Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 605(7910): 527-531, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35545676

RESUMEN

Gasdermins (GSDMs) are a family of pore-forming effectors that permeabilize the cell membrane during the cell death program pyroptosis1. GSDMs are activated by proteolytic removal of autoinhibitory carboxy-terminal domains, typically by caspase regulators1-9. However, no activator is known for one member of this family, GSDMA. Here we show that the major human pathogen group A Streptococcus (GAS) secretes a protease virulence factor, SpeB, that induces GSDMA-dependent pyroptosis. SpeB cleavage of GSDMA releases an active amino-terminal fragment that can insert into membranes to form lytic pores. GSDMA is primarily expressed in the skin10, and keratinocytes infected with SpeB-expressing GAS die of GSDMA-dependent pyroptosis. Mice have three homologues of human GSDMA, and triple-knockout mice are more susceptible to invasive infection by a pandemic hypervirulent M1T1 clone of GAS. These results indicate that GSDMA is critical in the immune defence against invasive skin infections by GAS. Furthermore, they show that GSDMs can act independently of host regulators as direct sensors of exogenous proteases. As SpeB is essential for tissue invasion and survival within skin cells, these results suggest that GSDMA can act akin to a guard protein that directly detects concerning virulence activities of microorganisms that present a severe infectious threat.


Asunto(s)
Piroptosis , Streptococcus pyogenes , Animales , Caspasas , Queratinocitos , Ratones , Proteínas Citotóxicas Formadoras de Poros , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/patogenicidad , Virulencia , Factores de Virulencia
2.
PLoS Pathog ; 19(4): e1011321, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37068092

RESUMEN

Group A Streptococcus (GAS, Streptococcus pyogenes) is a professional human pathogen that commonly infects the skin. Keratinocytes are one of the first cells to contact GAS, and by inducing inflammation, they can initiate the earliest immune responses to pathogen invasion. Here, we characterized the proinflammatory cytokine repertoire produced by primary human keratinocytes and surrogate cell lines commonly used in vitro. Infection induces several cytokines and chemokines, but keratinocytes constitutively secrete IL-18 in a form that is inert (pro-IL-18) and lacks proinflammatory activity. Canonically, IL-18 activation and secretion are coupled through a single proteolytic event that is regulated intracellularly by the inflammasome protease caspase-1 in myeloid cells. The pool of extracellular pro-IL-18 generated by keratinocytes is poised to sense extracellular proteases. It is directly processed into a mature active form by SpeB, a secreted GAS protease that is a critical virulent factor during skin infection. This mechanism contributes to the proinflammatory response against GAS, resulting in T cell activation and the secretion of IFN-γ. Under these conditions, isolates of several other major bacterial pathogens and microbiota of the skin were found to not have significant IL-18-maturing ability. These results suggest keratinocyte-secreted IL-18 is a sentinel that sounds an early alarm that is highly sensitive to GAS, yet tolerant to non-invasive members of the microbiota.


Asunto(s)
Infecciones Bacterianas , Interleucina-18 , Humanos , Infecciones Bacterianas/metabolismo , Citocinas/metabolismo , Inflamación , Interleucina-18/metabolismo , Queratinocitos/metabolismo , Péptido Hidrolasas/metabolismo
3.
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.
J Infect Dis ; 223(7): 1260-1264, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32808035

RESUMEN

Invasive group A Streptococcus (GAS) in immunocompetent individuals is largely linked to hypervirulent strains. Congenital immunodeficiencies and those acquired from chronic disease or immunosuppressant drugs also increase risk of severe illness. We recovered GAS from the blood of a patient receiving a biologic inhibitor of interleukin 6 (IL-6). Growth of this serotype M4 isolate in human blood or a murine bacteremia model was promoted by interleukin 1 or IL-6 inhibition. Hyperinvasive M1T1 GAS was unaffected by IL-6 in both models. These findings based on a natural experiment introduce IL-6 signaling deficiencies as a risk factor for invasive GAS.


Asunto(s)
Inmunoterapia/efectos adversos , Interleucina-6 , Infecciones Oportunistas/microbiología , Infecciones Estreptocócicas , Animales , Humanos , Interleucina-6/antagonistas & inhibidores , Ratones , Infecciones Estreptocócicas/tratamiento farmacológico , Streptococcus pyogenes
5.
Mol Pharm ; 18(8): 3171-3180, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34279974

RESUMEN

Current treatment of chronic wounds has been critically limited by various factors, including bacterial infection, biofilm formation, impaired angiogenesis, and prolonged inflammation. Addressing these challenges, we developed a multifunctional wound dressing-based three-pronged approach for accelerating wound healing. The multifunctional wound dressing, composed of nanofibers, functional nanoparticles, natural biopolymers, and selected protein and peptide, can target multiple endogenous repair mechanisms and represents a promising alternative to current wound healing products.


Asunto(s)
Anexina A1/administración & dosificación , Antiinflamatorios/administración & dosificación , Vendajes , Diabetes Mellitus Experimental/complicaciones , Proteínas Relacionadas con la Folistatina/administración & dosificación , Péptidos/administración & dosificación , Infecciones Estafilocócicas/complicaciones , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Herida Quirúrgica/complicaciones , Herida Quirúrgica/tratamiento farmacológico , Cicatrización de Heridas/efectos de los fármacos , Infección de Heridas/complicaciones , Infección de Heridas/tratamiento farmacológico , Células 3T3 , Animales , Materiales Biocompatibles/administración & dosificación , Biopolímeros/química , Supervivencia Celular/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Células HaCaT , Humanos , Nanopartículas Magnéticas de Óxido de Hierro/química , Masculino , Ensayo de Materiales/métodos , Ratones , Nanofibras/química , Ratas , Ratas Wistar , Infecciones Estafilocócicas/microbiología , Resultado del Tratamiento , Infección de Heridas/microbiología
6.
J Biol Chem ; 294(34): 12846-12854, 2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31300552

RESUMEN

Chronic heart failure and cardiac arrhythmias have high morbidity and mortality, and drugs for the prevention and management of these diseases are a large part of the pharmaceutical market. Among these drugs are plant-derived cardiac glycosides, which have been used by various cultures over millennia as both medicines and poisons. We report that digoxin and related compounds activate the NLRP3 inflammasome in macrophages and cardiomyocytes at concentrations achievable during clinical use. Inflammasome activation initiates the maturation and release of the inflammatory cytokine IL-1ß and the programmed cell death pathway pyroptosis in a caspase-1-dependent manner. Notably, the same fluxes of potassium and calcium cations that affect heart contraction also induce inflammasome activation in human but not murine cells. Pharmaceuticals that antagonize these fluxes, including glyburide and verapamil, also inhibit inflammasome activation by cardiac glycosides. Cardiac glycoside-induced cellular cytotoxicity and IL-1ß signaling are likewise antagonized by inhibitors of the NLRP3 inflammasome or the IL-1 receptor-targeting biological agent anakinra. Our results inform on the molecular mechanism by which the inflammasome integrates the diverse signals that activate it through secondary signals like cation flux. Furthermore, this mechanism suggests a contribution of the inflammasome to the toxicity and adverse events associated with cardiac glycosides use in humans and that targeted anti-inflammatories could provide an additional adjunct therapeutic countermeasure.


Asunto(s)
Digoxina/antagonistas & inhibidores , Inflamasomas/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Citocinas/análisis , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Digoxina/farmacología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
7.
Infect Immun ; 88(10)2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32719155

RESUMEN

Group A Streptococcus (GAS) is the etiologic agent of numerous high-morbidity and high-mortality diseases. Infections are typically highly proinflammatory. During the invasive infection necrotizing fasciitis, this is in part due to the GAS protease SpeB directly activating interleukin-1ß (IL-1ß) independent of the canonical inflammasome pathway. The upper respiratory tract is the primary site for GAS colonization, infection, and transmission, but the host-pathogen interactions at this site are still largely unknown. We found that in the murine nasopharynx, SpeB enhanced IL-1ß-mediated inflammation and the chemotaxis of neutrophils. However, neutrophilic inflammation did not restrict infection and instead promoted GAS replication and disease. Inhibiting IL-1ß or depleting neutrophils, which both promote invasive infection, prevented GAS infection of the nasopharynx. Mice pretreated with penicillin became more susceptible to GAS challenge, and this reversed the attenuation from neutralization or depletion of IL-1ß, neutrophils, or SpeB. Collectively, our results suggest that SpeB is essential to activate an IL-1ß-driven neutrophil response. Unlike during invasive tissue infections, this is beneficial in the upper respiratory tract because it disrupts colonization resistance mediated by the microbiota. This provides experimental evidence that the notable inflammation of strep throat, which presents with significant swelling, pain, and neutrophil influx, is not an ineffectual immune response but rather is a GAS-directed remodeling of this niche for its pathogenic benefit.


Asunto(s)
Nasofaringe/inmunología , Receptores Tipo I de Interleucina-1/inmunología , Transducción de Señal/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/patogenicidad , Animales , Antibacterianos/efectos adversos , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Caspasa 1/genética , Caspasa 1/inmunología , Quimiotaxis de Leucocito , Exotoxinas/genética , Exotoxinas/inmunología , Inflamación , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1beta/inmunología , Ratones , Nasofaringe/microbiología , Neutrófilos/inmunología , Faringitis/genética , Faringitis/inmunología , Faringitis/microbiología , Receptores Tipo I de Interleucina-1/genética , Transducción de Señal/efectos de los fármacos , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/efectos de los fármacos , Streptococcus pyogenes/genética , Streptococcus pyogenes/crecimiento & desarrollo , Virulencia/efectos de los fármacos , Virulencia/genética
8.
Hum Mol Genet ; 27(1): 41-52, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29048558

RESUMEN

Although Zika virus (ZIKV) infection is often asymptomatic, in some cases, it can lead to birth defects in newborns or serious neurologic complications in adults. However, little is known about the interplay between immune and neural cells that could contribute to the ZIKV pathology. To understand the mechanisms at play during infection and the antiviral immune response, we focused on neural precursor cells (NPCs)-microglia interactions. Our data indicate that human microglia infected with the current circulating Brazilian ZIKV induces a similar pro-inflammatory response found in ZIKV-infected human tissues. Importantly, using our model, we show that microglia interact with ZIKV-infected NPCs and further spread the virus. Finally, we show that Sofosbuvir, an FDA-approved drug for Hepatitis C, blocked viral infection in NPCs and therefore the transmission of the virus from microglia to NPCs. Thus, our model provides a new tool for studying neuro-immune interactions and a platform to test new therapeutic drugs.


Asunto(s)
Infección por el Virus Zika/inmunología , Virus Zika/patogenicidad , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas/patología , Microglía/patología , Modelos Biológicos , Células-Madre Neurales/patología , Sofosbuvir/farmacología , Virus Zika/metabolismo
9.
Biochim Biophys Acta ; 1848(11 Pt B): 3047-54, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25701232

RESUMEN

Cationic antimicrobial peptides (CAMPs) are critical front line contributors to host defense against invasive bacterial infection. These immune factors have direct killing activity toward microbes, but many pathogens are able to resist their effects. Group A Streptococcus, group B Streptococcus and Streptococcus pneumoniae are among the most common pathogens of humans and display a variety of phenotypic adaptations to resist CAMPs. Common themes of CAMP resistance mechanisms among the pathogenic streptococci are repulsion, sequestration, export, and destruction. Each pathogen has a different array of CAMP-resistant mechanisms, with invasive disease potential reflecting the utilization of several mechanisms that may act in synergy. Here we discuss recent progress in identifying the sources of CAMP resistance in the medically important Streptococcus genus. Further study of these mechanisms can contribute to our understanding of streptococcal pathogenesis, and may provide new therapeutic targets for therapy and disease prevention. This article is part of a Special Issue entitled: Bacterial Resistance to Antimicrobial Peptides.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana , Infecciones Estreptocócicas/metabolismo , Streptococcus/metabolismo , Animales , Antibacterianos/uso terapéutico , Péptidos Catiónicos Antimicrobianos/inmunología , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Proteínas Bacterianas/inmunología , Transporte Biológico , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Inmunidad Innata , Proteínas de Transporte de Membrana/metabolismo , Viabilidad Microbiana , Proteolisis , Transducción de Señal , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/prevención & control , Streptococcus/efectos de los fármacos , Streptococcus/inmunología , Streptococcus/patogenicidad
10.
ACS Pharmacol Transl Sci ; 7(3): 855-862, 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38481694

RESUMEN

Accurate assessment of nanomedicines' antibacterial properties is pivotal for their effective use in both in vitro and in vivo settings. Conventional antibacterial activity assessment methods, involving bacterial coculture with compounds on agar plates, may not fully suit nanomedicines due to their susceptibility to alterations in physicochemical properties induced by biological fluids. Furthermore, these biological fluids might even enhance the bacterial growth. This study introduces a novel, rigorous, and reproducible methodology for evaluating nanomedicine antibacterial properties using cell culture media (i.e., DMEM-FBS10%). To assess the antibacterial activity of the nanoparticles in cell culture media, superparamagnetic iron oxide nanoparticles (SPIONs) were chosen as the model nanomedicine due to their clinical significance. A comparative analysis between the traditional and our proposed methods yielded contrasting outcomes, shedding light on the significant impact of biological fluids on nanoparticle antibacterial activities. While the conventional approach suggested the antibacterial effectiveness of SPIONs against Staphylococcus aureus, our innovative method unveiled a substantial increase in bacterial growth in the presence of biological fluids. More specifically, we found a significant increase in bacterial growth when exposed to bare SPIONs at various concentrations, while the formation of a protein corona on SPION surfaces could markedly reduce the observed bacterial growth compared to the control group. These findings underscore the necessity for more refined evaluation techniques that can better replicate the in vivo environment when studying the nanomedicine's antibacterial capabilities.

11.
Methods Mol Biol ; 2641: 163-169, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37074649

RESUMEN

The cytokine interleukin-1ß (IL-1ß) is a major mediator of inflammation in autoinflammatory disease and the host response to infection. IL-1ß is stored within cells in an inert form, which requires proteolytic removal of an amino-terminal fragment in order to bind the IL-1 receptor complex and have pro-inflammatory activity. This cleavage event is canonically carried out by inflammasome-activated caspase proteases, but microbe and host proteases can also generate unique active forms. Post-translational regulation of IL-1ß and the diversity in resulting products can present challenges to the evaluation of IL-1ß activation. This chapter details methods and important controls for the accurate and sensitive measurement of IL-1ß activation within biological samples.


Asunto(s)
Inflamasomas , Piroptosis , Piroptosis/fisiología , Interleucina-1beta/metabolismo , Inflamasomas/metabolismo , Caspasas , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Caspasa 1/metabolismo
12.
ACS Infect Dis ; 9(2): 270-282, 2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36669138

RESUMEN

LasB elastase is a broad-spectrum exoprotease and a key virulence factor of Pseudomonas aeruginosa, a major pathogen causing lung damage and inflammation in acute and chronic respiratory infections. Here, we describe the chemical optimization of specific LasB inhibitors with druglike properties and investigate their impact in cellular and animal models of P. aeruginosa infection. Competitive inhibition of LasB was demonstrated through structural and kinetic studies. In vitro LasB inhibition was confirmed with respect to several host target proteins, namely, elastin, IgG, and pro-IL-1ß. Furthermore, inhibition of LasB-mediated IL-1ß activation was demonstrated in macrophage and mouse lung infection models. In mice, intravenous administration of inhibitors also resulted in reduced bacterial numbers at 24 h. These highly potent, selective, and soluble LasB inhibitors constitute valuable tools to study the proinflammatory impact of LasB in P. aeruginosa infections and, most importantly, show clear potential for the clinical development of a novel therapy for life-threatening respiratory infections caused by this opportunistic pathogen.


Asunto(s)
Pseudomonas aeruginosa , Factores de Virulencia , Animales , Ratones , Cinética , Modelos Animales , Elastasa Pancreática
13.
iScience ; 25(9): 104947, 2022 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-36065192

RESUMEN

Biomaterial-associated microbial contaminations in biologically conducive three-dimensional (3D) tissue-engineered constructs have significantly limited the clinical applications of scaffold systems. To prevent such infections, antimicrobial biomaterials are rapidly evolving. Yet, the use of such materials in bioprinting-based approaches of scaffold fabrication has not been examined. This study introduces a new generation of bacteriostatic gelatin methacryloyl (GelMA)-based bioinks, incorporated with varying doses of antibacterial superparamagnetic iron oxide nanoparticles (SPIONs). The SPION-laden GelMA scaffolds showed significant resistance against the Staphylococcus aureus growth, while providing a contrast in magnetic resonance imaging. We simulated the bacterial contamination of cellular 3D GelMA scaffolds in vitro and demonstrated the significant effect of functionalized scaffolds in inhibiting bacterial growth, while maintaining cell viability and growth. Together, these results present a new promising class of functionalized bioinks to 3D bioprint tissue-engineered scaffold with markedly enhanced properties for the use in a variety of in vitro and clinical applications.

14.
Front Cell Infect Microbiol ; 11: 704099, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34295841

RESUMEN

Group A Streptococcus is an obligate human pathogen that is a major cause of infectious morbidity and mortality. It has a natural tropism for the oropharynx and skin, where it causes infections with excessive inflammation due to its expression of proinflammatory toxins and other virulence factors. Inflammation directly contributes to the severity of invasive infections, toxic shock syndrome, and the induction of severe post-infection autoimmune disease caused by autoreactive antibodies. This review discusses what is known about how the virulence factors of Group A Streptococcus induce inflammation and how this inflammation can promote disease. Understanding of streptococcal pathogenesis and the role of hyper-immune activation during infection may provide new therapeutic targets to treat the often-fatal outcome of severe disease.


Asunto(s)
Choque Séptico , Infecciones Estreptocócicas , Humanos , Streptococcus pyogenes , Virulencia , Factores de Virulencia
15.
Front Microbiol ; 12: 760255, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34803985

RESUMEN

Group A Streptococcus (GAS; Streptococcus pyogenes) is a nearly ubiquitous human pathogen responsible for a significant global disease burden. No vaccine exists, so antibiotics are essential for effective treatment. Despite a lower incidence of antimicrobial resistance than many pathogens, GAS is still a top 10 cause of death due to infections worldwide. The morbidity and mortality are primarily a consequence of the immune sequelae and invasive infections that are difficult to treat with antibiotics. GAS has remained susceptible to penicillin and other ß-lactams, despite their widespread use for 80 years. However, the failure of treatment for invasive infections with penicillin has been consistently reported since the introduction of antibiotics, and strains with reduced susceptibility to ß-lactams have emerged. Furthermore, isolates responsible for outbreaks of severe infections are increasingly resistant to other antibiotics of choice, such as clindamycin and macrolides. This review focuses on the challenges in the treatment of GAS infection, the mechanisms that contribute to antibiotic failure, and adjunctive therapeutics. Further understanding of these processes will be necessary for improving the treatment of high-risk GAS infections and surveillance for non-susceptible or resistant isolates. These insights will also help guide treatments against other leading pathogens for which conventional antibiotic strategies are increasingly failing.

16.
Polymers (Basel) ; 13(7)2021 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-33808295

RESUMEN

Current strategies for regeneration of large bone fractures yield limited clinical success mainly due to poor integration and healing. Multidisciplinary approaches in design and development of functional tissue engineered scaffolds are required to overcome these translational challenges. Here, a new generation of hyperelastic bone (HB) implants, loaded with superparamagnetic iron oxide nanoparticles (SPIONs), are 3D bioprinted and their regenerative effect on large non-healing bone fractures is studied. Scaffolds are bioprinted with the geometry that closely correspond to that of the bone defect, using an osteoconductive, highly elastic, surgically friendly bioink mainly composed of hydroxyapatite. Incorporation of SPIONs into HB bioink results in enhanced bacteriostatic properties of bone grafts while exhibiting no cytotoxicity. In vitro culture of mouse embryonic cells and human osteoblast-like cells remain viable and functional up to 14 days on printed HB scaffolds. Implantation of damage-specific bioprinted constructs into a rat model of femoral bone defect demonstrates significant regenerative effect over the 2-week time course. While no infection, immune rejection, or fibrotic encapsulation is observed, HB grafts show rapid integration with host tissue, ossification, and growth of new bone. These results suggest a great translational potential for 3D bioprinted HB scaffolds, laden with functional nanoparticles, for hard tissue engineering applications.

18.
J Cataract Refract Surg ; 46(7): 1047-1050, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32427643

RESUMEN

PURPOSE: To test the antiseptic efficacy of povidone-iodine when mixed with topical lidocaine gel. SETTING: Emory School of Medicine, Atlanta, Georgia, USA. DESIGN: Experimental study. METHODS: Staphylococcus epidermidis, Staphylococcus aureus, viridans streptococci (Streptococcus sanguinis), Escherichia coli, and Pseudomonas aeruginosa were grown on blood agar plates with povidone-iodine and/or lidocaine gel. The efficacy of sterilization was quantified by surviving bacterial colony-forming units. RESULTS: Combination of povidone-iodine and lidocaine gel prevented bacterial growth to levels similar to povidone-iodine alone. Application of lidocaine gel to bacteria before povidone-iodine treatment allowed bacterial growth similar to controls not exposed to povidone-iodine. CONCLUSIONS: Povidone-iodine must be applied before lidocaine gel for effective antisepsis, but admixture of povidone-iodine with lidocaine gel was also effective and may reduce the risk of endophthalmitis associated with lidocaine gel use. This strategy offers a practice-changing alternative for preoperative prophylaxis in procedures requiring topical anesthesia.


Asunto(s)
Anestesia , Antiinfecciosos Locales , Humanos , Lidocaína , Povidona Yodada , Staphylococcus epidermidis
19.
EBioMedicine ; 60: 102984, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32979835

RESUMEN

BACKGROUND: Pulmonary damage by Pseudomonas aeruginosa during cystic fibrosis lung infection and ventilator-associated pneumonia is mediated both by pathogen virulence factors and host inflammation. Impaired immune function due to tissue damage and inflammation, coupled with pathogen multidrug resistance, complicates the management of these deep-seated infections. Pathological inflammation during infection is driven by interleukin-1ß (IL-1ß), but the molecular processes involved are not fully understood. METHODS: We examined IL-1ß activation in a pulmonary model infection of Pseudomonas aeruginosa and in vitro using genetics, specific inhibitors, recombinant proteins, and targeted reporters of protease activity and IL-1ß bioactivity. FINDINGS: Caspase-family inflammasome proteases canonically regulate maturation of this proinflammatory cytokine, but we report that plasticity in IL-1ß proteolytic activation allows for its direct maturation by the pseudomonal protease LasB. LasB promotes IL-1ß activation, neutrophilic inflammation, and destruction of lung architecture characteristic of severe P. aeruginosa pulmonary infection. INTERPRETATION: Preservation of lung function and effective immune clearance may be enhanced by selectively controlling inflammation. Discovery of this IL-1ß regulatory mechanism provides a distinct target for anti-inflammatory therapeutics, such as matrix metalloprotease inhibitors that inhibit LasB and limit inflammation and pathology during P. aeruginosa pulmonary infections. FUNDING: Full details are provided in the Acknowledgements section.


Asunto(s)
Interacciones Huésped-Patógeno , Interleucina-1beta/metabolismo , Pseudomonas aeruginosa/enzimología , Serina Endopeptidasas/metabolismo , Animales , Biomarcadores , Fibrosis Quística/complicaciones , Fibrosis Quística/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática , Inmunohistoquímica , Inflamasomas/metabolismo , Mediadores de Inflamación , Metaloproteasas/antagonistas & inhibidores , Ratones , Ratones Noqueados , Modelos Biológicos , Neumonía Bacteriana/etiología , Neumonía Bacteriana/metabolismo , Neumonía Bacteriana/patología , Unión Proteica , Infecciones por Pseudomonas/etiología , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/patología
20.
Methods Mol Biol ; 501: 253-65, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19066826

RESUMEN

The fate of lysogens following prophage induction has assumed added significance with the finding that in many pathogens virulence genes are carried on prophages and, in some, the production and/or release of the virulence factor is under control of the phage lytic regulatory program. We outline a method for identifying and characterizing from a total lysogen population, the subpopulation in which the prophage is induced. The prophage is genetically altered so that on induction it does not go through the lytic pathway, but does express a resolvase that acts at a reporter cassette located at another site on the bacterial chromosome to irreversibly change the resistance of the bacterium from tetracycline to chloramphenicol. Thus, induced derivatives survive and are easily identified even if they make up a small fraction of the population.


Asunto(s)
Lisogenia/genética , Profagos/genética , Bacteriófagos/genética , ADN Viral/genética , ADN Viral/aislamiento & purificación , Regulación Viral de la Expresión Génica , Modelos Biológicos , Reacción en Cadena de la Polimerasa , Recombinasas/genética , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA