Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Infect Immun ; 92(6): e0014124, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38722166

RESUMEN

The human-specific bacterial pathogen group A Streptococcus (GAS) is a significant cause of morbidity and mortality. Macrophages are important to control GAS infection, but previous data indicate that GAS can persist in macrophages. In this study, we detail the molecular mechanisms by which GAS survives in THP-1 macrophages. Our fluorescence microscopy studies demonstrate that GAS is readily phagocytosed by macrophages, but persists within phagolysosomes. These phagolysosomes are not acidified, which is in agreement with our findings that GAS cannot survive in low pH environments. We find that the secreted pore-forming toxin Streptolysin O (SLO) perforates the phagolysosomal membrane, allowing leakage of not only protons but also large proteins including the lysosomal protease cathepsin B. Additionally, GAS recruits CD63/LAMP-3, which may contribute to lysosomal permeabilization, especially in the absence of SLO. Thus, although GAS does not inhibit fusion of the lysosome with the phagosome, it has multiple mechanisms to prevent proper phagolysosome function, allowing for persistence of the bacteria within the macrophage. This has important implications for not only the initial response but also the overall functionality of the macrophages, which may lead to the resulting pathologies in GAS infection. Our data suggest that therapies aimed at improving macrophage function may positively impact patient outcomes in GAS infection.


Asunto(s)
Proteínas Bacterianas , Lisosomas , Macrófagos , Streptococcus pyogenes , Estreptolisinas , Streptococcus pyogenes/inmunología , Humanos , Macrófagos/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Lisosomas/metabolismo , Lisosomas/microbiología , Estreptolisinas/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Fagosomas/microbiología , Fagosomas/metabolismo , Células THP-1 , Fagocitosis , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/metabolismo , Catepsina B/metabolismo , Concentración de Iones de Hidrógeno
2.
Infect Immun ; 91(11): e0025823, 2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-37874162

RESUMEN

The pro-inflammatory cytokine IL-6 regulates antimicrobial responses that are broadly crucial in the defense against infection. Our prior work shows that IL-6 promotes the killing of the M4 serotype group A Streptococcus (GAS) but does not impact the globally disseminated M1T1 serotype associated with invasive infections. Using in vitro and in vivo infection models, we show that IL-6 induces phagocyte reactive oxygen species (ROS) that are responsible for the differential susceptibility of M4 and M1T1 GAS to IL-6-mediated defenses. Clinical isolates naturally deficient in capsule, or M1T1 strains deficient in capsule production, are sensitive to this ROS killing. The GAS capsule is made of hyaluronic acid, an antioxidant that detoxifies ROS and can protect acapsular M4 GAS when added exogenously. During in vitro interactions with macrophages and neutrophils, acapsular GAS can also be rescued with the antioxidant N-acetylcysteine, suggesting this is a major virulence contribution of the capsule. In an intradermal infection model with gp91phox -/- (chronic granulomatous disease [CGD]) mice, phagocyte ROS production had a modest effect on bacterial proliferation and the cytokine response but significantly limited the size of the bacterial lesion in the skin. These data suggest that the capsule broadly provides enhanced resistance to phagocyte ROS but is not essential for invasive infection. Since capsule-deficient strains are observed across several GAS serotypes and are competent for transmission and both mild and invasive infections, additional host or microbe factors may contribute to ROS detoxification during GAS infections.


Asunto(s)
Ácido Hialurónico , Infecciones Estreptocócicas , Animales , Ratones , Especies Reactivas de Oxígeno , Antioxidantes , Interleucina-6 , Neutrófilos/microbiología , Streptococcus pyogenes , Infecciones Estreptocócicas/microbiología , Proteínas Bacterianas
4.
Annu Rev Microbiol ; 68: 439-58, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25002085

RESUMEN

The development of a severe invasive bacterial infection in an otherwise healthy individual is one of the most striking and fascinating aspects of human medicine. A small cadre of gram-positive pathogens of the genera Streptococcus and Staphylococcus stand out for their unique invasive disease potential and sophisticated ability to counteract the multifaceted components of human innate defense. This review illustrates how these leading human disease agents evade host complement deposition and activation, impede phagocyte recruitment and activation, resist the microbicidal activities of host antimicrobial peptides and reactive oxygen species, escape neutrophil extracellular traps, and promote and accelerate phagocyte cell death through the action of pore-forming cytolysins. Understanding the molecular basis of bacterial innate immune resistance can open new avenues for therapeutic intervention geared to disabling specific virulence factors and resensitizing the pathogen to host innate immune clearance.


Asunto(s)
Bacterias Grampositivas/inmunología , Infecciones por Bacterias Grampositivas/inmunología , Infecciones por Bacterias Grampositivas/microbiología , Inmunidad Innata , Animales , Bacterias Grampositivas/genética , Bacterias Grampositivas/patogenicidad , Humanos , Evasión Inmune , Virulencia
5.
PLoS Genet ; 8(10): e1003007, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23093945

RESUMEN

Autophagy is the mechanism by which cytoplasmic components and organelles are degraded by the lysosomal machinery in response to diverse stimuli including nutrient deprivation, intracellular pathogens, and multiple forms of cellular stress. Here, we show that the membrane-associated E3 ligase RNF5 regulates basal levels of autophagy by controlling the stability of a select pool of the cysteine protease ATG4B. RNF5 controls the membranal fraction of ATG4B and limits LC3 (ATG8) processing, which is required for phagophore and autophagosome formation. The association of ATG4B with-and regulation of its ubiquitination and stability by-RNF5 is seen primarily under normal growth conditions. Processing of LC3 forms, appearance of LC3-positive puncta, and p62 expression are higher in RNF5(-/-) MEF. RNF5 mutant, which retains its E3 ligase activity but does not associate with ATG4B, no longer affects LC3 puncta. Further, increased puncta seen in RNF5(-/-) using WT but not LC3 mutant, which bypasses ATG4B processing, substantiates the role of RNF5 in early phases of LC3 processing and autophagy. Similarly, RNF-5 inactivation in Caenorhabditis elegans increases the level of LGG-1/LC3::GFP puncta. RNF5(-/-) mice are more resistant to group A Streptococcus infection, associated with increased autophagosomes and more efficient bacterial clearance by RNF5(-/-) macrophages. Collectively, the RNF5-mediated control of membranalATG4B reveals a novel layer in the regulation of LC3 processing and autophagy.


Asunto(s)
Autofagia , Infecciones Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Proteínas de la Membrana/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Infecciones Bacterianas/genética , Infecciones Bacterianas/mortalidad , Caenorhabditis elegans/metabolismo , Línea Celular , Membrana Celular/metabolismo , Estabilidad de Enzimas , Predisposición Genética a la Enfermedad , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Fagosomas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Transporte de Proteínas , Proteolisis , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
6.
Infect Immun ; 82(10): 4011-20, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25024366

RESUMEN

Streptococcal collagen-like protein 1 (Scl-1) is one of the most highly expressed proteins in the invasive M1T1 serotype group A Streptococcus (GAS), a globally disseminated clone associated with higher risk of severe invasive infections. Previous studies using recombinant Scl-1 protein suggested a role in cell attachment and binding and inhibition of serum proteins. Here, we studied the contribution of Scl-1 to the virulence of the M1T1 clone in the physiological context of the live bacterium by generating an isogenic strain lacking the scl-1 gene. Upon subcutaneous infection in mice, wild-type bacteria induced larger lesions than the Δscl mutant. However, loss of Scl-1 did not alter bacterial adherence to or invasion of skin keratinocytes. We found instead that Scl-1 plays a critical role in GAS resistance to human and murine phagocytic cells, allowing the bacteria to persist at the site of infection. Phenotypic analyses demonstrated that Scl-1 mediates bacterial survival in neutrophil extracellular traps (NETs) and protects GAS from antimicrobial peptides found within the NETs. Additionally, Scl-1 interferes with myeloperoxidase (MPO) release, a prerequisite for NET production, thereby suppressing NET formation. We conclude that Scl-1 is a virulence determinant in the M1T1 GAS clone, allowing GAS to subvert innate immune functions that are critical in clearing bacterial infections.


Asunto(s)
Evasión Inmune , Neutrófilos/inmunología , Neutrófilos/microbiología , Streptococcus pyogenes/inmunología , Streptococcus pyogenes/metabolismo , Factores de Virulencia/metabolismo , Animales , Adhesión Bacteriana , Células Cultivadas , Eliminación de Gen , Humanos , Queratinocitos/microbiología , Ratones , Ratones Endogámicos C57BL , Viabilidad Microbiana , Fagocitos/inmunología , Fagocitos/microbiología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/genética , Factores de Virulencia/genética
7.
BMC Microbiol ; 11: 120, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21619648

RESUMEN

BACKGROUND: The secreted enzyme EndoS, an endoglycosidase from Streptococcus pyogenes, hydrolyzes the N-linked glycan of the constant region of immunoglobulin G (IgG) heavy chain and renders the antibody unable to interact with Fc receptors and elicit effector functions. In this study we couple targeted allelic replacement mutagenesis and heterologous expression to elucidate the contribution of EndoS to group A Streptococcus (GAS) phagocyte resistance and pathogenicity in vitro and in vivo. RESULTS: Knocking out the EndoS gene in GAS M1T1 background revealed no significant differences in bacterial survival in immune cell killing assays or in a systemic mouse model of infection. However, exogenous addition and heterologous expression of EndoS was found to increase GAS resistance to killing by neutrophils and monocytes in vitro. Additionally, heterologous expression of EndoS in M49 GAS increased mouse virulence in vivo. CONCLUSIONS: We conclude that in a highly virulent M1T1 background, EndoS has no significant impact on GAS phagocyte resistance and pathogenicity. However, local accumulation or high levels of expression of EndoS in certain GAS strains may contribute to virulence.


Asunto(s)
Glicósido Hidrolasas/metabolismo , Inmunoglobulina G/metabolismo , Fagocitos/inmunología , Fagocitos/microbiología , Streptococcus pyogenes/enzimología , Streptococcus pyogenes/patogenicidad , Animales , Modelos Animales de Enfermedad , Eliminación de Gen , Glicósido Hidrolasas/genética , Humanos , Ratones , Viabilidad Microbiana , Fagocitosis , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/inmunología , Virulencia
8.
Cell Microbiol ; 10(10): 2078-90, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18637021

RESUMEN

The extracellular protozoan parasite Trichomonas vaginalis causes the most prevalent non-viral sexually transmitted human infection, yet the pathogenesis of infection is poorly understood, and host cell receptors have not been described. The surface of T. vaginalis is covered with a glycoconjugate called lipophosphoglycan (LPG), which plays a role in the adherence and cytotoxicity of parasites to human cells. T. vaginalis LPG contains high amounts of galactose, making this polysaccharide a candidate for recognition by the galactose-binding galectin family of lectins. Here we show that galectin-1 (gal-1) is expressed by human cervical epithelial cells and binds T. vaginalis LPG. Gal-1 binds to parasites in a carbohydrate-dependent manner that is inhibited in the presence of T. vaginalis LPG. Addition of purified gal-1 to cervical epithelial cells also enhances parasite binding, while a decrease in gal-1 expression by small interfering RNA (siRNA) transfection decreases parasite binding. In contrast, the related galectin-7 (gal-7) does not bind T. vaginalis in a carbohydrate-dependent manner, and is unable to mediate attachment of parasites to host cells. Our data are consistent with the presence of multiple host cell receptors for T. vaginalis of which gal-1 is the first to be identified and highlight the importance of glycoconjugates in host-pathogen interactions.


Asunto(s)
Células Epiteliales/parasitología , Galectina 1/metabolismo , Glicoesfingolípidos/metabolismo , Receptores de Superficie Celular/metabolismo , Trichomonas vaginalis/fisiología , Animales , Adhesión Celular , Línea Celular , Cuello del Útero/parasitología , Femenino , Silenciador del Gen , Humanos
9.
Front Immunol ; 9: 327, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29535718

RESUMEN

The resurgence of Group A Streptococcus (GAS) infections in the past two decades has been a rising major public health concern. Due to a large number of GAS infections occurring in the skin, mast cells (MCs), innate immune cells known to localize to the dermis, could play an important role in controlling infection. MCs can exert their antimicrobial activities either early during infection, by degranulation and release of antimicrobial proteases and the cathelicidin-derived antimicrobial peptide LL-37, or by forming antibacterial MC extracellular traps (MCETs) in later stages of infection. We demonstrate that MCs do not directly degranulate in response to GAS, reducing their ability to control bacterial growth in early stages of infection. However, MC granule components are highly cytotoxic to GAS due to the pore-forming activity of LL-37, while MC granule proteases do not significantly affect GAS viability. We therefore confirmed the importance of MCETs by demonstrating their capacity to reduce GAS survival. The data therefore suggests that LL-37 from MC granules become embedded in MCETs, and are the primary effector molecule by which MCs control GAS infection. Our work underscores the importance of a non-traditional immune effector cell, utilizing a non-conventional mechanism, in the defense against an important human pathogen.


Asunto(s)
Degranulación de la Célula/inmunología , Trampas Extracelulares/inmunología , Mastocitos/inmunología , Enfermedades Cutáneas Bacterianas/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Péptidos Catiónicos Antimicrobianos/inmunología , Línea Celular , Trampas Extracelulares/microbiología , Humanos , Mastocitos/microbiología , Mastocitos/patología , Vesículas Secretoras/inmunología , Vesículas Secretoras/microbiología , Vesículas Secretoras/patología , Enfermedades Cutáneas Bacterianas/microbiología , Enfermedades Cutáneas Bacterianas/patología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Catelicidinas
10.
Clin Ther ; 36(10): 1317-33, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25017183

RESUMEN

PURPOSE: Guidelines recommend daptomycin combination therapy as an option for methicillin-resistant Staphylococcus aureus (MRSA) bacteremia after vancomycin failure. Recent data suggest that combining daptomycin with a ß-lactam may have unique benefits; however, there are very limited clinical data regarding the use of ceftaroline with daptomycin. METHODS: All 26 cases from the 10 medical centers in which ceftaroline plus daptomycin was used for treatment of documented refractory staphylococcal bacteremia from March 2011 to November 2012 were included. In vitro (synergy studies, binding assays, cathelicidin LL-37 killing assays), and in vivo (virulence assays using a murine subcutaneous infection model) studies examining the effects of ceftaroline with daptomycin were also performed. FINDINGS: Daptomycin plus ceftaroline was used in 26 cases of staphylococcal bacteremia (20 MRSA, 2 vancomycin-intermediate S aureus, 2 methicillin-susceptible S aureus [MSSA], 2 methicillin-resistant S epidermidis). Bacteremia persisted for a median of 10 days (range, 3-23 days) on previous antimicrobial therapy. After daptomycin plus ceftaroline was started, the median time to bacteremia clearance was 2 days (range, 1-6 days). In vitro studies showed ceftaroline synergy against MRSA and enhanced MRSA killing by cathelicidin LL-37 and neutrophils. Ceftaroline also induced daptomycin binding in MSSA and MRSA to a comparable degree as nafcillin. MRSA grown in subinhibitory concentrations of ceftaroline showed attenuated virulence in a murine subcutaneous infection model. IMPLICATIONS: Ceftaroline plus daptomycin may be an option to hasten clearance of refractory staphylococcal bacteremia. Ceftaroline offers dual benefit via synergy with both daptomycin and sensitization to innate host defense peptide cathelicidin LL37, which could attenuate virulence of the pathogen.


Asunto(s)
Antibacterianos/uso terapéutico , Bacteriemia/tratamiento farmacológico , Cefalosporinas/uso terapéutico , Daptomicina/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/metabolismo , Cefalosporinas/farmacología , Daptomicina/farmacología , Quimioterapia Combinada , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Terapia Recuperativa , Staphylococcus/efectos de los fármacos , Staphylococcus/crecimiento & desarrollo , Catelicidinas , Ceftarolina
11.
mBio ; 4(4)2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23900173

RESUMEN

UNLABELLED: The Mac/IdeS protein of group A Streptococcus (GAS) is a secreted cysteine protease with cleavage specificity for IgG and is highly expressed in the GAS serotype M1T1 clone, which is the serotype most frequently isolated from patients with life-threatening invasive infections. While studies of Mac/IdeS with recombinant protein have shown that the protein can potentially prevent opsonophagocytosis of GAS by neutrophils, the role of the protein in immune evasion as physiologically produced by the living organism has not been studied. Here we examined the contribution of Mac/IdeS to invasive GAS disease by generating a mutant lacking Mac/IdeS in the hyperinvasive M1T1 background. While Mac/IdeS was highly expressed and proteolytically active in the hyperinvasive strain, elimination of the bacterial protease did not significantly influence GAS phagocytic uptake, oxidative-burst induction, cathelicidin sensitivity, resistance to neutrophil or macrophage killing, or pathogenicity in pre- or postimmune mouse infectious challenges. We conclude that in the highly virulent M1T1 background, Mac/IdeS is not essential for either phagocyte resistance or virulence. Given the conservation of Mac/IdeS and homologues across GAS strains, it is possible that Mac/IdeS serves another important function in GAS ecology or contributes to virulence in other strain backgrounds. IMPORTANCE: Group A Streptococcus (GAS) causes human infections ranging from strep throat to life-threatening conditions such as flesh-eating disease and toxic shock syndrome. Common disease-associated clones of GAS can cause both mild and severe infections because of a characteristic mutation and subsequent change in the expression of several genes that develops under host immune selection. One of these genes encodes Mac/IdeS, a protease that has been shown to cleave antibodies important to the immune defense system. In this study, we found that while Mac/IdeS is highly expressed in hypervirulent GAS, it does not significantly contribute to the ability of the bacteria to survive white blood cell killing or produce invasive infection in the mouse. These data underscore the importance of correlating studies on virulence factor function with physiologic expression levels and the complexity of streptococcal pathogenesis and contribute to our overall understanding of how GAS causes disease.


Asunto(s)
Proteínas Bacterianas/metabolismo , Inmunoglobulina G/metabolismo , Fagocitos/microbiología , Streptococcus pyogenes/inmunología , Streptococcus pyogenes/patogenicidad , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Línea Celular , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Eliminación de Gen , Humanos , Masculino , Ratones , Viabilidad Microbiana , Fagocitosis , Proteolisis , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/genética , Virulencia , Factores de Virulencia/genética
12.
J Mol Med (Berl) ; 90(9): 1079-89, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22371073

RESUMEN

Hypoxia inducible factor-1 (HIF-1) is a transcription factor that is a major regulator of energy homeostasis and cellular adaptation to low oxygen stress. HIF-1 is also activated in response to bacterial pathogens and supports the innate immune response of both phagocytes and keratinocytes. In this work, we show that a new pharmacological compound AKB-4924 increases HIF-1 levels and enhances the antibacterial activity of phagocytes and keratinocytes against both methicillin-sensitive and methicillin-resistant strains of Staphylococcus aureus in vitro. AKB-4924 is also effective in stimulating the killing capacity of keratinocytes against the important opportunistic skin pathogens Pseudomonas aeruginosa and Acinetobacter baumanii. The effect of AKB-4924 is mediated through the activity of host cells, as the compound exerts no direct antimicrobial activity. Administered locally as a single agent, AKB-4924 limits S. aureus proliferation and lesion formation in a mouse skin abscess model. This approach to pharmacologically boost the innate immune response via HIF-1 stabilization may serve as a useful adjunctive treatment for antibiotic-resistant bacterial infections.


Asunto(s)
Antibacterianos/uso terapéutico , Factor 1 Inducible por Hipoxia/inmunología , Inmunidad Innata/efectos de los fármacos , Piperazinas/uso terapéutico , Piridonas/uso terapéutico , Enfermedades Cutáneas Bacterianas/prevención & control , Piel/microbiología , Animales , Línea Celular , Femenino , Humanos , Factor 1 Inducible por Hipoxia/agonistas , Queratinocitos/efectos de los fármacos , Queratinocitos/inmunología , Queratinocitos/microbiología , Ratones , Fagocitos/efectos de los fármacos , Fagocitos/inmunología , Fagocitos/microbiología , Piperazinas/farmacología , Piridonas/farmacología , Piel/efectos de los fármacos , Piel/inmunología , Enfermedades Cutáneas Bacterianas/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA