Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 6: 25063, 2016 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-27121552

RESUMO

Multi-drug resistant bacteria are a persistent problem in modern health care, food safety and animal health. There is a need for new antimicrobials to replace over used conventional antibiotics. Here we describe engineered triple-acting staphylolytic peptidoglycan hydrolases wherein three unique antimicrobial activities from two parental proteins are combined into a single fusion protein. This effectively reduces the incidence of resistant strain development. The fusion protein reduced colonization by Staphylococcus aureus in a rat nasal colonization model, surpassing the efficacy of either parental protein. Modification of a triple-acting lytic construct with a protein transduction domain significantly enhanced both biofilm eradication and the ability to kill intracellular S. aureus as demonstrated in cultured mammary epithelial cells and in a mouse model of staphylococcal mastitis. Interestingly, the protein transduction domain was not necessary for reducing the intracellular pathogens in cultured osteoblasts or in two mouse models of osteomyelitis, highlighting the vagaries of exactly how protein transduction domains facilitate protein uptake. Bacterial cell wall degrading enzyme antimicrobials can be engineered to enhance their value as potent therapeutics.


Assuntos
Antibacterianos/metabolismo , Antibacterianos/uso terapêutico , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/efeitos dos fármacos , Animais , Portador Sadio/prevenção & controle , Células Cultivadas , Modelos Animais de Doenças , Humanos , Mastite/tratamento farmacológico , Camundongos , N-Acetil-Muramil-L-Alanina Amidase/genética , Osteomielite/tratamento farmacológico , Ratos , Proteínas Recombinantes de Fusão/genética , Resultado do Tratamento
2.
Cell Microbiol ; 15(7): 1154-67, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23298320

RESUMO

Streptococcus agalactiae (Group B Streptococcus, GBS) is a frequent commensal organism of the vaginal tract of healthy women. However, GBS can transition to a pathogen in susceptible hosts, but host and microbial factors that contribute to this conversion are not well understood. GBS CovR/S (CsrR/S) is a two component regulatory system that regulates key virulence elements including adherence and toxin production. We performed global transcription profiling of human vaginal epithelial cells exposed to WT, CovR deficient, and toxin deficient strains, and observed that insufficient regulation by CovR and subsequent increased toxin production results in a drastic increase in host inflammatory responses, particularly in cytokine signalling pathways promoted by IL-8 and CXCL2. Additionally, we observed that CovR regulation impacts epithelial cell attachment and intracellular invasion. In our mouse model of GBS vaginal colonization, we further demonstrated that CovR regulation promotes vaginal persistence, as infection with a CovR deficient strainresulted in a heightened host immune response as measured by cytokine production and neutrophil activation. Using CXCr2 KO mice, we determined that this immune alteration occurs, at least in part, via signalling through the CXCL2 receptor. Taken together, we conclude that CovR is an important regulator of GBS vaginal colonization and loss of this regulatory function may contribute to the inflammatory havoc seen during the course of infection.


Assuntos
Regulação da Expressão Gênica , Proteínas Repressoras/metabolismo , Transdução de Sinais , Streptococcus agalactiae/imunologia , Streptococcus agalactiae/patogenicidade , Vagina/imunologia , Vagina/microbiologia , Animais , Proteínas de Bactérias/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Feminino , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Camundongos Knockout , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/imunologia , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia
3.
Vaccine ; 30(1): 9-13, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-22044742

RESUMO

The emergence of community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA) is a serious health concern worldwide that requires new therapeutic approaches that extend beyond the development and use of new antibiotics. In this study, a conformationally biased, response-selective agonist of human C5a, known as EP67, was used to induce host innate immunity as a therapeutic method of reducing CA-MRSA infections. Using a murine model of dermonecrosis we show that EP67 treatment effectively limits CA-MRSA infection by promoting cytokine synthesis and neutrophil influx. In contrast, EP67 was ineffective in reducing lesion formation in C5a receptor (CD88(-/-)) knockout mice, indicating that EP67 activates host innate immunity by engagement of CD88 bearing cells. These results suggest that EP67 may serve as a novel immunotherapeutic for prevention and treatment of CA-MRSA dermal infection.


Assuntos
Fatores Imunológicos/administração & dosagem , Staphylococcus aureus Resistente à Meticilina/imunologia , Peptídeos/administração & dosagem , Infecções Cutâneas Estafilocócicas/tratamento farmacológico , Infecções Cutâneas Estafilocócicas/microbiologia , Animais , Complemento C5a/agonistas , Modelos Animais de Doenças , Feminino , Camundongos , Resultado do Tratamento
4.
J Bacteriol ; 193(24): 6834-42, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21984789

RESUMO

Streptococcus agalactiae (group B streptococcus [GBS]) is a Gram-positive bacterium found in the female rectovaginal tract and is capable of producing severe disease in susceptible hosts, including newborns and pregnant women. The vaginal tract is considered a major reservoir for GBS, and maternal vaginal colonization poses a significant risk to the newborn; however, little is known about the specific bacterial factors that promote GBS colonization and persistence in the female reproductive tract. We have developed in vitro models of GBS interaction with the human female cervicovaginal tract using human vaginal and cervical epithelial cell lines. Analysis of isogenic mutant GBS strains deficient in cell surface organelles such as pili and serine-rich repeat (Srr) proteins shows that these factors contribute to host cell attachment. As Srr proteins are heavily glycosylated, we confirmed that carbohydrate moieties contribute to the effective interaction of Srr-1 with vaginal epithelial cells. Antibody inhibition assays identified keratin 4 as a possible host receptor for Srr-1. Our findings were further substantiated in an in vivo mouse model of GBS vaginal colonization, where mice inoculated with an Srr-1-deficient mutant exhibited decreased GBS vaginal persistence compared to those inoculated with the wild-type (WT) parental strain. Furthermore, competition experiments in mice showed that WT GBS exhibited a significant survival advantage over the ΔpilA or Δsrr-1 mutant in the vaginal tract. Our results suggest that these GBS surface proteins contribute to vaginal colonization and may offer new insights into the mechanisms of vaginal niche establishment.


Assuntos
Adesinas Bacterianas/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/metabolismo , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/crescimento & desenvolvimento , Vagina/microbiologia , Adesinas Bacterianas/genética , Animais , Aderência Bacteriana , Proteínas de Bactérias/genética , Linhagem Celular , Feminino , Proteínas de Fímbrias/genética , Fímbrias Bacterianas/genética , Humanos , Camundongos , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/fisiologia
5.
Infect Immun ; 79(7): 2510-8, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21518787

RESUMO

Bacillus anthracis is a Gram-positive spore-forming bacterium that causes anthrax disease in humans and animals. Systemic infection is characterized by septicemia, toxemia, and meningitis, the main neurological complication associated with high mortality. We have shown previously that B. anthracis Sterne is capable of blood-brain barrier (BBB) penetration, establishing the classic signs of meningitis, and that infection is dependent on the expression of both major anthrax toxins, lethal toxin (LT) and edema toxin (ET). Here we further investigate the contribution of the individual toxins to BBB disruption using isogenic toxin mutants deficient in lethal factor, ΔLF, and edema factor, ΔEF. Acute infection with B. anthracis Sterne and the ΔLF mutant resulted in disruption of human brain microvascular endothelial cell (hBMEC) monolayer integrity and tight junction protein zona occludens-1, while the result for cells infected with the ΔEF mutant was similar to that for the noninfected control. A significant decrease in bacterial invasion of BBB endothelium in vitro was observed during infection with the ΔLF strain, suggesting a prominent role for LT in promoting BBB interaction. Further, treatment of hBMECs with purified LT or chemicals that mimic LT action on host signaling pathways rescued the hypoinvasive phenotype of the ΔLF mutant and resulted in increased bacterial uptake. We also observed that toxin expression reduced bacterial intracellular survival by inducing the bulk degradative autophagy pathway in host cells. Finally, in a murine model of anthrax meningitis, mice infected with the ΔLF mutant exhibited no mortality, brain bacterial load, or evidence of meningitis compared to mice infected with the parental or ΔEF strains.


Assuntos
Antraz/microbiologia , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Bacillus anthracis/patogenicidade , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Barreira Hematoencefálica/microbiologia , Meningites Bacterianas/microbiologia , Animais , Antraz/mortalidade , Antraz/patologia , Antígenos de Bactérias/biossíntese , Autofagia , Bacillus anthracis/genética , Toxinas Bacterianas/biossíntese , Barreira Hematoencefálica/patologia , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Células Endoteliais/patologia , Humanos , Proteínas de Membrana/metabolismo , Meningites Bacterianas/mortalidade , Meningites Bacterianas/patologia , Camundongos , Microvasos/patologia , Mutação , Fosfoproteínas/metabolismo , Junções Íntimas/ultraestrutura , Proteína da Zônula de Oclusão-1
6.
Cell Microbiol ; 12(11): 1576-88, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20557315

RESUMO

Streptococcus pneumoniae (SPN), the leading cause of meningitis in children and adults worldwide, is associated with an overwhelming host inflammatory response and subsequent brain injury. Here we examine the global response of the blood-brain barrier to SPN infection and the role of neuraminidase A (NanA), an SPN surface anchored protein recently described to promote central nervous system tropism. Microarray analysis of human brain microvascular endothelial cells (hBMEC) during infection with SPN or an isogenic NanA-deficient (ΔnanA) mutant revealed differentially activated genes, including neutrophil chemoattractants IL-8, CXCL-1, CXCL-2. Studies using bacterial mutants, purified recombinant NanA proteins and in vivo neutrophil chemotaxis assays indicated that pneumococcal NanA is necessary and sufficient to activate host chemokine expression and neutrophil recruitment during infection. Chemokine induction was mapped to the NanA N-terminal lectin-binding domain with a limited contribution of the sialidase catalytic activity, and was not dependent on the invasive capability of the organism. Furthermore, pretreatment of hBMEC with recombinant NanA protein significantly increased bacterial invasion, suggesting that NanA-mediated activation of hBMEC is a prerequisite for efficient SPN invasion. These findings were corroborated in an acute murine infection model where we observed less inflammatory infiltrate and decreased chemokine expression following infection with the ΔnanA mutant.


Assuntos
Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Células Endoteliais/metabolismo , Meningite Pneumocócica/microbiologia , Neuraminidase/metabolismo , Streptococcus pneumoniae/patogenicidade , Animais , Barreira Hematoencefálica/imunologia , Encéfalo/microbiologia , Quimiocina CCL20/metabolismo , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/metabolismo , Quimiotaxia de Leucócito , Células Endoteliais/imunologia , Células Endoteliais/microbiologia , Endotélio Vascular/metabolismo , Perfilação da Expressão Gênica , Humanos , Interleucina-8/metabolismo , Lectinas , Meningite Pneumocócica/imunologia , Camundongos , Mutação , Infiltração de Neutrófilos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Transdução de Sinais , Streptococcus pneumoniae/enzimologia , Streptococcus pneumoniae/genética
7.
J Mol Med (Berl) ; 88(6): 633-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20419283

RESUMO

Staphylococcus aureus is one of the most prevalent organisms responsible for nosocomial infections, and cases of community-acquired S. aureus infection have continued to increase despite widespread preventative measures. Pathologies attributed to S. aureus infection are diverse; ranging from dermal lesions to bacteremia, abscesses, and endocarditis. Reported cases of S. aureus-associated meningitis and brain abscesses have also increased in recent years, however, the precise mechanism whereby S. aureus leave the bloodstream and gain access to the central nervous system (CNS) are not known. Here we demonstrate for the first time that S. aureus efficiently adheres to and invades human brain microvascular endothelial cells (hBMEC), the single-cell layer which constitutes the blood-brain barrier (BBB). The addition of cytochalasin D, an actin microfilament aggregation inhibitor, strongly reduced bacterial invasion, suggesting an active hBMEC process is required for efficient staphylococcal uptake. Furthermore, mice injected with S. aureus exhibited significant levels of brain bacterial counts and histopathologic evidence of meningeal inflammation and brain abscess formation, indicating that S. aureus was able to breech the BBB in an experimental model of hematogenous meningitis. We found that a YpfP-deficient mutant, defective in lipoteichoic acid (LTA) membrane anchoring, exhibited a decreased ability to invade hBMEC and correlated to a reduced risk for the development of meningitis in vivo. Our results demonstrate that LTA-mediated penetration of the BBB may be a primary step in the pathogenesis of staphylococcal CNS disease.


Assuntos
Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/microbiologia , Membrana Celular/química , Lipopolissacarídeos/metabolismo , Staphylococcus aureus/patogenicidade , Ácidos Teicoicos/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular , Parede Celular/metabolismo , Células Endoteliais/citologia , Células Endoteliais/microbiologia , Humanos , Masculino , Camundongos , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/citologia , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
8.
J Bacteriol ; 191(23): 7165-73, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19820089

RESUMO

Anthrax is a zoonotic disease caused by the gram-positive spore-forming bacterium Bacillus anthracis. Human infection occurs after the ingestion, inhalation, or cutaneous inoculation of B. anthracis spores. The subsequent progression of the disease is largely mediated by two native virulence plasmids, pXO1 and pXO2, and is characterized by septicemia, toxemia, and meningitis. In order to produce meningitis, blood-borne bacteria must interact with and breach the blood-brain barrier (BBB) that is composed of a specialized layer of brain microvascular endothelial cells (BMEC). We have recently shown that B. anthracis Sterne is capable of penetrating the BBB in vitro and in vivo, establishing the classic signs of meningitis; however, the molecular mechanisms underlying the central nervous system (CNS) tropism are not known. Here, we show that attachment to and invasion of human BMEC by B. anthracis Sterne is mediated by the pXO1 plasmid and an encoded envelope factor, BslA. The results of studies using complementation analysis, recombinant BslA protein, and heterologous expression demonstrate that BslA is both necessary and sufficient to promote adherence to brain endothelium. Furthermore, mice injected with the BslA-deficient strain exhibited a significant decrease in the frequency of brain infection compared to mice injected with the parental strain. In addition, BslA contributed to BBB breakdown by disrupting tight junction protein ZO-1. Our results identify the pXO1-encoded BslA adhesin as a critical mediator of CNS entry and offer new insights into the pathogenesis of anthrax meningitis.


Assuntos
Antraz/microbiologia , Bacillus anthracis/patogenicidade , Proteínas de Bactérias/genética , Barreira Hematoencefálica/microbiologia , Adesinas Bacterianas/genética , Animais , Bacillus anthracis/genética , Aderência Bacteriana/genética , Linhagem Celular , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Microscopia , Plasmídeos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...