Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
JCI Insight ; 9(4)2024 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-38236641

RESUMO

BACKGROUNDT cell responses are impaired in Staphylococcus aureus-infected children, highlighting a potential mechanism of immune evasion. This study tested the hypotheses that toxin-specific antibodies protect immune cells from bacterial killing and are associated with improved T cell function following infection.METHODSS. aureus-infected and healthy children (N = 33 each) were prospectively enrolled. During acute infection and convalescence, we quantified toxin-specific IgG levels by ELISA, antibody function using a cell killing assay, and functional T cell responses by ELISPOT.RESULTSThere were no differences in toxin-specific IgG levels or ability to neutralize toxin-mediated immune cell killing between healthy and acutely infected children, but antibody levels and function increased following infection. Similarly, T cell function, which was impaired during acute infection, improved following infection. However, the response to infection was highly variable; up to half of children did not have improved antibody or T cell function. Serum from children with higher α-hemolysin-specific IgG levels more strongly protected immune cells against toxin-mediated killing. Importantly, children whose serum more strongly protected against toxin-mediated killing also had stronger immune responses to infection, characterized by more elicited antibodies and greater improvement in T cell function following infection.CONCLUSIONThis study demonstrates that, despite T cell impairment during acute infection, S. aureus elicits toxin-neutralizing antibodies. Individual antibody responses and T cell recovery are variable. These findings also suggest that toxin-neutralizing antibodies protect antigen-presenting cells and T cells, thereby promoting immune recovery. Finally, failure to elicit toxin-neutralizing antibodies may identify children at risk for prolonged T cell suppression.FUNDINGNIH National Institute of Allergy and Infectious Diseases R01AI125489 and Nationwide Children's Hospital.


Assuntos
Toxinas Bacterianas , Infecções Estafilocócicas , Criança , Humanos , Staphylococcus aureus , Linfócitos T , Anticorpos Antibacterianos , Infecções Estafilocócicas/microbiologia , Anticorpos Neutralizantes , Imunoglobulina G
2.
NPJ Vaccines ; 8(1): 3, 2023 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-36693884

RESUMO

Staphylococcus aureus infections are a major public health issue, and a vaccine is urgently needed. Despite a considerable promise in preclinical models, all vaccines tested thus far have failed to protect humans against S. aureus. Unlike laboratory mice, humans are exposed to S. aureus throughout life. In the current study, we hypothesized that prior exposure to S. aureus "imprints" the immune response to inhibit vaccine-mediated protection. We established a mouse model in which S. aureus skin and soft tissue infection (SSTI) is followed by vaccination and secondary SSTI. Unlike naïve mice, S. aureus-sensitized mice were incompletely protected against secondary SSTI by vaccination with the inactivated α-hemolysin (Hla) mutant HlaH35L. Inhibition of protection was specific for the HlaH35L vaccine and required hla expression during primary SSTI. Surprisingly, inhibition occurred at the level of vaccine-elicited effector T cells; hla expression during primary infection limited the expansion of T cells and dendritic cells and impaired vaccine-specific T cell responses. Importantly, the T cell-stimulating adjuvant CAF01 rescued inhibition and restored vaccine-mediated protection. Together, these findings identify a potential mechanism for the failure of translation of promising S. aureus vaccines from mouse models to clinical practice and suggest a path forward to prevent these devastating infections.

3.
mBio ; 13(5): e0147222, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36135382

RESUMO

Staphylococcus aureus is a ubiquitous Gram-positive bacterium and an opportunistic human pathogen. S. aureus pathogenesis relies on a complex network of regulatory factors that adjust gene expression. Two important factors in this network are CodY, a repressor protein responsive to nutrient availability, and the SaeRS two-component system (TCS), which responds to neutrophil-produced factors. Our previous work revealed that CodY regulates the secretion of many toxins indirectly via Sae through an unknown mechanism. We report that disruption of codY results in increased levels of phosphorylated SaeR (SaeR~P) and that codY mutant cell membranes contain a higher percentage of branched-chain fatty acids (BCFAs) than do wild-type membranes, prompting us to hypothesize that changes to membrane composition modulate the activity of the SaeS sensor kinase. Disrupting the lpdA gene encoding dihydrolipoyl dehydrogenase, which is critical for BCFA synthesis, significantly reduced the abundance of SaeR, phosphorylated SaeR, and BCFAs in the membrane, resulting in reduced toxin production and attenuated virulence. Lower SaeR levels could be explained in part by reduced stability. Sae activity in the lpdA mutant could be complemented genetically and chemically with exogenous short- or full-length BCFAs. Intriguingly, lack of lpdA also alters the activity of other TCSs, suggesting a specific BCFA requirement managing the basal activity of multiple TCSs. These results reveal a novel method of posttranscriptional virulence regulation via BCFA synthesis, potentially linking CodY activity to multiple virulence regulators in S. aureus. IMPORTANCE Two-component systems (TCSs) are an essential way that bacteria sense and respond to their environment. These systems are usually composed of a membrane-bound histidine kinase that phosphorylates a cytoplasmic response regulator. Because most of the histidine kinases are embedded in the membrane, lipids can allosterically regulate the activity of these sensors. In this study, we reveal that branched-chain fatty acids (BCFAs) are required for the activation of multiple TCSs in Staphylococcus aureus. Using both genetic and biochemical data, we show that the activity of the virulence activator SaeS and the phosphorylation of its response regulator SaeR are reduced in a branched-chain keto-acid dehydrogenase complex mutant and that defects in BCFA synthesis have far-reaching consequences for exotoxin secretion and virulence. Finally, we show that mutation of the global nutritional regulator CodY alters BCFA content in the membrane, revealing a potential mechanism of posttranscriptional regulation of the Sae system by CodY.


Assuntos
Infecções Estafilocócicas , Staphylococcus aureus , Humanos , Staphylococcus aureus/metabolismo , Regulação Bacteriana da Expressão Gênica , Histidina Quinase/metabolismo , Di-Hidrolipoamida Desidrogenase/genética , Di-Hidrolipoamida Desidrogenase/metabolismo , Histidina/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Infecções Estafilocócicas/microbiologia , Ácidos Graxos/metabolismo , Exotoxinas/metabolismo
4.
Front Immunol ; 13: 795792, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35983063

RESUMO

Infections caused by Staphylococcus aureus range from mild to severe and frequently recur. Emerging evidence suggests that the site and severity of infection drive the potency of elicited immune responses and susceptibility to recurrent infection. In this study, we used tractable mouse models of S. aureus skin infection (SSTI) and pneumonia to determine the relative magnitude of elicited protective immunity. Surprisingly, despite both SSTI and pneumonia eliciting antibody and local effector T cell responses, only SSTI elicited protective antibody and memory T cell responses and subsequent protection against secondary SSTI and pneumonia. The failure of pneumonia to elicit protective immunity was attributed to an inability of S. aureus pneumonia to elicit toxin-specific antibodies that confer protection during secondary infection and was associated with a failure to expand antigen-specific memory T cells. Taken together, these findings emphasize the importance of understanding protective immunity in the context of the tissue-specificity.


Assuntos
Infecções Estafilocócicas , Infecções Cutâneas Estafilocócicas , Animais , Camundongos , Recidiva Local de Neoplasia , Especificidade de Órgãos , Staphylococcus aureus
5.
Antimicrob Agents Chemother ; 66(4): e0227021, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35266822

RESUMO

Recurrent Staphylococcus aureus infections are common, suggesting a failure to elicit protective immunity. Given the emergence of antibiotic resistance, a vaccine is urgently needed, but there is no approved vaccine for S. aureus. While antibiotics are routinely used to treat S. aureus infections, their impact on the development of protective immunity is not understood. Using an established mouse model of S. aureus skin and soft tissue infection (SSTI), we observed that antibiotic therapy effectively resolved infection but failed to elicit protection against secondary (2°) SSTI. Key contributors to protective immunity, toxin-specific antibodies and interleukin-17A (IL-17A)-producing T cells, were not strongly elicited in antibiotic-treated mice. Delaying antibiotic treatment failed to resolve skin lesions but resulted in higher antibody levels after infection and strong protection against 2° SSTI, suggesting that the development of protective immunity requires a longer period of antigen exposure. We next investigated if combining α-hemolysin (Hla) vaccination with antibiotics during primary infection would both treat infection and generate durable protective immunity. This "therapeutic vaccination" approach resulted in rapid resolution of primary infection and protection against recurrent infection, demonstrating that concurrent vaccination could circumvent the deleterious effects of antibiotic therapy on elicited immune responses. Collectively, these findings suggest that protective immunity is thwarted by the rapid elimination of antigen during antibiotic treatment. However, vaccination in conjunction with antibiotic treatment can retain the benefits of antibiotic treatment while also establishing protective immunity.


Assuntos
Infecções dos Tecidos Moles , Infecções Estafilocócicas , Animais , Antibacterianos/uso terapêutico , Proteínas Hemolisinas , Camundongos , Infecções dos Tecidos Moles/tratamento farmacológico , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus
6.
J Infect Dis ; 225(1): 177-185, 2022 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-34145461

RESUMO

BACKGROUND: Staphylococcus aureus infections are common throughout the lifespan, with recurrent infections occurring in nearly half of infected children. There is no licensed vaccine, underscoring the need to better understand how S. aureus evades protective immunity. Despite much study, the relative contributions of antibodies and T cells to protection against S. aureus infections in humans are not fully understood. METHODS: We prospectively quantified S. aureus-specific antibody levels by ELISA and T-cell responses by ELISpot in S. aureus-infected and healthy children. RESULTS: S. aureus-specific antibody levels and T-cell responses increased with age in healthy children, suggesting a coordinated development of anti-staphylococcal immunity. Antibody levels against leukotoxin E (LukE) and Panton-Valentine leukocidin (LukS-PV), but not α-hemolysin (Hla), were higher in younger infected children, compared with healthy children; these differences disappeared in older children. We observed a striking impairment of global and S. aureus-specific T-cell function in children with invasive and noninvasive infection, suggesting that S. aureus-specific immune responses are dysregulated during childhood infection regardless of the infection phenotype. CONCLUSIONS: These findings identify a potential mechanism by which S. aureus infection actively evades adaptive immune responses, thereby preventing the development of protective immunity and maintaining susceptibility to recurrent infection.


Assuntos
Anticorpos Antibacterianos/sangue , Exotoxinas/imunologia , Leucocidinas/imunologia , Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas/epidemiologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus , Adolescente , Toxinas Bacterianas , Criança , Pré-Escolar , Ensaio de Imunoadsorção Enzimática , Feminino , Proteínas Hemolisinas/imunologia , Humanos , Lactente , Masculino , Estudos Prospectivos , Estudos Soroepidemiológicos , Linfócitos T , Adulto Jovem
7.
Front Immunol ; 12: 633672, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33692805

RESUMO

Recurrent S. aureus infections are common, suggesting that natural immune responses are not protective. All candidate vaccines tested thus far have failed to protect against S. aureus infections, highlighting an urgent need to better understand the mechanisms by which the bacterium interacts with the host immune system to evade or prevent protective immunity. Although there is evidence in murine models that both cellular and humoral immune responses are important for protection against S. aureus, human studies suggest that T cells are critical in determining susceptibility to infection. This review will use an "anatomic" approach to systematically outline the steps necessary in generating a T cell-mediated immune response against S. aureus. Through the processes of bacterial uptake by antigen presenting cells, processing and presentation of antigens to T cells, and differentiation and proliferation of memory and effector T cell subsets, the ability of S. aureus to evade or inhibit each step of the T-cell mediated response will be reviewed. We hypothesize that these interactions result in the redirection of immune responses away from protective antigens, thereby precluding the establishment of "natural" memory and potentially inhibiting the efficacy of vaccination. It is anticipated that this approach will reveal important implications for future design of vaccines to prevent these infections.


Assuntos
Desenho de Fármacos , Evasão da Resposta Imune , Memória Imunológica , Reinfecção/prevenção & controle , Infecções Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas/uso terapêutico , Staphylococcus aureus/imunologia , Linfócitos T/imunologia , Imunidade Adaptativa , Animais , Antígenos de Bactérias/imunologia , Epitopos , Humanos , Imunogenicidade da Vacina , Ativação Linfocitária , Reinfecção/imunologia , Reinfecção/microbiologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Vacinas Antiestafilocócicas/efeitos adversos , Vacinas Antiestafilocócicas/imunologia , Staphylococcus aureus/patogenicidade , Linfócitos T/microbiologia
8.
J Invest Dermatol ; 141(4): 810-820.e8, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32946878

RESUMO

The staphylococcal α-hemolysin is critical for the pathogenesis of Staphylococcus aureus skin and soft tissue infection. Vaccine and infection-elicited α-hemolysin-specific antibodies protect against S. aureus‒induced dermonecrosis, a key feature of skin and soft tissue infection. Many interactions between α-hemolysin and host cells have been identified that promote tissue damage and modulate immune responses, but the mechanisms by which protective adaptive responses cross talk with innate responses at the tissue level are not clear. Using an established mouse model of skin and soft tissue infection and a newly developed histopathologic scoring system, we observed pathologic correlates early after infection, predicting protection against dermonecrosis by anti-α-hemolysin antibody. Protection was characterized by robust neutrophilic inflammation and compartmentalization of bacteria into discrete abscesses, which led to the attenuation of dermonecrosis and enhancement of bacterial clearance later in the infection. The ultimate outcome of infection was driven by the recruitment of neutrophils within the first day after infection but not later. Antibody-mediated protection was dependent on toxin neutralization rather than on enhanced opsonophagocytic killing by neutrophils or protection against toxin-mediated neutrophil lysis. Together, these findings advance our understanding of the mechanisms by which the early synergism between antibody-mediated toxin neutralization and tissue-specific neutrophilic inflammation preserve tissue integrity during infection.


Assuntos
Anticorpos Antibacterianos/metabolismo , Anticorpos Neutralizantes/metabolismo , Toxinas Bacterianas/imunologia , Proteínas Hemolisinas/imunologia , Neutrófilos/imunologia , Pele/patologia , Infecções Cutâneas Estafilocócicas/imunologia , Animais , Anticorpos Antibacterianos/administração & dosagem , Anticorpos Antibacterianos/imunologia , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/imunologia , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Feminino , Voluntários Saudáveis , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Humanos , Imunização Passiva/métodos , Camundongos , Necrose/imunologia , Necrose/microbiologia , Necrose/patologia , Infiltração de Neutrófilos , Cultura Primária de Células , Pele/imunologia , Pele/microbiologia , Infecções Cutâneas Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/patologia , Staphylococcus aureus/imunologia
9.
Pediatrics ; 146(3)2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32759380

RESUMO

Staphylococcus aureus is a common cause of community and health care-associated bacteremia, with authors of recent studies estimating the incidence of S aureus bacteremia (SAB) in high-income countries between 8 and 26 per 100 000 children per year. Despite this, <300 children worldwide have ever been randomly assigned into clinical trials to assess the efficacy of treatment of SAB. A panel of infectious diseases physicians with clinical and research interests in pediatric SAB identified 7 key clinical questions. The available literature is systematically appraised, summarizing SAB management in children in relation to these priority clinical questions. The management of neonates, children, and adolescents with SAB is predominantly based on clinical experience and trial data extrapolated from adult studies, with limited high-quality evidence available to guide management. The optimal, comprehensive management strategies for SAB in children will remain unknown until the questions outlined are answered through prospective observational cohorts and inclusion of children with SAB in clinical trials.


Assuntos
Antibacterianos/uso terapêutico , Bacteriemia/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus , Adolescente , Fatores Etários , Algoritmos , Bacteriemia/diagnóstico , Bacteriemia/epidemiologia , Estudos de Casos e Controles , Infecções Relacionadas a Cateter/etiologia , Cefalosporinas/uso terapêutico , Criança , Pré-Escolar , Técnica Delphi , Esquema de Medicação , Ecocardiografia , Endocardite Bacteriana/diagnóstico por imagem , Endocardite Bacteriana/tratamento farmacológico , Glicopeptídeos/uso terapêutico , Humanos , Incidência , Lactente , Recém-Nascido , Injeções Intravenosas , Staphylococcus aureus Resistente à Meticilina , Estudos Observacionais como Assunto , Penicilinas/uso terapêutico , Ensaios Clínicos Controlados Aleatórios como Assunto , Encaminhamento e Consulta , Infecções Estafilocócicas/diagnóstico , Infecções Estafilocócicas/epidemiologia , Staphylococcus aureus/isolamento & purificação , Vancomicina/uso terapêutico , beta-Lactamas/uso terapêutico
10.
Sci Adv ; 6(14): eaaw7713, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32270029

RESUMO

Recurrent Staphylococcus aureus infections are common, despite robust immune responses. S. aureus infection elicited protective antibody and T cell responses in mice that expressed the Major Histocompatibility Complex (MHC) of the H-2d haplotype, but not H-2b, demonstrating that host genetics drives individual variability. Vaccination with a-toxin or leukotoxin E (LukE) elicited similar antibody and T cell responses in mice expressing H-2d or H-2b, but vaccine-elicited responses were inhibited by concomitant infection in H-2d-expressing mice. These findings suggested that competitive binding of microbial peptides to host MHC proteins determines the specificity of the immunodominant response, which was confirmed using LukE-derived peptide-MHC tetramers. A vaccine that elicited T cell and antibody responses protected mice that expressed H-2d or H-2b, demonstrating that vaccination can overcome MHC-restricted immunodominance. Together, these results define how host genetics determine whether immunity elicted by S. aureus is protective and provide a mechanistic roadmap for future vaccine design.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Tolerância Imunológica , Epitopos Imunodominantes/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia , Animais , Modelos Animais de Doenças , Exotoxinas/imunologia , Antígenos H-2/imunologia , Camundongos , Ligação Proteica , Infecções Estafilocócicas/prevenção & controle , Linfócitos T/imunologia , Linfócitos T/metabolismo , Vacinação
11.
J Biol Chem ; 294(10): 3369-3384, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30622143

RESUMO

Staphylococcus aureus is a major etiological agent of sepsis and induces endothelial cell (EC) barrier dysfunction and inflammation, two major hallmarks of acute lung injury. However, the molecular mechanisms of bacterial pathogen-induced EC barrier disruption are incompletely understood. Here, we investigated the role of microtubules (MT) in the mechanisms of EC barrier compromise caused by heat-killed S. aureus (HKSA). Using a customized monolayer permeability assay in human pulmonary EC and MT fractionation, we observed that HKSA-induced barrier disruption is accompanied by MT destabilization and increased histone deacetylase-6 (HDAC6) activity resulting from elevated reactive oxygen species (ROS) production. Molecular or pharmacological HDAC6 inhibition rescued barrier function in HKSA-challenged vascular endothelium. The HKSA-induced EC permeability was associated with impaired MT-mediated delivery of cytoplasmic linker-associated protein 2 (CLASP2) to the cell periphery, limiting its interaction with adherens junction proteins. HKSA-induced EC barrier dysfunction was also associated with increased Rho GTPase activity via activation of MT-bound Rho-specific guanine nucleotide exchange factor-H1 (GEF-H1) and was abolished by HDAC6 down-regulation. HKSA activated the NF-κB proinflammatory pathway and increased the expression of intercellular and vascular cell adhesion molecules in EC, an effect that was also HDAC6-dependent and mediated, at least in part, by a GEF-H1/Rho-dependent mechanism. Of note, HDAC6 knockout mice or HDAC6 inhibitor-treated WT mice were partially protected from vascular leakage and inflammation caused by both HKSA or methicillin-resistant S. aureus (MRSA). Our results indicate that S. aureus-induced, ROS-dependent up-regulation of HDAC6 activity destabilizes MT and thereby activates the GEF-H1/Rho pathway, increasing both EC permeability and inflammation.


Assuntos
Células Endoteliais/metabolismo , Microtúbulos/metabolismo , Staphylococcus aureus/fisiologia , Células Endoteliais/microbiologia , Desacetilase 6 de Histona/metabolismo , Temperatura Alta , Humanos , Inflamação/microbiologia , Viabilidade Microbiana , Oxirredução , Permeabilidade , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
12.
Ann Surg ; 267(4): 749-758, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28187042

RESUMO

OBJECTIVE: To determine whether intestinal colonization with methicillin-resistant Staphylococcus aureus (MRSA) can be the source of surgical site infections (SSIs). BACKGROUND: We hypothesized that gut-derived MRSA may cause SSIs via mechanisms in which circulating immune cells scavenge MRSA from the gut, home to surgical wounds, and cause infection (Trojan Horse Hypothesis). METHODS: MRSA gut colonization was achieved by disrupting the microbiota with antibiotics, imposing a period of starvation and introducing MRSA via gavage. Next, mice were subjected to a surgical injury (30% hepatectomy) and rectus muscle injury and ischemia before skin closure. All wounds were cultured before skin closure. To control for postoperative wound contamination, reiterative experiments were performed in mice in which the closed wound was painted with live MRSA for 2 consecutive postoperative days. To rule out extracellular bacteremia as a cause of wound infection, MRSA was injected intravenously in mice subjected to rectus muscle ischemia and injury. RESULTS: All wound cultures were negative before skin closure, ruling out intraoperative contamination. Out of 40 mice, 4 (10%) developed visible abscesses. Nine mice (22.5%) had MRSA positive cultures of the rectus muscle without visible abscesses. No SSIs were observed in mice injected intravenously with MRSA. Wounds painted with MRSA after closure did not develop infections. Circulating neutrophils from mice captured by flow cytometry demonstrated MRSA in their cytoplasm. CONCLUSIONS: Immune cells as Trojan horses carrying gut-derived MRSA may be a plausible mechanism of SSIs in the absence of direct contamination.


Assuntos
Intestinos/microbiologia , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Infecções Estafilocócicas/microbiologia , Infecção da Ferida Cirúrgica/microbiologia , Abscesso/microbiologia , Animais , Antibacterianos/administração & dosagem , Modelos Animais de Doenças , Hepatectomia , Isquemia , Masculino , Staphylococcus aureus Resistente à Meticilina/imunologia , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Reto do Abdome/irrigação sanguínea , Reto do Abdome/microbiologia , Reto do Abdome/cirurgia , Fatores de Risco , Virulência
13.
Hum Vaccin Immunother ; 13(7): 1609-1614, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28422567

RESUMO

Staphylococcus aureus is the leading cause of nosocomial and community-acquired infections, including soft tissue and skin infections and bacteremia. However, efforts to develop an effective vaccine against S. aureus infections have not been successful. We evaluated serotypes 5 and 8 capsule polysaccharides (CP) CRM197 conjugates as vaccine candidates in murine models of bacteremia, lethal sepsis, and skin infection. The conjugate vaccines elicited a good antibody response, and active immunization of CP5-CRM or CP8-CRM conjugates protected against staphylococcal bacteremia. In the skin infection model, CP8-CRM but not CP5-CRM protected against dermonecrosis, and CP8-CRM immunization significantly decreased the bacterial burden in the lesion. However, neither CP5-CRM nor CP8-CRM protected against mortality in the lethal sepsis model. The results indicate the capsular vaccines elicit protection against some, but not all, aspects of staphylococcal infection.


Assuntos
Polissacarídeos Bacterianos/imunologia , Sorogrupo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas/imunologia , Staphylococcus aureus/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Antibacterianos/sangue , Bacteriemia/microbiologia , Bacteriemia/prevenção & controle , Carga Bacteriana , Proteínas de Bactérias/administração & dosagem , Modelos Animais de Doenças , Feminino , Humanos , Camundongos Endogâmicos BALB C , Infecções Cutâneas Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas/administração & dosagem , Análise de Sobrevida , Vacinas Conjugadas/administração & dosagem , Vacinas Conjugadas/imunologia
14.
Pathogens ; 5(1)2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-26828524

RESUMO

Recurrent Staphylococcus aureus infections are common, suggesting that immunity elicited by these infections is not protective. We previously reported that S. aureus skin infection (SSTI) elicited antibody-mediated immunity against secondary SSTI in BALB/c mice. In this study, we investigated the role of humoral immunity and the IgG-binding proteins Sbi and SpA in S. aureus SSTI. We found that B lymphocyte-deficient µMT mice were highly susceptible to infection, compared with congenic BALB/c mice. Importantly, transfer of immune serum protected µMT mice, demonstrating an appropriate response to protective antibody. We found that deletion of sbi, but not spa, impaired virulence, as assessed by skin lesion severity, and that Sbi-mediated virulence required B lymphocytes/antibody. Furthermore, neither Sbi nor SpA impaired the elicited antibody response or protection against secondary SSTI. Taken together, these findings highlight a B lymphocyte/antibody-dependent role of Sbi in the pathogenesis of S. aureus SSTI, and demonstrate that neither Sbi nor SpA interfered with elicited antibody-mediated immunity.

15.
Infect Immun ; 83(9): 3712-21, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26169277

RESUMO

Recurrent Staphylococcus aureus skin and soft tissue infections (SSTIs) are common despite detectable antibody responses, leading to the belief that the immune response elicited by these infections is not protective. We recently reported that S. aureus USA300 SSTI elicits antibodies that protect against recurrent SSTI in BALB/c but not C57BL/6 mice, and in this study, we aimed to uncover the specificity of the protective antibodies. Using a proteomic approach, we found that S. aureus SSTI elicited broad polyclonal antibody responses in both BALB/c and C57BL/6 mice and identified 10 S. aureus antigens against which antibody levels were significantly higher in immune BALB/c serum. Four of the 10 antigens identified are regulated by the saeRS operon, suggesting a dominant role for saeRS in protection. Indeed, infection with USA300Δsae failed to protect against secondary SSTI with USA300, despite eliciting a strong polyclonal antibody response against antigens whose expression is not regulated by saeRS. Moreover, the antibody repertoire after infection with USA300Δsae lacked antibodies specific for 10 saeRS-regulated antigens, suggesting that all or a subset of these antigens are necessary to elicit protective immunity. Infection with USA300Δhla elicited modest protection against secondary SSTI, and complementation of USA300Δsae with hla restored protection but incompletely. Together, these findings support a role for both Hla and other saeRS-regulated antigens in eliciting protection and suggest that host differences in immune responses to saeRS-regulated antigens may determine whether S. aureus infection elicits protective or nonprotective immunity against recurrent infection.


Assuntos
Anticorpos Antibacterianos/imunologia , Proteínas de Bactérias/imunologia , Imunidade Humoral/imunologia , Proteínas Quinases/imunologia , Infecções Cutâneas Estafilocócicas/imunologia , Animais , Antígenos de Bactérias/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Proteômica , Staphylococcus aureus/imunologia , Fatores de Transcrição
16.
Curr Opin Infect Dis ; 28(3): 253-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25887613

RESUMO

PURPOSE OF REVIEW: Staphylococcus aureus is the most common cause of skin and soft tissue infections (SSTI) in the United States and elsewhere. Recurrent infections occur frequently in patients with S. aureus SSTI, underscoring the need to better understand the nature of protective immunity against these infections. Here, we review recent findings concerning the host factors that predispose to S. aureus SSTI. RECENT FINDINGS: Recurrent infections occur in nearly half of all patients with S. aureus SSTI. Epidemiologic and environmental factors, such as exposure to healthcare, age, and household contacts with S. aureus SSTI, and contaminated household fomites are associated with recurrence. The majority of the population has evidence of antistaphylococcal antibodies, but whether these are protective remains enigmatic. In contrast, recent clinical and experimental findings clearly highlight the critical roles of innate and T cell-mediated immunity in defense against these infections. S. aureus interferes with innate and adaptive immunity by a number of recently elucidated mechanisms. SUMMARY: Recurrent S. aureus SSTIs are common, suggesting incomplete or absent protective immunity among these patients. Our understanding of protective immunity against recurrent infections is incomplete, and further basic and translational investigation is urgently needed to design strategies to prevent and treat these infections.


Assuntos
Imunidade Adaptativa/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Celular , Imunidade Inata , Infecções Cutâneas Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade , Antibacterianos/uso terapêutico , Anticorpos Antibacterianos/sangue , Humanos , Higiene/normas , Fatores de Risco , Prevenção Secundária , Infecções Cutâneas Estafilocócicas/tratamento farmacológico , Infecções Cutâneas Estafilocócicas/imunologia , Infecções Cutâneas Estafilocócicas/patologia
17.
BMC Genomics ; 15: 1145, 2014 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-25527145

RESUMO

BACKGROUND: Staphylococcus aureus is a human pathogen responsible for substantial morbidity and mortality through its ability to cause a number of human infections including bacteremia, pneumonia and soft tissue infections. Of great concern is the emergence and dissemination of methicillin-resistant Staphylococcus aureus strains (MRSA) that are resistant to nearly all ß-lactams. The emergence of the USA300 MRSA genetic background among community associated S. aureus infections (CA-MRSA) in the USA was followed by the disappearance of USA400 CA-MRSA isolates. RESULTS: To gain a greater understanding of the potential fitness advantages and virulence capacity of S. aureus USA300 clones, we performed whole genome sequencing of 15 USA300 and 4 USA400 clinical isolates. A comparison of representative genomes of the USA300 and USA400 pulsotypes indicates a number of differences in mobile genome elements. We examined the in vitro gene expression profiles by microarray hybridization and the in vivo transcriptomes during lung infection in mice of a USA300 and a USA400 MRSA strain by performing complete genome qRT-PCR analysis. The unique presence and increased expression of 6 exotoxins in USA300 (12- to 600-fold) compared to USA400 may contribute to the increased virulence of USA300 clones. Importantly, we also observed the up-regulation of prophage genes in USA300 (compared with USA400) during mouse lung infection (including genes encoded by both prophages ΦSa2usa and ΦSa3usa), suggesting that these prophages may play an important role in vivo by contributing to the elevated virulence characteristic of the USA300 clone. CONCLUSIONS: We observed differences in the genetic content of USA300 and USA400 strains, as well as significant differences of in vitro and in vivo gene expression of mobile elements in a lung pneumonia model. This is the first study to document the global transcription differences between USA300 and USA400 strains during both in vitro and in vivo growth.


Assuntos
Infecções Comunitárias Adquiridas/microbiologia , Genoma Bacteriano , Staphylococcus aureus Resistente à Meticilina/genética , RNA Bacteriano/genética , Infecções Estafilocócicas/genética , Transcriptoma , Infecções Comunitárias Adquiridas/epidemiologia , Infecções Estafilocócicas/epidemiologia , Infecções Estafilocócicas/microbiologia , Estados Unidos/epidemiologia
18.
Infect Immun ; 82(5): 2125-34, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24614654

RESUMO

Although many microbial infections elicit an adaptive immune response that can protect against reinfection, it is generally thought that Staphylococcus aureus infections fail to generate protective immunity despite detectable T and B cell responses. No vaccine is yet proven to prevent S. aureus infections in humans, and efforts to develop one have been hampered by a lack of animal models in which protective immunity occurs. Our results describe a novel mouse model of protective immunity against recurrent infection, in which S. aureus skin and soft tissue infection (SSTI) strongly protected against secondary SSTI in BALB/c mice but much less so in C57BL/6 mice. This protection was dependent on antibody, because adoptive transfer of immune BALB/c serum or purified antibody into either BALB/c or C57BL/6 mice resulted in smaller skin lesions. We also identified an antibody-independent mechanism, because B cell-deficient mice were partially protected against secondary S. aureus SSTI and adoptive transfer of T cells from immune BALB/c mice resulted in smaller lesions upon primary infection. Furthermore, neutralization of interleukin-17A (IL-17A) abolished T cell-mediated protection in BALB/c mice, whereas neutralization of gamma interferon (IFN-γ) enhanced protection in C57BL/6 mice. Therefore, protective immunity against recurrent S. aureus SSTI was advanced by antibody and the Th17/IL-17A pathway and prevented by the Th1/IFN-γ pathway, suggesting that targeting both cell-mediated and humoral immunity might optimally protect against secondary S. aureus SSTI. These findings also highlight the importance of the mouse genetic background in the development of protective immunity against S. aureus SSTI.


Assuntos
Anticorpos Antibacterianos/imunologia , Interleucina-17/metabolismo , Infecções Cutâneas Estafilocócicas/imunologia , Transferência Adotiva , Animais , Regulação da Expressão Gênica , Interleucina-17/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções Cutâneas Estafilocócicas/microbiologia , Staphylococcus aureus
19.
PLoS One ; 8(7): e69508, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23861974

RESUMO

Staphylococcus aureus is the leading cause of skin infections. In a mouse model of S. aureus skin infection, we found that lesion size did not correlate with bacterial burden. Athymic nude mice had smaller skin lesions that contained lower levels of myeloperoxidase, IL-17A, and CXCL1, compared with wild type mice, although there was no difference in bacterial burden. T cell deficiency did not explain the difference in lesion size, because TCR ßδ (-/-) mice did not have smaller lesions, and adoptive transfer of congenic T cells into athymic nude mice prior to infection did not alter lesion size. The differences observed were specific to the skin, because mortality in a pneumonia model was not different between wild type and athymic nude mice. Thus, the clinical severity of S. aureus skin infection is driven by the inflammatory response to the bacteria, rather than bacterial burden, in a T cell independent manner.


Assuntos
Dermatite/imunologia , Dermatite/patologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/imunologia , Animais , Dermatite/microbiologia , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Nus , Infecções Estafilocócicas/microbiologia , Subpopulações de Linfócitos T/imunologia
20.
Antimicrob Agents Chemother ; 57(1): 83-95, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23070169

RESUMO

Staphylococcus aureus infections caused by strains that are resistant to all forms of penicillin, so-called methicillin-resistant S. aureus (MRSA) strains, have become common. One strategy to counter MRSA infections is to use compounds that resensitize MRSA to methicillin. S. aureus responds to diverse classes of cell wall-inhibitory antibiotics, like methicillin, using the two-component regulatory system VraSR (vra) to up- or downregulate a set of genes (the cell wall stimulon) that presumably facilitates resistance to these antibiotics. Accordingly, VraS and VraR mutations decrease resistance to methicillin, vancomycin, and daptomycin cell wall antimicrobials. vraS and vraR are encoded together on a transcript downstream of two other genes, which we call vraU and vraT (previously called yvqF). By producing nonpolar deletions in vraU and vraT in a USA300 MRSA clinical isolate, we demonstrate that vraT is essential for optimal expression of methicillin resistance in vitro, whereas vraU is not required for this phenotype. The deletion of vraT also improved the outcomes of oxacillin therapy in mouse models of lung and skin infection. Since vraT expressed in trans did not complement a vra operon deletion, we conclude that VraT does not inactivate the antimicrobial. Genome-wide transcriptional microarray experiments reveal that VraT facilitates resistance by playing a necessary regulatory role in the VraSR-mediated cell wall stimulon. Our data prove that VraTSR comprise a novel three-component regulatory system required to facilitate resistance to cell wall agents in S. aureus. We also provide the first in vivo proof of principle for using VraT as a sole target to resensitize MRSA to ß-lactams.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Ligação a DNA/genética , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Resistência a Meticilina/genética , Staphylococcus aureus Resistente à Meticilina/genética , Regulon , Sequência de Aminoácidos , Animais , Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Parede Celular/efeitos dos fármacos , Parede Celular/genética , Parede Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Perfilação da Expressão Gênica , Genes Reguladores , Masculino , Meticilina/farmacologia , Resistência a Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA