Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
1.
Carbohydr Res ; 534: 108977, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37949033

RESUMO

Herein, we report the total synthesis of the trisaccharide repeating unit of the O-antigen of Actinobacillus actinomycetemcomitans serotype f. The trisaccharide comprising of α-(1-2) and α-(1-3)-linked L-rhamnopyranosides backbone with the latter rhamnose containing a branching N-acetyl-d-galactosaminopyranoside at the C2-O via a ß-glycosidic bond was synthesized by two methods. Initially, the protected trisaccharide has been synthesized by step-wise assembly of the monosaccharide building blocks and subsequently the former was synthesized by the one-pot assembly of the latter components. The synthesized trisaccharide contains an aminoethyl linker appended as an O-glycoside at the reducing end, thereby providing scope for further conjugation for different applications.


Assuntos
Aggregatibacter actinomycetemcomitans , Antígenos O , Antígenos O/química , Aggregatibacter actinomycetemcomitans/química , Sorogrupo , Monossacarídeos , Glicosídeos , Trissacarídeos
2.
Microb Pathog ; 173(Pt A): 105843, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36374775

RESUMO

Each HACEK group pathogen, which can cause infective endocarditis, expresses type IVa pili. The type IVa major pilin PilA plays a role in bacterial colonization, virulence, twitching motility, and the uptake of extracellular DNA. The type IV prepilin homolog PilA of the periodontal pathogen A. actinomycetemcomitans (AaPilA) is linked to DNA uptake and natural competence. Our aim was to investigate the virulence properties and immunogenic potential of AaPilA. Since Neisseria meningitidis PilE, which shares sequence similarity with AaPilA, participates in sequestering host cytokines, we examined the ability of AaPilA to interact with various cytokines. Moreover, we investigated the structural characteristics of AaPilA with molecular modeling. AaPilA was conserved among A. actinomycetemcomitans strains. One of the 18 different natural variants, PilAD7S, is present in naturally competent strains. This variant interacted with DNA and bound interleukin (IL)-8 and tumor necrosis factor (TNF)-α. Specific anti-AaPilA antibodies were present in A. actinomycetemcomitans-positive periodontitis patient sera, and the production of reactive oxygen species from human neutrophils was less effectively induced by the ΔpilA mutant than by the wild-type strains. However, AaPilA did not stimulate human macrophages to produce proinflammatory cytokines, nor was it cytotoxic. The results strengthen our earlier hypothesis that the DNA uptake machinery of A. actinomycetemcomitans is involved in the sequestration of inflammatory cytokines. Furthermore, AaPilA stimulates host immune cells, such as B cells and neutrophils, making it a potential virulence factor.


Assuntos
Aggregatibacter actinomycetemcomitans , Citocinas , Humanos , Aggregatibacter actinomycetemcomitans/química , Citocinas/metabolismo , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/metabolismo , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , DNA/metabolismo , Leucócitos/metabolismo
3.
Comput Methods Programs Biomed ; 222: 106952, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35724475

RESUMO

The leukotoxin (LtxA) of Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans) is a protein exotoxin belonging to the repeat-in-toxin family (RTX). Numerous studies have demonstrated that LtxA may play a critical role in the pathogenicity of A. actinomycetemcomitans since hyper-leukotoxic strains have been associated with severe disease. Accordingly, considerable effort has been made to elucidate the mechanisms by which LtxA interacts with host cells and induce their death. However, these attempts have been hampered by the unavailability of a tertiary structure of the toxin, which limits the understanding of its molecular properties and mechanisms. In this paper, we used homology and template free modeling algorithms to build the complete tertiary model of LtxA at atomic level in its calcium-bound Holo-state. The resulting model was refined by energy minimization, validated by Molprobity and ProSA tools, and subsequently subjected to a cumulative 600ns of all-atom classical molecular dynamics simulation to evaluate its structural aspects. The druggability of the proposed model was assessed using Fpocket and FTMap tools, resulting in the identification of four putative cavities and fifteen binding hotspots that could be targeted by rational drug design tools to find new ligands to inhibit LtxA activity.


Assuntos
Aggregatibacter actinomycetemcomitans , Exotoxinas , Aggregatibacter actinomycetemcomitans/química , Aggregatibacter actinomycetemcomitans/metabolismo , Simulação por Computador , Exotoxinas/química , Exotoxinas/metabolismo , Exotoxinas/farmacologia
4.
Microbiology (Reading) ; 168(5)2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35551696

RESUMO

Aggregatibacter actinomycetemcomitans, a causative agent of periodontitis and non-oral diseases, synthesizes a trimeric extracellular matrix protein adhesin A (EmaA) that mediates collagen binding and biofilm formation. EmaA is found as two molecular forms, which correlate with the serotype of the bacterium. The canonical protein (b-EmaA), associated with serotypes b and c, has a monomeric molecular mass of 202 kDa. The collagen binding activity of b-EmaA is dependent on the presence of O-polysaccharide (O-PS), whereas biofilm activity is independent of O-PS synthesis. The EmaA associated with serotype a strains (a-EmaA) has a monomeric molecular mass of 173 kDa and differs in the amino acid sequence of the functional domain of the protein. In this study, a-emaA was confirmed to encode a protein that forms antenna-like appendages on the surface of the bacterium, which were found to be important for both collagen binding and biofilm formation. In an O-PS-deficient talose biosynthetic (tld) mutant strain, the electrophoretic mobility of the a-EmaA monomers was altered and the amount of membrane-associated EmaA was decreased when compared to the parent strain. The mass of biofilm formed remained unchanged. Interestingly, the collagen binding activity of the mutant strain was similar to the activity associated with the parent strain, which differs from that observed with the canonical b-EmaA isoform. These data suggest that the properties of the a-EmaA isoform are like those of b-EmaA, with the exception that collagen binding activity is independent of the presence or absence of the O-PS.


Assuntos
Aggregatibacter actinomycetemcomitans , Proteínas da Matriz Extracelular , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Aggregatibacter actinomycetemcomitans/química , Aggregatibacter actinomycetemcomitans/genética , Aggregatibacter actinomycetemcomitans/metabolismo , Colágeno/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Sorogrupo
5.
PLoS One ; 16(3): e0248308, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33667279

RESUMO

Periodontitis can result in tooth loss and the associated chronic inflammation can provoke several severe systemic health risks. Adjunctive to mechanical treatment of periodontitis and as alternatives to antibiotics, the use of probiotic bacteria was suggested. In this study, the inhibitory effect of the probiotic Streptococcus salivarius subsp. salivarius strains M18 and K12, Streptococcus oralis subsp. dentisani 7746, and Lactobacillus reuteri ATCC PTA 5289 on anaerobic periodontal bacteria and Aggregatibacter actinomycetemcomitans was tested. Rarely included in other studies, we also quantified the inverse effect of pathogens on probiotic growth. Probiotics and periodontal pathogens were co-incubated anaerobically in a mixture of autoclaved saliva and brain heart infusion broth. The resulting genome numbers of the pathogens and of the probiotics were measured by quantitative real-time PCR. Mixtures of the streptococcal probiotics were also used to determine their synergistic, additive, or antagonistic effects. The overall best inhibitor of the periodontal pathogens was L. reuteri ATCC PTA 5289, but the effect is coenzyme B12-, anaerobiosis-, as well as glycerol-dependent, and further modulated by L. reuteri strain DSM 17938. Notably, in absence of glycerol, the pathogen-inhibitory effect could even turn into a growth spurt. Among the streptococci tested, S. salivarius M18 had the most constant inhibitory potential against all pathogens, followed by K12 and S. dentisani 7746, with the latter still having significant inhibitory effects on P. intermedia and A. actinomycetemcomitans. Overall, mixtures of the streptococcal probiotics did inhibit the growth of the pathogens equally or-in the case of A. actinomycetemcomitans- better than the individual strains. P. gingivalis and F. nucleatum were best inhibited by pure cultures of S. salivarius K12 or S. salivarius M18, respectively. Testing inverse effects, the growth of S. salivarius M18 was enhanced when incubated with the periodontal pathogens minus/plus other probiotics. In contrast, S. oralis subsp. dentisani 7746 was not much influenced by the pathogens. Instead, it was significantly inhibited by the presence of other streptococcal probiotics. In conclusion, despite some natural limits such as persistence, the full potential for probiotic treatment is by far not utilized yet. Especially, further exploring concerted activity by combining synergistic strains, together with the application of oral prebiotics and essential supplements and conditions, is mandatory.


Assuntos
Anaerobiose/efeitos dos fármacos , Periodontite/tratamento farmacológico , Porphyromonas gingivalis/efeitos dos fármacos , Probióticos/farmacologia , Aggregatibacter actinomycetemcomitans/química , Aggregatibacter actinomycetemcomitans/crescimento & desenvolvimento , Antibiose/efeitos dos fármacos , Humanos , Limosilactobacillus reuteri/química , Limosilactobacillus reuteri/crescimento & desenvolvimento , Periodontite/microbiologia , Periodontite/patologia , Porphyromonas gingivalis/patogenicidade , Probióticos/química , Saliva/efeitos dos fármacos , Saliva/microbiologia , Streptococcus/química , Streptococcus/crescimento & desenvolvimento , Streptococcus mutans/química , Streptococcus mutans/crescimento & desenvolvimento , Streptococcus salivarius/química , Streptococcus salivarius/crescimento & desenvolvimento
6.
Carbohydr Polym ; 253: 117285, 2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-33278951

RESUMO

ß-glucans are potent immunomodulators, with effects on innate and adaptive immune responses via dectin-1 as the main receptor. In this study, we investigated the biological effect of ß-glucan from Schizophyllum commune, called Schizophyllan (SPG) on Interleukin-10 (IL-10) expression induced by a lipopolysaccharide (LPS) from Aggregatibacter actinomycetemcomitans in murine macrophages (J774.1). SPG and dectin-1 interaction up-regulates LPS-induced IL-10 expression. The regulative effect of SPG on IL-10 expression is dependent on prolongation of nuclear translocation activity of nuclear factor-kappa B (NF-κBα) pathway induced by LPS. We also found that LPS-induced phosphorylation of mitogen- and stress-activated protein kinase 1 (MSK1) and cAMP-responsive-element-binding protein (CREB), followed by up-regulation of IL-10, was stimulated by SPG priming via activation of the spleen tyrosine kinase (Syk). Our data indicate that SPG augments the anti-inflammatory response in murine macrophages which can be useful to create an intervention for periodontal disease treatment.


Assuntos
Adjuvantes Imunológicos/farmacologia , Aggregatibacter actinomycetemcomitans/química , Polissacarídeos Fúngicos/farmacologia , Interleucina-10/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , Schizophyllum/química , Sizofirano/farmacologia , Adjuvantes Imunológicos/metabolismo , Animais , Polissacarídeos Fúngicos/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , NF-kappa B/metabolismo , Infecções por Pasteurellaceae/tratamento farmacológico , Infecções por Pasteurellaceae/microbiologia , Doenças Periodontais/tratamento farmacológico , Doenças Periodontais/microbiologia , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sizofirano/metabolismo
7.
BMC Microbiol ; 20(1): 279, 2020 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-32917132

RESUMO

BACKGROUND: This study evaluated the effect of oral lactobacilli on the cytotoxicity and cytokine release from peripheral blood mononuclear cells (PBMCs) when exposed to Aggregatibacter actinomycetemcomitans subtypes in vitro. The supernatants and cell wall extracts (CWEs) of eight A. actinomycetemcomitans strains, representing different subtypes, and three Lactobacillus strains were used. The PBMCs from six blood donors were exposed to supernatants and CWEs of A. actinomycetemcomitans or Lactobacillus strains alone or combinations and untreated cells as control. The cytotoxicity was determined by trypan blue exclusion method and IL-1ß secretion by ELISA. TNF-α, IL-6, and IL-8 secretions were measured using Bioplex Multiplex Immunoassay. RESULTS: Supernatants or CWEs from all bacterial strains showed cytotoxicity and IL-1ß secretion and the subtypes of A. actinomycetemcomitans showed generally a significantly higher effect on PBMCs than that of the Lactobacillus strains. Two highly toxic A. actinomycetemcomitans strains (JP2 and JP2-like) induced a higher response than all other strains. When combined, Lactobacillus significantly reduced the toxicity and the IL-1ß secretion induced by A. acinomycetemcomitans. The effect varied between the subtypes and the reduction was highest for the JP2 and JP2-like strains. The Lactobacillus paracasei strain SD1 had a higher reducing effect than the other Lactobacillus strains. This strain had a consistent reducing effect on all subtypes of A. actinomycetemcomitans cytotoxicity, and release of IL-1ß, IL-6, IL-8, and TNF-α from PBMCs of the blood donors. A strong and significant variation in cytokine release between the six blood donors was noticed. CONCLUSIONS: Lactobacillus spp. and L. paracasei SD1 in particular, showed a limited but statistically significant reducing interaction with A. actinomycetemcomitans toxicity and release of cytokines in vitro.


Assuntos
Aggregatibacter actinomycetemcomitans/patogenicidade , Citocinas/metabolismo , Lactobacillus , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/microbiologia , Aggregatibacter actinomycetemcomitans/química , Aggregatibacter actinomycetemcomitans/isolamento & purificação , Parede Celular/química , Humanos , Lacticaseibacillus paracasei , Boca/microbiologia , Infecções por Pasteurellaceae/microbiologia , Probióticos/administração & dosagem
8.
J Biomol NMR ; 74(2-3): 147-159, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31932991

RESUMO

Resonance assignment of intrinsically disordered proteins is remarkably challenging due to scant chemical shift dispersion arising from conformational heterogeneity. The challenge is even greater if repeating segments are present in the amino acid sequence. To forward unambiguous resonance assignment of intrinsically disordered proteins, we present iHACANCO, HACACON and (HACA)CONCAHA, three Hα-detected 4D experiments with Cα as an additional dimension. In addition, we present (HACA)CON(CA)NH and (HACA)N(CA)CONH, new 4D Hα-start, HN-detect experiments which have two NH dimensions to enhance peak dispersion in a sequential walk through C', NH and HN, and provide more accurate NH/HN chemical shifts than those that can be obtained from a crowded 1H, 15N-HSQC spectrum. Application of these 4D experiments is demonstrated using BilRI (165 aa), an outer-membrane intrinsically disordered protein from the opportunistic oral pathogen Aggregatibacter actinomycetemcomitans. BilRI amino acid sequence encompasses three very similar repeats with a 13-residue identical stretch in two of them.


Assuntos
Aggregatibacter actinomycetemcomitans/química , Proteínas de Bactérias/química , Proteínas Intrinsicamente Desordenadas/química , Ressonância Magnética Nuclear Biomolecular
9.
Toxins (Basel) ; 11(11)2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31739483

RESUMO

Aggregatibacter actinomycetemcomitans is a Gram-negative bacterium that has been strongly associated with localized aggressive periodontitis. The capacity of A. actinomycetemcomitans to produce a leukotoxin (LtxA) that activates pyroptosis in macrophages and induces the release of endogenous danger signals is thought to play a key role in the disease process. The aim of the present study was to investigate the effects of cranberry proanthocyanidins (PACs) on gene expression and cytotoxic activities of LtxA. We showed that cranberry PACs dose-dependently attenuate the expression of genes making up the leukotoxin operon, including ltxB and ltxC, in the two strains of A. actinomycetemcomitans tested. Cranberry PACs (≥62.5 µg/mL) protected macrophages against the cytotoxic effect of purified LtxA. Moreover, cranberry PACs reduced caspase-1 activation in LtxA-treated macrophages and consequently decreased the release of both IL-1ß and IL-18, which are known as damage-associated molecular patterns (DAMPs) and contribute to the progression of periodontitis by increasing cell migration and osteoclastogenesis. In addition, cranberry PACs reduced the expression of genes encoding the P2X7 receptor and NALP3 (NACHT, LRR and PYD domains-containing protein 3), which play key roles in pore formation and cell death. Lastly, cranberry PACs blocked the binding of LtxA to macrophages and consequently reduced the LtxA-mediated cytotoxicity. In summary, the present study showed that cranberry PACs reduced LtxA gene expression in A. actinomycetemcomitans and neutralized the cytolytic and pro-inflammatory responses of human macrophages treated with LtxA. Given these properties, cranberry PACs may represent promising molecules for prevention and treatment of the aggressive form of periodontitis caused by A. actinomycetemcomitans.


Assuntos
Aggregatibacter actinomycetemcomitans/química , Exotoxinas/antagonistas & inibidores , Proantocianidinas/química , Vaccinium macrocarpon/química , Exotoxinas/farmacologia , Humanos , Células U937
10.
FASEB J ; 33(12): 13412-13422, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31545910

RESUMO

Among the main bacteria implicated in the pathology of periodontal disease, Aggregatibacter actinomycetemcomitans (Aa) is well known for causing loss of periodontal attachment and systemic disease. Recent studies have suggested that secreted extracellular RNAs (exRNAs) from several bacteria may be important in periodontitis, although their role is unclear. Emerging evidence indicates that exRNAs circulate in nanosized bilayered and membranous extracellular vesicles (EVs) known as outer membrane vesicles (OMVs) in gram-negative bacteria. In this study, we analyzed the small RNA expression profiles in activated human macrophage-like cells (U937) infected with OMVs from Aa and investigated whether these cells can harbor exRNAs of bacterial origin that have been loaded into the host RNA-induced silencing complex, thus regulating host target transcripts. Our results provide evidence for the cytoplasmic delivery and activity of microbial EV-derived small exRNAs in host gene regulation. The production of TNF-α was promoted by exRNAs via the TLR-8 and NF-κB signaling pathways. Numerous studies have linked periodontal disease to neuroinflammatory diseases but without elucidating specific mechanisms for the connection. We show here that intracardiac injection of Aa OMVs in mice showed successful delivery to the brain after crossing the blood-brain barrier, the exRNA cargos increasing expression of TNF-α in the mouse brain. The current study indicates that host gene regulation by microRNAs originating from OMVs of the periodontal pathogen Aa is a novel mechanism for host gene regulation and that the transfer of OMV exRNAs to the brain may cause neuroinflammatory diseases like Alzheimer's.-Han, E.-C., Choi, S.-Y., Lee, Y., Park, J.-W., Hong, S.-H., Lee, H.-J. Extracellular RNAs in periodontopathogenic outer membrane vesicles promote TNF-α production in human macrophages and cross the blood-brain barrier in mice.


Assuntos
Membrana Externa Bacteriana/metabolismo , Barreira Hematoencefálica/metabolismo , Vesículas Extracelulares/genética , Macrófagos/metabolismo , Doenças Periodontais/metabolismo , Pequeno RNA não Traduzido/genética , Fator de Necrose Tumoral alfa/metabolismo , Aggregatibacter actinomycetemcomitans/química , Aggregatibacter actinomycetemcomitans/genética , Animais , Vesículas Extracelulares/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Doenças Periodontais/microbiologia , Doenças Periodontais/patologia , RNA Bacteriano/genética , Células U937
11.
Mol Oral Microbiol ; 34(5): 183-193, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31271525

RESUMO

Studies have been conducted on the pathogenicity of periodontopathogens in cultures of endothelial cells on two-dimensional (2D) polystyrene surfaces, where the monolayer formed is not exposed to proteins of the subendothelial matrix. In this work, we developed a culture system by seeding human coronary artery endothelial cells (HCAECs) onto three-dimensional (3D) scaffolds of collagen type I, a subendothelial protein. The inflammatory responses of the HCAEC monolayers, formed either on 3D scaffolds or directly on a 2D polystyrene plate, to lipopolysaccharide (LPS) from Aggregatibacter actinomycetemcomitans (Aa) and Porphyromonas gingivalis (Pg) were evaluated. The transcription of 3 genes, the secretion of 40 cytokines and 2 prostanoids, and the adhesion of monocytes to 2D and 3D cultures with or without exposure to lipopolysaccharides (control) were assessed. HCAECs exhibited differences in transcriptional and secretory profiles between the 3D and 2D models. In addition, the inflammatory responses of HCAEC to Aa-LPS and Pg-LPS differed between the two models. In 3D cultures treated with Aa-LPS, the levels of IL-8, RANTES, G-CSF, ICAM-1, IL-6, and TXA2 were significantly higher than those in the controls. In 2D cultures treated with Aa-LPS, IL-8, RANTES, G-CSF, ICAM-1, TNF-RI, PGI2, and TXA2 levels were significantly higher than those in their controls. In the presence of Aa-LPS, monocyte adhesion did not differ between treated and control 3D cultures but was significantly higher in treated 2D cultures than in the controls. In response to Pg-LPS, cytokine-prostaglandin secretion and monocyte adhesion did not differ between 3D and 2D cultures. These data indicate that HCAECs respond differently to these two types of LPS.


Assuntos
Adesão Celular , Técnicas de Cultura de Células , Células Endoteliais , Lipopolissacarídeos , Alicerces Teciduais , Aggregatibacter actinomycetemcomitans/química , Humanos , Monócitos/fisiologia , Porphyromonas gingivalis/química
12.
mBio ; 10(4)2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31289186

RESUMO

The Escherichia coli hemolysin (HlyA) is a pore-forming exotoxin associated with severe complications of human urinary tract infections. HlyA is the prototype of the repeats-in-toxin (RTX) family, which includes LtxA from Aggregatibacter actinomycetemcomitans, a periodontal pathogen. The existence and requirement for a host cell receptor for these toxins are controversial. We performed an unbiased forward genetic selection in a mutant library of human monocytic cells, U-937, for host factors involved in HlyA cytotoxicity. The top candidate was the ß2 integrin ß subunit. Δß2 cell lines are approximately 100-fold more resistant than wild-type U-937 cells to HlyA, but remain sensitive to HlyA at high concentrations. Similarly, Δß2 cells are more resistant than wild-type U-937 cells to LtxA, as Δß2 cells remain LtxA resistant even at >1,000-fold-higher concentrations of the toxin. Loss of any single ß2 integrin α subunit, or even all four α subunits together, does not confer resistance to HlyA. HlyA and LtxA bind to the ß2 subunit, but not to αL, αM, or αX in far-Western blots. Genetic complementation of Δß2 cells with either ß2 or ß2 with a cytoplasmic tail deletion restores HlyA and LtxA sensitivity, suggesting that ß2 integrin signaling is not required for cytotoxicity. Finally, ß2 mutations do not alter sensitivity to unrelated pore-forming toxins, as wild-type or Δß2 cells are equally sensitive to Staphylococcus aureus α-toxin and Proteus mirabilis HpmA. Our studies show two RTX toxins use the ß2 integrin ß subunit alone to facilitate cytotoxicity, but downstream integrin signaling is dispensable.IMPORTANCE Urinary tract infections are one of the most common bacterial infections worldwide. Uropathogenic Escherichia coli strains are responsible for more than 80% of community-acquired urinary tract infections. Although we have known for nearly a century that severe infections stemming from urinary tract infections, including kidney or bloodstream infections are associated with expression of a toxin, hemolysin, from uropathogenic Escherichia coli, how hemolysin functions to enhance virulence is unknown. Our research defines the interaction of hemolysin with the ß2 integrin, a human white cell adhesion molecule, as a potential therapeutic target during urinary tract infections. The E. coli hemolysin is the prototype for a toxin family (RTX family) produced by a wide array of human and animal pathogens. Our work extends to the identification and characterization of the receptor for an additional member of the RTX family, suggesting that this interaction may be broadly conserved throughout the RTX toxin family.


Assuntos
Aggregatibacter actinomycetemcomitans/química , Antígenos CD18/química , Proteínas de Escherichia coli/química , Escherichia coli/química , Exotoxinas/química , Proteínas Hemolisinas/química , Aggregatibacter actinomycetemcomitans/genética , Toxinas Bacterianas/química , Antígenos CD18/genética , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Escherichia coli/genética , Teste de Complementação Genética , Humanos , Monócitos/microbiologia , Monócitos/patologia , Mutação , Ligação Proteica , Células U937
13.
J Bacteriol ; 201(16)2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31160398

RESUMO

Bacterial adhesion to host tissues is considered the first and critical step of microbial infection. The extracellular matrix protein adhesin A (EmaA) is a collagen-binding adhesin of the periodontal pathogen Aggregatibacter actinomycetemcomitans Three 202-kDa EmaA monomers form antenna-like structures on the bacterial surface with the functional domain located at the apical end. The structure of the 30-nm functional domain has been determined by three-dimensional (3D) electron tomography and subvolume averaging. The region exhibits a complex architecture composed of three subdomains (SI to SIII) and a linker between subdomains SII and SIII. However, the molecular interaction between the adhesin receptor complexes has yet to be revealed. This study provides the first detailed 3D structure of reconstituted EmaA/collagen complexes obtained using 3D electron tomography and image processing techniques. The observed interactions of EmaA with collagen were not to whole, intact fibrils, but rather to individual collagen triple helices dissociated from the fibrils. The majority of the contacts with the EmaA functional domain encompassed subdomains SII and SIII and in some cases the tip of the apical domain, involving SI. These data suggest a multipronged mechanism for the interaction of Gram-negative bacteria with collagen.IMPORTANCE Bacterial adhesion is a crucial step for bacterial colonization and infection. In recent years, the number of antibiotic-resistant strains has dramatically increased; therefore, there is a need to search for novel antimicrobial agents. Thus, great efforts are being devoted to develop a clear understanding of the bacterial adhesion mechanism for preventing infections. In host/pathogen interactions, once repulsive forces are overcome, adhesins recognize and tightly bind to specific receptors on the host cell or tissue components. Here, we present the first 3D structure of the interaction between the collagen-binding adhesin EmaA and collagen, which is critical for the development of endocarditis in humans.


Assuntos
Adesinas Bacterianas/química , Adesinas Bacterianas/metabolismo , Aggregatibacter actinomycetemcomitans/metabolismo , Colágeno/química , Colágeno/metabolismo , Infecções por Pasteurellaceae/metabolismo , Adesinas Bacterianas/genética , Aggregatibacter actinomycetemcomitans/química , Aggregatibacter actinomycetemcomitans/genética , Tomografia com Microscopia Eletrônica , Humanos , Infecções por Pasteurellaceae/microbiologia , Ligação Proteica , Domínios Proteicos
14.
Biochim Biophys Acta Gen Subj ; 1863(1): 191-198, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30342156

RESUMO

BACKGROUND: Catechins, polyphenols derived from tea leaves, have been shown to have antibacterial properties, through direct killing of bacteria as well as through inhibition of bacterial toxin activity. In particular, certain catechins have been shown to have bactericidal effects on the oral bacterium, Aggregatibacter actinomycetemcomitans, as well as the ability to inhibit a key virulence factor of this organism, leukotoxin (LtxA). The mechanism of catechin-mediated inhibition of LtxA has not been shown. METHODS: In this work, we studied the ability of six catechins to inhibit LtxA-mediated cytotoxicity in human white blood cells, using Trypan blue staining, and investigated the mechanism of action using a combination of techniques, including fluorescence and circular dichroism spectroscopy, confocal microscopy, and surface plasmon resonance. RESULTS: We found that all the catechins except (-)-catechin inhibited the activity of this protein, with the galloylated catechins having the strongest effect. Pre-incubation of the toxin with the catechins increased the inhibitory action, indicating that the catechins act on the protein, rather than the cell. The secondary structure of LtxA was dramatically altered in the presence of catechin, which resulted in an inhibition of toxin binding to cholesterol, an important initial step in the cytotoxic mechanism of the toxin. CONCLUSIONS: These results demonstrate that the catechins inhibit LtxA activity by altering its structure to prevent interaction with specific molecules present on the host cell surface. GENERAL SIGNIFICANCE: Galloylated catechins modify protein toxin structure, inhibiting the toxin from binding to the requisite molecules on the host cell surface.


Assuntos
Aggregatibacter actinomycetemcomitans/química , Toxinas Bacterianas/química , Catequina/química , Colesterol/química , Exotoxinas/química , Leucócitos/microbiologia , Membrana Celular/metabolismo , Sobrevivência Celular , Dicroísmo Circular , Humanos , Leucócitos/metabolismo , Fluidez de Membrana , Microscopia Confocal , Periodontite/terapia , Estrutura Secundária de Proteína , Ressonância de Plasmônio de Superfície , Células THP-1
15.
PLoS One ; 13(10): e0205871, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30335797

RESUMO

The oral bacterium, Aggregatibacter actinomycetemcomitans, which is associated with localized aggressive periodontitis, as well as systemic infections including endocarditis, produces numerous virulence factors, including a repeats-in-toxin (RTX) protein called leukotoxin (LtxA), which kills human immune cells. The strains of A. actinomycetemcomitans most closely associated with disease have been shown to produce the most LtxA, suggesting that LtxA plays a significant role in the virulence of this organism. LtxA, like many of the RTX toxins, can be divided into four functional domains: an N-terminal hydrophobic domain, which contains a significant fraction of hydrophobic residues and has been proposed to play a role in the membrane interaction of the toxin; the central domain, which contains two lysine residues that are the sites of post-translational acylation; the repeat domain that is characteristic of the RTX toxins, and a C-terminal domain thought to be involved in secretion. In its initial interaction with the host cell, LtxA must bind to both cholesterol and an integrin receptor, lymphocyte function-associated antigen-1 (LFA-1). While both interactions are essential for toxicity, the domains of LtxA involved remain unknown. We therefore undertook a series of experiments, including tryptophan quenching and trypsin digestion, to characterize the structure of LtxA upon interaction with membranes of various lipid compositions. Our results demonstrate that LtxA adopts a U-shaped conformation in the membrane, with the N- and C-terminal domains residing outside of the membrane.


Assuntos
Aggregatibacter actinomycetemcomitans/química , Proteínas de Bactérias/química , Colesterol/química , Proteínas Hemolisinas/química , Antígeno-1 Associado à Função Linfocitária/química , Fatores de Virulência/química , Aggregatibacter actinomycetemcomitans/crescimento & desenvolvimento , Aggregatibacter actinomycetemcomitans/patogenicidade , Sequência de Aminoácidos , Proteínas de Bactérias/isolamento & purificação , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Colesterol/metabolismo , Dimiristoilfosfatidilcolina/química , Dimiristoilfosfatidilcolina/metabolismo , Proteínas Hemolisinas/isolamento & purificação , Proteínas Hemolisinas/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Células Jurkat , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Lipossomos/química , Lipossomos/metabolismo , Antígeno-1 Associado à Função Linfocitária/metabolismo , Fosfatidiletanolaminas/química , Fosfatidiletanolaminas/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Proteólise , Tripsina/química , Fatores de Virulência/isolamento & purificação , Fatores de Virulência/metabolismo
16.
Virulence ; 9(1): 1205-1223, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30088437

RESUMO

Naturally competent bacteria acquire DNA from their surroundings to survive in nutrient-poor environments and incorporate DNA into their genomes as new genes for improved survival. The secretin HofQ from the oral pathogen Aggregatibacter actinomycetemcomitans has been associated with DNA uptake. Cytokine sequestering is a potential virulence mechanism in various bacteria and may modulate both host defense and bacterial physiology. The objective of this study was to elucidate a possible connection between natural competence and cytokine uptake in A. actinomycetemcomitans. The extramembranous domain of HofQ (emHofQ) was shown to interact with various cytokines, of which IL-8 exhibited the strongest interaction. The dissociation constant between emHofQ and IL-8 was 43 nM in static settings and 2.4 µM in dynamic settings. The moderate binding affinity is consistent with the hypothesis that emHofQ recognizes cytokines before transporting them into the cells. The interaction site was identified via crosslinking and mutational analysis. By structural comparison, relateda type I KH domain with a similar interaction site was detected in the Neisseria meningitidis secretin PilQ, which has been shown to participate in IL-8 uptake. Deletion of hofQ from the A. actinomycetemcomitans genome decreased the overall biofilm formation of this organism, abolished the response to cytokines, i.e., decreased eDNA levels in the presence of cytokines, and increased the susceptibility of the biofilm to tested ß-lactams. Moreover, we showed that recombinant IL-8 interacted with DNA. These results can be used in further studies on the specific role of cytokine uptake in bacterial virulence without interfering with natural-competence-related DNA uptake.


Assuntos
Aggregatibacter actinomycetemcomitans/química , Proteínas de Bactérias/genética , Citocinas/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Interleucina-8/metabolismo , Secretina/metabolismo , Aggregatibacter actinomycetemcomitans/genética , Aggregatibacter actinomycetemcomitans/patogenicidade , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/imunologia , Biofilmes/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Fímbrias/química , Proteínas de Fímbrias/genética , Humanos , Interleucina-8/imunologia , Periodontite/imunologia , Periodontite/microbiologia , Domínios e Motivos de Interação entre Proteínas/genética , Domínios e Motivos de Interação entre Proteínas/fisiologia , Secretina/imunologia , Virulência , beta-Lactamas/farmacologia
17.
Med Microbiol Immunol ; 207(5-6): 329-338, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30056510

RESUMO

Phosphorylcholine (ChoP) is covalently incorporated into bacterial surface structures, contributing to host mimicry and promoting adhesion to surfaces. Our aims were to determine the frequency of ChoP display among Aggregatibacter actinomycetemcomitans strains, to clarify which surface structures bear ChoP, and whether ChoP-positivity relates to serum killing. The tested oral (N = 67) and blood isolates (N = 27) represented 6 serotypes. Mab TEPC-15 was used for immunoblotting of cell lysates and fractions and for immunofluorescence microscopy of cell surface-bound ChoP. The lysates were denatured with urea for hidden ChoP or treated with proteinase K to test whether it binds to a protein. Three ChoP-positive and two ChoP-negative strains were subjected to serum killing in the presence/absence of CRP and using Ig-depleted serum as complement source. Cell lysates and the first soluble cellular fraction revealed a < 10 kDa band in immunoblots. Among 94 strains, 27 were ChoP positive. No difference was found in the prevalence of ChoP-positive oral (21/67) and blood (6/27) strains. Immunofluorescence microscopy corresponded to the immunoblot results. Proteinase K abolished ChoP reactivity, whereas urea did not change the negative result. The TEPC-15-reactive protein was undetectable in Δflp1 mutant strain. The survival rate of serotype-b strains in serum was 100% irrespective of ChoP, but that of serotype-a was higher in ChoP-positive (85%) than ChoP-negative (71%) strains. The results suggest that a third of rough-colony strains harbor ChoP and that ChoP is attached to fimbrial subunit protein Flp1. It further seems that ChoP-positivity does not enhance but may reduce A. actinomycetemcomitans susceptibility to serum killing.


Assuntos
Aggregatibacter actinomycetemcomitans/química , Aggregatibacter actinomycetemcomitans/imunologia , Proteínas de Bactérias/química , Fosforilcolina/análise , Aggregatibacter actinomycetemcomitans/isolamento & purificação , Proteínas de Bactérias/genética , Sangue/microbiologia , Atividade Bactericida do Sangue , Deleção de Genes , Humanos , Immunoblotting , Viabilidade Microbiana , Microscopia de Fluorescência , Boca/microbiologia , Infecções por Pasteurellaceae/microbiologia , Sorogrupo
18.
Infect Immun ; 86(3)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29229732

RESUMO

The Gram-negative bacterium Aggregatibacter actinomycetemcomitans is a causative agent of localized aggressive periodontitis. Critical to its infection process is the first and essential step of attachment, which is related to the coordinated functions of surface components comprised of proteins and extracellular polysaccharides. One such protein is the outer membrane trimeric autotransporter protein ApiA, a versatile virulence factor with numerous functions, including cell binding, invasion, serum resistance, autoaggregation, and induction of cytokine release. Here we report on the use of Escherichia coli strains expressing protein variants to define the separate functions ascribed to the N terminus and those related to the C terminus. Importantly, a hybrid protein that comprised the N terminus of trimeric ApiA and the ß-barrel domain of monomeric autotransporter Aae was constructed, which allowed the expression of a monomer surface-exposed domain of ApiA. Functional and phenotypic analyses demonstrated that the C terminus of ApiA forms an independent domain that is crucial for general stability and trimer formation, which appears to be associated with autoaggregation, biofilm formation, and surface expression. Importantly, the results show that the monomeric form of the N-terminal passenger domain of ApiA, while surface exposed, is sufficient for binding to buccal epithelial cells; however, it is not sufficient to allow aggregation and biofilm formation, strengthening the importance of the role of trimerization in these phenotypes.


Assuntos
Aggregatibacter actinomycetemcomitans/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Infecções por Pasteurellaceae/microbiologia , Sistemas de Secreção Tipo V/química , Sistemas de Secreção Tipo V/metabolismo , Aggregatibacter actinomycetemcomitans/química , Aggregatibacter actinomycetemcomitans/genética , Proteínas de Bactérias/genética , Humanos , Domínios Proteicos , Multimerização Proteica , Transporte Proteico , Sistemas de Secreção Tipo V/genética
19.
Anaerobe ; 48: 103-109, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28780429

RESUMO

Varying cytokine responses of human gingival epithelial cells (HGECs) by Aggregatibacter actinomycetemcomitans subtypes have been found. Most studies have used reference strains, whereas a few has evaluated the cytokine expression in response to clinical subtypes of this bacterial species. This study aimed to examine whether there was any difference in cytokine responses of HGECs stimulated with cell wall extract (CWE) from A. actinomycetemcomitans subtypes included clinical strains from Thai adult periodontitis, various serotypes and non-serotypeable strains, strains from deep or shallow pockets, and reference serotype strains. Totally 50 clinical strains and 7 reference strains of A. actinomycetemcomitans were analyzed for the expression of IL-1ß, IL-6, IL-8, and TNF-α mRNAs in HGECs by real time-PCR, and the IL-8 concentrations in cell-free supernatant measured using ELISA. An in vitro effect of released IL-8 on neutrophil migration was examined using transwell chambers. Result showed that among four cytokines studied, IL-8 mRNA was highly up-regulated by both clinical and reference strains. Serotype f revealed the highest expression compared to other serotypes. The JP2-like leukotoxin promoter gene and non-serotypeable (NS1 and NS2) demonstrated lower IL-8 responses compared to serotypeable strains, and IL-8 responses upon stimulation with clinical strains from deep pockets were also significantly lower than those isolated from shallow pockets (P < 0.01). Our findings suggest that the clinical isolates of A. actinomycetemcomitans associating with deep pockets, JP2-like leukotoxin promoter gene, NS1, and NS2 may interfere neutrophil function via minimal and immunosuppressing IL-8 responses, which may enhance their survival and virulence.


Assuntos
Aggregatibacter actinomycetemcomitans/química , Parede Celular/química , Gengiva/microbiologia , Bolsa Gengival/microbiologia , Interleucina-8/biossíntese , Periodontite/microbiologia , Aggregatibacter actinomycetemcomitans/imunologia , Aggregatibacter actinomycetemcomitans/isolamento & purificação , Movimento Celular/imunologia , Células Cultivadas , Exotoxinas/genética , Gengiva/patologia , Humanos , Interleucina-8/genética , Neutrófilos/imunologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/biossíntese
20.
Arch Oral Biol ; 82: 127-133, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28641178

RESUMO

OBJECTIVE: While different virulence factors have been reported of Aggregatibacter actinomycetemcomitans (Aa), there is little information about the stimulatory effect of its DNA. The main purpose of this study was to assess the inflammatory response of human gingival fibroblasts (HGFs) stimulated with A. actinomycetemcomitans DNA. DESIGN: Cytokine levels of IL-6, IL-1α and TNF-α were measured on the supernatant of HGFs activated with 10, 25, 50 and 100µg/ml DNA of Aa during 24h. Primary cultures of HGFs were infected with Aa and its DNA at different times and concentrations to compare its cytotoxic effect. Cell damage and adhesion of Aa to HGFs were evaluated under light microscopy and Scanning electron microscopy respectively. RESULTS: There was a statistical difference (p<0.05) in cytokine expression in HGFs activated by bacterial DNA with a dose dependent on IL-6 expression and a significantly elevated expression of IL-1α and TNF-α compared to Human DNA negative control. Substantial morphological alterations were observed after infection of A. actinomycetemcomitans in HGFs but not with bDNA exposure. Aggregatibacter actinomycetemcomitans showed a high rate of adhesion and cell damage to HGFs after 30min. CONCLUSIONS: Genomic DNA of A. actinomycetemcomitans could be a factor in the pathogenesis of periodontitis that might play a major role in the inflammatory response.


Assuntos
Aggregatibacter actinomycetemcomitans/química , DNA Bacteriano/química , Fibroblastos/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-6/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Aderência Bacteriana , Gengiva/citologia , Humanos , Técnicas In Vitro , Microscopia Eletrônica de Varredura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...