Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Type of study
Publication year range
1.
Methods Mol Biol ; 834: 93-109, 2012.
Article in English | MEDLINE | ID: mdl-22144356

ABSTRACT

In recent years there has been a growing interest in the precise and concerted assembly of multiple DNA fragments of diverse sizes, including chromosomes, and the fine tuning of gene expression levels and protein activity. Commercial DNA assembly solutions have not been conceived to support the cloning of very large or very small genetic elements or a combination of both. Here we summarize a series of protocols that allow the seamless, simultaneous, flexible, and highly efficient assembly of DNA elements of a wide range of sizes (up to hundred thousand base pairs). The protocols harness the power of homologous recombination and are performed either in vitro or within the living cells. The DNA fragments may or may not share homology at their ends. An efficient site-directed mutagenesis protocol enhanced by homologous recombination is also described.


Subject(s)
Genetic Engineering/methods , Homologous Recombination , Metabolic Engineering/methods , Mutagenesis, Site-Directed , Cloning, Molecular , Gene Order , Plasmids/genetics , Saccharomyces cerevisiae/genetics
2.
Methods Enzymol ; 498: 327-48, 2011.
Article in English | MEDLINE | ID: mdl-21601684

ABSTRACT

With the completion of myriad genome sequencing projects, genetic bioengineering has expanded into many applications including the integrated analysis of complex pathways, the construction of new biological parts and the redesign of existing, natural biological systems. All these areas require the precise and concerted assembly of multiple DNA fragments of various sizes, including chromosomes, and the fine-tuning of gene expression levels and protein activity. Current commercial cloning products are not robust enough to support the assembly of very large or very small genetic elements or a combination of both. In addition, current strategies are not flexible enough to allow further modifications to the original design without having to undergo complicated cloning strategies. Here, we present a set of protocols that allow the seamless, simultaneous, flexible, and highly efficient assembly of genetic material, designed for a wide size dynamic range (10s to 100,000s base pairs). The assembly can be performed either in vitro or within the living cells and the DNA fragments may or may not share homology at their ends. A novel site-directed mutagenesis approach enhanced by in vitro recombineering is also presented.


Subject(s)
DNA/chemical synthesis , Synthetic Biology/methods , Base Sequence , DNA/genetics , Escherichia coli/genetics , Genetic Engineering/methods , Genetic Vectors , Mutagenesis, Site-Directed , Oligonucleotides/chemistry , Oligonucleotides/genetics , Recombination, Genetic , Yeasts/genetics
3.
BMC Biotechnol ; 11: 57, 2011 May 23.
Article in English | MEDLINE | ID: mdl-21605442

ABSTRACT

BACKGROUND: G protein coupled receptors (GPCRs) represent the largest family of membrane proteins in the human genome and the richest source of targets for the pharmaceutical industry. A major limitation to characterizing GPCRs has been the difficulty in developing high-level heterologous expression systems that are cost effective. Reasons for these difficulties include inefficient transport and insertion in the plasma membrane and cytotoxicity. Additionally, GPCR purification requires detergents, which have a negative effect on receptor yields and stability. RESULTS: Here we report a detergent-free cell-free protein expression-based method to obtain pharmacologically active GPCRs in about 2 hours. Our strategy relies on the co-translational insertion of modified GPCRs into nanometer-sized planar membranes. As a model we employed an engineered ß2-adrenergic receptor in which the third intracellular loop has been replaced with T4 lysozyme (ß2AR -T4L). We demonstrated that nanolipoprotein particles (NLPs) are necessary for expression of active ß2AR -T4L in cell-free systems. The binding specificity of the NLP- ß2AR-T4L complex has been determined by competitive assays. Our results demonstrate that ß2AR-T4L synthesized in vitro depends on similar oxidative conditions as those required by an in vivo-expressed receptor. CONCLUSIONS: Although the activation of ß2AR-T4L requires the insertion of the T4 lysozyme sequence and the yield of that active protein limited, our results conceptually prove that cell-free protein expression could be used as a fast approach to express these valuable and notoriously difficult-to-express proteins.


Subject(s)
Lipid Bilayers/metabolism , Receptors, Adrenergic, beta-2/biosynthesis , Adrenergic beta-2 Receptor Antagonists/pharmacology , Bacteriophage T4/enzymology , Cell-Free System , Cloning, Molecular , Dihydroalprenolol/pharmacology , Humans , Lipid Bilayers/chemistry , Muramidase/biosynthesis , Muramidase/genetics , Nanostructures/chemistry , Protein Binding , Protein Folding , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics
4.
Protein Sci ; 20(2): 437-47, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21280134

ABSTRACT

Nanolipoprotein particles (NLPs) represent a unique nanometer-sized scaffold for supporting membrane proteins (MP). Characterization of their dynamic shape and association with MP in solution remains a challenge. Here, we present a rapid method of analysis by fluorescence correlation spectroscopy (FCS) to characterize bacteriorhodopsin (bR), a membrane protein capable of forming a NLP complex. By selectively labeling individual components of NLPs during cell-free synthesis, FCS enabled us to measure specific NLP diffusion times and infer size information for different NLP species. The resulting bR-loaded NLPs were shown to be dynamically discoidal in solution with a mean diameter of 7.8 nm. The insertion rate of bR in the complex was ∼55% based on a fit model incorporating two separate diffusion properties to best approximate the FCS data. More importantly, based on these data, we infer that membrane protein associated NLPs are thermodynamically constrained as discs in solution, while empty NLPs appear to be less constrained and dynamically spherical.


Subject(s)
Bacteriorhodopsins/chemistry , Lipoproteins/chemistry , Nanoparticles/chemistry , Bacteriorhodopsins/metabolism , Cell-Free System , Diffusion , Electrophoresis, Polyacrylamide Gel , Escherichia coli , Light , Linear Models , Microscopy, Atomic Force , Molecular Biology , Nanotechnology , Particle Size , Protein Engineering , Scattering, Radiation , Spectrometry, Fluorescence , Thermodynamics
5.
Trends Biotechnol ; 27(8): 455-60, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19616329

ABSTRACT

Structural and functional studies of membrane proteins have been severely hampered by difficulties in producing sufficient quantities of properly folded protein products. It is well established that cell-based expression of membrane proteins is generally problematic and frequently results in low yield, cell toxicity, protein aggregation and misfolding. Owing to its inherent open nature, cell-free protein expression has become a highly promising tool for the fast and efficient production of these difficult-to-express proteins. Here we review the most recent advances in this field, underscoring the potentials and weaknesses of the newly developed approaches and place specific emphasis on the use of nanolipoprotein particles (NLPs or nanodiscs).


Subject(s)
Biochemistry/methods , Biotechnology/methods , Membrane Proteins/biosynthesis , Cell-Free System
6.
Methods Mol Biol ; 498: 273-96, 2009.
Article in English | MEDLINE | ID: mdl-18988032

ABSTRACT

Membrane-associated proteins and protein complexes account for approximately a third or more of the proteins in the cell (1, 2). These complexes mediate essential cellular processes; including signal transduc-tion, transport, recognition, bioenergetics and cell-cell communication. In general, membrane proteins are challenging to study because of their insolubility and tendency to aggregate when removed from their protein lipid bilayer environment. This chapter is focused on describing a novel method for producing and solubilizing membrane proteins that can be easily adapted to high-throughput expression screening. This process is based on cell-free transcription and translation technology coupled with nanolipoprotein par ticles (NLPs), which are lipid bilayers confined within a ring of amphipathic protein of defined diameter. The NLPs act as a platform for inserting, solubilizing and characterizing functional membrane proteins. NLP component proteins (apolipoproteins), as well as membrane proteins can be produced by either traditional cell-based or as discussed here, cell-free expression methodologies.


Subject(s)
Lipoproteins/metabolism , Membrane Proteins/isolation & purification , Recombinant Proteins/isolation & purification , Animals , Biotinylation , Cell Fractionation/methods , Escherichia coli/genetics , Lipoproteins/chemistry , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Nanoparticles/chemistry , Protein Array Analysis , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Solubility
8.
Mol Cell Proteomics ; 7(11): 2246-53, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18603642

ABSTRACT

Here we demonstrate rapid production of solubilized and functional membrane protein by simultaneous cell-free expression of an apolipoprotein and a membrane protein in the presence of lipids, leading to the self-assembly of membrane protein-containing nanolipoprotein particles (NLPs). NLPs have shown great promise as a biotechnology platform for solubilizing and characterizing membrane proteins. However, current approaches are limited because they require extensive efforts to express, purify, and solubilize the membrane protein prior to insertion into NLPs. By the simple addition of a few constituents to cell-free extracts, we can produce membrane proteins in NLPs with considerably less effort. For this approach an integral membrane protein and an apolipoprotein scaffold are encoded by two DNA plasmids introduced into cell-free extracts along with lipids. For this study reported here we used plasmids encoding the bacteriorhodopsin (bR) membrane apoprotein and scaffold protein Delta1-49 apolipoprotein A-I fragment (Delta49A1). Cell free co-expression of the proteins encoded by these plasmids, in the presence of the cofactor all-trans-retinal and dimyristoylphosphatidylcholine, resulted in production of functional bR as demonstrated by a 5-nm shift in the absorption spectra upon light adaptation and characteristic time-resolved FT infrared difference spectra for the bR --> M transition. Importantly the functional bR was solubilized in discoidal bR.NLPs as determined by atomic force microscopy. A survey study of other membrane proteins co-expressed with Delta49A1 scaffold protein also showed significantly increased solubility of all of the membrane proteins, indicating that this approach may provide a general method for expressing membrane proteins enabling further studies.


Subject(s)
Apolipoprotein A-I/chemistry , Membrane Proteins/chemistry , Apolipoprotein A-I/genetics , Bacteriorhodopsins/chemistry , Bacteriorhodopsins/genetics , Base Sequence , DNA Primers/genetics , Halobacterium salinarum/genetics , Membrane Proteins/genetics , Microscopy, Atomic Force , Nanoparticles/chemistry , Peptide Fragments/chemistry , Peptide Fragments/genetics , Proteomics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Solubility , Spectroscopy, Fourier Transform Infrared
9.
J Proteome Res ; 7(8): 3535-42, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18557639

ABSTRACT

We report a cell-free approach for expressing and inserting integral membrane proteins into water-soluble particles composed of discoidal apolipoprotein-lipid bilayers. Proteins are inserted into the particles, circumventing the need of extracting and reconstituting the product into membrane vesicles. Moreover, the planar nature of the membrane support makes the protein freely accessible from both sides of the lipid bilayer. Complexes are successfully purified by means of the apoplipoprotein component or by the carrier protein. The method significantly enhances the solubility of a variety of membrane proteins with different functional roles and topologies. Analytical assays for a subset of model membrane proteins indicate that proteins are correctly folded and active. The approach provides a platform amenable to high-throughput structural and functional characterization of a variety of traditionally intractable drug targets.


Subject(s)
Lipid Bilayers/chemistry , Membrane Proteins/chemistry , Antiporters/biosynthesis , Antiporters/chemistry , Antiporters/genetics , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/genetics , Apolipoprotein E4/biosynthesis , Apolipoprotein E4/chemistry , Apolipoprotein E4/genetics , Bacteriorhodopsins/biosynthesis , Bacteriorhodopsins/chemistry , Bacteriorhodopsins/genetics , Chromatography, Gel , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Microscopy, Atomic Force , Solubility
10.
J Biotechnol ; 125(2): 194-7, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16621082

ABSTRACT

A novel method for generation of insect-based cell-free translation extracts is presented. The protocol can be completed in less than an hour, and the resulting extracts are extremely proficient in N-linked glycosylation and signal sequence processing. No specialized equipment other than that usually present in an ordinary biochemistry laboratory is required. The novel approach dramatically reduces cost and time while rendering enhanced lysates compared to previously published strategies.


Subject(s)
Protein Biosynthesis/genetics , Protein Processing, Post-Translational , Protein Sorting Signals/genetics , Spodoptera/metabolism , Animals , Biotechnology/methods , Cell Extracts/chemistry , Cell Line , Cell-Free System/metabolism , Glycosylation , Reproducibility of Results , Spodoptera/cytology
11.
J Am Chem Soc ; 128(14): 4508-9, 2006 Apr 12.
Article in English | MEDLINE | ID: mdl-16594652

ABSTRACT

A simple, rapid, in vitro cell-free protein expression system, Expressway NMR, is introduced and used to express the small ubiquitin-related modifier protein SUMO-1. This 12 kDa molecule is challenging for NMR as it has limited solubility and requires relatively high salt (200 mM) for stability in solution. Starting with the gene, the cell-free system, and milligram amounts of nitrogen-15 isotopically enriched amino acids, sufficient protein is produced in 4 h to obtain a high-resolution 2D HSQC spectrum of the protein in 40 min. This time would be closer to 10 min with the aid of a higher sensitivity salt-tolerant cryogenic NMR probe. With all protein purification steps included, and aggressive data processing using the filter diagonalization method (FDM), it is but 6 h from gene to heteronuclear single quantum coherence (HSQC). As the cell-free system is nearly background-free, it is also possible to work with the crude reaction mixture, in which case only a total of 5 h is required. Sample stability over time, whether crude extract or purified, was notable, with no significant change in the 15N-1H HSQC spectrum over 6 months at 4 degrees C (300 muM, pH 6.1, capped NMR tube). The combination of a turnkey, high-yield, protease-free in vitro protein expression system, an optimized sensitivity-enhanced HSQC pulse sequence, and FDM processing makes this scheme an attractive first step to rapidly assess the suitability of proteins for complete solution structure determination.

12.
Trends Biotechnol ; 23(3): 150-6, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15734558

ABSTRACT

Recent technical advances have revitalized cell-free expression systems to meet the increasing demands for protein synthesis. Cell-free systems offer several advantages over traditional cell-based expression methods, including the easy modification of reaction conditions to favor protein folding, decreased sensitivity to product toxicity and suitability for high-throughput strategies because of reduced reaction volumes and process time. Moreover, improvements in translation efficiency have resulted in yields that exceed a milligram of protein per milliliter of reaction mix. We review the advances on this expanding technology and highlight the growing list of associated applications.


Subject(s)
Cell-Free System , Protein Biosynthesis , Recombinant Proteins/biosynthesis , Directed Molecular Evolution/methods , Directed Molecular Evolution/trends , Protein Folding
SELECTION OF CITATIONS
SEARCH DETAIL
...