Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Immunol Rev ; 314(1): 210-228, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36345955

RESUMEN

Neutrophils or polymorphonuclear neutrophils (PMNs) are an important component of innate host defense. These phagocytic leukocytes are recruited to infected tissues and kill invading microbes. There are several general characteristics of neutrophils that make them highly effective as antimicrobial cells. First, there is tremendous daily production and turnover of granulocytes in healthy adults-typically 1011 per day. The vast majority (~95%) of these cells are neutrophils. In addition, neutrophils are mobilized rapidly in response to chemotactic factors and are among the first leukocytes recruited to infected tissues. Most notably, neutrophils contain and/or produce an abundance of antimicrobial molecules. Many of these antimicrobial molecules are toxic to host cells and can destroy host tissues. Thus, neutrophil activation and turnover are highly regulated processes. To that end, aged neutrophils undergo apoptosis constitutively, a process that contains antimicrobial function and proinflammatory capacity. Importantly, apoptosis facilitates nonphlogistic turnover of neutrophils and removal by macrophages. This homeostatic process is altered by interaction with microbes and their products, as well as host proinflammatory molecules. Microbial pathogens can delay neutrophil apoptosis, accelerate apoptosis following phagocytosis, or cause neutrophil cytolysis. Here, we review these processes and provide perspective on recent studies that have potential to impact this paradigm.


Asunto(s)
Antiinfecciosos , Neutrófilos , Humanos , Anciano , Neutrófilos/fisiología , Fagocitosis , Apoptosis , Muerte Celular
2.
J Surg Res ; 283: 428-437, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36434839

RESUMEN

INTRODUCTION: The use of prosthetic mesh in hernia repair provides a powerful tool to increase repair longevity, decrease recurrence rates, and facilitate complex abdominal wall reconstruction. Overall infection rates with mesh are low, but for those affected there is high morbidity and economic cost. The availability of a practicable small animal model would be advantageous for the preclinical testing of prophylactics, therapeutics, and new biomaterials. To this end, we have developed a novel mouse model for implantation of methicillin-resistant Staphylococcus aureus-infected surgical mesh and provide results from antibiotic and immunotherapeutic testing. MATERIALS AND METHODS: Implantation of surgical mesh between fascial planes of the mouse hind limb was used to approximate hernia repair in humans. Surgical mesh was inoculated with methicillin-resistant Staphylococcus aureus to test the efficacy of antibiotic therapy with daptomycin and/or immunotherapy to induce macrophage phagocytosis using antibody blockade of the CD47 "don't eat me" molecule. Clinical outcomes were assessed by daily ambulation scores of the animals and by enumeration of mesh-associated bacteria at predetermined end points. RESULTS: A single prophylactic treatment with daptomycin at the time of surgery led to improved ambulation scores and undetectable levels of bacteria in seven of eight mice by 21 days postinfection. Anti-CD47, an activator of macrophage phagocytosis, was ineffective when administered alone or in combination with daptomycin treatment. Ten days of daily antibiotic therapy begun 3 days after infection was ineffective at clearing infection. CONCLUSIONS: This fast and simple model allows rapid in vivo testing of novel antimicrobials and immunomodulators to treat surgical implant infections.


Asunto(s)
Daptomicina , Hernia Ventral , Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Humanos , Animales , Ratones , Mallas Quirúrgicas , Infecciones Estafilocócicas/microbiología , Antibacterianos/uso terapéutico , Modelos Animales de Enfermedad , Herniorrafia/métodos , Infección de la Herida Quirúrgica/prevención & control , Hernia Ventral/cirugía
3.
Artículo en Inglés | MEDLINE | ID: mdl-28115349

RESUMEN

Klebsiella pneumoniae is a prominent cause of nosocomial infections worldwide. Bloodstream infections caused by carbapenem-resistant K. pneumoniae, including the epidemic lineage known as multilocus sequence type 258 (ST258), are difficult to treat, and the rate of mortality from such infections is high. Thus, it is imperative that we gain a better understanding of host defense against this pathogen as a step toward developing novel therapies. Here we tested the hypothesis that the resistance of ST258 to bactericidal components of human blood, such as serum complement, is linked to virulence capacity in the context of bacteremia. There was significant variance in the survival of ST258 clinical isolates in heparinized human blood or normal human serum. The rate of survival of ST258 isolates in human blood was, in general, similar to that in normal human serum, suggesting a prominent role for complement (rather than leukocytes) in the healthy host defense against ST258 isolates and related organisms. Indeed, deposition of serum complement-the C5b to C9 (C5b-C9) membrane attack complex-onto the surface of ST258 isolates accompanied serum bactericidal activity. Human serum treated with pharmacological inhibitors of complement, depleted of antibody, or heated at 56°C for 30 min had significantly reduced or absent bactericidal activity. In contrast to heparinized blood from humans, that from BALB/c mice lacked bactericidal activity toward the ST258 isolates tested, but the virulence of these ST258 isolates in a mouse bacteremia model was inexplicably limited. Our data highlight the importance of the complement system in host defense against ST258 bacteremia, and we propose that there is the potential to enhance complement-mediated bactericidal activity using an antibody-based approach.


Asunto(s)
Antibacterianos/farmacología , Carbapenémicos/farmacología , Klebsiella pneumoniae/efectos de los fármacos , Animales , Antibacterianos/uso terapéutico , Bacteriemia/tratamiento farmacológico , Bacteriemia/microbiología , Carbapenémicos/uso terapéutico , Modelos Animales de Enfermedad , Humanos , Klebsiella pneumoniae/genética , Ratones , Ratones Endogámicos BALB C , Tipificación de Secuencias Multilocus , Virulencia/genética
4.
Proc Natl Acad Sci U S A ; 111(13): 4988-93, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24639510

RESUMEN

Infections caused by drug-resistant bacteria are a major problem worldwide. Carbapenem-resistant Klebsiella pneumoniae, most notably isolates classified as multilocus sequence type (ST) 258, have emerged as an important cause of hospital deaths. ST258 isolates are predominantly multidrug resistant, and therefore infections caused by them are difficult to treat. It is not known why the ST258 lineage is the most prevalent cause of multidrug-resistant K. pneumoniae infections in the United States and other countries. Here we tested the hypothesis that carbapenem-resistant ST258 K. pneumoniae is a single genetic clone that has disseminated worldwide. We sequenced to closure the genomes of two ST258 clinical isolates and used these genomes as references for comparative genome sequencing of 83 additional clinical isolates recovered from patients at diverse geographic locations worldwide. Phylogenetic analysis of the SNPs in the core genome of these isolates revealed that ST258 K. pneumoniae organisms are two distinct genetic clades. This unexpected finding disproves the single-clone hypothesis. Notably, genetic differentiation between the two clades results from an ∼ 215-kb region of divergence that includes genes involved in capsule polysaccharide biosynthesis. The region of divergence appears to be a hotspot for DNA recombination events, and we suggest that this region has contributed to the success of ST258 K. pneumoniae. Our findings will accelerate research on novel diagnostic, therapeutic, and vaccine strategies designed to prevent and/or treat infections caused by multidrug resistant K. pneumoniae.


Asunto(s)
Carbapenémicos/farmacología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/genética , Evolución Molecular , Klebsiella pneumoniae/clasificación , Klebsiella pneumoniae/genética , Tipificación de Secuencias Multilocus , Emparejamiento Base/genética , Secuencia de Bases , Genoma Bacteriano/genética , Geografía , Humanos , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/aislamiento & purificación , Datos de Secuencia Molecular , Filogenia , Plásmidos/genética , Polimorfismo de Nucleótido Simple/genética , Análisis de Secuencia de ADN
5.
J Infect Dis ; 213(10): 1615-22, 2016 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-26768252

RESUMEN

Carbapenem-resistant Klebsiella pneumoniae strains classified as multilocus sequence type 258 (ST258) are among the most widespread multidrug-resistant hospital-acquired pathogens. Treatment of infections caused by these organisms is difficult, and mortality is high. The basis for the success of ST258, outside of antibiotic resistance, remains incompletely determined. Here we tested the hypothesis that ST258K. pneumoniae has enhanced capacity to circumvent killing by human neutrophils, the primary cellular defense against bacterial infections. There was limited binding and uptake of ST258 by human neutrophils, and correspondingly, there was limited killing of bacteria. On the other hand, transmission electron microscopy revealed that any ingested organisms were degraded readily within neutrophil phagosomes, thus indicating that survival in the neutrophil assays is due to limited phagocytosis, rather than to microbicide resistance after uptake. Our findings suggest that enhancing neutrophil phagocytosis is a potential therapeutic approach for treatment of infection caused by carbapenem-resistant ST258K. pneumoniae.


Asunto(s)
Carbapenémicos/farmacología , Infecciones por Klebsiella/terapia , Klebsiella pneumoniae/inmunología , Neutrófilos/microbiología , Fagocitosis , Animales , Técnicas de Tipificación Bacteriana , Farmacorresistencia Bacteriana , Femenino , Humanos , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , Tipificación de Secuencias Multilocus , Neutrófilos/inmunología , Neutrófilos/metabolismo , Conejos , Especies Reactivas de Oxígeno/metabolismo
6.
Am J Pathol ; 185(6): 1518-27, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25749135

RESUMEN

Staphylococcus aureus causes many types of human infections and syndromes-most notably skin and soft tissue infections. Abscesses are a frequent manifestation of S. aureus skin and soft tissue infections and are formed, in part, to contain the nidus of infection. Polymorphonuclear leukocytes (neutrophils) are the primary cellular host defense against S. aureus infections and a major component of S. aureus abscesses. These host cells contain and produce many antimicrobial agents that are effective at killing bacteria, but can also cause non-specific damage to host tissues and contribute to the formation of abscesses. By comparison, S. aureus produces several molecules that also contribute to the formation of abscesses. Such molecules include those that recruit neutrophils, cause host cell lysis, and are involved in the formation of the fibrin capsule surrounding the abscess. Herein, we review our current knowledge of the mechanisms and processes underlying the formation of S. aureus abscesses, including the involvement of polymorphonuclear leukocytes, and provide a brief overview of therapeutic approaches.


Asunto(s)
Absceso/inmunología , Neutrófilos/inmunología , Enfermedades de la Piel/inmunología , Infecciones Estafilocócicas/inmunología , Absceso/metabolismo , Absceso/patología , Humanos , Infiltración Neutrófila , Neutrófilos/metabolismo , Neutrófilos/patología , Enfermedades de la Piel/metabolismo , Enfermedades de la Piel/patología , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/patología , Staphylococcus aureus
7.
J Immunol ; 192(10): 4709-17, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24729616

RESUMEN

Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) pose a significant threat to human health. Polymorphonuclear leukocytes (PMN) are the first responders during staphylococcal infection, but 15-50% of the initial ingested inoculum survives within the PMN phagosome and likely contributes directly or indirectly to disease pathogenesis. We hypothesize that surviving intracellular CA-MRSA undermine effective phagocyte-mediated defense by causing a decrease in macrophage uptake of PMN containing viable S. aureus and by promoting PMN lysis. In support of this hypothesis, PMN harboring viable CA-MRSA strain USA300 (PMN-SA) upregulated the "don't eat me" signal CD47, remained bound to the surface, and were inefficiently ingested by macrophages. In addition, coculture with PMN-SA altered the macrophage phenotype. Compared to macrophages fed USA300 alone, macrophages challenged with PMN-SA produced more IL-8 and less IL-1 receptor antagonist, TNF-α, activated caspase-1, and IL-1ß. Although they exhibited some features of apoptosis within 3 h following ingestion of S. aureus, including phosphatidylserine exposure and mitochondrial membrane depolarization, PMN-SA had sustained levels of proliferating cell nuclear Ag expression, absence of caspase activation, and underwent lysis within 6 h following phagocytosis. PMN lysis was dependent on receptor-interacting protein 1, suggesting that PMN-SA underwent programmed necrosis or necroptosis. These data are the first demonstration, to our knowledge, that bacteria can promote sustained expression of proliferating cell nuclear Ag and that human PMN undergo necroptosis. Together, these findings demonstrate that S. aureus surviving within PMN undermine the innate immune response and may provide insight into the pathogenesis of S. aureus disease.


Asunto(s)
Apoptosis/inmunología , Macrófagos/inmunología , Staphylococcus aureus Resistente a Meticilina/inmunología , Neutrófilos/inmunología , Antígeno CD47/inmunología , Caspasa 1/inmunología , Técnicas de Cocultivo , Femenino , Humanos , Interleucina-1beta/inmunología , Macrófagos/patología , Masculino , Necrosis/inmunología , Necrosis/patología , Neutrófilos/patología , Factor de Necrosis Tumoral alfa/inmunología
8.
J Immunol ; 191(12): 6022-9, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24190656

RESUMEN

Staphylococcus aureus secretes numerous virulence factors that facilitate evasion of the host immune system. Among these molecules are pore-forming cytolytic toxins, including Panton-Valentine leukocidin (PVL), leukotoxin GH (LukGH; also known as LukAB), leukotoxin DE, and γ-hemolysin. PVL and LukGH have potent cytolytic activity in vitro, and both toxins are proinflammatory in vivo. Although progress has been made toward elucidating the role of these toxins in S. aureus virulence, our understanding of the mechanisms that underlie the proinflammatory capacity of these toxins, as well as the associated host response toward them, is incomplete. To address this deficiency in knowledge, we assessed the ability of LukGH to prime human PMNs for enhanced bactericidal activity and further investigated the impact of the toxin on neutrophil function. We found that, unlike PVL, LukGH did not prime human neutrophils for increased production of reactive oxygen species nor did it enhance binding and/or uptake of S. aureus. Unexpectedly, LukGH promoted the release of neutrophil extracellular traps (NETs), which, in turn, ensnared but did not kill S. aureus. Furthermore, we found that electropermeabilization of human neutrophils, used as a separate means to create pores in the neutrophil plasma membrane, similarly induced formation of NETs, a finding consistent with the notion that NETs can form during nonspecific cytolysis. We propose that the ability of LukGH to promote formation of NETs contributes to the inflammatory response and host defense against S. aureus infection.


Asunto(s)
Proteínas Bacterianas/farmacología , Toxinas Bacterianas/farmacología , Leucocidinas/farmacología , Neutrófilos/inmunología , Staphylococcus aureus/patogenicidad , Proteínas Bacterianas/aislamiento & purificación , Toxinas Bacterianas/aislamiento & purificación , Permeabilidad de la Membrana Celular/efectos de los fármacos , Relación Dosis-Respuesta Inmunológica , Electroporación , Exocitosis/efectos de los fármacos , Espacio Extracelular , Humanos , Leucocidinas/aislamiento & purificación , Neutrófilos/efectos de los fármacos , Neutrófilos/ultraestructura , Proteínas Opsoninas/inmunología , Peroxidasa/metabolismo , Estallido Respiratorio/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/química , Staphylococcus aureus/inmunología , Superóxidos/metabolismo , Virulencia
10.
Proc Natl Acad Sci U S A ; 108(44): 18091-6, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22025717

RESUMEN

Staphylococcus aureus is a bacterial pathogen known to cause infections in epidemic waves. One such epidemic was caused by a clone known as phage-type 80/81, a penicillin-resistant strain that rose to world prominence in the late 1950s. The molecular underpinnings of the phage-type 80/81 outbreak have remained unknown for decades, nor is it understood why related S. aureus clones became epidemic in hospitals in the early 1990s. To better understand the molecular basis of these epidemics, we sequenced the genomes of eight S. aureus clinical isolates representative of the phage-type 80/81 clone, the Southwest Pacific clone [a community-associated methicillin-resistant S. aureus (MRSA) clone], and contemporary S. aureus clones, all of which are genetically related and belong to the same clonal complex (CC30). Genome sequence analysis revealed that there was coincident divergence of these clones from a recent common ancestor, a finding that resolves controversy about the evolutionary history of the lineage. Notably, we identified nonsynonymous SNPs in genes encoding accessory gene regulator C (agrC) and α-hemolysin (hla)--molecules important for S. aureus virulence--that were present in virtually all contemporary CC30 hospital isolates tested. Compared with the phage-type 80/81 and Southwest Pacific clones, contemporary CC30 hospital isolates had reduced virulence in mouse infection models, the result of SNPs in agrC and hla. We conclude that agr and hla (along with penicillin resistance) were essential for world dominance of phage-type 80/81 S. aureus, whereas key SNPs in contemporary CC30 clones restrict these pathogens to hospital settings in which the host is typically compromised.


Asunto(s)
Bacteriófagos/clasificación , Infecciones Estafilocócicas/epidemiología , Staphylococcus aureus/virología , Bacteriófagos/genética , Brotes de Enfermedades , Genoma Bacteriano , Genoma Viral , Humanos , Mutación , Filogenia , Polimorfismo de Nucleótido Simple , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Virulencia
11.
Proc Natl Acad Sci U S A ; 107(10): 4693-8, 2010 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-20179180

RESUMEN

Relatively little is understood about the dynamics of global host-pathogen transcriptome changes that occur during bacterial infection of mucosal surfaces. To test the hypothesis that group A Streptococcus (GAS) infection of the oropharynx provokes a distinct host transcriptome response, we performed genome-wide transcriptome analysis using a nonhuman primate model of experimental pharyngitis. We also identified host and pathogen biological processes and individual host and pathogen gene pairs with correlated patterns of expression, suggesting interaction. For this study, 509 host genes and seven biological pathways were differentially expressed throughout the entire 32-day infection cycle. GAS infection produced an initial widespread significant decrease in expression of many host genes, including those involved in cytokine production, vesicle formation, metabolism, and signal transduction. This repression lasted until day 4, at which time a large increase in expression of host genes was observed, including those involved in protein translation, antigen presentation, and GTP-mediated signaling. The interactome analysis identified 73 host and pathogen gene pairs with correlated expression levels. We discovered significant correlations between transcripts of GAS genes involved in hyaluronic capsule production and host endocytic vesicle formation, GAS GTPases and host fibrinolytic genes, and GAS response to interaction with neutrophils. We also identified a strong signal, suggesting interaction between host gammadelta T cells and genes in the GAS mevalonic acid synthesis pathway responsible for production of isopentenyl-pyrophosphate, a short-chain phospholipid that stimulates these T cells. Taken together, our results are unique in providing a comprehensive understanding of the host-pathogen interactome during mucosal infection by a bacterial pathogen.


Asunto(s)
Perfilación de la Expresión Génica , Macaca fascicularis/genética , Faringe/metabolismo , Streptococcus pyogenes/genética , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Vesículas Cubiertas por Clatrina/metabolismo , Citocinas/metabolismo , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Interacciones Huésped-Patógeno , Ácido Hialurónico/metabolismo , Macaca fascicularis/metabolismo , Macaca fascicularis/microbiología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Neutrófilos/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Faringitis/genética , Faringitis/microbiología , Faringe/microbiología , Faringe/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/fisiología
12.
J Infect Dis ; 206(8): 1185-93, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22872735

RESUMEN

BACKGROUND: Staphylococcus aureus produces numerous molecules that facilitate survival in the host. We recently identified a novel S. aureus leukotoxin (leukotoxin GH [LukGH]) using proteomics, but its role in virulence remains unclear. Here we investigated the role of LukGH in vivo. METHODS: We tested cytotoxicity of LukGH toward polymorphonuclear leukocytes (PMNs) from mice, rabbits, monkeys, and humans. LukGH was administered to mice, rabbits, and a cynomolgus monkey by subcutaneous or intradermal injection to assess cytotoxicity or host response in vivo. The effects of LukGH in vivo were compared with those of Panton-Valentine leukocidin (PVL), a well-characterized S. aureus leukotoxin. The contribution of LukGH to S. aureus infection was tested using mouse and rabbit infection models. RESULTS: Susceptibility of PMNs to LukGH was similar between humans and cynomolgus monkeys, and was greater than that of rabbits, which in turn was greater than that of mice. LukGH or PVL caused skin inflammation in rabbits and a monkey, but deletion of neither lukGH nor lukGH and lukS/F-PV reduced severity of USA300 infections in rabbits or mice. Rather, some disease parameters (eg, rabbit abscess size) were increased following infection with a lukGH and lukS/F-PV deletion strain. CONCLUSIONS: Our findings indicate that S. aureus leukotoxins enhance the host inflammatory response and influence the outcome of infection.


Asunto(s)
Exotoxinas/toxicidad , Inflamación/inducido químicamente , Staphylococcus aureus/patogenicidad , Factores de Virulencia/toxicidad , Animales , Modelos Animales de Enfermedad , Exotoxinas/administración & dosificación , Humanos , Inflamación/inmunología , Inyecciones Intradérmicas , Inyecciones Subcutáneas , Macaca fascicularis , Masculino , Ratones , Neutrófilos/inmunología , Neutrófilos/microbiología , Conejos , Infecciones Estafilocócicas/patología , Factores de Virulencia/administración & dosificación
13.
mBio ; 14(5): e0194923, 2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37671860

RESUMEN

IMPORTANCE: Klebsiella pneumoniae strains with a combination of multidrug resistance and hypervirulence genotypes (MDR hvKp) have emerged as a cause of human infections. The ability of these microbes to avoid killing by the innate immune system remains to be tested fully. To that end, we compared the ability of a global collection of hvKp and MDR hvKp clinical isolates to survive in human blood and resist phagocytic killing by human neutrophils. The two MDR hvKp clinical isolates tested (ST11 and ST147) were killed in human blood and by human neutrophils in vitro, whereas phagocytic killing of hvKp clinical isolates (ST23 and ST86) required specific antisera. Although the data were varied and often isolate specific, they are an important first step toward gaining an enhanced understanding of host defense against MDR hvKp.


Asunto(s)
Infecciones por Klebsiella , Klebsiella pneumoniae , Humanos , Virulencia/genética , Neutrófilos , Genotipo , Antibacterianos
14.
Phys Sportsmed ; 40(2): 13-21, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22759601

RESUMEN

The remarkable ability of Staphylococcus aureus to develop antibiotic resistance in conjunction with the emergence of highly virulent and/or transmissible strains has established the pathogen as a leading cause of human bacterial infections worldwide. Historically, methicillin-resistant S aureus (MRSA) was found almost exclusively in hospitals and/or health care-related facilities. However, in the late 1990s, community-associated MRSA strains emerged in the United States and rapidly became the leading cause of community-associated bacterial infections. An enhanced understanding of the pathogenesis and epidemiology of this bacterium is fundamental for the prevention and/or treatment of community-associated MRSA infections. This review highlights salient features of S aureus biology that contribute to the exceptional ability of this pathogen to cause human disease, as well as discusses, in brief, the established approaches for treatment and prevention of infection.


Asunto(s)
Atletas , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Infecciones Estafilocócicas/prevención & control , Antibacterianos/uso terapéutico , Toxinas Bacterianas/metabolismo , Infecciones Comunitarias Adquiridas/epidemiología , Infecciones Comunitarias Adquiridas/prevención & control , Drenaje , Humanos , Staphylococcus aureus Resistente a Meticilina/metabolismo , Educación del Paciente como Asunto , Medicina Deportiva , Infecciones Estafilocócicas/epidemiología , Infecciones Estafilocócicas/transmisión
15.
J Infect Dis ; 204(6): 937-41, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21849291

RESUMEN

Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) infections are frequently associated with strains harboring genes encoding Panton-Valentine leukocidin (PVL). The role of PVL in the success of the epidemic CA-MRSA strain USA300 remains unknown. Here we developed a skin and soft tissue infection model in rabbits to test the hypothesis that PVL contributes to USA300 pathogenesis and compare it with well-established virulence determinants: alpha-hemolysin (Hla), phenol-soluble modulin-alpha peptides (PSMα), and accessory gene regulator (Agr). The data indicate that Hla, PSMα, and Agr contribute to the pathogenesis of USA300 skin infections in rabbits, whereas a role for PVL could not be detected.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina/patogenicidad , Infecciones de los Tejidos Blandos/microbiología , Infecciones de los Tejidos Blandos/patología , Infecciones Cutáneas Estafilocócicas/microbiología , Infecciones Cutáneas Estafilocócicas/patología , Factores de Virulencia/metabolismo , Absceso/microbiología , Absceso/patología , Animales , Carga Bacteriana , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Modelos Animales de Enfermedad , Exotoxinas/genética , Exotoxinas/metabolismo , Femenino , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Histocitoquímica , Leucocidinas/genética , Leucocidinas/metabolismo , Microscopía , Conejos , Piel/microbiología , Piel/patología , Transactivadores/genética , Transactivadores/metabolismo , Virulencia , Factores de Virulencia/genética
16.
J Innate Immun ; 14(3): 167-181, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34628410

RESUMEN

Klebsiella pneumoniae (K. pneumoniae) is a Gram-negative commensal bacterium and opportunistic pathogen. In healthy individuals, the innate immune system is adept at protecting against K. pneumoniae infection. Notably, the serum complement system and phagocytic leukocytes (e.g., neutrophils) are highly effective at eliminating K. pneumoniae and thereby preventing severe disease. On the other hand, the microbe is a major cause of healthcare-associated infections, especially in individuals with underlying susceptibility factors, such as pre-existing severe illness or immune suppression. The burden of K. pneumoniae infections in hospitals is compounded by antibiotic resistance. Treatment of these infections is often difficult largely because the microbes are usually resistant to multiple antibiotics (multidrug resistant [MDR]). There are a limited number of treatment options for these infections and new therapies, and preventative measures are needed. Here, we review host defense against K. pneumoniae and discuss recent therapeutic measures and vaccine approaches directed to treat and prevent severe disease caused by MDR K. pneumoniae.


Asunto(s)
Infecciones por Klebsiella , Klebsiella pneumoniae , Antibacterianos/uso terapéutico , Proteínas del Sistema Complemento , Humanos , Inmunoterapia , Infecciones por Klebsiella/terapia
17.
Microbiol Spectr ; 10(6): e0151722, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36264264

RESUMEN

Carbapenem-resistant Klebsiella pneumoniae isolates classified as multilocus sequence type 258 (ST258) are a problem in health care settings in many countries globally. ST258 isolates are resistant to multiple classes of antibiotics and can cause life-threatening infections, such as pneumonia and sepsis, in susceptible individuals. Treatment strategies for such infections are limited. Understanding the response of K. pneumoniae to host factors in the presence of antibiotics could reveal mechanisms employed by the pathogen to evade killing in the susceptible host, as well as inform treatment of infections. Here, we investigated the ability of antibiotics at subinhibitory concentrations to alter K. pneumoniae capsular polysaccharide (CPS) production and survival in normal human serum (NHS). Unexpectedly, pretreatment with some of the antibiotics tested enhanced ST258 survival in NHS. For example, a subinhibitory concentration of mupirocin increased survival for 7 of 10 clinical isolates evaluated and there was increased cell-associated CPS for 3 of these isolates compared with untreated controls. Additionally, mupirocin pretreatment caused concomitant reduction in the deposition of the serum complement protein C5b-9 on the surface of these three isolates. Transcriptome analyses with a selected ST258 isolate (34446) indicated that genes implicated in the stringent response and/or serum resistance were upregulated following mupirocin treatment and/or culture in NHS. In conclusion, mupirocin and/or human serum causes changes in the K. pneumoniae transcriptome that likely contribute to the observed decrease in serum susceptibility via a multifactorial process. Whether these responses can be extended more broadly and thus impact clinical outcome in the human host merits further investigation. IMPORTANCE The extent to which commensal bacteria are altered by exposure to subinhibitory concentrations of antibiotics (outside resistance) remains incompletely determined. To gain a better understanding of this phenomenon, we tested the ability of selected antibiotics (at subinhibitory concentrations) to alter survival of ST258 clinical isolates in normal human serum. We found that exposure of ST258 to antibiotics at low concentrations differentially altered gene expression, capsule production, serum complement deposition, and bacterial survival. The findings were isolate and antibiotic dependent but provide insight into a potential confounding issue associated with ST258 infections.


Asunto(s)
Infecciones por Klebsiella , Neumonía , Sepsis , Humanos , Antibacterianos/farmacología , Antibacterianos/metabolismo , Klebsiella pneumoniae/metabolismo , Mupirocina/metabolismo , Infecciones por Klebsiella/tratamiento farmacológico , Infecciones por Klebsiella/microbiología
18.
Microbiol Spectr ; 10(2): e0271621, 2022 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-35389241

RESUMEN

Staphylococcus aureus remains a leading cause of skin and soft tissue infections (SSTIs) globally. In the United States, many of these infections are caused by isolates classified as USA300. Our understanding of the success of USA300 as a human pathogen is due in part to data obtained from animal infection models, including rabbit SSTI models. These animal models have been used to study S. aureus virulence and pathogenesis and to gain an enhanced understanding of the host response to infection. Although significant knowledge has been gained, the need to use a relatively high inoculum of USA300 (1 × 108 to 5 × 108 CFU) is a caveat of these infection models. As a step toward addressing this issue, we created mutations in USA300 that mimic those found in S. aureus strains with naturally occurring rabbit tropism-namely, single nucleotide polymorphisms in dltB and/or deletion of rot. We then developed a rabbit SSTI model that utilizes an inoculum of 106 USA300 CFU to cause reproducible disease and tested whether primary SSTI protects rabbits against severe reinfection caused by the same strain. Although there was modest protection against severe reinfection, primary infection and reinfection with rabbit-tropic USA300 strains failed to increase the overall level of circulating anti-S. aureus antibodies significantly. These findings provide additional insight into the host response to S. aureus. More work is needed to further develop a low-inoculum infection model that can be used to better test the potential of new therapeutics or vaccine target antigens. IMPORTANCE Animal models of S. aureus infection are important for evaluating bacterial pathogenesis and host immune responses. These animal infection models are often used as an initial step in the testing of vaccine antigens and new therapeutics. The extent to which animal models of S. aureus infection approximate human infections remains a significant consideration for translation of results to human clinical trials. Although significant progress has been made with rabbit models of S. aureus infection, one concern is the high inoculum needed to cause reproducible disease. Here, we generated USA300 strains that have tropism for rabbits and developed a rabbit SSTI model that uses fewer CFU than previous models.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Infecciones de los Tejidos Blandos , Infecciones Estafilocócicas , Infecciones Cutáneas Estafilocócicas , Vacunas , Animales , Staphylococcus aureus Resistente a Meticilina/genética , Conejos , Reinfección , Infecciones Estafilocócicas/microbiología , Infecciones Cutáneas Estafilocócicas/microbiología , Staphylococcus aureus , Estados Unidos
19.
Nat Microbiol ; 7(1): 62-72, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34873293

RESUMEN

Swift recruitment of phagocytic leucocytes is critical in preventing infection when bacteria breach through the protective layers of the skin. According to canonical models, this occurs via an indirect process that is initiated by contact of bacteria with resident skin cells and which is independent of the pathogenic potential of the invader. Here we describe a more rapid mechanism of leucocyte recruitment to the site of intrusion of the important skin pathogen Staphylococcus aureus that is based on direct recognition of specific bacterial toxins, the phenol-soluble modulins (PSMs), by circulating leucocytes. We used a combination of intravital imaging, ear infection and skin abscess models, and in vitro gene expression studies to demonstrate that this early recruitment was dependent on the transcription factor EGR1 and contributed to the prevention of infection. Our findings refine the classical notion of the non-specific and resident cell-dependent character of the innate immune response to bacterial infection by demonstrating a pathogen-specific high-alert mechanism involving direct recruitment of immune effector cells by secreted bacterial products.


Asunto(s)
Toxinas Bacterianas/inmunología , Linfocitos/inmunología , Infiltración Neutrófila/inmunología , Piel/inmunología , Piel/microbiología , Infecciones Cutáneas Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Femenino , Humanos , Microscopía Intravital/métodos , Ratones Endogámicos C57BL , Staphylococcus aureus/patogenicidad , Factores de Virulencia
20.
Infect Immun ; 79(1): 342-52, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20937760

RESUMEN

Staphylococcus aureus is a prominent human pathogen and a leading cause of community- and hospital-acquired bacterial infections worldwide. Herein, we describe the identification and characterization of the S. aureus 67.6-kDa hypothetical protein, named for the surface factor promoting resistance to oxidative killing (SOK) in this study. Sequence analysis showed that the SOK gene is conserved in all sequenced S. aureus strains and homologous to the myosin cross-reactive antigen of Streptococcus pyogenes. Immunoblotting and immunofluorescence analysis showed that SOK was copurified with membrane fractions and was exposed on the surface of S. aureus Newman and RN4220. Comparative analysis of wild-type S. aureus and an isogenic deletion strain indicated that SOK contributes to both resistance to killing by human neutrophils and to oxidative stress. In addition, the S. aureus sok deletion strain showed dramatically reduced aortic valve vegetation and bacterial cell number in a rabbit endocarditis model. These results, plus the suspected role of the streptococcal homologue in certain diseases such as acute rheumatic fever, suggest that SOK plays an important role in cardiovascular and other staphylococcal infections.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endocarditis Bacteriana/microbiología , Staphylococcus aureus/metabolismo , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Biología Computacional , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica/fisiología , Humanos , Neutrófilos/fisiología , Estrés Oxidativo , Polimorfismo de Longitud del Fragmento de Restricción , Conejos , Factores de Virulencia/química , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA