Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
1.
Front Cell Infect Microbiol ; 12: 1002230, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36389147

RESUMO

Group A Streptococcus (GAS, Streptococcus pyogenes) is a Gram-positive human pathogen that employs several secreted and surface-bound virulence factors to manipulate its environment, allowing it to cause a variety of disease outcomes. One such virulence factor is Streptolysin S (SLS), a ribosomally-produced peptide toxin that undergoes extensive post-translational modifications. The activity of SLS has been studied for over 100 years owing to its rapid and potent ability to lyse red blood cells, and the toxin has been shown to play a major role in GAS virulence in vivo. We have previously demonstrated that SLS induces hemolysis by targeting the chloride-bicarbonate exchanger Band 3 in erythrocytes, indicating that SLS is capable of targeting host proteins to promote cell lysis. However, the possibility that SLS has additional protein targets in other cell types, such as keratinocytes, has not been explored. Here, we use bioinformatics analysis and chemical inhibition studies to demonstrate that SLS targets the electroneutral sodium-bicarbonate cotransporter NBCn1 in keratinocytes during GAS infection. SLS induces NF-κB activation and host cytotoxicity in human keratinocytes, and these processes can be mitigated by treating keratinocytes with the sodium-bicarbonate cotransport inhibitor S0859. Furthermore, treating keratinocytes with SLS disrupts the ability of host cells to regulate their intracellular pH, and this can be monitored in real time using the pH-sensitive dye pHrodo Red AM in live imaging studies. These results demonstrate that SLS is a multifunctional bacterial toxin that GAS uses in numerous context-dependent ways to promote host cell cytotoxicity and increase disease severity. Studies to elucidate additional host targets of SLS have the potential to impact the development of therapeutics for severe GAS infections.


Assuntos
Infecções Estreptocócicas , Estreptolisinas , Humanos , Estreptolisinas/toxicidade , Estreptolisinas/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes , Queratinócitos/metabolismo , Inflamação
2.
Toxins (Basel) ; 14(5)2022 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-35622592

RESUMO

Melittin, the main toxic component in the venom of the European honeybee, interacts with natural and artificial membranes due to its amphiphilic properties. Rather than interacting with a specific receptor, melittin interacts with the lipid components, disrupting the lipid bilayer and inducing ion leakage and osmotic shock. This mechanism of action is shared with pneumolysin and other members of the cholesterol-dependent cytolysin family. In this manuscript, we investigated the inverse correlation for cholesterol dependency of these two toxins. While pneumolysin-induced damage is reduced by pretreatment with the cholesterol-depleting agent methyl-ß-cyclodextrin, the toxicity of melittin, after cholesterol depletion, increased. A similar response was also observed after a short incubation with lipophilic simvastatin, which alters membrane lipid organization and structure, clustering lipid rafts. Therefore, changes in toxin sensitivity can be achieved in cells by depleting cholesterol or changing the lipid bilayer organization.


Assuntos
Bicamadas Lipídicas , Meliteno , Animais , Proteínas de Bactérias , Abelhas , Colesterol , Meliteno/química , Meliteno/toxicidade , Estreptolisinas/toxicidade
3.
J Pharmacol Exp Ther ; 379(2): 117-124, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34389653

RESUMO

Streptolysin O (SLO) is produced by common hemolytic streptococci that cause a wide range of diseases from pharyngitis to life-threatening necrotizing fasciitis and toxic shock syndrome. Although the importance of SLO in invasive hemolytic streptococcus infection has been well demonstrated, the role of circulating SLO in noninvasive infection remains unclear. The aim of this study was to characterize the pharmacological effect of SLO on vascular functions, focusing on cellular signaling pathways. In control Wistar rats, SLO treatment (1-1000 ng/ml) impaired acetylcholine-induced endothelial-dependent relaxation in the aorta and second-order mesenteric artery in a dose-dependent manner without any effects on sodium nitroprusside-induced endothelium-independent relaxation or agonist-induced contractions. SLO also increased phosphorylation of the endothelial NO synthase (eNOS) inhibitory site at Thr495 in the aorta. Pharmacological analysis indicated that either endothelial dysfunction or eNOS phosphorylation was mediated by protein kinase Cß (PKCß), but not by the p38 mitogen-activated protein kinase pathway. Consistent with this, SLO increased phosphorylation levels of protein kinase C substrates in the aorta. In vivo study of control Wistar rats indicated that intravenous administration of SLO did not change basal blood pressure but significantly counteracted the acetylcholine-induced decrease in blood pressure. Interestingly, plasma anti-SLO IgG levels were significantly higher in 10- to 15-week-old spontaneously hypertensive rats compared with age-matched control rats (P < 0.05). These findings demonstrated that SLO causes vascular endothelial dysfunction, which is mediated by PKCß-induced phosphorylation of the eNOS inhibitory site. SIGNIFICANCE STATEMENT: This study showed for the first time that in vitro exposure of vascular tissues to SLO impairs endothelial function, an effect that is mediated by protein kinase C ß-induced phosphorylation of the endothelial NO synthase inhibitory site. Intravenous administration of SLO in control and hypertensive rats blunted the acetylcholine-induced decrease in blood pressure, providing evidence for a possible role of SLO in dysregulation of blood pressure.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Proteína Quinase C beta/metabolismo , Estreptolisinas/toxicidade , Vasoconstrição/efeitos dos fármacos , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/enzimologia , Proteínas de Bactérias/toxicidade , Relação Dose-Resposta a Droga , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/enzimologia , Técnicas de Cultura de Órgãos , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Ratos Wistar , Vasoconstrição/fisiologia
4.
Can J Microbiol ; 66(7): 435-445, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32191844

RESUMO

Pneumolysin (Ply) is a major virulence factor of Streptococcus pneumoniae. Ply-induced interferon-ß (IFN-ß) expression in host macrophages has been shown to be due to the accumulation of mitochondrial deoxyribonucleic acid (mtDNA) in the cytoplasm during S. pneumoniae infection. Our findings extend this work to show human bronchial epithelial cells that reside at the interface of inflammatory injury, BEAS-2B, adapt to local cues by altering mitochondrial states and releasing excess mtDNA. The results in this research showed that purified Ply induced the expression of IFN-ß in human epithelial cells, which was accompanied by mitochondrial damage both in vivo and in vitro. The observations also were supported by the increased mtDNA concentrations in the bronchial lavage fluid of mice infected with S. pneumoniae. In summary, our study demonstrated that Ply triggered the production of IFN-ß in epithelial cells, and this response was mediated by mtDNA released from Ply-damaged mitochondria. It displayed an impressive modulation of IFN-ß response to S. pneumoniae in epithelial cells.


Assuntos
Citosol/metabolismo , DNA Mitocondrial/metabolismo , Interferon beta/metabolismo , Mitocôndrias/efeitos dos fármacos , Estreptolisinas/toxicidade , Animais , Proteínas de Bactérias/toxicidade , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/microbiologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Streptococcus pneumoniae/patogenicidade
5.
PLoS One ; 15(3): e0219275, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32163417

RESUMO

Pathogenic bacteria often damage tissues by secreting toxins that form pores in cell membranes, and the most common pore-forming toxins are cholesterol-dependent cytolysins. During bacterial infections, glutamine becomes a conditionally essential amino acid, and glutamine is an important nutrient for immune cells. However, the role of glutamine in protecting tissue cells against pore-forming toxins is unclear. Here we tested the hypothesis that glutamine supports the protection of tissue cells against the damage caused by cholesterol-dependent cytolysins. Stromal and epithelial cells were sensitive to damage by the cholesterol-dependent cytolysins, pyolysin and streptolysin O, as determined by leakage of potassium and lactate dehydrogenase from cells, and reduced cell viability. However, glutamine deprivation increased the leakage of lactate dehydrogenase and reduced the viability of cells challenged with cholesterol-dependent cytolysins. Without glutamine, stromal cells challenged with pyolysin leaked lactate dehydrogenase (control vs. pyolysin, 2.6 ± 0.6 vs. 34.4 ± 4.5 AU, n = 12), which was more than three-fold the leakage from cells supplied with 2 mM glutamine (control vs. pyolysin, 2.2 ± 0.3 vs. 9.4 ± 1.0 AU). Glutamine cytoprotection did not depend on glutaminolysis, replenishing the Krebs cycle via succinate, changes in cellular cholesterol, or regulators of cell metabolism (AMPK and mTOR). In conclusion, although the mechanism remains elusive, we found that glutamine supports the protection of tissue cells against the damage caused by cholesterol-dependent cytolysins from pathogenic bacteria.


Assuntos
Colesterol/metabolismo , Citoproteção/efeitos dos fármacos , Citotoxinas/toxicidade , Glutamina/farmacologia , Animais , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Bovinos , Células HeLa , Proteínas Hemolisinas/toxicidade , Humanos , L-Lactato Desidrogenase/metabolismo , Estreptolisinas/toxicidade , Células Estromais/efeitos dos fármacos
6.
Microbiol Immunol ; 64(1): 10-22, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31584206

RESUMO

Streptolysin O (SLO) is a bacterial pore-forming toxin that is employed to permeabilize cell membranes in some biological experiments. SLO forms various types of pores with different shapes, increasing membrane ion permeability and subsequently inducing changes in membrane potential. To characterize the pores formed by SLO, the changes in membrane potential induced by SLO in rat lymphocytes were considered using flow cytometry with a voltage-sensitive fluorescent probe, bis-(1,3-dibutylbarbituric acid)trimethine oxonol (Oxonol). SLO caused three types of membrane potential responses accessed with Oxonol. One type induces a great decrease in Oxonol fluorescence (large hyperpolarization) that may be elicited via the increase of Ca2+ -dependent K+ permeability by SLO-induced influx of external Ca2+ . A second type is an increase in Oxonol fluorescence (depolarization) that may be caused by a nonspecific increase in membrane cation permeability. The third type is a small decrease in Oxonol fluorescence (small hyperpolarization), probably via an increase in Cl- permeability. That SLO transitionally changes membrane ion permeability may have implications in the pathology of pyogenic group streptococci infections in which SLO is thought to be one of the key virulence factors.


Assuntos
Toxinas Bacterianas/toxicidade , Linfócitos/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Estreptolisinas/toxicidade , Animais , Proteínas de Bactérias/toxicidade , Barbitúricos , Calcimicina , Cálcio , Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Modelos Animais de Doenças , Citometria de Fluxo , Corantes Fluorescentes , Masculino , Ratos , Ratos Wistar
7.
Toxins (Basel) ; 11(1)2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30577571

RESUMO

The largest superfamily of bacterial virulence factors is pore-forming toxins (PFTs). PFTs are secreted by both pathogenic and non-pathogenic bacteria. PFTs sometimes kill or induce pro-pathogen signaling in mammalian cells, all primarily through plasma membrane perforation, though the parameters that determine these outcomes are unclear. Membrane binding, calcium influx, pore size, and membrane repair are factors that influence PFT cytotoxicity. To test the contribution of membrane binding to cytotoxicity and repair, we compared the closely related, similarly-sized PFTs Perfringolysin O (PFO) from Clostridium perfringens and Streptolysin O (SLO) from Streptococcus pyogenes. Cell death kinetics for PFO and SLO were different because PFO increased in cytotoxicity over time. We introduced known L3 loop mutations that swap binding affinity between toxins and measured hemolytic activity, nucleated cell death kinetics and membrane repair using viability assays, and live cell imaging. Altered hemolytic activity was directly proportional to toxin binding affinity. In contrast, L3 loop alterations reduced nucleated cell death, and they had limited effects on cytotoxicity kinetics and membrane repair. This suggests other toxin structural features, like oligomerization, drives these parameters. Overall, these findings suggest that repair mechanisms and toxin oligomerization add constraints beyond membrane binding on toxin evolution and activity against nucleated cells.


Assuntos
Toxinas Bacterianas/toxicidade , Proteínas Hemolisinas/toxicidade , Estreptolisinas/toxicidade , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/genética , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Colesterol/metabolismo , Feminino , Proteínas Hemolisinas/genética , Hemólise/efeitos dos fármacos , Humanos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos Knockout , Proteínas Recombinantes/toxicidade , Estreptolisinas/genética
8.
Int J Mol Sci ; 19(4)2018 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-29641429

RESUMO

Pneumolysin (PLY), a member of the family of Gram-positive bacterial, cholesterol-dependent, ß-barrel pore-forming cytolysins, is the major protein virulence factor of the dangerous respiratory pathogen, Streptococcus pneumoniae (pneumococcus). PLY plays a major role in the pathogenesis of community-acquired pneumonia (CAP), promoting colonization and invasion of the upper and lower respiratory tracts respectively, as well as extra-pulmonary dissemination of the pneumococcus. Notwithstanding its role in causing acute lung injury in severe CAP, PLY has also been implicated in the development of potentially fatal acute and delayed-onset cardiovascular events, which are now recognized as being fairly common complications of this condition. This review is focused firstly on updating mechanisms involved in the immunopathogenesis of PLY-mediated myocardial damage, specifically the direct cardiotoxic and immunosuppressive activities, as well as the indirect pro-inflammatory/pro-thrombotic activities of the toxin. Secondly, on PLY-targeted therapeutic strategies including, among others, macrolide antibiotics, natural product antagonists, cholesterol-containing liposomes, and fully humanized monoclonal antibodies, as well as on vaccine-based preventive strategies. These sections are preceded by overviews of CAP in general, the role of the pneumococcus as the causative pathogen, the occurrence and types of CAP-associated cardiac complication, and the structure and biological activities of PLY.


Assuntos
Doenças Cardiovasculares/etiologia , Infecções Comunitárias Adquiridas/complicações , Pneumonia Pneumocócica/complicações , Estreptolisinas/toxicidade , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/toxicidade , Doenças Cardiovasculares/tratamento farmacológico , Infecções Comunitárias Adquiridas/tratamento farmacológico , Infecções Comunitárias Adquiridas/etiologia , Humanos , Pneumonia Pneumocócica/tratamento farmacológico , Pneumonia Pneumocócica/etiologia , Estreptolisinas/antagonistas & inibidores
9.
Sci Rep ; 8(1): 182, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29317705

RESUMO

Streptococcus pneumoniae (S.pn.) is the most common bacterial pathogen causing community acquired pneumonia. The pore-forming toxin pneumolysin (PLY) is the major virulence factor of S.pn. and supposed to affect alveolar epithelial cells thereby activating the immune system by liberation of danger-associated molecular patterns (DAMP). To test this hypothesis, we established a novel live-cell imaging based assay to analyse mitochondrial function and associated release of mitochondrial DNA (mtDNA) as DAMP in real-time. We first revealed that bacterially released PLY caused significant changes of the cellular ATP homeostasis and led to morphologic alterations of mitochondria in human alveolar epithelial cells in vitro and, by use of spectral live-tissue imaging, in human alveoli. This was accompanied by strong mitochondrial calcium influx and loss of mitochondrial membrane potential resulting in opening of the mitochondrial permeability transition pore and mtDNA release without activation of intrinsic apoptosis. Moreover, our data indicate cellular mtDNA liberation via microvesicles, which may contribute to S.pn. related pro-inflammatory immune activation in the human alveolar compartment.


Assuntos
Células Epiteliais Alveolares/efeitos dos fármacos , DNA Mitocondrial/metabolismo , Mitocôndrias/efeitos dos fármacos , Estreptolisinas/toxicidade , Trifosfato de Adenosina/metabolismo , Células Epiteliais Alveolares/metabolismo , Proteínas de Bactérias/toxicidade , Cálcio/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial
10.
Chem Pharm Bull (Tokyo) ; 65(6): 538-544, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28566646

RESUMO

Streptococcus pneumoniae (pneumococcus) is an important causative agent of acute invasive and non-invasive infections. Pneumolysin is one of a considerable number of virulence traits produced by pneumococcus that exhibits a variety of biological activities, thus making it a target of small molecule drug development. In this study, we aimed to evaluate the effect of morin, a natural compound that has no antimicrobial activity against S. pneumonia, is a potent neutralizer of pneumolysin-mediated cytotoxicity and genotoxicity by impairing oligomer formation, and possesses the capability of mitigating tissue damage caused by pneumococcus. These findings indicate that morin could be a potent candidate for a novel therapeutic or auxiliary substance to treat infections for which there are inadequate vaccines and that are resistant to traditional antibiotics.


Assuntos
Biopolímeros/metabolismo , Flavonoides/farmacologia , Streptococcus pneumoniae/metabolismo , Estreptolisinas/toxicidade , Células A549 , Animais , Proteínas de Bactérias/toxicidade , Feminino , Hemólise/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Streptococcus pneumoniae/efeitos dos fármacos
11.
Toxins (Basel) ; 9(1)2017 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-28098781

RESUMO

Cholesterol-dependent cytolysins (CDCs) are protein toxins that originate from Gram-positive bacteria and contribute substantially to their pathogenicity. CDCs bind membrane cholesterol and build prepores and lytic pores. Some effects of the toxins are observed in non-lytic concentrations. Two pathogens, Streptococcus pneumoniae and Listeria monocytogenes, cause fatal bacterial meningitis, and both produce toxins of the CDC family-pneumolysin and listeriolysin O, respectively. It has been demonstrated that pneumolysin produces dendritic varicosities (dendrite swellings) and dendritic spine collapse in the mouse neocortex, followed by synaptic loss and astrocyte cell shape remodeling without elevated cell death. We utilized primary glial cultures and acute mouse brain slices to examine the neuropathological effects of listeriolysin O and to compare it to pneumolysin with identical hemolytic activity. In cultures, listeriolysin O permeabilized cells slower than pneumolysin did but still initiated non-lytic astrocytic cell shape changes, just as pneumolysin did. In an acute brain slice culture system, listeriolysin O produced dendritic varicosities in an NMDA-dependent manner but failed to cause dendritic spine collapse and cortical astrocyte reorganization. Thus, listeriolysin O demonstrated slower cell permeabilization and milder glial cell remodeling ability than did pneumolysin and lacked dendritic spine collapse capacity but exhibited equivalent dendritic pathology.


Assuntos
Astrócitos/efeitos dos fármacos , Toxinas Bacterianas/toxicidade , Encéfalo/efeitos dos fármacos , Espinhas Dendríticas/efeitos dos fármacos , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Listeria monocytogenes/metabolismo , Neurotoxinas/toxicidade , Animais , Animais Recém-Nascidos , Astrócitos/patologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/genética , Encéfalo/patologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Espinhas Dendríticas/patologia , Proteínas de Choque Térmico/genética , Proteínas Hemolisinas/genética , Camundongos Endogâmicos C57BL , Neurotoxinas/genética , Cultura Primária de Células , Proteínas Recombinantes , Estreptolisinas/genética , Estreptolisinas/toxicidade
12.
Sci Rep ; 6: 38013, 2016 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-27892542

RESUMO

Streptococcus pneumoniae is a leading cause of bacterial pneumonia and is the principal cause of morbidity and mortality worldwide. Previous studies suggested that excessive activation of neutrophils results in the release of neutrophil elastase, which contributes to lung injury in severe pneumonia. Although both pneumococcal virulence factors and neutrophil elastase contribute to the development and progression of pneumonia, there are no studies analysing relationships between these factors. Here, we showed that pneumolysin, a pneumococcal pore-forming toxin, induced cell lysis in primary isolated human neutrophils, leading to the release of neutrophil elastase. Pneumolysin exerted minimal cytotoxicity against alveolar epithelial cells and macrophages, whereas neutrophil elastase induced detachment of alveolar epithelial cells and impaired phagocytic activity in macrophages. Additionally, activation of neutrophil elastase did not exert bactericidal activity against S. pneumoniae in vitro. P2X7 receptor, which belongs to a family of purinergic receptors, was involved in pneumolysin-induced cell lysis. These findings suggested that infiltrated neutrophils are the primary target cells of pneumolysin, and that S. pneumoniae exploits neutrophil-elastase leakage to induce the disruption of pulmonary immune defences, thereby causing lung injury.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Elastase de Leucócito/metabolismo , Neutrófilos/patologia , Streptococcus pneumoniae/patogenicidade , Estreptolisinas/metabolismo , Células Epiteliais Alveolares , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/toxicidade , Adesão Celular , Morte Celular , Linhagem Celular , Humanos , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Neutrófilos/efeitos dos fármacos , Neutrófilos/microbiologia , Fagocitose/fisiologia , Receptores Purinérgicos P2X7/metabolismo , Estreptolisinas/genética , Estreptolisinas/toxicidade
13.
Sci Rep ; 6: 34560, 2016 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-27703200

RESUMO

Although pneumococcal infection is a serious problem worldwide and has a high mortality rate, the molecular mechanisms underlying the lethality caused by pneumococcus remain elusive. Here, we show that BLT2, a G protein-coupled receptor for leukotriene B4 and 12(S)-hydroxyheptadecatrienoic acid (12-HHT), protects mice from lung injury caused by a pneumococcal toxin, pneumolysin (PLY). Intratracheal injection of PLY caused lethal acute lung injury (ALI) in BLT2-deficient mice, with evident vascular leakage and bronchoconstriction. Large amounts of cysteinyl leukotrienes (cysLTs), classically known as a slow reactive substance of anaphylaxis, were detected in PLY-treated lungs. PLY-dependent vascular leakage, bronchoconstriction, and death were markedly ameliorated by treatment with a CysLT1 receptor antagonist. Upon stimulation by PLY, mast cells produced cysLTs that activated CysLT1 expressed in vascular endothelial cells and bronchial smooth muscle cells, leading to lethal vascular leakage and bronchoconstriction. Treatment of mice with aspirin or loxoprofen inhibited the production of 12-HHT and increased the sensitivity toward PLY, which was also ameliorated by the CysLT1 antagonist. Thus, the present study identifies the molecular mechanism underlying PLY-dependent ALI and suggests the possible use of CysLT1 antagonists as a therapeutic tool to protect against ALI caused by pneumococcal infection.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/prevenção & controle , Permeabilidade Capilar/efeitos dos fármacos , Células Endoteliais/metabolismo , Receptores do Leucotrieno B4/metabolismo , Estreptolisinas/toxicidade , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Animais , Proteínas de Bactérias/toxicidade , Células Endoteliais/patologia , Mastócitos/metabolismo , Mastócitos/patologia , Camundongos , Camundongos Knockout , Receptores do Leucotrieno B4/genética
14.
J Vis Exp ; (114)2016 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-27585035

RESUMO

Many bacterial pathogens secrete potent toxins to aid in the destruction of host tissue, to initiate signaling changes in host cells or to manipulate immune system responses during the course of infection. Though methods have been developed to successfully purify and produce many of these important virulence factors, there are still many bacterial toxins whose unique structure or extensive post-translational modifications make them difficult to purify and study in in vitro systems. Furthermore, even when pure toxin can be obtained, there are many challenges associated with studying the specific effects of a toxin under relevant physiological conditions. Most in vitro cell culture models designed to assess the effects of secreted bacterial toxins on host cells involve incubating host cells with a one-time dose of toxin. Such methods poorly approximate what host cells actually experience during an infection, where toxin is continually produced by bacterial cells and allowed to accumulate gradually during the course of infection. This protocol describes the design of a permeable membrane insert-based bacterial infection system to study the effects of Streptolysin S, a potent toxin produced by Group A Streptococcus, on human epithelial keratinocytes. This system more closely mimics the natural physiological environment during an infection than methods where pure toxin or bacterial supernatants are directly applied to host cells. Importantly, this method also eliminates the bias of host responses that are due to direct contact between the bacteria and host cells. This system has been utilized to effectively assess the effects of Streptolysin S (SLS) on host membrane integrity, cellular viability, and cellular signaling responses. This technique can be readily applied to the study of other secreted virulence factors on a variety of mammalian host cell types to investigate the specific role of a secreted bacterial factor during the course of infection.


Assuntos
Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Queratinócitos/microbiologia , Membranas Artificiais , Streptococcus pyogenes/patogenicidade , Estreptolisinas/toxicidade , Trifosfato de Adenosina/metabolismo , Transporte Biológico , Western Blotting , Células Cultivadas , Contagem de Colônia Microbiana , Eletroforese em Gel de Poliacrilamida , Humanos , Queratinócitos/metabolismo , Microscopia de Fluorescência , NF-kappa B/metabolismo
15.
Biochim Biophys Acta ; 1860(11 Pt A): 2498-2509, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27481675

RESUMO

BACKGROUND: Streptococcus pneumoniae is a potent human pathogen. Its pore-forming exotoxin pneumolysin is instrumental for breaching the host's epithelial barrier and for the incapacitation of the immune system. METHODS AND RESULTS: Using a combination of life imaging and cryo-electron microscopy we show that pneumolysin, released by cultured bacteria, is capable of permeabilizing the plasmalemma of host cells. However, such permeabilization does not lead to cell lysis since pneumolysin is actively removed by the host cells. The process of pore elimination starts with the formation of pore-bearing plasmalemmal nanotubes and proceeds by the shedding of pores that are embedded in the membrane of released microvesicles. Pneumolysin prepores are likewise removed. The protein composition of the toxin-induced microvesicles, assessed by mass spectrometry, is suggestive of a Ca(2+)-triggered mechanism encompassing the proteins of the annexin family and members of the endosomal sorting complex required for transport (ESCRT) complex. CONCLUSIONS: S. pneumoniae releases sufficient amounts of pneumolysin to perforate the plasmalemma of host cells, however, the immediate cell lysis, which is frequently reported as a result of treatment with purified and artificially concentrated toxin, appears to be an unlikely event in vivo since the toxin pores are efficiently eliminated by microvesicle shedding. Therefore the dysregulation of cellular homeostasis occurring as a result of transient pore formation/elimination should be held responsible for the damaging toxin action. GENERAL SIGNIFICANCE: We have achieved a comprehensive view of a general plasma membrane repair mechanism after injury by a major bacterial toxin.


Assuntos
Membrana Celular/ultraestrutura , Streptococcus pneumoniae/patogenicidade , Estreptolisinas/farmacologia , Proteínas de Bactérias/farmacologia , Proteínas de Bactérias/toxicidade , Membrana Celular/efeitos dos fármacos , Membrana Celular/microbiologia , Permeabilidade da Membrana Celular , Células HEK293 , Células HeLa , Humanos , Estreptolisinas/toxicidade
16.
Infect Immun ; 84(5): 1457-69, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26930705

RESUMO

Streptococcus pneumoniae (the pneumococcus) is capable of invading the heart. Herein we observed that pneumococcal invasion of the myocardium occurred soon after development of bacteremia and was continuous thereafter. Using immunofluorescence microscopy (IFM), we observed that S. pneumoniae replication within the heart preceded visual signs of tissue damage in cardiac tissue sections stained with hematoxylin and eosin. Different S. pneumoniae strains caused distinct cardiac pathologies: strain TIGR4, a serotype 4 isolate, caused discrete pneumococcus-filled microscopic lesions (microlesions), whereas strain D39, a serotype 2 isolate, was, in most instances, detectable only using IFM and was associated with foci of cardiomyocyte hydropic degeneration and immune cell infiltration. Both strains efficiently invaded the myocardium, but cardiac damage was entirely dependent on the pore-forming toxin pneumolysin only for D39. Early microlesions caused by TIGR4 and microlesions formed by a TIGR4 pneumolysin-deficient mutant were infiltrated with CD11b(+) and Ly6G-positive neutrophils and CD11b(+) and F4/80-positive (F4/80(+)) macrophages. We subsequently demonstrated that macrophages in TIGR4-infected hearts died as a result of pneumolysin-induced necroptosis. The effector of necroptosis, phosphorylated mixed-lineage kinase domain-like protein (MLKL), was detected in CD11b(+) and F4/80(+) cells associated with microlesions. Likewise, treatment of infected mice and THP-1 macrophages in vitro with the receptor-interacting protein 1 kinase (RIP1) inhibitor necrostatin-5 promoted the formation of purulent microlesions and blocked cell death, respectively. We conclude that pneumococci that have invaded the myocardium are an important cause of cardiac damage, pneumolysin contributes to cardiac damage in a bacterial strain-specific manner, and pneumolysin kills infiltrated macrophages via necroptosis, which alters the immune response.


Assuntos
Bacteriemia/patologia , Morte Celular/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Miocardite/patologia , Infecções Pneumocócicas/patologia , Streptococcus pneumoniae/patogenicidade , Estreptolisinas/toxicidade , Animais , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/toxicidade , Linhagem Celular , Modelos Animais de Doenças , Feminino , Humanos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Proteínas Quinases/análise , Streptococcus pneumoniae/metabolismo , Estreptolisinas/metabolismo
17.
Sci Rep ; 5: 16945, 2015 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-26601609

RESUMO

Streptococcus dysgalactiae subsp. equisimilis (SDSE) has emerged as an important cause of severe skin and soft tissue infections, but little is known of the pathogenic mechanisms underlying tissue pathology. Patient samples and a collection of invasive and non-invasive group G SDSE strains (n = 69) were analyzed with respect to virulence factor expression and cytotoxic or inflammatory effects on human cells and 3D skin tissue models. SDSE strains efficiently infected the 3D-skin model and severe tissue pathology, inflammatory responses and altered production of host structural framework proteins associated with epithelial barrier integrity were evident already at 8 hours post-infection. Invasive strains were significantly more cytotoxic towards keratinocytes and expressed higher Streptokinase and Streptolysin O (SLO) activities, as compared to non-invasive strains. The opposite was true for Streptolysin S (SLS). Fractionation and proteomic analysis of the cytotoxic fractions implicated SLO as a factor likely contributing to the keratinocyte cytotoxicity and tissue pathology. Analyses of patient tissue biopsies revealed massive bacterial load, high expression of slo, as well as immune cell infiltration and pro-inflammatory markers. Our findings suggest the contribution of SLO to epithelial cytotoxicity and tissue pathology in SDSE tissue infections.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções Estreptocócicas/microbiologia , Streptococcus/metabolismo , Estreptolisinas/metabolismo , Proteínas de Bactérias/toxicidade , Técnicas de Cultura de Células , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/análise , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Proteína HMGB1/análise , Humanos , Interleucina-8/análise , Queratinócitos/citologia , Queratinócitos/metabolismo , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Proteoma/análise , Pele/metabolismo , Pele/microbiologia , Pele/patologia , Infecções dos Tecidos Moles/microbiologia , Infecções dos Tecidos Moles/patologia , Infecções Estreptocócicas/patologia , Streptococcus/patogenicidade , Estreptolisinas/toxicidade , Espectrometria de Massas em Tandem , Fatores de Virulência/metabolismo , Fatores de Virulência/toxicidade
18.
BMC Microbiol ; 15: 141, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26204951

RESUMO

BACKGROUND: Streptolysin S (SLS) is a cytolytic virulence factor produced by the human pathogen Streptococcus pyogenes and other Streptococcus species. Related "SLS-like" toxins have been characterized in select strains of Clostridium and Listeria, with homologous clusters bioinformatically identified in a variety of other species. SLS is a member of the thiazole/oxazole-modified microcin (TOMM) family of natural products. The structure of SLS has yet to be deciphered and many questions remain regarding its structure-activity relationships. RESULTS: In this work, we assessed the hemolytic activity of a series of C-terminally truncated SLS peptides expressed in SLS-deficient S. pyogenes. Our data indicate that while the N-terminal poly-heterocyclizable (NPH) region of SLS substantially contributes to its bioactivity, the variable C-terminal region of the toxin is largely dispensable. Through genome mining we identified additional SLS-like clusters in diverse Firmicutes, Spirochaetes and Actinobacteria. Among the Spirochaete clusters, naturally truncated SLS-like precursors were found in the genomes of three Lyme disease-causing Borrelia burgdorferi sensu lato (Bbsl) strains. Although unable to restore hemolysis in SLS-deficient S. pyogenes, a Bbsl SLS-like precursor peptide was converted to a cytolysin using purified SLS biosynthetic enzymes. A PCR-based screen demonstrated that SLS-like clusters are substantially more prevalent in Bbsl than inferred from publicly available genome sequences. CONCLUSIONS: The mutagenesis data described herein indicate that the minimal cytolytic unit of SLS encompasses the NPH region of the core peptide. Interestingly, this region is found in all characterized TOMM cytolysins, as well as the novel putative TOMM cytolysins we discovered. We propose that this conserved region represents the defining feature of the SLS-like TOMM family. We demonstrate the cytolytic potential of a Bbsl SLS-like precursor peptide, which has a core region of similar length to the SLS minimal cytolytic unit, when modified with purified SLS biosynthetic enzymes. As such, we speculate that some Borrelia have the potential to produce a TOMM cytolysin, although the biological significance of this finding remains to be determined. In addition to providing new insight into the structure-activity relationships of SLS, this study greatly expands the cytolysin group of TOMMs.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/toxicidade , Hemólise , Estreptolisinas/genética , Estreptolisinas/toxicidade , Análise por Conglomerados , Análise Mutacional de DNA , Humanos , Filogenia , Homologia de Sequência de Aminoácidos , Streptococcus pyogenes/metabolismo
19.
Sci Rep ; 5: 10624, 2015 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-26023727

RESUMO

Pneumonia remains one of the leading causes of death in both adults and children worldwide. Despite the adoption of a wide variety of therapeutics, the mortality from community-acquired pneumonia has remained relatively constant. Although viral and fungal acute airway infections can result in pneumonia, bacteria are the most common cause of community-acquired pneumonia, with Streptococcus pneumoniae isolated in nearly 50% of cases. Pneumolysin is a cholesterol-dependent cytolysin or pore-forming toxin produced by Streptococcus pneumonia and has been shown to play a critical role in bacterial pathogenesis. Airway epithelium is the initial site of many bacterial contacts and its barrier and mucosal immunity functions are central to infectious lung diseases. In our studies, we have shown that the prior exposure to statins confers significant resistance of airway epithelial cells to the cytotoxicity of pneumolysin. We decided to take this study one step further, assessing changes in both the transcriptome and lipidome of human airway epithelial cells exposed to toxin, statin or both. Our current work provides the first global view in human airway epithelial cells of both the transcriptome and the lipid interactions that result in cellular protection from pneumolysin.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Metabolismo dos Lipídeos , Metaboloma , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Estreptolisinas/toxicidade , Transcriptoma , Proteínas de Bactérias/toxicidade , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Análise por Conglomerados , Biologia Computacional , Células Epiteliais , Ácidos Graxos/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Lipídeos de Membrana/metabolismo , Substâncias Protetoras/farmacologia , Transdução de Sinais
20.
Am J Respir Cell Mol Biol ; 53(5): 689-702, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25874372

RESUMO

Statins are widely used to prevent cardiovascular disease. In addition to their inhibitory effects on cholesterol synthesis, statins have beneficial effects in patients with sepsis and pneumonia, although molecular mechanisms have mostly remained unclear. Using human airway epithelial cells as a proper in vitro model, we show that prior exposure to physiological nanomolar serum concentrations of simvastatin (ranging from 10-1,000 nM) confers significant cellular resistance to the cytotoxicity of pneumolysin, a pore-forming toxin and the main virulence factor of Streptococcus pneumoniae. This protection could be demonstrated with a different statin, pravastatin, or on a different toxin, α-hemolysin. Furthermore, through the use of gene silencing, pharmacological inhibitors, immunofluorescence microscopy, and biochemical and metabolic rescue approaches, we demonstrate that the mechanism of protection conferred by simvastatin at physiological nanomolar concentrations could be different from the canonical mevalonate pathways seen in most other mechanistic studies conducted with statins at micromolar levels. All of these data are integrated into a protein synthesis-dependent, calcium-dependent model showing the interconnected pathways used by statins in airway epithelial cells to elicit an increased resistance to pore-forming toxins. This research fills large gaps in our understanding of how statins may confer host cellular protection against bacterial infections in the context of airway epithelial cells without the confounding effect from the presence of immune cells. In addition, our discovery could be potentially developed into a host-centric strategy for the adjuvant treatment of pore-forming toxin associated bacterial infections.


Assuntos
Toxinas Bacterianas/antagonistas & inibidores , Células Epiteliais/efeitos dos fármacos , Proteínas Hemolisinas/antagonistas & inibidores , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Imunidade Inata/efeitos dos fármacos , Sinvastatina/farmacologia , Estreptolisinas/antagonistas & inibidores , Animais , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Linhagem Celular Transformada , Células Epiteliais/imunologia , Células Epiteliais/patologia , Proteínas Hemolisinas/toxicidade , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/imunologia , Injeções Intraperitoneais , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Pravastatina/imunologia , Pravastatina/farmacologia , Cultura Primária de Células , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia , Sinvastatina/imunologia , Staphylococcus aureus/química , Streptococcus pneumoniae/química , Estreptolisinas/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...