Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
J Biol Chem ; 294(42): 15304-15317, 2019 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-31439664

RESUMO

The aggregation of the tau protein into neurofibrillary tangles is believed to correlate with cognitive decline in several neurodegenerative disorders, including Alzheimer's disease. Recent studies suggest that tau's interactions with the cell membrane could serve as a toxicity pathway and also enhance fibrillation into paired helical filaments (PHFs). Conformational changes associated with tau-membrane interactions are poorly understood, and their characterization could improve our understanding of tau pathogenicity. In this study, we investigated the molecular level structural changes associated with the interaction of the tau hexapeptide PHF6 with model lipid membranes and characterized the effects of these interactions on membrane stability and peptide fibrillation. We used two PHF6 forms, the aggregation-prone PHF6 with N-terminal acetylation (Ac-PHF6) and the non-aggregation prone PHF6 with a standard N terminus (NH3+-PHF6). We found that both PHF6 peptides are neurotoxic and exhibit similar membrane-mediated changes, consisting of: 1) favorable interactions with anionic membranes, 2) membrane destabilization through lipid extraction, and 3) membrane-mediated fibrillation. The rate at which these changes occurred was the main difference between the two peptides. NH3+-PHF6 displayed slow membrane-mediated fibrillation after 6 days of incubation, whereas Ac-PHF6 adopted a ß-sheet conformation at the surface of the membrane within hours. Ac-PHF6 interactions with the membrane were also accompanied by membrane invagination and rapid membrane destabilization. Overall, our results reveal that membrane interactions could play a critical role in tau toxicity and fibrillation, and highlight that unraveling these interactions is important for significantly advancing the development of therapeutic strategies to manage tau-associated neurodegenerative diseases.


Assuntos
Membrana Celular/metabolismo , Emaranhados Neurofibrilares/metabolismo , Peptídeos/metabolismo , Proteínas tau/metabolismo , Acetilação , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Motivos de Aminoácidos , Membrana Celular/genética , Humanos , Emaranhados Neurofibrilares/genética , Peptídeos/genética , Peptídeos/toxicidade , Estrutura Secundária de Proteína , Proteínas tau/química , Proteínas tau/genética , Proteínas tau/toxicidade
2.
Proc Natl Acad Sci U S A ; 114(28): 7278-7282, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28642346

RESUMO

Photodynamic therapy, in which malignant tissue is killed by targeted light exposure following administration of a photosensitizer, can be a valuable treatment modality but currently relies on passive transport and local irradiation to avoid off-target oxidation. We present a system of excited-state control for truly local delivery of singlet oxygen. An anionic phenylene ethynylene oligomer is initially quenched by water, producing minimal fluorescence and no measurable singlet oxygen generation. When presented with a binding partner, in this case an oppositely charged surfactant, changes in solvent microenvironment result in fluorescence unquenching, restoration of intersystem crossing to the triplet state, and singlet oxygen generation, as assayed by transient absorption spectroscopy and chemical trapping. This solvation-controlled photosensitizer model has possible applications as a theranostic agent for, for example, amyloid diseases.


Assuntos
Alcinos/química , Derivados de Benzeno/química , Detergentes/química , Ésteres/química , Fármacos Fotossensibilizantes/química , Amiloide/química , Diagnóstico por Imagem , Transferência Ressonante de Energia de Fluorescência , Gases , Humanos , Micelas , Microscopia de Fluorescência , Oxigênio/química , Fotoquimioterapia , Fótons , Espécies Reativas de Oxigênio/metabolismo , Solventes/química , Espectrofotometria , Tensoativos
3.
Nano Lett ; 19(10): 7365-7369, 2019 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-31538793

RESUMO

Biomembranes are hard to compress laterally, and membrane area compressibility has not been associated with biological processes. Using X-ray surface scattering, we observed that bacterial Shiga toxin compresses lipid packing in a gel phase monolayer upon binding to its cellular receptor, the glycolipid Gb3. This toxin-induced reorganization of lipid packing reached beyond the immediate membrane patch that the protein was bound to, and linkers separating the Gb3 carbohydrate and ceramide moieties modulated the toxin's capacity to compress the membrane. Within a natural membrane, asymmetric compression of the toxin-bound leaflet could provide a mechanism to initiate narrow membrane bending, as observed upon toxin entry into cells. Such lipid compression and long-range membrane reorganization by glycolipid-binding proteins represent novel concepts in membrane biology that have direct implications for the construction of endocytic pits in clathrin-independent endocytosis.


Assuntos
Membrana Celular/metabolismo , Fosfatidiletanolaminas/metabolismo , Toxina Shiga/metabolismo , Shigella dysenteriae/metabolismo , Triexosilceramidas/metabolismo , Disenteria Bacilar/metabolismo , Endocitose , Humanos , Modelos Moleculares
4.
Langmuir ; 35(48): 16024-16036, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31509701

RESUMO

In Alzheimer's disease, the amyloid-beta peptide (Aß) is implicated in neuronal toxicity via interactions with the cell membrane. Monomeric Aß (Aßm) is intrinsically disordered, but it can adopt a range of aggregated conformations with varying toxicities from short fibrillar oligomers (FO), to globular nonfibrillar oligomers (NFO), and full-length amyloid fibrils. NFO is considered to be the most toxic, followed by fibrils, and finally Aßm. To elucidate molecular-level membrane interactions that contribute to their different toxicities, we used liquid surface X-ray scattering and Langmuir trough insertion assays to compare Aßm, FO, and NFO surface activities and interactions with anionic DMPG lipid monolayers at the air/water interface. All Aß species were highly surface active and rapidly adopted ß-sheet rich structures upon adsorption to the air/water interface. Likewise, all Aß species had affinity for the anionic membrane. Aßm rapidly converted to ß-sheet rich assemblies upon binding the membrane, and these aggregated structures of Aßm and FO disrupted hexagonally packed lipid domains and resulted in membrane thinning and instability. In contrast, NFO perturbed membrane structure by extracting lipids from the air/water interface and causing macroscale membrane deformations. Altogether, our results support two models for membrane-mediated Aß toxicity: fibril-induced reorganization of lipid packing and NFO-induced membrane destabilization and lipid extraction. This work provides a structural understanding of Aß neurotoxicity via membrane interactions and aids the effort in understanding early events in Alzheimer's disease and other neurodegenerative diseases.


Assuntos
Peptídeos beta-Amiloides/química , Adsorção , Ânions , Membranas Artificiais , Fosfolipídeos/química , Conformação Proteica
5.
Langmuir ; 35(2): 307-325, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30056722

RESUMO

In this retrospective, we first reviewed the synthesis of the oligo(phenylene-ethynylene) electrolytes (OPEs) we created in the past 10 years. Since the general antimicrobial activity of these OPEs had been reported in our previous account in Langmuir, we are focusing only on the unusual spectroscopic and photophysical properties of these OPEs and their complexes with anionic scaffolds and detergents in this Feature Article. We applied classical all-atom MD simulations to study the hydrogen bonding environment in the water surrounding the OPEs with and without detergents present. Our finding is that OPEs could form a unit cluster or unit aggregate with a few oppositely charged detergent molecules, indicating that the photostability and photoreactivity of these OPEs might be considerably altered with important consequences to their activity as antimicrobials and fluorescence-based sensors. Thus, in the following sections, we showed that OPE complexes with detergents exhibit enhanced light-activated biocidal activity compared to either OPE or detergent individually. We also found that similar complexes between certain OPEs and biolipids could be used to construct sensors for the enzyme activity. Finally, the OPEs could covalently bind to microsphere surfaces to make a bactericidal surface, which is simpler and more ordered than the surface grafted from microspheres with polyelectrolytes. In the Conclusions and Prospects section, we briefly summarize the properties of OPEs developed so far and future areas for investigation.

6.
Biophys J ; 114(5): 1103-1115, 2018 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-29539397

RESUMO

The influence of carbohydrate structure on the ordering of glycosphingolipids (GSLs) and surrounding phospholipids was investigated in monolayers at the air-water interface. Binary mixtures composed of GSLs, chosen to span a range of carbohydrate complexity, and zwitterionic dipalmitoylphosphatidylcholine phospholipid, were studied. X-ray reflectivity was used to measure the out-of-plane structure of the monolayers and characterize the extension and conformation of the GSL carbohydrates. Using synchrotron grazing incidence x-ray diffraction, the in-plane packing of the lipid acyl chains and the area per molecule within ordered domains were characterized at different mole ratios of the two components. Our findings indicate that GSL-containing mixtures, regardless of the carbohydrate size, enhance the ordering of the surrounding lipids, resulting in a larger fraction of ordered phase of the monolayer and greater dimensions of the ordered domains. Reduction of the averaged area per molecule within the ordered domains was also observed but only in the cases where there was a size mismatch between the phospholipid headgroups and GSL components, suggesting that the condensation mechanism involves the relief of steric interactions between headgroups in mixtures.


Assuntos
Carboidratos/química , Glicoesfingolipídeos/química , Difração de Raios X
7.
Langmuir ; 32(47): 12542-12551, 2016 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-27464311

RESUMO

Opportunistic bacteria and viruses are a worldwide health threat prompting the need to develop new targeting modalities. A class of novel synthetic poly(phenylene ethynylene) (PPE)-based oligomeric conjugated polyelectrolytes (OPEs) have demonstrated potent wide-spectrum biocidal activity. A subset of cationic OPEs display high antiviral activity against the MS2 bacteriophage. The oligomers have been found to inactivate the bacteriophage and perturb the morphology of the MS2 viral capsid. However, details of the initial binding and interactions between the OPEs and the viruses are not well understood. In this study, we use a multiscale computational approach, including random sampling, molecular dynamics, and electronic structure calculations, to gain an understanding of the molecular-level interactions of a series of OPEs that vary in length, charge, and functional groups with the MS2 capsid. Our results show that OPEs strongly bind to the MS2 capsid protein assembly with binding energies of up to -30 kcal/mol. Free-energy analysis shows that the binding is dominated by strong van der Waals interactions between the hydrophobic OPE backbone and the capsid surface and strong electrostatic free energy contributions between the OPE charged moieties and charged residues on the capsid surface. This knowledge provides molecular-level insight into how to tailor the OPEs to optimize viral capsid disruption and increase OPE efficacy to target amphiphilic protein coats of icosahedral-based viruses.


Assuntos
Proteínas do Capsídeo/química , Capsídeo/química , Levivirus , Polieletrólitos/química , Cátions , Simulação de Dinâmica Molecular , Polímeros
8.
Adv Exp Med Biol ; 863: 55-77, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26092626

RESUMO

Flavonoids are naturally occurring phytochemicals found in a variety of fruits and vegetables and offer color, flavor, aroma, nutritional and health benefits. Flavonoids have been found to play a neuroprotective role by inhibiting and/or modifying the self-assembly of the amyloid-ß (Aß) peptide into oligomers and fibrils, which are linked to the pathogenesis of Alzheimer's disease. The neuroprotective efficacy of flavonoids has been found to strongly depend on their structure and functional groups. Flavonoids may exist in monomeric, as well as di-, tri-, tetra- or polymeric form through C-C or C-O-C linkages. It has been shown that flavonoids containing two or more units, e.g., biflavonoids, exert greater biological activity than their respective monoflavonoids. For instance, biflavonoids have the ability to distinctly alter Aß aggregation and more effectively reduce the toxicity of Aß oligomers compared to the monoflavonoid moieties. Although the molecular mechanisms remain to be elucidated, flavonoids have been shown to alter the Aß aggregation pathway to yield non-toxic, unstructured Aß aggregates, as well as directly exerting a neuroprotective effect to cells. In this chapter, we review biflavonoid-mediated Aß aggregation and toxicity, and highlight the beneficial roles biflavonoids can potentially play in the prevention and treatment of Alzheimer's disease.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides/metabolismo , Biflavonoides/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Agregação Patológica de Proteínas , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Humanos , Agregação Patológica de Proteínas/tratamento farmacológico , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Relação Estrutura-Atividade
9.
Biopolymers ; 99(1): 55-62, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23097230

RESUMO

Elastin-like polypeptide (ELP) fusions have been designed to allow large-scale, nonchromatographic purification of many soluble proteins by using the inverse transition cycling (ITC) method; however, the sensitivity of the aqueous lower critical solubility phase transition temperature (T(t)) of ELPs to the addition of cosolutes, including detergents, may be a potential hindrance in purification of proteins with surface hydrophobicity in such a manner. To identify detergents that are known to solubilize such proteins (e.g., membrane proteins) and that have little effect on the T(t) of the ELP, we screened a number of detergents with respect to their effects on the T(t) and secondary structures of a model ELP (denoted here as ELP180). We found that mild detergents (e.g., n-dodecyl-ß-D-maltoside, Triton-X100, and 3-[(3-cholamidopropyl) dimethylamino]-1-propanesulfonate) do not alter the phase transition behavior or structure (as probed by circular dichroism) of ELP180. This result is in contrast to previous studies that showed a strong effect of other detergents (e.g., sodium dodecylsulfate) on the T(t) of ELPs. Our results clearly indicate that mild detergents do not preclude ITC-based separation of ELPs, and thus that ELP fusions may prove to be useful in the purification of detergent-solubilized recombinant hydrophobic proteins, including membrane proteins, which are otherwise notoriously difficult to extract and purify by conventional separation methods (e.g., chromatography).


Assuntos
Detergentes/química , Elastina/química , Peptídeos/química , Dicroísmo Circular , Modelos Biológicos , Estrutura Secundária de Proteína , Solubilidade , Temperatura
10.
Langmuir ; 29(34): 10635-47, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23738803

RESUMO

This Feature Article focuses on recent progress made in elucidating the intermolecular interactions between a novel class of synthetic phenylene ethynylene (PPE)-based conjugated polyelectrolyte polymers (CPEs) and oligomers (OPEs) and multiscale cellular targets that give rise to their remarkable broad spectrum biocidal activity. We first review the interactions and self-assembly behaviors of the CPEs and OPEs with a set of vital biomolecules, including lipids, proteins, and nucleic acids, that reveal the potential pathways by which synthetic biocidal agents could exert toxicity. An overview of the antimicrobial effects and mechanisms of the CPEs and OPEs on multiple clinically relevant pathogens is then presented, with an emphasis on the morphological damage induced by the biocidal compounds toward the pathogens. Finally, we discuss the cytotoxicity of these materials against mammalian cells and human tissues to explore the potential applications of the CPEs and OPEs as antiseptics. We also pose some unanswered questions about their antimicrobial mechanisms, which provide direction for a future study.


Assuntos
Anti-Infecciosos/química , Polímeros/química , Animais , Dicroísmo Circular , Interações Medicamentosas , Humanos
11.
Langmuir ; 29(2): 781-92, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23240979

RESUMO

A multiscale investigation was carried out to study the dark and light-enhanced bactericidal mechanisms of poly(phenylene ethynylene) (PPE)-based cationic conjugated polyelectrolytes (CPEs) and oligo-phenylene ethynylenes (OPEs). On the morphological scale, Gram-negative E. coli cells exposed to CPE and OPE compounds in the dark show damage to the cell envelope, plasma membrane, and in some cases the cytoplasm, while with UV-irradiation, E. coli sustained catastrophic damages to both the cell envelope and cytoplasm. In contrast, the Gram-positive S. epi bacteria appeared intact when exposed to CPE and OPE compounds in the dark but showed damages to the cell envelope with UV-irradiation. To better understand the molecular basis of CPE- and OPE-induced morphological changes and damages to bacteria, we investigated the effect of these compounds on model bacterial plasma membrane and bacterial proteins and plasmid DNA. Measurements of dark membrane perturbation activity of the CPEs and OPEs using model lipid membranes support a carpet or detergent-like mechanism by which the antimicrobial compounds induce membrane collapse and phase transitions. Under UV-irradiation, E. coli bacteria exposed to CPEs and OPEs showed covalent modifications and damages to both cellular protein and plasmid DNA, likely through oxidative pathways mediated by singlet oxygen and subsequent reactive oxygen species sensitized by the CPE and OPE compounds. Our finding thus show that the antimicrobial polymers and oligomers exert toxicity toward Gram-negative bacteria by disrupting the morphology and structures of cell envelope and cytoplasm, including cellular components such as proteins and DNA, while exert toxicity toward Gram-positive bacteria by binding to and disrupting just the cell wall.


Assuntos
Alcinos/química , Antibacterianos/farmacologia , Membrana Celular/efeitos dos fármacos , Parede Celular/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Éteres/química , Poliaminas/farmacologia , Staphylococcus epidermidis/efeitos dos fármacos , Antibacterianos/síntese química , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Membrana Celular/efeitos da radiação , Parede Celular/efeitos da radiação , Escherichia coli/química , Escherichia coli/efeitos da radiação , Bicamadas Lipídicas/efeitos da radiação , Oxirredução , Estresse Oxidativo , Plasmídeos/antagonistas & inibidores , Plasmídeos/química , Poliaminas/síntese química , Polieletrólitos , Polimerização , Oxigênio Singlete/química , Especificidade da Espécie , Staphylococcus epidermidis/química , Staphylococcus epidermidis/efeitos da radiação , Raios Ultravioleta
12.
Langmuir ; 29(37): 11713-23, 2013 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-24004419

RESUMO

Amyloid-ß peptide (Aß)-membrane interactions have been implicated in the formation of toxic oligomers that permeabilize membranes, allowing an influx of calcium ions and triggering cell death in the pathogenesis of Alzheimer's disease (AD). Curcumin, a small dietary polyphenolic molecule, has been shown to reduce Aß-induced toxicity and AD pathology. We investigate here the effect of curcumin on Aß40-induced toxicity in cultured human neuroblastoma SH-SY5Y cells and test a novel neuroprotection mechanism in which curcumin reduces Aß-membrane interactions and attenuates Aß-induced membrane disruptions. Predominantly monomeric Aß40 exerts toxicity toward SH-SY5Y cells and has been shown to insert spontaneously into anionic lipid monolayers at the air/water interface, resulting in the misfolding and assembly of Aß into ß-sheet-enriched oligomers. Concomitantly, membrane morphology and lipid packing are disrupted. Curcumin dose-dependently ameliorates Aß-induced neurotoxicity and reduces either the rate or extent of Aß insertion into anionic lipid monolayers. Moreover, curcumin reduces Aß-induced dye leakage from lipid-bilayer-covered, dye-loaded, porous silica microspheres. Because curcumin neither affects the inherent surface activity of Aß nor modifies the membrane properties, it reduces Aß insertion by directly attenuating Aß-membrane interactions and reducing Aß-induced membrane disruption. Although the exact molecular mechanism of curcumin's membrane protective effect remains unclear, this effect could in part contribute to curcumin's neuroprotective effect with respect to Aß-induced toxicity. Our work reveals a novel molecular mechanism by which curcumin reduces Aß-related pathology and toxicity and suggests a therapeutic strategy for preventing or treating AD by targeting the inhibition of Aß-induced membrane disruption.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Membrana Celular/efeitos dos fármacos , Curcumina/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Peptídeos beta-Amiloides/farmacologia , Curcumina/química , Relação Dose-Resposta a Droga , Humanos , Modelos Moleculares , Estrutura Molecular , Neurônios/patologia , Fármacos Neuroprotetores/química , Relação Estrutura-Atividade , Células Tumorais Cultivadas
13.
ACS Omega ; 8(11): 10148-10159, 2023 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-36969430

RESUMO

Selective photosensitized oxidation of amyloid protein aggregates is being investigated as a possible therapeutic strategy for treating Alzheimer's disease (AD). Photo-oxidation has been shown to degrade amyloid-ß (Aß) aggregates and ameliorate aggregate toxicity in vitro and reduce aggregate levels in the brains of AD animal models. To shed light on the mechanism by which photo-oxidation induces fibril destabilization, we carried out an all-atom molecular dynamics (MD) simulation to examine the effect of methionine (Met35) oxidation on the conformation and stability of a ß-sheet-rich Aß9-40 protofibril. Analyses of up to 1 µs simulations showed that the oxidation of the Met35 residues, which resulted in the addition of hydrophilic oxygens in the fibril core, reduced the overall conformational stability of the protofibril. Specifically, Met35 disrupted the hydrophobic interface that stabilizes the stacking of the two hexamers that comprise the protofibril. The oxidized protofibril is more solvent exposed and exhibits more backbone flexibility. However, the protofibril retained the underlying U-shaped architecture of each peptide upon oxidation, and although some loss of ß-sheets occurred, a significant portion remained. Our simulation results are thus consistent with our experimental observation that photo-oxidation of Aß40 fibril resulted in the dis-agglomeration and fragmentation of Aß fibrils but did not cause complete disruption of the fibrillar morphology or ß-sheet structures. The partial destabilization of Aß aggregates supports the further development of photosensitized platforms for the targeting and clearing of Aß aggregates as a therapeutic strategy for treating AD.

14.
ACS Appl Bio Mater ; 6(8): 2916-2924, 2023 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-37417798

RESUMO

The synthetic cationic conjugated polyelectrolytes and oligomers have demonstrated great effectiveness and versatility as antimicrobial materials. They have the ability to eliminate or render inactive various pathogens, including viruses like SARS-CoV-2, bacteria, and fungi. These pathogens can be rapidly eradicated when the polyelectrolytes and oligomers are applied as sprays, wipes, or coatings on solid surfaces. Inactivation of the pathogens occurs through two distinct processes: a non-light-activated process similar to Quats, and a more efficient and faster process that is triggered by light. These materials possess fluorescence and photosensitizing properties, enabling prolonged protection when coated on surfaces. The level of fluorescence exhibited by samples applied to nonfluorescent surfaces serves as an indicator of the coating's integrity and viability, making it easily detectable. Importantly, these materials demonstrate low toxicity towards mammalian cells and human skin, allowing for their safe use. While they can serve as durable coatings for pathogen protection, extended exposure to visible or ultraviolet light leads to their photochemical degradation. Our research also suggests that these materials act against pathogens through nonspecific mechanisms, minimizing the likelihood of pathogens developing resistance and rendering the materials ineffective.


Assuntos
COVID-19 , Desinfecção , Animais , Humanos , Polieletrólitos , COVID-19/prevenção & controle , SARS-CoV-2 , Mamíferos
15.
Biosensors (Basel) ; 13(2)2023 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-36831917

RESUMO

The identification of protein aggregates as biomarkers for neurodegeneration is an area of interest for disease diagnosis and treatment development. In this work, we present novel super luminescent conjugated polyelectrolyte molecules as ex vivo sensors for tau-paired helical filaments (PHFs) and amyloid-ß (Aß) plaques. We evaluated the use of two oligo-p-phenylene ethynylenes (OPEs), anionic OPE12- and cationic OPE24+, as stains for fibrillar protein pathology in brain sections of transgenic mouse (rTg4510) and rat (TgF344-AD) models of Alzheimer's disease (AD) tauopathy, and post-mortem brain sections from human frontotemporal dementia (FTD). OPE12- displayed selectivity for PHFs in fluorimetry assays and strong staining of neurofibrillary tangles (NFTs) in mouse and human brain tissue sections, while OPE24+ stained both NFTs and Aß plaques. Both OPEs stained the brain sections with limited background or non-specific staining. This novel family of sensors outperformed the gold-standard dye Thioflavin T in sensing capacities and co-stained with conventional phosphorylated tau (AT180) and Aß (4G8) antibodies. As the OPEs readily bind protein amyloids in vitro and ex vivo, they are selective and rapid tools for identifying proteopathic inclusions relevant to AD. Such OPEs can be useful in understanding pathogenesis and in creating in vivo diagnostically relevant detection tools for neurodegenerative diseases.


Assuntos
Doença de Alzheimer , Emaranhados Neurofibrilares , Camundongos , Humanos , Ratos , Animais , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Placa Amiloide , Proteínas tau , Doença de Alzheimer/diagnóstico , Encéfalo/metabolismo , Peptídeos beta-Amiloides , Coloração e Rotulagem , Etilenos/metabolismo
16.
Biochemistry ; 51(12): 2539-50, 2012 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-22401494

RESUMO

The misfolding and aggregation of the intrinsically disordered, microtubule-associated tau protein into neurofibrillary tangles is implicated in the pathogenesis of Alzheimer's disease. However, the mechanisms of tau aggregation and toxicity remain unknown. Recent work has shown that anionic lipid membranes can induce tau aggregation and that membrane permeabilization may serve as a pathway by which protein aggregates exert toxicity, suggesting that the plasma membrane may play dual roles in tau pathology. This prompted our investigation to assess tau's propensity to interact with membranes and to elucidate the mutually disruptive structural perturbations the interactions induce in both tau and the membrane. We show that although highly charged and soluble, the full-length tau (hTau40) is also highly surface active, selectively inserts into anionic DMPG lipid monolayers and induces membrane morphological changes. To resolve molecular-scale structural details of hTau40 associated with lipid membranes, X-ray and neutron scattering techniques are utilized. X-ray reflectivity indicates hTau40s presence underneath a DMPG monolayer and penetration into the lipid headgroups and tailgroups, whereas grazing incidence X-ray diffraction shows that hTau40 insertion disrupts lipid packing. Moreover, both air/water and DMPG lipid membrane interfaces induce the disordered hTau40 to partially adopt a more compact conformation with density similar to that of a folded protein. Neutron reflectivity shows that tau completely disrupts supported DMPG bilayers while leaving the neutral DPPC bilayer intact. Our results show that hTau40s strong interaction with anionic lipids induces tau structural compaction and membrane disruption, suggesting possible membrane-based mechanisms of tau aggregation and toxicity in neurodegenerative diseases.


Assuntos
Membrana Celular/química , Membrana Celular/metabolismo , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo , Humanos , Difração de Nêutrons , Ligação Proteica , Conformação Proteica , Solubilidade , Difração de Raios X
17.
Langmuir ; 28(1): 65-70, 2012 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-22148923

RESUMO

The bactericidal mechanisms of poly(phenylene ethynylene) (PPE)-based cationic conjugated polyelectrolytes (CPE) and oligo-phenylene ethynylenes (OPE) were investigated using electron/optical microscopy and small-angle X-ray scattering (SAXS). The ultrastructural analysis shows that polymeric PPE-Th can significantly remodel the bacterial outer membrane and/or the peptidoglycan layer, followed by the possible collapse of the bacterial cytoplasm membrane. In contrast, oligomeric end-only OPE (EO-OPE) possesses potent bacteriolysis activity, which efficiently disintegrates the bacterial cytoplasm membrane and induces the release of bacterial cell content. Using single giant vesicles and SAXS, we demonstrated that the membrane perturbation mechanism of EO-OPE against model bacterial membranes results from a 3D membrane phase transition or perturbation.


Assuntos
Bactérias/efeitos dos fármacos , Eletrólitos/farmacologia , Polímeros/farmacologia , Cátions , Escherichia coli/efeitos dos fármacos , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Espalhamento de Radiação
18.
ACS Appl Mater Interfaces ; 14(4): 4892-4898, 2022 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-35040619

RESUMO

This paper presents results of a study of a new cationic oligomer that contains end groups and a chromophore affording inactivation of SARS-CoV-2 by visible light irradiation in solution or as a solid coating on paper wipes and glass fiber filtration substrates. A key finding of this study is that the cationic oligomer with a central thiophene ring and imidazolium charged groups gives outstanding performance in both the killing of E. coli bacterial cells and inactivation of the virus at very short times. Our introduction of cationic N-methyl imidazolium groups enhances the light activation process for both E. coli and SARS-CoV-2 but dampens the killing of the bacteria and eliminates the inactivation of the virus in the dark. For the studies with this oligomer in solution at a concentration of 1 µg/mL and E. coli, we obtain 3 log killing of the bacteria with 10 min of irradiation with LuzChem cool white lights (mimicking indoor illumination). With the oligomer in solution at a concentration of 10 µg/mL, we observe 4 log inactivation (99.99%) in 5 min of irradiation and total inactivation after 10 min. The oligomer is quite active against E. coli on oligomer-coated paper wipes and glass fiber filter supports. The SARS-CoV-2 is also inactivated by oligomer-coated glass fiber filter papers. This study indicates that these oligomer-coated materials may be very useful as wipes and filtration materials.


Assuntos
Antivirais/farmacologia , COVID-19/terapia , SARS-CoV-2/efeitos da radiação , COVID-19/genética , COVID-19/virologia , Cátions/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/efeitos da radiação , Humanos , Luz , Fototerapia , SARS-CoV-2/patogenicidade , Raios Ultravioleta , Inativação de Vírus/efeitos dos fármacos , Inativação de Vírus/efeitos da radiação
19.
ACS Appl Mater Interfaces ; 14(13): 14871-14886, 2022 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-35344326

RESUMO

Photodynamic therapy (PDT) has been explored as a therapeutic strategy to clear toxic amyloid aggregates involved in neurodegenerative disorders such as Alzheimer's disease. A major limitation of PDT is off-target oxidation, which can be lethal for the surrounding cells. We have shown that a novel class of oligo-p-phenylene ethynylenes (OPEs) exhibit selective binding and fluorescence turn-on in the presence of prefibrillar and fibrillar aggregates of disease-relevant proteins such as amyloid-ß (Aß) and α-synuclein. Concomitant with fluorescence turn-on, OPE also photosensitizes singlet oxygen under illumination through the generation of a triplet state, pointing to the potential application of OPEs as photosensitizers in PDT. Herein, we investigated the photosensitizing activity of an anionic OPE for the photo-oxidation of Aß fibrils and compared its efficacy to the well-known but nonselective photosensitizer methylene blue (MB). Our results show that, while MB photo-oxidized both monomeric and fibrillar conformers of Aß40, OPE oxidized only Aß40 fibrils, targeting two histidine residues on the fibril surface and a methionine residue located in the fibril core. Oxidized fibrils were shorter and more dispersed but retained the characteristic ß-sheet rich fibrillar structure and the ability to seed further fibril growth. Importantly, the oxidized fibrils displayed low toxicity. We have thus discovered a class of novel theranostics for the simultaneous detection and oxidization of amyloid aggregates. Importantly, the selectivity of OPE's photosensitizing activity overcomes the limitation of off-target oxidation of traditional photosensitizers and represents an advancement of PDT as a viable strategy to treat neurodegenerative disorders.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Doença de Alzheimer/metabolismo , Amiloide/química , Peptídeos beta-Amiloides/metabolismo , Proteínas Amiloidogênicas , Humanos , Fragmentos de Peptídeos/química , Conformação Proteica em Folha beta
20.
Biophys J ; 101(1): 118-27, 2011 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-21723821

RESUMO

Lipid interfaces, ranging from cell membranes to thin surfactant layers that stabilize lung alveoli, are integral to living systems. Such interfaces are often subjected to mechanical forces, and because of their membrane-like geometry, they can easily deform by bending into localized folds. In this work, we explore the role of small molecules (i.e., glycerol) on the mechanical stability of model lung surfactant monolayers. We demonstrate that the presence of glycerol increases local monolayer bending stiffness by orders of magnitude. Our x-ray and neutron reflectivity measurements indicate that water is preferentially depleted, or glycerol is preferentially enriched, at the lipid headgroup/solvent interface, and that this glycerol-enriched layer extends O(10Å) beneath the monolayer with an adsorption free energy of -2.5 to -4.6 kJ/mol. The dramatic change in membrane bending stiffness in the presence of the sugar adlayer is understood in terms of two models: 1), lipid antiplasticization by glycerol; and 2), a continuum mechanical model of the viscous adlayer.


Assuntos
Glicerol/química , Fluidez de Membrana , Lipídeos de Membrana/química , Nêutrons , Análise Espectral , Temperatura , Viscosidade , Difração de Raios X , Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA