Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 290
Filtrar
1.
Med Microbiol Immunol ; 209(5): 621-630, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32607764

RESUMO

Outer inflammatory protein (OipA) is an important virulence factor of Helicobacter pylori (H. pylori), but the correlation between oipA copy number and its virulence remains unknown. The study was designed to investigate whether the duplicate oipA gene loci showed more virulent than one oipA gene in vitro. H. pylori strain CCS9803 (China Chongqing Strain 9803) that carries duplicate oipA loci was used to construct one or two oipA knockout mutant strain, which was further verified by qPCR and western blot. Gastric epithelial cells AGS and GES-1 were infected with wild-type (WT) or oipA mutants for 6 or 24 h. The expression levels of IL-8, bacterial adhesion, cell apoptosis and cell cycle were performed to analyze the function of oipA. The WT and oipA mutant strains induce significantly higher mRNA and protein levels of IL-8 than the uninfected group (P < 0.05), but only oipA2 mutants induced significantly decreased expression levels than the WT-infected group (P < 0.05). Adherence to gastric cells was significantly decreased by inactivated two oipA loci (P < 0.05). The WT strain caused a significant rising proportion of early apoptosis cell, which had dropped after duplicate oipA genes were both knockout (P < 0.05). WT and oipA1 mutants failed to affect cell cycle; however, the oipA2 mutants increased M phase and reduced S phase when compared to the uninfected group. In conclusion, our study demonstrated that oipA impacts IL-8 expression, adherence, cell apoptosis and cell cycle of gastric cells independent of its gene copy number.


Assuntos
Proteínas da Membrana Bacteriana Externa , Infecções por Helicobacter/microbiologia , Helicobacter pylori , Interleucina-8/metabolismo , Fatores de Virulência , Apoptose , Aderência Bacteriana , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/fisiologia , Ciclo Celular , Células Cultivadas , Variações do Número de Cópias de DNA , Células Epiteliais/microbiologia , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Helicobacter pylori/patogenicidade , Humanos , Virulência , Fatores de Virulência/genética , Fatores de Virulência/fisiologia
2.
Mol Microbiol ; 114(2): 214-229, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32239779

RESUMO

A large subfamily of the type IV secretion systems (T4SSs), termed the conjugation systems, transmit mobile genetic elements (MGEs) among many bacterial species. In the initiating steps of conjugative transfer, DNA transfer and replication (Dtr) proteins assemble at the origin-of-transfer (oriT) sequence as the relaxosome, which nicks the DNA strand destined for transfer and couples the nicked substrate with the VirD4-like substrate receptor. Here, we defined contributions of the Dtr protein TraK, a predicted member of the Ribbon-Helix-Helix (RHH) family of DNA-binding proteins, to transfer of DNA and protein substrates through the pKM101-encoded T4SS. Using a combination of cross-linking/affinity pull-downs and two-hybrid assays, we determined that TraK self-associates as a probable tetramer and also forms heteromeric contacts with pKM101-encoded TraI relaxase, VirD4-like TraJ receptor, and VirB11-like and VirB4-like ATPases, TraG and TraB, respectively. TraK also promotes stable TraJ-TraB complex formation and stimulates binding of TraI with TraB. Finally, TraK is required for or strongly stimulates the transfer of cognate (pKM101, TraI relaxase) and noncognate (RSF1010, MobA relaxase) substrates. We propose that TraK functions not only to nucleate pKM101 relaxosome assembly, but also to activate the TrapKM101 T4SS via interactions with the ATPase energy center positioned at the channel entrance.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Escherichia coli/metabolismo , Nucleoproteínas/metabolismo , Proteínas Periplásmicas/metabolismo , Sistemas de Secreção Tipo IV/metabolismo , Adenosina Trifosfatases/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas da Membrana Bacteriana Externa/fisiologia , Proteínas de Bactérias/metabolismo , Conjugação Genética/genética , DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/fisiologia , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiologia , Proteínas de Membrana/metabolismo , Nucleoproteínas/fisiologia , Proteínas Periplásmicas/fisiologia , Plasmídeos/genética
3.
Mol Microbiol ; 113(5): 1038-1051, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31975447

RESUMO

Cells interact with their surrounding environment through surface proteins. However, knowledge gaps remain in understanding how these important types of proteins are transported and anchored on the cell surface. In the Gram-negative social bacterium, Myxococcus xanthus, a putative C-terminal sorting tag (MYXO-CTERM) is predicted to help direct 34 different proteins onto the cell surface. Here we investigate the sorting pathway for MYXO-CTERM proteins by using the TraA cell surface receptor as a paradigm. Deleting this motif from TraA abolishes the cell surface anchoring and results in extracellular secretion. Our findings indicate that conserved cysteines within the MYXO-CTERM are posttranslationally modified and are required for TraA cell surface localization and function. A region immediately upstream of these residues is predicted to be disordered and removing this motif caused a secretion defect and blocked cell surface anchoring. We further show that the type II secretion system is required for translocation across the outer membrane and that a cysteine-rich region directs TraA to the T2SS. Similar results were found with another MYXO-CTERM protein indicating our findings can be generalized. Further, we show the universal distribution of MXYO-CTERM motif across the Myxococcales order and provide a working model for sorting of these proteins.


Assuntos
Proteínas da Membrana Bacteriana Externa/fisiologia , Membrana Celular/fisiologia , Myxococcus xanthus/fisiologia , Transporte Proteico , Receptores de Superfície Celular/fisiologia , Sistemas de Secreção Tipo II/fisiologia , Domínios e Motivos de Interação entre Proteínas , Processamento de Proteína Pós-Traducional
4.
PLoS Pathog ; 15(9): e1007972, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31487328

RESUMO

The biogenesis of bacterial cell-envelope polysaccharides requires the translocation, across the plasma membrane, of sugar sub-units that are produced inside the cytoplasm. To this end, the hydrophilic sugars are anchored to a lipid phosphate carrier (undecaprenyl phosphate (C55-P)), yielding membrane intermediates which are translocated to the outer face of the membrane. Finally, the glycan moiety is transferred to a nascent acceptor polymer, releasing the carrier in the "inactive" undecaprenyl pyrophosphate (C55-PP) form. Thus, C55-P is generated through the dephosphorylation of C55-PP, itself arising from either de novo synthesis or recycling. Two types of integral membrane C55-PP phosphatases were described: BacA enzymes and a sub-group of PAP2 enzymes (type 2 phosphatidic acid phosphatases). The human pathogen Helicobacter pylori does not contain BacA homologue but has four membrane PAP2 proteins: LpxE, LpxF, HP0350 and HP0851. Here, we report the physiological role of HP0851, renamed HupA, via multiple and complementary approaches ranging from a detailed biochemical characterization to the assessment of its effect on cell envelope metabolism and microbe-host interactions. HupA displays a dual function as being the main C55-PP pyrophosphatase (UppP) and phosphatidylglycerol phosphate phosphatase (PGPase). Although not essential in vitro, HupA was essential in vivo for stomach colonization. In vitro, the remaining UppP activity was carried out by LpxE in addition to its lipid A 1-phosphate phosphatase activity. Both HupA and LpxE have crucial roles in the biosynthesis of several cell wall polysaccharides and thus constitute potential targets for new therapeutic strategies.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Helicobacter pylori/metabolismo , Sequência de Aminoácidos , Animais , Proteínas da Membrana Bacteriana Externa/fisiologia , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Parede Celular/metabolismo , Proteínas de Ligação a DNA , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Feminino , Helicobacter pylori/patogenicidade , Camundongos , Camundongos Endogâmicos , Testes de Sensibilidade Microbiana , Fosfatidato Fosfatase , Monoéster Fosfórico Hidrolases/metabolismo , Fosfatos de Poli-Isoprenil/metabolismo , Polimixina B/farmacologia , Pirofosfatases/metabolismo , Estômago
5.
FASEB J ; 33(10): 10808-10818, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31262188

RESUMO

Colonization of the oropharynx is the initial step in Group A Streptococcus (GAS) pharyngeal infection. We have previously reported that the highly virulent M1T1 GAS clone attaches to oral epithelial cells via M1 protein interaction with blood group antigen carbohydrate structures. Here, we have identified that colonization of human oral epithelial cells by GAS serotypes M3 and M12 is mediated by human blood group antigens [ABO(H)] and Lewis (Le) antigen expression. Removal of linkage-specific fucose, galactose, N-acetylgalactosamine, and sialic acid modulated GAS colonization, dependent on host ABO(H) blood group and Le expression profile. Furthermore, N-linked glycans from human salivary glycoproteins, when released and purified, were potent inhibitors of M1, M3, and M12 GAS colonization ex vivo. These data highlight the important role played by human protein glycosylation patterns in GAS attachment to oral epithelial cell surfaces.-De Oliveira, D. M. P., Everest-Dass, A., Hartley-Tassell, L., Day, C. J., Indraratna, A., Brouwer, S., Cleary, A., Kautto, L., Gorman, J., Packer, N. H., Jennings, M. P., Walker, M. J., Sanderson-Smith, M. L. Human glycan expression patterns influence Group A streptococcal colonization of epithelial cells.


Assuntos
Interações entre Hospedeiro e Microrganismos/fisiologia , Polissacarídeos/metabolismo , Streptococcus pyogenes/patogenicidade , Antígenos de Bactérias/fisiologia , Aderência Bacteriana/imunologia , Aderência Bacteriana/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Antígenos de Grupos Sanguíneos/química , Proteínas de Transporte/fisiologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Glicosilação , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Técnicas In Vitro , Polissacarídeos/química , Polissacarídeos/imunologia , Ligação Proteica , Proteínas e Peptídeos Salivares/química , Proteínas e Peptídeos Salivares/imunologia , Proteínas e Peptídeos Salivares/metabolismo , Infecções Estreptocócicas/etiologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/crescimento & desenvolvimento , Streptococcus pyogenes/fisiologia , Virulência/fisiologia
6.
Sci Rep ; 9(1): 4656, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30874582

RESUMO

Peptidoglycan is a major component of the bacterial cell wall and thus a major determinant of cell shape. Its biosynthesis is initiated by several sequential reactions catalyzed by cytoplasmic Mur enzymes. Mur ligases (MurC, -D, -E, and -F) are essential for bacteria, metabolize molecules not present in eukaryotes, and are structurally and biochemically tractable. However, although many Mur inhibitors have been developed, few have shown promising antibacterial activity, prompting the hypothesis that within the cytoplasm, Mur enzymes could exist as a complex whose architecture limits access of small molecules to their active sites. This suggestion is supported by the observation that in many bacteria, mur genes are present in a single operon, and pairs of these genes often are fused to generate a single polypeptide. Here, we explored this genetic arrangement in the human pathogen Bordetella pertussis and show that MurE and MurF are expressed as a single, bifunctional protein. EM, small angle X-ray scattering (SAXS), and analytical centrifugation (AUC) revealed that the MurE-MurF fusion displays an elongated, flexible structure that can dimerize. Moreover, MurE-MurF interacted with the peripheral glycosyltransferase MurG, which formed discrete oligomers resembling 4- or 5-armed stars in EM images. The oligomeric structure of MurG may allow it to play a bona fide scaffolding role for a potential Mur complex, facilitating the efficient conveyance of peptidoglycan-building blocks toward the inner membrane leaflet. Our findings shed light on the structural determinants of a peptidoglycan formation complex involving Mur enzymes in bacterial cell wall formation.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Bordetella pertussis/genética , Bordetella pertussis/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Antibacterianos/farmacologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/fisiologia , Proteínas de Bactérias/metabolismo , Sítios de Ligação/fisiologia , Bordetella pertussis/patogenicidade , Domínio Catalítico/fisiologia , Parede Celular/metabolismo , Citoplasma/metabolismo , Glicosiltransferases/metabolismo , Glicosiltransferases/fisiologia , Humanos , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/fisiologia , Peptídeo Sintases/metabolismo , Peptidoglicano/biossíntese , Peptidoglicano/metabolismo , Ligação Proteica/fisiologia , Espalhamento a Baixo Ângulo , Difração de Raios X/métodos
7.
BMC Res Notes ; 11(1): 539, 2018 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-30068381

RESUMO

OBJECTIVE: The intestine is the major defensive barrier in the body by having more than 60% of the immune cells in the intestinal mucosa. The aim of this study was to evaluate the Toll like receptor (TLR) signaling pathways and immune response profiles, against outer membrane vesicles (OMVs) in pathogenic and non-pathogenic strains of Escherichia coli. RESULTS: Our results demonstrated that despite inducing inflammatory and regulatory responses to OMVs released by both strains, there is a remarkable difference in the nature and severity of these responses between the two strains. Following the production and release of OMV by the pathogenic strain, the expressions of the pro-inflammatory cytokines were significantly elevated, in comparison to the non-pathogenic strains. Eventually, our findings suggest that OMV released by the pathogen strain might be colonized, causing inflammation, eliminating the tight junctions of epithelial cells and damaging underlying cells, without the presence of IL-17 at the inflammation site. This could have happened to prevent the development of more severe inflammation, which could lead to the inhibition of colonization. The production of IL-10 is also preventing such inflammations. On the other hand, OMV released by non-pathogenic E. coli appears to influence intestinal homeostasis by causing more anti-inflammatory responses and mild inflammation.


Assuntos
Proteínas da Membrana Bacteriana Externa/fisiologia , Escherichia coli/fisiologia , Inflamação , Mucosa Intestinal , Transdução de Sinais , Células Epiteliais , Humanos
8.
Biochem Soc Trans ; 46(5): 1021-1027, 2018 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-30154095

RESUMO

As part of their life cycle, Gram-negative bacteria produce and release microvesicles (outer membrane vesicles, OMVs) consisting of spherical protrusions of the outer membrane that encapsulate periplasmic contents. OMVs produced by commensal bacteria in the gastrointestinal (GI) tract of animals are dispersed within the gut lumen with their cargo and enzymes being distributed across and throughout the GI tract. Their ultimate destination and fate is unclear although they can interact with and cross the intestinal epithelium using different entry pathways and access underlying immune cells in the lamina propria. OMVs have also been found in the bloodstream from which they can access various tissues and possibly the brain. The nanosize and non-replicative status of OMVs together with their resistance to enzyme degradation and low pH, alongside their ability to interact with the host, make them ideal candidates for delivering biologics to mucosal sites, such as the GI and the respiratory tract. In this mini-review, we discuss the fate of OMVs produced in the GI tract of animals with a focus on vesicles released by Bacteroides species and the use of OMVs as vaccine delivery vehicles and other potential applications.


Assuntos
Proteínas da Membrana Bacteriana Externa/fisiologia , Microbioma Gastrointestinal , Bactérias Gram-Negativas/fisiologia , Vesículas Secretórias/fisiologia , Animais , Bacteroides , Encéfalo/microbiologia , Células Epiteliais/microbiologia , Trato Gastrointestinal/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Sistema Imunitário , Mucosa Intestinal/microbiologia
9.
Sci Rep ; 8(1): 6340, 2018 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-29679057

RESUMO

Legionella pneumophila invades protozoa with an "accidental" ability to cause pneumonia upon transmission to humans. To support its nutrition during intracellular residence, L. pneumophila relies on host amino acids as the main source of carbon and energy to feed the TCA cycle. Despite the apparent lack of a requirement for glucose for L. pneumophila growth in vitro and intracellularly, the organism contains multiple amylases, which hydrolyze polysaccharides into glucose monomers. Here we describe one predicted putative amylase, LamB, which is uniquely present only in L. pneumophila and L. steigerwaltii among the ~60 species of Legionella. Our data show that LamB has a strong amylase activity, which is abolished upon substitutions of amino acids that are conserved in the catalytic pocket of amylases. Loss of LamB or expression of catalytically-inactive variants of LamB results in a severe growth defect of L. pneumophila in Acanthamoeba polyphaga and human monocytes-derived macrophages. Importantly, the lamB null mutant is severely attenuated in intra-pulmonary proliferation in the mouse model and is defective in dissemination to the liver and spleen. Our data show an essential role for LamB in intracellular replication of L. pneumophila in amoeba and human macrophages and in virulence in vivo.


Assuntos
Amilases/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Legionella pneumophila/metabolismo , Porinas/metabolismo , Receptores Virais/metabolismo , Amoeba , Amilases/fisiologia , Animais , Proteínas da Membrana Bacteriana Externa/fisiologia , Proteínas de Bactérias , Humanos , Legionella , Legionella pneumophila/patogenicidade , Macrófagos/fisiologia , Camundongos , Porinas/fisiologia , Receptores Virais/fisiologia , Virulência , Fatores de Virulência
10.
Can J Microbiol ; 64(6): 376-384, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29510061

RESUMO

Chlamydia trachomatis is the most common cause of bacterial sexually transmitted infections. The effect of antibiotic treatment is not satisfactory, and there is currently no vaccine to prevent C. trachomatis infection. Our results showed that Chlamydia virus CPG1 capsid protein Vp1 treatment significantly inhibited C. trachomatis growth in cell culture, and the inclusion numbers of different C. trachomatis serotypes were decreased. In addition, we conducted a preliminary investigation of the possible mechanisms behind the Vp1 inhibition effects and the C. trachomatis molecules targeted by Vp1. Using far-western blot and GST pull-down assay, we found that purified Vp1 can bind to the C. trachomatis outer membrane protein PmpI. PmpI polyclonal antibody treatment markedly reduced the inhibitory effect of Vp1 on C. trachomatis infectivity. On the basis of these experimental results, we infer that PmpI participates in the inhibitory effect of Vp1 and may be a potential receptor of Vp1 in the outer membrane of C. trachomatis. Our research provides clues regarding the molecular mechanisms underlying the interactions between chlamydia virus and chlamydia.


Assuntos
Anticorpos Antibacterianos/farmacologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Chlamydia trachomatis/efeitos dos fármacos , Proteínas Estruturais Virais/farmacologia , Chlamydia trachomatis/patogenicidade , Feminino , Células HeLa , Humanos
11.
PLoS One ; 11(8): e0160977, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27513341

RESUMO

Francisella tularensis is the causative agent of the lethal disease tularemia. Despite decades of research, little is understood about why F. tularensis is so virulent. Bacterial outer membrane proteins (OMPs) are involved in various virulence processes, including protein secretion, host cell attachment, and intracellular survival. Many pathogenic bacteria require metals for intracellular survival and OMPs often play important roles in metal uptake. Previous studies identified three F. tularensis OMPs that play roles in iron acquisition. In this study, we examined two previously uncharacterized proteins, FTT0267 (named fmvA, for Francisella metal and virulence) and FTT0602c (fmvB), which are homologs of the previously studied F. tularensis iron acquisition genes and are predicted OMPs. To study the potential roles of FmvA and FmvB in metal acquisition and virulence, we first examined fmvA and fmvB expression following pulmonary infection of mice, finding that fmvB was upregulated up to 5-fold during F. tularensis infection of mice. Despite sequence homology to previously-characterized iron-acquisition genes, FmvA and FmvB do not appear to be involved iron uptake, as neither fmvA nor fmvB were upregulated in iron-limiting media and neither ΔfmvA nor ΔfmvB exhibited growth defects in iron limitation. However, when other metals were examined in this study, magnesium-limitation significantly induced fmvB expression, ΔfmvB was found to express significantly higher levels of lipopolysaccharide (LPS) in magnesium-limiting medium, and increased numbers of surface protrusions were observed on ΔfmvB in magnesium-limiting medium, compared to wild-type F. tularensis grown in magnesium-limiting medium. RNA sequencing analysis of ΔfmvB revealed the potential mechanism for increased LPS expression, as LPS synthesis genes kdtA and wbtA were significantly upregulated in ΔfmvB, compared with wild-type F. tularensis. To provide further evidence for the potential role of FmvB in magnesium uptake, we demonstrated that FmvB was outer membrane-localized. Finally, ΔfmvB was found to be attenuated in mice and cytokine analyses revealed that ΔfmvB-infected mice produced lower levels of pro-inflammatory cytokines, including GM-CSF, IL-3, and IL-10, compared with mice infected with wild-type F. tularensis. Taken together, although the function of FmvA remains unknown, FmvB appears to play a role in magnesium uptake and F. tularensis virulence. These results may provide new insights into the importance of magnesium for intracellular pathogens.


Assuntos
Proteínas da Membrana Bacteriana Externa/fisiologia , Francisella tularensis/patogenicidade , Magnésio/metabolismo , Sequência de Aminoácidos , Animais , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/genética , Células Cultivadas , Citocinas/metabolismo , Feminino , Deleção de Genes , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Células Hep G2 , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Alinhamento de Sequência , Análise de Sequência de RNA , Regulação para Cima , Virulência/genética
12.
BMC Microbiol ; 16(1): 161, 2016 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-27448791

RESUMO

BACKGROUND: Iron homeostasis is an essential process over the entire lives of both hosts and bacterial pathogens, and also plays roles in many other metabolic functions. Currently, knowledge is limited on the iron scavenging mechanism of the cell envelope in the aquatic pathogen, Aeromonas hydrophila. To understand the iron homeostasis mechanism in A. hydrophila, a dimethyl labelling based quantitative proteomics method was used to compare the differential expression of cell envelope proteins under iron starvation. RESULTS: A total of 542 cell envelope proteins were identified by LC-MS/MS, with 66 down-regulated and 104 up-regulated proteins. Bioinformatics analysis showed that outer membrane siderophores, heme and iron receptors, periplasmic iron binding proteins, inner membrane ABC transporters and H(+)-ATP synthase subunits increased in abundance while iron-cluster proteins, electron transport chain and redox proteins were down-regulated. Further q-PCR validation, in vivo addition of exogenous metabolites, and an enzyme inhibition assay revealed that redox, the energy generation process, and ATP synthase elevated the susceptibility of A. hydrophila to iron starvation. CONCLUSIONS: Our study demonstrates that the redox and energy generation process, and ATP synthase in A. hydrophila may play critical roles in iron acquisition under conditions of iron-stress. An understanding of the iron scavenging mechanism may be helpful for the development of strategies for preventing and treating A. hydrophila infection.


Assuntos
Aeromonas hydrophila/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Ferro/metabolismo , Proteômica/métodos , Inanição/metabolismo , Estresse Fisiológico , Aeromonas hydrophila/enzimologia , Aeromonas hydrophila/genética , Proteínas da Membrana Bacteriana Externa/biossíntese , Proteínas da Membrana Bacteriana Externa/isolamento & purificação , Proteínas da Membrana Bacteriana Externa/fisiologia , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/isolamento & purificação , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/fisiologia , Regulação para Baixo , Complexo de Proteínas da Cadeia de Transporte de Elétrons , Ativação Enzimática , Ensaios Enzimáticos , Regulação Bacteriana da Expressão Gênica , Heme/metabolismo , Homeostase , Proteínas de Ligação ao Ferro , RNA Mensageiro/análise , Sideróforos/metabolismo , Espectrometria de Massas em Tandem , Regulação para Cima
13.
J Infect Dis ; 213(2): 314-23, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26153407

RESUMO

Interaction with the extracellular matrix (ECM) is one of the successful colonization strategies employed by nontypeable Haemophilus influenzae (NTHi). Here we identified Haemophilus lipoprotein e (P4) as a receptor for ECM proteins. Purified recombinant P4 displayed a high binding affinity for laminin (Kd = 9.26 nM) and fibronectin (Kd = 10.19 nM), but slightly less to vitronectin (Kd = 16.51 nM). A P4-deficient NTHi mutant showed a significantly decreased binding to these ECM components. Vitronectin acquisition conferred serum resistance to both P4-expressing NTHi and Escherichia coli transformants. P4-mediated bacterial adherence to pharynx, type II alveolar, and bronchial epithelial cells was mainly attributed to fibronectin. Importantly, a significantly reduced bacterial infection was observed in the middle ear of the Junbo mouse model when NTHi was devoid of P4. In conclusion, our data provide new insight into the role of P4 as an important factor for Haemophilus colonization and subsequent respiratory tract infection.


Assuntos
Aderência Bacteriana/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Esterases/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/classificação , Haemophilus influenzae/fisiologia , Lipoproteínas/fisiologia , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fibronectinas/fisiologia , Regulação Bacteriana da Expressão Gênica , Laminina/fisiologia , Proteína do Locus do Complexo MDS1 e EVI1 , Camundongos , Otite Média/microbiologia , Ligação Proteica , Proto-Oncogenes/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Vitronectina/fisiologia
14.
J Infect Dis ; 210(9): 1367-75, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24813471

RESUMO

Plague is transmitted by fleas or contaminated aerosols. To successfully produce disease, the causal agent (Yersinia pestis) must rapidly sense and respond to rapid variations in its environment. Here, we investigated the role of 2-component regulatory systems (2CSs) in plague because the latter are known to be key players in bacterial adaptation to environmental change. Along with the previously studied PhoP-PhoQ system, OmpR-EnvZ was the only one of Y. pestis' 23 other 2CSs required for production of bubonic, septicemic, and pneumonic plague. In vitro, OmpR-EnvZ was needed to counter serum complement and leukocytes but was not required for the secretion of antiphagocyte exotoxins. In vivo, Y. pestis lacking OmpR-EnvZ did not induce an early immune response in the skin and was fully virulent in neutropenic mice. We conclude that, throughout the course of Y. pestis infection, OmpR-EnvZ is required to counter toxic effectors secreted by polymorphonuclear leukocytes in the tissues.


Assuntos
Proteínas da Membrana Bacteriana Externa/fisiologia , Peste/microbiologia , Yersinia pestis/fisiologia , Animais , Proteínas do Sistema Complemento/imunologia , Feminino , Imunidade Inata , Macrófagos/microbiologia , Camundongos , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Yersinia pestis/imunologia , Yersinia pestis/patogenicidade
15.
Infect Immun ; 82(6): 2287-99, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24643539

RESUMO

Colonization of the human nasopharynx by Moraxella catarrhalis is presumed to involve attachment of this bacterium to the mucosa. DNA microarray analysis was used to determine whether attachment of M. catarrhalis to human bronchial epithelial (HBE) cells in vitro affected gene expression in this bacterium. Attachment affected expression of at least 454 different genes, with 163 being upregulated and 291 being downregulated. Among the upregulated genes was one (ORF113) previously annotated as encoding a protein with some similarity to outer membrane protein A (OmpA). The protein encoded by ORF113 was predicted to have a signal peptidase II cleavage site, and globomycin inhibition experiments confirmed that this protein was indeed a lipoprotein. The ORF113 protein also contained a predicted peptidoglycan-binding domain in its C-terminal half. The use of mutant and recombinant M. catarrhalis strains confirmed that the ORF113 protein was present in outer membrane preparations, and this protein was also shown to be at least partially exposed on the bacterial cell surface. A mutant unable to produce the ORF113 protein showed little or no change in its growth rate in vitro, in its ability to attach to HBE cells in vitro, or in its autoagglutination characteristics, but it did exhibit a reduced ability to survive in the chinchilla nasopharynx. This is the first report of a lipoprotein essential to the ability of M. catarrhalis to persist in an animal model.


Assuntos
Proteínas da Membrana Bacteriana Externa/fisiologia , Moraxella catarrhalis/patogenicidade , Infecções por Moraxellaceae/microbiologia , Doenças Nasofaríngeas/microbiologia , Animais , Aderência Bacteriana/fisiologia , Linhagem Celular , Chinchila , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Humanos , Proteínas de Membrana/metabolismo , Testes de Sensibilidade Microbiana , Moraxella catarrhalis/efeitos dos fármacos , Moraxella catarrhalis/genética , Análise de Sequência com Séries de Oligonucleotídeos , Peptídeos/farmacologia , Inibidores de Proteases/farmacologia
16.
Artigo em Inglês | MEDLINE | ID: mdl-24492845

RESUMO

Bacterial contact-dependent growth inhibition (CDI) is mediated by the CdiA/CdiB family of two-partner secretion proteins. CDI(+) cells bind to susceptible target bacteria and deliver a toxic effector domain derived from the carboxyl terminus of CdiA (CdiA-CT). More than 60 distinct CdiA-CT sequence types have been identified, and all CDI toxins characterized thus far display RNase, DNase, or pore-forming activities. CDI systems also encode CdiI immunity proteins, which specifically bind and inactivate cognate CdiA-CT toxins to prevent autoinhibition. CDI activity appears to be limited to target cells of the same species, suggesting that these systems play a role in competition between closely related bacteria. Recent work on the CDI system from uropathogenic Escherichia coli (UPEC 536) has revealed that its CdiA-CT toxin binds tightly to a cysteine biosynthetic enzyme (CysK) in the cytoplasm of target cells. The unanticipated complexity in the UPEC CDI pathway raises the possibility that these systems perform other functions in addition to growth inhibition. Finally, we propose that the phenomenon of CDI is more widespread than previously appreciated. Rhs (rearrangement hotspot) systems encode toxin-immunity pairs, some of which share significant sequence identity with CdiA-CT/CdiI proteins. A number of recent observations suggest that Rhs proteins mediate a distinct form of CDI.


Assuntos
Aderência Bacteriana/fisiologia , Inibição de Contato/fisiologia , Proteínas de Escherichia coli/fisiologia , Proteínas de Membrana/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Toxinas Bacterianas/metabolismo , Burkholderia/fisiologia , Escherichia coli/fisiologia , Proteínas de Escherichia coli/metabolismo , Proteínas de Membrana/metabolismo
17.
Infect Immun ; 82(4): 1372-81, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24421041

RESUMO

Persistent Helicobacter pylori infection induces chronic inflammation in the human gastric mucosa, which is associated with development of peptic ulceration, gastric atrophy, and gastric adenocarcinoma. It has been postulated that secretion of immunomodulatory molecules by H. pylori facilitates bacterial persistence, and membrane vesicles (MV), which have the potential to cross the gastric epithelial barrier, may mediate delivery of these molecules to host immune cells. However, bacterial MV effects on human immune cells remain largely uncharacterized to date. In the present study, we investigated the immunomodulatory effects of H. pylori MV with and without the vacuolating cytotoxin, VacA, which inhibits human T cell activity. We show a high degree of variability in the toxin content of vesicles between two H. pylori strains (SS1 and 60190). Vesicles from the more toxigenic 60190 strain contain more VacA (s1i1 type) than vesicles from the SS1 strain (s2i2 VacA), but engineering the SS1 strain to produce s1i1 VacA did not increase the toxin content of its vesicles. Vesicles from all strains tested, including a 60190 isogenic mutant null for VacA, strongly induced interleukin-10 (IL-10) and IL-6 production by human peripheral blood mononuclear cells independently of the infection status of the donor. Finally, we show that H. pylori MV induce T cell apoptosis and that this is enhanced by, but not completely dependent on, the carriage of VacA. Together, these findings suggest a role for H. pylori MV in the stimulation of innate pro- and anti-inflammatory responses and in the suppression of T cell immunity.


Assuntos
Apoptose/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Apoptose/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Bactérias/imunologia , Células Cultivadas , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Células Jurkat/fisiologia
18.
Trends Biochem Sci ; 39(1): 8-16, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24316304

RESUMO

Drug export from cells is a major factor in the acquisition of cellular resistance to antimicrobial and cancer chemotherapy, and poses a significant threat to future clinical management of disease. Many of the proteins that catalyse drug efflux do so with remarkably low substrate specificity, a phenomenon known as multidrug transport. For these reasons we need a greater understanding of drug recognition and transport in multidrug pumps to inform research that attempts to circumvent their action. Structural and computational studies have been heralded as being great strides towards a full elucidation of multidrug recognition and transport. In this review we summarise these advances and ask how close we are to a molecular understanding of this remarkable phenomenon.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Transportadores de Cassetes de Ligação de ATP/química , Animais , Proteínas da Membrana Bacteriana Externa/antagonistas & inibidores , Proteínas da Membrana Bacteriana Externa/química , Transporte Biológico , Farmacorresistência Bacteriana , Resistencia a Medicamentos Antineoplásicos , Humanos , Modelos Moleculares , Conformação Proteica
19.
Rev. bras. parasitol. vet ; 22(2): 253-259, Apr.-June 2013. graf, ilus
Artigo em Inglês | LILACS, VETINDEX | ID: lil-679424

RESUMO

Despite our current knowledge of the immunology, pathology, and genetics of Anaplasma marginale, prevention in cattle is currently based on old standbys, including live attenuated vaccines, antibiotic treatment, and maintaining enzootic stability in cattle herds. In the present study, we evaluated the use of an immunostimulant complex (ISCOMATRIX) adjuvant, associated with a pool of recombinant major surface proteins (rMSP1a, rMSP1b, rMSP4 and rMSP5) to improve the humoral immune response triggered in calves mainly by IgG2. Ten calves were divided in three groups: 4 calves were inoculated with the ISCOMATRIX/rMSPs (G1); 2 calves were inoculated with ISCOMATRIX adjuvant (G2); and 4 calves received saline (G3). Three inoculations were administered at 21-day intervals. In G1, the calves showed significant increases in total IgG, IgG1 and IgG2 levels 21 days after the second inoculation, compared to the control group (p < 0.05), and G1 calves remained above the cut-off value 28 days after the third inoculation (p < 0.05). The post-immunized sera from calves in G1 reacted specifically for each of the rMSPs used. In conclusion, the ISCOMATRIX/rMSPs induced antigen-specific seroconversion in calves. Therefore, additional testing to explore the protection induced by rMSPs, both alone and in conjunction with proteins previously identified as subdominant epitopes, is warranted.


Apesar dos avanços da imunologia, patologia e genética de Anaplasma marginale, a prevenção em bovinos ainda é baseada nas vacinas vivas atenuadas, na terapia com antibiótico e estabilidade enzoótica dos rebanhos bovinos. No presente estudo, avaliou-se o uso de um complexo imunoestimulante (ISCOMATRIX), associado às proteínas recombinantes de superfície (rMSP1a, rMSP1b, rMSP4 e rMSP5) para melhorar a resposta imune humoral desencadeada em bezerros, principalmente por IgG2. Dez animais foram divididos em três grupos: 4 bezerros foram inoculados com o ISCOMATRIX/rMSPs (G1), 2 bezerros foram inoculados com ISCOMATRIX adjuvante (G2) e 4 bezerros receberam salina (G3). Três doses vacinais foram administradas em intervalos de 21 dias. No G1, os bezerros apresentaram aumentos significativos nos níveis de IgG total, IgG1 e IgG2 21 dias após a segunda inoculação, em comparação com o grupo de controle (p <0,05). Nos bezerros do G1 esses níveis de anticorpos permaneceram acima do ponto de corte 28 dias após a terceira inoculação (p < 0,05). Os soros pós-imunização de bezerros do G1 reagiram especificamente com cada uma das rMSPs utilizadas. Em conclusão, o ISCOMATRIX/rMSPs induziu soroconversão antígeno-específica em bezerros. Portanto, se justifica a realização de ensaios adicionais para explorar a proteção induzida pela rMSPs, tanto sozinhas como em conjunto com novas proteínas identificadas com epítopos subdominantes.


Assuntos
Animais , Adjuvantes Imunológicos , Anaplasma marginale , Proteínas da Membrana Bacteriana Externa/fisiologia , Bovinos/imunologia , Colesterol , Fosfolipídeos , Saponinas , Saponinas/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Colesterol/administração & dosagem , Combinação de Medicamentos , Fosfolipídeos/administração & dosagem
20.
J Neuroinflammation ; 10: 47, 2013 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-23587438

RESUMO

BACKGROUND: Central nervous system (CNS) invasion by bacteria of the genus Brucella results in an inflammatory disorder called neurobrucellosis. We have recently demonstrated that B. abortus infects microglia and astrocytes, eliciting the production of a variety of pro-inflammatory cytokines which contribute to CNS damage. Matrix metalloproteinases (MMP) have been implicated in inflammatory tissue destruction in a range of pathological situations in the CNS. Increased MMP secretion is induced by pro-inflammatory cytokines in a variety of CNS diseases characterized by tissue-destructive pathology. METHODS: In this study, the molecular mechanisms that regulate MMP secretion from Brucella-infected astrocytes in vitro were investigated. MMP-9 was evaluated in culture supernatants by ELISA, zymography and gelatinolytic activity. Involvement of mitogen-activated protein kinases (MAPK) signaling pathways was evaluated by Western blot and using specific inhibitors. The role of TNF-α was evaluated by ELISA and by assays with neutralizing antibodies. RESULTS: B. abortus infection induced the secretion of MMP-9 from murine astrocytes in a dose-dependent fashion. The phenomenon was independent of bacterial viability and was recapitulated by L-Omp19, a B. abortus lipoprotein model, but not its LPS. B. abortus and L-Omp19 readily activated p38 and Erk1/2 MAPK, thus enlisting these pathways among the kinase pathways that the bacteria may address as they invade astrocytes. Inhibition of p38 or Erk1/2 significantly diminished MMP-9 secretion, and totally abrogated production of this MMP when both MAPK pathways were inhibited simultaneously. A concomitant abrogation of B. abortus- and L-Omp19-induced TNF-α production was observed when p38 and Erk1/2 pathways were inhibited, indicating that TNF-α could be implicated in MMP-9 secretion. MMP-9 secretion induced by B. abortus or L-Omp19 was completely abrogated when experiments were conducted in the presence of a TNF-α neutralizing antibody. MMP-9 activity was detected in cerebrospinal fluid (CSF) samples from patients suffering from neurobrucellosis. CONCLUSIONS: Our results indicate that the inflammatory response elicited by B. abortus in astrocytes would lead to the production of MMP-9 and that MAPK may play a role in this phenomenon. MAPK inhibition may thus be considered as a strategy to control inflammation and CNS damage in neurobrucellosis.


Assuntos
Brucella abortus , Brucelose/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/fisiologia , Animais , Anticorpos Bloqueadores/farmacologia , Antígenos de Bactérias/fisiologia , Astrócitos/metabolismo , Astrócitos/microbiologia , Astrócitos/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Citocinas/metabolismo , Gelatinases/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Lipopolissacarídeos/farmacologia , Lipoproteínas/farmacologia , Lipoproteínas/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Cultura Primária de Células , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA