Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Phys Chem Lett ; 12(23): 5587-5592, 2021 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-34109791

RESUMEN

The confinement effect of biological ion channels regulates the transport of molecules and ions due to angstrom-sized pores. The structure of the potassium channel has a selection region (3-4 Å), a cavity (10 Å), and a gated region, while ZIF-8 has intrinsic pores with a 3.4 Å aperture and an 11.6 Å cavity similar to those of the potassium channel. Inspired by this, we constructed the glass/ZIF-8 hybrid membrane through an electrochemical growth process to explore the kinetics of the ion transmembrane by I-V curves and electrochemical impedance spectroscopy. These complementary approaches yield highly correlated results that show that ion transportation of the ZIF-8 membrane follows Arrhenius behavior. The rates of ions are controlled by the transmembrane activation energy, in which the ionic charge and radius play an important role.


Asunto(s)
Imidazoles/farmacocinética , Estructuras Metalorgánicas/farmacocinética , Metales Alcalinotérreos/farmacocinética , Nanotecnología/métodos , Canales de Potasio/farmacocinética , Imidazoles/química , Canales Iónicos/química , Canales Iónicos/farmacocinética , Transporte Iónico/fisiología , Cinética , Estructuras Metalorgánicas/química , Metales Alcalinotérreos/química , Canales de Potasio/química
2.
Cell Mol Biol (Noisy-le-grand) ; 66(4): 243-249, 2020 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-32583783

RESUMEN

Phytol (PHY), a chlorophyll-derived diterpenoid, exhibits numerous pharmacological properties, including antioxidant, antimicrobial, and anticancer activities. This study evaluates the anti-diarrheal effect of phytol (PHY) along with its possible mechanism of action through in-vivo and in-silico models. The effect of PHY was investigated on castor oil-induced diarrhea in Swiss mice by using prazosin, propranolol, loperamide, and nifedipine as standards with or without PHY. PHY at 50 mg/kg (p.o.) and all other standards exhibit significant (p < 0.05) anti-diarrheal effect in mice. The effect was prominent in the loperamide and propranolol groups. PHY co-treated with prazosin and propranolol was found to increase in latent periods along with a significant reduction in diarrheal section during the observation period than other individual or combined groups. Furthermore, molecular docking studies also suggested that PHY showed better interactions with the α- and ß-adrenergic receptors, especially with α-ADR1a and ß-ADR1. In the former case, PHY showed interaction with hydroxyl group of Ser192 at a distance of 2.91Å, while in the latter it showed hydrogen bond interactions with Thr170 and Lys297 with a distance of 2.65 and 2.72Å, respectively. PHY exerted significant anti-diarrheal effect in Swiss mice, possibly through blocking α- and ß-adrenergic receptors.


Asunto(s)
Simulación por Computador , Diarrea/tratamiento farmacológico , Modelos Biológicos , Fitol/uso terapéutico , Secuencia de Aminoácidos , Animales , Aceite de Ricino , Modelos Animales de Enfermedad , Ayuno , Humanos , Canales Iónicos/química , Canales Iónicos/metabolismo , Masculino , Ratones , Simulación del Acoplamiento Molecular , Fitol/farmacología , Receptores Adrenérgicos alfa 1/química , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos beta/química , Receptores Adrenérgicos beta/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Homología de Secuencia de Aminoácido
3.
Chem Rec ; 20(7): 730-742, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31944562

RESUMEN

An artificial cell membrane that is composed of bilayer lipid membranes (BLMs) with transmembrane proteins incorporated within them represents a well-defined system for the analysis of membrane proteins, especially ion channel proteins that are major targets for drug design. Because the BLM system has a high compatibility with recently developed cell-free expression systems, it has attracted attention as a next-generation drug screening system. However, three issues associated with BLM systems, i. e., their instability, the need for non-volatile organic solvents and a low efficiency of ion channel incorporation, have limited their use as a drug screening platform. In this personal account, we discuss our recent approaches to address these issues based on microfabrication. We also discuss the potential for using the BLM system combined with cell-free expression systems as a drug screening system for future personalized medicine.


Asunto(s)
Canales Iónicos/química , Membrana Dobles de Lípidos/química , Evaluación Preclínica de Medicamentos
4.
Int J Mol Sci ; 20(18)2019 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-31540178

RESUMEN

Ion channels contribute fundamental properties to cell membranes. Although highly diverse in conductivity, structure, location, and function, many of them can be regulated by common mechanisms, such as voltage or (de-)phosphorylation. Primarily considering ion channels involved in the nociceptive system, this review covers more novel and less known features. Accordingly, we outline noncanonical operation of voltage-gated sodium, potassium, transient receptor potential (TRP), and hyperpolarization-activated cyclic nucleotide (HCN)-gated channels. Noncanonical features discussed include properties as a memory for prior voltage and chemical exposure, alternative ion conduction pathways, cluster formation, and silent subunits. Complementary to this main focus, the intention is also to transfer knowledge between fields, which become inevitably more separate due to their size.


Asunto(s)
Canales Iónicos/metabolismo , Dolor/etiología , Dolor/metabolismo , Animales , Susceptibilidad a Enfermedades , Descubrimiento de Drogas , Humanos , Activación del Canal Iónico , Canales Iónicos/química , Canales Iónicos/genética , Dolor/tratamiento farmacológico
5.
Int J Mol Sci ; 20(2)2019 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-30669290

RESUMEN

Calmodulin (CaM) is the principal Ca2+ sensor in eukaryotic cells, orchestrating the activity of hundreds of proteins. Disease causing mutations at any of the three genes that encode identical CaM proteins lead to major cardiac dysfunction, revealing the importance in the regulation of excitability. In turn, some mutations at the CaM binding site of ion channels cause similar diseases. Here we provide a summary of the two sides of the partnership between CaM and ion channels, describing the diversity of consequences of mutations at the complementary CaM binding domains.


Asunto(s)
Calmodulina/genética , Calmodulina/metabolismo , Susceptibilidad a Enfermedades , Canales Iónicos/genética , Canales Iónicos/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio , Calmodulina/química , Regulación de la Expresión Génica , Humanos , Activación del Canal Iónico , Canales Iónicos/química , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Sensibilidad y Especificidad , Transducción de Señal , Relación Estructura-Actividad
6.
Methods Mol Biol ; 1800: 313-326, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29934900

RESUMEN

Ion channels are membrane proteins involved in almost all physiological processes, including neurotransmission, muscle contraction, pace-making activity, secretion, electrolyte and water balance, immune response, and cell proliferation. Due to their broad distribution in human body and physiological roles, ion channels are attractive targets for drug discovery and safety pharmacology. Over the years ion channels have been associated to many genetic diseases ("channelopathies"). For most of these diseases the therapy is mainly empirical and symptomatic, often limited by lack of efficacy and tolerability for a number of patients. The search for the development of new and more specific therapeutic approaches is therefore strongly pursued. At the same time acquired channelopathies or dangerous side effects (such as proarrhythmic risk) can develop as a consequence of drugs unexpectedly targeting ion channels. Several noncardiovascular drugs are known to block cardiac ion channels, leading to potentially fatal delayed ventricular repolarization. Thus, the search of reliable preclinical cardiac safety testing in early stage of drug discovery is mandatory. To fulfill these needs, both ion channels drug discovery and toxicology strategies are evolving toward comprehensive research approaches integrating ad hoc designed in silico predictions and experimental studies for a more reliable and quick translation of results to the clinic side.Here we discuss two examples of how the combination of in silico methods and patch clamp experiments can help addressing drug discovery and safety issues regarding ion channels.


Asunto(s)
Descubrimiento de Drogas/métodos , Canales Iónicos/química , Modelos Moleculares , Farmacovigilancia , Animales , Cardiotoxicidad , Bases de Datos de Compuestos Químicos , Evaluación Preclínica de Medicamentos , Humanos , Canales Iónicos/agonistas , Canales Iónicos/antagonistas & inhibidores , Ligandos , Células Musculares/citología , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Técnicas de Placa-Clamp , Relación Estructura-Actividad Cuantitativa , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Toxicología/métodos
7.
Electrophoresis ; 39(3): 496-503, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29193178

RESUMEN

Combining high-resolution imaging and electrophysiological recordings is key for various types of experimentation on lipid bilayers and ion channels. Here, we propose an integrated biosensing platform consisting of a microfluidic cartridge and a dedicated chip-holder to conduct such dual measurements on suspended lipid bilayers, in a user-friendly manner. To illustrate the potential of the integrated platform, we characterize lipid bilayers in terms of thickness and fluidity while simultaneously monitoring single ion channel currents. For that purpose, POPC lipid bilayers are supplemented with a fluorescently-tagged phospholipid (NBD-PE, 1% mol) for Fluorescence Recovery After Photobleaching (FRAP) measurements and a model ion channel (gramicidin, 1 nM). These combined measurements reveal that NBD-PE has no effect on the lipid bilayer thickness while gramicidin induces thinning of the membrane. Furthermore, the presence of gramicidin does not alter the lipid bilayer fluidity. Surprisingly, in lipid bilayers supplemented with both probes, a reduction in gramicidin open probability and lifetime is observed compared to lipid bilayers with gramicidin only, suggesting an influence of NBD-PE on the gramicidin ion function. Altogether, our proposed microfluidic biosensing platform in combination with the herein presented multi-parametric measurement scheme paves the way to explore the interdependent relationship between lipid bilayer properties and ion channel function.


Asunto(s)
Técnicas Biosensibles/instrumentación , Canales Iónicos/química , Membrana Dobles de Lípidos/química , Técnicas Analíticas Microfluídicas/instrumentación , Microscopía Confocal/instrumentación , Colorantes Fluorescentes/química , Gramicidina/química , Dispositivos Laboratorio en un Chip , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química
8.
Ann N Y Acad Sci ; 1411(1): 83-95, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29106710

RESUMEN

The central nervous system (CNS) plays a vital role in regulating energy balance and metabolism. Over the last 50 years, studies in animal models have allowed us to identify critical CNS regions involved in these processes and even crucial cell populations. Now, techniques for genetically and anatomically targeted manipulation of specific neural populations using light (optogenetic), ligands (chemogenetic), or magnetic fields (radiogenetic/magnetogenetic) allow detailed investigation of circuits involved in metabolic regulation. In this review, we provide a brief overview of recent studies using light- and magnetic field-regulated neural activity to investigate the neural circuits contributing to metabolic control.


Asunto(s)
Sistema Nervioso Central/fisiología , Metabolismo Energético/fisiología , Animales , Apetito/efectos de los fármacos , Apetito/fisiología , Apetito/efectos de la radiación , Campos Electromagnéticos , Diseño de Equipo , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Conducta Alimentaria/efectos de la radiación , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Glucosa/metabolismo , Homeostasis/fisiología , Humanos , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiología , Canales Iónicos/química , Canales Iónicos/efectos de los fármacos , Rayos Láser , Nanopartículas de Magnetita , Neuronas/efectos de los fármacos , Neuronas/efectos de la radiación , Optogenética , Ingeniería de Proteínas , Receptores Acoplados a Proteínas G/agonistas
9.
New Phytol ; 216(1): 46-61, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28722226

RESUMEN

Contents 46 I. 46 II. 47 III. 50 IV. 53 V. 56 VI. 57 58 58 References 58 SUMMARY: Stomatal guard cells control leaf CO2 intake and concomitant water loss to the atmosphere. When photosynthetic CO2 assimilation is limited and the ratio of CO2 intake to transpiration becomes suboptimal, guard cells, sensing the rise in CO2 concentration in the substomatal cavity, deflate and the stomata close. Screens for mutants that do not close in response to experimentally imposed high CO2 atmospheres identified the guard cell-expressed Slowly activating anion channel, SLAC1, as the key player in the regulation of stomatal closure. SLAC1 evolved, though, before the emergence of guard cells. In Arabidopsis, SLAC1 is the founder member of a family of anion channels, which comprises four homologues. SLAC1 and SLAH3 mediate chloride and nitrate transport in guard cells, while SLAH1, SLAH2 and SLAH3 are engaged in root nitrate and chloride acquisition, and anion translocation to the shoot. The signal transduction pathways involved in CO2 , water stress and nutrient-sensing activate SLAC/SLAH via distinct protein kinase/phosphatase pairs. In this review, we discuss the role that SLAC/SLAH channels play in guard cell closure, on the one hand, and in the root-shoot continuum on the other, along with the molecular basis of the channels' anion selectivity and gating.


Asunto(s)
Aniones/metabolismo , Canales Iónicos/metabolismo , Nitrógeno/metabolismo , Fósforo/metabolismo , Estomas de Plantas/fisiología , Secuencia de Aminoácidos , Activación del Canal Iónico , Canales Iónicos/química , Estomas de Plantas/citología
10.
Biomed Res Int ; 2016: 3981478, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27631006

RESUMEN

Conotoxins are a kind of neurotoxin which can specifically interact with potassium, sodium type, and calcium channels. They have become potential drug candidates to treat diseases such as chronic pain, epilepsy, and cardiovascular diseases. Thus, correctly identifying the types of ion channel-targeted conotoxins will provide important clue to understand their function and find potential drugs. Based on this consideration, we developed a new computational method to rapidly and accurately predict the types of ion-targeted conotoxins. Three kinds of new properties of residues were proposed to use in pseudo amino acid composition to formulate conotoxins samples. The support vector machine was utilized as classifier. A feature selection technique based on F-score was used to optimize features. Jackknife cross-validated results showed that the overall accuracy of 94.6% was achieved, which is higher than other published results, demonstrating that the proposed method is superior to published methods. Hence the current method may play a complementary role to other existing methods for recognizing the types of ion-target conotoxins.


Asunto(s)
Conotoxinas/química , Canales Iónicos/química , Simulación del Acoplamiento Molecular/métodos , Mapeo de Interacción de Proteínas/métodos , Análisis de Secuencia de Proteína/métodos , Algoritmos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión , Aprendizaje Automático , Datos de Secuencia Molecular , Reconocimiento de Normas Patrones Automatizadas/métodos , Unión Proteica
11.
PLoS One ; 11(3): e0150761, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26963710

RESUMEN

Since the first Hodgkin and Huxley ion channel model was described in the 1950s, there has been an explosion in mathematical models to describe ion channel function. As experimental data has become richer, models have concomitantly been improved to better represent ion channel kinetic processes, although these improvements have generally resulted in more model complexity and an increase in the number of parameters necessary to populate the models. Models have also been developed to explicitly model drug interactions with ion channels. Recent models of drug-channel interactions account for the discrete kinetics of drug interaction with distinct ion channel state conformations, as it has become clear that such interactions underlie complex emergent kinetics such as use-dependent block. Here, we describe an approach for developing a model for ion channel drug interactions. The method describes the process of extracting rate constants from experimental electrophysiological function data to use as initial conditions for the model parameters. We then describe implementation of a parameter optimization method to refine the model rate constants describing ion channel drug kinetics. The algorithm takes advantage of readily available parallel computing tools to speed up the optimization. Finally, we describe some potential applications of the platform including the potential for gaining fundamental mechanistic insights into ion channel function and applications to in silico drug screening and development.


Asunto(s)
Simulación por Computador , Canales Iónicos/química , Modelos Moleculares , Preparaciones Farmacéuticas/química , Animales , Evaluación Preclínica de Medicamentos/métodos , Humanos
12.
PLoS One ; 10(8): e0136346, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26317868

RESUMEN

In the present study, patch-clamp techniques have been used to investigate the effect of trimethyltin chloride (Met3SnCl) on the slow vacuolar (SV) channels in vacuoles from red beet (Beta vulgaris L.) taproots. Activity of SV channels has been measured in whole-vacuole and cytosolic side-out patch configurations. It was found that addition of trimethyltin chloride to the bath solution suppressed, in a concentration-dependent manner, SV currents in red beet vacuoles. The time constant, τ, increased significantly in the presence of the organotin. When single channel activity was analyzed, only little channel activity could be recorded at 100 µM Met3SnCl. Trimethyltin chloride added to the bath medium significantly decreased (by ca. threefold at 100 µM Met3SnCl and at 100 mV voltage, as compared to the control medium) the open probability of single channels. Single channel recordings obtained in the presence and absence of trimethyltin chloride showed that the organotin only slightly (by <10%) decreased the unitary conductance of single channels. It was also found that Met3SnCl significantly diminished the number of SV channel openings, whereas it did not change the opening times of the channels. Taking into account the above and the fact that under the here applied experimental conditions (pH = 7.5) Met3SnCl is a non-dissociated (more lipophilic) compound, we suggest that the suppression of SV currents observed in the presence of the organotin results probably from its hydrophobic properties allowing this compound to translocate near the selectivity filter of the channel.


Asunto(s)
Beta vulgaris/metabolismo , Canales Iónicos/metabolismo , Proteínas de Plantas/metabolismo , Raíces de Plantas/metabolismo , Compuestos de Trimetilestaño/química , Vacuolas/metabolismo , Beta vulgaris/química , Canales Iónicos/química , Proteínas de Plantas/química , Raíces de Plantas/química , Vacuolas/química
13.
PLoS One ; 10(6): e0126510, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26030803

RESUMEN

BACKGROUND: The current standard care therapy for hepatitis C virus (HCV) infection consists of two regimes, namely interferon-based and interferon-free treatments. The treatment through the combination of ribavirin and pegylated interferon is expensive, only mildly effective, and is associated with severe side effects. In 2011, two direct-acting antiviral (DAA) drugs, boceprevir and telaprevir, were licensed that have shown enhanced sustained virologic response (SVR) in phase III clinical trial, however, these interferon-free treatments are more sensitive to HCV genotype 1 infection. The variable nature of HCV, and the limited number of inhibitors developed thus aim in expanding the repertoire of available drug targets, resulting in targeting the virus assembly therapeutically. AIM: We conducted this study to predict the 3D structure of the p7 protein from the HCV genotypes 3 and 4. Approximately 63 amino acid residues encoded in HCV render this channel sensitive to inhibitors, making p7 a promising target for novel therapies. HCV p7 protein forms a small membrane known as viroporin, and is essential for effective self-assembly of large channels that conduct cation assembly and discharge infectious virion particles. METHOD: In this study, we screened drugs and flavonoids known to disrupt translation and production of HCV proteins, targeted against the active site of p7 residues of HCV genotype 3 (GT3) (isolatek3a) and HCV genotype 4a (GT4) (isolateED43). Furthermore, we conducted a quantitative structure-activity relationship and docking interaction study. RESULTS: The drug NB-DNJ formed the highest number of hydrogen bond interactions with both modeled p7 proteins with high interaction energy, followed by BIT225. A flavonoid screen demonstrated that Epigallocatechin gallate (EGCG), nobiletin, and quercetin, have more binding modes in GT3 than in GT4. Thus, the predicted p7 protein molecule of HCV from GT3 and GT4 provides a general avenue to target structure-based antiviral compounds. CONCLUSIONS: We hypothesize that the inhibitors of viral p7 identified in this screen may be a new class of potent agents, but further confirmation in vitro and in vivo is essential. This structure-guided drug design for both GT3 and GT4 can lead to the identification of drug-like natural compounds, confirming p7 as a new target in the rapidly increasing era of HCV.


Asunto(s)
Productos Biológicos/farmacología , Hepacivirus/genética , Canales Iónicos/química , Simulación del Acoplamiento Molecular , Proteínas Virales/química , Secuencia de Aminoácidos , Antivirales/farmacología , Sitios de Unión , Secuencia de Consenso , Evaluación Preclínica de Medicamentos , Flavonoides/farmacología , Genotipo , Hepacivirus/efectos de los fármacos , Humanos , Canales Iónicos/metabolismo , Ligandos , Datos de Secuencia Molecular , Filogenia , Relación Estructura-Actividad Cuantitativa , Alineación de Secuencia , Proteínas Virales/metabolismo
14.
Acc Chem Res ; 48(7): 1947-60, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26103428

RESUMEN

Light is a fascinating phenomenon that ties together physics, chemistry, and biology. It is unmatched in its ability to confer information with temporal and spatial precision and has been used to map objects on the scale of tens of nanometers (10(-8) m) to light years (10(16) m). This information, gathered through super-resolution microscopes or space-based telescopes, is ultimately funneled through the human visual system, which is a miracle in itself. It allows us to see the Andromeda galaxy at night, an object that is 2.5 million light years away and very dim, and ski the next day in bright sunlight at an intensity that is 12 orders of magnitude higher. Human vision is only one of many photoreceptive systems that have evolved on earth and are found in all kingdoms of life. These systems rely on molecular photoswitches, such as retinal or tetrapyrrols, which undergo transient bond isomerizations or bond formations upon irradiation. The set of chromophores that have been employed in Nature for this purpose is surprisingly small. Nevertheless, they control a wide variety of biological functions, which have recently been significantly increased through the rapid development of optogenetics. Optogenetics originated as an effort to control neural function with genetically encoded photoreceptors that use abundant chromophores, in particular retinal. It now covers a variety of cellular functions other than excitability and has revolutionized the control of biological pathways in neuroscience and beyond. Chemistry has provided a large repertoire of synthetic photoswitches with highly tunable properties. Like their natural counterparts, these chromophores can be attached to proteins to effectively put them under optical control. This approach has enabled a new type of synthetic photobiology that has gone under various names to distinguish it from optogenetics. We now call it photopharmacology. Here we trace our involvement in this field, starting with the first light-sensitive potassium channel (SPARK) and concluding with our most recent work on photoswitchable fatty acids. Instead of simply providing a historical account of our efforts, we discuss the design criteria that guided our choice of molecules and receptors. As such, we hope to provide a roadmap to success in photopharmacology and make a case as to why synthetic photoswitches, properly designed and made available through well-planned and efficient syntheses, should have a bright future in biology and medicine.


Asunto(s)
Luz , Fototerapia , Humanos , Canales Iónicos/química , Canales Iónicos/metabolismo , Ligandos
15.
Analyst ; 139(7): 1732-9, 2014 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-24527488

RESUMEN

As one of the powerful molecular recognition elements, the functional DNA probes have been successfully utilized to construct various biosensors. However, the accurate readout of the recognition event of DNA probe binding to the specific target by label-free means is still challenging. Here, a simple and label-free electrochemiluminescence (ECL) method for sensing the recognition event of DNA probe to sequence-specific DNA is developed. Oxalate is used as an ECL co-reactant and p53 tumor suppressor gene as a model of target analyte. In the ECL sensing platform, the nanochannel structural film, which contains silica-sol, chitosan and Ru(bpy)3(2+), is prepared by an electrochemical deposition method. Then, DNA probes are attached onto the surface of the nanochannel-based composite film electrode based on the stronger interaction between DNA probes and chitosan embedded in the ECL composite film. These nanochannels were capped by the DNA probes. As a result, the mass-transfer channel between the Ru(bpy)3(2+) embedded in the nanochannel-based composite film and the ECL co-reactant in the bulk solution was greatly blocked and a weak ECL signal was observed. Conversely, in the presence of target sequences, the hybridizing reaction of targets with DNA probes could result in the escape of the DNA probes from the composite film due to the rigid structure of the duplex DNA. Thus, these nanochannels were uncapped and a stronger ECL signal was detected. Our results show that this ECL method could effectively discriminate complementary from single-base mismatch DNA sequences. Under the optimal conditions, the linear range for target DNA was from 1.0 × 10(-11) to 1.0 × 10(-9) mol L(-1) with a detect limit of 2.7 × 10(-12) mol L(-1). This work demonstrates that porous structures on the silica-chitosan composite film can provide a label-free and general platform to measure the change of DNA configuration.


Asunto(s)
Técnicas Biosensibles/métodos , Sondas de ADN/química , ADN de Cadena Simple/química , Técnicas Electroquímicas/métodos , Canales Iónicos/química , Mediciones Luminiscentes/métodos , Secuencia de Bases , Técnicas Biosensibles/instrumentación , Quitosano/química , Técnicas Electroquímicas/instrumentación , Electrodos , Mediciones Luminiscentes/instrumentación , Nanoporos , Compuestos Organometálicos/química , Dióxido de Silicio/química
16.
ACS Appl Mater Interfaces ; 5(16): 7931-6, 2013 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-23844847

RESUMEN

In this work, the synthetic alumina nanochannels with bi-, tri-, and tetra-branched geometry structures exhibited ionic current rectifications with nonlinear I-V curves. Such diode performance of the branched alumina nanochannel is mainly dependent on the cooperative asymmetry of the branched structure and the surface-charge distribution on inner walls. By regulating the geometry, electrolyte pH, and solution concentration, the tunable ionic rectification properties are effectively obtained including both the rectification ratios and the rectifying direction that were deduced from the converted ion selectivity. This nanofluidic diode may open up a new opportunity for the application of the complex nanofluidic devices in contrast to previously reported channels to provide molecular analysis, controlled mass transport, drug release, and various logic gate operations.


Asunto(s)
Óxido de Aluminio/química , Sistemas de Liberación de Medicamentos , Nanoestructuras/química , Humanos , Concentración de Iones de Hidrógeno , Canales Iónicos/química , Iones/química
17.
Nanoscale ; 5(14): 6224-42, 2013 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-23743952

RESUMEN

Gold nanoparticles (AuNPs) are widely used as contrast agents in electron microscopy as well as for diagnostic tests. Due to their unique optical and electrical properties and their small size, there is also a growing field of potential applications in medical fields of imaging and therapy, for example as drug carriers or as active compounds in thermotherapy. Besides their intrinsic optical properties, facile surface decoration with (bio)functional ligands renders AuNPs ideally suited for many industrial and medical applications. However, novel AuNPs may have toxicological profiles differing from bulk and therefore a thorough analysis of the quantitative structure-activity relationship (QSAR) is required. Several mechanisms are proposed that cause adverse effects of nanoparticles in biological systems. Catalytic generation of reactive species due to the large and chemically active surface area of nanomaterials is well established. Because nanoparticles approach the size of biological molecules and subcellular structures, they may overcome natural barriers by active or passive uptake. Ultrasmall AuNPs with sizes of 2 nm or less may even behave as molecular ligands. These types of potential interactions would imply a size and ligand-dependent behaviour of any nanomaterial towards biological systems. Thus, to fully understand their QSAR, AuNPs bioactivity should be analysed in biological systems of increasing complexity ranging from cell culture to whole animal studies.


Asunto(s)
Medios de Contraste/síntesis química , Oro/química , Nanopartículas del Metal/química , Animales , Barrera Hematoencefálica/metabolismo , Medios de Contraste/farmacocinética , Medios de Contraste/uso terapéutico , ADN/química , ADN/metabolismo , Portadores de Fármacos/química , Humanos , Canales Iónicos/química , Canales Iónicos/metabolismo , Nanopartículas del Metal/uso terapéutico , Neoplasias/tratamiento farmacológico , Relación Estructura-Actividad Cuantitativa , Distribución Tisular
18.
Small ; 8(7): 1001-5, 2012 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22311804

RESUMEN

The nanochannel (in a porous layer) and ion-channel (in a barrier layer) hybrid structure of anodic alumina is used as a protein-trapping device. The transmembrane potential drives the electromigration of the charged proteins (FITC-labeled) into the nanochannels, but electromigration across the barrier layer is impossible due to the size-exclusion effect. As a result, the proteins can be continuously trapped in the nanochannels.


Asunto(s)
Óxido de Aluminio/química , Canales Iónicos/química , Nanotecnología/métodos , Nanotubos/química , Proteínas/química
19.
J Gen Physiol ; 139(1): 3-17, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22155736

RESUMEN

Voltage-gated ion channels are crucial for electrical activity and chemical signaling in a variety of cell types. Structure-activity studies involving electrophysiological characterization of mutants are widely used and allow us to quickly realize the energetic effects of a mutation by measuring macroscopic currents and fitting the observed voltage dependence of conductance to a Boltzmann equation. However, such an approach is somewhat limiting, principally because of the inherent assumption that the channel activation is a two-state process. In this analysis, we show that the area delineated by the gating charge displacement curve and its ordinate axis is related to the free energy of activation of a voltage-gated ion channel. We derive a parameter, the median voltage of charge transfer (V(m)), which is proportional to this area, and prove that the chemical component of free energy change of a system can be obtained from the knowledge of V(m) and the maximum number of charges transferred. Our method is not constrained by the number or connectivity of intermediate states and is applicable to instances in which the observed responses show a multiphasic behavior. We consider various models of ion channel gating with voltage-dependent steps, latent charge movement, inactivation, etc. and discuss the applicability of this approach in each case. Notably, our method estimates a net free energy change of approximately -14 kcal/mol associated with the full-scale activation of the Shaker potassium channel, in contrast to -2 to -3 kcal/mol estimated from a single Boltzmann fit. Our estimate of the net free energy change in the system is consistent with those derived from detailed kinetic models (Zagotta et al. 1994. J. Gen. Physiol. doi:10.1085/jgp.103.2.321). The median voltage method can reliably quantify the magnitude of free energy change associated with activation of a voltage-dependent system from macroscopic equilibrium measurements. This will be particularly useful in scanning mutagenesis experiments.


Asunto(s)
Potenciales de la Membrana/fisiología , Proteínas Musculares/metabolismo , Canales de Sodio/metabolismo , Animales , ADN Complementario/metabolismo , Canales Iónicos/química , Canales Iónicos/genética , Canales Iónicos/metabolismo , Proteínas Musculares/química , Proteínas Musculares/genética , Oocitos/metabolismo , Ratas , Canales de Sodio/química , Canales de Sodio/genética , Termodinámica , Xenopus laevis/metabolismo
20.
Biomed Microdevices ; 13(4): 671-82, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21487665

RESUMEN

Solid-state nanopore sensors are highly versatile platforms for the rapid, label-free electrical detection and analysis of single molecules, applicable to next generation DNA sequencing. The versatility of this technology allows for both large scale device integration and interfacing with biological systems. Here we report on the development of a hybrid biological solid-state nanopore platform that incorporates a highly mobile lipid bilayer on a single solid-state Al(2)O(3) nanopore sensor, for the potential reconstitution of ion channels and biological nanopores. Such a system seeks to combine the superior electrical, thermal, and mechanical stability of Al(2)O(3) solid-state nanopores with the chemical specificity of biological nanopores. Bilayers on Al(2)O(3) exhibit higher diffusivity than those formed on TiO(2) and SiO(2) substrates, attributed to the presence of a thick hydration layer on Al(2)O(3), a key requirement to preserving the biological functionality of reconstituted membrane proteins. Molecular dynamics simulations demonstrate that the electrostatic repulsion between the dipole of the DOPC headgroup and the positively charged Al(2)O(3) surface may be responsible for the enhanced thickness of this hydration layer. Lipid bilayer coated Al(2)O(3) nanopore sensors exhibit excellent electrical properties and enhanced mechanical stability (GΩ seals for over 50 h), making this technology ideal for use in ion channel electrophysiology, the screening of ion channel active drugs and future integration with biological nanopores such as α-hemolysin and MspA for rapid single molecule DNA sequencing. This technology can find broad application in bio-nanotechnology.


Asunto(s)
Óxido de Aluminio/química , Técnicas Biosensibles , Membrana Dobles de Lípidos/química , Nanoporos , Óxido de Aluminio/síntesis química , ADN/química , Recuperación de Fluorescencia tras Fotoblanqueo/métodos , Canales Iónicos/química , Membrana Dobles de Lípidos/síntesis química , Modelos Biológicos , Simulación de Dinámica Molecular , Nanotecnología , Fosfatidilcolinas/química , Análisis de Secuencia de ADN , Dióxido de Silicio/química , Coloración y Etiquetado , Titanio/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA