistry , Trastuzumab/genetics , Trastuzumab/immunology , Xenograft Model Antitumor Assays
18.
Microbiology (Reading) ; 165(8): 876-890, 2019 08.
Article in English | MEDLINE | ID: mdl-31162026

ABSTRACT

Diphtheria toxin is one of the best investigated bacterial toxins and the major virulence factor of toxigenic Corynebacterium diphtheriae and Corynebacterium ulcerans strains. However, also diphtheria toxin-free strains of these two species can cause severe infections in animals and humans, indicating the presence of additional virulence factors. In this study, we present a first characterization of two proteins with cytotoxic effect in corynebacteria. A putative ribosome-binding protein (AEG80717, CULC809_00177), first annotated in a genome sequencing project of C. ulcerans strain 809, was investigated in detail together with a homologous protein identified in C. diphtheriae strain HC04 (AEX80148, CDHC04_0155) in this study. The corresponding proteins show striking structural similarity to Shiga-like toxins. Interaction of wild-type, mutant and complementation as well as overexpression strains with invertebrate model systems and cell lines were investigated. Depending on the presence of the corresponding genes, detrimental effects were observed in vivo in two invertebrate model systems, Caenorhabditis elegans and Galleria mellonella, and on various animal and human epithelial and macrophage cell lines in vitro. Taken together, our results support the idea that pathogenicity of corynebacteria is a multifactorial process and that new virulence factors may influence the outcome of potentially fatal corynebacterial infections.


Subject(s)
Corynebacterium diphtheriae/genetics , Corynebacterium/genetics , Cytotoxins/biosynthesis , Exotoxins/genetics , Virulence Factors/genetics , Animals , Bacterial Proteins/biosynthesis , Corynebacterium/pathogenicity , Corynebacterium Infections/microbiology , Corynebacterium diphtheriae/pathogenicity , Cytotoxins/genetics , Diphtheria/microbiology , Diphtheria Toxin , Exotoxins/biosynthesis , Humans , Virulence Factors/biosynthesis
19.
Curr Issues Mol Biol ; 32: 435-472, 2019.
Article in English | MEDLINE | ID: mdl-31166177

ABSTRACT

Streptococcus pyogenes (Group A Streptococcus, GAS) is a strictly human bacterial pathogen. Since the mid-1980s, GAS M1T1 clone has been the most prevalent and globally disseminated serotype and is the culprit causing invasive and severe streptococcal infections, urging a better understanding of the emergence of hypervirulent M1T1 clone from an evolutionary perspective. This review highlights the molecular and evolutionary events leading to pandemic M1T1 strains, and discusses the pressure driving the genetic acquisition of novel virulence genes and the selection of hypervirulent isolates in host. By understanding the evolutionary selection and pressures that select and shape the pandemic M1T1 clone, we could potentially develop new therapeutic strategies to tackle challenges when dealing with the globally disseminated M1T1 GAS clone.


Subject(s)
Gene Expression Regulation, Bacterial , Genome, Bacterial , Pandemics , Streptococcal Infections/epidemiology , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Clone Cells , Cytotoxins/genetics , Cytotoxins/metabolism , Deoxyribonuclease I/genetics , Deoxyribonuclease I/metabolism , Evolution, Molecular , Exotoxins/genetics , Exotoxins/metabolism , Host-Pathogen Interactions/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Recombination, Genetic , Selection, Genetic , Streptococcal Infections/microbiology , Streptococcal Infections/pathology , Streptococcus Phages/genetics , Streptococcus Phages/metabolism , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/virology , Transduction, Genetic , Virulence
20.
Curr Issues Mol Biol ; 32: 521-560, 2019.
Article in English | MEDLINE | ID: mdl-31166179

ABSTRACT

Streptococci are common human pathogens, colonizing multiple parts of the human body such as the upper respiratory tract, urethra, gastrointestinal tract, and oral cavity. Since they cause a variety of serious infections including heart diseases, meningitis, and oral diseases, streptococci are considered to play an important role in human diseases. Two critical steps in the pathogenesis of streptococcal infection are the adhesion to and invasion of host cells. This invasion is a strategy of streptococci to evade the host immune response and antibiotic therapy, as well as to penetrate to deeper tissues. To establish interaction between bacteria and host cells, adhesion is the initial step. To effectively adhere to host cells, streptococci express multiple adhesins, and the expression of different adhesins may lead to distinct mechanisms of subsequent invasion. The binding of streptococcal molecules to host proteins triggers downstream signal transduction in the host cells, leading to the uptake of bacteria. In this review, we present the adhesion and invasion mechanisms of different streptococci and the interaction with host cells leading to internalization.


Subject(s)
Adhesins, Bacterial/genetics , Bacterial Adhesion/genetics , Gene Expression Regulation, Bacterial , Streptococcal Infections/microbiology , Streptococcus/genetics , Streptococcus/pathogenicity , Adhesins, Bacterial/metabolism , Biofilms/growth & development , Cytotoxins/genetics , Cytotoxins/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , Fibronectins/genetics , Fibronectins/metabolism , Host-Pathogen Interactions/genetics , Humans , Lung/microbiology , Lung/pathology , Mouth/microbiology , Mouth/pathology , Protein Binding , Signal Transduction , Streptococcal Infections/metabolism , Streptococcal Infections/pathology , Streptococcus/growth & development , Streptococcus/metabolism , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...