Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.326
Filter
1.
Nature ; 629(8010): 165-173, 2024 May.
Article in English | MEDLINE | ID: mdl-38632398

ABSTRACT

Streptomyces are a genus of ubiquitous soil bacteria from which the majority of clinically utilized antibiotics derive1. The production of these antibacterial molecules reflects the relentless competition Streptomyces engage in with other bacteria, including other Streptomyces species1,2. Here we show that in addition to small-molecule antibiotics, Streptomyces produce and secrete antibacterial protein complexes that feature a large, degenerate repeat-containing polymorphic toxin protein. A cryo-electron microscopy structure of these particles reveals an extended stalk topped by a ringed crown comprising the toxin repeats scaffolding five lectin-tipped spokes, which led us to name them umbrella particles. Streptomyces coelicolor encodes three umbrella particles with distinct toxin and lectin composition. Notably, supernatant containing these toxins specifically and potently inhibits the growth of select Streptomyces species from among a diverse collection of bacteria screened. For one target, Streptomyces griseus, inhibition relies on a single toxin and that intoxication manifests as rapid cessation of vegetative hyphal growth. Our data show that Streptomyces umbrella particles mediate competition among vegetative mycelia of related species, a function distinct from small-molecule antibiotics, which are produced at the onset of reproductive growth and act broadly3,4. Sequence analyses suggest that this role of umbrella particles extends beyond Streptomyces, as we identified umbrella loci in nearly 1,000 species across Actinobacteria.


Subject(s)
Antibiosis , Bacterial Proteins , Bacterial Toxins , Streptomyces , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Antibiosis/drug effects , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Bacterial Proteins/ultrastructure , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Bacterial Toxins/pharmacology , Cryoelectron Microscopy , Lectins/chemistry , Lectins/genetics , Lectins/metabolism , Lectins/ultrastructure , Microbial Sensitivity Tests , Models, Molecular , Streptomyces/chemistry , Streptomyces/drug effects , Streptomyces/genetics , Streptomyces/growth & development , Streptomyces coelicolor/chemistry , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Streptomyces griseus/drug effects , Streptomyces griseus/genetics , Streptomyces griseus/growth & development , Streptomyces griseus/metabolism
2.
Rheumatology (Oxford) ; 63(3): 882-890, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37481715

ABSTRACT

OBJECTIVE: The p.E148Q variant in pyrin is present in different populations at a frequency of up to 29%, and has been associated with diseases, including vasculitis and FMF. The pathogenicity of p.E148Q in FMF is unclear, even when observed in cis or in trans to a single, typically recessive, pathogenic mutation. We performed functional validation to determine whether p.E148Q increases the ability of pyrin to form an active inflammasome complex in cell lines. METHODS: We interrogated the Australian Autoinflammatory Disease RegistrY (AADRY) to find candidate inheritance patterns for the p.E148Q variant in pyrin. Different pyrin variant combinations were tested in HEK293T cells stably expressing the adaptor protein apoptosis-associated speck-like (ASC), which were analysed by flow cytometry to visualize inflammasome formation, with and without stimulation by Clostridioides difficile toxin B (TcdB). Inflammasome-dependent cytokine secretion was also quantified by ELISA of supernatants from THP-1 cells transduced with lentiviral expression vectors. RESULTS: In AADRY, we observed the p.E148Q allele in individuals with autoinflammatory diseases alone or in conjunction with other pyrin variants. Two FMF families harboured the allele p.E148Q-M694I in cis with dominant heritability. In vitro, p.E148Q pyrin could spontaneously potentiate inflammasome formation, with increased IL-1ß and IL-18 secretion. p.E148Q in cis to classical FMF mutations provided significant potentiation of inflammasome formation. CONCLUSION: The p.E148Q variant in pyrin potentiates inflammasome activation in vitro. In cis, this effect is additive to known pathogenic FMF mutations. In some families, this increased effect could explain why FMF segregates as an apparently dominant disease.


Subject(s)
Inflammasomes , Pyrin , Humans , Australia , Bacterial Toxins/pharmacology , HEK293 Cells , Inflammasomes/genetics , Mutation , Pyrin/genetics
3.
Nature ; 622(7983): 611-618, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37699522

ABSTRACT

Clostridioides difficile infection (CDI) is a major cause of healthcare-associated gastrointestinal infections1,2. The exaggerated colonic inflammation caused by C. difficile toxins such as toxin B (TcdB) damages tissues and promotes C. difficile colonization3-6, but how TcdB causes inflammation is unclear. Here we report that TcdB induces neurogenic inflammation by targeting gut-innervating afferent neurons and pericytes through receptors, including the Frizzled receptors (FZD1, FZD2 and FZD7) in neurons and chondroitin sulfate proteoglycan 4 (CSPG4) in pericytes. TcdB stimulates the secretion of the neuropeptides substance P (SP) and calcitonin gene-related peptide (CGRP) from neurons and pro-inflammatory cytokines from pericytes. Targeted delivery of the TcdB enzymatic domain, through fusion with a detoxified diphtheria toxin, into peptidergic sensory neurons that express exogeneous diphtheria toxin receptor (an approach we term toxogenetics) is sufficient to induce neurogenic inflammation and recapitulates major colonic histopathology associated with CDI. Conversely, mice lacking SP, CGRP or the SP receptor (neurokinin 1 receptor) show reduced pathology in both models of caecal TcdB injection and CDI. Blocking SP or CGRP signalling reduces tissue damage and C. difficile burden in mice infected with a standard C. difficile strain or with hypervirulent strains expressing the TcdB2 variant. Thus, targeting neurogenic inflammation provides a host-oriented therapeutic approach for treating CDI.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Neurogenic Inflammation , Neurons, Afferent , Pericytes , Animals , Mice , Bacterial Toxins/administration & dosage , Bacterial Toxins/pharmacology , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Calcitonin Gene-Related Peptide/metabolism , Clostridioides difficile/pathogenicity , Clostridium Infections/microbiology , Neurogenic Inflammation/chemically induced , Neurogenic Inflammation/microbiology , Neurogenic Inflammation/pathology , Pericytes/drug effects , Pericytes/microbiology , Pericytes/pathology , Receptors, Neurokinin-1/metabolism , Substance P/antagonists & inhibitors , Substance P/metabolism , Neurons, Afferent/drug effects , Neurons, Afferent/microbiology , Neurons, Afferent/pathology , Inflammation Mediators/metabolism , Cecum/drug effects , Cecum/metabolism , Signal Transduction/drug effects
4.
Braz J Med Biol Res ; 56: e12659, 2023.
Article in English | MEDLINE | ID: mdl-37075347

ABSTRACT

Dendritic cells (DCs) are antigen-presenting cells that drive the differentiation of T CD4+ cells into different profiles according to the nature of the antigen or immunomodulator. Propolis is a resinous product made by bees that has numerous pharmacological properties, including an immunomodulatory action. To assess whether propolis can modulate the activation of CD4+ T cells by stimulating DCs with heat-labile enterotoxin B subunit (EtxB) or lipopolysaccharide (LPS), we aimed to elucidate the mechanisms affected by propolis in the differential activation of T lymphocytes. Cell viability, lymphocyte proliferation, gene expression (GATA-3 and RORc), and cytokine production (interleukin (IL)-4 and IL-17A) were analyzed. Propolis, EtxB, and LPS induced a higher lymphoproliferation compared with the control. Propolis induced GATA-3 expression and, in combination with EtxB, maintained the baseline levels. Propolis alone or in combination with LPS inhibited RORc expression. EtxB alone and in combination with propolis increased IL-4 production. Propolis in combination with LPS prevented LPS-induced IL-17A production. These results opened perspectives for the study of biological events that may be favored by propolis by promoting Th2 activation or helping in the treatment of inflammatory conditions mediated by Th17 cells.


Subject(s)
Bacterial Toxins , Escherichia coli Proteins , Propolis , Bacterial Toxins/pharmacology , Lipopolysaccharides/pharmacology , Propolis/pharmacology , Interleukin-17 , Th17 Cells , Escherichia coli Proteins/pharmacology , Dendritic Cells , Th2 Cells
5.
Folia Med (Plovdiv) ; 65(1): 116-123, 2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36855983

ABSTRACT

AIM: The present study was conducted in an attempt to find possible direct mechanisms of action of Clostridium difficile toxins A and B (TCdA and TCdB) on contractility of isolated rat intestinal smooth muscles, as the contractive pathways affected by the toxins and responsible for motility disorders remain unclear. MATERIALS AND METHODS: Adult male Wistar rats were used in our experiments. Longitudinal smooth muscle (SM) preparations of proximal colon were isolated and their contractile activity was isometrically registered. The samples were mounted in tissue baths and exogenously treated with acetylcholine (ACh), serotonin (5-HT), dopamine, norepinephrine, TCdA and TCdB. The potential of TCdA and TCdB to affect the action of these mediators on SM activity was examined. RESULTS: The experiments have shown that exciting action of ACh and 5-HT on colonic contractility is enhanced by TCdA rather than TCdB. Conversely, relaxing effect of dopamine and norepinephrine on contractile activity of colonic SM is under impact of TCdB but not TcdA. TCdA has a stronger direct effect on in vitro SM sensitivity to ACh and 5-HT than TCdB. CONCLUSIONS: TCdA and TCdB affect directly the contractile reactivity of isolated rat colon smooth muscle. TCdA has a stronger direct effect on smooth muscle sensitivity to acetylcholine and 5-HT than TCdB. Such a trend has not been established for dopamine and norepinephrine.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Male , Animals , Rats , Rats, Wistar , Acetylcholine/pharmacology , Bacterial Toxins/pharmacology , Dopamine/pharmacology , Serotonin/pharmacology , Colon , Muscle, Smooth , Norepinephrine/pharmacology
6.
Curr Cancer Drug Targets ; 23(5): 388-399, 2023.
Article in English | MEDLINE | ID: mdl-36424771

ABSTRACT

BACKGROUND: Bacterial pore-forming toxins, BinA and BinB together known as the binary toxin are potent insecticidal proteins, that share structural homology with antitumor bacterial parasporin-2 protein. The underlying molecular mechanism of Bin toxin-induced cancer cell cytotoxicity requires more knowledge to understand whether the toxin induced human cytotoxic effects occur in the same way as that of parasporin-2 or not. METHODS: In this study, anticancer properties of Lysinibacillus sphaericus derived Bin toxin on HK1 were evaluated through MTT assay, morphological analysis and lactate dehydrogenase efflux assay. Induction of apoptosis was determined from RT-qPCR, caspase activity and cytochrome c release assay. Internalization pattern of Bin toxin in HK1 cells was studied by confocal laser-scanning microscopic analysis. RESULTS: Activated Bin toxin had strong cytocidal activity to HK1 cancer cell line at 24 h postinoculation. Both BinA and BinB treated HK1 cells showed significant inhibition of cell viability at 12 µM. Induction of apoptotic mediators from RT-qPCR and caspase activity analyses indicated the activation of programmed cell death in HK1 cells in response to Bin toxin treatment. Internalization pattern of Bin toxin studied by using confocal microscopy indicated the localization of BinA on cell surface and internalization of BinB in the cytoplasm of cancer cells as well as colocalization of BinA with BinB. Evaluation of cytochrome c release also showed the association of BinB and BinA+BinB with mitochondria. CONCLUSION: Bin toxin is a cytotoxic protein that induces cytotoxic and apoptotic events in HK1 cells, and may have high therapeutic potential as an anti-cancer agent.


Subject(s)
Apoptosis , Bacterial Toxins , Cytochromes c , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Humans , Apoptosis/drug effects , Caspases , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Neoplasms/drug therapy , Bacterial Toxins/pharmacology , Bacterial Toxins/therapeutic use
7.
Cells ; 11(22)2022 11 18.
Article in English | MEDLINE | ID: mdl-36429089

ABSTRACT

Clostridioides bacteria are responsible for life threatening infections. Here, we show that in addition to actin, the binary toxins CDT, C2I, and Iota from Clostridioides difficile, botulinum, and perfrigens, respectively, ADP-ribosylate the actin-related protein Arp2 of Arp2/3 complex and its additional components ArpC1, ArpC2, and ArpC4/5. The Arp2/3 complex is composed of seven subunits and stimulates the formation of branched actin filament networks. This activity is inhibited after ADP-ribosylation of Arp2. Translocation of the ADP-ribosyltransferase component of CDT toxin into human colon carcinoma Caco2 cells led to ADP-ribosylation of cellular Arp2 and actin followed by a collapse of the lamellipodial extensions and F-actin network. Exposure of isolated mouse colon pieces to CDT toxin induced the dissolution of the enterocytes leading to luminal aggregation of cellular debris and the collapse of the mucosal organization. Thus, we identify the Arp2/3 complex as hitherto unknown target of clostridial ADP-ribosyltransferases.


Subject(s)
Actin-Related Protein 2-3 Complex , Bacterial Toxins , Animals , Mice , Humans , Actin-Related Protein 2-3 Complex/metabolism , Clostridioides , Actins/metabolism , Bacterial Toxins/pharmacology , Bacterial Toxins/metabolism , Caco-2 Cells , ADP Ribose Transferases/pharmacology , ADP Ribose Transferases/metabolism , ADP-Ribosylation , Adenosine Diphosphate/metabolism
8.
BMC Complement Med Ther ; 22(1): 261, 2022 Oct 07.
Article in English | MEDLINE | ID: mdl-36207726

ABSTRACT

BACKGROUND: As the largest organ, the skin has been frequently affected by trauma, chemical materials, toxins, bacterial pathogens, and free radicals. Recently, many attempts have been made to develop natural nanogels that, besides hydrating the skin, could also be used as antioxidant or antibacterial agents. METHODS: In this study, the chemical composition of the Mentha spicata essential oil was first investigated using GC-MS analysis. Its nanoemulsion-based nanogel was then investigated; successful loading of the essential oil in the nanogel was confirmed using FTIR analysis. Besides, nanogel's antioxidative, anticancer, and antibacterial activities were investigated. RESULTS: Carvone (37.1%), limonene (28.5%), borneol (3.9%), ß-pinene (3.3%), and pulegone (3.3%) were identified as five major compounds in the essential oil. By adding carboxymethylcellulose (3.5% w/v) to the optimal nanoemulsion containing the essential oil (droplet size of 196 ± 8 nm), it was gelified. The viscosity was fully fitted with a common non-Newtonian viscosity regression, the Carreau-Yasuda model. The antioxidant effect of the nanogel was significantly more potent than the essential oil (P < 0.001) at all examined concentrations (62.5-1000 µg/mL). Furthermore, the potency of the nanogel with an IC50 value of 55.0 µg/mL was substantially more (P < 0.001) than the essential oil (997.4 µg/mL). Also, the growth of Staphylococcus aureus and Escherichia coli after treatment with 1000 µg/mL nanogel was about 50% decreased compared to the control group. Besides, the prepared electrospun polycaprolactone-hydroxypropyl methylcellulose nanofibers mat with no cytotoxic, antioxidant, or antibacterial effects was proposed as lesion dressing after treatment with the nanogel. High potency, natural ingredients, and straightforward preparation are advantages of the prepared nanogel. Therefore, it could be considered for further consideration in vivo studies.


Subject(s)
Bacterial Toxins , Mentha spicata , Nanofibers , Oils, Volatile , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Bacterial Toxins/pharmacology , Carboxymethylcellulose Sodium/pharmacology , Escherichia coli , Free Radicals/pharmacology , Hypromellose Derivatives/pharmacology , Limonene/pharmacology , Mentha spicata/chemistry , Microbial Sensitivity Tests , Nanogels , Oils, Volatile/chemistry , Oils, Volatile/pharmacology , Polyesters , Polyethylene Glycols , Polyethyleneimine
9.
Proc Natl Acad Sci U S A ; 119(28): e2201423119, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35867758

ABSTRACT

Treatments for advanced and recurrent ovarian cancer remain a challenge due to a lack of potent, selective, and effective therapeutics. Here, we developed the basis for a transformative anticancer strategy based on anthrax toxin that has been engineered to be selectively activated by the catalytic power of zymogen-activating proteases on the surface of malignant tumor cells to induce cell death. Exposure to the engineered toxin is cytotoxic to ovarian tumor cell lines and ovarian tumor spheroids derived from patient ascites. Preclinical studies demonstrate that toxin treatment induces tumor regression in several in vivo ovarian cancer models, including patient-derived xenografts, without adverse side effects, supportive of progression toward clinical evaluation. These data lay the groundwork for developing therapeutics for treating women with late-stage and recurrent ovarian cancers, utilizing a mechanism distinct from current anticancer therapies.


Subject(s)
Antigens, Bacterial , Antineoplastic Agents , Bacterial Toxins , Ovarian Neoplasms , Prodrugs , Serine Proteases , Antigens, Bacterial/pharmacology , Antigens, Bacterial/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bacterial Toxins/pharmacology , Bacterial Toxins/therapeutic use , Cell Line, Tumor , Enzyme Precursors/metabolism , Female , Humans , Neoplasm Recurrence, Local , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Prodrugs/pharmacology , Prodrugs/therapeutic use , Serine Proteases/metabolism , Spheroids, Cellular , Xenograft Model Antitumor Assays
10.
Insect Biochem Mol Biol ; 147: 103799, 2022 08.
Article in English | MEDLINE | ID: mdl-35662624

ABSTRACT

The Cry48Aa/Cry49Aa toxin of Lysinibacillus sphaericus shows specific toxicity towards larvae of Culex spp. Individual Cry48Aa and Cry49Aa subunits interact with distinct target sites in the larval midgut and overcome the resistance of Culex to the Bin toxin. However, the toxin-binding proteins have not yet been identified. The present study aimed to identify Cry48Aa-binding proteins in Culex quinquefasciatus. Pulldown assays using C. quinquefasciatus midgut brush-border membrane fractions (BBMFs) identified a class of proteins, including aminopeptidases (APNs), protease m1 zinc metalloproteases, alkaline phosphatases (ALPs), and maltases, that could be potentially involved in the mode of action of this toxin. RNA interference analysis showed that silenced larvae treated with dsRNA of the alpha-glucosidase (named Glu71) gene were more tolerant of the Cry48Aa/Cry49Aa toxin, which induced less than 20% mortality. The amino acid sequence of Glu71 exhibited 42% identity with Cqm1/Cpm1, which acted as a Bin toxin receptor. Toxin binding assays showed that Cry48Aa had a high specific binding capacity for the Glu71 protein, whereas Cry49Aa exhibited no specific binding. Overall, our results showed that Glu71 is a Cry48-binding protein involved in Cry48Aa/Cry49Aa toxicity.


Subject(s)
Bacillaceae , Bacillus , Bacterial Toxins , Culex , Animals , Bacillaceae/genetics , Bacillus/metabolism , Bacterial Toxins/metabolism , Bacterial Toxins/pharmacology , Culex/genetics , Culex/metabolism , Larva/genetics , Larva/metabolism , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism
11.
Adv Healthc Mater ; 11(14): e2200036, 2022 07.
Article in English | MEDLINE | ID: mdl-35481905

ABSTRACT

Antibiotic resistance is a severe global health threat and hence demands rapid action to develop novel therapies, including microscale drug delivery systems. Herein, a hierarchical microparticle system is developed to achieve bacteria-activated single- and dual-antibiotic drug delivery for preventing methicillin-resistant Staphylococcus aureus (MRSA) bacterial infections. The designed system is based on a capsosome structure, which consists of a mesoporous silica microparticle coated in alternating layers of oppositely charged polymers and antibiotic-loaded liposomes. The capsosomes are engineered and shown to release their drug payloads in the presence of MRSA toxins controlled by the Agr quorum sensing system. MRSA-activated single drug delivery of vancomycin and synergistic dual delivery of vancomycin together with an antibacterial peptide successfully kills MRSA in vitro. The capability of capsosomes to selectively deliver their cargo in the presence of bacteria, producing a bactericidal effect to protect the host organism, is confirmed in vivo using a Drosophila melanogaster MRSA infection model. Thus, the capsosomes serve as a versatile multidrug, subcompartmentalized microparticle system for preventing antibiotic-resistant bacterial infections, with potential applications to protect wounds or medical device implants from infections.


Subject(s)
Bacterial Toxins , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Animals , Anti-Bacterial Agents/chemistry , Bacterial Toxins/pharmacology , Drosophila melanogaster , Microbial Sensitivity Tests , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Vancomycin/chemistry , Vancomycin/pharmacology
12.
Cytokine ; 153: 155862, 2022 05.
Article in English | MEDLINE | ID: mdl-35306427

ABSTRACT

Group 3 innate lymphocytes (ILC3s) are important immune cells within mucosal tissues and protect against bacterial infections. They can be activated in response to the innate cytokines IL-23 or IL-1ß, which rapidly increases their production of effector molecules that regulate barrier functions. Pathogens can subvert these anti-bacterial effects to evade mucosal defenses to infect the host. Bacillus anthracis, the causative agent of anthrax, produces two major toxins that can modulate the immune response. We have previously shown that lethal toxin downmodulates the function of ILC3s. On the other hand, edema toxin has been shown promote T helper 17 (Th17) cell differentiation, adaptive counterparts of ILC3s, via elevation of cyclic adenosine monophosphate (cAMP). We hypothesized that edema toxin may also modulate ILC3 function. In this study, we show that edema toxin has the opposite effect of lethal toxin; edema toxin directly activates ILC3s independently of innate cytokine stimulation. Treatment of a mouse ILC3-like cell line with edema toxin, a potent adenylate cyclase, upregulated production of the cytokine IL-22, a major effector molecule of ILC3s and a critical factor in maintaining mucosal barriers. Forskolin treatment phenocopied the effect observed with edema toxin and led to an increase in CREB phosphorylation in ILC3s. This observation has potential implications for a role for cAMP signaling in the activation of ILC3s.


Subject(s)
Bacterial Toxins , Animals , Antigens, Bacterial , Bacterial Toxins/pharmacology , Cyclic AMP , Cytokines , Edema , Immunity, Innate , Interleukins , Lymphocytes , Mice , Interleukin-22
13.
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
14.
Sci Rep ; 12(1): 2794, 2022 02 18.
Article in English | MEDLINE | ID: mdl-35181693

ABSTRACT

Type II toxin-antitoxin (TA) systems are widespread in bacteria and are involved in important cell features, such as cell growth inhibition and antimicrobial tolerance, through the induction of persister cells. Overall, these characteristics are associated with bacterial survival under stress conditions and represent a significant genetic mechanism to be explored for antibacterial molecules. We verified that even though Xylella fastidiosa and Xanthomonas citri subsp. citri share closely related genomes, they have different Type II TA system contents. One important difference is the absence of mqsRA in X. citri. The toxin component of this TA system has been shown to inhibit the growth of X. fastidiosa. Thus, the absence of mqsRA in X. citri led us to explore the possibility of using the MqsR toxin to impair X. citri growth. We purified MqsR and confirmed that the toxin was able to inhibit X. citri. Subsequently, transgenic citrus plants producing MqsR showed a significant reduction in citrus canker and citrus variegated chlorosis symptoms caused, respectively, by X. citri and X. fastidiosa. This study demonstrates that the use of toxins from TA systems is a promising strategy to be explored aiming bacterial control.


Subject(s)
Bacterial Toxins/genetics , Citrus/microbiology , Disease Resistance/genetics , Plant Diseases/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Toxins/pharmacology , Biotechnology , Citrus/genetics , Escherichia coli Proteins/genetics , Plant Diseases/microbiology , Plant Leaves/genetics , Plant Leaves/microbiology , Plants, Genetically Modified/genetics , Virulence/genetics , Xanthomonas/genetics , Xanthomonas/pathogenicity , Xylella/genetics , Xylella/pathogenicity
15.
Toxins (Basel) ; 14(1)2022 01 17.
Article in English | MEDLINE | ID: mdl-35051041

ABSTRACT

Botulinum neurotoxin (BoNT) is used for the treatment of a number of ailments. The activity of the toxin that is isolated from bacterial cultures is frequently tested in the mouse lethality assay. Apart from the ethical concerns inherent to this assay, species-specific differences in the affinity for different BoNT serotypes give rise to activity results that differ from the activity in humans. Thus, BoNT/B is more active in mice than in humans. The current study shows that the stimulus-dependent release of a luciferase from a differentiated human neuroblastoma-based reporter cell line (SIMA-hPOMC1-26-Gluc) was inhibited by clostridial and recombinant BoNT/A to the same extent, whereas both clostridial and recombinant BoNT/B inhibited the release to a lesser extent and only at much higher concentrations, reflecting the low activity of BoNT/B in humans. By contrast, the genetically modified BoNT/B-MY, which has increased affinity for human synaptotagmin, and the BoNT/B protein receptor inhibited luciferase release effectively and with an EC50 comparable to recombinant BoNT/A. This was due to an enhanced uptake into the reporter cells of BoNT/B-MY in comparison to the recombinant wild-type toxin. Thus, the SIMA-hPOMC1-26-Gluc cell assay is a versatile tool to determine the activity of different BoNT serotypes providing human-relevant dose-response data.


Subject(s)
Bacterial Toxins/toxicity , Botulinum Toxins, Type A/toxicity , Mutation , Bacterial Toxins/genetics , Bacterial Toxins/pharmacology , Biological Assay , Botulinum Toxins, Type A/genetics , Botulinum Toxins, Type A/pharmacology , Cell Line , Humans , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Recombinant Proteins/toxicity
16.
Viruses ; 14(1)2022 01 14.
Article in English | MEDLINE | ID: mdl-35062352

ABSTRACT

Respiratory disease in horses is caused by a multifactorial complex of infectious agents and environmental factors. An important pathogen in horses is equine herpesvirus type 1 (EHV-1). During co-evolution with this ancient alphaherpesvirus, the horse's respiratory tract has developed multiple antiviral barriers. However, these barriers can become compromised by environmental threats. Pollens and mycotoxins enhance mucosal susceptibility to EHV-1 by interrupting cell junctions, allowing the virus to reach its basolateral receptor. Whether bacterial toxins also play a role in this impairment has not been studied yet. Here, we evaluated the role of α-hemolysin (Hla) and adenylate cyclase (ACT), toxins derived from the facultative pathogenic bacterium Staphylococcus aureus (S. aureus) and the primary pathogen Bordetella bronchiseptica (B. bronchiseptica), respectively. Equine respiratory mucosal explants were cultured at an air-liquid interface and pretreated with these toxins, prior to EHV-1 inoculation. Morphological analysis of hematoxylin-eosin (HE)-stained sections of the explants revealed a decreased epithelial thickness upon treatment with both toxins. Additionally, the Hla toxin induced detachment of epithelial cells and a partial loss of cilia. These morphological changes were correlated with increased EHV-1 replication in the epithelium, as assessed by immunofluorescent stainings and confocal microscopy. In view of these results, we argue that the ACT and Hla toxins increase the susceptibility of the epithelium to EHV-1 by disrupting the epithelial barrier function. In conclusion, this study is the first to report that bacterial exotoxins increase the horse's sensitivity to EHV-1 infection. Therefore, we propose that horses suffering from infection by S. aureus or B. bronchiseptica may be more susceptible to EHV-1 infection.


Subject(s)
Bacterial Toxins/pharmacology , Bordetella bronchiseptica/metabolism , Herpesviridae Infections/drug therapy , Herpesviridae Infections/veterinary , Herpesvirus 1, Equid/drug effects , Horse Diseases/virology , Respiratory Tract Diseases/virology , Staphylococcus aureus/metabolism , Animals , Epithelial Cells/virology , Hemolysin Proteins , Horses , Respiratory Mucosa/drug effects , Respiratory Mucosa/virology , Virus Replication/drug effects
17.
Autophagy ; 18(9): 2050-2067, 2022 09.
Article in English | MEDLINE | ID: mdl-34989311

ABSTRACT

Clostridioides difficile infection (CDI) is a common cause of nosocomial diarrhea. TcdB is a major C. difficile exotoxin that activates macrophages to promote inflammation and epithelial damage. Lysosome impairment is a known trigger for inflammation. Herein, we hypothesize that TcdB could impair macrophage lysosomal function to mediate inflammation during CDI. Effects of TcdB on lysosomal function and the downstream pro-inflammatory SQSTM1/p62-NFKB (nuclear factor kappa B) signaling were assessed in cultured macrophages and in a murine CDI model. Protective effects of two lysosome activators (i.e., vitamin D3 and carbamazepine) were assessed. Results showed that TcdB inhibited CTNNB1/ß-catenin activity to downregulate MITF (melanocyte inducing transcription factor) and its direct target genes encoding components of lysosomal membrane vacuolar-type ATPase, thereby suppressing lysosome acidification in macrophages. The resulting lysosomal dysfunction then impaired autophagic flux and activated SQSTM1-NFKB signaling to drive the expression of IL1B/IL-1ß (interleukin 1 beta), IL8 and CXCL2 (chemokine (C-X-C motif) ligand 2). Restoring MITF function by enforced MITF expression or restoring lysosome acidification with 1α,25-dihydroxyvitamin D3 or carbamazepine suppressed pro-inflammatory cytokine expression in vitro. In mice, gavage with TcdB-hyperproducing C. difficile or injection of TcdB into ligated colon segments caused prominent MITF downregulation in macrophages. Vitamin D3 and carbamazepine lessened TcdB-induced lysosomal dysfunction, inflammation and histological damage. In conclusion, TcdB inhibits the CTNNB1-MITF axis to suppress lysosome acidification and activates the downstream SQSTM1-NFKB signaling in macrophages during CDI. Vitamin D3 and carbamazepine protect against CDI by restoring MITF expression and lysosomal function in mice.Abbreviations: ATP6V0B: ATPase H+ transporting V0 subunit b; ATP6V0C: ATPase H+ transporting V0 subunit c; ATP6V0E1: ATPase H+ transporting V0 subunit e1; ATP6V1H: ATPase H+ transporting V1 subunit H; CBZ: carbamazepine; CDI: C. difficile infection; CXCL: chemokine C-X-X motif ligand; IL: interleukin; LAMP1: lysosomal-associated membrane protein 1; LC3: microtubule-associated protein 1 light chain 3; LEF: lymphoid enhancer binding factor 1; MITF: melanocyte inducing transcription factor; NFKB: nuclear factor kappa B; PMA: phorbol 12-myristate 13-acetate; TcdA: Clostridial toxin A; TcdB: Clostridial toxin B; TFE3: transcription factor E3; TFEB: transcription factor EB.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Clostridium Infections , Vacuolar Proton-Translocating ATPases , Animals , Autophagy , Bacterial Proteins/metabolism , Bacterial Toxins/pharmacology , Carbamazepine/metabolism , Carbamazepine/pharmacology , Cholecalciferol/pharmacology , Clostridium Infections/metabolism , Hydrogen-Ion Concentration , Inflammation/metabolism , Lysosomes/metabolism , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Sequestosome-1 Protein/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
18.
Int J Mol Sci ; 24(1)2022 Dec 28.
Article in English | MEDLINE | ID: mdl-36613943

ABSTRACT

Cytolethal distending toxin (CDT) is produced by a range of Gram-negative pathogenic bacteria such as Campylobacter jejuni. CDT represents an important virulence factor that is a heterotrimeric complex composed of CdtA, CdtB, and CdtC. CdtA and CdtC constitute regulatory subunits whilst CdtB acts as the catalytic subunit exhibiting phosphatase and DNase activities, resulting in cell cycle arrest and cell death. Extracellular vesicle (EV) secretion is an evolutionarily conserved process that is present throughout all kingdoms. Mammalian EVs play important roles in regular cell-to-cell communications but can also spread pathogen- and host-derived molecules during infections to alter immune responses. Here, we demonstrate that CDT targets the endo-lysosomal compartment, partially evading lysosomal degradation and exploiting unconventional secretion (EV release), which is largely involved in bacterial infections. CDT-like effects are transferred by Caco-2 cells to uninfected heterologous U937 and homologous Caco-2 cells. The journey of EVs derived from CDT-treated Caco-2 cells is associated with both intestinal and myeloid tumour cells. EV release represents the primary route of CDT dissemination, revealing an active toxin as part of the cargo. We demonstrated that bacterial toxins could represent suitable tools in cancer therapy, highlighting both the benefits and limitations. The global cell response involves a moderate induction of apoptosis and autophagic features may play a protective role against toxin-induced cell death. EVs from CDT-treated Caco-2 cells represent reliable CDT carriers, potentially suitable in colorectal cancer treatments. Our data present a potential bacterial-related biotherapeutic supporting a multidrug anticancer protocol.


Subject(s)
Bacterial Toxins , Campylobacter jejuni , Humans , Bacterial Toxins/pharmacology , Bacterial Toxins/metabolism , Caco-2 Cells , Campylobacter jejuni/metabolism , Cell Proliferation , Gram-Negative Bacteria/metabolism , U937 Cells
19.
Semin Cancer Biol ; 86(Pt 3): 753-768, 2022 11.
Article in English | MEDLINE | ID: mdl-34271147

ABSTRACT

It is a major concern to treat cancer successfully, due to the distinctive pathophysiology of cancer cells and the gradual manifestation of resistance. Specific action, adverse effects and development of resistance has prompted the urgent requirement of exploring alternative anti-tumour treatment therapies. The naturally derived microbial toxins as a therapy against cancer cells are a promisingly new dimension. Various important microbial toxins such as Diphtheria toxin, Vibrio cholera toxin, Aflatoxin, Patulin, Cryptophycin-55, Chlorella are derived from several bacterial, fungal and algal species. These agents act on different biotargets such as inhibition of protein synthesis, reduction in cell growth, regulation of cell cycle and many cellular processes. Bacterial toxins produce actions primarily by targeting protein moieties and some immunomodulation and few acts through DNA. Fungal toxins appear to have more DNA damaging activity and affect the cell cycle. Algal toxins produce alteration in mitochondrial phosphorylation. In conclusion, microbial toxins and their metabolites appear to have a great potential to provide a promising option for the treatment and management to combat cancer.


Subject(s)
Bacterial Toxins , Chlorella , Neoplasms , Humans , Bacterial Toxins/pharmacology , Cholera Toxin/pharmacology , Neoplasms/drug therapy
20.
Toxins (Basel) ; 13(12)2021 12 10.
Article in English | MEDLINE | ID: mdl-34941719

ABSTRACT

This study was conducted to compare the potential ameliorative effects between probiotic Bacillus subtilis and biodegradable Bacillus subtilis on zearalenone (ZEN) toxicosis in gilts. Thirty-six Landrace×Yorkshire gilts (average BW = 64 kg) were randomly divided into four groups: (1) Normal control diet group (NC) fed the basal diet containing few ZEN (17.5 µg/kg); (2) ZEN contaminated group (ZC) fed the contaminated diet containing an exceeded limit dose of ZEN (about 300 µg/kg); (3) Probiotic agent group (PB) fed the ZC diet with added 5 × 109 CFU/kg of probiotic Bacillus subtilis ANSB010; (4) Biodegradable agent group (DA) fed the ZC diet with added 5 × 109 CFU/kg of biodegradable Bacillus subtilis ANSB01G. Results showed that Bacillus subtilis ANSB010 and ANSB01G isolated from broiler intestinal chyme had similar inhibitory activities against common pathogenic bacteria. In addition, the feed conversion ratio and the vulva size in DA group were significantly lower than ZC group (p < 0.05). The levels of IgG, IgM, IL-2 and TNFα in the ZC group were significantly higher than PB and DA groups (p < 0.05). The levels of estradiol and prolactin in the ZC group was significantly higher than those of the NC and DA groups (p < 0.05). Additionally, the residual ZEN in the feces of the ZC and PB groups were higher than those of the NC and DA groups (p < 0.05). In summary, the ZEN-contaminated diet had a damaging impact on growth performance, plasma immune function and hormone secretion of gilts. Although probiotic and biodegradable Bacillus subtilis have similar antimicrobial capacities, only biodegradable Bacillus subtilis could eliminate these negative effects through its biodegradable property to ZEN.


Subject(s)
Animal Feed/toxicity , Bacillus subtilis , Bacterial Toxins/pharmacology , Bacterial Toxins/therapeutic use , Probiotics/therapeutic use , Protective Agents/therapeutic use , Zearalenone/toxicity , Animals , Female , Probiotics/pharmacology , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...