Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
J Biol Chem ; 299(12): 105321, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37802313

RESUMEN

Staphylococcus aureus (S. aureus) is a serious global pathogen that causes a diverse range of invasive diseases. S. aureus utilizes a family of pore-forming toxins, known as bi-component leukocidins, to evade the host immune response and promote infection. Among these is LukAB (leukocidin A/leukocidin B), a toxin that assembles into an octameric ß-barrel pore in the target cell membrane, resulting in host cell death. The established cellular receptor for LukAB is CD11b of the Mac-1 complex. Here, we show that hydrogen voltage-gated channel 1 is also required for the cytotoxicity of all major LukAB variants. We demonstrate that while each receptor is sufficient to recruit LukAB to the plasma membrane, both receptors are required for maximal lytic activity. Why LukAB requires two receptors, and how each of these receptors contributes to pore-formation remains unknown. To begin to resolve this, we performed an alanine scanning mutagenesis screen to identify mutations that allow LukAB to maintain cytotoxicity without CD11b. We discovered 30 mutations primarily localized in the stem domains of LukA and LukB that enable LukAB to exhibit full cytotoxicity in the absence of CD11b. Using crosslinking, electron microscopy, and hydroxyl radical protein footprinting, we show these mutations increase the solvent accessibility of the stem domain, priming LukAB for oligomerization. Together, our data support a model in which CD11b binding unlatches the membrane penetrating stem domains of LukAB, and this change in flexibility promotes toxin oligomerization.


Asunto(s)
Proteínas Bacterianas , Leucocidinas , Staphylococcus aureus , Toxinas Biológicas , Humanos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Membrana Celular/metabolismo , Leucocidinas/genética , Leucocidinas/metabolismo , Leucocidinas/toxicidad , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Toxinas Biológicas/metabolismo , Mutación , Unión Proteica/genética , Dominios Proteicos , Línea Celular , Células CHO , Cricetulus , Animales
2.
Life Sci Alliance ; 6(6)2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36977592

RESUMEN

Staphylococcus aureus causes severe infections such as pneumonia and sepsis depending on the pore-forming toxin Panton-Valentine leukocidin (PVL). PVL kills and induces inflammation in macrophages and other myeloid cells by interacting with the human cell surface receptor, complement 5a receptor 1 (C5aR1). C5aR1 expression is tighly regulated and may thus modulate PVL activity, although the mechanisms involved remain incompletely understood. Here, we used a genome-wide CRISPR/Cas9 screen and identified F-box protein 11 (FBXO11), an E3 ubiquitin ligase complex member, to promote PVL toxicity. Genetic deletion of FBXO11 reduced the expression of C5aR1 at the mRNA level, whereas ectopic expression of C5aR1 in FBXO11-/- macrophages, or priming with LPS, restored C5aR1 expression and thereby PVL toxicity. In addition to promoting PVL-mediated killing, FBXO11 dampens secretion of IL-1ß after NLRP3 activation in response to bacterial toxins by reducing mRNA levels in a BCL-6-dependent and BCL-6-independent manner. Overall, these findings highlight that FBXO11 regulates C5aR1 and IL-1ß expression and controls macrophage cell death and inflammation following PVL exposure.


Asunto(s)
Toxinas Bacterianas , Proteínas F-Box , Humanos , Neutrófilos/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Exotoxinas/metabolismo , Exotoxinas/toxicidad , Inflamación/genética , Inflamación/metabolismo , Macrófagos/metabolismo , Muerte Celular/genética , Leucocidinas/farmacología , Leucocidinas/toxicidad , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas F-Box/genética , Proteínas F-Box/metabolismo
3.
Toxins (Basel) ; 15(1)2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36668859

RESUMEN

Background: Panton−Valentine Leukocidin sustains a strong cytotoxic activity, targeting immune cells and, consequently, perforating the plasma membrane and inducing cell death. The present study is aimed to examine the individual effect of ascorbic acid and nicotinamide on PVL cytotoxicity ex vivo, as well as their effect on granulocytes viability when treated with PVL. Materials and Methods: The PVL cytotoxicity assay was performed in triplicates using the commercial Cytotoxicity Detection Kit PLUS (LDH). LDH release was measured to determine cell damage and cell viability was measured via flow cytometry. Results and discussion: A clear reduction in PVL cytotoxicity was demonstrated (p < 0.001). Treatment with ascorbic acid at 5 mg/mL has shown a 3-fold reduction in PVL cytotoxicity; likewise, nicotinamide illustrated a 4-fold reduction in PVL cytotoxicity. Moreover, granulocytes' viability after PVL treatment was maintained when incubated with 5 mg/mL of ascorbic acid and nicotinamide. Conclusions: our findings illustrated that ascorbic acid and nicotinamide exhibit an inhibitory effect on PVL cytotoxicity and promote cell viability, as the cytotoxic effect of the toxin is postulated to be neutralized by antioxidant incubation. Further investigations are needed to assess whether these antioxidants may be viable options in PVL cytotoxicity attenuation in PVL-associated diseases.


Asunto(s)
Ácido Ascórbico , Toxinas Bacterianas , Leucocidinas , Niacinamida , Humanos , Ácido Ascórbico/química , Ácido Ascórbico/farmacología , Toxinas Bacterianas/toxicidad , Exotoxinas/toxicidad , Leucocidinas/toxicidad , Niacinamida/química , Niacinamida/farmacología
4.
Nat Commun ; 12(1): 6193, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702812

RESUMEN

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.


Asunto(s)
Membrana Celular/metabolismo , Pared Celular/metabolismo , Staphylococcus aureus/metabolismo , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Citotoxinas/metabolismo , Citotoxinas/toxicidad , Humanos , Leucocidinas/metabolismo , Leucocidinas/toxicidad , Lipopolisacáridos/genética , Lipopolisacáridos/metabolismo , Lisina/genética , Lisina/metabolismo , Ratones , Fagocitos/efectos de los fármacos , Fosfatidilgliceroles/genética , Fosfatidilgliceroles/metabolismo , Transporte de Proteínas , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Ácidos Teicoicos/genética , Ácidos Teicoicos/metabolismo , Factores de Virulencia/toxicidad
5.
Jpn J Infect Dis ; 74(4): 352-358, 2021 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-33518621

RESUMEN

Necrotizing pneumonia caused by Panton-Valentine leukocidin (PVL)-positive community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA) has high mortality rates and is currently a serious clinical issue. PVL is a two-component toxin (LukS-PV and LukF-PV). It can cause necrosis in target cells by forming pores consisting of an octamer comprised of LukS-PV and LukF-PV. However, considering the specificity of PVL towards several target cells and species, the specific effect of PVL remains controversial. Therefore, we focused on necrotizing pneumonia caused by PVL-positive S. aureus and clarified the effect of PVL on alveolar macrophages, which play a central role in innate immunity in the alveolar space. We constructed recombinant PVL (rPVL) components and stimulated alveolar macrophages isolated from rabbits to evaluate cytotoxicity and pro-inflammatory cytokine release. Recombinant LukS-PV (rLukS-PV), but not recombinant LukF-PV (rLukF-PV), induced pro-inflammatory cytokine release. Specifically, tumor necrosis factor (TNF)-α release was mediated by the C5a receptor (C5aR) expressed on rabbit alveolar macrophages, and the toxicity of rPVL, consisting of rLukS-PV and rLukF-PV, towards rabbit alveolar macrophages was mediated by the same receptor. Overall, our findings shed light on the C5aR-mediated cytotoxic effect of PVL on alveolar macrophages, which may be useful for understanding the mechanism of necrotizing pneumonia caused by PVL.


Asunto(s)
Toxinas Bacterianas/toxicidad , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Exotoxinas/toxicidad , Leucocidinas/toxicidad , Macrófagos Alveolares/efectos de los fármacos , Receptor de Anafilatoxina C5a/metabolismo , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Conejos , Receptor de Anafilatoxina C5a/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/toxicidad
6.
Toxins (Basel) ; 12(11)2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33233557

RESUMEN

Staphylococcus aureus is a major human pathogen, inducing several infections ranging from the benign to the life-threatening, such as necrotising pneumonia. S. aureus is capable of producing a great variety of virulence factors, such as bicomponent pore-forming leucocidin, which take part in the physiopathology of staphylococcal infection. In necrotising pneumonia, Panton-Valentine leucocidin (PVL) induces not only lung injury and necrosis, but also leukopenia, regarded as a major factor of a poor prognosis. The aim of the present study was to evaluate the effect of bicomponent pore-forming leucocidin, PVL and gamma haemolysin on bone marrow leucocytes, to better understand the origin of leukopenia. Using multi-parameter cytometry, the expression of leucocidin receptors (C5aR, CXCR1, CXCR2, and CCR2) was assessed and toxin-induced lysis was measured for each bone marrow leucocyte population. We observed that PVL resulted in myeloid-derived cells lysis according to their maturation and their C5aR expression; it also induced monocytes lysis according to host susceptibility. Haemolysin gamma A, B, and C (HlgABC) displayed cytotoxicity to monocytes and natural killer cells, hypothetically through CXCR2 and CXCR1 receptors, respectively. Taken together, the data suggest that PVL and HlgABC can lyse bone marrow leucocytes. Nevertheless, the origin of leukopenia in severe staphylococcal infection is predominantly peripheral, since immature cells stay insensitive to leucocidins.


Asunto(s)
Toxinas Bacterianas/toxicidad , Exotoxinas/toxicidad , Proteínas Hemolisinas/toxicidad , Leucocidinas/toxicidad , Leucocitos/efectos de los fármacos , Staphylococcus aureus , Células de la Médula Ósea/citología , Supervivencia Celular/efectos de los fármacos , Humanos
7.
Cell Microbiol ; 22(11): e13251, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32779854

RESUMEN

During acute Pseudomonas aeruginosa infection, the inflammatory response is essential for bacterial clearance. Neutrophil recruitment can be initiated following the assembly of an inflammasome within sentinel macrophages, leading to activation of caspase-1, which in turn triggers macrophage pyroptosis and IL-1ß/IL-18 maturation. Inflammasome formation can be induced by a number of bacterial determinants, including Type III secretion systems (T3SSs) or pore-forming toxins, or, alternatively, by lipopolysaccharide (LPS) via caspase-11 activation. Surprisingly, previous studies indicated that a T3SS-induced inflammasome increased pathogenicity in mouse models of P. aeruginosa infection. Here, we investigated the immune reaction of mice infected with a T3SS-negative P. aeruginosa strain (IHMA879472). Virulence of this strain relies on ExlA, a secreted pore-forming toxin. IHMA879472 promoted massive neutrophil infiltration in infected lungs, owing to efficient priming of toll-like receptors, and thus enhanced the expression of inflammatory proteins including pro-IL-1ß and TNF-α. However, mature-IL-1ß and IL-18 were undetectable in wild-type mice, suggesting that ExlA failed to effectively activate caspase-1. Nevertheless, caspase-1/11 deficiency improved survival following infection with IHMA879472, as previously described for T3SS+ bacteria. We conclude that the detrimental effect associated with the ExlA-induced inflammasome is probably not due to hyperinflammation, rather it stems from another inflammasome-dependent process.


Asunto(s)
Inflamasomas/inmunología , Leucocidinas/toxicidad , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/patogenicidad , Animales , Citocinas/biosíntesis , Inflamasomas/metabolismo , Inflamación , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Pulmón/inmunología , Pulmón/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Infiltración Neutrófila , Fragmentos de Péptidos/metabolismo , Pseudomonas aeruginosa/crecimiento & desarrollo , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/metabolismo , Sistemas de Secreción Tipo III , Virulencia
8.
Medicine (Baltimore) ; 99(20): e19617, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32443285

RESUMEN

To describe the epidemiological, clinical, laboratory, and radiological features and the management of adult patients who experienced a relapse between 2003 and 2015 of an acute hematogenous osteomyelitis acquired in childhood.A retrospective multicentric cohort study was conducted in 5 centers in France.Thirty-seven patients were included. The median age was 40 years (28-56), and 26 (70%) were male. The first site of infection was the distal femur (n = 23, 62%). The median time between the osteomyelitis in childhood and the relapse in adulthood was 26 years (13-45). Thirty-four (92%) patients reported inflammatory local clinical manifestations, 17 (46%) draining fistula, 10 (27%) fever. Most patients had intramedullary gadolinium deposition (with or without abscess) on magnetic resonance imaging. Most relapses were monomicrobial infections (82%). Staphylococcus aureus was the most commonly found microorganism (82%), expressing a small colony variant phenotype in 3 cases. Most patients (97%) had a surgical treatment, and the median duration of antibiotics for the relapse was 12 weeks. All patients had a favorable outcome, no patient died and no further relapse occurred. We count 2 femoral fractures on osteotomy site.Osteomyelitis in childhood can relapse later in adulthood, especially in patients with lack of care during the initial episode. Osteotomy and prolonged antimicrobial therapy are required for clinical remission.


Asunto(s)
Osteomielitis/epidemiología , Adulto , Anciano , Toxinas Bacterianas/toxicidad , Exotoxinas/toxicidad , Femenino , Francia/epidemiología , Humanos , Leucocidinas/toxicidad , Masculino , Persona de Mediana Edad , Osteomielitis/diagnóstico por imagen , Osteomielitis/microbiología , Osteomielitis/terapia , Recurrencia , Estudios Retrospectivos , Infecciones Estafilocócicas/complicaciones , Adulto Joven
9.
Toxins (Basel) ; 12(2)2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32041354

RESUMEN

Staphylococcal bi-component pore-forming toxins, also known as leukocidins, target and lyse human phagocytes in a receptor-dependent manner. S-components of the leukocidins Panton-Valentine leukocidin (PVL), γ-haemolysin AB (HlgAB) and CB (HlgCB), and leukocidin ED (LukED) specifically employ receptors that belong to the class of G-protein coupled receptors (GPCRs). Although these receptors share a common structural architecture, little is known about the conserved characteristics of the interaction between leukocidins and GPCRs. In this study, we investigated host cellular pathways contributing to susceptibility towards S. aureus leukocidin cytotoxicity. We performed a genome-wide CRISPR/Cas9 library screen for toxin-resistance in U937 cells sensitized to leukocidins by ectopic expression of different GPCRs. Our screen identifies post-translational modification (PTM) pathways involved in the sulfation and sialylation of the leukocidin-receptors. Subsequent validation experiments show differences in the impact of PTM moieties on leukocidin toxicity, highlighting an additional layer of refinement and divergence in the staphylococcal host-pathogen interface. Leukocidin receptors may serve as targets for anti-staphylococcal interventions and understanding toxin-receptor interactions will facilitate the development of innovative therapeutics. Variations in the genes encoding PTM pathways could provide insight into observed differences in susceptibility of humans to infections with S. aureus.


Asunto(s)
Interacciones Microbiota-Huesped/genética , Leucocidinas/toxicidad , Procesamiento Proteico-Postraduccional , Receptores Acoplados a Proteínas G/metabolismo , Infecciones Estafilocócicas/patología , Staphylococcus aureus/patogenicidad , Sistemas CRISPR-Cas , Técnicas de Cultivo de Célula , Supervivencia Celular/genética , Farmacorresistencia Bacteriana/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Células HEK293 , Humanos , Leucocidinas/genética , Leucocidinas/metabolismo , Fagocitos/microbiología , Fagocitos/patología , Unión Proteica , Receptores Acoplados a Proteínas G/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Células U937
10.
Rev Mal Respir ; 36(3): 359-363, 2019 Mar.
Artículo en Francés | MEDLINE | ID: mdl-30704806
11.
mBio ; 10(1)2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30602580

RESUMEN

Staphylococcus aureus is a human pathogen responsible for high morbidity and mortality worldwide. Recurrent infections with this bacterium are common, suggesting that S. aureus thwarts the development of sterilizing immunity. S. aureus strains that cause disease in humans produce up to five different bicomponent toxins (leukocidins) that target and lyse neutrophils, innate immune cells that represent the first line of defense against S. aureus infections. However, little is known about the role of leukocidins in blunting adaptive immunity. Here, we explored the effects of leukocidins on human dendritic cells (DCs), antigen-presenting cells required for the development of adaptive immunity. Using an ex vivo infection model of primary human monocyte-derived dendritic cells, we found that S. aureus, including strains from different clonal complexes and drug resistance profiles, effectively kills DCs despite efficient phagocytosis. Although all purified leukocidins could kill DCs, infections with live bacteria revealed that S. aureus targets and kills DCs primarily via the activity of leukocidin LukAB. Moreover, using coculture experiments performed with DCs and autologous CD4+ T lymphocytes, we found that LukAB inhibits DC-mediated activation and proliferation of primary human T cells. Taken together, the data determined in the study reveal a novel immunosuppressive strategy of S. aureus whereby the bacterium blunts the development of adaptive immunity via LukAB-mediated injury of DCs.IMPORTANCE Antigen-presenting cells such as dendritic cells (DCs) fulfill an indispensable role in the development of adaptive immunity by producing proinflammatory cytokines and presenting microbial antigens to lymphocytes to trigger a faster, specific, and long-lasting immune response. Here, we studied the effect of Staphylococcus aureus toxins on human DCs. We discovered that the leukocidin LukAB hinders the development of adaptive immunity by targeting human DCs. The ability of S. aureus to blunt the function of DCs could help explain the high frequency of recurrent S. aureus infections. Taken together, the results from this study suggest that therapeutically targeting the S. aureus leukocidins may boost effective innate and adaptive immune responses by protecting innate leukocytes, enabling proper antigen presentation and T cell activation.


Asunto(s)
Proteínas Bacterianas/toxicidad , Células Dendríticas/efectos de los fármacos , Células Dendríticas/fisiología , Evasión Inmune , Leucocidinas/toxicidad , Infecciones Estafilocócicas/patología , Staphylococcus aureus/patogenicidad , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/microbiología , Humanos , Activación de Linfocitos , Modelos Biológicos
12.
BMC Microbiol ; 18(1): 181, 2018 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-30419818

RESUMEN

BACKGROUND: Staphylococcus aureus is a leading cause of Gram-positive bacterial infections worldwide; however, the treatment of S. aureus infection has become increasingly difficult due to the prevalence of methicillin-resistant S. aureus strains, highlighting the urgent need for the development of novel strategies. The complexity of S. aureus pathogenesis relies on virulence factors. Recent studies have demonstrated that leukocidins expressed by the majority of clinical isolates play important roles in the pathogenesis of S. aureus. RESULTS: In this study, we developed three human monoclonal antibodies against all F-components of leukocidins HlgABC, LukSF, and LukED with high affinity. These antibodies were found to be capable of blocking leukocidin-mediated cell lysis in vitro. Furthermore, the antibodies dramatically reduced disease progression and mortality after S. aureus infection in vivo. CONCLUSIONS: Our findings revealed that neutralizing bicomponent leukocidins may be a promising strategy to combat infections caused by S. aureus.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Proteínas Hemolisinas/inmunología , Leucocidinas/inmunología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/mortalidad , Staphylococcus aureus/fisiología , Animales , Anticuerpos Monoclonales/inmunología , Progresión de la Enfermedad , Eritrocitos/efectos de los fármacos , Femenino , Proteínas Hemolisinas/toxicidad , Hemólisis/efectos de los fármacos , Humanos , Leucocidinas/toxicidad , Ratones Endogámicos BALB C , Pruebas de Neutralización , Conejos , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética
13.
Toxins (Basel) ; 10(11)2018 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-30400375

RESUMEN

: Panton-Valentine leukocidin (PVL) retinal intoxication induces glial activation and inflammatory response via the interaction with retinal neurons. In this study, rabbit retinal explant was used as a model to study neuronal and glial consequences of PVL intoxication. Retinal explants were treated with different concentrations of PVL. PVL location and neuronal and glial changes were examined using immunohistochemistry. Some inflammatory factors were quantified using RT-qPCR at 4 and 8 h. These results were compared with those of control explants. PVL co-localized rapidly with retinal ganglion cells and with horizontal cells. PVL induced Müller and microglial cell activation. Retinal structure was altered and some amacrine and microglial cells underwent apoptosis. Glial activation and cell apoptosis increased in a PVL concentration- and time-dependent manner. IL-6 and IL-8 expression increased in PVL-treated explants but less than in control explants, which may indicate that other factors were responsible for glial activation and retinal apoptosis. On retinal explants, PVL co-localized with neuronal cells and induced glial activation together with microglial apoptosis, which confirms previous results observed in in vivo model. Rabbit retinal explant seems to be suitable model to further study the process of PVL leading to glial activation and retinal cells apoptosis.


Asunto(s)
Toxinas Bacterianas/toxicidad , Exotoxinas/toxicidad , Leucocidinas/toxicidad , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Células Cultivadas , Mediadores de Inflamación/metabolismo , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Neuronas/patología , Conejos
15.
Artículo en Inglés | MEDLINE | ID: mdl-28589102

RESUMEN

Some bacterial pathogens modulate signaling pathways of eukaryotic cells in order to subvert the host response for their own benefit, leading to successful colonization and invasion. Pathogenic bacteria produce multiple compounds that generate favorable conditions to their survival and growth during infection in eukaryotic hosts. Many bacterial toxins can alter the cell cycle progression of host cells, impairing essential cellular functions and impeding host cell division. This review summarizes current knowledge regarding cyclomodulins, a heterogeneous family of bacterial effectors that induce eukaryotic cell cycle alterations. We discuss the mechanisms of actions of cyclomodulins according to their biochemical properties, providing examples of various cyclomodulins such as cycle inhibiting factor, γ-glutamyltranspeptidase, cytolethal distending toxins, shiga toxin, subtilase toxin, anthrax toxin, cholera toxin, adenylate cyclase toxins, vacuolating cytotoxin, cytotoxic necrotizing factor, Panton-Valentine leukocidin, phenol soluble modulins, and mycolactone. Special attention is paid to the benefit provided by cyclomodulins to bacteria during colonization of the host.


Asunto(s)
Bacterias/patogenicidad , Fenómenos Fisiológicos Bacterianos , Toxinas Bacterianas/metabolismo , Ciclo Celular/efectos de los fármacos , Células Eucariotas/microbiología , Toxina de Adenilato Ciclasa/toxicidad , Animales , Antígenos Bacterianos/toxicidad , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/toxicidad , Toxina del Cólera/toxicidad , Células Eucariotas/efectos de los fármacos , Exotoxinas/toxicidad , Interacciones Huésped-Parásitos , Humanos , Leucocidinas/toxicidad , Macrólidos/toxicidad , Toxina Shiga/toxicidad , Transducción de Señal , Factores de Virulencia/toxicidad
16.
Artículo en Inglés | MEDLINE | ID: mdl-28596942

RESUMEN

Coagulase-negative staphylococci (CoNS) are important nosocomial pathogens and the leading cause of sepsis. The second most frequently implicated species, after Staphylococcus epidermidis, is Staphylococcus haemolyticus. However, we have a significant lack of knowledge about what causes virulence of S. haemolyticus, as virulence factors of this pathogen have remained virtually unexplored. In contrast to the aggressive pathogen Staphylococcus aureus, toxin production has traditionally not been associated with CoNS. Recent findings have suggested that phenol-soluble modulins (PSMs), amphipathic peptide toxins with broad cytolytic activity, are widespread in staphylococci, but there has been no systematic assessment of PSM production in CoNS other than S. epidermidis. Here, we identified, purified, and characterized PSMs of S. haemolyticus. We found three PSMs of the ß-type, which correspond to peptides that before were described to have anti-gonococcal activity. We also detected an α-type PSM that has not previously been described. Furthermore, we confirmed that S. haemolyticus does not produce a δ-toxin, as results from genome sequencing had indicated. All four S. haemolyticus PSMs had strong pro-inflammatory activity, promoting neutrophil chemotaxis. Notably, we identified in particular the novel α-type PSM, S. haemolyticus PSMα, as a potent hemolysin and leukocidin. For the first time, our study describes toxins of this important staphylococcal pathogen with the potential to have a significant impact on virulence during blood infection and sepsis.


Asunto(s)
Toxinas Bacterianas/toxicidad , Infecciones Estafilocócicas/metabolismo , Staphylococcus haemolyticus/metabolismo , Staphylococcus haemolyticus/patogenicidad , Factores de Virulencia , Virulencia , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/aislamiento & purificación , Proteínas Hemolisinas/toxicidad , Hemólisis , Humanos , Leucocidinas/toxicidad , Neutrófilos/efectos de los fármacos , Sepsis/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus/patogenicidad , Staphylococcus aureus/patogenicidad , Staphylococcus epidermidis/patogenicidad , Staphylococcus haemolyticus/genética , Virulencia/genética , Factores de Virulencia/genética , Factores de Virulencia/aislamiento & purificación , Factores de Virulencia/toxicidad
17.
Emerg Infect Dis ; 22(1): 96-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26690308

RESUMEN

Staphylococcus aureus strains that produce Panton-Valentine leukocidin are known to cause community infections. We describe an outbreak of skin abscesses caused by Panton-Valentine leukocidin-producing methicillin-susceptible S. aureus (clonal complex 121) in a professional rugby team in France during July 2010-February 2011. Eight team members were carriers; 7 had skin abscesses.


Asunto(s)
Toxinas Bacterianas/toxicidad , Exotoxinas/toxicidad , Leucocidinas/toxicidad , Infecciones Cutáneas Estafilocócicas/epidemiología , Adolescente , Adulto , Atletas , Brotes de Enfermedades , Fútbol Americano , Francia/epidemiología , Humanos , Masculino , Meticilina/uso terapéutico , Infecciones Cutáneas Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Adulto Joven
18.
PLoS One ; 10(9): e0137874, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26367030

RESUMEN

S. aureus vaccine development has proven particularly difficult. The conventional approach to achieve sterile immunity through opsonophagocytic killing has been largely unsuccessful. S. aureus is highly toxigenic and a great body of evidence suggests that a successful future vaccine for this organism should target extracellular toxins which are responsible for host tissue destruction and immunosuppression. Major staphylococcal toxins are alpha toxin (a single subunit hemolysin) along with a group of bicomponent pore-forming toxins (BCPFT), namely Panton-Valentine leukocidin (PVL), gamma hemolysins (HlgCB and AB), LukAB and LukED. In our previous report, an attenuated mutant of LukS-PV (PVL- S subunit) named as "LukS-mut9" elicited high immunogenic response as well as provided a significant protection in a mouse sepsis model. Recent discovery of PVL receptors shows that mice lack receptors for this toxin, thus the reported protection of mice with the PVL vaccine may relate to cross protective responses against other homologous toxins. This manuscript addresses this issue by demonstrating that polyclonal antibody generated by LukS-mut9 can neutralize other canonical and non-canonical leukotoxin pairs. In this report, we also demonstrated that several potent toxins can be created by non-canonical pairing of subunits. Out of 5 pairs of canonical and 8 pairs of non-canonical toxins tested, anti-LukS-mut9 polyclonal antibodies neutralized all except for LukAB. We also studied the potential hemolytic activities of canonical and noncanonical pairs among biocomponent toxins and discovered that a novel non-canonical pair consisting of HlgA and LukD is a highly toxic combination. This pair can lyse RBC from different species including human blood far better than alpha hemolysin. Moreover, to follow-up our last report, we explored the correlation between the levels of pre-existing antibodies to new sets of leukotoxins subunits and clinical outcomes in adult patients with S. aureus bacteremia. We found that there is an inversed correlation between the antibody titer to sepsis for leukotoxins LukS-mut9, LukF-PV, HlgC, LukE and LukAB, suggesting the risk of sepsis was significantly lower in the patients with higher antibody titer against those toxins.


Asunto(s)
Proteínas Bacterianas/inmunología , Leucocidinas/inmunología , Vacunas Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Adulto , Animales , Anticuerpos/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/toxicidad , Reacciones Cruzadas , Pruebas Inmunológicas de Citotoxicidad , Exotoxinas/genética , Exotoxinas/inmunología , Exotoxinas/toxicidad , Femenino , Humanos , Leucocidinas/genética , Leucocidinas/toxicidad , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Pruebas de Neutralización , Conejos , Infecciones Estafilocócicas/microbiología , Vacunas Estafilocócicas/farmacología , Staphylococcus aureus/patogenicidad , Vacunas Atenuadas/inmunología
19.
Lett Appl Microbiol ; 61(2): 158-64, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25963798

RESUMEN

UNLABELLED: α-Defensins produced by neutrophils are important effector molecules of the innate immune system. In addition to their microbicidal effects, α-defensins have the ability to neutralize bacterial toxins. Panton-Valentine leukocidin (PVL) is the hallmark of community-acquired methicillin-resistant Staphylococcus aureus. Staphylococcus aureus that produce PVL are responsible for severe diseases, including necrotizing pneumonia. Polymorphonuclear neutrophils (PMNs) are the target cells of PVL action. The goal of this study was to elucidate the effect of a group of α-defensins known as the human neutrophil peptides (HNPs) on the interactions between LukS-PV and LukF-PV, which compose PVL, and human PMNs. We observed that HNPs bound to both subunits of PVL and significantly decreased PVL pore formation in PMNs, with a maximum inhibition of 27%. When various HNP molecules were tested individually under the same conditions, we observed that HNP3, but not HNP1 or 2, decreased pore formation. Similarly, HNP3 significantly decreased PVL-induced PMN lysis, with a maximum inhibition of 31%. Interestingly, HNPs did not affect LukS-PV LukF-PV oligomerization, LukS-PV LukF-PV binding to PMNs or calcium influx induced by PVL in PMNs. Our results suggest that HNP3 partially protects neutrophils against PVL by interfering with the conformational changes of PVL required to form a functional pore. SIGNIFICANCE AND IMPACT OF THE STUDY: Panton-Valentine leukocidin (PVL) is a pore-forming toxin produced by Staphylococcus aureus, responsible for neutrophil damage and key player of severe staphylococcal diseases. Antimicrobial peptides produced by neutrophils (HNP1-3) neutralize several other bacterial cytotoxins. We examined the impact of human neutrophil peptides (HNPs) on PVL cytotoxicity against human neutrophils and we found that HNPs bind to both LukS and LukF components of PVL, thereby inhibiting pore formation and neutrophil lysis. Our results suggest that HNP3 may impair PVL conformational changes required to form a functional pore and provide insight into the pathogenesis of PVL-related staphylococcal infection, with potential impact on the disease outcome.


Asunto(s)
Toxinas Bacterianas/toxicidad , Exotoxinas/toxicidad , Leucocidinas/toxicidad , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Neutrófilos/enzimología , Infecciones Estafilocócicas/inmunología , alfa-Defensinas/metabolismo , Proteínas Bacterianas/metabolismo , Humanos , Leucocidinas/metabolismo , Neutrófilos/inmunología
20.
Ned Tijdschr Geneeskd ; 158: A7540, 2014.
Artículo en Holandés | MEDLINE | ID: mdl-25269632

RESUMEN

A twelve-year-old girl was admitted to the paediatric department with progressive joint problems following a mild trauma. Diagnostic follow-up showed a Staphylococcus aureus infection. Generally, S. aureus infection mainly involves the skin and the nasal mucosa. In some cases it can cause a fulminant invasive infection in previously healthy young patients. If this is the case, it is important to consider PVL (Panton-Valentine-leukocidin)-positive S. aureus and underlying immune deficiencies when performing a differential diagnosis. In addition, it is important to remember that previous trauma can cause increased susceptibility to septic arthritis or osteomyelitis in children, as is seen in this case. Aggressive treatment of an invasive S. aureus infection is important even in apparently healthy young patients, to prevent morbidity or mortality.


Asunto(s)
Antibacterianos/uso terapéutico , Artritis Infecciosa/diagnóstico , Infecciones Estafilocócicas/diagnóstico , Staphylococcus aureus/efectos de los fármacos , Artritis Infecciosa/tratamiento farmacológico , Toxinas Bacterianas/biosíntesis , Toxinas Bacterianas/toxicidad , Niño , Diagnóstico Diferencial , Exotoxinas/biosíntesis , Exotoxinas/toxicidad , Femenino , Humanos , Leucocidinas/biosíntesis , Leucocidinas/toxicidad , Osteomielitis/diagnóstico , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA