Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Biochemistry ; 56(1): 260-270, 2017 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-27931094

RESUMO

Infection by the opportunistic pathogen Pseudomonas aeruginosa (PA) is accompanied by the secretion of virulence factors such as the secondary metabolite rhamnolipid (RL) as well as an array of bacterial enzymes, including the peptidase elastase. The human immune system tries to counter this via defensive proteins such as lysozyme (HLZ). HLZ targets the bacterial cell wall but may also have other antimicrobial activities. The enzyme contains four disulfide bonds and shows high thermodynamic stability and resistance to proteolytic attack. Here we show that RL promotes HLZ degradation by several unrelated peptidases, including the PA elastase and human peptidases. This occurs although RL does not by itself denature HLZ. Nevertheless, RL binds in a sufficiently high stoichiometry (8:1 RL:HLZ) to neutralize the highly cationic surface of HLZ. The initial cleavage sites agree well with the domain boundaries of HLZ. Thus, binding of RL to native HLZ may be sufficient to allow proteolytic attack at slightly exposed sites on the protein, leading to subsequent degradation. Furthermore, biofilms of RL-producing strains of PA are protected better against high concentrations of HLZ than RL-free PA strains are. We conclude that pathogen-produced RL may weaken host defenses by facilitating degradation of key host proteins.


Assuntos
Proteínas de Bactérias/metabolismo , Glicolipídeos/metabolismo , Muramidase/metabolismo , Peptídeo Hidrolases/metabolismo , Pseudomonas aeruginosa/metabolismo , Ânions/metabolismo , Proteínas de Bactérias/química , Biofilmes , Dicroísmo Circular , Eletroforese em Gel de Poliacrilamida , Glicolipídeos/química , Humanos , Espectrometria de Massas/métodos , Modelos Moleculares , Muramidase/química , Elastase Pancreática/metabolismo , Peptídeo Hidrolases/química , Ligação Proteica , Domínios Proteicos , Proteólise , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/fisiologia , Eletricidade Estática , Fatores de Virulência/metabolismo
2.
J Biol Chem ; 291(51): 26540-26553, 2016 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-27784787

RESUMO

Epigallocatechin-3-gallate (EGCG) is the major polyphenol in green tea. It has antimicrobial properties and disrupts the ordered structure of amyloid fibrils involved in human disease. The antimicrobial effect of EGCG against the opportunistic pathogen Pseudomonas aeruginosa has been shown to involve disruption of quorum sensing (QS). Functional amyloid fibrils in P. aeruginosa (Fap) are able to bind and retain quorum-sensing molecules, suggesting that EGCG interferes with QS through structural remodeling of amyloid fibrils. Here we show that EGCG inhibits the ability of Fap to form fibrils; instead, EGCG stabilizes protein oligomers. Existing fibrils are remodeled by EGCG into non-amyloid aggregates. This fibril remodeling increases the binding of pyocyanin, demonstrating a mechanism by which EGCG can affect the QS function of functional amyloid. EGCG reduced the amyloid-specific fluorescent thioflavin T signal in P. aeruginosa biofilms at concentrations known to exert an antimicrobial effect. Nanoindentation studies showed that EGCG reduced the stiffness of biofilm containing Fap fibrils but not in biofilm with little Fap. In a combination treatment with EGCG and tobramycin, EGCG had a moderate effect on the minimum bactericidal eradication concentration against wild-type P. aeruginosa biofilms, whereas EGCG had a more pronounced effect when Fap was overexpressed. Our results provide a direct molecular explanation for the ability of EGCG to disrupt P. aeruginosa QS and modify its biofilm and strengthens the case for EGCG as a candidate in multidrug treatment of persistent biofilm infections.


Assuntos
Amiloide/biossíntese , Proteínas de Bactérias/biossíntese , Biofilmes/efeitos dos fármacos , Catequina/análogos & derivados , Farmacorresistência Bacteriana/efeitos dos fármacos , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Pseudomonas aeruginosa/fisiologia , Tobramicina/farmacologia , Benzotiazóis , Biofilmes/crescimento & desenvolvimento , Catequina/farmacologia , Humanos , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/metabolismo , Tiazóis/farmacologia
3.
J Biol Chem ; 290(33): 20590-600, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26109065

RESUMO

Archaea are renowned for their ability to thrive in extreme environments, although they can be found in virtually all habitats. Their adaptive success is linked to their unique cell envelopes that are extremely resistant to chemical and thermal denaturation and that resist proteolysis by common proteases. Here we employ amyloid-specific conformation antibodies and biophysical techniques to show that the extracellular cell wall sheaths encasing the methanogenic archaea Methanosaeta thermophila PT are functional amyloids. Depolymerization of sheaths and subsequent MS/MS analyses revealed that the sheaths are composed of a single major sheath protein (MspA). The amyloidogenic nature of MspA was confirmed by in vitro amyloid formation of recombinant MspA under a wide range of environmental conditions. This is the first report of a functional amyloid from the archaeal domain of life. The amyloid nature explains the extreme resistance of the sheath, the elastic properties that allow diffusible substrates to penetrate through expandable hoop boundaries, and how the sheaths are able to split and elongate outside the cell. The archaeal sheath amyloids do not share homology with any of the currently known functional amyloids and clearly represent a new function of the amyloid protein fold.


Assuntos
Amiloide/fisiologia , Methanosarcinales/fisiologia , Amiloide/biossíntese , Methanosarcinales/metabolismo , Microscopia Eletrônica de Transmissão , Espectrometria de Massas em Tandem
4.
Biochim Biophys Acta ; 1854(8): 882-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25796141

RESUMO

The medium-length fungal peptaibol SPF-5506-A(4) has been shown to inhibit formation of the Aß peptide involved in Alzheimer''s disease. As Aß is a cleavage-product from the membrane-bound APP protein, we hypothesized that SPF-5506-A(4)'s activity might be linked to membrane interactions in general. Here we describe the synthesis, structure and membrane interactions of SPF-5506-A4. The challenging synthesis was carried out on solid phase and a detailed conformational analysis in solution revealed a ß-bend ribbon spiral core structure with flexible termini. Investigations of its membrane activity revealed low hemolytic activity, limited inhibition of both Gram-positive and Gram-negative cell growth and a preference for an overall negatively charged membrane surface mimicking the bacterial cell surface. SPF-5506-A(4) is the first peptaibol to be shown to facilitate leakage of large (4.6 nm diameter) fluorescence-labeled dextran from vesicles while leaving the vesicles intact. We conclude that SPF-5506-A(4) follows the toroidal pore model in its mode of action.


Assuntos
Permeabilidade da Membrana Celular , Membrana Eritrocítica/química , Modelos Químicos , Oligopeptídeos/química , Escherichia coli/química , Bactérias Gram-Positivas/química , Hemólise , Humanos , Estrutura Secundária de Proteína
5.
Chemistry ; 22(24): 8358-67, 2016 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-27144376

RESUMO

Incorporation of silicon-containing amino acids in peptides is known to endow the peptide with desirable properties such as improved proteolytic stability and increased lipophilicity. In the presented study, we demonstrate that incorporation of ß-silicon-ß3-amino acids into the antimicrobial peptide alamethicin provides the peptide with improved membrane permeabilizing properties. A robust synthetic procedure for the construction of ß-silicon-ß3-amino acids was developed and the amino acid analogues were incorporated into alamethicin at different positions of the hydrophobic face of the amphipathic helix by using SPPS. The incorporation was shown to provide up to 20-fold increase in calcein release as compared with wild-type alamethicin.


Assuntos
Alameticina/análogos & derivados , Aminoácidos/química , Anti-Infecciosos/química , Peptídeos Catiônicos Antimicrobianos/química , Silício/química , Alameticina/síntese química , Alameticina/farmacologia , Anti-Infecciosos/síntese química , Anti-Infecciosos/farmacologia , Peptídeos Catiônicos Antimicrobianos/síntese química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Dicroísmo Circular , Lipossomos/química , Lipossomos/metabolismo , Estrutura Secundária de Proteína , Técnicas de Síntese em Fase Sólida
6.
J Biol Chem ; 289(27): 19098-109, 2014 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-24860098

RESUMO

ATP is as an extracellular signaling molecule able to amplify the cell lysis inflicted by certain bacterial toxins including the two RTX toxins α-hemolysin (HlyA) from Escherichia coli and leukotoxin A (LtxA) from Aggregatibacter actinomycetemcomitans. Inhibition of P2X receptors completely blocks the RTX toxin-induced hemolysis over a larger concentration range. It is, however, at present not known how the ATP that provides the amplification is released from the attacked cells. Here we show that both HlyA and LtxA trigger acute release of ATP from human erythrocytes that preceded and were not caused by cell lysis. This early ATP release did not occur via previously described ATP-release pathways in the erythrocyte. Both HlyA and LtxA were capable of triggering ATP release in the presence of the pannexin 1 blockers carbenoxolone and probenecid, and the HlyA-induced ATP release was found to be similar in erythrocytes from pannexin 1 wild type and knock-out mice. Moreover, the voltage-dependent anion channel antagonist TRO19622 had no effect on ATP release by either of the toxins. Finally, we showed that both HlyA and LtxA were able to release ATP from ATP-loaded lipid (1-palmitoyl-2-oleoyl-phosphatidylcholine) vesicles devoid of any erythrocyte channels or transporters. Again we were able to show that this happened in a non-lytic fashion, using calcein-containing vesicles as controls. These data show that both toxins incorporate into lipid vesicles and allow ATP to be released. We suggest that both toxins cause acute ATP release by letting ATP pass the toxin pores in both human erythrocytes and artificial membranes.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/farmacologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/farmacologia , Proteínas Hemolisinas/química , Proteínas Hemolisinas/farmacologia , Aggregatibacter actinomycetemcomitans , Animais , Conexinas/deficiência , Conexinas/genética , Eritrócitos/citologia , Técnicas de Inativação de Genes , Hemoglobinas/metabolismo , Hemólise/efeitos dos fármacos , Humanos , Membranas Artificiais , Camundongos , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Fosfatidilcolinas/metabolismo , Porosidade
7.
J Biol Chem ; 289(31): 21299-310, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24907278

RESUMO

Oligomeric species of various proteins are linked to the pathogenesis of different neurodegenerative disorders. Consequently, there is intense focus on the discovery of novel inhibitors, e.g. small molecules and antibodies, to inhibit the formation and block the toxicity of oligomers. In Parkinson disease, the protein α-synuclein (αSN) forms cytotoxic oligomers. The flavonoid epigallocatechin gallate (EGCG) has previously been shown to redirect the aggregation of αSN monomers and remodel αSN amyloid fibrils into disordered oligomers. Here, we dissect EGCG's mechanism of action. EGCG inhibits the ability of preformed oligomers to permeabilize vesicles and induce cytotoxicity in a rat brain cell line. However, EGCG does not affect oligomer size distribution or secondary structure. Rather, EGCG immobilizes the C-terminal region and moderately reduces the degree of binding of oligomers to membranes. We interpret our data to mean that the oligomer acts by destabilizing the membrane rather than by direct pore formation. This suggests that reduction (but not complete abolition) of the membrane affinity of the oligomer is sufficient to prevent cytotoxicity.


Assuntos
Biopolímeros/antagonistas & inibidores , Catequina/análogos & derivados , alfa-Sinucleína/antagonistas & inibidores , Biopolímeros/metabolismo , Biopolímeros/toxicidade , Varredura Diferencial de Calorimetria , Catequina/farmacologia , Permeabilidade da Membrana Celular , Dicroísmo Circular , Técnicas In Vitro , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Ressonância Magnética Nuclear Biomolecular , Estrutura Secundária de Proteína , alfa-Sinucleína/metabolismo , alfa-Sinucleína/toxicidade
8.
J Membr Biol ; 248(3): 487-96, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25801603

RESUMO

The biological activity of antimicrobial peptides is believed to be closely linked to their ability to perturb bacterial membranes. This makes it important to understand the basis of their membrane-binding properties. Here, we present a biophysical analysis of the interactions of the antimicrobial peptide Novicidin (Nc) with ether- and ester-linked C14 phospholipid vesicles below and above the lipid phase transition temperature (t p). These interactions are strongly dependent on whether the lipids contain ether or ester linkages. Nc is in random coil state in solution but undergoes a large increase in α-helicity in ether vesicles, and to a much smaller extent in ester vesicles, around the t p. This structure is lost at higher temperatures. Steady-state fluorescence and stopped-flow kinetics using fluorophore-labeled Nc reveal that Nc binds more strongly to ether vesicles than to ester vesicles below the t p, while there is no significant difference above the t p. This may reflect ether lipid interdigitation in the gel phase. Isothermal titration calorimetry reveals that partitioning of Nc into both lipids is exothermic and thus enthalpy driven. The higher enthalpy associated with binding to ether lipid may be linked to Nc's higher propensity to form α-helical structure in this lipid. The large effect of the ether-ester interchange reveals that membrane-AMP interactions can be strongly modulated by charge-neutral head group changes.


Assuntos
Peptídeos Catiônicos Antimicrobianos/química , Bicamadas Lipídicas/química , Dimiristoilfosfatidilcolina/química , Cinética , Ligação Proteica , Termodinâmica , Lipossomas Unilamelares/química
9.
Biochim Biophys Acta ; 1834(6): 996-1002, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23562965

RESUMO

We have investigated the interactions between the antimicrobial peptide Novicidin (Nc) and vesicles containing the phospholipid DOPC, with various amounts of DOPG and cholesterol using circular dichroism spectroscopy, calcein release, equilibrium dialysis and isothermal titration calorimetry. Nc adopts a random coil structure in the absence of lipids and in the presence of vesicles containing 100% DOPC. Lipids with 25-40% DOPG induce the highest level of helicity in Nc; higher DOPG levels lead to lower helicity levels and an altered tertiary arrangement of the peptide. However, the ability of Nc to permeabilize vesicles correlates not with helicity but rather with its overall membrane affinity, which is enthalpically favorable but opposed by entropy. Permeabilization declines with increasing mole percentage PG. Changes in helicity correlate with changes in enthalpy, reflecting the enthalpy of helix formation, but not with affinity. There is also a large favorable enthalpic interaction between Nc and lipids in the absence of negative charge and structural changes. Cholesterol slightly reduces membrane permeabilization but has little effect on Nc affinity and secondary structure, and probably protects the membrane by inducing the liquid ordered state. We conclude that helicity is not a prerequisite for activity, and charge-charge interactions are not the only major driving force for AMP interactions with membranes. Our data are compatible with a model in which a superficial binding mode with a large membrane surface binding area per peptide is more efficient than a more intimate embedding within the membrane environment.


Assuntos
Peptídeos Catiônicos Antimicrobianos/química , Colesterol/química , Bicamadas Lipídicas/química , Fosfolipídeos/química , Dicroísmo Circular/métodos , Permeabilidade , Fosfatidilcolinas/química , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Termodinâmica
10.
Biochim Biophys Acta ; 1834(3): 677-87, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23313095

RESUMO

Ribosomal protein S6 fibrillates readily at slightly elevated temperatures and acidic pH. We find that S6 fibrillation is retarded rather than favored when the protein concentration is increased above a threshold concentration of around 3.5mg/mL. We name this threshold concentration C(FR), the concentration at which fibrillation is retarded. Our data are consistent with a model in which this inhibition is due to the formation of an off-pathway oligomeric species with native-like secondary structure. The oligomeric species dominates at high protein concentrations but exists in dynamic equilibrium with the monomer so that seeding with fibrils can overrule oligomer formation and favors fibrillation under C(FR) conditions. Thus, fibrillation competes with formation of off-pathway oligomers, probably due to a monomeric conversion step that is required to commit the protein to the fibrillation pathway. The S6 oligomer is resistant to pepsin digestion. We also report that S6 forms different types of fibrils dependent on protein concentration. Our observations highlight the multitude of conformational states available to proteins under destabilizing conditions.


Assuntos
Conformação Proteica , Multimerização Proteica , Proteínas Quinases S6 Ribossômicas/química , Proteínas Quinases S6 Ribossômicas/metabolismo , Dicroísmo Circular , Eletroforese em Gel de Poliacrilamida , Concentração de Íons de Hidrogênio , Cinética , Microscopia de Força Atômica , Modelos Químicos , Modelos Moleculares , Mutação , Proteólise , Proteínas Quinases S6 Ribossômicas/genética , Espectroscopia de Infravermelho com Transformada de Fourier , Temperatura , Fatores de Tempo
11.
Appl Environ Microbiol ; 80(24): 7758-70, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25304506

RESUMO

Epsilon-poly-l-lysine (ε-PL) is a natural antimicrobial cationic peptide which is generally regarded as safe (GRAS) as a food preservative. Although its antimicrobial activity is well documented, its mechanism of action is only vaguely described. The aim of this study was to clarify ε-PL's mechanism of action using Escherichia coli and Listeria innocua as model organisms. We examined ε-PL's effect on cell morphology and membrane integrity and used an array of E. coli deletion mutants to study how specific outer membrane components affected the action of ε-PL. We furthermore studied its interaction with lipid bilayers using membrane models. In vitro cell studies indicated that divalent cations and the heptose I and II phosphate groups in the lipopolysaccharide layer of E. coli are critical for ε-PL's binding efficiency. ε-PL removed the lipopolysaccharide layer and affected cell morphology of E. coli, while L. innocua underwent minor morphological changes. Propidium iodide staining showed that ε-PL permeabilized the cytoplasmic membrane in both species, indicating the membrane as the site of attack. We compared the interaction with neutral or negatively charged membrane systems and showed that the interaction with ε-PL relied on negative charges on the membrane. Suspended membrane vesicles were disrupted by ε-PL, and a detergent-like disruption of E. coli membrane was confirmed by atomic force microscopy imaging of supported lipid bilayers. We hypothesize that ε-PL destabilizes membranes in a carpet-like mechanism by interacting with negatively charged phospholipid head groups, which displace divalent cations and enforce a negative curvature folding on membranes that leads to formation of vesicles/micelles.


Assuntos
Antibacterianos/metabolismo , Antibacterianos/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Listeria/metabolismo , Polilisina/metabolismo , Transporte Biológico , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Escherichia coli/química , Listeria/química , Listeria/efeitos dos fármacos , Polilisina/farmacologia
12.
J Biol Chem ; 287(50): 42361-72, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23093408

RESUMO

Antimicrobial peptides are a new class of antibiotics that are promising for pharmaceutical applications because they have retained efficacy throughout evolution. One class of antimicrobial peptides are the defensins, which have been found in different species. Here we describe a new fungal defensin, eurocin. Eurocin acts against a range of Gram-positive human pathogens but not against Gram-negative bacteria. Eurocin consists of 42 amino acids, forming a cysteine-stabilized α/ß-fold. The thermal denaturation data point shows the disulfide bridges being responsible for the stability of the fold. Eurocin does not form pores in cell membranes at physiologically relevant concentrations; it does, however, lead to limited leakage of a fluorophore from small unilamellar vesicles. Eurocin interacts with detergent micelles, and it inhibits the synthesis of cell walls by binding equimolarly to the cell wall precursor lipid II.


Assuntos
Anti-Infecciosos/química , Defensinas/química , Eurotium/química , Proteínas Fúngicas/química , Lipídeos de Membrana/química , Dobramento de Proteína , Uridina Difosfato Ácido N-Acetilmurâmico/análogos & derivados , Anti-Infecciosos/farmacologia , Defensinas/farmacologia , Proteínas Fúngicas/farmacologia , Bactérias Gram-Positivas/crescimento & desenvolvimento , Bactérias Gram-Positivas/metabolismo , Infecções por Bactérias Gram-Positivas/metabolismo , Humanos , Lipídeos de Membrana/metabolismo , Micelas , Estrutura Secundária de Proteína , Uridina Difosfato Ácido N-Acetilmurâmico/química , Uridina Difosfato Ácido N-Acetilmurâmico/metabolismo
13.
Biophys J ; 98(4): 576-85, 2010 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-20159154

RESUMO

Pardaxin is a 33-amino-acid neurotoxin from the Red Sea Moses sole Pardachirus marmoratus, whose mode of action shows remarkable sensitivity to lipid chain length and charge, although the effect of pH is unclear. Here we combine optical spectroscopy and dye release experiments with laser scanning confocal microscopy and natural abundance (13)C solid-state nuclear magnetic resonance to provide a more complete picture of how pardaxin interacts with lipids. The kinetics and efficiency of release of entrapped calcein is highly sensitive to pH. In vesicles containing zwitterionic lipids (PC), release occurs most rapidly at low pH, whereas in vesicles containing 20% anionic lipid (PG), release occurs most rapidly at high pH. Pardaxin forms stable or transient pores in PC vesicles that allow release of contents without loss of vesicle integrity, whereas the inclusion of PG promotes total vesicle collapse. In agreement with this, solid-state nuclear magnetic resonance reveals that pardaxin takes up a trans-membrane orientation in 14-O-PC/6-O-PC bicelles, whereas the inclusion of 14-0-PG restricts it to contacts with lipid headgroups, promoting membrane lysis. Pore formation in zwitterionic vesicles is more efficient than lysis of anionic vesicles, suggesting that electrostatic interactions may trap pardaxin in several suboptimal interconverting conformations on the membrane surface.


Assuntos
Venenos de Peixe/metabolismo , Lipossomas Unilamelares/química , Lipossomas Unilamelares/metabolismo , Sequência de Aminoácidos , Venenos de Peixe/química , Fluoresceínas/metabolismo , Concentração de Íons de Hidrogênio , Cinética , Metabolismo dos Lipídeos , Lipídeos/química , Espectroscopia de Ressonância Magnética , Microscopia Confocal , Dados de Sequência Molecular , Permeabilidade , Porosidade , Conformação Proteica , Prótons
14.
Protein Sci ; 28(3): 633-642, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30592554

RESUMO

Functional amyloid (FA) is widespread in bacteria and serves multiple purposes such as strengthening of biofilm and contact with eukaryotic hosts. Unlike pathological amyloid, FA has been subjected to evolutionary optimization which is likely to be reflected in the aggregation mechanism. FA from different bacteria, including Escherichia coli (CsgA) and Pseudomonas (FapC), contains a number of imperfect repeats which may be key to efficient aggregation. Here we report on the aggregative behavior of FapC constructs which represent all single, double, and triple deletions of the protein's three imperfect repeats. Analysis of the fibrillation kinetics by the program Amylofit reveals that the removal of these repeats increases the tendency of the growing fibrils to fragment and also generally increases aggregation half-times. Remarkably, even the mutant lacking all three repeats was able to fibrillate, although fibrillation was much more irregular and led to significantly altered and destabilized fibrils. We conclude that imperfect repeats can promote fibrillation efficiency thanks to their modular design, though the context of the imperfect repeats also plays a significant role.


Assuntos
Amiloide/química , Proteínas Amiloidogênicas/química , Pseudomonas/química , Sequência de Aminoácidos , Amiloide/ultraestrutura , Humanos , Cinética , Modelos Moleculares , Agregados Proteicos , Infecções por Pseudomonas/microbiologia
15.
FEBS Lett ; 592(9): 1484-1496, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29572816

RESUMO

The Parkinson's disease-associated protein α-synuclein (αSN) is natively unfolded but its structure can be modulated by membranes and surfactants. The opportunistic pathogen Pseudomonas aeruginosa (PA) produces and secretes the biosurfactant rhamnolipid (RL) which modulates bacterial biofilm. Here, we show that monomeric RL enhances the ability of αSN to permeabilize membranes, while micellar RL rapidly induces protein ß-sheet structure with a worm-like fibrillary appearance, which cannot seed RL-free fibrillation but transforms into linear fibrils faster than αSN fibrillating on its own. Exposure to αSN reduces the degree of biofilm formation by PA unless RL is present. Our data suggest that RL interactions with αSN may affect both αSN aggregation and cell toxicity, potentially implicating microbiomic metabolites in the origin and propagation of Parkinson's disease.


Assuntos
Biofilmes/crescimento & desenvolvimento , Glicolipídeos/farmacologia , Agregados Proteicos/efeitos dos fármacos , Pseudomonas aeruginosa/fisiologia , alfa-Sinucleína/química , Biofilmes/efeitos dos fármacos , Humanos , Cinética , Micelas , Permeabilidade/efeitos dos fármacos , Conformação Proteica em Folha beta/efeitos dos fármacos , Pseudomonas aeruginosa/efeitos dos fármacos , alfa-Sinucleína/metabolismo , alfa-Sinucleína/farmacologia
16.
Front Microbiol ; 7: 1711, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27877155

RESUMO

Biosurfactants are of growing interest as sustainable alternatives to fossil-fuel-derived chemical surfactants, particularly for the detergent industry. To realize this potential, it is necessary to understand how they affect proteins which they may encounter in their applications. However, knowledge of such interactions is limited. Here, we present a study of the interactions between the model protein apo-α-lactalbumin (apo-aLA) and the biosurfactant sophorolipid (SL) produced by the yeast Starmerella bombicola. SL occurs both as an acidic and a lactonic form; the lactonic form (lactSL) is sparingly soluble and has a lower critical micelle concentration (cmc) than the acidic form [non-acetylated acidic sophorolipid (acidSL)]. We show that acidSL affects apo-aLA in a similar way to the related glycolipid biosurfactant rhamnolipid (RL), with the important difference that RL is also active below the cmc in contrast to acidSL. Using isothermal titration calorimetry data, we show that acidSL has weak and saturable interactions with apo-aLA at low concentrations; due to the relatively low cmc of acidSL (which means that the monomer concentration is limited to ca. 0-1 mM SL), it is only possible to observe interactions with monomeric acidSL at high apo-aLA concentrations. However, the denaturation kinetics of apo-aLA in the presence of acidSL are consistent with a collaboration between monomeric and micellar surfactant species, similar to RL and non-ionic or zwitterionic surfactants. Inclusion of diacetylated lactonic sophorolipid (lactSL) as mixed micelles with acidSL lowers the cmc and this effectively reduces the rate of unfolding, emphasizing that SL like other biosurfactants is a gentle anionic surfactant. Our data highlight the potential of these biosurfactants for future use in the detergent and pharmaceutical industry.

17.
Front Microbiol ; 6: 1099, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26500638

RESUMO

The success of Pseudomonas species as opportunistic pathogens derives in great part from their ability to form stable biofilms that offer protection against chemical and mechanical attack. The extracellular matrix of biofilms contains numerous biomolecules, and it has recently been discovered that in Pseudomonas one of the components includes ß-sheet rich amyloid fibrils (functional amyloid) produced by the fap operon. However, the role of the functional amyloid within the biofilm has not yet been investigated in detail. Here we investigate how the fap-based amyloid produced by Pseudomonas affects biofilm hydrophobicity and mechanical properties. Using atomic force microscopy imaging and force spectroscopy, we show that the amyloid renders individual cells more resistant to drying and alters their interactions with hydrophobic probes. Importantly, amyloid makes Pseudomonas more hydrophobic and increases biofilm stiffness 20-fold. Deletion of any one of the individual members of in the fap operon (except the putative chaperone FapA) abolishes this ability to increase biofilm stiffness and correlates with the loss of amyloid. We conclude that amyloid makes major contributions to biofilm mechanical robustness.

18.
J Phys Chem B ; 116(26): 7652-9, 2012 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-22676384

RESUMO

Bacterial resistance to classical antibiotics is a serious medical problem, which continues to grow. Small antimicrobial peptides represent a potential solution and are increasingly being developed as novel therapeutic agents. Many of these peptides owe their antibacterial activity to the formation of trans-membrane ion-channels resulting in cell lysis. However, to further develop the field of peptide antibiotics, a thorough understanding of their mechanism of action is needed. Alamethicin belongs to a class of peptides called peptaibols and represents one of these antimicrobial peptides. To examine the dynamics of assembly and to facilitate a thorough structural evaluation of the alamethicin ion-channels, we have applied click chemistry for the synthesis of templated alamethicin multimers covalently attached to cyclodextrin-scaffolds. Using oriented circular dichroism, calcein release assays, and single-channel current measurements, the α-helices of the templated multimers were demonstrated to insert into lipid bilayers forming highly efficient and remarkably stable ion-channels.


Assuntos
Alameticina/química , Antibacterianos/química , Peptídeos Catiônicos Antimicrobianos/química , Permeabilidade da Membrana Celular/efeitos dos fármacos , Ciclodextrinas/química , Farmacorresistência Bacteriana , Alameticina/farmacologia , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Dicroísmo Circular , Ciclodextrinas/farmacologia , Canais Iônicos/química , Bicamadas Lipídicas/química , Técnicas de Patch-Clamp , Peptaibols/química , Estrutura Secundária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA