Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Microbiol Spectr ; 10(3): e0117521, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35647689

RESUMO

Staphylococcus aureus infections have become a major challenge in health care due to increasing antibiotic resistance. We aimed to design small molecule inhibitors of S. aureus surface proteins to be developed as colonization inhibitors. We identified allantodapsone in an initial screen searching for inhibitors of clumping factors A and B (ClfA and ClfB). We used microbial adhesion assays to investigate the effect of allantodapsone on extracellular matrix protein interactions. Allantodapsone inhibited S. aureus Newman adhesion to fibrinogen with an IC50 of 21.3 µM (95% CI 4.5-102 µM), minimum adhesion inhibitory concentration (MAIC) of 100 µM (40.2 µg/mL). Additionally, allantodapsone inhibited adhesion of Lactococcus lactis strains exogenously expressing the clumping factors to fibrinogen (L. lactis ClfA, IC50 of 3.8 µM [95% CI 1.0-14.3 µM], MAIC 10 µM, 4.0 µg/mL; and L. lactis ClfB, IC50 of 11.0 µM [95% CI 0.9-13.6 µM], MAIC 33 µM, 13.3 µg/mL), indicating specific inhibition. Furthermore, the dapsone and alloxan fragments of allantodapsone did not have any inhibitory effect. Adhesion of S. aureus Newman to L2v loricrin is dependent on the expression of ClfB. Allantodapsone caused a dose dependent inhibition of S. aureus adhesion to the L2v loricrin fragment, with full inhibition at 40 µM (OD600 0.11 ± 0.01). Furthermore, recombinant ClfB protein binding to L2v loricrin was inhibited by allantodapsone (P < 0.0001). Allantodapsone also demonstrated dose dependent inhibition of S. aureus Newman adhesion to cytokeratin 10 (CK10). Allantodapsone is the first small molecule inhibitor of the S. aureus clumping factors with potential for development as a colonization inhibitor. IMPORTANCE S. aureus colonization of the nares and the skin provide a reservoir of bacteria that can be transferred to wounds that can ultimately result in systemic infections. Antibiotic resistance can make these infections difficult to treat with significant associated morbidity and mortality. We have identified and characterized a first-in-class small molecule inhibitor of the S. aureus clumping factors A and B, which has the potential to be developed further as a colonization inhibitor.


Assuntos
Queratinas/metabolismo , Infecções Estafilocócicas , Staphylococcus aureus , Adesinas Bacterianas/metabolismo , Aderência Bacteriana/fisiologia , Fibrinogênio/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo
2.
Sci Rep ; 11(1): 22799, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34815454

RESUMO

Staphylococcus aureus is the cause of a spectrum of diseases in humans and animals. The molecular basis of this pathogenicity lies in the expression of a variety of virulence factors, including proteins that mediate adherence to the host plasma and extracellular matrix proteins. In this study, we discovered that the iron-regulated surface determinant B (IsdB) protein, besides being involved in iron transport and vitronectin binding, interacts with von Willebrand Factor (vWF). IsdB-expressing bacteria bound to both soluble and immobilized vWF. The binding of recombinant IsdB to vWF was blocked by heparin and reduced at high ionic strength. Furthermore, treatment with ristocetin, an allosteric agent that promotes the exposure of the A1 domain of vWF, potentiates the binding of IsdB to vWF. Both near-iron transporter motifs NEAT1 and NEAT2 of IsdB individually bound recombinant A1 domain with KD values in the micromolar range. The binding of IsdB and adhesion of S. aureus expressing IsdB to monolayers of activated endothelial cells was significantly inhibited by a monoclonal antibody against the A1 domain and by IsdB reactive IgG from patients with staphylococcal endocarditis. This suggests the importance of IsdB in adherence of S. aureus to the endothelium colonization and as potential therapeutic target.


Assuntos
Aderência Bacteriana , Proteínas de Transporte de Cátions/metabolismo , Endotélio Vascular/metabolismo , Staphylococcus aureus/fisiologia , Fator de von Willebrand/metabolismo , Proteínas de Transporte de Cátions/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Ligação Proteica , Fator de von Willebrand/genética
4.
Clin Microbiol Rev ; 34(2)2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33361142

RESUMO

Staphylococcus lugdunensis is a species of coagulase-negative staphylococcus (CoNS) that causes serious infections in humans akin to those of S. aureus It was often misidentified as S. aureus, but this has been rectified by recent routine use of matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) in diagnostic laboratories. It encodes a diverse array of virulence factors for adhesion, cytotoxicity, and innate immune evasion, but these are less diverse than those encoded by S. aureus It expresses an iron-regulated surface determinant (Isd) system combined with a novel energy-coupling factor (ECF) mechanism for extracting heme from hemoproteins. Small cytolytic S. lugdunensis synergistic hemolysins (SLUSH), peptides related to phenol-soluble modulins of S. aureus, act synergistically with ß-toxin to lyse erythrocytes. S. lugdunensis expresses a novel peptide antibiotic, lugdunin, that can influence the nasal and skin microbiota. Endovascular infections are initiated by bacterial adherence to fibrinogen promoted by a homologue of Staphylococcus aureus clumping factor A and to von Willebrand factor on damaged endothelium by an uncharacterized mechanism. S. lugdunensis survives within mature phagolysosomes of macrophages without growing and is released only following apoptosis. This differs fundamentally from S. aureus, which actively grows and expresses bicomponent leukotoxins that cause membrane damage and could contribute to survival in the infected host. S. lugdunensis is being investigated as a probiotic to eradicate S. aureus from the nares of carriers. However, this is contraindicated by its innate virulence. Studies to obtain a deeper understanding of S. lugdunensis colonization, virulence, and microbiome interactions are therefore warranted.


Assuntos
Infecções Estafilocócicas , Staphylococcus lugdunensis , Humanos , Ferro , Staphylococcus aureus , Fatores de Virulência
5.
Front Microbiol ; 11: 1829, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32849430

RESUMO

Staphylococcus epidermidis is a ubiquitous commensal of human skin. The widespread use of indwelling medical devices in modern medicine provides an opportunity for it to cause infections. Disease causing isolates can come from many different genetic backgrounds. Multiply antibiotic resistant strains have spread globally. S. epidermidis has a smaller repertoire of cell wall anchored (CWA) surface proteins than Staphylococcus aureus. Nevertheless, these CWA proteins promote adhesion to components of the extracellular matrix including collagen, fibrinogen, and fibronectin and contribute to the formation of biofilm. The A domain of the accumulation associated protein Aap can promote adhesion to unconditioned biomaterial but must be removed proteolytically to allow accumulation to proceed by homophilic Zn2+-dependent interactions. Mature biofilm contains amyloid structures formed by Aap and the small basic protein (Sbp). The latter contributes to the integrity of both protein and polysaccharide biofilm matrices. Several other CWA proteins can also promote S. epidermidis biofilm formation.

6.
J Biol Chem ; 295(29): 10008-10022, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32499371

RESUMO

Staphylococcus aureus is an important bacterial pathogen that can cause a wide spectrum of diseases in humans and other animals. S. aureus expresses a variety of virulence factors that promote infection with this pathogen. These include cell-surface proteins that mediate adherence of the bacterial cells to host extracellular matrix components, such as fibronectin and fibrinogen. Here, using immunoblotting, ELISA, and surface plasmon resonance analysis, we report that the iron-regulated surface determinant B (IsdB) protein, besides being involved in heme transport, plays a novel role as a receptor for the plasma and extracellular matrix protein vitronectin (Vn). Vn-binding activity was expressed by staphylococcal strains grown under iron starvation conditions when Isd proteins are expressed. Recombinant IsdB bound Vn dose dependently and specifically. Both near-iron transporter motifs NEAT1 and NEAT2 of IsdB individually bound Vn in a saturable manner, with KD values in the range of 16-18 nm Binding of Vn to IsdB was specifically blocked by heparin and reduced at high ionic strength. Furthermore, IsdB-expressing bacterial cells bound significantly higher amounts of Vn from human plasma than did an isdB mutant. Adherence to and invasion of epithelial and endothelial cells by IsdB-expressing S. aureus cells was promoted by Vn, and an αvß3 integrin-blocking mAb or cilengitide inhibited adherence and invasion by staphylococci, suggesting that Vn acts as a bridge between IsdB and host αvß3 integrin.


Assuntos
Proteínas de Transporte de Cátions/química , Staphylococcus aureus/química , Vitronectina/química , Proteínas de Transporte de Cátions/metabolismo , Humanos , Integrina alfaVbeta3/química , Integrina alfaVbeta3/metabolismo , Ligação Proteica , Staphylococcus aureus/metabolismo , Vitronectina/metabolismo
7.
mBio ; 10(6)2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31848274

RESUMO

Staphylococcus epidermidis is a significant opportunistic pathogen of humans. Molecular studies in this species have been hampered by the presence of restriction-modification (RM) systems that limit introduction of foreign DNA. Here, we establish the complete genomes and methylomes for seven clinically significant, genetically diverse S. epidermidis isolates and perform the first systematic genomic analyses of the type I RM systems within both S. epidermidis and Staphylococcus aureus Our analyses revealed marked differences in the gene arrangement, chromosomal location, and movement of type I RM systems between the two species. Unlike S. aureus, S. epidermidis type I RM systems demonstrate extensive diversity even within a single genetic lineage. This is contrary to current assumptions and has important implications for approaching the genetic manipulation of S. epidermidis Using Escherichia coli plasmid artificial modification (PAM) to express S. epidermidishsdMS, we readily overcame restriction barriers in S. epidermidis and achieved electroporation efficiencies equivalent to those of modification-deficient mutants. With these functional experiments, we demonstrated how genomic data can be used to predict both the functionality of type I RM systems and the potential for a strain to be electroporation proficient. We outline an efficient approach for the genetic manipulation of S. epidermidis strains from diverse genetic backgrounds, including those that have hitherto been intractable. Additionally, we identified S. epidermidis BPH0736, a naturally restriction-defective, clinically significant, multidrug-resistant ST2 isolate, as an ideal candidate for molecular studies.IMPORTANCEStaphylococcus epidermidis is a major cause of hospital-acquired infections, especially those related to implanted medical devices. Understanding how S. epidermidis causes disease and devising ways to combat these infections have been hindered by an inability to genetically manipulate clinically significant hospital-adapted strains. Here, we provide the first comprehensive analyses of the barriers to the uptake of foreign DNA in S. epidermidis and demonstrate that these are distinct from those described for S. aureus Using these insights, we demonstrate an efficient approach for the genetic manipulation of S. epidermidis to enable the study of clinical isolates for the first time.


Assuntos
Biologia Computacional , Mineração de Dados , Desoxirribonucleases de Sítio Específico do Tipo I/genética , Epigenoma , Epigenômica , Perfilação da Expressão Gênica , Staphylococcus epidermidis/fisiologia , Mapeamento Cromossômico , Biologia Computacional/métodos , Elementos de DNA Transponíveis , Desoxirribonucleases de Sítio Específico do Tipo I/química , Desoxirribonucleases de Sítio Específico do Tipo I/metabolismo , Epigenômica/métodos , Evolução Molecular , Interações Hospedeiro-Patógeno , Humanos , Filogenia , Plasmídeos/genética , Plasmídeos/metabolismo , Fagos de Staphylococcus/genética , Staphylococcus epidermidis/classificação , Staphylococcus epidermidis/virologia
8.
Trends Microbiol ; 27(11): 927-941, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31375310

RESUMO

The microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) are a family of proteins that are defined by the presence of two adjacent IgG-like folded subdomains. These promote binding to ligands by mechanisms that involve major conformational changes exemplified by the binding to fibrinogen by the 'dock-lock-latch' mechanism or to collagen by the 'collagen hug'. Clumping factors A and B are two such MSCRAMMs that have several important roles in the pathogenesis of Staphylococcus aureus infections. MSCRAMM architecture, ligand binding, and roles in infection and colonization are examined with a focus on recent developments with clumping factors.


Assuntos
Adesinas Bacterianas/fisiologia , Cocos Gram-Positivos/fisiologia , Adesinas Bacterianas/química , Proteínas de Bactérias/química , Proteínas de Transporte/química , Parede Celular/química , Parede Celular/fisiologia , Coagulase/química , Humanos , Ligantes , Ligação Proteica , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/fisiologia , Staphylococcus epidermidis/fisiologia
9.
Microbiol Spectr ; 7(4)2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31267926

RESUMO

The surface of Staphylococcus aureus is decorated with over 20 proteins that are covalently anchored to peptidoglycan by the action of sortase A. These cell wall-anchored (CWA) proteins can be classified into several structural and functional groups. The largest is the MSCRAMM family, which is characterized by tandemly repeated IgG-like folded domains that bind peptide ligands by the dock lock latch mechanism or the collagen triple helix by the collagen hug. Several CWA proteins comprise modules that have different functions, and some individual domains can bind different ligands, sometimes by different mechanisms. For example, the N-terminus of the fibronectin binding proteins comprises an MSCRAMM domain which binds several ligands, while the C-terminus is composed of tandem fibronectin binding repeats. Surface proteins promote adhesion to host cells and tissue, including components of the extracellular matrix, contribute to biofilm formation by stimulating attachment to the host or indwelling medical devices followed by cell-cell accumulation via homophilic interactions between proteins on neighboring cells, help bacteria evade host innate immune responses, participate in iron acquisition from host hemoglobin, and trigger invasion of bacteria into cells that are not normally phagocytic. The study of genetically manipulated strains using animal infection models has shown that many CWA proteins contribute to pathogenesis. Fragments of CWA proteins have the potential to be used in multicomponent vaccines to prevent S. aureus infections.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Membrana/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Animais , Aderência Bacteriana , Proteínas de Bactérias/genética , Humanos , Proteínas de Membrana/genética , Staphylococcus aureus/genética
10.
Pathogens ; 8(1)2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30609641

RESUMO

Staphylococcus aureus is a major cause of corneal infections that can cause reduced vision, even blindness. Secreted toxins cause tissue damage and inflammation resulting in scars that lead to vision loss. Identifying tissue damaging proteins is a prerequisite to limiting these harmful reactions. The present study characterized a previously unrecognized S. aureus toxin. This secreted toxin was purified from strain Newman ΔhlaΔhlg, the N-terminal sequence determined, the gene cloned, and the purified recombinant protein was tested in the rabbit cornea. The virulence of a toxin deletion mutant was compared to its parent and the mutant after gene restoration (rescue strain). The toxin (23 kDa) had an N-terminal sequence matching the Newman superantigen-like protein SSL1. An SSL1 homodimer (46 kDa) had proteolytic activity as demonstrated by zymography and cleavage of a synthetic substrate, collagens, and cytokines (IL-17A, IFN-γ, and IL-8); the protease was susceptible to serine protease inhibitors. As compared to the parent and rescue strains, the ssl1 mutant had significantly reduced virulence, but not reduced bacterial growth, in vivo. The ocular isolates tested had the ssl1 gene, with allele type 2 being the predominant type. SSL1 is a protease with corneal virulence and activity on host defense and structural proteins.

11.
J Biol Chem ; 294(10): 3588-3602, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30622139

RESUMO

Staphylococcus aureus is a Gram-positive bacterium that can cause both superficial and deep-seated infections. Histones released by neutrophils kill bacteria by binding to the bacterial cell surface and causing membrane damage. We postulated that cell wall-anchored proteins protect S. aureus from the bactericidal effects of histones by binding to and sequestering histones away from the cell envelope. Here, we focused on S. aureus strain LAC and by using an array of biochemical assays, including surface plasmon resonance and ELISA, discovered that fibronectin-binding protein B (FnBPB) is the main histone receptor. FnBPB bound all types of histones, but histone H3 displayed the highest affinity and bactericidal activity and was therefore investigated further. H3 bound specifically to the A domain of recombinant FnBPB with a KD of 86 nm, ∼20-fold lower than that for fibrinogen. Binding apparently occurred by the same mechanism by which FnBPB binds to fibrinogen, because FnBPB variants defective in fibrinogen binding also did not bind H3. An FnBPB-deletion mutant of S. aureus LAC bound less H3 and was more susceptible to its bactericidal activity and to neutrophil extracellular traps, whereas an FnBPB-overexpressing mutant bound more H3 and was more resistant than the WT. FnBPB bound simultaneously to H3 and plasminogen, which after activation by tissue plasminogen activator cleaved the bound histone. We conclude that FnBPB provides a dual immune-evasion function that captures histones and prevents them from reaching the bacterial membrane and simultaneously binds plasminogen, thereby promoting its conversion to plasmin to destroy the bound histone.


Assuntos
Adesinas Bacterianas/metabolismo , Anti-Infecciosos/farmacologia , Histonas/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/metabolismo , Anti-Infecciosos/metabolismo , Parede Celular/efeitos dos fármacos , Parede Celular/metabolismo , Histonas/metabolismo , Concentração Osmolar , Plasminogênio/metabolismo , Ligação Proteica , Staphylococcus aureus/citologia
12.
Trends Microbiol ; 27(1): 26-38, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30031590

RESUMO

The use of ß-lactam antibiotics to treat infections caused by Staphylococcus aureus has been severely compromised by the acquisition by horizontal gene transfer of a gene that encodes the ß-lactam-insensitive penicillin-binding protein PBP2a. This allows methicillin-resistant S. aureus (MRSA) to proliferate in the presence of ß-lactam antibiotics. Paradoxically the dependence on PBP2a for the essential transpeptidase activity in cell wall peptidoglycan biosynthesis is the 'Achilles heel' of MRSA. Compounds that disrupt the divisome, wall teichoic acid, and functional membrane microdomains act synergistically with ß-lactams against MRSA. These include drugs such as statins that are widely used in human medicine. The antibiotics vancomycin and daptomycin are also synergistic with ß-lactams, and combinations have been employed to treat persistent MRSA infections. An additional benefit of exposing MRSA to ß-lactams could be a reduction in virulence mediated by interfering with the global regulator Agr. The mechanistic basis of synergy is discussed, and the possibility that ß-lactams can be resurrected to combat MRSA infections is explored.


Assuntos
Antibacterianos/uso terapêutico , Sinergismo Farmacológico , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , beta-Lactamas/uso terapêutico , Antibacterianos/farmacologia , Daptomicina/farmacologia , Daptomicina/uso terapêutico , Quimioterapia Combinada/métodos , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Vancomicina/farmacologia , Vancomicina/uso terapêutico , beta-Lactamas/farmacologia
14.
Trends Microbiol ; 26(6): 484-497, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29233606

RESUMO

Staphylococcus aureus is frequently isolated from the skin of atopic dermatitis (AD) patients during flares. The normal microbiota is disrupted and the diversity of the microorganisms on the skin is reduced. Many species that produce inhibitors of S. aureus growth decline. Strains from S. aureus clonal complex 1 are enriched among AD sufferers whereas the CC30 strains most frequently isolated from nasal carriers in the normal population are much rarer in AD. S. aureus expresses several molecules that contribute to the intensity of symptoms, including δ-toxin which stimulates mast cells, α-toxin which damages keratinocytes, phenol-soluble modulins which stimulate cytokine release by keratinocytes, protein A which triggers inflammatory responses from keratinocytes, superantigens which trigger B cell expansion and cytokine release, and proinflammatory lipoproteins. Proteases contribute to disruption of the epidermal barrier. S. aureus isolated from AD patients adheres to the deformed corneocytes from AD patients in a clumping factor B-dependent fashion. Novel targeted therapies for AD have recently been introduced to clinical practice with many more in development, including monoclonal antibodies that specifically target cytokines and their receptors, and a bacteriophage lysin that eliminates S. aureus from AD skin.


Assuntos
Dermatite Atópica/microbiologia , Pele/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus , Animais , Anticorpos Monoclonais/uso terapêutico , Toxinas Bacterianas/imunologia , Dermatite Atópica/imunologia , Dermatite Atópica/terapia , Proteínas Hemolisinas/imunologia , Humanos , Queratinócitos/imunologia , Camundongos , Pele/patologia
15.
mBio ; 8(5)2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28874469

RESUMO

Invasive bacterial pathogens can capture host plasminogen (Plg) and allow it to form plasmin. This process is of medical importance as surface-bound plasmin promotes bacterial spread by cleaving tissue components and favors immune evasion by degrading opsonins. In Staphylococcus aureus, Plg binding is in part mediated by cell surface fibronectin-binding proteins (FnBPs), but the underlying molecular mechanism is not known. Here, we use single-cell and single-molecule techniques to demonstrate that FnBPs capture Plg by a sophisticated activation mechanism involving fibrinogen (Fg), another ligand found in the blood. We show that while FnBPs bind to Plg through weak (∼200-pN) molecular bonds, direct interaction of the adhesins with Fg through the high-affinity dock, lock, and latch mechanism dramatically increases the strength of the FnBP-Plg bond (up to ∼2,000 pN). Our results point to a new model in which the binding of Fg triggers major conformational changes in the FnBP protein, resulting in the buried Plg-binding domains being projected and exposed away from the cell surface, thereby promoting strong interactions with Plg. This study demonstrated a previously unidentified role for a ligand-binding interaction by a staphylococcal cell surface protein, i.e., changing the protein orientation to activate a cryptic biological function.IMPORTANCEStaphylococcus aureus captures human plasminogen (Plg) via cell wall fibronectin-binding proteins (FnBPs), but the underlying molecular mechanism is not known. Here we show that the forces involved in the interaction between Plg and FnBPs on the S. aureus surface are weak. However, we discovered that binding of fibrinogen to FnBPs dramatically strengthens the FnBP-Plg bond, therefore revealing an unanticipated role for Fg in the capture of Plg by S. aureus These experiments favor a model where Fg-induced conformational changes in FnBPs promote their interaction with Plg. This work uncovers a previously undescribed activation mechanism for a staphylococcal surface protein, whereby ligand-binding elicits a cryptic biological function.


Assuntos
Adesinas Bacterianas/metabolismo , Fibrinogênio/metabolismo , Interações Hospedeiro-Patógeno , Plasminogênio/metabolismo , Staphylococcus aureus/metabolismo , Adesinas Bacterianas/química , Adesinas Bacterianas/genética , Aderência Bacteriana , Humanos , Ligantes , Proteínas de Membrana/metabolismo , Ligação Proteica , Conformação Proteica , Staphylococcus aureus/patogenicidade
16.
Food Funct ; 8(6): 2231-2242, 2017 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-28608892

RESUMO

The development of emulsion-based products through optimisation of ingredients, reduction in energy-input during manufacture, while fulfilling healthy attributes, are major objectives within the food industry. Instant emulsions can meet these features, but comprehensive studies are necessary to investigate the effect of the initial formulation on the final microstructure and, in turn, on the in vitro lipolysis, comprising the double aim of this work. The instant emulsion is formed within 1.5-3 min after pouring the aqueous phase into the oil phase which contains a mixture of emulsifier (Tween 20), swelling particles (Sephadex) and thickeners (hydroxypropylmethylcellulose, HPMC, and guar gum, GG) under mild shearing (180 rpm). The creation of oil-in-water emulsions is monitored in situ by viscosity analysis, the final microstructure visualised by microscopy and the release of free fatty acids under simulated intestinal conditions quantified by titration. Increasing the concentration and molecular weight (Mw) of GG leads to smaller emulsion droplets due to increased bulk viscosity upon shearing. This droplet size reduction is magnified when increasing the Mw of HPMC or swelling capacity of viscosifying particles. In addition, in the absence of the emulsifier Tween 20, the sole use of high-Mw HPMC is effective in emulsification due to combined increased bulk viscosity and interfacial activity. Hence, optimisation of the ingredient choice and usage level is possible when designing microstructures. Finally, emulsions with larger droplet size (>20 µm) display a slower rate and lower extent of lipolysis, while finer emulsions (droplet size ≤20 µm) exhibit maximum rate and extent profiles. This correlates with the extent of emulsion destabilisation observed under intestinal conditions.


Assuntos
Emulsões/química , Polissacarídeos/química , Emulsificantes/química , Galactanos/química , Lipólise , Mananas/química , Peso Molecular , Tamanho da Partícula , Gomas Vegetais/química , Polissorbatos/química
17.
Epigenomics ; 9(6): 823-832, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28523967

RESUMO

AIM: Validation of sequencing-based DNA methylation data is an important step for meaningful translation of findings. However, there has been limited assessment of different platforms to validate methylation data from next generation sequencing. METHODS: We performed a comparative methylation analysis between the genome-wide platform of reduced representation bisulfite sequencing with a targeted, Sequenom EpiTyper platform (four genes were analyzed in 15 cell lines covering 52 CpG sites). RESULTS: We show that the accuracy of validation substantially improves if results from multiple and adjacent CpG sites are combined rather than at single CpG sites. We demonstrate increased read number improves accuracy of reduced representation bisulfite sequencing results. Further, by using series of replicates, we document variation in samples analyzed by Sequenom EpiTyper, indicating the importance of including replicates to increase precision. CONCLUSION: The results reveal potential sources of bias and provide a guideline for refining study design for DNA methylation analysis.


Assuntos
Metilação de DNA , Sequenciamento Completo do Genoma/métodos , Linhagem Celular , Linhagem Celular Tumoral , Ilhas de CpG , Humanos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Sequenciamento Completo do Genoma/normas
18.
Trends Microbiol ; 25(7): 512-514, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28495024

RESUMO

Live-cell nanoscopy has contributed significantly to assessing the inhibition of adhesion of uropathogenic Escherichia coli and Staphylococcus aureus by glycoconjugates and monoclonal antibodies, respectively, and of S. aureus surface attachment and cell-cell association by a synthetic peptide. This new technology shows promise for the development of antiadhesion therapies against bacterial pathogens.


Assuntos
Aderência Bacteriana/efeitos dos fármacos , Nanotecnologia/métodos , Infecções Estafilocócicas/terapia , Biofilmes/crescimento & desenvolvimento , Escherichia coli/efeitos dos fármacos , Escherichia coli/fisiologia , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/terapia , Peptídeos/farmacologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia
19.
Infect Immun ; 85(6)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28373353

RESUMO

Staphylococcus aureus skin infection is a frequent and recurrent problem in children with the common inflammatory skin disease atopic dermatitis (AD). S. aureus colonizes the skin of the majority of children with AD and exacerbates the disease. The first step during colonization and infection is bacterial adhesion to the cornified envelope of corneocytes in the outer layer, the stratum corneum. Corneocytes from AD skin are structurally different from corneocytes from normal healthy skin. The objective of this study was to identify bacterial proteins that promote the adherence of S. aureus to AD corneocytes. S. aureus strains from clonal complexes 1 and 8 were more frequently isolated from infected AD skin than from the nasal cavity of healthy children. AD strains had increased ClfB ligand binding activity compared to normal nasal carriage strains. Adherence of single S. aureus bacteria to corneocytes from AD patients ex vivo was studied using atomic force microscopy. Bacteria expressing ClfB recognized ligands distributed over the entire corneocyte surface. The ability of an isogenic ClfB-deficient mutant to adhere to AD corneocytes compared to that of its parent clonal complex 1 clinical strain was greatly reduced. ClfB from clonal complex 1 strains had a slightly higher binding affinity for its ligand than ClfB from strains from other clonal complexes. Our results provide new insights into the first step in the establishment of S. aureus colonization in AD patients. ClfB is a key adhesion molecule for the interaction of S. aureus with AD corneocytes and represents a target for intervention.


Assuntos
Adesinas Bacterianas/metabolismo , Dermatite Atópica/microbiologia , Células Epiteliais/microbiologia , Infecções Cutâneas Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Adesinas Bacterianas/genética , Aderência Bacteriana , Pré-Escolar , Feminino , Proteínas Filagrinas , Humanos , Masculino , Cavidade Nasal/microbiologia , Deleção de Sequência , Pele/citologia , Pele/microbiologia , Staphylococcus aureus/genética
20.
FEMS Microbiol Rev ; 41(3): 430-449, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28419231

RESUMO

The major targets for antibiotics in staphylococci are (i) the cell envelope, (ii) the ribosome and (iii) nucleic acids. Several novel targets emerged from recent targeted drug discovery programmes including the ClpP protease and FtsZ from the cell division machinery. Resistance can either develop by horizontal transfer of resistance determinants encoded by mobile genetic elements viz plasmids, transposons and the staphylococcal cassette chromosome or by mutations in chromosomal genes. Horizontally acquired resistance can occur by one of the following mechanisms: (i) enzymatic drug modification and inactivation, (ii) enzymatic modification of the drug binding site, (iii) drug efflux, (iv) bypass mechanisms involving acquisition of a novel drug-resistant target, (v) displacement of the drug to protect the target. Acquisition of resistance by mutation can result from (i) alteration of the drug target that prevents the inhibitor from binding, (ii) derepression of chromosomally encoded multidrug resistance efflux pumps and (iii) multiple stepwise mutations that alter the structure and composition of the cell wall and/or membrane to reduce drug access to its target. This review focuses on development of resistance to currently used antibiotics and examines future prospects for new antibiotics and informed use of drug combinations.


Assuntos
Antibacterianos/farmacologia , Membrana Celular/efeitos dos fármacos , Parede Celular/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla/genética , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética , Transferência Genética Horizontal , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...