Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(51): e2213116119, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36512492

RESUMO

New antimicrobials are needed for the treatment of extensively drug-resistant Acinetobacter baumannii. The de novo pyrimidine biosynthetic enzyme dihydroorotate dehydrogenase (DHODH) is a validated drug target for malaria and human autoimmune diseases. We provide genetic evidence that A. baumannii DHODH (AbDHODH) is essential for bacterial survival in rodent infection models. We chemically validate the target by repurposing a unique library of ~450 triazolopyrimidine/imidazopyrimidine analogs developed for our malaria DHODH program to identify 21 compounds with submicromolar activity on AbDHODH. The most potent (DSM186, DHODH IC50 28 nM) had a minimal inhibitory concentration of ≤1 µg/ml against geographically diverse A. baumannii strains, including meropenem-resistant isolates. A structurally related analog (DSM161) with a long in vivo half-life conferred significant protection in the neutropenic mouse thigh infection model. Encouragingly, the development of resistance to these compounds was not identified in vitro or in vivo. Lastly, the X-ray structure of AbDHODH bound to DSM186 was solved to 1.4 Å resolution. These data support the potential of AbDHODH as a drug target for the development of antimicrobials for the treatment of A. baumannii and potentially other high-risk bacterial infections.


Assuntos
Acinetobacter baumannii , Humanos , Camundongos , Animais , Di-Hidro-Orotato Desidrogenase , Testes de Sensibilidade Microbiana , Meropeném , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/química , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico
2.
Antimicrob Agents Chemother ; 67(1): e0103322, 2023 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-36475717

RESUMO

Limited therapeutic options dictate the need for new classes of antimicrobials active against carbapenem-resistant Acinetobacter baumannii. Presented data confirm and extend penicillin binding protein 7/8 (PBP 7/8) as a high-value target in the CR A. baumannii strain HUMC1. PBP 7/8 was essential for optimal growth/survival of HUMC1 in ex vivo human ascites and in a rat subcutaneous abscess model; in a mouse pneumonia model, the absence of PBP 7/8 decreased lethality 11-fold. The loss of PBP 7/8 resulted in increased permeability, sensitivity to complement, and lysozyme-mediated bactericidal activity. These changes did not appear to be due to alterations in the cellular fatty acid composition or capsule production. However, a decrease in lipid A and an increase in coccoidal cells and cell aggregation were noted. The compromise of the stringent permeability barrier in the PBP 7/8 mutant was reflected by an increased susceptibility to several antimicrobials. Importantly, expression of ampC was not significantly affected by the loss of PBP 7/8 and serial passage of the mutant strain in human ascites over 7 days did not yield revertants possessing a wild-type phenotype. In summary, these data and other features support PBP 7/8 as a high-value drug target for extensively drug-resistant and CR A. baumannii. Our results guide next-stage studies; the determination that the inactivation of PBP 7/8 results in an increased sensitivity to lysozyme enables the design of a high-throughput screening assay to identify small molecule compounds that can specifically inhibit PBP 7/8 activity.


Assuntos
Acinetobacter baumannii , Camundongos , Animais , Humanos , Ratos , Proteínas de Ligação às Penicilinas/genética , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Muramidase/metabolismo , Ascite , Testes de Sensibilidade Microbiana , Carbapenêmicos/farmacologia , Carbapenêmicos/metabolismo , Antibacterianos/farmacologia , Antibacterianos/metabolismo
3.
PLoS Pathog ; 17(2): e1009291, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33529209

RESUMO

Acinetobacter baumannii is a highly antibiotic-resistant bacterial pathogen for which novel therapeutic approaches are needed. Unfortunately, the drivers of virulence in A. baumannii remain uncertain. By comparing genomes among a panel of A. baumannii strains we identified a specific gene variation in the capsule locus that correlated with altered virulence. While less virulent strains possessed the intact gene gtr6, a hypervirulent clinical isolate contained a spontaneous transposon insertion in the same gene, resulting in the loss of a branchpoint in capsular carbohydrate structure. By constructing isogenic gtr6 mutants, we confirmed that gtr6-disrupted strains were protected from phagocytosis in vitro and displayed higher bacterial burden and lethality in vivo. Gtr6+ strains were phagocytized more readily and caused lower bacterial burden and no clinical illness in vivo. We found that the CR3 receptor mediated phagocytosis of gtr6+, but not gtr6-, strains in a complement-dependent manner. Furthermore, hypovirulent gtr6+ strains demonstrated increased virulence in vivo when CR3 function was abrogated. In summary, loss-of-function in a single capsule assembly gene dramatically altered virulence by inhibiting complement deposition and recognition by phagocytes across multiple A. baumannii strains. Thus, capsular structure can determine virulence among A. baumannii strains by altering bacterial interactions with host complement-mediated opsonophagocytosis.


Assuntos
Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/patogenicidade , Cápsulas Bacterianas/fisiologia , Fagócitos/virologia , Fagocitose , Polissacarídeos Bacterianos/química , Virulência , Infecções por Acinetobacter/genética , Infecções por Acinetobacter/metabolismo , Animais , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Fagócitos/metabolismo , Células RAW 264.7
4.
JCI Insight ; 9(2)2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38258902

RESUMO

With antimicrobial resistance (AMR) emerging as a major threat to global health, monoclonal antibodies (MAbs) have become a promising means to combat difficult-to-treat AMR infections. Unfortunately, in contrast with standard antimicrobials, for which there are well-validated clinical laboratory methodologies to determine whether an infecting pathogen is susceptible or resistant to a specific antimicrobial drug, no assays have been described that can inform clinical investigators or clinicians regarding the clinical efficacy of a MAb against a specific pathogenic strain. Using Acinetobacter baumannii as a model organism, we established and validated 2 facile clinical susceptibility assays, which used flow cytometry and latex bead agglutination, to determine susceptibility (predicting in vivo efficacy) or resistance (predicting in vivo failure) of 1 newly established and 3 previously described anti-A. baumannii MAbs. These simple assays exhibited impressive sensitivity, specificity, and reproducibility, with clear susceptibility breakpoints that predicted the in vivo outcomes in our preclinical model with excellent fidelity. These MAb susceptibility assays have the potential to enable and facilitate clinical development and deployment of MAbs that generally target the surface of microbes.


Assuntos
Acinetobacter baumannii , Anti-Infecciosos , Reprodutibilidade dos Testes , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Imunoterapia
5.
mBio ; 15(2): e0286723, 2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-38231533

RESUMO

Distinguishing hypervirulent (hvKp) from classical Klebsiella pneumoniae (cKp) strains is important for clinical care, surveillance, and research. Some combinations of iucA, iroB, peg-344, rmpA, and rmpA2 are most commonly used, but it is unclear what combination of genotypic or phenotypic markers (e.g., siderophore concentration, mucoviscosity) most accurately predicts the hypervirulent phenotype. Furthermore, acquisition of antimicrobial resistance may affect virulence and confound identification. Therefore, 49 K. pneumoniae strains that possessed some combinations of iucA, iroB, peg-344, rmpA, and rmpA2 and had acquired resistance were assembled and categorized as hypervirulent hvKp (hvKp) (N = 16) or cKp (N = 33) via a murine infection model. Biomarker number, siderophore production, mucoviscosity, virulence plasmid's Mash/Jaccard distances to the canonical pLVPK, and Kleborate virulence score were measured and evaluated to accurately differentiate these pathotypes. Both stepwise logistic regression and a CART model were used to determine which variable was most predictive of the strain cohorts. The biomarker count alone was the strongest predictor for both analyses. For logistic regression, the area under the curve for biomarker count was 0.962 (P = 0.004). The CART model generated the classification rule that a biomarker count = 5 would classify the strain as hvKP, resulting in a sensitivity for predicting hvKP of 94% (15/16), a specificity of 94% (31/33), and an overall accuracy of 94% (46/49). Although a count of ≥4 was 100% (16/16) sensitive for predicting hvKP, the specificity and accuracy decreased to 76% (25/33) and 84% (41/49), respectively. These findings can be used to inform the identification of hvKp.IMPORTANCEHypervirulent Klebsiella pneumoniae (hvKp) is a concerning pathogen that can cause life-threatening infections in otherwise healthy individuals. Importantly, although strains of hvKp have been acquiring antimicrobial resistance, the effect on virulence is unclear. Therefore, it is of critical importance to determine whether a given antimicrobial resistant K. pneumoniae isolate is hypervirulent. This report determined which combination of genotypic and phenotypic markers could most accurately identify hvKp strains with acquired resistance. Both logistic regression and a machine-learning prediction model demonstrated that biomarker count alone was the strongest predictor. The presence of all five of the biomarkers iucA, iroB, peg-344, rmpA, and rmpA2 was most accurate (94%); the presence of ≥4 of these biomarkers was most sensitive (100%). Accurately identifying hvKp is vital for surveillance and research, and the availability of biomarker data could alert the clinician that hvKp is a consideration, which, in turn, would assist in optimizing patient care.


Assuntos
Infecções por Klebsiella , Klebsiella pneumoniae , Humanos , Animais , Camundongos , Infecções por Klebsiella/epidemiologia , Biomarcadores , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Sideróforos
6.
bioRxiv ; 2023 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-37961280

RESUMO

Distinguishing hypervirulent (hvKp) from classical Klebsiella pneumoniae (cKp) strains is important for clinical care, surveillance, and research. Some combination of iucA, iroB, peg-344, rmpA, and rmpA2 are most commonly used, but it is unclear what combination of genotypic or phenotypic markers (e.g. siderophore concentration, mucoviscosity) most accurately predicts the hypervirulent phenotype. Further, acquisition of antimicrobial resistance may affect virulence and confound identification. Therefore, 49 K. pneumoniae strains that possessed some combination of iucA, iroB, peg-344, rmpA, and rmpA2 and had acquired resistance were assembled and categorized as hypervirulent hvKp (hvKp) (N=16) or cKp (N=33) via a murine infection model. Biomarker number, siderophore production, mucoviscosity, virulence plasmid's Mash/Jaccard distances to the canonical pLVPK, and Kleborate virulence score were measured and evaluated to accurately differentiate these pathotypes. Both stepwise logistic regression and a CART model were used to determine which variable was most predictive of the strain cohorts. The biomarker count alone was the strongest predictor for both analyses. For logistic regression the area under the curve for biomarker count was 0.962 (P = 0.004). The CART model generated the classification rule that a biomarker count = 5 would classify the strain as hvKP, resulting in a sensitivity for predicting hvKP of 94% (15/16), a specificity of 94% (31/33), and an overall accuracy of 94% (46/49). Although a count of ≥ 4 was 100% (16/16) sensitive for predicting hvKP, the specificity and accuracy decreased to 76% (25/33) and 84% (41/49) respectively. These findings can be used to inform the identification of hvKp. Importance: Hypervirulent Klebsiella pneumoniae (hvKp) is a concerning pathogen that can cause life-threatening infections in otherwise healthy individuals. Importantly, although strains of hvKp have been acquiring antimicrobial resistance, the effect on virulence is unclear. Therefore, it is of critical importance to determine whether a given antimicrobial resistant K. pneumoniae isolate is hypervirulent. This report determined which combination of genotypic and phenotypic markers could most accurately identify hvKp strains with acquired resistance. Both logistic regression and a machine-learning prediction model demonstrated that biomarker count alone was the strongest predictor. The presence of all 5 of the biomarkers iucA, iroB, peg-344, rmpA, and rmpA2 was most accurate (94%); the presence of ≥ 4 of these biomarkers was most sensitive (100%). Accurately identifying hvKp is vital for surveillance and research, and the availability of biomarker data could alert the clinician that hvKp is a consideration, which in turn would assist in optimizing patient care.

7.
FEMS Immunol Med Microbiol ; 53(1): 65-71, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18384367

RESUMO

Infections outside of the intestinal tract due to pathogenic strains of Escherichia coli result in significant morbidity, mortality and increased healthcare costs. The ability of these strains to cause both mucosal and systemic infections, as well as recurrent infections due to the same (homologous) strain suggests the hypothesis that strains of E. coli that cause infection outside of the intestinal tract possess proteases that are capable of cleaving IgA1, IgA2, sIgA or IgG. To test this hypothesis the ability of eight E. coli strains, isolated from sites outside of the urinary tract and 14 homologous and 11 heterologous strains of E. coli that were isolated from women with recurrent UTI, to cleave IgA1, IgA2, sIgA or IgG was evaluated. Our experimental design allowed for detection of cell-associated and secreted immunoglobulin proteases in both log and stationary phase. Surprisingly, none of these 33 human clinical isolates when grown in iron depleted Luria-Bertani medium or human urine were able to degrade the immunoglobulins assessed. Despite previous studies suggesting otherwise, the findings from this study support the concept that strains of E. coli that cause infection outside of the intestinal tract do not possess proteases that cleave the human immunoglobulins IgA1, IgA2, sIgA or IgG.


Assuntos
Infecções por Escherichia coli/microbiologia , Escherichia coli/enzimologia , Escherichia coli/imunologia , Imunoglobulina A/metabolismo , Imunoglobulina G/metabolismo , Eletroforese em Gel de Poliacrilamida , Escherichia coli/isolamento & purificação , Infecções por Escherichia coli/imunologia , Feminino , Humanos , Peptídeo Hidrolases/metabolismo , Serina Endopeptidases/metabolismo
8.
FEMS Microbiol Lett ; 226(2): 355-61, 2003 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-14553933

RESUMO

Gram-negative enteric bacilli are agents of life-threatening pneumonia. The role of the bacterial capsule and O-antigen moiety of lipopolysaccharide in the pathogenesis of Gram-negative pneumonia was assessed. In a rat model of pneumonia the LD(50) of a wild-type extraintestinal pathogenic Escherichia coli strain (CP9) was significantly less than its isogenic derivatives deficient in capsule (CP9.137), O-antigen (CP921) or both capsule and O-antigen (CP923) (P< or =0.003). Studies using complement depleted or neutropenic animals established that both neutrophils and complement are important for the pulmonary clearance of E. coli. Data from these studies also support that capsule and O-antigen serve, at least in part, to counter the complement and neutrophil components of the pulmonary host defense response. Lastly, the contribution of E. coli versus neutrophils in causing lung injury was examined. Findings suggest that E. coli virulence factors and/or non-neutrophil host factors are more important mediators of lung injury than neutrophils. These findings extend our understanding of Gram-negative pneumonia and have treatment implications.


Assuntos
Proteínas do Sistema Complemento/imunologia , Infecções por Escherichia coli/imunologia , Escherichia coli/patogenicidade , Neutrófilos/imunologia , Pneumonia Bacteriana/imunologia , Polissacarídeos Bacterianos/fisiologia , Animais , Cápsulas Bacterianas/fisiologia , Modelos Animais de Doenças , Escherichia coli/genética , Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Genes Bacterianos , Mutagênese Insercional , Antígenos O/fisiologia , Pneumonia Bacteriana/microbiologia , Polissacarídeos Bacterianos/imunologia , Ratos , Fatores de Virulência/fisiologia
9.
Infect Immun ; 71(12): 7164-9, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14638809

RESUMO

It would be medically and economically desirable to prevent the millions of annual extraintestinal infections and the thousands of associated deaths due to Escherichia coli. Outer membrane proteins are potential vaccine candidates for the prevention of these infections. This study tested the hypotheses that the siderophore receptor IroN is antigenic and that an IroN-specific antibody response confers protection in vivo. Subcutaneous immunization with denatured IroN resulted in a significant IroN immunoglobulin G (IgG)-specific response in serum (P < 0.0001) but not a systemic or mucosal IroN-specific IgA response. In a mouse model of ascending urinary tract infection, subcutaneous immunization with denatured IroN conferred significant protection against renal (P = 0.0135 and 0.0095 in two independent experiments), but not bladder, infection. These data, together with the previously demonstrated role of IroN in virulence, its expression in human biologic fluids, and its prevalence among extraintestinal pathogenic E. coli strains, support further studies on the role of IroN as a vaccine candidate.


Assuntos
Anticorpos Antibacterianos/sangue , Vacinas Bacterianas/administração & dosagem , Infecções por Escherichia coli/prevenção & controle , Proteínas de Escherichia coli/imunologia , Receptores de Superfície Celular/imunologia , Infecções Urinárias/prevenção & controle , Animais , Vacinas Bacterianas/imunologia , Modelos Animais de Doenças , Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/administração & dosagem , Proteínas de Escherichia coli/metabolismo , Humanos , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Superfície Celular/administração & dosagem , Receptores de Superfície Celular/metabolismo , Infecções Urinárias/microbiologia
10.
Infect Immun ; 70(12): 7156-60, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12438401

RESUMO

IroN was recently identified in the extracellular pathogenic Escherichia coli strain CP9. In this study experimental evidence demonstrating that IroN mediates utilization of the siderophore enterobactin was obtained, thereby establishing IroN as a catecholate siderophore receptor. In a mouse model of ascending urinary tract infection the presence of iroN contributed significantly to CP9's ability to colonize the mouse bladder, kidneys, and urine, evidence that IroN is a urovirulence factor. However, growth in human urine ex vivo and adherence to uroepithelial cells in vitro were equivalent for an isogenic mutant deficient in IroN (CP82) and its wild-type parent (CP9). Taken together, these findings establish that IroN is a siderophore receptor and a urovirulence factor. However, uncertainty exists as to the mechanism(s) via which IroN contributes to urovirulence.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Enterobactina/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Receptores de Superfície Celular/metabolismo , Infecções Urinárias/microbiologia , Animais , Aderência Bacteriana , Proteínas da Membrana Bacteriana Externa/genética , Células Cultivadas , Modelos Animais de Doenças , Escherichia coli/crescimento & desenvolvimento , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Feminino , Humanos , Camundongos , Dados de Sequência Molecular , Ratos , Receptores de Superfície Celular/genética , Bexiga Urinária/citologia , Bexiga Urinária/microbiologia , Urina/microbiologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA