Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Cell Infect Microbiol ; 14: 1328185, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38510967

RESUMO

Pseudomonas aeruginosa is an important human opportunistic pathogen responsible for a wide range of infections. The complement system is the main early host defense mechanism to control these infections. P. aeruginosa counteracts complement attack by binding Factor H (FH), a complement regulator that inactivates C3b, preventing the formation of the C3-convertase and complement amplification on the bacterial surface. Factor H-related proteins (FHRs) are a group of plasma proteins evolutionarily related to FH that have been postulated to interfere in this bacterial mechanism of resisting complement. Here, we show that FHR-1 binds to P. aeruginosa via the outer membrane protein OprG in a lipopolysaccharide (LPS) O antigen-dependent manner. Binding assays with purified components or with FHR-1-deficient serum supplemented with FHR-1 show that FHR-1 competes with FH for binding to P. aeruginosa. Blockage of FH binding to C3b deposited on the bacteria reduces FH-mediated cofactor activity of C3b degradation, increasing the opsonization of the bacteria and the formation of the potent chemoattractant C5a. Overall, our findings indicate that FHR-1 is a host factor that promotes complement activation, facilitating clearance of P. aeruginosa by opsonophagocytosis.


Assuntos
Proteínas Sanguíneas , Fator H do Complemento , Pseudomonas aeruginosa , Humanos , Fator H do Complemento/metabolismo , Pseudomonas aeruginosa/metabolismo , Opsonização , Ligação Proteica , Proteínas do Sistema Complemento/metabolismo , Bactérias/metabolismo
2.
Front Cell Infect Microbiol ; 13: 1191806, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37424774

RESUMO

Pseudomonas aeruginosa is a common cause of hospital-acquired infections, including central line-associated bloodstream infections and ventilator-associated pneumonia. Unfortunately, effective control of these infections can be difficult, in part due to the prevalence of multi-drug resistant strains of P. aeruginosa. There remains a need for novel therapeutic interventions against P. aeruginosa, and the use of monoclonal antibodies (mAb) is a promising alternative strategy to current standard of care treatments such as antibiotics. To develop mAbs against P. aeruginosa, we utilized ammonium metavanadate, which induces cell envelope stress responses and upregulates polysaccharide expression. Mice were immunized with P. aeruginosa grown with ammonium metavanadate and we developed two IgG2b mAbs, WVDC-0357 and WVDC-0496, directed against the O-antigen lipopolysaccharide of P. aeruginosa. Functional assays revealed that WVDC-0357 and WVDC-0496 directly reduced the viability of P. aeruginosa and mediated bacterial agglutination. In a lethal sepsis model of infection, prophylactic treatment of mice with WVDC-0357 and WVDC-0496 at doses as low as 15 mg/kg conferred 100% survival against challenge. In both sepsis and acute pneumonia models of infection, treatment with WVDC-0357 and WVDC-0496 significantly reduced bacterial burden and inflammatory cytokine production post-challenge. Furthermore, histopathological examination of the lungs revealed that WVDC-0357 and WVDC-0496 reduced inflammatory cell infiltration. Overall, our results indicate that mAbs directed against lipopolysaccharide are a promising therapy for the treatment and prevention of P. aeruginosa infections.


Assuntos
Anticorpos Antibacterianos , Anticorpos Monoclonais , Lipopolissacarídeos , Infecções por Pseudomonas , Pseudomonas aeruginosa , Animais , Feminino , Camundongos , Anticorpos Antibacterianos/imunologia , Anticorpos Monoclonais/imunologia , Aderência Bacteriana , Carga Bacteriana/imunologia , Convalescença , Mediadores da Inflamação/imunologia , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/imunologia , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/prevenção & controle , Pseudomonas aeruginosa/imunologia , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/prevenção & controle , Sepse/imunologia , Sepse/microbiologia , Sepse/prevenção & controle
3.
Front Cell Infect Microbiol ; 13: 1117844, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37124031

RESUMO

The rise of antimicrobial-resistant bacterial infections is a crucial health concern in the 21st century. In particular, antibiotic-resistant Pseudomonas aeruginosa causes difficult-to-treat infections associated with high morbidity and mortality. Unfortunately, the number of effective therapeutic interventions against antimicrobial-resistant P. aeruginosa infections continues to decline. Therefore, discovery and development of alternative treatments are necessary. Here, we present pre-clinical efficacy studies on an anti-P. aeruginosa therapeutic monoclonal antibody. Using hybridoma technology, we generated a monoclonal antibody and characterized its binding to P. aeruginosa in vitro using ELISA and fluorescence correlation spectroscopy. We also characterized its function in vitro and in vivo against P. aeruginosa. The anti-P. aeruginosa antibody (WVDC-5244) bound P. aeruginosa clinical strains of various serotypes in vitro, even in the presence of alginate exopolysaccharide. In addition, WVDC-5244 induced opsonophagocytic killing of P. aeruginosa in vitro in J774.1 murine macrophage, and complement-mediated killing. In a mouse model of acute pneumonia, prophylactic administration of WVDC-5244 resulted in an improvement of clinical disease manifestations and reduction of P. aeruginosa burden in the respiratory tract compared to the control groups. This study provides promising pre-clinical efficacy data on a new monoclonal antibody with therapeutic potential for P. aeruginosa infections.


Assuntos
Pneumonia , Infecções por Pseudomonas , Camundongos , Animais , Pseudomonas aeruginosa , Pneumonia/microbiologia , Anticorpos Monoclonais/uso terapêutico , Hibridomas/metabolismo , Proteínas do Sistema Complemento , Infecções por Pseudomonas/microbiologia
4.
Microorganisms ; 11(3)2023 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-36985237

RESUMO

The increasing emergence of multidrug resistant isolates of P. aeruginosa causes major problems in hospitals worldwide. This concern is particularly significant in bloodstream infections that progress rapidly, with a high number of deaths within the first hours and without time to select the most appropriate treatment. In fact, despite improvements in antimicrobial therapy and hospital care, P. aeruginosa bacteremia remains fatal in about 30% of cases. The complement system is a main defensive mechanism in blood against this pathogen. This system can mark bacteria for phagocytosis or directly lyse it via the insertion of a membrane attack complex in the bacterial membrane. P. aeruginosa exploits different strategies to resist complement attack. In this review for the special issue on "bacterial pathogens associated with bacteriemia", we present an overview of the interactions between P. aeruginosa and the complement components and strategies used by this pathogen to prevent recognition and killing by the complement system. A thorough understanding of these interactions will be critical in order to develop drugs to counteract bacterial evasion mechanisms.

5.
NPJ Vaccines ; 7(1): 143, 2022 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-36357402

RESUMO

Whole cell vaccines are complex mixtures of antigens, immunogens, and sometimes adjuvants that can trigger potent and protective immune responses. In some instances, such as whole cell Bordetella pertussis vaccination, the immune response to vaccination extends beyond the pathogen the vaccine was intended for and contributes to protection against other clinically significant pathogens. In this study, we describe how B. pertussis whole cell vaccination protects mice against acute pneumonia caused by Pseudomonas aeruginosa. Using ELISA and western blot, we identified that B. pertussis whole cell vaccination induces production of antibodies that bind to lab-adapted and clinical strains of P. aeruginosa, regardless of immunization route or adjuvant used. The cross-reactive antigens were identified using immunoprecipitation, mass spectrometry, and subsequent immunoblotting. We determined that B. pertussis GroEL and OmpA present in the B. pertussis whole cell vaccine led to production of antibodies against P. aeruginosa GroEL and OprF, respectively. Finally, we showed that recombinant B. pertussis OmpA was sufficient to induce protection against P. aeruginosa acute murine pneumonia. This study highlights the potential for use of B. pertussis OmpA as a vaccine antigen for prevention of P. aeruginosa infection, and the potential of broadly protective antigens for vaccine development.

6.
Urolithiasis ; 50(6): 737-742, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36064981

RESUMO

The use of double J ureteral stents can lead to several adverse effects, as urinary infection. Bacteria tend to colonize the stent surface, leading to the formation of bacterial biofilms. The presence of urease-producing bacteria increase the urine pH leading to the incrustation and blockage of the stent. On the other hand, these large crystalline masses function as niduses, allowing the attachment of even more bacteria and decreasing its exposure to antibiotics. The aim of this in vitro study was to assess the effect of phytate on the attachment of bacteria to the catheter surface under conditions that favor crystallization. Catheter sections were incubated in a synthetic urine medium (pH 6.5) in the presence or absence of Pseudomonas aeruginosa and phytate. Amount of calcium deposits was measured using an Arsenazo III colorimetric method and the number of attached bacteria to the stent was determined. Differences were assessed using an ANOVA with a Bonferroni post hoc test. The formation of calcium phosphate deposits (brushite and hydroxyapatite) and oxalate crystals (COM), as were as the amount of bacteria decreased when phytate was present. Thus, phytate successfully decreased bacterial adhesion by inhibiting the formation of crystalline deposits.


Assuntos
Cálcio , Ácido Fítico , Humanos , Cristalização , Urease , Arsenazo III , Fosfatos de Cálcio/química , Oxalato de Cálcio/química , Bactérias , Durapatita , Stents/efeitos adversos , Antibacterianos
7.
Virulence ; 13(1): 215-224, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35094639

RESUMO

Cystic fibrosis (CF) disease is characterized by an intense airway inflammatory response mediated by neutrophils and chronic respiratory infections caused by P. aeruginosa. High levels of the complement component C5a, the strongest neutrophil chemoattractant molecule, are commonly found in the CF lung and have been associated with a worsening of the disease. In this study, we investigated how the isolates from CF patients modulate the levels of C5a and identified the bacterial factors involved. We demonstrated that most isolates from airway chronic infections induce the production and accumulation of C5a, an effect attributable to the loss of C5a cleavage by the exoproteases alkaline protease (AprA) and elastase B (LasB). Furthermore, we found that lack of the bacterial protease-dependent C5a degradation is due to mutations in the master regulator LasR. Thus, complementation of a non-C5a-cleaving CF isolate with a functional wild-type LasR restored its ability to express both proteases, cleave C5a and reduce neutrophil recruitment in vitro. These findings suggest that the non-cleaving C5a phenotype acquired by the LasR variants frequently isolated in CF patients may account for the strong neutrophilia and general neutrophil dysfunction predisposing toward increased inflammation and reduced bacterial clearance described in CF patients.


Assuntos
Complemento C5a/análise , Fibrose Cística , Infecções por Pseudomonas , Fibrose Cística/complicações , Fibrose Cística/microbiologia , Humanos , Infiltração de Neutrófilos , Peptídeo Hidrolases/metabolismo , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/metabolismo , Sistema Respiratório
8.
Front Cell Infect Microbiol ; 11: 816356, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35145924

RESUMO

Pseudomonas aeruginosa is a major cause of nosocomial bloodstream infections. This microorganism secretes two major proteases, alkaline protease A (AprA) and elastase B (LasB). Despite several in vitro studies having demonstrated that both purified proteases cleave a number of components of the immune system, their contribution to P. aeruginosa bloodstream infections in vivo remains poorly investigated. In this study, we used a set of isogenic mutants deficient in AprA, LasB or both to demonstrate that these exoproteases are sufficient to cleave the complement component C3, either soluble or deposited on the bacteria. Nonetheless, exoprotease-deficient mutants were as virulent as the wild-type strain in a murine model of systemic infection, in Caenorhabditis elegans and in Galleria mellonella. Consistently, the effect of the exoproteases on the opsonization of P. aeruginosa by C3 became evident four hours after the initial interaction of the complement with the microorganism and was not crucial to survival in blood. These results indicate that exoproteases AprA and LasB, although conferring the capacity to cleave C3, are not essential for the virulence of P. aeruginosa bloodstream infections.


Assuntos
Proteínas de Bactérias , Metaloendopeptidases , Infecções por Pseudomonas , Sepse , Animais , Proteínas de Bactérias/genética , Endopeptidases , Metaloendopeptidases/genética , Camundongos , Elastase Pancreática/genética , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/genética , Virulência
9.
Environ Microbiol ; 19(10): 4278-4286, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28805355

RESUMO

Pseudomonas aeruginosa adaptation to survive in the host hinges on its ability to probe the environment and respond appropriately. Rapid adaptation is often mediated by two-component regulatory systems, such as the PhoP/PhoQ system that responds to Mg2+ ion concentration. However, there is limited information about the role of PhoQ in P. aeruginosa bloodstream infections. We used a murine model of systemic infection to test the virulence of a PhoQ-deficient mutant. Mutation of PhoQ impaired the virulence and the ability to cause bacteremia of P. aeruginosa. In the presence of blood concentrations of Mg2+ , a PhoQ mutant bound more C3 and was more susceptible to complement-mediated opsonophagocytosis than the parent strain, suggesting a direct effect of the Mg2+ on the modulation of expression of a bacterial component controlled by the PhoP/PhoQ system. Ligand blot analysis, C3 binding experiments and opsonophagocytosis assays identified this component as the outer membrane protein OprH, expression of which impaired the virulence of P. aeruginosa in a murine model of systemic infection. We demonstrate that expression of PhoQ is essential to detect Mg2+ and reduce the expression of OprH, a previously unrecognized C3 binding molecule that promotes the opsonophagocytosis of P. aeruginosa.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/metabolismo , Complemento C3/imunologia , Regulação Bacteriana da Expressão Gênica , Fagocitose/imunologia , Pseudomonas aeruginosa/imunologia , Animais , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Humanos , Magnésio , Masculino , Camundongos , Ligação Proteica/imunologia , Pseudomonas aeruginosa/genética , Virulência
10.
J Infect Dis ; 214(9): 1449-1455, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27543671

RESUMO

Surfactant protein A (SP-A) plays a critical role in the clearance of Pseudomonas aeruginosa from the lung. However, there is limited information about the interaction of this protein with P. aeruginosa isolates from individuals with cystic fibrosis (CF). We characterized the interplay between SP-A and a collection of isogenic sequential isolates from 7 patients with CF. We identified outer membrane protein OprH as a novel ligand for SP-A on P. aeruginosa The last-available (late) isolates from patients with CF bound significantly less SP-A than their respective first-available (early) isolates. This difference could be associated with a reduction in the expression of OprH. Binding of SP-A to OprH promoted phagocytic killing; thus, late CF isolates were at least 2-fold more resistant to SP-A-mediated killing by human macrophages than their respective early isolates. We postulate that the reduction of OprH expression is a previously unrecognized adaptation of P. aeruginosa to the lung of individuals with CF that facilitates the escape of the microorganism from SP-A-mediated phagocytic killing.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Fibrose Cística/metabolismo , Fibrose Cística/microbiologia , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Proteína A Associada a Surfactante Pulmonar/metabolismo , Humanos , Pulmão/metabolismo , Pulmão/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA