Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Microbiol ; 102(1): 137-51, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27353397

RESUMO

Neisseria gonorrhoeae produces two transferrin binding proteins, TbpA and TbpB, which together enable efficient iron transport from human transferrin. We demonstrate that expression of the tbp genes is controlled by MisR, a response regulator in the two-component regulatory system that also includes the sensor kinase MisS. The tbp genes were up-regulated in the misR mutant under iron-replete conditions but were conversely down-regulated in the misR mutant under iron-depleted conditions. The misR mutant was capable of transferrin-iron uptake at only 50% of wild-type levels, consistent with decreased tbp expression. We demonstrate that phosphorylated MisR specifically binds to the tbpBA promoter and that MisR interacts with five regions upstream of the tbpB start codon. These analyses confirm that MisR directly regulates tbpBA expression. The MisR binding sites in the gonococcus are only partially conserved in Neisseria meningitidis, which may explain why tbpBA was not MisR-regulated in previous studies using this related pathogen. This is the first report of a trans-acting protein factor other than Fur that can directly contribute to gonococcal tbpBA regulation.


Assuntos
Neisseria gonorrhoeae/genética , Proteína A de Ligação a Transferrina/genética , Proteína B de Ligação a Transferrina/genética , Sítios de Ligação , Ferro/metabolismo , Neisseria gonorrhoeae/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Transativadores/metabolismo , Transferrina/metabolismo , Proteína A de Ligação a Transferrina/metabolismo , Proteína B de Ligação a Transferrina/metabolismo
2.
PLoS One ; 10(12): e0144347, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26641098

RESUMO

Autophagy, an ancient homeostasis mechanism for macromolecule degradation, performs an important role in host defense by facilitating pathogen elimination. To counteract this host defense strategy, bacterial pathogens have evolved a variety of mechanisms to avoid or otherwise dysregulate autophagy by phagocytic cells so as to enhance their survival during infection. Neisseria gonorrhoeae is a strictly human pathogen that causes the sexually transmitted infection, gonorrhea. Phosphoethanolamine (PEA) addition to the 4' position of the lipid A (PEA-lipid A) moiety of the lipooligosaccharide (LOS) produced by gonococci performs a critical role in this pathogen's ability to evade innate defenses by conferring decreased susceptibility to cationic antimicrobial (or host-defense) peptides, complement-mediated killing by human serum and intraleukocytic killing by human neutrophils compared to strains lacking this PEA decoration. Heretofore, however, it was not known if gonococci can evade autophagy and if so, whether PEA-lipid A contributes to this ability. Accordingly, by using murine macrophages and human macrophage-like phagocytic cell lines we investigated if PEA decoration of gonococcal lipid A modulates autophagy formation. We report that infection with PEA-lipid A-producing gonococci significantly reduced autophagy flux in murine and human macrophages and enhanced gonococcal survival during their association with macrophages compared to a PEA-deficient lipid A mutant. Our results provide further evidence that PEA-lipid A produced by gonococci is a critical component in the ability of this human pathogen to evade host defenses.


Assuntos
Autofagia/fisiologia , Etanolaminas , Lipídeo A/metabolismo , Macrófagos/microbiologia , Neisseria gonorrhoeae/patogenicidade , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular/microbiologia , Quimiocinas/metabolismo , Dissacarídeos/farmacologia , Interações Hospedeiro-Patógeno , Humanos , Lipídeo A/química , Camundongos , Neisseria gonorrhoeae/metabolismo , Fagossomos/metabolismo , Fagossomos/microbiologia , Fosfatos Açúcares/farmacologia
3.
PLoS One ; 9(1): e87688, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24489950

RESUMO

Neisseria gonorrhoeae is a strict human pathogen that causes the sexually transmitted infection termed gonorrhea. The gonococcus can survive extracellularly and intracellularly, but in both environments the bacteria must acquire iron from host proteins for survival. However, upon infection the host uses a defensive response by limiting the bioavailability of iron by a number of mechanisms including the enhanced expression of hepcidin, the master iron-regulating hormone, which reduces iron uptake from the gut and retains iron in macrophages. The host also secretes the antibacterial protein NGAL, which sequesters bacterial siderophores and therefore inhibits bacterial growth. To learn whether intracellular gonococci can subvert this defensive response, we examined expression of host genes that encode proteins involved in modulating levels of intracellular iron. We found that N. gonorrhoeae can survive in association (tightly adherent and intracellular) with monocytes and macrophages and upregulates a panel of its iron-responsive genes in this environment. We also found that gonococcal infection of human monocytes or murine macrophages resulted in the upregulation of hepcidin, NGAL, and NRAMP1 as well as downregulation of the expression of the gene encoding the short chain 3-hydroxybutyrate dehydrogenase (BDH2); BDH2 catalyzes the production of the mammalian siderophore 2,5-DHBA involved in chelating and detoxifying iron. Based on these findings, we propose that N. gonorrhoeae can subvert the iron-limiting innate immune defenses to facilitate iron acquisition and intracellular survival.


Assuntos
Imunidade Inata , Ferro/metabolismo , Macrófagos/metabolismo , Neisseria gonorrhoeae/imunologia , Regulação para Baixo , Humanos , Hidroxibutirato Desidrogenase/genética , Macrófagos/imunologia , Neisseria gonorrhoeae/metabolismo , Virulência
4.
Antimicrob Agents Chemother ; 56(3): 1491-501, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22214775

RESUMO

Previous studies have shown that the MpeR transcriptional regulator produced by Neisseria gonorrhoeae represses the expression of mtrF, which encodes a putative inner membrane protein (MtrF). MtrF works as an accessory protein with the Mtr efflux pump, helping gonococci to resist high levels of diverse hydrophobic antimicrobials. Regulation of mpeR has been reported to occur by an iron-dependent mechanism involving Fur (ferric uptake regulator). Collectively, these observations suggest the presence of an interconnected regulatory system in gonococci that modulates the expression of efflux pump protein-encoding genes in an iron-responsive manner. Herein, we describe this connection and report that levels of gonococcal resistance to a substrate of the mtrCDE-encoded efflux pump can be modulated by MpeR and the availability of free iron. Using microarray analysis, we found that the mtrR gene, which encodes a direct repressor (MtrR) of mtrCDE, is an MpeR-repressed determinant in the late logarithmic phase of growth when free iron levels would be reduced due to bacterial consumption. This repression was enhanced under conditions of iron limitation and resulted in increased expression of the mtrCDE efflux pump operon. Furthermore, as judged by DNA-binding analysis, MpeR-mediated repression of mtrR was direct. Collectively, our results indicate that both genetic and physiologic parameters (e.g., iron availability) can influence the expression of the mtr efflux system and modulate levels of gonococcal susceptibility to efflux pump substrates.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Proteínas Reguladoras de Ferro/genética , Ferro/metabolismo , Neisseria gonorrhoeae/genética , Proteínas de Bactérias/metabolismo , Farmacorresistência Bacteriana Múltipla/genética , Loci Gênicos , Humanos , Proteínas Reguladoras de Ferro/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Neisseria gonorrhoeae/efeitos dos fármacos , Óperon , Plasmídeos , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
5.
Infect Immun ; 79(12): 4764-76, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21947770

RESUMO

Neisseria gonorrhoeae is an obligate human pathogen that causes the common sexually transmitted infection gonorrhea. Gonococcal infections cause significant morbidity, particularly among women, as the organism ascends to the upper reproductive tract, resulting in pelvic inflammatory disease, ectopic pregnancy, and infertility. In the last few years, antibiotic resistance rates have risen dramatically, leading to severe restriction of treatment options for gonococcal disease. Gonococcal infections do not elicit protective immunity, nor is there an effective vaccine to prevent the disease. Thus, further understanding of the expression, function, and regulation of surface antigens could lead to better treatment and prevention modalities in the future. In the current study, we determined that an iron-repressed regulator, MpeR, interacted specifically with the DNA sequence upstream of fetA and activated FetA expression. Interestingly, MpeR was previously shown to regulate the expression of gonococcal antimicrobial efflux systems. We confirmed that the outer membrane transporter FetA allows gonococcal strain FA1090 to utilize the xenosiderophore ferric enterobactin as an iron source. However, we further demonstrated that FetA has an extended range of substrates that encompasses other catecholate xenosiderophores, including ferric salmochelin and the dimers and trimers of dihydroxybenzoylserine. We demonstrated that fetA is part of an iron-repressed, MpeR-activated operon which putatively encodes other iron transport proteins. This is the first study to describe a regulatory linkage between antimicrobial efflux and iron transport in N. gonorrhoeae. The regulatory nidus that links these systems, MpeR, is expressed exclusively by pathogenic neisseriae and is therefore expected to be an important virulence factor.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Ferro/farmacologia , Neisseria gonorrhoeae/efeitos dos fármacos , Neisseria gonorrhoeae/metabolismo , Fatores de Transcrição/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , DNA Bacteriano/genética , Enterobactina/genética , Enterobactina/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação , Periplasma/metabolismo , Ligação Proteica , Fatores de Transcrição/genética , Regulação para Cima
6.
PLoS One ; 5(10): e13627, 2010 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-21049021

RESUMO

Capsular polysaccharides (CPS) are a major virulence factor in meningococcal infections and form the basis for serogroup designation and protective vaccines. Our work has identified meningococcal CPS as a pro-inflammatory ligand that functions through TLR2 and TLR4-MD2-dependent activation. We hypothesized that human cationic host defense peptides interact with CPS and influence its biologic activity. Accordingly, the interaction of meningococcal CPS with the human-derived cationic peptide LL-37, which is expressed by phagocytic and epithelial cells that interface with meningococci during infection, was investigated. LL-37 neutralized the pro-inflammatory activity of endotoxin-free CPS as assessed by TLR2 and TLR4-MD-2-dependent release of TNFα, IL-6 and IL-8 from human and murine macrophages. The cationic and hydrophobic properties of LL-37 were crucial for this inhibition, which was due to binding of LL-37 to CPS. LL-37 also inhibited the ability of meningococcal CPS to induce nitric oxide release, as well as TNFα and CXCL10 (IP-10) release from TLR4-sufficient and TLR4-deficient murine macrophages. Truncated LL-37 analogs, especially those that retained the antibacterial domain, inhibited vaccine grade CPS and meningococcal CPS prepared from the major serogroups (A, B C, Y and W135). Thus, LL-37 interaction with CPS was independent of specific glucan structure. We conclude that the capacity of meningococcal CPS to activate macrophages via TLR2 and TLR4-MD-2 can be inhibited by the human cationic host defense peptide LL-37 and propose that this impacts CPS-based vaccine responses.


Assuntos
Peptídeos Catiônicos Antimicrobianos/fisiologia , Neisseria meningitidis/metabolismo , Polissacarídeos Bacterianos/metabolismo , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/química , Linhagem Celular , Citocinas/metabolismo , Humanos , Macrófagos/metabolismo , Camundongos , Dados de Sequência Molecular , Catelicidinas
7.
Mol Microbiol ; 70(2): 462-78, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18761689

RESUMO

The MtrC-MtrD-MtrE efflux pump system confers resistance to macrolide antibiotics and antimicrobial substances of the host innate defence. Clinical isolates with increased resistance to erythromycin and azithromycin frequently harbour mutations in the mtrR structural gene, which encodes a repressor of the mtrCDE operon, or the mtrR promoter region. The MtrC-MtrD-MtrE system is important for gonococcal survival in the murine genital tract, and derepression of the mtrCDE operon via deletion of mtrR confers increased fitness in vivo. Here we compared isogenic strains with naturally occurring mtrR locus mutations for differences in mtrCDE expression and pump-related phenotypes. Mutations upstream of mtrC, including those within the MtrR binding region and a novel mutation that increases mtrC RNA stability conferred the highest levels of derepression as measured by mtrCDE transcription and resistance to antibiotics, progesterone and antimicrobial peptides. In contrast, mutations within the mtrR coding sequence conferred low to intermediate levels of derepression. In vivo, the mtr mutants were more fit than the wild-type strain, the degree to which paralleled in vitro resistance gradients. These studies establish a hierarchy of mtrR locus mutations with regard to regulation of pump efflux, and suggest selection for more derepressed mutants may occur during mixed infections.


Assuntos
Antibacterianos/farmacologia , Proteínas da Membrana Bacteriana Externa/biossíntese , Proteínas de Bactérias/biossíntese , Farmacorresistência Bacteriana , Lipoproteínas/biossíntese , Proteínas de Membrana/biossíntese , Proteínas de Membrana Transportadoras/biossíntese , Mutação , Neisseria gonorrhoeae/efeitos dos fármacos , Neisseria gonorrhoeae/patogenicidade , Animais , Peptídeos Catiônicos Antimicrobianos/farmacologia , Azitromicina/farmacologia , Proteínas de Bactérias/genética , Eritromicina/farmacologia , Feminino , Perfilação da Expressão Gênica , Gonorreia , Camundongos , Camundongos Endogâmicos BALB C , Neisseria gonorrhoeae/genética , Progesterona/farmacologia , Regiões Promotoras Genéticas , Estabilidade de RNA , Proteínas Repressoras/genética , Virulência
8.
J Bacteriol ; 189(14): 5399-402, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17496077

RESUMO

Retraction of type IV pili is mediated by PilT. We show that loss of pilT function leads to upregulation of mtrF (multiple transferable resistance) and two operons encoding putative ABC transporters in Neisseria gonorrhoeae MS11. This effect occurs indirectly through the transcriptional regulator FarR, which until now has been shown to regulate only farAB. L-Glutamine can reverse pilT downregulation of the ABC transporter operons and mtrF.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Adenosina Trifosfatases/fisiologia , Proteínas de Bactérias/genética , Proteínas Motores Moleculares/fisiologia , Neisseria gonorrhoeae/genética , Óperon , Adenosina Trifosfatases/genética , Proteínas de Bactérias/fisiologia , Farmacorresistência Bacteriana/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Glutamina/farmacologia , Proteínas Motores Moleculares/genética , Neisseria gonorrhoeae/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
9.
Cell Microbiol ; 7(9): 1251-62, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16098213

RESUMO

Antimicrobial peptides (AMPs), in addition to their antibacterial properties, are also chemotactic and signalling molecules that connect the innate and adaptive immune responses. The role of AMP [alpha defensins, LL-37, a cathepsin G-derived peptide (CG117-136), protegrins (PG-1), polymyxin B (PMX) and LLP1] in modulating the respiratory burst response in human and murine macrophages in the presence of bacterial endotoxin [lipopolysaccharide (LPS) or lipooligosaccharide (LOS)] was investigated. AMP were found to neutralize endotoxin induction of nitric oxide and TNFalpha release in macrophages in a dose-dependent manner. In contrast, macrophages primed overnight with AMP and LOS or LPS significantly enhanced reactive oxygen species (ROS) release compared with cells primed with endotoxin or AMP alone, while no responses were seen in unprimed cells. This enhanced ROS release by macrophages was seen in all cell lines including those obtained from C3H/HeJ (TLR4-/-) mice. Similar effects were also seen when AMP and endotoxin were added directly with zymosan to trigger phagocytosis and the respiratory burst in unprimed RAW 264.7 and C3H/HeJ macrophages. Amplification of ROS release was also demonstrated in a cell-free system of xanthine and xanthine oxidase. Although AMP inhibited cytokine and nitric oxide induction by endotoxin in a TLR4-dependent manner, AMP and endotoxin amplified ROS release in a TLR4-independent manner possibly by exerting a prolonged catalytic effect on the ROS generating enzymes such as the NADPH-oxidase complex.


Assuntos
Peptídeos Catiônicos Antimicrobianos/fisiologia , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Explosão Respiratória , Fator de Necrose Tumoral alfa/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/farmacologia , Linhagem Celular , Humanos , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Polimixina B/metabolismo , Polimixina B/farmacologia , Proteínas/farmacologia , Proteínas/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , alfa-Defensinas/farmacologia , alfa-Defensinas/fisiologia , Catelicidinas
10.
J Bacteriol ; 187(15): 5214-23, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16030215

RESUMO

Several genetic systems that allow the use of iron-protoporphyrin IX (heme) have been described for the pathogenic bacterium Neisseria meningitidis. However, many questions about the process of heme acquisition and utilization remain to be answered. To isolate and analyze unidentified genes that play a role in heme iron uptake and utilization, a Himar1 transposon mutant library was screened in N. meningitidis serogroup A strain IR4162. One locus identified by transposon mutagenesis conferred protection against heme toxicity. A mutant with a deletion in a gene termed ght (gene of hydrophobic agent tolerance) within this locus was susceptible to heme and other hydrophobic agents compared to the parental strain. Transcriptional analysis indicated that ght is cotranscribed with an upstream open reading frame NMA2149. Uncharacterized orthologues of ght were identified in many other gram-negative bacteria. We present genetic evidence for the importance of ght in resistance to hydrophobic agents and its potential role in interaction with other hydrophobic agent resistance mechanisms within N. meningitidis.


Assuntos
Genes Bacterianos , Heme/metabolismo , Ferro/metabolismo , Neisseria meningitidis/genética , Neisseria meningitidis/metabolismo , Sequência de Aminoácidos , Teste de Complementação Genética , Heme/toxicidade , Metaloporfirinas/toxicidade , Dados de Sequência Molecular , Neisseria meningitidis/efeitos dos fármacos , Fases de Leitura Aberta , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
11.
Infect Immun ; 72(10): 5712-21, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15385470

RESUMO

Cathepsin G (CatG) is a serine protease found in the azurophilic granules of monocytes that is known to have antimicrobial properties, but its role in Mycobacterium tuberculosis infection is unknown. We found that M. tuberculosis infection of human THP-1 monocytic cells induced the down-regulation of CatG mRNA expression, as demonstrated by gene array analysis and reverse transcription-PCR. This was associated with a concomitant decrease in CatG protein and enzymatic activity. In contrast, the expression of lysosomal cathepsins B and D was up-regulated in infected cells. This effect was also observed when THP-1 cells were induced to differentiate into adherent macrophages by exposure to bacterial lipopolysaccharide (LPS). In agreement with this, CatG expression was null in adherent macrophages isolated from bronchoalveolar lavages and normal blood. We wanted to determine if the down-regulation of CatG would be relevant to M. tuberculosis infection. First, we found that addition of CatG to THP-1 cells prior to infection resulted in decreased bacillary viability, presumably due to extracellular killing of bacilli. However, pretreatment of cells with LPS, which decreases intracellular CatG expression, resulted in increased bacillary viability. Second, we found that CatG cationic peptides killed M. tuberculosis bacilli and were five- to sevenfold more bactericidal than full-length CatG. These observations suggest that M. tuberculosis infection of human monocytic cells results in a "cathepsin switch" with down-regulation of CatG rendering M. tuberculosis bacilli more viable. Therefore, the down-regulation of CatG in macrophages is advantageous to M. tuberculosis bacilli and possibly is an important mechanism by which M. tuberculosis is able to evade the host immune defenses.


Assuntos
Catepsinas/metabolismo , Regulação para Baixo , Espaço Intracelular/microbiologia , Monócitos/metabolismo , Monócitos/microbiologia , Mycobacterium tuberculosis/citologia , Mycobacterium tuberculosis/fisiologia , Lavagem Broncoalveolar , Catepsina G , Catepsinas/biossíntese , Catepsinas/genética , Catepsinas/farmacologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Humanos , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/microbiologia , Monócitos/citologia , Monócitos/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/farmacologia , Serina Endopeptidases
12.
J Bacteriol ; 186(3): 730-9, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14729699

RESUMO

A spontaneous point mutation in pilQ (pilQ1) resulted in phenotypic suppression of a hemoglobin (Hb) receptor mutant (hpuAB mutant), allowing gonococci to grow on Hb as the sole source of iron. PilQ, formerly designated OMP-MC, is a member of the secretin family of proteins located in the outer membrane and is required for pilus biogenesis. The pilQ1 mutant also showed decreased piliation and transformation efficiency. Insertional inactivation of pilQ1 resulted in the loss of the Hb utilization phenotype and decreased entry of free heme. Despite the ability of the pilQ1 mutant to use Hb for iron acquisition and porphyrin, there was no demonstrable binding of Hb to the cell surface. The pilQ1 mutant was more sensitive to the toxic effect of free heme in growth medium and hypersensitive to the detergent Triton X-100 and multiple antibiotics. Double mutation in pilQ1 and tonB had no effect on these phenotypes, but a double pilQ1 pilT mutant showed a reduction in Hb-dependent growth and decreased sensitivity to heme and various antimicrobial agents. Insertional inactivation of wild-type pilQ also resulted in reduced entry of heme, Triton X-100, and some antibiotics. These results show that PilQ forms a channel that allows entry of heme and certain antimicrobial compounds and that a gain-of function point mutation in pilQ results in TonB-independent, PilT-dependent increase of entry.


Assuntos
Anti-Infecciosos/farmacocinética , Proteínas de Fímbrias/fisiologia , Heme/metabolismo , Neisseria gonorrhoeae/química , Adenosina Trifosfatases/fisiologia , Proteínas de Bactérias/fisiologia , Fímbrias Bacterianas/fisiologia , Proteínas de Membrana/fisiologia , Proteínas Motores Moleculares/fisiologia , Fenótipo , Mutação Puntual , Transporte Proteico , Transformação Bacteriana
13.
Infect Immun ; 71(10): 5576-82, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14500476

RESUMO

Active efflux of antimicrobial substances is likely to be an important bacterial defense against inhibitory host factors inherent to different body sites. Two well-characterized multidrug resistance efflux systems (MtrCDE and FarAB-MtrE) exist in Neisseria gonorrhoeae, a bacterial pathogen of the human genital mucosae. In vitro studies suggest that the MtrCDE and FarAB-MtrE efflux systems protect the gonococcus from hydrophobic antimicrobial substances that are likely to be present on mucosal surfaces. Here we report that a functional MtrCDE efflux system, but not a functional FarAB-MtrE system, enhances experimental gonococcal genital tract infection in female mice. Specifically, the recovery of mtrD and mtrE mutants, but not a farB mutant, from mice inoculated with mutant or wild-type gonococci was reduced compared with that of the wild-type strain. Competitive-infection experiments confirmed the survival disadvantage of MtrCDE-deficient gonococci. This report is the first direct evidence that a multidrug resistance efflux system enhances survival of a bacterial pathogen in the genital tract. Additionally, experiments using ovariectomized mice showed that MtrCDE-deficient gonococci were more rapidly cleared from mice that were capable of secreting gonadal hormones. MtrCDE-deficient gonococci were more sensitive to nonphysiological concentrations of progesterone in vitro than were wild-type or FarAB-MtrE-deficient gonococci. These results suggest that progesterone may play an inhibitory role in vivo. However, hormonally regulated factors rather than progesterone itself may be responsible for the more rapid clearance of mtr-deficient gonococci from intact mice.


Assuntos
Proteínas de Bactérias , Proteínas de Membrana Transportadoras , Neisseria gonorrhoeae/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Farmacorresistência Bacteriana Múltipla , Estradiol/metabolismo , Estradiol/farmacologia , Feminino , Gonorreia/imunologia , Gonorreia/metabolismo , Gonorreia/microbiologia , Técnicas In Vitro , Lipoproteínas/genética , Lipoproteínas/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Neisseria gonorrhoeae/efeitos dos fármacos , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/imunologia , Progesterona/metabolismo , Progesterona/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA