Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 210
Filtrar
1.
Toxins (Basel) ; 14(12)2022 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-36548771

RESUMO

Candida albicans produces an important virulence factor, the hypha-associated Ece1-derived secreted peptide toxin candidalysin, which is crucial for the establishment of mucosal and systemic infections. C. albicans has also long been known to be hemolytic, yet the hemolytic factor has not been clearly identified. Here, we show that candidalysin is the hemolytic factor of C. albicans. Its hemolytic activity is modulated by fragments of another Ece1 peptide, P7. Hemolysis by candidalysin can be neutralized by the purinergic receptor antagonist pyridoxal-phosphate-6-azophenyl-2',4'-disulfonic acid (PPADS). PPADS also affects candidalysin's ability to intercalate into synthetic membranes. We also describe the neutralization potential of two anti-candidalysin nanobodies, which are promising candidates for future anti-Candida therapy. This work provides evidence that the historically proposed hemolytic factor of C. albicans is in fact candidalysin and sheds more light on the complex roles of this toxin in C. albicans biology and pathogenicity.


Assuntos
Candida albicans , Hemólise , Proteínas Fúngicas , Candida , Mucosa , Fatores de Virulência/toxicidade
2.
Oxid Med Cell Longev ; 2022: 3060579, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35340215

RESUMO

Pyocyanin (PCN) is a redox-active secondary metabolite produced by Pseudomonas aeruginosa as its primary virulence factor. Several studies have reported the cytotoxic potential of PCN and its role during infection establishment and progression. Considering its ability to diffuse through biological membranes, it is hypothesized that PCN can gain entry into the brain and induce oxidative stress across the blood-brain barrier (BBB), ultimately contributing towards reactive oxygen species (ROS) mediated neurodegeneration. Potential roles of PCN in the central nervous system (CNS) have never been evaluated, hence the study aimed to evaluate PCN's probable penetration into CNS through blood-brain barrier (BBB) using both in silico and in vivo (Balb/c mice) approaches and the impact of ROS generation via commonly used tests: Morris water maze test, novel object recognition, elevated plus maze test, and tail suspension test. Furthermore, evidence for ROS generation in the brain was assessed using glutathione S-transferase assay. PCN demonstrated BBB permeability albeit in minute quantities. A significant hike was observed in ROS generation (P < 0.0001) along with changes in behavior indicating PCN permeability across BBB and potentially affecting cognitive functions. This is the first study exploring the potential role of PCN in influencing the cognitive functions of test animals.


Assuntos
Disfunção Cognitiva , Piocianina , Animais , Barreira Hematoencefálica/metabolismo , Camundongos , Permeabilidade , Pseudomonas/metabolismo , Pseudomonas aeruginosa , Piocianina/metabolismo , Piocianina/farmacologia , Fatores de Virulência/metabolismo , Fatores de Virulência/toxicidade
3.
Front Immunol ; 13: 815775, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154132

RESUMO

Many species of bacteria produce toxins such as cholesterol-dependent cytolysins that form pores in cell membranes. Membrane pores facilitate infection by releasing nutrients, delivering virulence factors, and causing lytic cell damage - cytolysis. Oxysterols are oxidized forms of cholesterol that regulate cellular cholesterol and alter immune responses to bacteria. Whether oxysterols also influence the protection of cells against pore-forming toxins is unresolved. Here we tested the hypothesis that oxysterols stimulate the intrinsic protection of epithelial cells against damage caused by cholesterol-dependent cytolysins. We treated epithelial cells with oxysterols and then challenged them with the cholesterol-dependent cytolysin, pyolysin. Treating HeLa cells with 27-hydroxycholesterol, 25-hydroxycholesterol, 7α-hydroxycholesterol, or 7ß-hydroxycholesterol reduced pyolysin-induced leakage of lactate dehydrogenase and reduced pyolysin-induced cytolysis. Specifically, treatment with 10 ng/ml 27-hydroxycholesterol for 24 h reduced pyolysin-induced lactate dehydrogenase leakage by 88%, and reduced cytolysis from 74% to 1%. Treating HeLa cells with 27-hydroxycholesterol also reduced pyolysin-induced leakage of potassium ions, prevented mitogen-activated protein kinase cell stress responses, and limited alterations in the cytoskeleton. Furthermore, 27-hydroxycholesterol reduced pyolysin-induced damage in lung and liver epithelial cells, and protected against the cytolysins streptolysin O and Staphylococcus aureus α-hemolysin. Although oxysterols regulate cellular cholesterol by activating liver X receptors, cytoprotection did not depend on liver X receptors or changes in total cellular cholesterol. However, oxysterol cytoprotection was partially dependent on acyl-CoA:cholesterol acyltransferase (ACAT) reducing accessible cholesterol in cell membranes. Collectively, these findings imply that oxysterols stimulate the intrinsic protection of epithelial cells against pore-forming toxins and may help protect tissues against pathogenic bacteria.


Assuntos
Bactérias/química , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Proteínas Hemolisinas/toxicidade , Oxisteróis/farmacologia , Fatores de Virulência/toxicidade , Proteínas de Bactérias/química , Toxinas Bacterianas/química , Células Epiteliais/metabolismo , Células HeLa , Proteínas Hemolisinas/química , Humanos , Fatores de Virulência/química
4.
Nat Commun ; 12(1): 6193, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34702812

RESUMO

Staphylococcus aureus bi-component pore-forming leukocidins are secreted toxins that directly target and lyse immune cells. Intriguingly, one of the leukocidins, Leukocidin AB (LukAB), is found associated with the bacterial cell envelope in addition to secreted into the extracellular milieu. Here, we report that retention of LukAB on the bacterial cells provides S. aureus with a pre-synthesized active toxin that kills immune cells. On the bacteria, LukAB is distributed as discrete foci in two distinct compartments: membrane-proximal and surface-exposed. Through genetic screens, we show that a membrane lipid, lysyl-phosphatidylglycerol (LPG), and lipoteichoic acid (LTA) contribute to LukAB deposition and release. Furthermore, by studying non-covalently surface-bound proteins we discovered that the sorting of additional exoproteins, such as IsaB, Hel, ScaH, and Geh, are also controlled by LPG and LTA. Collectively, our study reveals a multistep secretion system that controls exoprotein storage and protein translocation across the S. aureus cell wall.


Assuntos
Membrana Celular/metabolismo , Parede Celular/metabolismo , Staphylococcus aureus/metabolismo , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/toxicidade , Citotoxinas/metabolismo , Citotoxinas/toxicidade , Humanos , Leucocidinas/metabolismo , Leucocidinas/toxicidade , Lipopolissacarídeos/genética , Lipopolissacarídeos/metabolismo , Lisina/genética , Lisina/metabolismo , Camundongos , Fagócitos/efeitos dos fármacos , Fosfatidilgliceróis/genética , Fosfatidilgliceróis/metabolismo , Transporte Proteico , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Ácidos Teicoicos/genética , Ácidos Teicoicos/metabolismo , Fatores de Virulência/toxicidade
5.
FASEB J ; 35(10): e21889, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34569656

RESUMO

Many species of pathogenic bacteria secrete toxins that form pores in mammalian cell membranes. These membrane pores enable the delivery of virulence factors into cells, result in the leakage of molecules that bacteria can use as nutrients, and facilitate pathogen invasion. Inflammatory responses to bacteria are regulated by the side-chain-hydroxycholesterols 27-hydroxycholesterol and 25-hydroxycholesterol, but their effect on the intrinsic protection of cells against pore-forming toxins is unclear. Here, we tested the hypothesis that 27-hydroxycholesterol and 25-hydroxycholesterol help protect cells against pore-forming toxins. We treated bovine endometrial epithelial and stromal cells with 27-hydroxycholesterol or 25-hydroxycholesterol, and then challenged the cells with pyolysin, which is a cholesterol-dependent cytolysin from Trueperella pyogenes that targets these endometrial cells. We found that treatment with 27-hydroxycholesterol or 25-hydroxycholesterol protected both epithelial and stomal cells against pore formation and the damage caused by pyolysin. The oxysterols limited pyolysin-induced leakage of potassium and lactate dehydrogenase from cells, and reduced cytoskeletal changes and cytolysis. This oxysterol cytoprotection against pyolysin was partially dependent on reducing cytolysin-accessible cholesterol in the cell membrane and on activating liver X receptors. Treatment with 27-hydroxycholesterol also protected the endometrial cells against Staphylococcus aureus α-hemolysin. Using mass spectrometry, we found 27-hydroxycholesterol and 25-hydroxycholesterol in uterine and follicular fluid. Furthermore, epithelial cells released additional 25-hydroxycholesterol in response to pyolysin. In conclusion, both 27-hydroxycholesterol and 25-hydroxycholesterol increased the intrinsic protection of bovine endometrial cells against pore-forming toxins. Our findings imply that side-chain-hydroxycholesterols may help defend the endometrium against pathogenic bacteria.


Assuntos
Bactérias/química , Proteínas de Bactérias/toxicidade , Endométrio/metabolismo , Proteínas Hemolisinas/toxicidade , Hidroxicolesteróis/farmacologia , Fatores de Virulência/toxicidade , Animais , Proteínas de Bactérias/química , Bovinos , Feminino , Proteínas Hemolisinas/química , Células Estromais/metabolismo , Fatores de Virulência/química
6.
Toxins (Basel) ; 13(1)2020 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-33375750

RESUMO

Mono-ADP-ribosyltransferase toxins are often key virulence factors produced by pathogenic bacteria as tools to compromise the target host cell. These toxins are enzymes that use host cellular NAD+ as the substrate to modify a critical macromolecule target in the host cell machinery. This post-translational modification of the target macromolecule (usually protein or DNA) acts like a switch to turn the target activity on or off resulting in impairment of a critical process or pathway in the host. One approach to stymie bacterial pathogens is to curtail the toxic action of these factors by designing small molecules that bind tightly to the enzyme active site and prevent catalytic function. The inactivation of these toxins/enzymes is targeted for the site of action within the host cell and small molecule therapeutics can function as anti-virulence agents by disarming the pathogen. This represents an alternative strategy to antibiotic therapy with the potential as a paradigm shift that may circumvent multi-drug resistance in the offending microbe. In this review, work that has been accomplished during the past two decades on this approach to develop anti-virulence compounds against mono-ADP-ribosyltransferase toxins will be discussed.


Assuntos
ADP Ribose Transferases/toxicidade , Antídotos , Toxinas Bacterianas , Fatores de Virulência/antagonistas & inibidores , Animais , Humanos , Fatores de Virulência/metabolismo , Fatores de Virulência/toxicidade
7.
Pak J Pharm Sci ; 33(3): 915-922, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-33191213

RESUMO

Pseudomonas aeruginosa (PA) is one of the most clinically significant nosocomial infectious agents. Clinical significance of this bacterium is intensified due to the phenomenon of its natural tendency for acquiring drug resistance mechanisms. PA produces pyocyanin (PCN), an important redox-active virulence factor. PCN has been detected in higher quantities in sputum samples of PA infected Cystic Fibrosis patients. PCN producing PA strains were isolated and characterized. Genomic 16s rRNA gene segment was amplified and sequenced (GenBank accession # jx280426). PCN was extracted and purified. In silico analysis yielded permeability and cytotoxic potential of PCN in modeled cell lines. PCN has high intestinal absorption, plasma protein binding potential, and permeability across biological membranes. Oral toxicity study in in silico rodent model classified PCN in class IV 'harmful if swallowed' (ld50 0.3-2g/kg). Cytotoxicity was assessed by oxidative stress levels in different organs in balb/c mice induced by intra peritoneal PCN injection. Significant alterations in oxidative stress levels in different organs of balb/c mice were observed. Increased levels of oxidative stress were observed in lungs, and heart, lower in liver and spleen while muscle tissues showed no significant difference in comparison to control.


Assuntos
Pseudomonas aeruginosa/metabolismo , Piocianina/toxicidade , Fatores de Virulência/toxicidade , Animais , Células CACO-2 , Cães , Humanos , Absorção Intestinal , Células Madin Darby de Rim Canino , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos , Permeabilidade , Ligação Proteica , Pseudomonas aeruginosa/patogenicidade , Piocianina/metabolismo , Medição de Risco , Testes de Toxicidade , Fatores de Virulência/metabolismo
8.
J Fish Dis ; 43(10): 1229-1236, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32974952

RESUMO

Streptococcus agalactiae secrete virulence factors believed to be able of killing host tissues, especially under elevated water temperature. A direct effect of S. agalactiae secretory products on tilapia cells was tested on the tilapia kidney (TK-1) cell culture. The bacteria were cultured under four different temperature levels: 22, 29, 32 and 37°C; the cell-free portion was processed through SDS-PAGE; and distinct bands were identified by LC-MS/MS. At least, three virulence factors were identified, Bsp, PcsB and CAMP factor, with increasing levels as the cultured temperature rose. Expressions of bsp, pcsB and cfb were also up-regulated with the rising of the temperature in S. agalactiae culture. The supernatant from the bacteria cultured under specified temperatures was added into TK-1 cell-cultured wells. Morphological damage and mortality of the cultured cells, as determined by MTT method, were increased progressively from the supernatant treatment according to the rise of temperature in S. agalactiae culture. This study suggests that the production of the three virulence factors of S. agalactiae reported herein is temperature-dependent, and it is likely that CAMP factor directly kills the TK-1 cells since the other two types of protein are involved in S. agalactiae cell division and the bacterial adherence to host tissues.


Assuntos
Proteínas de Bactérias/toxicidade , Streptococcus agalactiae/patogenicidade , Tilápia/microbiologia , Fatores de Virulência/toxicidade , Animais , Aderência Bacteriana , Linhagem Celular , Doenças dos Peixes/microbiologia , Temperatura
9.
Infect Immun ; 88(9)2020 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-32571989

RESUMO

Staphylococcus aureus is a major human pathogen, and the emergence of antibiotic-resistant strains is making all types of S. aureus infections more challenging to treat. With a pressing need to develop alternative control strategies to use alongside or in place of conventional antibiotics, one approach is the targeting of established virulence factors. However, attempts at this have had little success to date, suggesting that we need to better understand how this pathogen causes disease if effective targets are to be identified. To address this, using a functional genomics approach, we have identified a small membrane-bound protein that we have called MspA. Inactivation of this protein results in the loss of the ability of S. aureus to secrete cytolytic toxins, protect itself from several aspects of the human innate immune system, and control its iron homeostasis. These changes appear to be mediated through a change in the stability of the bacterial membrane as a consequence of iron toxicity. These pleiotropic effects on the ability of the pathogen to interact with its host result in significant impairment in the ability of S. aureus to cause infection in both a subcutaneous and sepsis model of infection. Given the scale of the effect the inactivation of MspA causes, it represents a unique and promising target for the development of a novel therapeutic approach.


Assuntos
Bacteriemia/microbiologia , Evasão da Resposta Imune , Infecções Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade , Fatores de Virulência/genética , Células A549 , Animais , Bacteriemia/imunologia , Bacteriemia/patologia , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Eritrócitos/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Heme/imunologia , Heme/metabolismo , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/imunologia , Homeostase/imunologia , Humanos , Ferro/imunologia , Ferro/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutação , Fagocitose , Proteômica/métodos , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/patologia , Infecções Cutâneas Estafilocócicas/imunologia , Infecções Cutâneas Estafilocócicas/patologia , Toxoide Estafilocócico/genética , Toxoide Estafilocócico/imunologia , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia , Células THP-1 , Virulência , Fatores de Virulência/imunologia , Fatores de Virulência/toxicidade , alfa-Defensinas/genética , alfa-Defensinas/imunologia
10.
Toxins (Basel) ; 12(2)2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-32033406

RESUMO

Intestinal microbiota exerts protective effects against the infection of various bacterial pathogens, including Listeria monocytogenes, a major foodborne pathogen whose infection can lead to a disease (listeriosis) with a high fatality rate. As a strategy to mitigate the action of the intestinal microbiota, pathogens often produce antimicrobial proteinaceous compounds such as bacteriocins. In this review, we summarize the information currently available for the well-characterized L. monocytogenes bacteriocin listeriolysin S, with the emphasis on its intriguing mode of action as a virulence factor, which promotes the infection of L. monocytogenes by changing the composition of the intestinal microbiota. We then discuss another intriguing L. monocytogenes bacteriocin Lmo2776 that specifically inhibits the inflammogenic species, Prevotella copri, in the intestinal microbiota, reducing superfluous inflammation while weakening virulence. In addition, we describe relatively less studied phage tail-like Listeria bacteriocins (monocins) and elaborate on the possibility that these monocins could be involved in enhancing pathogenicity. In spite of the burgeoning interest in the roles played by the intestinal microbiota against the L. monocytogenes infection, our understanding on the virulence factors affecting the intestinal microbiota is still lacking, calling for further studies on bacteriocins that could function as novel virulence factors.


Assuntos
Bacteriocinas , Microbioma Gastrointestinal/efeitos dos fármacos , Listeria monocytogenes/patogenicidade , Listeriose/microbiologia , Fatores de Virulência , Animais , Bacteriocinas/genética , Bacteriocinas/toxicidade , Interações Hospedeiro-Patógeno , Humanos , Virulência , Fatores de Virulência/genética , Fatores de Virulência/toxicidade
11.
Nat Commun ; 11(1): 760, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-32029733

RESUMO

Inflammasomes are important for host defence against pathogens and homeostasis with commensal microbes. Here, we show non-haemolytic enterotoxin (NHE) from the neglected human foodborne pathogen Bacillus cereus is an activator of the NLRP3 inflammasome and pyroptosis. NHE is a non-redundant toxin to haemolysin BL (HBL) despite having a similar mechanism of action. Via a putative transmembrane region, subunit C of NHE initiates binding to the plasma membrane, leading to the recruitment of subunit B and subunit A, thus forming a tripartite lytic pore that is permissive to efflux of potassium. NHE mediates killing of cells from multiple lineages and hosts, highlighting a versatile functional repertoire in different host species. These data indicate that NHE and HBL operate synergistically to induce inflammation and show that multiple virulence factors from the same pathogen with conserved function and mechanism of action can be exploited for sensing by a single inflammasome.


Assuntos
Bacillus cereus/patogenicidade , Enterotoxinas/toxicidade , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Proteínas de Bactérias/toxicidade , Linhagem Celular , Enterotoxinas/química , Feminino , Proteínas Hemolisinas/toxicidade , Interações entre Hospedeiro e Microrganismos , Especificidade de Hospedeiro , Humanos , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Piroptose/efeitos dos fármacos , Fatores de Virulência/toxicidade
12.
Biotechnol Lett ; 42(1): 125-133, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31720976

RESUMO

OBJECTIVE: We aimed to investigate the expression of a novel small cysteine-rich (SCR) effector protein SCR96 from the phytopathogenic oomycete Phytophthora cactorum in mammalian cells, its bioactivity and to exploit its polyclonal antibody. RESULTS: The gene encoding the SCR effector protein SCR96 was codon-optimized, custom-synthesized, cloned into pcDNA3.1(-) and overexpressed in human embryonic kidney (HEK) 293-6E cells. The recombinant protein SCR96 was prone to aggregation and purified with its monomer to homogeneity with a predicted molecular weight of 8.9 kDa. SCR96 exhibited strong phytotoxic activity on tomato seedlings at 24 h post treatment with 4.2 µg of the purified protein. An anti-SCR96 polyclonal antibody was prepared by immunization of New Zealand white rabbits. The good-titer antibody had a detection sensitivity at 6.25-ng level and could specifically detect the SCR96 protein expressed either in yeast, or in tomato leaves. CONCLUSIONS: Transient production of the SCR effector protein SCR96 in mammalian cells is reliable, providing sufficient recombinant protein that can be utilized for analysis of its phytotoxic activity and preparation of its polyclonal antibody.


Assuntos
Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/toxicidade , Phytophthora/metabolismo , Solanum lycopersicum/efeitos dos fármacos , Fatores de Virulência/biossíntese , Fatores de Virulência/toxicidade , Animais , Anticorpos/imunologia , Proteínas Fúngicas/genética , Proteínas Fúngicas/imunologia , Células HEK293 , Humanos , Phytophthora/genética , Coelhos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/toxicidade , Plântula/efeitos dos fármacos , Fatores de Virulência/genética , Fatores de Virulência/imunologia
13.
J Vis Exp ; (152)2019 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-31710035

RESUMO

Intracellular bacteria secrete virulence factors called effector proteins into the host cytosol that act to subvert host proteins and/or their associated biological pathways to the benefit of the bacterium. Identification of putative bacterial effector proteins has become more manageable due to advances in bacterial genome sequencing and the advent of algorithms that allow in silico identification of genes encoding secretion candidates and/or eukaryotic-like domains. However, identification of these important virulence factors is only an initial step. Naturally, the goal is to determine the molecular function of effector proteins and elucidate how they interact with the host. In recent years, techniques like the yeast two-hybrid screen and large-scale immunoprecipitations coupled with mass spectrometry have aided in the identification of protein-protein interactions. Although identification of a host binding partner is the crucial first step toward elucidating the molecular function of a bacterial effector protein, sometimes the host protein is found to have multiple biological functions (e.g., actin, clathrin, tubulin), or the bacterial protein may not physically bind host proteins, depriving the researcher of crucial information about the precise host pathway being manipulated. A modified yeast toxicity screen coupled with a suppressor screen has been adapted to identify host pathways impacted by bacterial effector proteins. The toxicity screen relies on a toxic effect in yeast caused by the effector protein interfering with the host biological pathways, which often manifests as a growth defect. Expression of a yeast genomic library is used to identify host factors that suppress the toxicity of the bacterial effector protein and thus identify proteins in the pathway that the effector protein targets. This protocol contains detailed instructions for both the toxicity and suppressor screens. These techniques can be performed in any lab capable of molecular cloning and cultivation of yeast and Escherichia coli.


Assuntos
Proteínas de Bactérias/fisiologia , Proteínas de Bactérias/toxicidade , Saccharomyces cerevisiae/efeitos dos fármacos , Bactérias/genética , Técnicas Genéticas , Fatores de Virulência/metabolismo , Fatores de Virulência/toxicidade
14.
PLoS Pathog ; 15(9): e1008029, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31545853

RESUMO

Although Escherichia coli Nissle 1917 (EcN) has been used therapeutically for over a century, the determinants of its probiotic properties remain elusive. EcN produces two siderophore-microcins (Mcc) responsible for an antagonistic activity against other Enterobacteriaceae. EcN also synthesizes the genotoxin colibactin encoded by the pks island. Colibactin is a virulence factor and a putative pro-carcinogenic compound. Therefore, we aimed to decouple the antagonistic activity of EcN from its genotoxic activity. We demonstrated that the pks-encoded ClbP, the peptidase that activates colibactin, is required for the antagonistic activity of EcN. The analysis of a series of ClbP mutants revealed that this activity is linked to the transmembrane helices of ClbP and not the periplasmic peptidase domain, indicating the transmembrane domain is involved in some aspect of Mcc biosynthesis or secretion. A single amino acid substitution in ClbP inactivates the genotoxic activity but maintains the antagonistic activity. In an in vivo salmonellosis model, this point mutant reduced the clinical signs and the fecal shedding of Salmonella similarly to the wild type strain, whereas the clbP deletion mutant could neither protect nor outcompete the pathogen. The ClbP-dependent antibacterial effect was also observed in vitro with other E. coli strains that carry both a truncated form of the Mcc gene cluster and the pks island. In such strains, siderophore-Mcc synthesis also required the glucosyltransferase IroB involved in salmochelin production. This interplay between colibactin, salmochelin, and siderophore-Mcc biosynthetic pathways suggests that these genomic islands were co-selected and played a role in the evolution of E. coli from phylogroup B2. This co-evolution observed in EcN illustrates the fine margin between pathogenicity and probiotic activity, and the need to address both the effectiveness and safety of probiotics. Decoupling the antagonistic from the genotoxic activity by specifically inactivating ClbP peptidase domain opens the way to the safe use of EcN.


Assuntos
Escherichia coli/fisiologia , Mutagênicos/toxicidade , Probióticos/uso terapêutico , Animais , Antibiose/genética , Antibiose/fisiologia , Bacteriocinas/genética , Bacteriocinas/metabolismo , Bacteriocinas/toxicidade , Vias Biossintéticas/genética , Enterobactina/análogos & derivados , Enterobactina/genética , Enterobactina/fisiologia , Enterobactina/toxicidade , Escherichia coli/genética , Escherichia coli/patogenicidade , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiologia , Feminino , Genes Bacterianos , Ilhas Genômicas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Família Multigênica , Mutação , Peptídeo Hidrolases/química , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/fisiologia , Peptídeos/genética , Peptídeos/fisiologia , Peptídeos/toxicidade , Policetídeos/toxicidade , Probióticos/toxicidade , Domínios Proteicos , Salmonelose Animal/microbiologia , Salmonelose Animal/terapia , Salmonella typhimurium , Sideróforos/genética , Sideróforos/fisiologia , Sideróforos/toxicidade , Fatores de Virulência/genética , Fatores de Virulência/fisiologia , Fatores de Virulência/toxicidade
15.
PLoS One ; 14(6): e0214035, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31163020

RESUMO

The genetic determinants of bacterial pathogenicity are highly variable between species and strains. However, a factor that is commonly associated with virulent Gram-negative bacteria, including many Aeromonas spp., is the type 3 secretion system (T3SS), which is used to inject effector proteins into target eukaryotic cells. In this study, we developed a bioinformatics pipeline to identify T3SS effector proteins, applied this approach to the genomes of 105 Aeromonas strains isolated from environmental, mutualistic, or pathogenic contexts and evaluated the cytotoxicity of the identified effectors through their heterologous expression in yeast. The developed pipeline uses a two-step approach, where candidate Aeromonas gene families are initially selected using Hidden Markov Model (HMM) profile searches against the Virulence Factors DataBase (VFDB), followed by strict comparisons against positive and negative control datasets, greatly reducing the number of false positives. This approach identified 21 Aeromonas T3SS likely effector families, of which 8 represent known or characterized effectors, while the remaining 13 have not previously been described in Aeromonas. We experimentally validated our in silico findings by assessing the cytotoxicity of representative effectors in Saccharomyces cerevisiae BY4741, with 15 out of 21 assayed proteins eliciting a cytotoxic effect in yeast. The results of this study demonstrate the utility of our approach, combining a novel in silico search method with in vivo experimental validation, and will be useful in future research aimed at identifying and authenticating bacterial effector proteins from other genera.


Assuntos
Aeromonas/patogenicidade , Sistemas de Secreção Tipo III/metabolismo , Fatores de Virulência/toxicidade , Aeromonas/genética , Aeromonas/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/toxicidade , Biologia Computacional/métodos , Simulação por Computador , Genoma Bacteriano , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/efeitos dos fármacos , Fatores de Virulência/genética
16.
Toxins (Basel) ; 11(6)2019 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-31212980

RESUMO

Clostridium difficile induces antibiotic-associated diarrhea due to the release of toxin A (TcdA) and toxin B (TcdB), the latter being its main virulence factor. The epidemic strain NAP1/027 has an increased virulence attributed to different factors. We compared cellular intoxication by TcdBNAP1 with that by the reference strain VPI 10463 (TcdBVPI). In a mouse ligated intestinal loop model, TcdBNAP1 induced higher neutrophil recruitment, cytokine release, and epithelial damage than TcdBVPI. Both toxins modified the same panel of small GTPases and exhibited similar in vitro autoprocessing kinetics. On the basis of sequence variations in the frizzled-binding domain (FBD), we reasoned that TcdBVPI and TcdBNAP1 might have different receptor specificities. To test this possibility, we used a TcdB from a NAP1 variant strain (TcdBNAP1v) unable to glucosylate RhoA but with the same receptor-binding domains as TcdBNAP1. Cells were preincubated with TcdBNAP1v to block cellular receptors, prior to intoxication with either TcdBVPI or TcdBNAP1. Preincubation with TcdBNAP1v blocked RhoA glucosylation by TcdBNAP1 but not by TcdBVPI, indicating that the toxins use different host factors for cell entry. This crucial difference might explain the increased biological activity of TcdBNAP1 in the intestine, representing a contributing factor for the increased virulence of the NAP1/027 strain.


Assuntos
Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Enterotoxinas/toxicidade , Interações entre Hospedeiro e Microrganismos , Fatores de Virulência/toxicidade , Células 3T3 , Animais , Fenômenos Fisiológicos Bacterianos , Sobrevivência Celular/efeitos dos fármacos , Clostridioides difficile/fisiologia , Infecções por Clostridium/imunologia , Citocinas/imunologia , Células HeLa , Humanos , Intestinos/efeitos dos fármacos , Intestinos/imunologia , Intestinos/microbiologia , Masculino , Camundongos , Neutrófilos/imunologia , Receptores de Superfície Celular/metabolismo
17.
Toxins (Basel) ; 11(3)2019 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-30832302

RESUMO

Staphylococcus aureus is an important bacterial pathogen causing bovine mastitis, but little is known about the virulence factor and the inflammatory responses in the mammary infection. Staphylococcal enterotoxin C (SEC) is the most frequent toxin produced by S. aureus, isolated from bovine mastitis. To investigate the pathogenic activity of SEC in the inflammation of the mammary gland and the immune responses in an animal model, mouse mammary glands were injected with SEC, and the clinical signs, inflammatory cell infiltration, and proinflammatory cytokine production in the mammary glands were assessed. SEC induced significant inflammatory reactions in the mammary gland, in a dose-dependent manner. SEC-injected mammary glands showed a severe inflammation with inflammatory cell infiltration and tissue damage. In addition, interleukin (IL)-1ß and IL-6 production in the SEC-injected mammary glands were significantly higher than those in the PBS control glands. Furthermore, the SEC-induced inflammation and tissue damage in the mammary gland were specifically inhibited by anti-SEC antibody. These results indicated, for the first time, that SEC can directly cause inflammation, proinflammatory cytokine production, and tissue damage in mammary glands, suggesting that SEC might play an important role in the development of mastitis associated with S. aureus infection. This finding offers an opportunity to develop novel treatment strategies for reduction of mammary tissue damage in mastitis.


Assuntos
Toxinas Bacterianas/toxicidade , Enterotoxinas/toxicidade , Mastite , Fatores de Virulência/toxicidade , Animais , Modelos Animais de Doenças , Feminino , Interleucina-1beta/imunologia , Interleucina-6/imunologia , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/imunologia , Glândulas Mamárias Animais/patologia , Mastite/imunologia , Mastite/patologia , Camundongos Endogâmicos BALB C
18.
Anaerobe ; 56: 51-56, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30771459

RESUMO

Fusobacterium necrophorum is a Gram negative, rod-shaped and aero tolerant anaerobe. In animals, it is an opportunistic pathogen frequently associated with necrotic infections, generally called necrobacillosis, such as calf diphtheria, foot rot and liver abscesses in cattle. Two subspecies exist: subsp. necrophorum and subsp. funduliforme. Among several virulence factors, leukotoxin (Lkt) is considered to be a major factor and a protective antigen. The objective of the study was to utilize BL3 cells and measure the release of lactic dehydrogenase to quantify Lkt activity of F. necrophorum. The assay was used to examine the effects of storage and handling conditions, growth media, polymyxin B addition on the cytotoxicity and evaluate Lkt activities of F. necrophorum strains isolated from bovine liver abscesses and foot rot. The Lkt activity peaked at 9 h of incubation. There was a significant decrease in the cytotoxicity measured in the samples after each freeze and thaw cycle. No difference was observed in the cytotoxicity for the samples handled aerobically versus anaerobically. Lkt activities of strains grown in anaerobic Brain-Heart Infusion broth were higher compared to Vegitone broth. A small reduction in the cytotoxicity activity was observed after the addition of polymyxin. The Lkt activity was consistently higher in strains of subsp. necrophorum than subsp. funduliforme of liver abscess origin. Among the strains isolated from cattle foot rot, Lkt activities of subsp. necrophorum strains appear to be much more variable. Use of BL3 cells in combination of lactic acid dehydrogenase assay appears to be a simple and valid assay to measure Lkt activity of F. necrophorum.


Assuntos
Doenças dos Bovinos/microbiologia , Exotoxinas/toxicidade , Infecções por Fusobacterium/veterinária , Fusobacterium necrophorum/isolamento & purificação , Fusobacterium necrophorum/patogenicidade , Fatores de Virulência/toxicidade , Animais , Bovinos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Pododermatite Necrótica dos Ovinos/microbiologia , Infecções por Fusobacterium/microbiologia , L-Lactato Desidrogenase/análise , Abscesso Hepático/microbiologia , Abscesso Hepático/veterinária
19.
J Cancer Res Clin Oncol ; 145(1): 49-63, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30542789

RESUMO

PURPOSE: Human microbiota comprises of a variety of organisms ranging from bacterial species to viruses, fungi, and protozoa which are present on the epidermal and mucosal barriers of the body. It plays a key role in health and survival of the host by regulation of the systemic functions. Its apparent functions in modulation of the host immune system, inducing carcinogenesis and regulation of the response to the cancer therapy through a variety of mechanisms such as bacterial dysbiosis, production of genotoxins, pathobionts, and disruption of the host metabolism are increasingly becoming evident. METHODS: Different electronic databases such as PubMed, Google Scholar, and Web of Science were searched for relevant literature which has been reviewed in this article. RESULTS: Characterization of the microbiome particularly gut microbiota, understanding of the host-microbiota interactions, and its potential for therapeutic exploitation are necessary for the development of novel anticancer therapeutic strategies with better efficacy and lowered off-target side effects. CONCLUSION: In this review, the role of microbiota is explained in carcinogenesis, mechanisms of microbiota-mediated carcinogenesis, and role of gut microbiota in modulation of cancer therapy.


Assuntos
Bactérias/patogenicidade , Interações entre Hospedeiro e Microrganismos , Microbiota/fisiologia , Neoplasias/microbiologia , Animais , Bactérias/isolamento & purificação , Toxinas Bacterianas/toxicidade , Carcinogênese , Disbiose , Humanos , Neoplasias/patologia , Neoplasias/terapia , Receptores de Reconhecimento de Padrão/metabolismo , Receptores Toll-Like/metabolismo , Fatores de Virulência/toxicidade
20.
Med Microbiol Immunol ; 208(1): 25-32, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30386929

RESUMO

Gram-negative bacterium Legionella is able to proliferate intracellularly in mammalian host cells and amoeba, which became known in 1976 since they caused a large outbreak of pneumonia. It had been reported that different strains of Legionella pneumophila, Legionella micdadei, Legionella longbeachae, and Legionella feeleii caused human respiratory diseases, which were known as Pontiac fever or Legionnaires' disease. However, the differences of the virulence traits among the strains of the single species and the pathogenesis of the two diseases that were due to the bacterial virulence factors had not been well elucidated. L. feeleii is an important pathogenic organism in Legionellae, which attracted attention due to cause an outbreak of Pontiac fever in 1981 in Canada. In published researches, it has been found that L. feeleii serogroup 2 (ATCC 35849, LfLD) possess mono-polar flagellum, and L. feeleii serogroup 1 (ATCC 35072, WRLf) could secrete some exopolysaccharide (EPS) materials to the surrounding. Although the virulence of the L. feeleii strain was evidenced that could be promoted, the EPS might be dispensable for the bacteria that caused Pontiac fever. Based on the current knowledge, we focused on bacterial infection in human and murine host cells, intracellular growth, cytopathogenicity, stimulatory capacity of cytokines secretion, and pathogenic effects of the EPS of L. feeleii in this review.


Assuntos
Citocinas/metabolismo , Legionella/patogenicidade , Doença dos Legionários/microbiologia , Doença dos Legionários/patologia , Polissacarídeos Bacterianos/metabolismo , Fatores de Virulência/metabolismo , Animais , Linhagem Celular , Humanos , Legionella/crescimento & desenvolvimento , Camundongos , Polissacarídeos Bacterianos/toxicidade , Virulência , Fatores de Virulência/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...