Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 41(16): e110410, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35698800

RESUMEN

Although amyloid fibres are highly stable protein aggregates, a specific combination of human Hsp70 system chaperones can disassemble them, including fibres formed of α-synuclein, huntingtin, or Tau. Disaggregation requires the ATPase activity of the constitutively expressed Hsp70 family member, Hsc70, together with the J domain protein DNAJB1 and the nucleotide exchange factor Apg2. Clustering of Hsc70 on the fibrils appears to be necessary for disassembly. Here we use atomic force microscopy to show that segments of in vitro assembled α-synuclein fibrils are first coated with chaperones and then undergo bursts of rapid, unidirectional disassembly. Cryo-electron tomography and total internal reflection fluorescence microscopy reveal fibrils with regions of densely bound chaperones, preferentially at one end of the fibre. Sub-stoichiometric amounts of Apg2 relative to Hsc70 dramatically increase recruitment of Hsc70 to the fibres, creating localised active zones that then undergo rapid disassembly at a rate of ~ 4 subunits per second. The observed unidirectional bursts of Hsc70 loading and unravelling may be explained by differences between the two ends of the polar fibre structure.


Asunto(s)
Proteínas HSP70 de Choque Térmico , alfa-Sinucleína , Amiloide/metabolismo , Proteínas Amiloidogénicas/metabolismo , Proteínas del Choque Térmico HSC70/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Agregado de Proteínas , Unión Proteica , alfa-Sinucleína/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34716276

RESUMEN

Gram-negative bacteria are surrounded by a protective outer membrane (OM) with phospholipids in its inner leaflet and lipopolysaccharides (LPS) in its outer leaflet. The OM is also populated with many ß-barrel outer-membrane proteins (OMPs), some of which have been shown to cluster into supramolecular assemblies. However, it remains unknown how abundant OMPs are organized across the entire bacterial surface and how this relates to the lipids in the membrane. Here, we reveal how the OM is organized from molecular to cellular length scales, using atomic force microscopy to visualize the OM of live bacteria, including engineered Escherichia coli strains and complemented by specific labeling of abundant OMPs. We find that a predominant OMP in the E. coli OM, the porin OmpF, forms a near-static network across the surface, which is interspersed with barren patches of LPS that grow and merge with other patches during cell elongation. Embedded within the porin network is OmpA, which forms noncovalent interactions to the underlying cell wall. When the OM is destabilized by mislocalization of phospholipids to the outer leaflet, a new phase appears, correlating with bacterial sensitivity to harsh environments. We conclude that the OM is a mosaic of phase-separated LPS-rich and OMP-rich regions, the maintenance of which is essential to the integrity of the membrane and hence to the lifestyle of a gram-negative bacterium.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Membrana Externa Bacteriana/metabolismo , Condensados Biomoleculares/fisiología , Membrana Externa Bacteriana/fisiología , Membrana Celular/metabolismo , Pared Celular/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Membrana Dobles de Lípidos/metabolismo , Lipopolisacáridos/metabolismo , Simulación de Dinámica Molecular , Fosfolípidos/metabolismo , Porinas/metabolismo
3.
EMBO J ; 38(4)2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30643019

RESUMEN

The immune system kills bacteria by the formation of lytic membrane attack complexes (MACs), triggered when complement enzymes cleave C5. At present, it is not understood how the MAC perturbs the composite cell envelope of Gram-negative bacteria. Here, we show that the role of C5 convertase enzymes in MAC assembly extends beyond the cleavage of C5 into the MAC precursor C5b. Although purified MAC complexes generated from preassembled C5b6 perforate artificial lipid membranes and mammalian cells, these components lack bactericidal activity. In order to permeabilize both the bacterial outer and inner membrane and thus kill a bacterium, MACs need to be assembled locally by the C5 convertase enzymes. Our data indicate that C5b6 rapidly loses the capacity to form bactericidal pores; therefore, bacterial killing requires both in situ conversion of C5 and immediate insertion of C5b67 into the membrane. Using flow cytometry and atomic force microscopy, we show that local assembly of C5b6 at the bacterial surface is required for the efficient insertion of MAC pores into bacterial membranes. These studies provide basic molecular insights into MAC assembly and bacterial killing by the immune system.


Asunto(s)
Actividad Bactericida de la Sangre , Membrana Celular/metabolismo , Convertasas de Complemento C3-C5/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Bacterias Gramnegativas/crecimiento & desarrollo , Hemólisis , Permeabilidad de la Membrana Celular , Activación de Complemento , Bacterias Gramnegativas/metabolismo , Humanos
4.
PLoS Pathog ; 16(6): e1008606, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32569291

RESUMEN

An important effector function of the human complement system is to directly kill Gram-negative bacteria via Membrane Attack Complex (MAC) pores. MAC pores are assembled when surface-bound convertase enzymes convert C5 into C5b, which together with C6, C7, C8 and multiple copies of C9 forms a transmembrane pore that damages the bacterial cell envelope. Recently, we found that bacterial killing by MAC pores requires local conversion of C5 by surface-bound convertases. In this study we aimed to understand why local assembly of MAC pores is essential for bacterial killing. Here, we show that rapid interaction of C7 with C5b6 is required to form bactericidal MAC pores on Escherichia coli. Binding experiments with fluorescently labelled C6 show that C7 prevents release of C5b6 from the bacterial surface. Moreover, trypsin shaving experiments and atomic force microscopy revealed that this rapid interaction between C7 and C5b6 is crucial to efficiently anchor C5b-7 to the bacterial cell envelope and form complete MAC pores. Using complement-resistant clinical E. coli strains, we show that bacterial pathogens can prevent complement-dependent killing by interfering with the anchoring of C5b-7. While C5 convertase assembly was unaffected, these resistant strains blocked efficient anchoring of C5b-7 and thus prevented stable insertion of MAC pores into the bacterial cell envelope. Altogether, these findings provide basic molecular insights into how bactericidal MAC pores are assembled and how bacteria evade MAC-dependent killing.


Asunto(s)
Actividad Bactericida de la Sangre , Membrana Celular/metabolismo , Pared Celular/metabolismo , Complemento C5/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Escherichia coli/metabolismo , Proteínas del Sistema Complemento/metabolismo , Células HEK293 , Humanos
5.
Methods ; 193: 68-79, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33548405

RESUMEN

We present TopoStats, a Python toolkit for automated editing and analysis of Atomic Force Microscopy images. The program automates identification and tracing of individual molecules in circular and linear conformations without user input. TopoStats was able to identify and trace a range of molecules within AFM images, finding, on average, ~90% of all individual molecules and molecular assemblies within a wide field of view, and without the need for prior processing. DNA minicircles of varying size, DNA origami rings and pore forming proteins were identified and accurately traced with contour lengths of traces typically within 10 nm of the predicted contour length. TopoStats was also able to reliably identify and trace linear and enclosed circular molecules within a mixed population. The program is freely available via GitHub (https://github.com/afm-spm/TopoStats) and is intended to be modified and adapted for use if required.


Asunto(s)
Microscopía de Fuerza Atómica , Automatización de Laboratorios , ADN
6.
Biophys J ; 120(9): 1565-1577, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33617830

RESUMEN

In the nuclear pore complex, intrinsically disordered proteins (FG Nups), along with their interactions with more globular proteins called nuclear transport receptors (NTRs), are vital to the selectivity of transport into and out of the cell nucleus. Although such interactions can be modeled at different levels of coarse graining, in vitro experimental data have been quantitatively described by minimal models that describe FG Nups as cohesive homogeneous polymers and NTRs as uniformly cohesive spheres, in which the heterogeneous effects have been smeared out. By definition, these minimal models do not account for the explicit heterogeneities in FG Nup sequences, essentially a string of cohesive and noncohesive polymer units, and at the NTR surface. Here, we develop computational and analytical models that do take into account such heterogeneity in a minimal fashion and compare them with experimental data on single-molecule interactions between FG Nups and NTRs. Overall, we find that the heterogeneous nature of FG Nups and NTRs does play a role in determining equilibrium binding properties but is of much greater significance when it comes to unbinding and binding kinetics. Using our models, we predict how binding equilibria and kinetics depend on the distribution of cohesive blocks in the FG Nup sequences and of the binding pockets at the NTR surface, with multivalency playing a key role. Finally, we observe that single-molecule binding kinetics has a rather minor influence on the diffusion of NTRs in polymer melts consisting of FG-Nup-like sequences.


Asunto(s)
Proteínas Intrínsecamente Desordenadas , Poro Nuclear , Transporte Activo de Núcleo Celular , Proteínas Intrínsecamente Desordenadas/metabolismo , Cinética , Poro Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo
7.
Phys Rep ; 921: 1-53, 2021 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-35892075

RESUMEN

The hallmark of eukaryotic cells is the nucleus that contains the genome, enclosed by a physical barrier known as the nuclear envelope (NE). On the one hand, this compartmentalization endows the eukaryotic cells with high regulatory complexity and flexibility. On the other hand, it poses a tremendous logistic and energetic problem of transporting millions of molecules per second across the nuclear envelope, to facilitate their biological function in all compartments of the cell. Therefore, eukaryotes have evolved a molecular "nanomachine" known as the Nuclear Pore Complex (NPC). Embedded in the nuclear envelope, NPCs control and regulate all the bi-directional transport between the cell nucleus and the cytoplasm. NPCs combine high molecular specificity of transport with high throughput and speed, and are highly robust with respect to molecular noise and structural perturbations. Remarkably, the functional mechanisms of NPC transport are highly conserved among eukaryotes, from yeast to humans, despite significant differences in the molecular components among various species. The NPC is the largest macromolecular complex in the cell. Yet, despite its significant complexity, it has become clear that its principles of operation can be largely understood based on fundamental physical concepts, as have emerged from a combination of experimental methods of molecular cell biology, biophysics, nanoscience and theoretical and computational modeling. Indeed, many aspects of NPC function can be recapitulated in artificial mimics with a drastically reduced complexity compared to biological pores. We review the current physical understanding of the NPC architecture and function, with the focus on the critical analysis of experimental studies in cells and artificial NPC mimics through the lens of theoretical and computational models. We also discuss the connections between the emerging concepts of NPC operation and other areas of biophysics and bionanotechnology.

8.
Faraday Discuss ; 232(0): 236-255, 2021 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-34545865

RESUMEN

Perforin is a pore forming protein used by cytotoxic T lymphocytes to remove cancerous or virus-infected cells during the immune response. During the response, the lymphocyte membrane becomes refractory to perforin function by accumulating densely ordered lipid rafts and externalizing negatively charged lipid species. The dense membrane packing lowers the capacity of perforin to bind, and the negatively charged lipids scavenge any residual protein before pore formation. Using atomic force microscopy on model membrane systems, we here provide insight into the molecular basis of perforin lipid specificity.


Asunto(s)
Lípidos , Linfocitos T Citotóxicos , Perforina , Proteínas Citotóxicas Formadoras de Poros
9.
Anal Chem ; 92(5): 4082-4093, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-31995983

RESUMEN

Exosomes are endocytic lipid-membrane bound bodies with the potential to be used as biomarkers in cancer and neurodegenerative disease. The limitations and scarcity of current exosome characterization approaches have led to a growing demand for translational techniques, capable of determining their molecular composition and physical properties in physiological fluids. Here, we investigate label-free immunosensing, using a quartz crystal microbalance with dissipation monitoring (QCM-D), to detect exosomes by exploiting their surface protein profile. Exosomes expressing the transmembrane protein CD63 were isolated by size-exclusion chromatography from cell culture media. QCM-D sensors functionalized with anti-CD63 antibodies formed a direct immunoassay toward CD63-positive exosomes in 75% v/v serum, exhibiting a limit-of-detection of 2.9 × 108 and 1.4 × 108 exosome sized particles (ESPs)/mL for frequency and dissipation response, respectively, i.e., clinically relevant concentrations. Our proof-of-concept findings support the adoption of dual-mode acoustic analysis of exosomes, leveraging both frequency and dissipation monitoring for use in bioanalytical characterization.


Asunto(s)
Exosomas/química , Tecnicas de Microbalanza del Cristal de Cuarzo/métodos , Anticuerpos/inmunología , Exosomas/metabolismo , Humanos , Inmunoensayo , Límite de Detección , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Nanopartículas/química , Nanopartículas/metabolismo , Tetraspanina 30/metabolismo
10.
Semin Cell Dev Biol ; 68: 42-51, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28506890

RESUMEN

The nuclear pore complex (NPC) is the selective gateway through which all molecules must pass when entering or exiting the nucleus. It is a cog in the gene expression pathway, an entrance to the nucleus exploited by viruses, and a highly-tuned nanoscale filter. The NPC is a large proteinaceous assembly with a central lumen occluded by natively disordered proteins, known as FG-nucleoporins (or FG-nups). These FG-nups, along with a family of soluble proteins known as nuclear transport receptors (NTRs), form the selective transport barrier. Although much is known about the transport cycle and the necessity of NTRs for chaperoning cargo molecules through the NPC, the mechanism by which NTRs and NTR•cargo complexes translocate the selective transport barrier is not well understood. How can disordered FG-nups and soluble NTRs form a transport barrier that is selective, ATP-free, and fast? In this work, we review various mechanical approaches - both experimental and theoretical/computational - employed to better understand the morphology of the FG-nups, and their role in nucleocytoplasmic transport. Recent experiments on FG-nups tethered to planar surfaces, coupled with quantitative modelling work suggests that FG-nup morphologies are the result of a finely balanced system with significant contributions from FG-nup cohesiveness and entropic repulsion, and from NTR•FG-nup binding avidity; whilst AFM experiments on intact NPCs suggest that the FG-nups are sufficiently cohesive to form condensates in the centre of the NPC lumen, which may transiently dissolve to facilitate the transport of larger cargoes.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Proteínas de Complejo Poro Nuclear/metabolismo , Poro Nuclear/metabolismo , Humanos
11.
Analyst ; 144(23): 6944-6952, 2019 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-31620716

RESUMEN

Atomic force microscopy (AFM) provides an effective, label-free technique enabling the imaging of live bacteria under physiological conditions with nanometre precision. However, AFM is a surface scanning technique, and the accuracy of its performance requires the effective and reliable immobilisation of bacterial cells onto substrates. Here, we compare the effectiveness of various chemical approaches to facilitate the immobilisation of Escherichia coli onto glass cover slips in terms of bacterial adsorption, viability and compatibility with correlative imaging by fluorescence microscopy. We assess surface functionalisation using gelatin, poly-l-lysine, Cell-Tak™, and Vectabond®. We describe how bacterial immobilisation, viability and suitability for AFM experiments depend on bacterial strain, buffer conditions and surface functionalisation. We demonstrate the use of such immobilisation by AFM images that resolve the porin lattice on the bacterial surface; local degradation of the bacterial cell envelope by an antimicrobial peptide (Cecropin B); and the formation of membrane attack complexes on the bacterial membrane.


Asunto(s)
Células Inmovilizadas/ultraestructura , Escherichia coli/ultraestructura , Microscopía de Fuerza Atómica/métodos , Tampones (Química) , Adhesión Celular , Membrana Celular/metabolismo , Gelatina/química , Polilisina/química , Porinas/metabolismo , Propilaminas/química , Silanos/química
12.
J Cell Sci ; 129(11): 2125-33, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27179071

RESUMEN

The membrane attack complex and perforin proteins (MACPFs) and bacterial cholesterol-dependent cytolysins (CDCs) are two branches of a large and diverse superfamily of pore-forming proteins that function in immunity and pathogenesis. During pore formation, soluble monomers assemble into large transmembrane pores through conformational transitions that involve extrusion and refolding of two α-helical regions into transmembrane ß-hairpins. These transitions entail a dramatic refolding of the protein structure, and the resulting assemblies create large holes in cellular membranes, but they do not use any external source of energy. Structures of the membrane-bound assemblies are required to mechanistically understand and modulate these processes. In this Commentary, we discuss recent advances in the understanding of assembly mechanisms and molecular details of the conformational changes that occur during MACPF and CDC pore formation.


Asunto(s)
Colesterol/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Citotoxinas/metabolismo , Perforina/metabolismo , Animales , Complejo de Ataque a Membrana del Sistema Complemento/química , Citotoxinas/química , Humanos , Modelos Moleculares , Complejos Multiproteicos/metabolismo , Perforina/química
13.
Phys Biol ; 15(6): 060201, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29863490

RESUMEN

Increasing numbers of physicists engage in research activities that address biological questions from physics perspectives or strive to develop physics insights from active biological processes. The on-going development and success of such activities morph our ways of thinking about what it is to 'do biophysics' and add to our understanding of the physics of life. Many scientists in this research and teaching landscape are homed in physics departments. A challenge for a hosting department is how to group, name and structure such biophysicists to best add value to their emerging research and teaching but also to the portfolio of the whole department. Here we discuss these issues and speculate on strategies.


Asunto(s)
Biofisica/organización & administración , Universidades/organización & administración , Investigación/organización & administración
15.
Proc Natl Acad Sci U S A ; 110(22): 8918-23, 2013 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-23671080

RESUMEN

Antimicrobial peptides are postulated to disrupt microbial phospholipid membranes. The prevailing molecular model is based on the formation of stable or transient pores although the direct observation of the fundamental processes is lacking. By combining rational peptide design with topographical (atomic force microscopy) and chemical (nanoscale secondary ion mass spectrometry) imaging on the same samples, we show that pores formed by antimicrobial peptides in supported lipid bilayers are not necessarily limited to a particular diameter, nor they are transient, but can expand laterally at the nano-to-micrometer scale to the point of complete membrane disintegration. The results offer a mechanistic basis for membrane poration as a generic physicochemical process of cooperative and continuous peptide recruitment in the available phospholipid matrix.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Membrana Dobles de Lípidos/química , Nanotecnología/métodos , Fosfolípidos/metabolismo , Secuencia de Aminoácidos , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/genética , Cromatografía Líquida de Alta Presión , Dicroismo Circular , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Microscopía de Fuerza Atómica , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Fosfolípidos/química , Ingeniería de Proteínas , Espectrometría de Masa de Ion Secundario
16.
Small ; 10(16): 3257-61, 2014 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-24740866

RESUMEN

Based on soft-touch atomic force microscopy, a method is described to reconstruct the secondary structure of single extended biomolecules, without the need for crystallization. The method is tested by accurately reproducing the dimensions of the B-DNA crystal structure. Importantly, intramolecular variations in groove depth of the DNA double helix are resolved, which would be inaccessible for methods that rely on ensemble-averaging.


Asunto(s)
ADN/química , Microscopía de Fuerza Atómica/métodos , Conformación de Ácido Nucleico , Análisis de Fourier
17.
J Mol Recognit ; 27(11): 669-75, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25277091

RESUMEN

Atomic force microscopy (AFM) is a unique tool for imaging membrane proteins in near-native environment (embedded in a membrane and in buffer solution) at ~1 nm spatial resolution. It has been most successful on membrane proteins reconstituted in 2D crystals and on some specialized and densely packed native membranes. Here, we report on AFM imaging of purified plasma membranes from Xenopus laevis oocytes, a commonly used system for the heterologous expression of membrane proteins. Isoform M23 of human aquaporin 4 (AQP4-M23) was expressed in the X. laevis oocytes following their injection with AQP4-M23 cRNA. AQP4-M23 expression and incorporation in the plasma membrane were confirmed by the changes in oocyte volume in response to applied osmotic gradients. Oocyte plasma membranes were then purified by ultracentrifugation on a discontinuous sucrose gradient, and the presence of AQP4-M23 proteins in the purified membranes was established by Western blotting analysis. Compared with membranes without over-expressed AQP4-M23, the membranes from AQP4-M23 cRNA injected oocytes showed clusters of structures with lateral size of about 10 nm in the AFM topography images, with a tendency to a fourfold symmetry as may be expected for higher-order arrays of AQP4-M23. In addition, but only infrequently, AQP4-M23 tetramers could be resolved in 2D arrays on top of the plasma membrane, in good quantitative agreement with transmission electron microscopy analysis and the current model of AQP4. Our results show the potential and the difficulties of AFM studies on cloned membrane proteins in native eukaryotic membranes.


Asunto(s)
Acuaporina 4/química , Membrana Celular/ultraestructura , Microscopía de Fuerza Atómica/métodos , Oocitos/ultraestructura , Xenopus laevis/metabolismo , Animales , Acuaporina 4/metabolismo , Western Blotting , Membrana Celular/metabolismo , Femenino , Humanos , Oocitos/citología , Oocitos/metabolismo , Xenopus laevis/crecimiento & desarrollo
18.
Biophys J ; 105(12): 2781-9, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24359750

RESUMEN

Nuclear transport receptors (NTRs) mediate nucleocytoplasmic transport via their affinity for unstructured proteins (polymers) in the nuclear pore complex (NPC). Here, we have modeled the effect of NTRs on polymeric structure in the nanopore confinement of the NPC central conduit. The model explicitly takes into account inter- and intramolecular interactions, as well as the finite size of the NTRs (∼20% of the NPC channel diameter). It reproduces various proposed scenarios for the channel structure, ranging from a central polymer condensate (selective phase) to brushlike polymer arrangements localized at the channel wall (virtual gate, reduction of dimensionality), with the transport receptors lining the polymer surface. In addition, it predicts a new structure in which NTRs become an integral part of the transport barrier by forming a cross-linked network with the unstructured proteins stretching across the pore. The model provides specific and distinctive predictions for the equilibrium spatial distributions of NTRs for these different scenarios that can be experimentally verified by, e.g., superresolution fluorescence microscopy. Moreover, it suggests mechanisms by which globular macromolecules (colloidal particles) can cause polymer-coated nanopores to switch between open and closed configurations, a possible explanation of the biological function of the NPC, and suggests potential technological applications for filtration and single-molecule sensing.


Asunto(s)
Modelos Biológicos , Poro Nuclear/química , Transporte Activo de Núcleo Celular , Animales , Poro Nuclear/metabolismo
19.
Nano Lett ; 12(7): 3846-50, 2012 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-22731615

RESUMEN

Structural variability and flexibility are crucial factors for biomolecular function. Here we have reduced the invasiness and enhanced the spatial resolution of atomic force microscopy (AFM) to visualize, for the first time, different structural conformations of the two polynucleotide strands in the DNA double helix, for single molecules under near-physiological conditions. This is achieved by identifying and tracking the anomalous resonance behavior of nanoscale AFM cantilevers in the immediate vicinity of the sample.


Asunto(s)
ADN/química , Microscopía de Fuerza Atómica , Nanoestructuras/química , Conformación de Ácido Nucleico , Plásmidos/química
20.
Sensors (Basel) ; 12(5): 6497-507, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22778654

RESUMEN

Micromechanic resonators provide a small-volume and potentially high-throughput method to determine rheological properties of fluids. Here we explore the accuracy in measuring mass density and viscosity of ethanol-water and glycerol-water model solutions, using a simple and easily implemented model to deduce the hydrodynamic effects on resonating cantilevers of various length-to-width aspect ratios. We next show that these measurements can be extended to determine the alcohol percentage of both model solutions and commercial beverages such as beer, wine and liquor. This demonstrates how micromechanical resonators can be used for quality control of every-day drinks.


Asunto(s)
Bebidas/análisis , Etanol/análisis , Modelos Teóricos , Reología , Soluciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA