Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
mBio ; 12(1)2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33622728

RESUMEN

Severe infections caused by multidrug-resistant Klebsiella pneumoniae sequence type 258 (ST258) highlight the need for new therapeutics with activity against this pathogen. Bacteriophage (phage) therapy is an alternative treatment approach for multidrug-resistant bacterial infections that has shown efficacy in experimental animal models and promise in clinical case reports. In this study, we assessed microbiologic, histopathologic, and survival outcomes following systemic administration of phage in ST258-infected mice. We found that prompt treatment with two phages, either individually or in combination, rescued mice with K. pneumoniae ST258 bacteremia. Among the three treatment groups, mice that received combination phage therapy demonstrated the greatest increase in survival and the lowest frequency of phage resistance among bacteria recovered from mouse blood and tissue. Our findings support the utility of phage therapy as an approach for refractory ST258 infections and underscore the potential of this treatment modality to be enhanced through strategic phage selection.IMPORTANCE Infections caused by multidrug-resistant K. pneumoniae pose a serious threat to at-risk patients and present a therapeutic challenge for clinicians. Bacteriophage (phage) therapy is an alternative treatment approach that has been associated with positive clinical outcomes when administered experimentally to patients with refractory bacterial infections. Inasmuch as these experimental treatments are prepared for individual patients and authorized for compassionate use only, they lack the rigor of a clinical trial and therefore cannot provide proof of efficacy. Here, we demonstrate that administration of viable phage provides effective treatment for multidrug-resistant K. pneumoniae (sequence type 258 [ST258]) bacteremia in a murine infection model. Moreover, we compare outcomes among three distinct phage treatment groups and identify potential correlates of therapeutic phage efficacy. These findings constitute an important first step toward optimizing and assessing phage therapy's potential for the treatment of severe ST258 infection in humans.


Asunto(s)
Antibacterianos/uso terapéutico , Bacteriófagos/fisiología , Infecciones por Klebsiella/terapia , Terapia de Fagos , Animales , Antibacterianos/farmacología , Bacteriemia/terapia , Farmacorresistencia Bacteriana Múltiple , Femenino , Infecciones por Klebsiella/sangre , Klebsiella pneumoniae/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL
2.
mBio ; 10(6)2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31848292

RESUMEN

Klebsiella pneumoniae is a human gut communal organism and notorious opportunistic pathogen. The relative high burden of asymptomatic colonization by K. pneumoniae is often compounded by multidrug resistance-a potential problem for individuals with significant comorbidities or other risk factors for infection. A carbapenem-resistant K. pneumoniae strain classified as multilocus sequence type 258 (ST258) is widespread in the United States and is usually multidrug resistant. Thus, treatment of ST258 infections is often difficult. Inasmuch as new preventive and/or therapeutic measures are needed for treatment of such infections, we developed an ST258 pneumonia model in cynomolgus macaques and tested the ability of an ST258 capsule polysaccharide type 2 (CPS2) vaccine to moderate disease severity. Compared with sham-vaccinated animals, those vaccinated with ST258 CPS2 had significantly less disease as assessed by radiography 24 h after intrabronchial installation of 108 CFU of ST258. All macaques vaccinated with CPS2 ultimately developed ST258-specific antibodies that significantly enhanced serum bactericidal activity and killing of ST258 by macaque neutrophils ex vivo Consistent with a protective immune response to CPS2, transcripts encoding inflammatory mediators were increased in infected lung tissues obtained from CPS-vaccinated animals compared with control, sham-vaccinated macaques. Taken together, our data provide support for the idea that vaccination with ST258 CPS can be used to prevent or moderate infections caused by ST258. As with studies performed decades earlier, we propose that this prime-boost vaccination approach can be extended to include multiple capsule types.IMPORTANCE Multidrug-resistant bacteria continue to be a major problem worldwide, especially among individuals with significant comorbidities and other risk factors for infection. K. pneumoniae is among the leading causes of health care-associated infections, and the organism is often resistant to multiple classes of antibiotics. A carbapenem-resistant K. pneumoniae strain known as multilocus sequence type 258 (ST258) is the predominant carbapenem-resistant Enterobacteriaceae in the health care setting in the United States. Infections caused by ST258 are often difficult to treat and new prophylactic measures and therapeutic approaches are needed. To that end, we developed a lower respiratory tract infection model in cynomolgus macaques in which to test the ability of ST258 CPS to protect against severe ST258 infection.


Asunto(s)
Vacunas Bacterianas/inmunología , Farmacorresistencia Bacteriana Múltiple , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/inmunología , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/prevención & control , Animales , Biopsia , Inmunización , Infecciones por Klebsiella/diagnóstico , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/prevención & control , Primates , Radiografía , Infecciones del Sistema Respiratorio/diagnóstico , Transcriptoma , Vacunación
3.
PLoS One ; 11(6): e0158293, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27336691

RESUMEN

Staphylococcus aureus produces numerous factors that facilitate survival in the human host. S. aureus coagulase (Coa) and von Willebrand factor-binding protein (vWbp) are known to clot plasma through activation of prothrombin and conversion of fibrinogen to fibrin. In addition, S. aureus clumping factor A (ClfA) binds fibrinogen and contributes to platelet aggregation via a fibrinogen- or complement-dependent mechanism. Here, we evaluated the contribution of Coa, vWbp and ClfA to S. aureus pathogenesis in a rabbit model of skin and soft tissue infection. Compared to skin abscesses caused by the Newman wild-type strain, those caused by isogenic coa, vwb, or clfA deletion strains, or a strain deficient in coa and vwb, were significantly smaller following subcutaneous inoculation in rabbits. Unexpectedly, we found that fibrin deposition and abscess capsule formation appear to be independent of S. aureus coagulase activity in the rabbit infection model. Similarities notwithstanding, S. aureus strains deficient in coa and vwb elicited reduced levels of several proinflammatory molecules in human blood in vitro. Although a specific mechanism remains to be determined, we conclude that S. aureus Coa, vWbp and ClfA contribute to abscess formation in rabbits.


Asunto(s)
Absceso/microbiología , Coagulasa/metabolismo , Infecciones de los Tejidos Blandos/microbiología , Infecciones Cutáneas Estafilocócicas/microbiología , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Absceso/patología , Animales , Proteínas Bacterianas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Conejos , Infecciones de los Tejidos Blandos/patología , Infecciones Cutáneas Estafilocócicas/patología
4.
J Infect Dis ; 212 Suppl 2: S404-9, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26022440

RESUMEN

Stat1(-/-) mice lack a response to interferon α, ß, and γ, allowing for replication of nonadapted wild-type (wt) Ebolavirus and Marburgvirus. We sought to establish a mouse model for efficacy testing of live attenuated recombinant vesicular stomatitis virus (rVSV)-based filovirus vaccine vectors using wt Ebolavirus and Marburgvirus challenge strains. While infection of immunocompetent mice with different rVSV-based filovirus vectors did not cause disease, infection of Stat1(-/-) mice with the same vectors resulted in systemic infection and lethal outcome for the majority of tested rVSVs. Despite differences in viral loads, organ tropism was remarkably similar between rVSV filovirus vaccine vectors and rVSVwt, with the exception of the brain. In conclusion, Stat1(-/-) mice are not an appropriate immunocompromised mouse model for efficacy testing of live attenuated, replication-competent rVSV vaccine vectors.


Asunto(s)
Filoviridae/inmunología , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/genética , Vacunas Atenuadas/inmunología , Estomatitis Vesicular/inmunología , Vacunas Virales/inmunología , Animales , Chlorocebus aethiops , Modelos Animales de Enfermedad , Ebolavirus/inmunología , Infecciones por Filoviridae/genética , Infecciones por Filoviridae/inmunología , Infecciones por Filoviridae/virología , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Fiebre Hemorrágica Ebola/genética , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/virología , Enfermedad del Virus de Marburg/genética , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/virología , Marburgvirus/inmunología , Ratones , Factor de Transcripción STAT1/inmunología , Células Vero , Carga Viral/inmunología , Replicación Viral/genética , Replicación Viral/inmunología
5.
Vaccine ; 33(24): 2823-9, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25865472

RESUMEN

Nipah virus is a zoonotic paramyxovirus that causes severe respiratory and/or encephalitic disease in humans, often resulting in death. It is transmitted from pteropus fruit bats, which serve as the natural reservoir of the virus, and outbreaks occur on an almost annual basis in Bangladesh or India. Outbreaks are small and sporadic, and several cases of human-to-human transmission have been documented as an important feature of the epidemiology of Nipah virus disease. There are no approved countermeasures to combat infection and medical intervention is supportive. We recently generated a recombinant replication-competent vesicular stomatitis virus-based vaccine that encodes a Nipah virus glycoprotein as an antigen and is highly efficacious in the hamster model of Nipah virus disease. Herein, we show that this vaccine protects African green monkeys, a well-characterized model of Nipah virus disease, from disease one month after a single intramuscular administration of the vaccine. Vaccination resulted in a rapid and strong virus-specific immune response which inhibited virus shedding and replication. This vaccine platform provides a rapid means to afford protection from Nipah virus in an outbreak situation.


Asunto(s)
Infecciones por Henipavirus/prevención & control , Virus Nipah/inmunología , Vacunas de ADN/administración & dosificación , Vesiculovirus/inmunología , Vacunas Virales/administración & dosificación , Animales , Chlorocebus aethiops , Cricetinae , Modelos Animales de Enfermedad , Femenino , Glicoproteínas/administración & dosificación , Glicoproteínas/genética , Glicoproteínas/inmunología , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/virología , Inmunidad Celular , Masculino , Virus Nipah/genética , Virus Nipah/fisiología , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas de ADN/inmunología , Vesiculovirus/genética , Proteínas del Envoltorio Viral/inmunología , Carga Viral , Vacunas Virales/inmunología , Viremia
6.
J Virol ; 88(14): 8139-52, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24807726

RESUMEN

Live attenuated cold-adapted (ca) H5N1, H7N3, H6N1, and H9N2 influenza vaccine viruses replicated in the respiratory tract of mice and ferrets, and 2 doses of vaccines were immunogenic and protected these animals from challenge infection with homologous and heterologous wild-type (wt) viruses of the corresponding subtypes. However, when these vaccine candidates were evaluated in phase I clinical trials, there were inconsistencies between the observations in animal models and in humans. The vaccine viruses did not replicate well and immune responses were variable in humans, even though the study subjects were seronegative with respect to the vaccine viruses before vaccination. Therefore, we sought a model that would better reflect the findings in humans and evaluated African green monkeys (AGMs) as a nonhuman primate model. The distribution of sialic acid (SA) receptors in the respiratory tract of AGMs was similar to that in humans. We evaluated the replication of wt and ca viruses of avian influenza (AI) virus subtypes H5N1, H6N1, H7N3, and H9N2 in the respiratory tract of AGMs. All of the wt viruses replicated efficiently, while replication of the ca vaccine viruses was restricted to the upper respiratory tract. Interestingly, the patterns and sites of virus replication differed among the different subtypes. We also evaluated the immunogenicity and protective efficacy of H5N1, H6N1, H7N3, and H9N2 ca vaccines. Protection from wt virus challenge correlated well with the level of serum neutralizing antibodies. Immune responses were slightly better when vaccine was delivered by both intranasal and intratracheal delivery than when it was delivered intranasally by sprayer. We conclude that live attenuated pandemic influenza virus vaccines replicate similarly in AGMs and human subjects and that AGMs may be a useful model to evaluate the replication of ca vaccine candidates. Importance: Ferrets and mice are commonly used for preclinical evaluation of influenza vaccines. However, we observed significant inconsistencies between observations in humans and in these animal models. We used African green monkeys (AGMs) as a nonhuman primate (NHP) model for a comprehensive and comparative evaluation of pairs of wild-type and pandemic live attenuated influenza virus vaccines (pLAIV) representing four subtypes of avian influenza viruses and found that pLAIVs replicate similarly in AGMs and humans and that AGMs can be useful for evaluation of the protective efficacy of pLAIV.


Asunto(s)
Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Enfermedades de los Primates/prevención & control , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Chlorocebus aethiops , Modelos Animales de Enfermedad , Femenino , Humanos , Vacunas contra la Influenza/administración & dosificación , Gripe Humana , Masculino , Ratones , Infecciones por Orthomyxoviridae/inmunología , Pandemias , Enfermedades de los Primates/inmunología , Sistema Respiratorio/virología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología
7.
Virulence ; 4(8): 707-15, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24104465

RESUMEN

Staphylococcus aureus community-acquired pneumonia is often associated with influenza or an influenza-like syndrome. Morbidity and mortality due to methicillin-resistant S. aureus (MRSA) or influenza and pneumonia, which includes bacterial co-infection, are among the top causes of death by infectious diseases in the United States. We developed a non-lethal influenza A virus (IAV) (H3N2)/S. aureus co-infection model in cynomolgus macaques (Macaca fascicularis) to test the hypothesis that seasonal IAV infection predisposes non-human primates to severe S. aureus pneumonia. Infection and disease progression were monitored by clinical assessment of animal health; analysis of blood chemistry, nasal swabs, and X-rays; and gross pathology and histopathology of lungs from infected animals. Seasonal IAV infection in healthy cynomolgus macaques caused mild pneumonia, but unexpectedly, did not predispose these animals to subsequent severe infection with the community-associated MRSA clone USA300. We conclude that in our co-infection model, seasonal IAV infection alone is not sufficient to promote severe S. aureus pneumonia in otherwise healthy non-human primates. The implication of these findings is that comorbidity factors in addition to IAV infection are required to predispose individuals to secondary S. aureus pneumonia.


Asunto(s)
Coinfección/microbiología , Coinfección/virología , Subtipo H3N2 del Virus de la Influenza A/crecimiento & desarrollo , Interacciones Microbianas , Infecciones por Orthomyxoviridae/complicaciones , Neumonía Estafilocócica/complicaciones , Staphylococcus aureus/crecimiento & desarrollo , Animales , Coinfección/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Pulmón/patología , Macaca fascicularis , Masculino , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/patología
8.
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
9.
Curr Top Microbiol Immunol ; 357: 243-57, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21956160

RESUMEN

Animal models of ricin toxicosis are necessary for testing the efficacy of therapeutic measures, as well studying the mechanisms by which ricin exerts its toxicity in intact animals. Because ricin can serve as a particularly well-characterized model of tissue damage, and the host response to that damage, studies of the mechanisms of ricin toxicity may have more general applicability. For example, our studies of the molecular mechanisms underlying the development of ricin-induced hypoglycemia may help elucidate the relationship of type II diabetes, insulin resistance, and inflammation. Studies in non-human primates are most relevant for testing and developing agents having clinical utility. But these animals are expensive and limited in quantity, and so rodents are used for most mechanistic studies.


Asunto(s)
Modelos Animales , Ricina/envenenamiento , Administración por Inhalación , Administración Oral , Animales , Inyecciones , Intestino Delgado/patología , Pulmón/patología , Macaca , Ratones , Ricina/administración & dosificación , Estómago/patología
10.
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
11.
Mol Cell Biochem ; 355(1-2): 75-82, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21519920

RESUMEN

Inactivation of the Staphylococcus aureus tricarboxylic acid (TCA) cycle delays the resolution of cutaneous ulcers in a mouse soft tissue infection model. In this study, it was observed that cutaneous lesions in mice infected with wild-type or isogenic aconitase mutant S. aureus strains contained comparable inflammatory infiltrates, suggesting the delayed resolution was independent of the recruitment of immune cells. These observations led us to hypothesize that staphylococcal metabolism can modulate the host immune response. Using an in vitro model system involving RAW 264.7 cells, the authors observed that cells cultured with S. aureus aconitase mutant strains produced significantly lower amounts of nitric oxide (NO(•)) and an inducible nitric oxide synthase as compared to those cells exposed to wild-type bacteria. Despite the decrease in NO(•) synthesis, the expression of antigen-presentation and costimulatory molecules was similar in cells cultured with wild-type and those cultured with aconitase mutant bacteria. The data suggest that staphylococci can evade innate immune responses and potentially enhance their ability to survive in infected hosts by altering their metabolism. This may also explain the occurrence of TCA cycle mutants in clinical S. aureus isolates.


Asunto(s)
Ciclo del Ácido Cítrico , Óxido Nítrico/metabolismo , Infecciones Cutáneas Estafilocócicas/metabolismo , Staphylococcus aureus/genética , Aconitato Hidratasa/genética , Inmunidad Adaptativa , Animales , Antígenos CD/metabolismo , Proteínas Bacterianas/genética , Línea Celular , Técnicas de Cocultivo , Expresión Génica , Técnicas de Inactivación de Genes , Antígenos de Histocompatibilidad Clase II/metabolismo , Interacciones Huésped-Patógeno , Interleucina-6/metabolismo , Ratones , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Infecciones Cutáneas Estafilocócicas/patología , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Receptor Toll-Like 2/metabolismo , Úlcera/microbiología , Úlcera/patología , Virulencia
12.
J Infect Dis ; 202(7): 1050-8, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20726702

RESUMEN

Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) infections are predominantly those affecting skin and soft tissues. Although progress has been made, our knowledge of the molecules that contribute to the pathogenesis of CA-MRSA skin infections is incomplete. We tested the hypothesis that alpha-hemolysin (Hla) contributes to the severity of USA300 skin infections in mice and determined whether vaccination against Hla reduces disease severity. Isogenic hla-negative (Deltahla) strains caused skin lesions in a mouse infection model that were significantly smaller than those caused by wild-type USA300 and Newman strains. Moreover, infection due to wild-type strains produced dermonecrotic skin lesions, whereas there was little or no dermonecrosis in mice infected with Deltahla strains. Passive immunization with Hla-specific antisera or active immunization with a nontoxigenic form of Hla significantly reduced the size of skin lesions caused by USA300 and prevented dermonecrosis. We conclude that Hla is a potential target for therapeutics or vaccines designed to moderate severe S. aureus skin infections.


Asunto(s)
Toxinas Bacterianas/antagonistas & inhibidores , Proteínas Hemolisinas/antagonistas & inhibidores , Inmunización Pasiva/métodos , Inmunización/métodos , Infecciones Cutáneas Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Proteínas Hemolisinas/deficiencia , Humanos , Ratones , Ratones Endogámicos BALB C , Piel/patología , Infecciones Cutáneas Estafilocócicas/microbiología , Staphylococcus aureus/patogenicidad
13.
Infect Immun ; 78(9): 3660-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20547745

RESUMEN

Chlamydia trachomatis is a human pathogen of global importance. An obstacle to studying the pathophysiology of human chlamydial disease is the lack of a suitable murine model of C. trachomatis infection. Mice are less susceptible to infection with human isolates due in part to innate mouse-specific host defense mechanisms to which human strains are sensitive. Another possible factor that influences the susceptibility of mice to infection is that human isolates are commonly cultivated in vitro prior to infection of mice; therefore, virulence genes could be lost as a consequence of negative selective pressure. We tested this hypothesis by infecting innate immunity-deficient C3H/HeJ female mice intravaginally with a human serovar D urogenital isolate that had undergone multiple in vitro passages. We observed early and late infection clearance phenotypes. Strains of each phenotype were isolated and then used to reinfect naïve mice. Following infection, the late-clearance strain was significantly more virulent. It caused unvarying infections of much longer durations with greater infectious burdens that naturally ascended to the upper genital tract, causing salpingitis. Despite contrasting in vivo virulence characteristics, the strains exhibited no differences in the results of in vitro infectivity assays or sensitivities to gamma interferon. Genome sequencing of the strains revealed mutations that localized to a single gene (CT135), implicating it as a critical virulence factor. Mutations in CT135 were not unique to serovar D but were also found in multiple oculogenital reference strains. Our findings provide new information about the pathogenomics of chlamydial infection and insights for improving murine models of infection using human strains.


Asunto(s)
Infecciones por Chlamydia/etiología , Chlamydia trachomatis/patogenicidad , Mutación del Sistema de Lectura , Enfermedades de los Genitales Femeninos/etiología , Factores de Virulencia/genética , Animales , Secuencia de Bases , Infecciones por Chlamydia/patología , Chlamydia trachomatis/genética , Femenino , Enfermedades de los Genitales Femeninos/patología , Ratones , Ratones Endogámicos C3H , Datos de Secuencia Molecular , Fenotipo , Polimorfismo Genético
14.
Proc Natl Acad Sci U S A ; 107(12): 5587-92, 2010 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-20231457

RESUMEN

Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) is epidemic in the United States, even rivaling HIV/AIDS in its public health impact. The pandemic clone USA300, like other CA-MRSA strains, expresses Panton-Valentine leukocidin (PVL), a pore-forming toxin that targets polymorphonuclear leukocytes (PMNs). PVL is thought to play a key role in the pathogenesis of necrotizing pneumonia, but data from rodent infection models are inconclusive. Rodent PMNs are less susceptible than human PMNs to PVL-induced cytolysis, whereas rabbit PMNs, like those of humans, are highly susceptible to PVL-induced cytolysis. This difference in target cell susceptibility could affect results of experimental models. Therefore, we developed a rabbit model of necrotizing pneumonia to compare the virulence of a USA300 wild-type strain with that of isogenic PVL-deletion mutant and -complemented strains. PVL enhanced the capacity of USA300 to cause severe lung necrosis, pulmonary edema, alveolar hemorrhage, hemoptysis, and death, hallmark clinical features of fatal human necrotizing pneumonia. Purified PVL instilled directly into the lung caused lung inflammation and injury by recruiting and lysing PMNs, which damage the lung by releasing cytotoxic granule contents. These findings provide insights into the mechanism of PVL-induced lung injury and inflammation and demonstrate the utility of the rabbit for studying PVL-mediated pathogenesis.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Toxinas Bacterianas/toxicidad , Exotoxinas/toxicidad , Leucocidinas/toxicidad , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Neumonía Estafilocócica/etiología , Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/patología , Animales , Toxinas Bacterianas/genética , Modelos Animales de Enfermedad , Exotoxinas/genética , Eliminación de Gen , Genes Bacterianos , Prueba de Complementación Genética , Humanos , Técnicas In Vitro , Leucocidinas/genética , Staphylococcus aureus Resistente a Meticilina/genética , Neutrófilos/patología , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/patología , Conejos , Virulencia/genética
15.
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
16.
Am J Pathol ; 176(3): 1346-54, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20093487

RESUMEN

Panton-Valentine leukocidin (PVL) is a two-component cytolytic toxin epidemiologically linked to community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) infections, including serious invasive infections caused by the epidemic clone referred to as strain USA300. Although PVL has long been known to be a S. aureus virulence molecule in vitro, the relative contribution of this leukotoxin to invasive CA-MRSA infections such as pneumonia remains controversial. We developed a nonhuman primate model of CA-MRSA pneumonia and used it to test the hypothesis that PVL contributes to lower respiratory tract infections caused by S. aureus strain USA300. The lower respiratory tract disease observed in this monkey model mimicked the clinical and pathological features of early mild to moderate S. aureus pneumonia in humans, including fine-structure histopathology. In this experiment using a large sample of monkeys and multiple time points of examination, no involvement of PVL in virulence could be detected. Compared with the wild-type parental USA300 strain, the isogenic PVL deletion-mutant strain caused equivalent lower respiratory tract pathology. We conclude that PVL does not contribute to lower respiratory tract infection in this nonhuman primate model of human CA-MRSA pneumonia.


Asunto(s)
Toxinas Bacterianas/toxicidad , Exotoxinas/toxicidad , Leucocidinas/toxicidad , Infecciones del Sistema Respiratorio/complicaciones , Infecciones del Sistema Respiratorio/microbiología , Infecciones Estafilocócicas/complicaciones , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/química , Proteínas de Fase Aguda/metabolismo , Animales , Citocinas/sangre , Pulmón/efectos de los fármacos , Pulmón/microbiología , Pulmón/patología , Macaca fascicularis/microbiología , Masculino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/fisiología , Técnicas Microbiológicas , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/microbiología , Arteria Pulmonar/patología , Infecciones del Sistema Respiratorio/sangre , Infecciones del Sistema Respiratorio/patología , Infecciones Estafilocócicas/sangre , Infecciones Estafilocócicas/patología , Staphylococcus aureus/efectos de los fármacos
17.
J Infect Dis ; 200(5): 724-34, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19604047

RESUMEN

sThe role of Panton-Valentine leukocidin (PVL) in Staphylococcus aureus pathogenesis is controversial. Here, we show that an unintended point mutation in the agr P2 promoter of S. aureus caused the phenotypes in gene regulation and murine pneumonia attributed to PVL by earlier investigators. In agreement with other studies that failed to detect similar effects of PVL using community-associated methicillin-resistant S. aureus strains, we found no significant effect of PVL on gene expression or pathogenesis after we repaired the mutation. These findings provide further evidence that PVL does not have a major impact on S. aureus pathogenesis. Moreover, our results demonstrate that a single nucleotide polymorphism in an intergenic region can dramatically affect bacterial physiology and virulence. Finally, our work emphasizes the need to frequently evaluate the integrity of the S. aureus agr locus.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Toxinas Bacterianas/biosíntesis , Exotoxinas/biosíntesis , Leucocidinas/biosíntesis , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Neumonía Estafilocócica/microbiología , Mutación Puntual , Regiones Promotoras Genéticas , Transactivadores/biosíntesis , Factores de Virulencia/biosíntesis , Animales , Secuencia de Bases , Peso Corporal , Infecciones Comunitarias Adquiridas/microbiología , Regulación Bacteriana de la Expresión Génica , Humanos , Staphylococcus aureus Resistente a Meticilina/fisiología , Ratones , Datos de Secuencia Molecular , Virulencia
18.
J Infect Dis ; 194(12): 1761-70, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17109350

RESUMEN

Methicillin-resistant Staphylococcus aureus (MRSA) remains a major problem in hospitals, and it is now spreading in the community. A single toxin, Panton-Valentine leukocidin (PVL), has been linked by epidemiological studies to community-associated MRSA (CA-MRSA) disease. However, the role that PVL plays in the pathogenesis of CA-MRSA has not been tested directly. To that end, we used mouse infection models to compare the virulence of PVL-positive with that of PVL-negative CA-MRSA representing the leading disease-causing strains. Unexpectedly, strains lacking PVL were as virulent in mouse sepsis and abscess models as those containing the leukotoxin. Isogenic PVL-negative (lukS/F-PV knockout) strains of USA300 and USA400 were as lethal as wild-type strains in a sepsis model, and they caused comparable skin disease. Moreover, lysis of human neutrophils and pathogen survival after phagocytosis were similar between wild-type and mutant strains. Although the toxin may be a highly linked epidemiological marker for CA-MRSA strains, we conclude that PVL is not the major virulence determinant of CA-MRSA.


Asunto(s)
Exotoxinas/fisiología , Leucocidinas/fisiología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/patogenicidad , Absceso/microbiología , Animales , Animales no Consanguíneos , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Exotoxinas/genética , Exotoxinas/metabolismo , Femenino , Humanos , Inmunocompetencia , Leucocidinas/genética , Leucocidinas/metabolismo , Leucocitos Mononucleares/microbiología , Meticilina/farmacología , Resistencia a la Meticilina , Ratones , Ratones Pelados , Neutrófilos/microbiología , Mutación Puntual , Sepsis/microbiología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/metabolismo , Virulencia
19.
Am J Pathol ; 169(3): 927-42, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16936267

RESUMEN

Molecular mechanisms mediating group A Streptococcus (GAS)-host interactions remain poorly understood but are crucial for diagnostic, therapeutic, and vaccine development. An optimized high-density microarray was used to analyze the transcriptome of GAS during experimental mouse soft tissue infection. The transcriptome of a wild-type serotype M1 GAS strain and an isogenic transcriptional regulator knockout mutant (covR) also were compared. Array datasets were verified by quantitative real-time reverse transcriptase-polymerase chain reaction and in situ immunohistochemistry. The results unambiguously demonstrate that coordinated expression of proven and putative GAS virulence factors is directed toward overwhelming innate host defenses leading to severe cellular damage. We also identified adaptive metabolic responses triggered by nutrient signals and hypoxic/acidic conditions in the host, likely facilitating pathogen persistence and proliferation in soft tissues. Key discoveries included that oxidative stress genes, virulence genes, genes related to amino acid and maltodextrin utilization, and several two-component transcriptional regulators were highly expressed in vivo. This study is the first global analysis of the GAS transcriptome during invasive infection. Coupled with parallel analysis of the covR mutant strain, novel insights have been made into the regulation of GAS virulence in vivo, resulting in new avenues for targeted therapeutic and vaccine research.


Asunto(s)
Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica/genética , Proteínas Represoras/genética , Infecciones de los Tejidos Blandos/genética , Infecciones Estreptocócicas/genética , Streptococcus pyogenes/genética , Factores de Virulencia/biosíntesis , Animales , Perfilación de la Expresión Génica , Inmunidad Innata/genética , Ratones , Estrés Oxidativo/genética , Infecciones de los Tejidos Blandos/metabolismo , Infecciones de los Tejidos Blandos/microbiología , Infecciones de los Tejidos Blandos/patología , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/patología , Infecciones Estreptocócicas/prevención & control , Vacunas Estreptocócicas/genética , Streptococcus pyogenes/patogenicidad , Transcripción Genética/genética , Vacunación
20.
J Immunol ; 175(6): 3907-19, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16148137

RESUMEN

Polymorphonuclear leukocytes (PMNs, or neutrophils) are critical for human innate immunity and kill most invading bacteria. However, pathogens such as Staphylococcus aureus avoid destruction by PMNs to survive, thereby causing human infections. The molecular mechanisms used by pathogens to circumvent killing by the immune system remain largely undefined. To that end, we studied S. aureus pathogenesis and bacteria-PMN interactions using strains originally isolated from individuals with community-acquired (CA) and hospital-acquired infections. Compared with strains from hospital infections (COL and MRSA252), strain MW2 and a methicillin-susceptible relative, MnCop, were significantly more virulent in a mouse model of S. aureus infection, and caused the greatest level of pathology in major vital organs. Although phagocytosis of each strain triggered production of reactive oxygen species and granule-phagosome fusion, those from CA infections were significantly more resistant to killing by human PMNs and caused greater host cell lysis. Microarray analysis of the strains during neutrophil phagocytosis identified genes comprising a global S. aureus response to human innate host defense. Genes involved in capsule synthesis, gene regulation, oxidative stress, and virulence, were up-regulated following ingestion of the pathogen. Notably, phagocytosis of strains from CA infections induced changes in gene expression not observed in the other strains, including up-regulation of genes encoding virulence factors and hypothetical proteins. Our studies reveal a gene transcription program in a prominent human pathogen that likely contributes to evasion of innate host defense.


Asunto(s)
Neutrófilos/inmunología , Staphylococcus aureus/patogenicidad , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Humanos , Inmunidad Innata , Fagocitosis , Infecciones Estafilocócicas/etiología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/transmisión , Staphylococcus aureus/genética , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...