Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Struct Biol ; 197(1): 3-12, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-26873782

RESUMO

Single-molecule force spectroscopy sheds light onto the free energy landscapes governing protein folding and molecular recognition. Since only a single molecule or single molecular complex is probed at any given point in time, the technique is capable of identifying low-probability conformations within a large ensemble of possibilities. It furthermore allows choosing certain unbinding pathways through careful selection of the points at which the force acts on the protein or molecular complex. This review focuses on recent innovations in construct design, site-specific bioconjugation, measurement techniques, instrumental advances, and data analysis methods for improving workflow, throughput, and data yield of AFM-based single-molecule force spectroscopy experiments. Current trends that we highlight include customized fingerprint domains, peptide tags for site-specific covalent surface attachment, and polyproteins that are formed through mechanostable receptor-ligand interactions. Recent methods to improve measurement stability, signal-to-noise ratio, and force precision are presented, and theoretical considerations, analysis methods, and algorithms for analyzing large numbers of force-extension curves are further discussed. The various innovations identified here will serve as a starting point to researchers in the field looking for opportunities to push the limits of the technique further.


Assuntos
Peptídeos/química , Poliproteínas/química , Dobramento de Proteína , Imagem Individual de Molécula/métodos , Algoritmos , Microscopia de Força Atômica , Poliproteínas/ultraestrutura
2.
Nat Methods ; 11(11): 1127-1130, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25194847

RESUMO

Single-molecule force spectroscopy enables mechanical testing of individual proteins, but low experimental throughput limits the ability to screen constructs in parallel. We describe a microfluidic platform for on-chip expression, covalent surface attachment and measurement of single-molecule protein mechanical properties. A dockerin tag on each protein molecule allowed us to perform thousands of pulling cycles using a single cohesin-modified cantilever. The ability to synthesize and mechanically probe protein libraries enables high-throughput mechanical phenotyping.


Assuntos
Técnicas Analíticas Microfluídicas , Análise de Sequência com Séries de Oligonucleotídeos , Análise Serial de Proteínas/métodos , Clostridium thermocellum/genética , Ensaios de Triagem em Larga Escala , Microscopia de Força Atômica/métodos , Biblioteca de Peptídeos
3.
Nanoscale ; 11(2): 407-411, 2019 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-30604815

RESUMO

Single-molecule cut-and-paste facilitates bottom-up directed assembly of nanoscale biomolecular networks in defined geometries and enables analysis with spatio-temporal resolution. However, arrangement of diverse molecules of interest requires versatile handling systems. The novel DNA-free, genetically encodable scheme described here utilises an orthogonal handling strategy to promote arrangement of enzymes and enzyme networks.


Assuntos
Enzimas Imobilizadas/química , Nanoestruturas/química , Nanotecnologia/métodos , Enzimas Imobilizadas/metabolismo , Corantes Fluorescentes , Proteínas Imobilizadas/química , Proteínas Imobilizadas/metabolismo , Microscopia de Força Atômica , Microscopia de Fluorescência , Modelos Moleculares , Nanoestruturas/ultraestrutura
4.
ACS Nano ; 12(7): 6734-6747, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-29851456

RESUMO

The mechanical properties of DNA nanostructures are of widespread interest as applications that exploit their stability under constant or intermittent external forces become increasingly common. We explore the force response of DNA origami in comprehensive detail by combining AFM single molecule force spectroscopy experiments with simulations using oxDNA, a coarse-grained model of DNA at the nucleotide level, to study the unravelling of an iconic origami system: the Rothemund tile. We contrast the force-induced melting of the tile with simulations of an origami 10-helix bundle. Finally, we simulate a recently proposed origami biosensor, whose function takes advantage of origami behavior under tension. We observe characteristic stick-slip unfolding dynamics in our force-extension curves for both the Rothemund tile and the helix bundle and reasonable agreement with experimentally observed rupture forces for these systems. Our results highlight the effect of design on force response: we observe regular, modular unfolding for the Rothemund tile that contrasts with strain-softening of the 10-helix bundle which leads to catastropic failure under monotonically increasing force. Further, unravelling occurs straightforwardly from the scaffold ends inward for the Rothemund tile, while the helix bundle unfolds more nonlinearly. The detailed visualization of the yielding events provided by simulation allows preferred pathways through the complex unfolding free-energy landscape to be mapped, as a key factor in determining relative barrier heights is the extensional release per base pair broken. We shed light on two important questions: how stable DNA nanostructures are under external forces and what design principles can be applied to enhance stability.


Assuntos
DNA/química , Nanoestruturas/química , Pareamento de Bases , Fenômenos Biomecânicos , Técnicas Biossensoriais , Microscopia de Força Atômica , Simulação de Dinâmica Molecular , Nanoestruturas/ultraestrutura , Nanotecnologia/métodos , Conformação de Ácido Nucleico , Estresse Mecânico
5.
ACS Nano ; 11(6): 6346-6354, 2017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28591514

RESUMO

Single-molecule force spectroscopy (SMFS) is by now well established as a standard technique in biophysics and mechanobiology. In recent years, the technique has benefitted greatly from new approaches to bioconjugation of proteins to surfaces. Indeed, optimized immobilization strategies for biomolecules and refined purification schemes are being steadily adapted and improved, which in turn has enhanced data quality. In many previously reported SMFS studies, poly(ethylene glycol) (PEG) was used to anchor molecules of interest to surfaces and/or cantilever tips. The limitation, however, is that PEG exhibits a well-known trans-trans-gauche to all-trans transition, which results in marked deviation from standard polymer elasticity models such as the worm-like chain, particularly at elevated forces. As a result, the assignment of unfolding events to protein domains based on their corresponding amino acid chain lengths is significantly obscured. Here, we provide a solution to this problem by implementing unstructured elastin-like polypeptides as linkers to replace PEG. We investigate the suitability of tailored elastin-like polypeptides linkers and perform direct comparisons to PEG, focusing on attributes that are critical for single-molecule force experiments such as linker length, monodispersity, and bioorthogonal conjugation tags. Our results demonstrate that by avoiding the ambiguous elastic response of mixed PEG/peptide systems and instead building the molecular mechanical systems with only a single bond type with uniform elastic properties, we improve data quality and facilitate data analysis and interpretation in force spectroscopy experiments. The use of all-peptide linkers allows alternative approaches for precisely defining elastic properties of proteins linked to surfaces.


Assuntos
Elastina/química , Peptídeos/química , Imagem Individual de Molécula/métodos , Aminoácidos/química , Fenômenos Biomecânicos , Elasticidade , Escherichia coli/genética , Proteínas Imobilizadas/química , Polietilenoglicóis/química , Conformação Proteica , Desdobramento de Proteína
6.
Elife ; 42015 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-26519733

RESUMO

Receptor-ligand pairs are ordinarily thought to interact through a lock and key mechanism, where a unique molecular conformation is formed upon binding. Contrary to this paradigm, cellulosomal cohesin-dockerin (Coh-Doc) pairs are believed to interact through redundant dual binding modes consisting of two distinct conformations. Here, we combined site-directed mutagenesis and single-molecule force spectroscopy (SMFS) to study the unbinding of Coh:Doc complexes under force. We designed Doc mutations to knock out each binding mode, and compared their single-molecule unfolding patterns as they were dissociated from Coh using an atomic force microscope (AFM) cantilever. Although average bulk measurements were unable to resolve the differences in Doc binding modes due to the similarity of the interactions, with a single-molecule method we were able to discriminate the two modes based on distinct differences in their mechanical properties. We conclude that under native conditions wild-type Doc from Clostridium thermocellum exocellulase Cel48S populates both binding modes with similar probabilities. Given the vast number of Doc domains with predicted dual binding modes across multiple bacterial species, our approach opens up new possibilities for understanding assembly and catalytic properties of a broad range of multi-enzyme complexes.


Assuntos
Celulossomas/química , Celulossomas/metabolismo , Clostridium thermocellum/enzimologia , Celulossomas/genética , Clostridium thermocellum/genética , Microscopia de Força Atômica , Modelos Moleculares , Mutagênese Sítio-Dirigida , Ligação Proteica , Conformação Proteica , Análise Espectral
7.
Nat Commun ; 5: 5635, 2014 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-25482395

RESUMO

Challenging environments have guided nature in the development of ultrastable protein complexes. Specialized bacteria produce discrete multi-component protein networks called cellulosomes to effectively digest lignocellulosic biomass. While network assembly is enabled by protein interactions with commonplace affinities, we show that certain cellulosomal ligand-receptor interactions exhibit extreme resistance to applied force. Here, we characterize the ligand-receptor complex responsible for substrate anchoring in the Ruminococcus flavefaciens cellulosome using single-molecule force spectroscopy and steered molecular dynamics simulations. The complex withstands forces of 600-750 pN, making it one of the strongest bimolecular interactions reported, equivalent to half the mechanical strength of a covalent bond. Our findings demonstrate force activation and inter-domain stabilization of the complex, and suggest that certain network components serve as mechanical effectors for maintaining network integrity. This detailed understanding of cellulosomal network components may help in the development of biocatalysts for production of fuels and chemicals from renewable plant-derived biomass.


Assuntos
Celulossomas/química , Ruminococcus/química , Biomassa , Biofísica , Cálcio/química , Catálise , Adesão Celular , Simulação por Computador , Ligação de Hidrogênio , Íons , Ligantes , Microscopia de Força Atômica , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Distribuição Normal , Ligação Proteica , Conformação Proteica , Dobramento de Proteína
8.
J Vis Exp ; (82): e50950, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-24378772

RESUMO

Cellulosomes are discrete multienzyme complexes used by a subset of anaerobic bacteria and fungi to digest lignocellulosic substrates. Assembly of the enzymes onto the noncatalytic scaffold protein is directed by interactions among a family of related receptor-ligand pairs comprising interacting cohesin and dockerin modules. The extremely strong binding between cohesin and dockerin modules results in dissociation constants in the low picomolar to nanomolar range, which may hamper accurate off-rate measurements with conventional bulk methods. Single-molecule force spectroscopy (SMFS) with the atomic force microscope measures the response of individual biomolecules to force, and in contrast to other single-molecule manipulation methods (i.e. optical tweezers), is optimal for studying high-affinity receptor-ligand interactions because of its ability to probe the high-force regime (>120 pN). Here we present our complete protocol for studying cellulosomal protein assemblies at the single-molecule level. Using a protein topology derived from the native cellulosome, we worked with enzyme-dockerin and carbohydrate binding module-cohesin (CBM-cohesin) fusion proteins, each with an accessible free thiol group at an engineered cysteine residue. We present our site-specific surface immobilization protocol, along with our measurement and data analysis procedure for obtaining detailed binding parameters for the high-affinity complex. We demonstrate how to quantify single subdomain unfolding forces, complex rupture forces, kinetic off-rates, and potential widths of the binding well. The successful application of these methods in characterizing the cohesin-dockerin interaction responsible for assembly of multidomain cellulolytic complexes is further described.


Assuntos
Proteínas de Ciclo Celular/química , Celulossomas/química , Proteínas Cromossômicas não Histona/química , Proteínas Imobilizadas/química , Microscopia de Força Atômica/métodos , Complexos Multienzimáticos/química , Receptores de Superfície Celular/química , Proteínas de Ciclo Celular/metabolismo , Celulossomas/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas Imobilizadas/metabolismo , Ligantes , Complexos Multienzimáticos/metabolismo , Receptores de Superfície Celular/metabolismo , Coesinas
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa