Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.110
Filtrar
1.
Mol Microbiol ; 116(5): 1281-1297, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34581467

RESUMO

Many bacteria can migrate from a free-living, planktonic state to an attached, biofilm existence. One factor regulating this transition in the facultative plant pathogen Agrobacterium tumefaciens is the ExoR-ChvG-ChvI system. Periplasmic ExoR regulates the activity of the ChvG-ChvI two-component system in response to environmental stress, most notably low pH. ChvI impacts hundreds of genes, including those required for type VI secretion, virulence, biofilm formation, and flagellar motility. Previous studies revealed that activated ChvG-ChvI represses expression of most of class II and class III flagellar biogenesis genes, but not the master motility regulator genes visN, visR, and rem. In this study, we characterized the integration of the ExoR-ChvG-ChvI and VisNR-Rem pathways. We isolated motile suppressors of the non-motile ΔexoR mutant and thereby identified the previously unannotated mirA gene encoding a 76 amino acid protein. We report that the MirA protein interacts directly with the Rem DNA-binding domain, sequestering Rem and preventing motility gene activation. The ChvG-ChvI pathway activates mirA expression and elevated mirA is sufficient to block motility. This study reveals how the ExoR-ChvG-ChvI pathway prevents flagellar motility in A. tumefaciens. MirA is also conserved among other members of the Rhizobiales suggesting similar mechanisms of motility regulation.


Assuntos
Agrobacterium tumefaciens/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Proteínas de Bactérias/fisiologia , Regulação Bacteriana da Expressão Gênica , Proteínas Quinases/fisiologia , Estresse Fisiológico , Fatores de Transcrição/fisiologia , Sequência de Aminoácidos , Genes Bacterianos , Ligação Proteica , Virulência
2.
Food Funct ; 12(8): 3597-3610, 2021 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-33900345

RESUMO

Akkermansia muciniphila is a probiotic inhabiting host intestinal mucus layers and displays evident easing or therapeutic effects on host enteritis and metabolic disorders such as obesity and diabetes. The outer membrane protein Amuc_1100 of A. muciniphila is likely to play a crucial role during the interaction with the host. 5-HT is a neurotransmitter and a key signal molecule regulating the gastrointestinal tract functions and other organs, which is involved in diverse physiological and pathological processes. This study demonstrated that Amuc_1100 could promote the expression of the 5-HT synthesis rate-limiting enzyme Tph1 in RIN-14B cells and reduce the expression of the serotonin reuptake transporter (SERT) in Caco-2 cells through direct interaction with TLR2, thereby improving 5-HT biosynthesis and extracellular availability. Using antibiotic-treated mice as animal models, we found that after gavage with A. muciniphila or Amuc_1100, Tph1 expression increased and SERT expression decreased in colon tissues. The 5-HT concentrations in colon tissues and blood were markedly elevated simultaneously. We also found that A. muciniphila or Amuc_1100 improved the gastrointestinal motility function and restored gut microbiota abundance and species diversity in antibiotic-treated mice. These results suggest that A. muciniphila can regulate the host intestinal 5-HT system via its outer membrane protein Amuc_1100 and TLR2. This mechanism represented an important approach through which A. muciniphila interacts with the host and further influences 5-HT-related physiological functions. These results advance the understanding of interplay mechanisms between the gut microbiota and the host, which could be the basis for new intervention strategies for related diseases.


Assuntos
Akkermansia/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Microbioma Gastrointestinal/fisiologia , Intestinos/efeitos dos fármacos , Serotonina/biossíntese , Receptor 2 Toll-Like/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/farmacologia , Células CACO-2 , Linhagem Celular , Células Enterocromafins/efeitos dos fármacos , Células Enterocromafins/metabolismo , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos
3.
Mol Microbiol ; 115(3): 412-424, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33283907

RESUMO

The type II secretion system (T2SS) is a multi-protein complex used by many bacteria to move substrates across their cell membrane. Substrates released into the environment serve as local and long-range effectors that promote nutrient acquisition, biofilm formation, and pathogenicity. In both animals and plants, the T2SS is increasingly recognized as a key driver of virulence. The T2SS spans the bacterial cell envelope and extrudes substrates through an outer membrane secretin channel using a pseudopilus. An inner membrane assembly platform and a cytoplasmic motor controls pseudopilus assembly. This microreview focuses on the structure and mechanism of the T2SS. Advances in cryo-electron microscopy are enabling increasingly elaborate sub-complexes to be resolved. However, key questions remain regarding the mechanism of pseudopilus extension and retraction, and how this is coupled with the choreography of the substrate moving through the secretion system. The T2SS is part of an ancient type IV filament superfamily that may have been present within the last universal common ancestor (LUCA). Overall, mechanistic principles that underlie T2SS function have implication for other closely related systems such as the type IV and tight adherence pilus systems.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/fisiologia , Fímbrias Bacterianas/química , Fímbrias Bacterianas/fisiologia , Sistemas de Secreção Tipo II/química , Sistemas de Secreção Tipo II/fisiologia , Sequência de Aminoácidos , Animais , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/fisiologia , Fenômenos Fisiológicos Bacterianos , Microscopia Crioeletrônica , Humanos , Modelos Moleculares , Conformação Proteica , Secretina/metabolismo , Fatores de Virulência/química , Fatores de Virulência/fisiologia
4.
PLoS One ; 15(11): e0242593, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33227031

RESUMO

Bacterial efflux pumps are an important pathogenicity trait because they extrude a variety of xenobiotics. Our laboratory previously identified in silico Burkholderia collagen-like protein 8 (Bucl8) in the hazardous pathogens Burkholderia pseudomallei and Burkholderia mallei. We hypothesize that Bucl8, which contains two predicted tandem outer membrane efflux pump domains, is a component of a putative efflux pump. Unique to Bucl8, as compared to other outer membrane proteins, is the presence of an extended extracellular region containing a collagen-like (CL) domain and a non-collagenous C-terminus (Ct). Molecular modeling and circular dichroism spectroscopy with a recombinant protein, corresponding to this extracellular CL-Ct portion of Bucl8, demonstrated that it adopts a collagen triple helix, whereas functional assays screening for Bucl8 ligands identified binding to fibrinogen. Bioinformatic analysis of the bucl8 gene locus revealed it resembles a classical efflux-pump operon. The bucl8 gene is co-localized with downstream fusCDE genes encoding fusaric acid (FA) resistance, and with an upstream gene, designated as fusR, encoding a LysR-type transcriptional regulator. Using reverse transcriptase (RT)-qPCR, we defined the boundaries and transcriptional organization of the fusR-bucl8-fusCDE operon. We found exogenous FA induced bucl8 transcription over 80-fold in B. pseudomallei, while deletion of the entire bucl8 locus decreased the minimum inhibitory concentration of FA 4-fold in its isogenic mutant. We furthermore showed that the putative Bucl8-associated pump expressed in the heterologous Escherichia coli host confers FA resistance. On the contrary, the Bucl8-associated pump did not confer resistance to a panel of clinically-relevant antimicrobials in Burkholderia and E. coli. We finally demonstrated that deletion of the bucl8-locus drastically affects the growth of the mutant in L-broth. We determined that Bucl8 is a component of a novel tetrapartite efflux pump, which confers FA resistance, fibrinogen binding, and optimal growth.


Assuntos
Burkholderia mallei/metabolismo , Burkholderia pseudomallei/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas da Membrana Bacteriana Externa/fisiologia , Burkholderia/genética , Burkholderia/metabolismo , Burkholderia mallei/genética , Burkholderia pseudomallei/genética , Colágeno/metabolismo , Farmacorresistência Bacteriana Múltipla/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Genes Bacterianos/efeitos dos fármacos , Óperon/efeitos dos fármacos , Fatores de Transcrição/metabolismo
5.
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
6.
Trends Microbiol ; 28(9): 706-708, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32466989

RESUMO

The bacterial cell envelope plays essential roles in controlling cell shape, division, pathogenicity, and resistance against external stresses. In Escherichia coli, peptidoglycan (PG) has long been thought to be the primary component that conveys mechanical strength to the envelope. But a recent publication demonstrates the key contribution of the lipoprotein Lpp in defining the stiffness of the cell envelope and its sensitivity to drugs.


Assuntos
Membrana Celular/fisiologia , Farmacorresistência Bacteriana , Escherichia coli/citologia , Escherichia coli/fisiologia , Lipoproteínas/fisiologia , Peptidoglicano , Proteínas da Membrana Bacteriana Externa/fisiologia , Fenômenos Biomecânicos , Microscopia de Força Atômica
7.
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
8.
Future Microbiol ; 15: 143-157, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-32073314

RESUMO

Rresistance-nodulation-division (RND) efflux pumps in Gram-negative bacteria remove multiple, structurally distinct classes of antimicrobials from inside bacterial cells therefore directly contributing to multidrug resistance. There is also emerging evidence that many other mechanisms of antibiotic resistance rely on the intrinsic resistance conferred by RND efflux. In addition to their role in antibiotic resistance, new information has become available about the natural role of RND pumps including their established role in virulence of many Gram-negative organisms. This review also discusses the recent advances in understanding the regulation and structure of RND efflux pumps.


Assuntos
Farmacorresistência Bacteriana Múltipla , Regulação Bacteriana da Expressão Gênica , Bactérias Gram-Negativas/fisiologia , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/fisiologia , Antibacterianos/farmacologia , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/fisiologia , Transporte Biológico , Virulência
9.
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
10.
Biochim Biophys Acta Biomembr ; 1862(1): 183031, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31374213

RESUMO

Electrostatic side chain contacts can contribute substantial interaction energy terms to the stability of proteins. The impact of electrostatic interactions on the structure and architecture of outer membrane proteins is however not well studied compared to soluble proteins. Here, we report the results of a systematic study of all charged side chains of the E. coli outer membrane protein X (OmpX). The data identify three distinct salt-bridge clusters in the core of OmpX that contribute significantly to protein stability in dodecylphosphocholine detergent micelles. The three clusters form an "electrostatic core" of the membrane protein OmpX, corresponding in its architectural role to the hydrophobic core of soluble proteins. This article is part of a Special Issue entitled: Molecular biophysics of membranes and membrane proteins.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Proteínas de Escherichia coli/química , Escherichia coli/química , Hidrolases/química , Eletricidade Estática , Proteínas da Membrana Bacteriana Externa/fisiologia , Proteínas de Escherichia coli/fisiologia , Hidrolases/fisiologia , Interações Hidrofóbicas e Hidrofílicas , Micelas , Modelos Moleculares , Conformação Proteica , Estabilidade Proteica , Estrutura Quaternária de Proteína
11.
J Fish Dis ; 43(2): 275-284, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31779054

RESUMO

Vibrio alginolyticus is one of the most serious causative agents of diseases in cultured marine fish and shellfish. However, the characteristics of virulence factors in pathogenic V. alginolyticus are poorly known. To gain insight into fish diseases caused by V. alginolyticus, we carried out two-dimensional gel electrophoresis (2-DE) combined with MALDI-TOF mass spectrometry to identify uniquely expressed proteins in the disease-causing V. alginolyticus. V. alginolyticus strains were isolated from marine environments and diseased fish obtained from southern Thailand. We identified seven unique proteins in the disease-causing V. alginolyticus strain. Among those, the outer membrane protein A (OmpA) had the strongest expression. Therefore, the function of this protein was further analysed. To investigate the role of OmpA protein, an in-frame deletion mutant of ompA was constructed using the homologous recombination method. Although the ompA mutant V. alginolyticus strain (ΔompA) grew normally, the mutant exhibited a significant defect in the swarming ability and the biofilm formation. Furthermore, Galleria mellonella larvae injected with the mutant bacteria had a significantly greater survival percentage than those injected with the wild-type strain, demonstrating that OmpA protein is required for the pathogenicity of V. alginolyticus. Together, this study suggests a potential target for vaccine development against pathogenic V. alginolyticus strain.


Assuntos
Proteínas da Membrana Bacteriana Externa/fisiologia , Doenças dos Peixes/microbiologia , Vibrioses/microbiologia , Vibrio alginolyticus/patogenicidade , Fatores de Virulência/fisiologia , Animais , Eletroforese em Gel Bidimensional , Tailândia , Vibrio alginolyticus/genética
13.
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
14.
Mol Microbiol ; 112(6): 1757-1768, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31550057

RESUMO

Most bacteria are surrounded by a complex cell envelope. As with many biological processes, studies of envelope assembly have benefited from cell-based assays for detecting protein-protein interactions. These assays use simple readouts and lack a protein purification requirement, making them ideal for early stage investigations. The most widely used two-hybrid interaction assay for proteins involved in envelope biogenesis is based on the reconstitution of adenylate cyclase activity from a split enzyme. Because adenylate cyclase is only functional in the cytoplasm, both protein fusions used in the assay must have a terminus located in this compartment. However, many envelope assembly factors are wholly extracytoplasmic. Detecting interactions involving such proteins using two-hybrid systems has therefore been problematic. To address this issue, we developed a cytological assay in Escherichia coli based on PopZ from Caulobacter crescentus. Here, we demonstrate the utility of this PopZ-Linked Apical Recruitment (POLAR) method for detecting interactions between proteins located in different cellular compartments. Additionally, we report that recruitment of an active peptidoglycan synthase to the cell pole is detrimental for E. coli and that interactions between proteins in the inner and outer membranes of the Gram-negative envelope may provide a mechanism for recruiting protein complexes to subpolar sites.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Parede Celular/fisiologia , Mapeamento de Interação de Proteínas/métodos , Membrana Externa Bacteriana/metabolismo , Proteínas da Membrana Bacteriana Externa/fisiologia , Proteínas de Bactérias/metabolismo , Caulobacter crescentus/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Divisão Celular , Membrana Celular/metabolismo , Parede Celular/metabolismo , Centrômero/metabolismo , Cromossomos Bacterianos/metabolismo , Citoplasma/metabolismo , Escherichia coli/metabolismo , Peptidoglicano/metabolismo , Mapas de Interação de Proteínas/fisiologia
15.
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
16.
Biochim Biophys Acta Biomembr ; 1861(10): 183021, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31306626

RESUMO

OmpG is a general diffusion pore in the E. coli outer membrane with a molecular architecture comprising a 14-stranded ß-barrel scaffold and unique structural features. In contrast to other non-specific porins, OmpG lacks a central constriction zone and has an exceptionally wide pore diameter of about 13 Å. The equatorial plane of OmpG harbors an annulus of four alternating basic and acidic patches whose function is only poorly characterized. We have investigated the role of charge distribution for ion selectivity and sugar transport with the help of OmpG variants mutated in the annulus. Substituting the glutamate residues of the annulus for histidines or alanines led to a strong reduction in cation selectivity. Replacement of the glutamates in the annulus by histidine residues also disfavored the passage of pentoses and hexoses relative to disaccharides. Our results demonstrate that despite the wide pore diameter, an annulus only consisting of two opposing basic patches confers reduced cation and monosaccharide transport compared to OmpG wild type. Furthermore, randomization of charged residues in the annulus had the potential to abolish pH-dependency of sugar transport. Our results indicate that E15, E31, R92, R111 and R211 in the annulus form electrostatic interactions with R228, E229 and D232 in loop L6 that influence pH-dependency of sugar transport.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Escherichia coli/metabolismo , Porinas/química , Arginina/metabolismo , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/fisiologia , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiologia , Ácido Glutâmico/metabolismo , Concentração de Íons de Hidrogênio , Porinas/genética , Porinas/metabolismo , Porinas/fisiologia , Especificidade por Substrato/fisiologia , Açúcares/metabolismo
17.
Lab Invest ; 99(11): 1650-1660, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31253864

RESUMO

Understanding bacterial adhesion is challenging and critical to our understanding of the initial stages of the pathogenesis of endovascular bacterial infections. The vascular endothelial cell (EC) is the main target of Rickettsia, an obligately intracellular bacterium that causes serious systemic disease in humans and animals. But the mechanism(s) underlying bacterial adherence to ECs under shear stress from flowing blood prior to activation are unknown for any bacteria. Although host surface annexin a2 (ANXA2) has been identified to participate in efficient bacterial invasion of epithelial cells, direct evidence is lacking in the field of bacterial infections of ECs. In the present study, we employ a novel, anatomically based, in vivo quantitative bacterial-adhesion-to-vascular-EC system, combined with atomic force microscopy (AFM), to examine the role of endothelial luminal surface ANXA2 during rickettsial adherence to ECs. We also examined whether ANXA2 antibody affected binding of Staphylococcus aureus to ECs. We found that deletion of ANXA2 impeded rickettsial attachment to the ECs in vitro and blocked rickettsial adherence to the blood vessel luminal surface in vivo. The AFM studies established that EC surface ANXA2 acts as an adherence receptor for rickettsiae, and that rickettsial adhesin OmpB is the associated bacterial ligand. Furthermore, pretreatment of ECs with anti-ANXA2 antibody reduced EC surface-associated S. aureus. We conclude that the endothelial surface ANXA2 plays an important role in initiating pathogen-host interactions, ultimately leading to bacterial anchoring on the vascular luminal surface.


Assuntos
Anexina A2/fisiologia , Aderência Bacteriana/fisiologia , Células Endoteliais/microbiologia , Células Endoteliais/fisiologia , Animais , Anexina A2/deficiência , Anexina A2/genética , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/fisiologia , Fenômenos Biomecânicos , Modelos Animais de Doenças , Interações entre Hospedeiro e Microrganismos/fisiologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Força Atômica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Rickettsia/patogenicidade , Rickettsia/fisiologia , Infecções por Rickettsia/microbiologia , Staphylococcus aureus/patogenicidade , Staphylococcus aureus/fisiologia
18.
PLoS Pathog ; 15(3): e1007385, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30845186

RESUMO

The six-component maintenance of lipid asymmetry (Mla) system is responsible for retrograde transport of phospholipids, ensuring the barrier function of the Gram-negative cell envelope. Located within the outer membrane, MlaA (VacJ) acts as a channel to shuttle phospholipids from the outer leaflet. We identified Neisseria gonorrhoeae MlaA (ngo2121) during high-throughput proteomic mining for potential therapeutic targets against this medically important human pathogen. Our follow-up phenotypic microarrays revealed that lack of MlaA results in a complex sensitivity phenome. Herein we focused on MlaA function in cell envelope biogenesis and pathogenesis. We demonstrate the existence of two MlaA classes among 21 bacterial species, characterized by the presence or lack of a lipoprotein signal peptide. Purified truncated N. gonorrhoeae MlaA elicited antibodies that cross-reacted with a panel of different Neisseria. Little is known about MlaA expression; we provide the first evidence that MlaA levels increase in stationary phase and under anaerobiosis but decrease during iron starvation. Lack of MlaA resulted in higher cell counts during conditions mimicking different host niches; however, it also significantly decreased colony size. Antimicrobial peptides such as polymyxin B exacerbated the size difference while human defensin was detrimental to mutant viability. Consistent with the proposed role of MlaA in vesicle biogenesis, the ΔmlaA mutant released 1.7-fold more membrane vesicles. Comparative proteomics of cell envelopes and native membrane vesicles derived from ΔmlaA and wild type bacteria revealed enrichment of TadA-which recodes proteins through mRNA editing-as well as increased levels of adhesins and virulence factors. MlaA-deficient gonococci significantly outcompeted (up to 16-fold) wild-type bacteria in the murine lower genital tract, suggesting the growth advantage or increased expression of virulence factors afforded by inactivation of mlaA is advantageous in vivo. Based on these results, we propose N. gonorrhoeae restricts MlaA levels to modulate cell envelope homeostasis and fine-tune virulence.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Neisseria gonorrhoeae/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Bactérias , Proteínas da Membrana Bacteriana Externa/fisiologia , Proteínas de Bactérias , Transporte Biológico , Membrana Celular , Parede Celular , Proteínas de Escherichia coli , Gonorreia , Bactérias Gram-Negativas/metabolismo , Humanos , Neisseria gonorrhoeae/fisiologia , Fosfolipídeos/metabolismo , Fosfolipídeos/fisiologia , Filogenia , Proteômica , Virulência , Fatores de Virulência
19.
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
20.
Med Sci Monit Basic Res ; 25: 45-52, 2019 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-30739907

RESUMO

BACKGROUND Recent studies have shown that Escherichia coli induced digestive tract diseases may be related to outer membrane vesicles (OMVs) induced intestinal double-strand breaks (DSBs) in intestinal epithelial cells. This study aimed to compare the impact of OMVs forces on DSBs in intestinal epithelial Caco-2 cells, and provide a new treatment for digestive diseases caused by E. coli. MATERIAL AND METHODS E.coli OMVs were prepared and co-cultured with Caco-2 cells. The uptake of OMVs by Caco-2 cells was observed by confocal microscopy. The γ-H2AX protein was detected by western-blots. The DSBs caused by OMVs was detected by single cell gel electrophoresis. RESULTS The particle size analyzer showed that the average diameters of OMVs centrifuged at 20 000×g and 50 000×g were 217.5±7.29 nm and 186.3±6.59 nm (P<0.05), respectively. Transmission electron microscopy of the OMVs revealed a lipid bilayer structure with a variety of different sizes. Confocal fluorescence microscopy revealed that OMVs almost completely entered Caco-2 cells after 24 hours. The ratio of γ-H2AX protein band gray value normalized data in the OMVs centrifuged at 20 000×g and 50 000×g, and the control group (without OMVs) were 2.23±0.18, 1.58±0.20, 1±0.30 (P<0.05), respectively, while DNA levels of the comet tail (TailDNA%, TDNA%) were 72.21±14.61%, 23.11±4.98%, and 1.02±1.41% (P<0.05), respectively. The corresponding DNA damage was categorized as high (grade 3), moderate (grade 2), and no damage (grade 0). CONCLUSIONS Different sizes of OMVs induced different degrees of DNA damage in intestinal epithelial Caco-2 cells.


Assuntos
Proteínas da Membrana Bacteriana Externa/fisiologia , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Células CACO-2/fisiologia , Escherichia coli/patogenicidade , Escherichia coli/fisiologia , Proteínas de Escherichia coli/fisiologia , Humanos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...