Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Langmuir ; 37(4): 1337-1352, 2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33478220

RESUMO

Previous studies have demonstrated the potential for non-steroidal anti-inflammatory drugs (NSAIDs), in particular aspirin, to be used as chemopreventives for colorectal cancer; however, a range of unwanted gastrointestinal side effects limit their effectiveness. Due to the role of bismuth in the treatment of gastrointestinal disorders, it is hypothesized that bismuth-coordinated NSAIDs (BiNSAIDs) could be used to combat the gastrointestinal side effects of NSAIDs while still maintaining their chemopreventive potential. To further understand the biological activity of these compounds, the present study examined four NSAIDs, namely, tolfenamic acid (tolfH), aspirin (aspH), indomethacin (indoH), and mefenamic acid (mefH) and their analogous homoleptic BiNSAIDs ([Bi(L)3]n), to determine how these compounds interact with biological membrane mimics composed of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) or a mixture of POPC and cholesterol. Electrical impedance spectroscopy studies revealed that each of the NSAIDs and BiNSAIDs influenced membrane conductance, suggesting that temporary pore formation may play a key role in the previously observed cytotoxicity of tolfH and Bi(tolf)3. Quartz crystal microbalance with dissipation monitoring showed that all the compounds were able to interact with membrane mimics composed of solely POPC or POPC/cholesterol. Finally, neutron reflectometry studies showed changes in membrane thickness and composition. The location of the compounds within the bilayer could not be determined with certainty; however, a complex interplay of interactions governs the location of small molecules, such as NSAIDs, within lipid membranes. The charged nature of the parent NSAIDs means that interactions with the polar headgroup region are most likely with larger hydrophobic sections, potentially leading to deeper penetration.


Assuntos
Bicamadas Lipídicas , Preparações Farmacêuticas , Anti-Inflamatórios não Esteroides/toxicidade , Bismuto/toxicidade , Concentração de Íons de Hidrogênio , Fosfatidilcolinas
2.
Langmuir ; 34(31): 9141-9152, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-29999320

RESUMO

The interfacial structures of a range of amphiphilic molecules are studied with both "soft" and "hard" hydrophobic substrates. Neutron reflection and quartz crystal microbalance with dissipation measurements highlight the differences between the adsorbed structures adopted by sodium dodecyl sulfate (SDS), cetyltrimethylammonium bromide (C16TAB), and the "AM1" surface active peptide. At the soft siloxane/water interface, small molecular surfactants form loosely packed layers, with the hydrophobic tails penetrating into the oily layer, and an area per surfactant molecule that is significantly less than previously reported for the air/water interface. Neutron reflection measurements, supported by quartz crystal microbalance studies, indicate that for C16TAB, approximately 30 ± 8% of the alkyl tail penetrates into the poly(dimethylsiloxane) (PDMS) layer, whereas 20 ± 5% of the alkyl tail of SDS is located in the PDMS. For the engineered peptide surfactant AM1 (21 residues), it was found that one face of the α helix penetrated into the PDMS film. In contrast, penetration of the surfactant tails was not observed against hard solidlike hydrophobic surfaces made from octadecyltrichlorosilane (OTS) for any of the molecular species studied. At the OTS/water interface, C16TAB and SDS were seen to adsorb as larger aggregates and not as monolayers. Amphiphilic adsorption (amount, structural conformation) at the PDMS/water interface is shown to be different from that at both the air/water interface and the hard OTS/water interface, illustrating that interfacial structures cannot be predicted by the surfactant packing parameter alone. The bound PDMS layer is shown to be a useful proxy for the oil/water interface in surface and stabilization studies, with hydrophobic components of the molecules able to penetrate into the oily PDMS.

3.
Soft Matter ; 13(43): 7953-7961, 2017 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-29038804

RESUMO

The interfacial properties of nanoscale materials have profound influence on biodistribution and stability as well as the effectiveness of sophisticated surface-encoded properties such as active targeting to cell surface receptors. Tailorable nanocarrier emulsions (TNEs) are a novel class of oil-in-water emulsions stabilised by molecularly-engineered biosurfactants that permit single-pot stepwise surface modification with related polypeptides that may be chemically conjugated or genetically fused to biofunctional moieties. We have probed the structure and function of poly(ethylene glycol) (PEG) used to decorate TNEs in this way. The molecular weight of PEG decorating TNEs has considerable impact on the ζ-potential of the emulsion particles, related to differential interfacial thickness of the PEG layer as determined by X-ray reflectometry. By co-modifying TNEs with an antibody fragment, we show that the molecular weight and density of PEG governs the competing parameters of accessibility of the targeting moiety and of shielding the interface from non-specific interactions with the environment. The fundamental understanding of the molecular details of the PEG layer that we present provides valuable insights into the structure-function relationship for soft nanomaterial interfaces. This work therefore paves the way for further rational design of TNEs and other nanocarriers that must interact with their environment in controlled and predictable ways.

4.
J Biol Chem ; 290(52): 31101-12, 2015 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-26542805

RESUMO

Perforin is an essential component in the cytotoxic lymphocyte-mediated cell death pathway. The traditional view holds that perforin monomers assemble into pores in the target cell membrane via a calcium-dependent process and facilitate translocation of cytotoxic proteases into the cytoplasm to induce apoptosis. Although many studies have examined the structure and role of perforin, the mechanics of pore assembly and granzyme delivery remain unclear. Here we have employed quartz crystal microbalance with dissipation monitoring (QCM-D) to investigate binding and assembly of perforin on lipid membranes, and show that perforin monomers bind to the membrane in a cooperative manner. We also found that cholesterol influences perforin binding and activity on intact cells and model membranes. Finally, contrary to current thinking, perforin efficiently binds membranes in the absence of calcium. When calcium is added to perforin already on the membrane, the QCM-D response changes significantly, indicating that perforin becomes membranolytic only after calcium binding.


Assuntos
Cálcio/química , Colesterol/química , Membranas Artificiais , Perforina/química , Técnicas de Microbalança de Cristal de Quartzo/métodos , Animais , Cálcio/metabolismo , Colesterol/metabolismo , Camundongos , Perforina/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
5.
Chem Sci ; 14(14): 3730-3741, 2023 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-37035708

RESUMO

The self-assembly of peptides into supramolecular structures has been linked to neurodegenerative diseases but has also been observed in functional roles. Peptides are physiologically exposed to crowded environments of biomacromolecules, and particularly cellular membrane lipids. Previous research has shown that membranes can both accelerate and inhibit peptide self-assembly. Here, we studied the impact of membrane models that mimic cellular oxidative stress and compared this to mammalian and bacterial membranes. Using molecular dynamics simulations and experiments, we propose a model that explains how changes in peptide-membrane binding, electrostatics, and peptide secondary structure stabilization determine the nature of peptide self-assembly. We explored the influence of zwitterionic (POPC), anionic (POPG) and oxidized (PazePC) phospholipids, as well as cholesterol, and mixtures thereof, on the self-assembly kinetics of the amyloid ß (1-40) peptide (Aß40), linked to Alzheimer's disease, and the amyloid-forming antimicrobial peptide uperin 3.5 (U3.5). We show that the presence of an oxidized lipid had similar effects on peptide self-assembly as the bacterial mimetic membrane. While Aß40 fibril formation was accelerated, U3.5 aggregation was inhibited by the same lipids at the same peptide-to-lipid ratio. We attribute these findings and peptide-specific effects to differences in peptide-membrane adsorption with U3.5 being more strongly bound to the membrane surface and stabilized in an α-helical conformation compared to Aß40. Different peptide-to-lipid ratios resulted in different effects. We found that electrostatic interactions are a primary driving force for peptide-membrane interaction, enabling us to propose a model for predicting how cellular changes might impact peptide self-assembly in vivo.

6.
Biochim Biophys Acta ; 1808(7): 1811-7, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21414289

RESUMO

The multifarious Tat peptide derived from the HIV-1 virus exhibits antimicrobial activity. In this article, we use Quartz Crystal Microbalance with Dissipation monitoring (QCM-D) to investigate the mechanisms of action of Tat (44-57) and Tat (49-57) on bacterial-mimetic 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC)/1,2-dimyristoyl-sn-glycero-3-phospho-rac-(1-glycerol) (sodium salt) (DMPG) membranes. The results reveal that both peptides disrupt DMPC/DMPG membranes via a surface-active (carpet-like) mechanism. The magnitude of this disruption is dependent on both membrane and peptide properties. Firstly, less disruption was observed on the more negatively charged membranes. Secondly, less disruption was observed for the longer and slightly more hydrophobic Tat (44-57) peptide. As a comparison, the behaviour of the two Tat peptides on mammalian-mimetic DMPC/cholesterol membranes was investigated. Consistent with the literature no membrane disruption was observed. These results suggest that both electrostatic and hydrophobic interactions, as well as peptide geometry, determine the antimicrobial activity of Tat. This should guide the development of more potent Tat antibiotics.


Assuntos
Permeabilidade da Membrana Celular , Produtos do Gene tat/metabolismo , Lipídeos de Membrana/metabolismo , Sequência de Aminoácidos , Animais , Colesterol/metabolismo , Produtos do Gene tat/química , Mamíferos , Dados de Sequência Molecular , Fosfolipídeos/metabolismo
7.
Eur Biophys J ; 40(4): 437-46, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21161523

RESUMO

The increasing prevalence of antibiotic-resistant bacteria is becoming a public health crisis. Antimicrobial peptides (AMPs) are a promising solution, because bacterial resistance is less likely. Quartz crystal microbalance with dissipation monitoring (QCM-D) is a versatile and valuable technique for investigation of these peptides. This article looks at the different approaches to the interpretation of QCM-D data, showing how to extract the maximum information from the data. Five AMPs of diverse charge, length and activity are used as case studies: caerin 1.1 wild-type, two caerin 1.1 mutants (Gly15Gly19-caerin 1.1 and Ala15Ala19-caerin 1.1), aurein 1.2 and oncocin. The interaction between the AMP and a 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) membrane is analysed inter alia using frequency-dissipation plots (∆f-∆D plots) to ascertain the mechanism of action of the AMP. The ∆f-∆D plot can then be used to provide a fingerprint for the AMP-membrane interaction. Building up a database of these fingerprints for all known AMPs will enable the relationship between AMP structure and membrane activity to be better understood, hopefully leading to the future development of antibiotics without bacterial resistance.


Assuntos
Anti-Infecciosos/análise , Peptídeos Catiônicos Antimicrobianos/análise , Técnicas de Microbalança de Cristal de Quartzo/métodos , Sequência de Aminoácidos , Anti-Infecciosos/química , Anti-Infecciosos/metabolismo , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/classificação , Peptídeos Catiônicos Antimicrobianos/metabolismo , Membrana Celular/química , Dimiristoilfosfatidilcolina/química , Dimiristoilfosfatidilcolina/metabolismo , Humanos , Dados de Sequência Molecular
8.
Front Cell Dev Biol ; 4: 39, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27243004

RESUMO

The Gram-negative bacterium Pseudomonas aeruginosa is a life-threatening nosocomial pathogen due to its generally low susceptibility toward antibiotics. Furthermore, many strains have acquired resistance mechanisms requiring new antimicrobials with novel mechanisms to enhance treatment options. Proline-rich antimicrobial peptides, such as the apidaecin analog Api137, are highly efficient against various Enterobacteriaceae infections in mice, but less active against P. aeruginosa in vitro. Here, we extended our recent work by optimizing lead peptides Api755 (gu-OIORPVYOPRPRPPHPRL-OH; gu = N,N,N',N'-tetramethylguanidino, O = L-ornithine) and Api760 (gu-OWORPVYOPRPRPPHPRL-OH) by incorporation of Ile-Orn- and Trp-Orn-motifs, respectively. Api795 (gu-O(IO)2RPVYOPRPRPPHPRL-OH) and Api794 (gu-O(WO)3RPVYOPRPRPPHPRL-OH) were highly active against P. aeruginosa with minimal inhibitory concentrations of 8-16 and 8-32 µg/mL against Escherichia coli and Klebsiella pneumoniae. Assessed using a quartz crystal microbalance, these peptides inserted into a membrane layer and the surface activity increased gradually from Api137, over Api795, to Api794. This mode of action was confirmed by transmission electron microscopy indicating some membrane damage only at the high peptide concentrations. Api794 and Api795 were highly stable against serum proteases (half-life times >5 h) and non-hemolytic to human erythrocytes at peptide concentrations of 0.6 g/L. At this concentration, Api795 reduced the cell viability of HeLa cells only slightly, whereas the IC50 of Api794 was 0.23 ± 0.09 g/L. Confocal fluorescence microscopy revealed no colocalization of 5(6)-carboxyfluorescein-labeled Api794 or Api795 with the mitochondria, excluding interactions with the mitochondrial membrane. Interestingly, Api795 was localized in endosomes, whereas Api794 was present in endosomes and the cytosol. This was verified using flow cytometry showing a 50% higher uptake of Api794 in HeLa cells compared with Api795. The uptake was reduced for both peptides by 50 and 80%, respectively, after inhibiting endocytotic uptake with dynasore. In summary, Api794 and Api795 were highly active against P. aeruginosa in vitro. Both peptides passed across the bacterial membrane efficiently, most likely then disturbing the ribosome assembly, and resulting in further intracellular damage. Api795 with its IOIO-motif, which was particularly active and only slightly toxic in vitro, appears to represent a promising third generation lead compound for the development of novel antibiotics against P. aeruginosa.

9.
Metallomics ; 7(2): 371-84, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25608481

RESUMO

Compounds which are able to destabilize the lysosomal membrane have been proposed as interesting candidates for targeted anticancer drugs due to the pronounced lysosomal changes in cancer cells. For this purpose, metallocene derivatives of a cell penetrating polyarginine peptide M­(Arg)9(Phe)2Lys­NH2 (where M = ferrocene carboxylate or ruthenocene carboxylate) were designed and their biological activities were investigated in detail. The ferrocenoyl- and ruthenocenoyl polyarginine bioconjugates were synthesized via Fmoc solid-phase peptide synthesis (SPPS) protocols on a microwave-assisted synthesizer. After HPLC purification >98% purity was observed for all conjugates. Their interaction with supported biomimetic membranes was investigated on a quartz crystal microbalance (QCM) and revealed a very strong binding of the metallocene peptides and their metal-free congeners to an artificial eukaryotic membrane model (DMPC­cholesterol). To demonstrate their antiproliferative utility as cytotoxic compounds for a targeted anticancer drug, cell viability (by the crystal violet assay), apoptosis (flow cytometry, Ann V/PI staining), induction of reactive oxygen species (ROS, by flow cytometry with dihydroethidium staining), and changes in cancer cell metabolism, e.g. respiration and glycolysis, were studied. Our results reveal only a weak toxicity for the metal-free polyarginine peptide, which could be significantly enhanced (to ca. 50 µM against HeLa cells in the best case) by coupling ferrocene or ruthenocene carboxylates to the N-terminus of the peptide. The investigation of the cellular uptake and intracellular localization by fluorescence microscopy revealed an enhanced vesicular disruption by the metallocene bioconjugate compared to the metal-free derivative which could be triggered by light and chemicals. Further studies of apoptosis, respiration, glycolysis and ROS formation reveal the superior characteristics of the metallocene compounds. While most cells remain viable even at 300 µM of the metal free bioconjugate 1, most cells are dead or in late stages of apoptosis at 200 µM of the ruthenocene derivative 3, and at 100 µM of the most active ferrocene derivative 2, however, all show very little sign of necrosis. Also, the metal free compound 1 does not induce ROS formation but both metallocene­polyarginine bioconjugates are clearly associated with enhanced intracellular ROS levels, with levels for the redox-active ferrocene derivative being two times higher than for the structurally very similar but redox-silent ruthenocene derivative. We propose that such metallocene­polyarginine peptides induce lysosomal membrane permeabilization and thereby could be developed towards targeted anticancer drugs.


Assuntos
Vesículas Citoplasmáticas/metabolismo , Lisossomos/metabolismo , Compostos Organometálicos/metabolismo , Peptídeos/metabolismo , Apoptose , Ciclo Celular , Fluoresceína-5-Isotiocianato/metabolismo , Violeta Genciana/metabolismo , Células HeLa , Células Hep G2 , Humanos , Concentração Inibidora 50 , Espécies Reativas de Oxigênio/metabolismo , Padrões de Referência
10.
ACS Chem Biol ; 7(7): 1281-91, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22594381

RESUMO

The emergence of multiple-drug-resistant (MDR) bacterial pathogens in hospitals (nosocomial infections) presents a global threat of growing importance, especially for Gram-negative bacteria with extended spectrum ß-lactamase (ESBL) or the novel New Delhi metallo-ß-lactamase 1 (NDM-1) resistance. Starting from the antibacterial peptide apidaecin 1b, we have optimized the sequence to treat systemic infections with the most threatening human pathogens, such as Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Acinetobacter baumannii. The lead compound Api88 enters bacteria without lytic effects at the membrane and inhibits chaperone DnaK at the substrate binding domain with a K(D) of 5 µmol/L. The Api88-DnaK crystal structure revealed that Api88 binds with a seven residue long sequence (PVYIPRP), in two different modes. Mice did not show any sign of toxicity when Api88 was injected four times intraperitoneally at a dose of 40 mg/kg body weight (BW) within 24 h, whereas three injections of 1.25 mg/kg BW and 5 mg/kg BW were sufficient to rescue all animals in lethal sepsis models using pathogenic E. coli strains ATCC 25922 and Neumann, respectively. Radioactive labeling showed that Api88 enters all organs investigated including the brain and is cleared through both the liver and kidneys at similar rates. In conclusion, Api88 is a novel, highly promising, 18-residue peptide lead compound with favorable in vitro and in vivo properties including a promising safety margin.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Desenho de Fármacos , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Peptídeos/química , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Farmacorresistência Bacteriana Múltipla/fisiologia , Feminino , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Peptídeos/genética , Resultado do Tratamento
11.
J Med Chem ; 53(14): 5240-7, 2010 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-20565063

RESUMO

Small proline-rich antimicrobial peptides (AMP) have attracted considerable interest, as they target specific intracellular bacterial components and do not act by lytic mechanisms. Here, a novel peptide, termed oncocin (VDKPPYLPRPRPPRRIYNR-NH(2)), is reported that was optimized for the treatment of Gram-negative pathogens. Its minimal inhibitory concentrations in tryptic soy broth medium ranged from 0.125 to 8 microg/mL for 34 different strains and clinical isolates of Enterobacteriaceae and nonfermenters, such as Escherichia coli , Pseudomonas aeruginosa , and Acinetobacter baumannii . Substitutions of two arginine residues by ornithine increased the half-lives in full mouse serum from about 20 min to greater than 180 min and the activity. Both optimized oncocin derivatives were neither toxic to human cell lines nor hemolytic to human erythrocytes. They could freely penetrate lipid membranes and were washed out completely without any sign of lytic activity, as assessed by quartz crystal microbalance. Fluorescence labeled peptides entered the periplasmic space within 20 min at room temperature and homogeneously stained E. coli within 50 min. In conclusion, the optimized oncocin represents a very promising candidate for future in vivo work and may serve as a novel lead compound for an antibacterial drug class.


Assuntos
Antibacterianos/química , Peptídeos Catiônicos Antimicrobianos/química , Bactérias Gram-Negativas/efeitos dos fármacos , Peptídeos/química , Sequência de Aminoácidos , Animais , Antibacterianos/farmacologia , Antibacterianos/toxicidade , Peptídeos Catiônicos Antimicrobianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular , Hemólise , Humanos , Membranas Artificiais , Camundongos , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Peptídeos/farmacologia , Peptídeos/toxicidade , Permeabilidade , Relação Estrutura-Atividade
12.
Biomaterials ; 30(4): 682-9, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19000635

RESUMO

Supported phospholipid bilayers are frequently used to establish a pseudo-physiological environment required for the study of protein function or the design of enzyme-based biosensors and biocatalytic reactors. These membranes are deposited from bilayer vesicles (liposomes) that rupture and fuse into a planar membrane upon adhesion to a surface. However, the morphology and homogeneity of the resulting layer is affected by the characteristics of the precursor liposome suspension and the substrate. Here we show that two distinct liposome populations contribute to membrane formation--equilibrium liposomes and small unilamellar vesicles. Liposome deposition onto carboxylic acid terminated self-assembled monolayers resulted in planar mono- and multilayer, vesicular and composite membranes, as a function of liposome size and composition. Quartz crystal microbalance data provided estimates for layer thicknesses and sheer moduli and were used for classification of the final structure. Finally, atomic force microscopy data illustrated the inherently inhomogeneous and dynamic nature of these membranes.


Assuntos
Ácidos Carboxílicos/química , Bicamadas Lipídicas/química , Fosfolipídeos/química , 1,2-Dipalmitoilfosfatidilcolina/química , Lipossomos/química , Microscopia de Força Atômica , Modelos Químicos , Tamanho da Partícula , Fosfatidilgliceróis/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA