Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
PLoS Pathog ; 13(9): e1006603, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28945820

RESUMEN

Various bacterial toxins circumvent host defenses through overproduction of cAMP. In a previous study, we showed that edema factor (EF), an adenylate cyclase from Bacillus anthracis, disrupts endocytic recycling mediated by the small GTPase Rab11. As a result, cargo proteins such as cadherins fail to reach inter-cellular junctions. In the present study, we provide further mechanistic dissection of Rab11 inhibition by EF using a combination of Drosophila and mammalian systems. EF blocks Rab11 trafficking after the GTP-loading step, preventing a constitutively active form of Rab11 from delivering cargo vesicles to the plasma membrane. Both of the primary cAMP effector pathways -PKA and Epac/Rap1- contribute to inhibition of Rab11-mediated trafficking, but act at distinct steps of the delivery process. PKA acts early, preventing Rab11 from associating with its effectors Rip11 and Sec15. In contrast, Epac functions subsequently via the small GTPase Rap1 to block fusion of recycling endosomes with the plasma membrane, and appears to be the primary effector of EF toxicity in this process. Similarly, experiments conducted in mammalian systems reveal that Epac, but not PKA, mediates the activity of EF both in cell culture and in vivo. The small GTPase Arf6, which initiates endocytic retrieval of cell adhesion components, also contributes to junctional homeostasis by counteracting Rab11-dependent delivery of cargo proteins at sites of cell-cell contact. These studies have potentially significant practical implications, since chemical inhibition of either Arf6 or Epac blocks the effect of EF in cell culture and in vivo, opening new potential therapeutic avenues for treating symptoms caused by cAMP-inducing toxins or related barrier-disrupting pathologies.


Asunto(s)
Antígenos Bacterianos/farmacología , Toxinas Bacterianas/farmacología , Edema/metabolismo , Endosomas/efectos de los fármacos , Uniones Intercelulares/efectos de los fármacos , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/metabolismo , Adenilil Ciclasas/metabolismo , Animales , Cadherinas/metabolismo , Línea Celular , Endosomas/metabolismo , Uniones Intercelulares/metabolismo , Transporte de Proteínas/efectos de los fármacos , Proteínas de Unión al GTP rab/metabolismo
2.
J Infect Dis ; 217(2): 270-279, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29099935

RESUMEN

Background: Necrotizing fasciitis (NF) retains a very high mortality rate despite prompt and adequate antibiotic treatment and surgical debridement. Necrotizing fasciitis has recently been associated with Streptococcus dysgalactiae subspecies equisimilis (SDSE). Methods: We investigated the causes of a very severe clinical manifestation of SDSE-NF by assessing both host and pathogen factors. Results: We found a lack of streptokinase-function blocking antibodies in the patient resulting in increased streptokinase-mediated fibrinolysis and bacterial spread. At the same time, the clinical SDSE isolate produced very high levels of streptokinase. Exogenous immunoglobulin Gs (ex-IgGs) efficiently blocked streptokinase-mediated fibrinolysis in vitro, indicating a protective role against the action of streptokinase. In vivo, SDSE infection severity was also attenuated by ex-IgGs in a NF mouse model. Conclusions: These findings illustrate for the first time that the lack of specific antibodies against streptococcal virulence factors, such as streptokinase, may contribute to NF disease severity. This can be counteracted by ex-IgGs.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Fascitis Necrotizante/patología , Infecciones Estreptocócicas/patología , Streptococcus/patogenicidad , Estreptoquinasa/antagonistas & inhibidores , Factores de Virulencia/antagonistas & inhibidores , Adulto , Animales , Fascitis Necrotizante/microbiología , Femenino , Fibrinolíticos/inmunología , Fibrinolíticos/metabolismo , Interacciones Huésped-Patógeno , Humanos , Ratones Endogámicos C57BL , Infecciones Estreptocócicas/microbiología , Streptococcus/inmunología , Estreptoquinasa/inmunología , Factores de Virulencia/inmunología
3.
J Biol Chem ; 291(27): 13964-13973, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27226531

RESUMEN

Emerging antibiotic resistance among pathogenic bacteria is an issue of great clinical importance, and new approaches to therapy are urgently needed. Anacardic acid, the primary active component of cashew nut shell extract, is a natural product used in the treatment of a variety of medical conditions, including infectious abscesses. Here, we investigate the effects of this natural product on the function of human neutrophils. We find that anacardic acid stimulates the production of reactive oxygen species and neutrophil extracellular traps, two mechanisms utilized by neutrophils to kill invading bacteria. Molecular modeling and pharmacological inhibitor studies suggest anacardic acid stimulation of neutrophils occurs in a PI3K-dependent manner through activation of surface-expressed G protein-coupled sphingosine-1-phosphate receptors. Neutrophil extracellular traps produced in response to anacardic acid are bactericidal and complement select direct antimicrobial activities of the compound.


Asunto(s)
Ácidos Anacárdicos/farmacología , Anacardium/química , Antibacterianos/farmacología , Trampas Extracelulares/metabolismo , Neutrófilos/efectos de los fármacos , Humanos , Lisofosfolípidos/metabolismo , Estallido Respiratorio , Esfingosina/análogos & derivados , Esfingosina/metabolismo
4.
FASEB J ; 29(5): 1859-68, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25630970

RESUMEN

Bicuspid aortic valves calcify at a significantly higher rate than normal aortic valves, a process that involves increased inflammation. Because we have previously found that bicuspid aortic valve experience greater stretch, we investigated the potential connection between stretch and inflammation in human aortic valve interstitial cells (AVICs). Microarray, quantitative PCR (qPCR), and protein assays performed on AVICs exposed to cyclic stretch showed that stretch was sufficient to increase expression of interleukin and metalloproteinase family members by more than 1.5-fold. Conditioned medium from stretched AVICs was sufficient to activate leukocytes. microRNA sequencing and qPCR experiments demonstrated that miR-148a-3p was repressed in both stretched AVICs (43% repression) and, as a clinical correlate, human bicuspid aortic valves (63% reduction). miR-148a-3p was found to be a novel repressor of IKBKB based on data from qPCR, luciferase, and Western blot experiments. Furthermore, increasing miR-148a-3p levels in AVICs was sufficient to decrease NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) signaling and NF-κB target gene expression. Our data demonstrate that stretch-mediated activation of inflammatory pathways is at least partly the result of stretch-repression of miR-148a-3p and a consequent failure to repress IKBKB. To our knowledge, we are the first to report that cyclic stretch of human AVICs activates inflammatory genes in a tissue-autonomous manner via a microRNA that regulates a central inflammatory pathway.


Asunto(s)
Válvula Aórtica/anomalías , Biomarcadores/metabolismo , Enfermedades de las Válvulas Cardíacas/metabolismo , Quinasa I-kappa B/metabolismo , Inflamación/genética , MicroARNs/genética , FN-kappa B/metabolismo , Válvula Aórtica/inmunología , Válvula Aórtica/metabolismo , Enfermedad de la Válvula Aórtica Bicúspide , Western Blotting , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Enfermedades de las Válvulas Cardíacas/inmunología , Humanos , Quinasa I-kappa B/genética , Inflamación/inmunología , Inflamación/patología , Monocitos/citología , Monocitos/metabolismo , FN-kappa B/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Fisiológico
5.
PLoS Pathog ; 9(7): e1003469, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23853591

RESUMEN

Recruitment of the serine protease plasmin is central to the pathogenesis of many bacterial species, including Group A streptococcus (GAS), a leading cause of morbidity and mortality globally. A key process in invasive GAS disease is the ability to accumulate plasmin at the cell surface, however the role of host activators of plasminogen in this process is poorly understood. Here, we demonstrate for the first time that the urokinase-type plasminogen activator (uPA) contributes to plasmin recruitment and subsequent invasive disease initiation in vivo. In the absence of a source of host plasminogen activators, streptokinase (Ska) was required to facilitate cell surface plasmin acquisition by GAS. However, in the absence of Ska, host activators were sufficient to promote cell surface plasmin acquisition by GAS strain 5448 during incubation with plasminogen or human plasma. Furthermore, GAS were able mediate a significant increase in the activation of zymogen pro-uPA in human plasma. In order to assess the contribution of uPA to invasive GAS disease, a previously undescribed transgenic mouse model of infection was employed. Both C57/black 6J, and AlbPLG1 mice expressing the human plasminogen transgene, were significantly more susceptible to invasive GAS disease than uPA-/- mice. The observed decrease in virulence in uPA-/-mice was found to correlate directly with a decrease in bacterial dissemination and reduced cell surface plasmin accumulation by GAS. These findings have significant implications for our understanding of GAS pathogenesis, and research aimed at therapeutic targeting of plasminogen activation in invasive bacterial infections.


Asunto(s)
Resistencia a la Enfermedad , Plasminógeno/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Cruzamientos Genéticos , Susceptibilidad a Enfermedades , Precursores Enzimáticos/sangre , Precursores Enzimáticos/metabolismo , Fibrinolisina/metabolismo , Heterocigoto , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Plasminógeno/genética , Proteolisis , Infecciones Estreptocócicas/sangre , Infecciones Estreptocócicas/metabolismo , Streptococcus pyogenes/patogenicidad , Estreptoquinasa/metabolismo , Propiedades de Superficie , Activador de Plasminógeno de Tipo Uroquinasa/sangre , Activador de Plasminógeno de Tipo Uroquinasa/genética , Virulencia
6.
Infect Immun ; 82(3): 1132-40, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24366250

RESUMEN

Bacillus anthracis, the causative agent of anthrax, relies on multiple virulence factors to subvert the host immune defense. Using Caenorhabditis elegans as an infection model, we screened approximately 5,000 transposon mutants of B. anthracis Sterne for decreased virulence. One of the attenuated mutants resulted in loss of expression of yceG and yceH, the last two genes in a six-gene cluster of tellurite resistance genes. We generated an analogous insertional mutant to confirm the phenotype and characterize the role of yceGH in resistance to host defenses. Loss of yceGH rendered the mutants more sensitive to tellurite toxicity as well as to host defenses such as reactive oxygen species and the cathelicidin family of antimicrobial peptides. Additionally, we see decreased survival in mammalian models of infection, including human whole blood and in mice. We identify a novel role for the yceGH genes in B. anthracis Sterne virulence and suggest that C. elegans is a useful infection model to study anthrax pathogenesis.


Asunto(s)
Carbunco/inmunología , Bacillus anthracis/genética , Bacillus anthracis/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Telurio/inmunología , Animales , Carbunco/microbiología , Caenorhabditis elegans/inmunología , Caenorhabditis elegans/microbiología , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Mutación/genética , Mutación/inmunología , Virulencia/genética , Virulencia/inmunología , Factores de Virulencia/genética , Factores de Virulencia/inmunología
7.
FASEB J ; 27(7): 2633-43, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23531597

RESUMEN

In Western countries, invasive infections caused by M1T1 serotype group A Streptococcus (GAS) are epidemiologically linked to mutations in the control of virulence regulatory 2-component operon (covRS). In indigenous communities and developing countries, severe GAS disease is associated with genetically diverse non-M1T1 GAS serotypes. Hypervirulent M1T1 covRS mutant strains arise through selection by human polymorphonuclear cells for increased expression of GAS virulence factors such as the DNase Sda1, which promotes neutrophil resistance. The GAS bacteremia isolate NS88.2 (emm 98.1) is a covS mutant that exhibits a hypervirulent phenotype and neutrophil resistance yet lacks the phage-encoded Sda1. Here, we have employed a comprehensive systems biology (genomic, transcriptomic, and proteomic) approach to identify NS88.2 virulence determinants that enhance neutrophil resistance in the non-M1T1 GAS genetic background. Using this approach, we have identified streptococcal collagen-like protein A and general stress protein 24 proteins as NS88.2 determinants that contribute to survival in whole blood and neutrophil resistance in non-M1T1 GAS. This study has revealed new factors that contribute to GAS pathogenicity that may play important roles in resisting innate immune defenses and the development of human invasive infections.


Asunto(s)
Proteínas Bacterianas/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Animales , Adhesión Bacteriana/genética , Adhesión Bacteriana/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Electroforesis en Gel Bidimensional , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano/genética , Genómica/métodos , Interacciones Huésped-Patógeno/inmunología , Humanos , Ratones , Viabilidad Microbiana/genética , Viabilidad Microbiana/inmunología , Mutación , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteoma/genética , Proteoma/metabolismo , Proteómica/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Infecciones Estreptocócicas/sangre , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Virulencia/genética , Virulencia/inmunología
8.
Nat Med ; 13(8): 981-5, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17632528

RESUMEN

Most invasive bacterial infections are caused by species that more commonly colonize the human host with minimal symptoms. Although phenotypic or genetic correlates underlying a bacterium's shift to enhanced virulence have been studied, the in vivo selection pressures governing such shifts are poorly understood. The globally disseminated M1T1 clone of group A Streptococcus (GAS) is linked with the rare but life-threatening syndromes of necrotizing fasciitis and toxic shock syndrome. Mutations in the GAS control of virulence regulatory sensor kinase (covRS) operon are associated with severe invasive disease, abolishing expression of a broad-spectrum cysteine protease (SpeB) and allowing the recruitment and activation of host plasminogen on the bacterial surface. Here we describe how bacteriophage-encoded GAS DNase (Sda1), which facilitates the pathogen's escape from neutrophil extracellular traps, serves as a selective force for covRS mutation. The results provide a paradigm whereby natural selection exerted by the innate immune system generates hypervirulent bacterial variants with increased risk of systemic dissemination.


Asunto(s)
Desoxirribonucleasa I/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/enzimología , Streptococcus pyogenes/patogenicidad , Animales , Supervivencia Celular , Desoxirribonucleasa I/genética , Humanos , Inmunidad Innata , Ratones , Neutrófilos/citología , Neutrófilos/microbiología , Fenotipo , Selección Genética , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/genética , Virulencia
9.
Biochem J ; 455(1): 107-18, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23865566

RESUMEN

Many bacteria have evolved ways to interact with glycosylation functions of the immune system of their hosts. Streptococcus pyogenes [GAS (group A Streptococcus)] secretes the enzyme EndoS that cleaves glycans on human IgG and impairs the effector functions of the antibody. The ndoS gene, encoding EndoS, has, until now, been thought to be conserved throughout the serotypes. However, in the present study, we identify EndoS2, an endoglycosidase in serotype M49 GAS strains. We characterized EndoS2 and the corresponding ndoS2 gene using sequencing, bioinformatics, phylogenetic analysis, recombinant expression and LC-MS analysis of glycosidic activity. This revealed that EndoS2 is present exclusively, and highly conserved, in serotype M49 of GAS and is only 37% identical with EndoS. EndoS2 showed endo-ß-N-acetylglucosaminidase activity on all N-linked glycans of IgG and on biantennary and sialylated glycans of AGP (α1-acid glycoprotein). The enzyme was found to act only on native IgG and AGP and to be specific for free biantennary glycans with or without terminal sialylation. GAS M49 expression of EndoS2 was monitored in relation to carbohydrates present in the culture medium and was linked to the presence of sucrose. We conclude that EndoS2 is a unique endoglycosidase in serotype M49 and differs from EndoS of other GAS strains by targeting both IgG and AGP. EndoS2 expands the repertoire of GAS effectors that modify key glycosylated molecules of host defence.


Asunto(s)
Proteínas Bacterianas/metabolismo , Glicósido Hidrolasas/metabolismo , Inmunoglobulina G/metabolismo , Orosomucoide/metabolismo , Streptococcus pyogenes/enzimología , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Secuencia de Carbohidratos , Secuencia Conservada , Escherichia coli/genética , Escherichia coli/metabolismo , Glicósido Hidrolasas/química , Glicósido Hidrolasas/genética , Interacciones Huésped-Patógeno , Humanos , Inmunoglobulina G/química , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Datos de Secuencia Molecular , Orosomucoide/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Streptococcus pyogenes/química , Streptococcus pyogenes/genética , Especificidad por Sustrato , Sacarosa/metabolismo
10.
J Clin Invest ; 134(9)2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38502193

RESUMEN

Chimeric antigen receptor (CAR) designs that incorporate pharmacologic control are desirable; however, designs suitable for clinical translation are needed. We designed a fully human, rapamycin-regulated drug product for targeting CD33+ tumors called dimerizaing agent-regulated immunoreceptor complex (DARIC33). T cell products demonstrated target-specific and rapamycin-dependent cytokine release, transcriptional responses, cytotoxicity, and in vivo antileukemic activity in the presence of as little as 1 nM rapamycin. Rapamycin withdrawal paused DARIC33-stimulated T cell effector functions, which were restored following reexposure to rapamycin, demonstrating reversible effector function control. While rapamycin-regulated DARIC33 T cells were highly sensitive to target antigen, CD34+ stem cell colony-forming capacity was not impacted. We benchmarked DARIC33 potency relative to CD19 CAR T cells to estimate a T cell dose for clinical testing. In addition, we integrated in vitro and preclinical in vivo drug concentration thresholds for off-on state transitions, as well as murine and human rapamycin pharmacokinetics, to estimate a clinically applicable rapamycin dosing schedule. A phase I DARIC33 trial has been initiated (PLAT-08, NCT05105152), with initial evidence of rapamycin-regulated T cell activation and antitumor impact. Our findings provide evidence that the DARIC platform exhibits sensitive regulation and potency needed for clinical application to other important immunotherapy targets.


Asunto(s)
Leucemia Mieloide Aguda , Lectina 3 Similar a Ig de Unión al Ácido Siálico , Sirolimus , Linfocitos T , Animales , Femenino , Humanos , Masculino , Ratones , Inmunoterapia Adoptiva , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Receptores Quiméricos de Antígenos/inmunología , Lectina 3 Similar a Ig de Unión al Ácido Siálico/inmunología , Lectina 3 Similar a Ig de Unión al Ácido Siálico/metabolismo , Sirolimus/farmacología , Sirolimus/administración & dosificación , Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Biol Chem ; 287(49): 40891-7, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23038245

RESUMEN

The bacterial pathogen Group A Streptococcus (GAS) colonizes epithelial and mucosal surfaces and can cause a broad spectrum of human disease. Through the secreted plasminogen activator streptokinase (Ska), GAS activates human plasminogen into plasmin and binds it to the bacterial surface. The resulting surface plasmin protease activity has been proposed to play a role in disrupting tissue barriers, promoting invasive spread of the bacterium. We investigated whether this surface protease activity could aid the immune evasion role through degradation of the key innate antimicrobial peptide LL-37, the human cathelicidin. Cleavage products of plasmin-degraded LL-37 were analyzed by matrix-assisted laser desorption ionization mass spectrometry. Ska-deficient GAS strains were generated by targeted allelic exchange mutagenesis and confirmed to lack surface plasmin activity after growth in human plasma or media supplemented with plasminogen and fibrinogen. Loss of surface plasmin activity left GAS unable to efficiently degrade LL-37 and increased bacterial susceptibility to killing by the antimicrobial peptide. When mice infected with GAS were simultaneously treated with the plasmin inhibitor aprotinin, a significant reduction in the size of necrotic skin lesions was observed. Together these data reveal a novel immune evasion strategy of the human pathogen: co-opting the activity of a host protease to evade peptide-based innate host defenses.


Asunto(s)
Catelicidinas/química , Catelicidinas/metabolismo , Alelos , Secuencia de Aminoácidos , Animales , Anticoagulantes/química , Péptidos Catiónicos Antimicrobianos , Fibrinolisina/química , Humanos , Sistema Inmunológico , Inmunidad Innata , Cinética , Ratones , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Mutagénesis , Plasminógeno/química , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Streptococcus pyogenes/metabolismo , Estreptoquinasa/química , Estreptoquinasa/metabolismo , Factores de Virulencia/metabolismo
12.
J Biol Chem ; 287(17): 13889-98, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22371493

RESUMEN

Staphylococcus aureus causes a wide range of human disease ranging from localized skin and soft tissue infections to potentially lethal systemic infections. S. aureus has the biosynthetic ability to generate numerous virulence factors that assist in circumventing the innate immune system during disease pathogenesis. Recent studies have uncovered a set of extracellular peptides produced by community-associated methicillin-resistant S. aureus (CA-MRSA) with homology to the phenol-soluble modulins (PSMs) from Staphylococcus epidermidis. CA-MRSA PSMs contribute to skin infection and recruit and lyse neutrophils, and truncated versions of these peptides possess antimicrobial activity. In this study, novel CA-MRSA PSM derivatives were discovered by the use of microbial imaging mass spectrometry. The novel PSM derivatives are compared with their parent full-length peptides for changes in hemolytic, cytolytic, and neutrophil-stimulating activity. A potential contribution of the major S. aureus secreted protease aureolysin in processing PSMs is demonstrated. Finally, we show that PSM processing occurs in multiple CA-MRSA strains by structural confirmation of additional novel derivatives. This work demonstrates that IMS can serve as a useful tool to go beyond genome predictions and expand our understanding of the important family of small peptide virulence factors.


Asunto(s)
Espectrometría de Masas/métodos , Staphylococcus aureus Resistente a Meticilina/metabolismo , Fenol/química , Secuencia de Aminoácidos , Animales , Antiinfecciosos/farmacología , Infecciones Bacterianas/metabolismo , Proteínas Bacterianas/química , Toxinas Bacterianas/química , Eritrocitos/citología , Hemólisis , Humanos , Inmunosupresores/farmacología , Metaloendopeptidasas/química , Ratones , Datos de Secuencia Molecular , Neutrófilos/citología , Neutrófilos/metabolismo , Homología de Secuencia de Aminoácido , Ovinos , Piel/metabolismo , Piel/microbiología , Infecciones Cutáneas Estafilocócicas/microbiología , Factores de Virulencia/química
13.
FASEB J ; 26(11): 4675-84, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22878963

RESUMEN

The past 50 years has witnessed the emergence of new viral and bacterial pathogens with global effect on human health. The hyperinvasive group A Streptococcus (GAS) M1T1 clone, first detected in the mid-1980s in the United States, has since disseminated worldwide and remains a major cause of severe invasive human infections. Although much is understood regarding the capacity of this pathogen to cause disease, much less is known of the precise evolutionary events selecting for its emergence. We used high-throughput technologies to sequence a World Health Organization strain collection of serotype M1 GAS and reconstructed its phylogeny based on the analysis of core genome single-nucleotide polymorphisms. We demonstrate that acquisition of a 36-kb genome segment from serotype M12 GAS and the bacteriophage-encoded DNase Sda1 led to increased virulence of the M1T1 precursor and occurred relatively early in the molecular evolutionary history of this strain. The more recent acquisition of the phage-encoded superantigen SpeA is likely to have provided selection advantage for the global dissemination of the M1T1 clone. This study provides an exemplar for the evolution and emergence of virulent clones from microbial populations existing commensally or causing only superficial infection.


Asunto(s)
Evolución Biológica , Pandemias , Infecciones Estreptocócicas/epidemiología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/microbiología , Exotoxinas/genética , Exotoxinas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Genoma Bacteriano , Salud Global , Interacciones Huésped-Patógeno , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Neutrófilos/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fagocitosis , Filogenia , Streptococcus pyogenes/clasificación , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Transcriptoma , Virulencia
14.
Microbiology (Reading) ; 157(Pt 9): 2485-2492, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21719540

RESUMEN

Microbial competition exists in the general environment, such as soil or aquatic habitats, upon or within unicellular or multicellular eukaryotic life forms. The molecular actions that govern microbial competition, leading to niche establishment and microbial monopolization, remain undetermined. The emerging technology of imaging mass spectrometry (IMS) enabled the observation that there is directionality in the metabolic output of the organism Bacillus subtilis when co-cultured with Staphylococcus aureus. The directionally released antibiotic alters S. aureus virulence factor production and colonization. Therefore, IMS provides insight into the largely hidden nature of competitive microbial encounters and niche establishment, and provides a paradigm for future antibiotic discovery.


Asunto(s)
Bacillus subtilis/metabolismo , Viabilidad Microbiana , Staphylococcus aureus/metabolismo , Animales , Bacillus subtilis/aislamiento & purificación , Técnicas de Cocultivo , Ácidos Grasos/metabolismo , Femenino , Humanos , Lipopéptidos/metabolismo , Masculino , Espectrometría de Masas , Ratones , Microscopía Fluorescente , Oligopéptidos/metabolismo , Péptidos Cíclicos/metabolismo , Staphylococcus aureus/aislamiento & purificación
15.
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
16.
J Infect Dis ; 202(1): 11-9, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20507231

RESUMEN

BACKGROUND: The recent resurgence of invasive group A streptococcal disease has been paralleled by the emergence of the M1T1 clone. Recently, invasive disease initiation has been linked to mutations in the covR/S 2-component regulator. We investigated whether a fitness cost is associated with the covS mutation that counterbalances hypervirulence. METHODS: Wild-type M1T1 group A Streptococcus and an isogenic covS-mutant strain derived from animal passage were compared for adherence to human laryngeal epithelial cells, human keratinocytes, or fibronectin; biofilm formation; and binding to intact mouse skin. Targeted mutagenesis of capsule expression of both strains was performed for analysis of its unique contribution to the observed phenotypes. RESULTS: The covS-mutant bacteria showed reduced capacity to bind to epithelial cell layers as a consequence of increased capsule expression. The covS-mutant strain also had reduced capacity to bind fibronectin and to form biofilms on plastic and epithelial cell layers. A defect in skin adherence of the covS-mutant strain was demonstrated in a murine model. CONCLUSION: Reduced colonization capacity provides a potential explanation for why the covS mutation, which confers hypervirulence, has not become fixed in the globally disseminated M1T1 group A Streptococcus clone, but rather may arise anew under innate immune selection in individual patients.


Asunto(s)
Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Animales , Adhesión Bacteriana , Línea Celular , Células Epiteliales/microbiología , Fibronectinas/metabolismo , Humanos , Queratinocitos/microbiología , Ratones , Fenotipo , Unión Proteica , Piel/microbiología , Virulencia
17.
Nat Commun ; 6: 8369, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26458291

RESUMEN

Tamoxifen is a selective oestrogen receptor modulator widely used for the treatment of breast cancer. In addition to its activity as an oestrogen receptor agonist/antagonist, tamoxifen also modulates sphingolipid biosynthesis, which has been shown to play an important role in the regulation of neutrophil activity. Here, we find that tamoxifen stimulation enhances several pro-inflammatory pathways in human neutrophils, including chemotaxis, phagocytosis and neutrophil extracellular trap (NET) formation. The enhancement of NET production occurs via a ceramide/PKCζ-mediated pathway, and treatment with synthetic ceramide is sufficient to promote NET formation. Pretreatment of human neutrophils with tamoxifen boosts neutrophil bactericidal capacity against a variety of pathogens in vitro and enhances clearance of the leading human pathogen methicillin-resistant Staphylococcus aureus in vivo. Our results suggest that tamoxifen, and the lipid signalling pathways it modulates, merit further exploration as targets for boosting host innate immune function.


Asunto(s)
Ceramidas/metabolismo , Trampas Extracelulares/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología , Animales , Femenino , Voluntarios Sanos , Humanos , Inmunidad Innata/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina , Ratones , Neutrófilos/metabolismo , Proteína Quinasa C/metabolismo
18.
mBio ; 6(2): e00133, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25759502

RESUMEN

UNLABELLED: The M1T1 clone of group A Streptococcus (GAS) is associated with severe invasive infections, including necrotizing fasciitis and septicemia. During invasive M1T1 GAS disease, mutations in the covRS regulatory system led to upregulation of an ADP-ribosyltransferase, SpyA. Surprisingly, a GAS ΔspyA mutant was resistant to killing by macrophages and caused higher mortality with impaired bacterial clearance in a mouse intravenous challenge model. GAS expression of SpyA triggered macrophage cell death in association with caspase-1-dependent interleukin 1ß (IL-1ß) production, and differences between wild-type (WT) and ΔspyA GAS macrophage survival levels were lost in cells lacking caspase-1, NOD-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein (ASC), or pro-IL-1ß. Similar in vitro findings were identified in macrophage studies performed with pseudomonal exotoxin A, another ADP-ribosylating toxin. Thus, SpyA triggers caspase-1-dependent inflammatory cell death in macrophages, revealing a toxin-triggered IL-1ß-dependent innate immune response pathway critical in defense against invasive bacterial infection. IMPORTANCE: Group A Streptococcus (GAS) is a leading human pathogen capable of producing invasive infections even in healthy individuals. GAS bacteria produce a toxin called SpyA that modifies host proteins through a process called ADP ribosylation. We describe how macrophages, frontline defenders of the host innate immune system, respond to SpyA by undergoing a specialized form of cell death in which they are activated to release the proinflammatory cytokine molecule interleukin 1ß (IL-1ß). Release of IL-1ß activates host immune cell clearance of GAS, as we demonstrated in tissue culture models of macrophage bacterial killing and in vivo mouse infectious-challenge experiments. Similar macrophage responses to a related toxin of Pseudomonas bacteria were also shown. Thus, macrophages recognize certain bacterial toxins to activate a protective immune response in the host.


Asunto(s)
ADP Ribosa Transferasas/inmunología , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Streptococcus pyogenes/enzimología , Streptococcus pyogenes/inmunología , ADP Ribosa Transferasas/genética , Animales , Supervivencia Celular , Modelos Animales de Enfermedad , Eliminación de Gen , Ratones , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/genética , Virulencia
19.
EBioMedicine ; 2(7): 690-8, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26288841

RESUMEN

Antibiotic resistance poses an increasingly grave threat to the public health. Of pressing concern, rapid spread of carbapenem-resistance among multidrug-resistant (MDR) Gram-negative rods (GNR) is associated with few treatment options and high mortality rates. Current antibiotic susceptibility testing guiding patient management is performed in a standardized manner, identifying minimum inhibitory concentrations (MIC) in bacteriologic media, but ignoring host immune factors. Lacking activity in standard MIC testing, azithromycin (AZM), the most commonly prescribed antibiotic in the U.S., is never recommended for MDR GNR infection. Here we report a potent bactericidal action of AZM against MDR carbapenem-resistant isolates of Pseudomonas aeruginosa, Klebsiella pneumoniae, and Acinetobacter baumannii. This pharmaceutical activity is associated with enhanced AZM cell penetration in eukaryotic tissue culture media and striking multi-log-fold synergies with host cathelicidin antimicrobial peptide LL-37 or the last line antibiotic colistin. Finally, AZM monotherapy exerts clear therapeutic effects in murine models of MDR GNR infection. Our results suggest that AZM, currently ignored as a treatment option, could benefit patients with MDR GNR infections, especially in combination with colistin.


Asunto(s)
Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Azitromicina/farmacología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Animales , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Azitromicina/uso terapéutico , Catelicidinas/farmacología , Catelicidinas/uso terapéutico , Permeabilidad de la Membrana Celular/efectos de los fármacos , Colistina/farmacología , Colistina/uso terapéutico , Medios de Cultivo , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Bacterias Gramnegativas/ultraestructura , Humanos , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana
20.
J Inflamm (Lond) ; 10(1): 28, 2013 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-23937964

RESUMEN

BACKGROUND: Hypoxia inducible factor-1 (HIF-1) is a major regulator of the cellular adaption to low oxygen stress and the innate immune function of myeloid cells. Treatment with the novel HIF-1 stabilizing drug AKB-4924 has been shown to enhance the bactericidal activity of keratinocytes as well as phagocytic cells. In this study, we sought to investigate the effect of pharmacological boosting of HIF-1 with AKB-4924 in keratinocytes and their contribution to the innate immune response. FINDINGS: Treatment with the novel HIF-1 stabilizing drug AKB-4924 can increase keratinocyte production of pro-inflammatory cytokines in vitro and enhance neutrophil recruitment in vivo. CONCLUSIONS: HIF plays an important role in cytokine production by keratinocytes and in neutrophil recruitment to the skin. The HIF-boosting drug AKB-4924 has the potential to enhance the immune response even in the complex environment of bacterial skin infections.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA