Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 524
Filter
1.
Nature ; 603(7900): 315-320, 2022 03.
Article in English | MEDLINE | ID: mdl-35197633

ABSTRACT

Colibactin is a chemically unstable small-molecule genotoxin that is produced by several different bacteria, including members of the human gut microbiome1,2. Although the biological activity of colibactin has been extensively investigated in mammalian systems3, little is known about its effects on other microorganisms. Here we show that colibactin targets bacteria that contain prophages, and induces lytic development through the bacterial SOS response. DNA, added exogenously, protects bacteria from colibactin, as does expressing a colibactin resistance protein (ClbS) in non-colibactin-producing cells. The prophage-inducing effects that we observe apply broadly across different phage-bacteria systems and in complex communities. Finally, we identify bacteria that have colibactin resistance genes but lack colibactin biosynthetic genes. Many of these bacteria are infected with predicted prophages, and we show that the expression of their ClbS homologues provides immunity from colibactin-triggered induction. Our study reveals a mechanism by which colibactin production could affect microbiomes and highlights a role for microbial natural products in influencing population-level events such as phage outbreaks.


Subject(s)
Bacteria , Bacterial Toxins , Peptides , Polyketides , Prophages , Virus Activation , Bacteria/drug effects , Bacteria/virology , Bacterial Toxins/metabolism , Bacterial Toxins/pharmacology , Bacteriolysis/drug effects , Microbial Interactions/drug effects , Peptides/metabolism , Peptides/pharmacology , Polyketides/metabolism , Polyketides/pharmacology , Prophages/drug effects , Prophages/physiology , SOS Response, Genetics/drug effects , Virus Activation/drug effects
2.
Int J Mol Sci ; 23(3)2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35163197

ABSTRACT

Problems connected with biofilm-related infections and antibiotic resistance necessitate the investigation and development of novel treatment strategies. Given their unique characteristics, one of the most promising alternatives to conventional antibiotics are bacteriophages. In the in vitro and in vivo larva model study, we demonstrate that phages vB_SauM-A, vB_SauM-C, and vB_SauM-D are effective antibiofilm agents. The exposure of biofilm to phages vB_SauM-A and vB_SauM-D led to 2-3 log reductions in the colony-forming unit number in most of the multidrug-resistant S. aureus strains. It was found that phage application reduced the formed biofilms independently of the used titer. Moreover, the study demonstrated that bacteriophages are more efficient in biofilm biomass removal and reduction in staphylococci count when compared to the antibiotics used. The scanning electron microscopy analysis results are in line with colony forming unit (CFU) counting but not entirely consistent with crystal violet (CV) staining. Additionally, phages vB_SauM-A, vB_SauM-C, and vB_SauM-D can significantly increase the survival rate and extend the survival time of Galleria mellonella larvae.


Subject(s)
Anti-Bacterial Agents/pharmacology , Staphylococcal Infections/therapy , Staphylococcus aureus/drug effects , Bacteriolysis/drug effects , Bacteriolysis/genetics , Bacteriophages/genetics , Bacteriophages/pathogenicity , Biofilms/drug effects , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/genetics , Genome, Viral/genetics , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Phage Therapy/methods , Staphylococcal Infections/drug therapy , Staphylococcus aureus/growth & development
3.
BMC Microbiol ; 22(1): 3, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34979918

ABSTRACT

BACKGROUND: Members of the Bacillus genus produce a large variety of antimicrobial peptides including linear or cyclic lipopeptides and thiopeptides, that often have a broad spectrum of action against Gram-positive and Gram-negative bacteria. We have recently reported that SF214, a marine isolated strain of Bacillus pumilus, produces two different antimicrobials specifically active against either Staphylococcus aureus or Listeria monocytogenes. The anti-Staphylococcus molecule has been previously characterized as a pumilacidin, a nonribosomally synthesized lipopetide composed of a mixture of cyclic heptapeptides linked to fatty acids of variable length. RESULTS: Our analysis on the anti-Listeria molecule of B. pumilus SF214 indicated that it is a peptide slightly smaller than 10 kDa, produced during the exponential phase of growth, stable at a wide range of pH conditions and resistant to various chemical treatments. The peptide showed a lytic activity against growing but not resting cells of Listeria monocytogenes and appeared extremely specific being inactive also against L. innocua, a close relative of L. monocytogenes. CONCLUSIONS: These findings indicate that the B. pumilus peptide is unusual with respect to other antimicrobials both for its time of synthesis and secretion and for its strict specificity against L. monocytogenes. Such specificity, together with its stability, propose this new antimicrobial as a tool for potential biotechnological applications in the fight against the dangerous food-borne pathogen L. monocytogenes.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Antimicrobial Peptides/pharmacology , Bacillus pumilus/metabolism , Listeria monocytogenes/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/metabolism , Bacillus pumilus/genetics , Bacillus pumilus/growth & development , Bacteriolysis/drug effects , Cell Wall/drug effects , Genome, Bacterial/genetics , Hot Temperature , Hydrogen-Ion Concentration , Listeria monocytogenes/growth & development , Molecular Weight , Protein Stability , Species Specificity
4.
Sci Rep ; 12(1): 1245, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35075218

ABSTRACT

Bacteriophage endolysins degrade the bacterial cell wall and are therefore considered promising antimicrobial alternatives to fight pathogens resistant to conventional antibiotics. Gram-positive bacteria are usually considered easy targets to exogenously added endolysins, since their cell walls are not shielded by an outer membrane. However, in nutrient rich environments these bacteria can also tolerate endolysin attack if they keep an energized cytoplasmic membrane. Hence, we have hypothesized that the membrane depolarizing action of antimicrobial peptides (AMPs), another attractive class of alternative antibacterials, could be explored to overcome bacterial tolerance to endolysins and consequently improve their antibacterial potential. Accordingly, we show that under conditions supporting bacterial growth, Staphylococcus aureus becomes much more susceptible to the bacteriolytic action of endolysins if an AMP is also present. The bactericidal gain resulting from the AMP/endolysin combined action ranged from 1 to 3 logs for different S. aureus strains, which included drug-resistant clinical isolates. In presence of an AMP, as with a reduced content of cell wall teichoic acids, higher endolysin binding to cells is observed. However, our results indicate that this higher endolysin binding alone does not fully explain the higher susceptibility of S. aureus to lysis in these conditions. Other factors possibly contributing to the increased endolysin susceptibility in presence of an AMP are discussed.


Subject(s)
Antimicrobial Peptides/pharmacology , Bacteriolysis/drug effects , Endopeptidases/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Viral Proteins/pharmacology , Drug Synergism , Microbial Sensitivity Tests , Teichoic Acids
5.
FEBS Open Bio ; 11(3): 705-713, 2021 03.
Article in English | MEDLINE | ID: mdl-33480189

ABSTRACT

Combinations of human lysozyme (hLYS) and antimicrobial peptides (AMPs) are known to exhibit either additive or synergistic activity, and as a result, they have therapeutic potential for persistent and antibiotic-resistant infections. We examined hLYS activity against Pseudomonas aeruginosa when combined with six different AMPs. In contrast to prior reports, we discovered that some therapeutically relevant AMPs manifest striking antagonistic interactions with hLYS across particular concentration ranges. We further found that the synthetic AMP Tet009 can inhibit hLYS-mediated bacterial lysis. To the best of our knowledge, these results represent the first observations of antagonism between hLYS and AMPs, and they advise that future development of lytic enzyme and AMP combination therapies considers the potential for antagonistic interactions.


Subject(s)
Antimicrobial Peptides/pharmacology , Muramidase/adverse effects , Pseudomonas aeruginosa/growth & development , Antimicrobial Peptides/chemistry , Bacteriolysis/drug effects , Drug Antagonism , Humans , Pseudomonas aeruginosa/drug effects
6.
J Appl Microbiol ; 130(6): 1902-1912, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33107183

ABSTRACT

AIMS: The objective of this work was to study the antibacterial specificity and antibacterial effect of endolysins isolated from colibacteriophages RB43, RB49 and T5-as manifested on the exponential and stationary cell cultures of diverse bacteria depending on the growth stage, structure of peptidoglycan (PG) and antibiotic resistance. METHODS AND RESULTS: Enzyme activity was assayed by the spectrophotometric method. Antimicrobial activity was estimated by the number of colony forming units (CFUs), with the results represented as logarithmic units. Morphological examination of bacterial cells was conducted using phase-contrast and scanning electron microscopy. The enzymes EndoT5, endolysin of bacteriophage T5, EndoRB43, endolysin of bacteriophage RB43 and EndoRB49, endolysin of bacteriophage RB49 turned out to be much less bacteriospecific than the corresponding Escherichia coli phages; they lysed bacteria of the genera Bacillus, Cellulomonas and Sporosarcina, whose PGs had different structures (A1γ, A4α and A4ß) and chemical modifications (amidation). The specific lytic activity of phage enzymes was independent of the antibiotic resistance of bacterial cells and was higher when the cells were in the exponential, rather than stationary, growth phase. The analysis of morphological changes showed that the intermediate stage of the endolysin-induced lysis of bacterial cells was the formation of spheroplasts and protoplasts. CONCLUSIONS: Endolysins of colibacteriophages RB49, RB43 and T5 have a wide spectrum of antibacterial action, which includes a number of diverse micro-organisms with different PG structures. SIGNIFICANCE AND IMPACT OF THE STUDY: This is a study of the bacterial selectivity of enzymes degrading bacterial cell wall in relation to the chemical structure of PG. It is shown that endolysins of bacteriophages RB49 and RB43 efficiently lyse cell wall of Gram-positive bacteria of the genus Bacillus and Gram-negative bacteria of the genus Pseudomonas (including an antibiotic-resistant strain). The number of bacterial cells is reduced by 3-6 orders of magnitude, which indicates good prospects for using these enzymes in biotechnology.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Bacteria/drug effects , Bacteriolysis/drug effects , Coliphages/enzymology , Endopeptidases/isolation & purification , Anti-Bacterial Agents/pharmacology , Bacteria/chemistry , Bacteria/classification , Bacteria/cytology , Biotechnology , Cell Wall/chemistry , Coliphages/classification , Endopeptidases/pharmacology , Peptidoglycan/analysis
7.
Biochimie ; 180: 158-168, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33181225

ABSTRACT

Hydroxychavicol (HC), found abundantly in Piper betle leaves is credited with antimicrobial property. Previously we had shown HC induces reactive oxygen species mediated DNA damage in bacterial cells. HC also resulted in membrane compromise revealing its pleiotropic effects on cellular targets. The kinetics and exact sequence of events leading to inhibition of growth and cell death in E. coli after HC treatment remains poorly understood. We show that sub-lethal concentration (125 µg/mL) of HC causes cellular filamentation within 1 h of treatment, while a higher concentration (750 µg/mL) induces cell breakage. HC-treated cells were found to experience oxidative stress as early as 10 min, while evidence of membrane damage was apparent at 30 min. DNA damage repair genes were found to be activated at 60 min. Interestingly, HC-induced cell permeabilization was inhibited and enhanced by external Mg2+ and EDTA, respectively, suggesting that HC damages the outer membrane. Kinetic experiments revealed that HC-treated cells underwent oxidative stress, membrane damage and DNA damage in that order. Because gram negative bacteria such as E. coli are refractory to several antibiotics due to the presence of the outer membrane, we hypothesized that HC pretreatment would sensitize E. coli to hydrophobic antibiotics. Our study reveals for the first time that HC could sensitize bacteria to clinically used antibiotics due to its outer membrane damaging property.


Subject(s)
Anti-Infective Agents/pharmacology , Escherichia coli/drug effects , Eugenol/analogs & derivatives , Animals , Anti-Bacterial Agents/pharmacology , Bacteriolysis/drug effects , Cell Membrane/drug effects , DNA Damage , DNA Repair/drug effects , Edetic Acid/pharmacology , Escherichia coli/cytology , Escherichia coli/ultrastructure , Eugenol/chemistry , Eugenol/pharmacology , Hydrophobic and Hydrophilic Interactions , Kinetics , Magnesium/pharmacology , Mice , Microbial Sensitivity Tests , Microbial Viability/drug effects , Reactive Oxygen Species/metabolism
8.
mBio ; 11(6)2020 12 15.
Article in English | MEDLINE | ID: mdl-33323510

ABSTRACT

Streptococcus pneumoniae, a major cause of pneumonia, sepsis, and meningitis worldwide, has the nasopharynges of small children as its main ecological niche. Depletion of pneumococci from this niche would reduce the disease burden and could be achieved using small molecules with narrow-spectrum antibacterial activity. We identified the alkylated dicyclohexyl carboxylic acid 2CCA-1 as a potent inducer of autolysin-mediated lysis of S. pneumoniae, while having low activity against Staphylococcus aureus 2CCA-1-resistant strains were found to have inactivating mutations in fakB3, known to be required for uptake of host polyunsaturated fatty acids, as well as through inactivation of the transcriptional regulator gene fabT, vital for endogenous, de novo fatty acid synthesis regulation. Structure activity relationship exploration revealed that, besides the central dicyclohexyl group, the fatty acid-like structural features of 2CCA-1 were essential for its activity. The lysis-inducing activity of 2CCA-1 was considerably more potent than that of free fatty acids and required growing bacteria, suggesting that 2CCA-1 needs to be metabolized to exert its antimicrobial activity. Total lipid analysis of 2CCA-1 treated bacteria identified unique masses that were modeled to 2CCA-1 containing lysophosphatidic and phosphatidic acid in wild-type but not in fakB3 mutant bacteria. This suggests that 2CCA-1 is metabolized as a fatty acid via FakB3 and utilized as a phospholipid building block, leading to accumulation of toxic phospholipid species. Analysis of FabT-mediated fakB3 expression elucidates how the pneumococcus could ensure membrane homeostasis and concurrent economic use of host-derived fatty acids.IMPORTANCE Fatty acid biosynthesis is an attractive antibiotic target, as it affects the supply of membrane phospholipid building blocks. In Streptococcus pneumoniae, it is not sufficient to target only the endogenous fatty acid synthesis machinery, as uptake of host fatty acids may bypass this inhibition. Here, we describe a small-molecule compound, 2CCA-1, with potent bactericidal activity that upon interactions with the fatty acid binding protein FakB3, which is present in a limited number of Gram-positive species, becomes metabolized and incorporated as a toxic phospholipid species. Resistance to 2CCA-1 developed specifically in fakB3 and the regulatory gene fabT These mutants reveal a regulatory connection between the extracellular polyunsaturated fatty acid metabolism and endogenous fatty acid synthesis in S. pneumoniae, which could ensure balance between efficient scavenging of host polyunsaturated fatty acids and membrane homeostasis. The data might be useful in the identification of narrow-spectrum treatment strategies to selectively target members of the Lactobacillales such as S. pneumoniae.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carboxylic Acids/pharmacology , Fatty Acids, Unsaturated/biosynthesis , Fatty Acids/pharmacology , Streptococcus pneumoniae/drug effects , Anti-Bacterial Agents/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriolysis/drug effects , Biosynthetic Pathways/drug effects , Carboxylic Acids/chemistry , Drug Resistance, Bacterial , Fatty Acids/chemistry , Gene Expression Regulation, Bacterial , Mutation , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Streptococcus pneumoniae/cytology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism
9.
Eur J Pharm Biopharm ; 151: 181-188, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32315699

ABSTRACT

The increasing prevalence of multidrug resistant bacteria has warranted the search for new antimicrobial agents as existing antibiotics lose their potency. Among these, bacteriophage therapy, as well as the administration of specific bacteriolysis agents, i.e., lytic enzymes, have emerged as attractive alternatives. Nebulizers offer the possibility for delivering these therapeutics directly to the lung, which is particularly advantageous as a non-invasive and direct route to treat bacterial lung infections. Nevertheless, nebulizers can often result in significant degradation of the bacteriophage or protein, both structurally and functionally, due to the large stresses the aerosolization process imposes on these entities. In this work, we assess the capability of a novel low-cost and portable hybrid surface and bulk acoustic wave platform (HYDRA) to nebulize a Myoviridae bacteriophage (phage K) and lytic enzyme (lysostaphin) that specifically targets Staphylococcus aureus. Besides its efficiency in producing phage or protein-laden aerosols within the 1-5 µm respirable range for optimum delivery to the lower respiratory tract where lung infections commonly take place, we observe that the HYDRA platform-owing to the efficiency of driving the aerosolization process at relatively low powers and high frequencies (approximately 10 MHz)-does not result in appreciable denaturation of the phages or proteins, such that the loss of antimicrobial activity following nebulization is minimized. Specifically, a low (0.1 log10 (pfu/ml)) titer loss was obtained with the phages, resulting in a high viable respirable fraction of approximately 90%. Similarly, minimal loss of antimicrobial activity was obtained with lysostaphin upon nebulization wherein its minimum inhibitory concentration (0.5 µg/ml) remained unaltered as compared with the non-nebulized control. These results therefore demonstrate the potential of the HYDRA nebulization platform as a promising strategy for pulmonary administration of alternative antimicrobial agents to antibiotics for the treatment of lung diseases caused by pathogenic bacteria.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemistry , Lung/drug effects , Staphylococcus aureus/drug effects , Acoustics , Administration, Inhalation , Aerosols/administration & dosage , Aerosols/chemistry , Bacteriolysis/drug effects , Bacteriophages/drug effects , Microbial Sensitivity Tests/methods , Myoviridae/drug effects , Nebulizers and Vaporizers , Staphylococcal Infections/drug therapy
10.
Nat Chem Biol ; 16(1): 24-30, 2020 01.
Article in English | MEDLINE | ID: mdl-31686030

ABSTRACT

Lysostaphin is a bacteriolytic enzyme targeting peptidoglycan, the essential component of the bacterial cell envelope. It displays a very potent and specific activity toward staphylococci, including methicillin-resistant Staphylococcus aureus. Lysostaphin causes rapid cell lysis and disrupts biofilms, and is therefore a therapeutic agent of choice to eradicate staphylococcal infections. The C-terminal SH3b domain of lysostaphin recognizes peptidoglycans containing a pentaglycine crossbridge and has been proposed to drive the preferential digestion of staphylococcal cell walls. Here we elucidate the molecular mechanism underpinning recognition of staphylococcal peptidoglycan by the lysostaphin SH3b domain. We show that the pentaglycine crossbridge and the peptide stem are recognized by two independent binding sites located on opposite sides of the SH3b domain, thereby inducing a clustering of SH3b domains. We propose that this unusual binding mechanism allows synergistic and structurally dynamic recognition of S. aureus peptidoglycan and underpins the potent bacteriolytic activity of this enzyme.


Subject(s)
Lysostaphin/chemistry , Peptidoglycan/chemistry , Staphylococcus aureus/chemistry , Bacteriolysis/drug effects , Biofilms , Cell Wall/chemistry , Chromatography, High Pressure Liquid , DNA Mutational Analysis , Glycine/chemistry , Ligands , Magnetic Resonance Spectroscopy , Mutagenesis, Site-Directed , Peptides/chemistry , Protein Binding , Protein Domains , Recombinant Proteins/chemistry , src Homology Domains
11.
Pol J Microbiol ; 68(4): 477-491, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31880892

ABSTRACT

This study explored a potential treatment against methicillin-resistant Staphylococcus aureus (MRSA) infections that combines thioridazine (TZ), an efflux pump inhibitor, and miconazole (MCZ), an autolysis inducer, with the anti-microbial drug cloxacillin (CXN). In vitro, the combination treatment of TZ and MCZ significantly reduced 4096-fold (Σ (FIC) = 0.1 - 1.25) the MIC value of CXN against S. aureus. In vivo, the combination therapy significantly relieved breast redness and swelling in mice infected with either clinical or standard strains of S. aureus. Meanwhile, the number of bacteria isolated from the MRSA135-infected mice decreased significantly (p = 0.0427 < 0.05) after the combination therapy when compared to monotherapy. Moreover, the number of bacteria isolated from the mice infected with a reference S. aureus strain also decreased significantly (p = 0.0191 < 0.05) after the combination therapy when compared to monotherapy. The pathological changes were more significant in the CXN-treated group when compared to mice treated with a combination of three drugs. In addition, we found that combination therapy reduced the release of the bacteria-stimulated cytokines such as IL-6, IFN-γ, and TNF-α. Cytokine assays in serum revealed that CXN alone induced IL-6, IFN-γ, and TNF-α in the mouse groups infected with ATCC 29213 or MRSA135, and the combination of these three drugs significantly reduced IL-6, IFN-γ, and TNF-α concentrations. Also, the levels of TNF-α and IFN-γ in mice treated with a combination of three drugs were significantly lower than in the CXN-treated group. Given the synergistic antibacterial activity of CXN, we concluded that the combination of CXN with TZ, and MCZ could be developed as a novel therapeutic strategy against S. aureus.This study explored a potential treatment against methicillin-resistant Staphylococcus aureus (MRSA) infections that combines thioridazine (TZ), an efflux pump inhibitor, and miconazole (MCZ), an autolysis inducer, with the anti-microbial drug cloxacillin (CXN). In vitro, the combination treatment of TZ and MCZ significantly reduced 4096-fold (Σ (FIC) = 0.1 ­ 1.25) the MIC value of CXN against S. aureus. In vivo, the combination therapy significantly relieved breast redness and swelling in mice infected with either clinical or standard strains of S. aureus. Meanwhile, the number of bacteria isolated from the MRSA135-infected mice decreased significantly (p = 0.0427 < 0.05) after the combination therapy when compared to monotherapy. Moreover, the number of bacteria isolated from the mice infected with a reference S. aureus strain also decreased significantly (p = 0.0191 < 0.05) after the combination therapy when compared to monotherapy. The pathological changes were more significant in the CXN-treated group when compared to mice treated with a combination of three drugs. In addition, we found that combination therapy reduced the release of the bacteria-stimulated cytokines such as IL-6, IFN-γ, and TNF-α. Cytokine assays in serum revealed that CXN alone induced IL-6, IFN-γ, and TNF-α in the mouse groups infected with ATCC 29213 or MRSA135, and the combination of these three drugs significantly reduced IL-6, IFN-γ, and TNF-α concentrations. Also, the levels of TNF-α and IFN-γ in mice treated with a combination of three drugs were significantly lower than in the CXN-treated group. Given the synergistic antibacterial activity of CXN, we concluded that the combination of CXN with TZ, and MCZ could be developed as a novel therapeutic strategy against S. aureus.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Drug Resistance, Bacterial , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , beta-Lactams/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriolysis/drug effects , Cloxacillin/pharmacology , Drug Therapy, Combination , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Miconazole/pharmacology , Microbial Sensitivity Tests , Staphylococcal Infections/drug therapy , Staphylococcus aureus/cytology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Thioridazine/pharmacology
12.
Viruses ; 11(10)2019 09 20.
Article in English | MEDLINE | ID: mdl-31546996

ABSTRACT

The lytic cassette of Lactococcus lactis prophage TP712 contains a putative membrane protein of unknown function (Orf54), a holin (Orf55), and a modular endolysin with a N-terminal glycoside hydrolase (GH_25) catalytic domain and two C-terminal LysM domains (Orf56, LysTP712). In this work, we aimed to study the mode of action of the endolysin LysTP712. Inducible expression of the holin-endolysin genes seriously impaired growth. The growth of lactococcal cells overproducing the endolysin LysTP712 alone was only inhibited upon the dissipation of the proton motive force by the pore-forming bacteriocin nisin. Processing of a 26-residues signal peptide is required for LysTP712 activation, since a truncated version without the signal peptide did not impair growth after membrane depolarization. Moreover, only the mature enzyme displayed lytic activity in zymograms, while no lytic bands were observed after treatment with the Sec inhibitor sodium azide. LysTP712 might belong to the growing family of multimeric endolysins. A C-terminal fragment was detected during the purification of LysTP712. It is likely to be synthesized from an alternative internal translational start site located upstream of the cell wall binding domain in the lysin gene. Fractions containing this fragment exhibited enhanced activity against lactococcal cells. However, under our experimental conditions, improved in vitro inhibitory activity of the enzyme was not observed upon the supplementation of additional cell wall binding domains in. Finally, our data pointed out that changes in the lactococcal cell wall, such as the degree of peptidoglycan O-acetylation, might hinder the activity of LysTP712. LysTP712 is the first secretory endolysin from a lactococcal phage described so far. The results also revealed how the activity of LysTP712 might be counteracted by modifications of the bacterial peptidoglycan, providing guidelines to exploit the biotechnological potential of phage endolysins within industrially relevant lactococci and, by extension, other bacteria.


Subject(s)
Endopeptidases/metabolism , Lactococcus lactis/virology , Prophages/physiology , Siphoviridae/physiology , Acetylation , Bacteriolysis/drug effects , Cell Wall/metabolism , Endopeptidases/chemistry , Endopeptidases/genetics , Lactococcus lactis/growth & development , Lactococcus lactis/metabolism , Nisin/pharmacology , Peptidoglycan/metabolism , Prophages/genetics , Prophages/metabolism , Protein Domains , Protein Sorting Signals , Siphoviridae/genetics , Siphoviridae/metabolism
13.
Can J Microbiol ; 65(12): 895-903, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31479619

ABSTRACT

Bacteriocins are antimicrobial peptides, produced by Gram-positive bacteria such as lactococci and staphylococci, that have limited bactericidal action against Gram-negative bacteria. The aim of this paper was to study the sensitivity of three strains of Escherichia coli to bacteriocins: nisin (as Nisaplin®) and two staphylococcal peptides (warnerin and hominin) during sucrose-induced osmotic stress. We found that all peptides in a 0.3 g·mL-1 sucrose solution significantly reduced the number of viable E. coli. The most pronounced antibacterial effect was achieved by nisin against E. coli K-12 (3 log reduction). Slightly less bactericidal effects were observed with warnerin (1 mg·mL-1) and hominin (1 mg·mL-1) in sucrose solution. The lytic activity of staphylococcal peptides was detected by decreased optical density and viable cell counts. Moreover, it was confirmed by the increased amount of DNA and protein in the medium and the morphological changes detected by atomic force microscopy after 20 h of treatment. Zymographic analysis revealed the release of lytic enzymes from E. coli cells after treatment with staphylococcal peptides and sucrose. These results indicated that the antimicrobial action of peptides can be extended to Gram-negative bacteria via combination with high concentrations of sucrose.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteriocins/pharmacology , Escherichia coli/drug effects , Osmotic Pressure , Sucrose/pharmacology , Anti-Bacterial Agents/metabolism , Bacteriocins/metabolism , Bacteriolysis/drug effects , Gram-Positive Bacteria/metabolism , Microbial Viability/drug effects
14.
PLoS Pathog ; 15(9): e1008044, 2019 09.
Article in English | MEDLINE | ID: mdl-31518377

ABSTRACT

ß-lactam antibiotics interfere with cross-linking of the bacterial cell wall, but the killing mechanism of this important class of antibiotics is not fully understood. Serendipitously we found that sub-lethal doses of ß-lactams rescue growth and prevent spontaneous lysis of Staphylococcus aureus mutants lacking the widely conserved chaperone ClpX, and we reasoned that a better understanding of the clpX phenotypes could provide novel insights into the downstream effects of ß-lactam binding to the PBP targets. Super-resolution imaging revealed that clpX cells display aberrant septum synthesis, and initiate daughter cell separation prior to septum completion at 30°C, but not at 37°C, demonstrating that ClpX becomes critical for coordinating the S. aureus cell cycle as the temperature decreases. FtsZ localization and dynamics were not affected in the absence of ClpX, suggesting that ClpX affects septum formation and autolytic activation downstream of Z-ring formation. Interestingly, oxacillin antagonized the septum progression defects of clpX cells and prevented lysis of prematurely splitting clpX cells. Strikingly, inhibitors of wall teichoic acid (WTA) biosynthesis that work synergistically with ß-lactams to kill MRSA synthesis also rescued growth of the clpX mutant, as did genetic inactivation of the gene encoding the septal autolysin, Sle1. Taken together, our data support a model in which Sle1 causes premature splitting and lysis of clpX daughter cells unless Sle1-dependent lysis is antagonized by ß-lactams or by inhibiting an early step in WTA biosynthesis. The finding that ß-lactams and inhibitors of WTA biosynthesis specifically prevent lysis of a mutant with dysregulated autolytic activity lends support to the idea that PBPs and WTA biosynthesis play an important role in coordinating cell division with autolytic splitting of daughter cells, and that ß-lactams do not kill S. aureus simply by weakening the cell wall.


Subject(s)
Bacterial Proteins/physiology , Endopeptidase Clp/physiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriolysis/drug effects , Bacteriolysis/physiology , Cell Wall/drug effects , Cell Wall/metabolism , Cytoskeletal Proteins/metabolism , Endopeptidase Clp/genetics , Humans , Models, Biological , Mutation , Oxacillin/pharmacology , Staphylococcus aureus/genetics , Teichoic Acids/biosynthesis , Tunicamycin/pharmacology , beta-Lactams/pharmacology
15.
Future Microbiol ; 14: 949-955, 2019 07.
Article in English | MEDLINE | ID: mdl-31373220

ABSTRACT

Aim: Bile salts promote the specific autolysis of pneumococcal cells, allowing the differentiation between Streptococcus pneumoniae and other viridans group streptococci (VGS). Material & methods: One hundred clinical VGS isolates identified by amplification and sequencing of 16S rRNA, groEL and sodA genes were analyzed with different variants of bile-solubility tests: tube testing read by naked eye; tube testing where the lysis was measured as the decrease of turbidity with a densitometer; and direct testing on blood agar plate. Results: As expected, all S. pneumoniae isolates were fully lysed in the presence of bile salts except for one isolate that partially lysate in tube testing as well as on the blood agar plate. None of the VGS were lysed by bile salts. Conclusion: Bile-solubility testing is an accurate and technically nondemanding method to discriminate between S. pneumoniae and other VGS species.


Subject(s)
Bacteriological Techniques/methods , Bile Acids and Salts , Streptococcal Infections/diagnosis , Streptococcal Infections/microbiology , Streptococcus pneumoniae/isolation & purification , Viridans Streptococci/isolation & purification , Bacteriolysis/drug effects , Bile Acids and Salts/pharmacology , Diagnosis, Differential , Humans , Sensitivity and Specificity , Solubility
16.
Elife ; 82019 04 09.
Article in English | MEDLINE | ID: mdl-30964003

ABSTRACT

Penicillin and related antibiotics disrupt cell wall synthesis to induce bacteriolysis. Lysis in response to these drugs requires the activity of cell wall hydrolases called autolysins, but how penicillins misactivate these deadly enzymes has long remained unclear. Here, we show that alterations in surface polymers called teichoic acids (TAs) play a key role in penicillin-induced lysis of the Gram-positive pathogen Streptococcus pneumoniae (Sp). We find that during exponential growth, Sp cells primarily produce lipid-anchored TAs called lipoteichoic acids (LTAs) that bind and sequester the major autolysin LytA. However, penicillin-treatment or prolonged stationary phase growth triggers the degradation of a key LTA synthase, causing a switch to the production of wall-anchored TAs (WTAs). This change allows LytA to associate with and degrade its cell wall substrate, thus promoting osmotic lysis. Similar changes in surface polymer assembly may underlie the mechanism of antibiotic- and/or growth phase-induced lysis for other important Gram-positive pathogens.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteriolysis/drug effects , Biosynthetic Pathways/drug effects , Penicillins/pharmacology , Streptococcus pneumoniae/drug effects , Teichoic Acids/biosynthesis
17.
Article in English | MEDLINE | ID: mdl-30863725

ABSTRACT

Streptococcosis is recognized as a leading infectious disease in the swine industry. Streptococcus suis serotype 2 is regarded as the most virulent species, which threatens human and pig health and causes serious economic losses. In this study, multiple in vitro and in vivo effects of MP1102 on multidrug resistant S. suis was studied for the first time. MP1102 exhibited significant antibacterial activity against S. suis (minimum inhibitory concentration, MIC = 0.028-0.228 µM), rapid bacteriocidal action, a longer postantibiotic effect than ceftriaxone, and a synergistic or additive effect with lincomycin, penicillin, and ceftriaxone (FICI = 0.29-0.96). No resistant mutants appeared after 30 serial passages of S. suis in the presence of MP1102. Flow cytometric analysis and electron microscopy observations showed that MP1102 destroyed S. suis cell membrane integrity and affected S. suis cell ultrastructure and membrane morphology. Specifically, a significantly wrinkled surface, intracellular content leakage, and cell lysis were noted, establishing a cyto-basis of nonresistance to this pathogen. DNA gel retardation and circular dichroism analysis indicated that MP1102 interacted with DNA by binding to DNA and changing the DNA conformation, even leading to the disappearance of the helical structure. This result further supported the mechanistic basis of nonresistance via interaction with an intracellular target, which could serve as a means of secondary injury after MP1102 is transported across the membrane. Upon treatment with 2.5-5.0 mg/kg MP1102, the survival of mice challenged with S. suis was 83.3-100%. MP1102 decreased bacterial translocation in liver, lung, spleen, and blood; inhibited the release of interleukin-1ß and tumor necrosis factor-α; and relieved the lung, liver, and spleen from acute injury induced by S. suis. These results suggest that MP1102 is a potent novel antibacterial agent for the treatment of porcine streptococcal disease.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial , Streptococcal Infections/drug therapy , Streptococcal Infections/microbiology , Streptococcus suis/drug effects , Animal Structures/microbiology , Animal Structures/pathology , Animals , Bacteriolysis/drug effects , Cell Membrane/drug effects , Cell Membrane/ultrastructure , DNA, Bacterial/drug effects , Disease Models, Animal , Drug Synergism , Mice , Microbial Sensitivity Tests , Microbial Viability/drug effects , Nucleic Acid Conformation/drug effects , Serogroup , Streptococcal Infections/pathology , Streptococcus suis/classification , Streptococcus suis/physiology , Streptococcus suis/ultrastructure , Survival Analysis
18.
Article in English | MEDLINE | ID: mdl-30670427

ABSTRACT

Bacteriophage-derived lysins are cell-wall-hydrolytic enzymes that represent a potential new class of antibacterial therapeutics in development to address burgeoning antimicrobial resistance. CF-301, the lead compound in this class, is in clinical development as an adjunctive treatment to potentially improve clinical cure rates of Staphylococcus aureus bacteremia and infective endocarditis (IE) when used in addition to antibiotics. In order to profile the activity of CF-301 in a clinically relevant milieu, we assessed its in vitro activity in human blood versus in a conventional testing medium (cation-adjusted Mueller-Hinton broth [caMHB]). CF-301 exhibited substantially greater potency (32 to ≥100-fold) in human blood versus caMHB in three standard microbiologic testing formats (e.g., broth dilution MICs, checkerboard synergy, and time-kill assays). We demonstrated that CF-301 acted synergistically with two key human blood factors, human serum lysozyme (HuLYZ) and human serum albumin (HSA), which normally have no nascent antistaphylococcal activity, against a prototypic methicillin-resistant S. aureus (MRSA) strain (MW2). Similar in vitro enhancement of CF-301 activity was also observed in rabbit, horse, and dog (but not rat or mouse) blood. Two well-established MRSA IE models in rabbit and rat were used to validate these findings in vivo by demonstrating comparable synergistic efficacy with standard-of-care anti-MRSA antibiotics at >100-fold lower lysin doses in the rabbit than in the rat model. The unique properties of CF-301 that enable bactericidal potentiation of antimicrobial activity via activation of "latent" host factors in human blood may have important therapeutic implications for durable improvements in clinical outcomes of serious antibiotic-resistant staphylococcal infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteriolysis/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Staphylococcal Infections/drug therapy , Animals , Bacteremia/drug therapy , Bacteremia/microbiology , Bacteriophages/metabolism , Dogs , Drug Synergism , Endocarditis, Bacterial/drug therapy , Endocarditis, Bacterial/microbiology , Horses/microbiology , Humans , Methicillin/pharmacology , Mice , Microbial Sensitivity Tests/methods , Rabbits , Rats , Staphylococcal Infections/microbiology
19.
J Infect Dis ; 219(9): 1439-1447, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30476337

ABSTRACT

Emergence of multidrug-resistant (MDR) bacterial infections is a major problem in clinical medicine. Development of new strategies such as phage therapy may be a novel approach for treatment of life-threatening infections caused by MDR bacteria. A newly isolated phage, MMI-Ps1, with strong lytic activity was used for treatment of acute lung infection with Pseudomonas aeruginosa in a mouse model. Intranasal administration of a single dose of MMI-Ps1 immediately after infection provided a significant level of protection and increased the survival duration. Moreover, treatment of infected mice with phage as late as 12 hours after infection was still protective. Our in vitro results are the first to show the synergistic elimination of serum-resistant Pseudomonas strains by phage and complement. Phage therapy increases the efficacy of complement-mediated lysis of serum-resistant P. aeruginosa strains, indicating the importance of an intact complement system in clearing Pseudomonas infection during phage therapy.


Subject(s)
Bacteriophages , Complement System Proteins/therapeutic use , Lung Diseases/microbiology , Lung Diseases/therapy , Phage Therapy , Pseudomonas Infections/therapy , Acute Disease , Administration, Intranasal , Animals , Bacteriolysis/drug effects , Caudovirales , Colony Count, Microbial , Complement System Proteins/pharmacology , Disease Models, Animal , Drug Resistance, Multiple, Bacterial , Female , In Vitro Techniques , Lung Diseases/prevention & control , Mice , Mice, Inbred BALB C , Pseudomonas Infections/microbiology , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/physiology
20.
Microb Pathog ; 127: 60-69, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30513367

ABSTRACT

Bacterial infections caused by antibiotic resistant bacteria are the leading cause of morbidity and mortality after burn injuries. This issue has driven the need for promising antimicrobial drugs to eradication of bacterial pathogens. Accordingly, we aimed to determine the therapeutic value of melittin, as a natural Antimicrobial peptide (AMP), in eradication of extensively drug-resistant (XDR) Acinetobacter spp. on a mouse model of third degree burn infection. Melittin killed all examined XDR isolates at 4 µg/mL up to 3 h. Melittin caused significant fluorescence release from XDR isolates at the minimum dose of 0.062 µg/mL. Vesicle formation on the membrane and squeezing of bacteria followed by cell lysis indicated the membranolytic effect of melittin. Melittin at 32 µg/mL completely eradicated the colonized XDR bacteria on infected burn mice during 2 h. No toxicity was observed on injured or healthy derma, as well as circulating Red Blood Cells (RBCs) in the examined mice. Potent promising antibacterial activity of melittin and the lack of toxicity at the therapeutic dose can clarify that melittin can be implemented as a topical drug lead in a preclinical trial of third degree burn infections.


Subject(s)
Acinetobacter Infections/drug therapy , Acinetobacter/drug effects , Anti-Infective Agents/administration & dosage , Burns/complications , Drug Resistance, Multiple, Bacterial , Melitten/administration & dosage , Wound Infection/drug therapy , Acinetobacter Infections/microbiology , Animals , Anti-Infective Agents/adverse effects , Bacteriolysis/drug effects , Cell Membrane/drug effects , Disease Models, Animal , Melitten/adverse effects , Mice , Microbial Viability/drug effects , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...