Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Bases de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cell Microbiol ; 21(6): e13015, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30702185

RESUMO

Epithelial cell shedding is a defence mechanism against infectious microbes that use these cells as an infection foothold and that eliminate microbes from infection foci by removing infected cells. Mycoplasma pneumoniae, a causative agent of respiratory infections, is known to adhere to and colonise the surface of ciliated airway epithelial cells; it produces a large amount of hydrogen peroxide, indicating its capability of regulating hydrogen peroxide-induced infected cell detachment. In this study, we found that M. pneumoniae reduces exogenous hydrogen peroxide-induced detachment of the infected cells from culture plates. This cell detachment occurred dependently of DNA damage-initiated, poly (ADP-ribose) polymerase 1 (PARP1)-mediated cell death, or parthanatos. In cells infected with M. pneumoniae, exogenous hydrogen peroxide failed to induce DNA damage-initiated poly (ADP-ribose) (PAR) synthesis and concomitant increased cytoplasmic membrane rupture, both of which are biochemical hallmarks of parthanatos. The impairment of PAR synthesis was attributed to a reduction in the amount of cytosolic nicotinamide adenine dinucleotide (NAD), a substrate of PARP1, caused by M. pneumoniae. On the other hand, nonadherent mutant strains of M. pneumoniae showed a lower ability to reduce cell detachment than wild-type strains, but the extent to which NAD was decreased in infected cells was comparable to that seen in the wild-type strain. We found that NAD depletion could induce PARP1-independent cell detachment pathways following stimulation with hydrogen peroxide and that M. pneumoniae could also regulate PARP1-independent cell detachment in a cytoadhesion-dependent manner. These results suggest that M. pneumoniae might regulate infected cell detachment induced by hydrogen peroxide that it produces itself, and such a mechanism may contribute to sustaining the bacterial infection.


Assuntos
Células Epiteliais/microbiologia , Peróxido de Hidrogênio/toxicidade , Mycoplasma pneumoniae/metabolismo , Parthanatos/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/metabolismo , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Mycoplasma pneumoniae/genética , Mycoplasma pneumoniae/patogenicidade , NAD/metabolismo , Poli(ADP-Ribose) Polimerase-1/genética , Espécies Reativas de Oxigênio/metabolismo
2.
Microbiol Immunol ; 64(4): 280-295, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31907968

RESUMO

Mucin is a glycoprotein that is the primary component of the mucus overlaying the epithelial tissues. Because mucin functions as a first line of the innate immune system, Pseudomonas aeruginosa appears to require interaction with mucin to establish infection in the host. However, the interactions between P. aeruginosa and mucin have been poorly understood. In this study, using in vivo expression technology (IVET), we attempted to identify mucin-inducible promoters that are likely to be involved in the establishment of P. aeruginosa infection. The IVET analysis revealed that the genes encoding glycosidases, sulfatases, and peptidases that are thought to be required for the utilization of mucin as a nutrient are present in 13 genes downstream of the identified promoters. Our results indicated that, among them, sdsA1 encoding a secreted sulfatase plays a central role in the degradation of mucin. It was then demonstrated that disruption of sdsA1 leads to a decreased release of sulfate from mucin and sulfated sugars. Furthermore, the sdsA1 mutant showed a reduction in the ability of mucin gel penetration and an attenuation of virulence in leukopenic mice compared with the wild-type strain. Collectively, these results suggest that SdsA1 plays an important role as a virulence factor of P. aeruginosa.


Assuntos
Proteínas de Bactérias/imunologia , Mucina-3/metabolismo , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/enzimologia , Sulfatases/imunologia , Fatores de Virulência/imunologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos ICR , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/patogenicidade , Suínos , Virulência
3.
Cell Microbiol ; 19(3)2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27603754

RESUMO

Neutrophils play an important role in antimicrobial defense as the first line of innate immune system. Recently, the release of neutrophil extracellular traps (NETs) has been identified as a killing mechanism of neutrophils against invading microbes. Mycoplasma pneumoniae, a causative agent of respiratory infection, has been shown to be resistant to in vitro killing by neutrophils, suggesting that the bacterium might circumvent bactericidal activity of NETs. In this study, we investigated whether M. pneumoniae possesses resistance mechanisms against the NETs-mediated killing of neutrophils and found that the bacterium degrades the NETs induced upon M. pneumoniae infection. The NETs-degrading ability of M. pneumoniae required the production of a secreted nuclease, Mpn491, capable of using Mg2+ as a cofactor for its hydrolytic activity. Moreover, the inactivation of the nuclease resulted in increased susceptibility of M. pneumoniae to the NETs-mediated killing of neutrophils. The results suggest that M. pneumoniae employs Mpn491 as a means for evading the killing mechanism of neutrophils.


Assuntos
Desoxirribonucleases/metabolismo , Armadilhas Extracelulares/metabolismo , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Mycoplasma pneumoniae/enzimologia , Mycoplasma pneumoniae/patogenicidade , Neutrófilos/imunologia , Hidrólise , Magnésio/metabolismo , Mycoplasma pneumoniae/imunologia
4.
Infect Immun ; 82(7): 3076-86, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24799628

RESUMO

Mycoplasma pneumoniae causes pneumonia, tracheobronchitis, pharyngitis, and asthma in humans. The pathogenesis of M. pneumoniae infection is attributed to excessive immune responses. We previously demonstrated that M. pneumoniae lipoproteins induced inflammatory responses through Toll-like receptor 2 (TLR2). In the present study, we demonstrated that M. pneumoniae induced strong inflammatory responses in macrophages derived from TLR2 knockout (KO) mice. Cytokine production in TLR2 KO macrophages was increased compared with that in the macrophages of wild-type (WT) mice. Heat-killed, antibiotic-treated, and overgrown M. pneumoniae failed to induce inflammatory responses in TLR2 KO macrophages. 3-Methyladenine and chloroquine, inhibitors of autophagy, decreased the induction of inflammatory responses in TLR2 KO macrophages. These inflammatory responses were also inhibited in macrophages treated with the TLR4 inhibitor VIPER and those obtained from TLR2 and TLR4 (TLR2/4) double-KO mice. Two mutants that lacked the ability to induce inflammatory responses in TLR2 KO macrophages were obtained by transposon mutagenesis. The transposons were inserted in atpC encoding an ATP synthase F0F1 ε subunit and F10_orf750 encoding hypothetical protein MPN333. These mutants showed deficiencies in cytadherence. These results suggest that cytadherence of M. pneumoniae induces inflammatory responses through TLR4 and autophagy.


Assuntos
Autofagia/fisiologia , Aderência Bacteriana/fisiologia , Infecções por Mycoplasma/imunologia , Mycoplasma pneumoniae/fisiologia , Receptor 4 Toll-Like/metabolismo , Animais , Regulação da Expressão Gênica/imunologia , Inflamação/metabolismo , Macrófagos , Camundongos , Camundongos Knockout , Mutação , Infecções por Mycoplasma/microbiologia , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética
5.
Cell Microbiol ; 15(7): 1168-81, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23311922

RESUMO

PA3535 (EprS), an autotransporter (AT) protein of Pseudomonas aeruginosa, is predicted to contain a serine protease motif. The eprS encodes a 104.5 kDa protein with a 30-amino-acid-long signal peptide, a 51.2 kDa amino-terminal secreted passenger domain and a 50.1 kDa carboxyl-terminal outer membrane channel formed translocator. Although the majority of AT proteins have been reported to be virulence factors, little is known about the functions of EprS in the pathogenicity of P. aeruginosa. In this study, we performed functional analyses of recombinant EprS secreted by Escherichia coli. The proteolytic activity of EprS was markedly decreased by changing Ser to Ala at position 308 or by serine protease inhibitors. EprS preferred to cleave substrates that terminated with arginine or lysine residues. Thus, these results indicate that EprS, a serine protease, displays the substrate specificity, cleaving after basic residues. We demonstrated that EprS activates NF-κB-driven promoters through protease-activated receptor (PAR)-1, -2 or -4 and induces IL-8 production through PAR-2 in a human bronchiole epithelial cell line. Moreover, EprS cleaved the peptides corresponding to the tethered ligand region of PAR-1, -2 and -4 at a specific site with exposure oftheir tethered ligands. Collectively, these results suggest that EprS activates host inflammatory responses through PARs.


Assuntos
Inflamação/patologia , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/patogenicidade , Receptores Ativados por Proteinase/metabolismo , Serina Proteases/metabolismo , Fatores de Virulência/metabolismo , Linhagem Celular , Células Epiteliais/microbiologia , Escherichia coli/genética , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
6.
J Biol Chem ; 287(16): 13170-81, 2012 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-22303020

RESUMO

Bacterial lipoproteins/lipopeptides inducing host innate immune responses are sensed by mammalian Toll-like receptor 2 (TLR2). These bacterial lipoproteins are structurally divided into two groups, diacylated or triacylated lipoproteins, by the absence or presence of an amide-linked fatty acid. The presence of diacylated lipoproteins has been predicted in low-GC content gram-positive bacteria and mycoplasmas based on the absence of one modification enzyme in their genomes; however, we recently determined triacylated structures in low-GC gram-positive Staphylococcus aureus, raising questions about the actual lipoprotein structure in other low-GC content gram-positive bacteria. Here, through intensive MS analyses, we identified a novel and unique bacterial lipoprotein structure containing an N-acyl-S-monoacyl-glyceryl-cysteine (named the lyso structure) from low-GC gram-positive Enterococcus faecalis, Bacillus cereus, Streptococcus sanguinis, and Lactobacillus bulgaricus. Two of the purified native lyso-form lipoproteins induced proinflammatory cytokine production from mice macrophages in a TLR2-dependent and TLR1-independent manner but with a different dependence on TLR6. Additionally, two other new lipoprotein structures were identified. One is the "N-acetyl" lipoprotein structure containing N-acetyl-S-diacyl-glyceryl-cysteine, which was found in five gram-positive bacteria, including Bacillus subtilis. The N-acetyl lipoproteins induced the proinflammatory cytokines through the TLR2/6 heterodimer. The other was identified in a mycoplasma strain and is an unusual diacyl lipoprotein structure containing two amino acids before the lipid-modified cysteine residue. Taken together, our results suggest the existence of novel TLR2-stimulating lyso and N-acetyl forms of lipoproteins that are conserved in low-GC content gram-positive bacteria and provide clear evidence for the presence of yet to be identified key enzymes involved in the bacterial lipoprotein biosynthesis.


Assuntos
Bactérias Gram-Positivas/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Receptor 2 Toll-Like/imunologia , Receptor 2 Toll-Like/metabolismo , Animais , Bacillus cereus/imunologia , Bacillus cereus/metabolismo , Bacillus subtilis/imunologia , Bacillus subtilis/metabolismo , Enterococcus faecalis/imunologia , Enterococcus faecalis/metabolismo , Geobacillus/imunologia , Geobacillus/metabolismo , Bactérias Gram-Positivas/metabolismo , Lactobacillus/imunologia , Lactobacillus/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia por Mycoplasma/imunologia , Pneumonia por Mycoplasma/metabolismo , Streptococcus sanguis/imunologia , Streptococcus sanguis/metabolismo , Receptor 1 Toll-Like/genética , Receptor 1 Toll-Like/imunologia , Receptor 1 Toll-Like/metabolismo , Receptor 2 Toll-Like/genética , Receptor 6 Toll-Like/genética , Receptor 6 Toll-Like/imunologia , Receptor 6 Toll-Like/metabolismo
7.
J Infect Chemother ; 19(2): 261-71, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23229613

RESUMO

Streptococcus pneumoniae has been reported to form biofilms. Many different surface molecules, including capsular polysaccharide (CPS), may play a fundamental role in pneumococcal biofilm development. We designed a CPS mutant, TIGR4cps4D(-), from the TIGR4 strain and detected enhanced biofilm formation. The pathogenic diversities of the mutant were also investigated with the in vitro expression levels of pavA, lytA, IgA1, piaA, psaA, ply, and spxB. The mean OD595 of TIGR4cps4D(-) biofilm was 1.77 and 1.74, whereas that of TIGR4 was 0.76 and 0.33 on day 1 and day 2, respectively. Scanning electron microscopy and confocal laser scanning microscopy showed TIGR4cps4D(-) formed a biofilm that was significantly thicker than that formed by TIGR4 (~12.22 vs. ~6.29 µm). Compared to TIGR4, the gene expression of lytA, IgA1, and, psaA in TIGR4cps4D(-) was 1.9 × 10(-5)-, 2.4 × 10(-5)-, and 3.2 × 10(-3) fold lower under the planktonic condition, and 1.9 × 10(-5)- and 9.7 × 10(-5) fold lower in biofilms, respectively. Furthermore, TIGR4cps4D(-) seemed to induce less cell death, compared to the results of TIGR4 (21.38 vs. 33.47 %, after a 5-h exposure; P < 0.05). Our data indicate that impaired pneumococcal CPS may increase biofilm formation and be involved in inhibition of virulence, possibly by influencing the gene expression.


Assuntos
Biofilmes , Polissacarídeos Bacterianos/fisiologia , Streptococcus pneumoniae/fisiologia , Virulência/genética , Linhagem Celular , Engenharia Genética , Interações Hospedeiro-Patógeno , Humanos , Microscopia Confocal , Mutação , Infecções Pneumocócicas/microbiologia , Polissacarídeos Bacterianos/genética , Polissacarídeos Bacterianos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/metabolismo , Streptococcus pneumoniae/patogenicidade
8.
Inflamm Res ; 61(5): 493-501, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22270622

RESUMO

OBJECTIVE: To investigate the involvement of reactive oxygen species (ROS) in the activation of Janus kinase2 (Jak2)/signal transducers and activators of transcription3 (Stat3), and IL-8 expression in pulmonary epithelial cells stimulated with lipid-associated membrane proteins (LAMP) from Mycoplasma pneumoniae using a known antioxidant, N-acetylcysteine (NAC). METHODS: Pulmonary epithelial A549 cells were treated with or without NAC in the presence or absence of LAMP. Intracellular ROS levels were detected by fluorescent analysis for fluorescent dichlorofluorescein. mRNA expression of IL-8 was analyzed by reverse transcription-polymerase chain reaction. IL-8 protein in the medium was determined by enzyme-linked immunosorbent assay. Activation of Jak2/Stat3 was determined by the increases in phospho-specific forms of Jak2/Stat3 compared to total forms of Jak2/Stat3 by western blotting. Stat3-DNA binding activity was assessed by electrophoretic mobility shift assay. RESULTS: LAMP increased the level of ROS, phosphorylation of Jak2/Stat3, Stat3-DNA binding activity, and IL-8 expression in A549 cells, which were inhibited by NAC dose-dependently. CONCLUSION: LAMP of M. pneumoniae induces the production of ROS, Jak2/Stat3 activation, and IL-8 induction in A549 cells. Antioxidants such as NAC may be beneficial for preventing pulmonary inflammation caused by M. pneumoniae.


Assuntos
Proteínas de Bactérias/farmacologia , Interleucina-8/genética , Janus Quinase 2/fisiologia , Pulmão/metabolismo , Proteínas de Membrana/farmacologia , Mycoplasma pneumoniae/patogenicidade , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT3/fisiologia , Acetilcisteína/farmacologia , Linhagem Celular , DNA/metabolismo , Ativação Enzimática , Células Epiteliais/metabolismo , Humanos , NF-kappa B/fisiologia , Receptores Toll-Like/fisiologia
9.
Infect Immun ; 79(1): 211-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21041488

RESUMO

Pseudomonas aeruginosa-derived large extracellular protease (LepA) and hemolytic phospholipase C (PlcH) are considered to play an important role in the pathogenicity of this organism. Although bacterial growth appears to be closely related to virulence, little is known about whether LepA and PlcH participate in the growth and virulence of P. aeruginosa. In this study, we investigated whether LepA and PlcH contribute to the virulence and growth of P. aeruginosa using a wild-type strain and mutants. The growth rate of the isogenic lepA single mutant was lower than that of the wild-type strain in a minimal medium containing serum albumin or hemoglobin as the sole carbon and nitrogen source. Furthermore, the growth rate of the lepA plcH double mutant decreased greatly compared with that of the wild-type strain in a minimal medium containing erythrocytes as a sole nutrient source for growth. Thus, these results indicate that cooperation between LepA and PlcH would contribute to the utilization of erythrocytes as a sole nutrient source for the growth of P. aeruginosa. In addition, mouse infection experiments demonstrated that the virulence of the lepA and plcH single mutants was attenuated, and the numbers of the mutants were lower than the numbers of the wild-type strain in peritoneal lavage fluid and whole-blood specimens. In particular, the virulence and growth rate of the lepA plcH double mutant were markedly lower than those of the wild-type strain. Collectively, these results suggest that LepA and PlcH contribute to the in vivo virulence and growth of P. aeruginosa.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/enzimologia , Fosfolipases Tipo C/metabolismo , Albuminas , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Meios de Cultura/química , Eritrócitos , Humanos , Ferro , Masculino , Camundongos , Mutação , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Ovinos/sangue , Fosfolipases Tipo C/classificação , Fosfolipases Tipo C/genética , Virulência
10.
Immunology ; 133(1): 51-61, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21320122

RESUMO

Pathogenesis of Mycoplasma pneumoniae infection is considered to be in part attributed to excessive immune responses. Mycoplasma pneumoniae shows strong cytoadherence to host cells and this cytoadherence is thought to be involved in the progression of pneumonia. However, the interaction between the cytoadherence and the immune responses is not known in detail. In this study, we demonstrated that the induction of pro-inflammatory cytokines in the human monocyte cell line THP-1 is dependent on the property of cytoadherence of M. pneumoniae. A wild-type strain of M. pneumoniae with cytoadherence ability induced pro-inflammatory cytokines such as tumour necrosis factor-α and interleukin-1ß (IL-1ß). Whereas, heat-killed M. pneumoniae and cytoadherence-deficient mutants of M. pneumoniae caused significantly less production of pro-inflammatory cytokines than the wild-type strain. The wild-type strain induced pro-inflammatory cytokines in an endocytosis-independent manners, but the induction by heat-killed M. pneumoniae and cytoadherence-deficient mutants was dependent on endocytosis. Moreover, the wild-type strain induced caspase-1 production and ATP efflux, promoting the maturation of IL-1ß and release of the pro-IL-1ß precursor, whereas heat-killed M. pneumoniae and the cytoadherence-deficient mutants failed to induce them. These data suggest that the cytoadherence ability of M. pneumoniae activates immune responses and is involved in the pathogenesis of M. pneumoniae infection.


Assuntos
Aderência Bacteriana/imunologia , Citocinas/biossíntese , Inflamação/imunologia , Monócitos/imunologia , Mycoplasma pneumoniae/patogenicidade , Western Blotting , Linhagem Celular , Citocinas/imunologia , Endocitose/imunologia , Ensaio de Imunoadsorção Enzimática , Humanos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Inflamação/metabolismo , Inflamação/microbiologia , Monócitos/microbiologia , Mycoplasma pneumoniae/imunologia , Transdução de Sinais/imunologia
11.
Microbiol Immunol ; 55(8): 582-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21605160

RESUMO

In innate immunity, cationic antimicrobial peptides including cathelin-related antimicrobial peptide (CRAMP) are known to play critical roles in protecting the host from infection by invasive microbes, including Gram-positive and -negative bacteria. However, little is known about the interactions between CRAMP and mycoplasmas. In the present study, the antimicrobial activity of CRAMP against M. pneumoniae and the expression of CRAMP in bronchoalveolar lavage fluid (BALF) of M. pneumoniae-infected mice was examined. CRAMP at 10-20 µg/mL reduced the growth of two strains of M. pneumoniae by 100 to 1000-fold. The amount of CRAMP in the BALF of M. pneumoniae-infected mice was 20∼25 ng/mL by ELISA. The presence of mature CRAMP in BALF was observed by Western blotting. Neutrophils in BALF showed a fair amount of CRAMP in their cytoplasm by immunofluorescence. Furthermore, the addition of M. pneumoniae resulted in the release of a large amount of CRAMP from neutrophils induced by thioglycolate. These results suggest that CRAMP from neutrophils may play an important role in protection against M. pneumoniae infection.


Assuntos
Catelicidinas/metabolismo , Mycoplasma pneumoniae/imunologia , Neutrófilos/imunologia , Pneumonia por Mycoplasma/imunologia , Animais , Peptídeos Catiônicos Antimicrobianos , Western Blotting , Líquido da Lavagem Broncoalveolar/química , Citoplasma/química , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Mycoplasma pneumoniae/crescimento & desenvolvimento , Mycoplasma pneumoniae/patogenicidade , Doenças dos Roedores/imunologia
12.
J Pept Sci ; 16(11): 607-12, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20648478

RESUMO

The widespread natural sources-derived cationic peptides have been reported to reveal bacterial killing and/or growth-inhibiting properties. Correspondingly, a number of artificial peptides have been designed to understand antibacterial mechanism of the cationic peptides. These peptides are expected to be an alternative antibiotic against drug-resistant pathogenic bacteria because major antimicrobial mechanism of cationic peptides involves bacterial membrane disorder, although those availabilities have not been well evaluated. In this study, cationic peptides containing Aib were prepared to evaluate the availability as an antimicrobial agent, especially against representative pathogenic bacteria. Among them, BRBA20, consisting of five repeated Aib-Arg-Aib-Ala sequences, showed strong antibacterial activity against both Gram-negative and Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus. Additionally, growth of Serratia marcescens and multidrug-resistant Pseudomonas aeruginosa, known as proteases-secreting pathogenic bacteria, were also completely inhibited by BRBA20 under 20 µg/ml peptide concentrations. Our results suggested availabilities of Aib-derived amphiphilicity and protease resistance in the design of artificial antimicrobial peptides. Comparing BRBA20 with BKBA20, it was also concluded that Arg residue is the preferred cationic source than Lys for antimicrobial action of amphiphilic helices.


Assuntos
Peptídeos Catiônicos Antimicrobianos/química , Tensoativos/química , Ácidos Aminoisobutíricos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Cátions , Permeabilidade da Membrana Celular/efeitos dos fármacos , Desenho de Fármacos , Escherichia coli/efeitos dos fármacos , Bicamadas Lipídicas/química , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Pseudomonas aeruginosa/efeitos dos fármacos , Serratia marcescens/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Tensoativos/farmacologia
13.
Cell Microbiol ; 10(7): 1491-504, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18331590

RESUMO

The Pseudomonas aeruginosa-derived alkaline protease (AprA), elastase A (LasA), elastase B (LasB) and protease IV are considered to play an important role in pathogenesis of this organism. Although the sequence analysis of P. aeruginosa genome predicts the presence of several genes encoding other potential proteases in the genome, little has been known about the proteases involving in pathogenesis. Recently, Porphyromonas gingivalis gingipains and Serratia marcescens serralysin have been shown to activate protease-activated receptor 2 (PAR-2), thereby modulating host inflammatory and immune responses. Accordingly, we hypothesized that unknown protease(s) from P. aeruginosa would also modulate such responses through PARs. In this study, we found that P. aeruginosa produces a novel large exoprotease (LepA) distinct from known proteases such as AprA, LasA, LasB and protease IV. Sequence analysis of LepA showed a molecular feature of the proteins transported by the two-partner secretion pathway. Our results indicated that LepA activates NF-kappaB-driven promoter through human PAR-1, -2 or -4 and cleaves the peptides corresponding to the tethered ligand region of human PAR-1, -2 and -4 at a specific site with exposure of their tethered ligands. Considered together, these results suggest that LepA would require PARs to modulate various host responses against bacterial infection.


Assuntos
Proteínas de Bactérias/metabolismo , Endopeptidases/metabolismo , NF-kappa B/metabolismo , Pseudomonas aeruginosa/enzimologia , Receptores Ativados por Proteinase/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Células COS , Bovinos , Linhagem Celular , Chlorocebus aethiops , Endopeptidases/genética , Regulação da Expressão Gênica , Humanos , Interleucina-8/metabolismo , Dados de Sequência Molecular , NF-kappa B/genética , Peptídeos/genética , Peptídeos/metabolismo , Regiões Promotoras Genéticas , Receptores Ativados por Proteinase/genética , Trombina/metabolismo , Tripsina/metabolismo
14.
Infect Immun ; 76(8): 3672-8, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18474641

RESUMO

Mycoplasma genitalium is a sexually transmitted bacterial pathogen that causes nongonococcal chlamydia-negative urethritis, mucopurulent cervicitis, endometritis, pelvic inflammatory disease, and tubal factor infertility in humans. However, pathogenic agents that induce inflammatory responses have not been identified in M. genitalium. In this study, we examined the involvement of Toll-like receptors (TLRs) in activation of the immune response by a lipoprotein from M. genitalium and their active component responsible for NF-kappaB activation. The Triton X-114 detergent phase of M. genitalium was found to induce NF-kappaB through TLR2. The active component of the Triton X-114 detergent phase was a lipoprotein precursor, MG149. The activation of NF-kappaB by MG149 was inhibited by a dominant negative (DN) construct of TLR1 but not by a DN construct of TLR6. These results indicate that the activation of NF-kappaB by MG149 is dependent on TLR1 and TLR2. A synthetic lipopeptide derived from MG149 containing three acyl chains also induced NF-kappaB through TLR1 and TLR2. Thus, the results show that MG149, a triacylated lipoprotein from M. genitalium, activates NF-kappaB through TLR1 and TLR2.


Assuntos
Lipoproteínas/imunologia , Mycoplasma genitalium/imunologia , NF-kappa B/metabolismo , Receptor 1 Toll-Like/imunologia , Receptor 2 Toll-Like/imunologia , Sequência de Aminoácidos , Linhagem Celular , Humanos , Lipoproteínas/isolamento & purificação , Dados de Sequência Molecular , Peptídeos/síntese química , Peptídeos/imunologia
15.
Infect Immun ; 76(1): 270-7, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17954722

RESUMO

The pathogenesis of Mycoplasma pneumoniae infection is considered to be in part attributable to excessive immune responses. In this study, we investigated whether synthetic lipopeptides of subunit b of F0F1-type ATPase (F0F1-ATPase), NF-kappaB-activating lipoprotein 1 (N-ALP1), and N-ALP2 (named FAM20, sN-ALP1, and sN-ALP2, respectively) derived from M. pneumoniae induce cytokine and chemokine production and leukocyte infiltration in vivo. Intranasal administration of FAM20 and sN-ALP2 induced infiltration of leukocyte cells and production of chemokines and cytokines in bronchoalveolar lavage fluid, but sN-ALP1 failed to do so. The activity of FAM20 was notably higher than that of sN-ALP2. FAM20 and sN-ALP2 induced tumor necrosis factor alpha (TNF-alpha) through Toll-like receptor 2 in mouse peritoneal macrophages. Moreover, in the range of low concentrations of lipopeptides, FAM20 showed relatively high activity of inducing TNF-alpha in mouse peritoneal macrophages compared to synthetic lipopeptides such as MALP-2 and FSL-1, derived from Mycoplasma fermentans and Mycoplasma salivarium, respectively. These findings indicate that the F0F1-ATPase might be a key molecule in inducing cytokines and chemokines contributing to inflammatory responses during M. pneumoniae infection in vivo.


Assuntos
Inflamação/induzido quimicamente , Lipoproteínas/toxicidade , Pulmão/efeitos dos fármacos , Mycoplasma pneumoniae/metabolismo , Administração Intranasal , Animais , Citocinas , Regulação da Expressão Gênica/efeitos dos fármacos , Leucócitos , Lipoproteínas/administração & dosagem , Lipoproteínas/síntese química , Lipoproteínas/metabolismo , Pulmão/citologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Conformação Proteica
16.
J Pharmacol Toxicol Methods ; 55(3): 248-53, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17141531

RESUMO

INTRODUCTION: The cynomolgus monkey, one of a number of primate species phylogenetically close to humans, is commonly used in cardiovascular research, but a method for determination of the RR interval-corrected QT interval in this species needs greater consideration. The objectives of this study were to determine a method for evaluating QT interval in cynomolgus monkeys individually, disregarding RR interval change artifacts, and to investigate prerequisite information for this method. METHODS: The physiological QT-RR relationship for practical evaluation of QT interval was recorded and analyzed by 24-hour telemetric ECG monitoring. A linear model for log-transformed QT and RR intervals was used to correct the QT interval from RR interval change artifacts for each animal. Sample size was also estimated based on the simulation results. RESULTS: Histograms showed that both QT and RR intervals had a right-heavy tail distribution. QT interval corrected individually by the linear model formula showed smaller within-animal variability than QTb and QTf, which were corrected by Bazett's formula and Fridericia's formula. The simulation results showed that the individual correction factor, beta(i), could be reliably estimated when at least 24 pairs of QT-RR baseline data were available. DISCUSSION: As with humans, QT interval in cynomolgus monkeys varies widely between individuals. Therefore, a method for correcting QT interval individually should be considered, whenever extensive untreated data are available.


Assuntos
Eletrocardiografia/veterinária , Frequência Cardíaca , Animais , Interpretação Estatística de Dados , Modelos Lineares , Síndrome do QT Longo/veterinária , Macaca fascicularis , Masculino , Modelos Animais , Especificidade da Espécie , Telemetria
17.
Mol Immunol ; 43(12): 1972-81, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16423398

RESUMO

The antimicrobial protein cathelicidin is considered to play an important role in the defense mechanisms against bacterial infection. Recent studies show that sodium butyrate induces cathelicidin gene expression in human colonic, gastric and hepatic cells. However, little is known about the precise regulatory mechanisms underlying sodium butyrate-induced cathelicidin gene expression. In this study, we examined the regulatory mechanisms involved in sodium butyrate-induced cathelicidin gene expression using a human lung epithelial cell line, EBC-1. Our results indicate that sodium butyrate induces both cathelicidin mRNA and protein expression. Moreover, deletion or mutation of a putative activator protein-1 (AP-1) binding site in the cathelicidin gene promoter abrogated the response to sodium butyrate stimulation. Three different mitogen-activated protein (MAP) kinase inhibitors suppressed sodium butyrate-induced transactivation of the cathelicidin promoter. Electrophoretic mobility shift assays (EMSA) showed that nuclear extracts prepared from sodium butyrate-stimulated EBC-1 cells generated specific binding to probe including a putative AP-1 binding site in the cathelicidin gene promoter. Furthermore, chromatin immunoprecipitation (ChIP) assays demonstrated that sodium butyrate augmented histone acetylation of the cathelicidin promoter in EBC-1 cells. Therefore, these results indicate that AP-1 and histone acetylation of the cathelicidin promoter play a critical role in the regulation of inducible cathelicidin gene expression in EBC-1 cells stimulated with sodium butyrate.


Assuntos
Peptídeos Catiônicos Antimicrobianos/imunologia , Butiratos/farmacologia , Regulação da Expressão Gênica/imunologia , Histonas/metabolismo , Pulmão/citologia , Fator de Transcrição AP-1/imunologia , Regulação para Cima/efeitos dos fármacos , Acetilação , Peptídeos Catiônicos Antimicrobianos/genética , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Catelicidinas , Linhagem Celular , Células Epiteliais/citologia , Deleção de Genes , Regulação da Expressão Gênica/genética , Humanos , Regiões Promotoras Genéticas , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Fator de Transcrição AP-1/genética
18.
J Vet Med Sci ; 79(9): 1472-1479, 2017 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-28757508

RESUMO

First-generation cephalosporins such as cefazolin (CEZ) have been widely used for mastitis treatment in dairy cattle. However, the use of antibiotics results in the presence of antibiotic residues in milk, which is used for human consumption. Nisin A, a bacteriocin produced by Lactococcus lactis, has been used as a broad-spectrum food preservative for over 50 years. Therefore, a combination of CEZ and nisin A might provide an extended activity spectrum against mastitis pathogens and reduce the antibiotic dose for mastitis treatment. This study aimed to evaluate the combined effect of CEZ and nisin A against mastitis pathogens using the checkerboard and time-kill assays. In the checkerboard assay, the CEZ-nisin A combination exhibited a synergistic effect against Staphylococcus aureus (n=20/20) and Enterococcus faecalis (n=13/18), and meanwhile exhibited a mostly additive effect against Staphylococcus intermedius (n=12/20), Streptococcus agalactiae (n=10/10), Streptococcus dysgalactiae (n=18/18), and Escherichia coli (n=14/18). There were no indifferent or antagonistic effects between CEZ and nisin A. In the time-kill assay, the CEZ-nisin A combination at 0.5 × or 1 × minimum inhibitory concentration exhibited synergistic reduction of bacterial growth by over 3 log10 colony forming units per ml relative to that observed with either antimicrobial substance alone. These results suggest that the CEZ-nisin A combination can be used for developing an intramammary infusion for mastitis treatment, with lower antibiotic concentrations than normal.


Assuntos
Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Cefazolina/farmacologia , Mastite Bovina/microbiologia , Nisina/farmacologia , Animais , Bovinos , Sinergismo Farmacológico , Enterococcus/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Mastite Bovina/tratamento farmacológico , Testes de Sensibilidade Microbiana , Staphylococcus/efeitos dos fármacos , Streptococcus/efeitos dos fármacos , Fatores de Tempo
19.
PLoS One ; 12(1): e0169367, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28046014

RESUMO

To specify critical factors responsible for Pseudomonas aeruginosa penetration through the Caco-2 cell epithelial barrier, we analyzed transposon insertion mutants that demonstrated a dramatic reduction in penetration activity relative to P. aeruginosa PAO1 strain. From these strains, mutations could be grouped into five classes, specifically flagellin-associated genes, pili-associated genes, heat-shock protein genes, genes related to the glycolytic pathway, and biosynthesis-related genes. Of these mutants, we here focused on the serA mutant, as the association between this gene and penetration activity is yet unknown. Inactivation of the serA gene caused significant repression of bacterial penetration through Caco-2 cell monolayers with decreased swimming and swarming motilities, bacterial adherence, and fly mortality rate, as well as repression of ExoS secretion; however, twitching motility was not affected. Furthermore, L-serine, which is known to inhibit the D-3-phosphoglycerate dehydrogenase activity of the SerA protein, caused significant reductions in penetration through Caco-2 cell monolayers, swarming and swimming motilities, bacterial adherence to Caco-2 cells, and virulence in flies in the wild-type P. aeruginosa PAO1 strain. Together, these results suggest that serA is associated with bacterial motility and adherence, which are mediated by flagella that play a key role in the penetration of P. aeruginosa through Caco-2 cell monolayers. Oral administration of L-serine to compromised hosts might have the potential to interfere with bacterial translocation and prevent septicemia caused by P. aeruginosa through inhibition of serA function.


Assuntos
Translocação Bacteriana/genética , Genes Bacterianos , Pseudomonas aeruginosa/genética , Sistemas de Secreção Bacterianos/genética , Células CACO-2 , Flagelos/efeitos dos fármacos , Flagelos/metabolismo , Humanos , Movimento , Mutação/genética , Fosfoglicerato Desidrogenase/metabolismo , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/patogenicidade , Serina/farmacologia , Virulência
20.
Int J Antimicrob Agents ; 26(5): 396-402, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16226432

RESUMO

Nisin, an amphipathic antibiotic peptide, is produced by a number of strains of Lactococcus lactis subsp. lactis. It has been employed as a food preservative as it has a high antibacterial activity with a relatively low toxicity for humans. Nisin is known to exert a high antibacterial activity against Gram-positive but not Gram-negative bacteria. However, purified nisin Z was found to show an antibacterial activity both against Gram-positive and Gram-negative bacteria. To clarify the mechanisms of activity, nisin Z and purified nisin Z were tested for their antibacterial activities in a high-salt environment. The activity of nisin Z against Staphylococcus aureus was stable even in the presence of NaCl at 100 mM, showing ca. 2log colony-forming unit (CFU) reduction. In contrast, the activity of nisin Z against Escherichia coli was highly sensitive to the same concentration of NaCl, and CFU reduction was not observed. Furthermore, purified nisin Z caused the permeabilisation both of S. aureus and E. coli cytoplasmic membranes. The permeabilisation of E. coli but not S. aureus cytoplasmic membranes was remarkably reduced in a high-salt environment. Moreover, vancomycin inhibited the nisin Z-induced permeabilisation of the S. aureus cytoplasmic membrane. These results suggest that nisin Z utilises two distinct mechanisms of antibacterial activity: a high-salt-sensitive mechanism for E. coli and a high-salt-insensitive mechanism for S. aureus.


Assuntos
Antibacterianos/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Nisina/análogos & derivados , Antibacterianos/isolamento & purificação , Membrana Celular/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Positivas/metabolismo , Humanos , Técnicas In Vitro , Nisina/isolamento & purificação , Nisina/farmacologia , Permeabilidade/efeitos dos fármacos , Cloreto de Sódio/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/metabolismo , Vancomicina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA