Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nat Chem Biol ; 16(10): 1078-1086, 2020 10.
Article in English | MEDLINE | ID: mdl-32719556

ABSTRACT

The p53 homolog TAp63α is the transcriptional key regulator of genome integrity in oocytes. After DNA damage, TAp63α is activated by multistep phosphorylation involving multiple phosphorylation events by the kinase CK1, which triggers the transition from a dimeric and inactive conformation to an open and active tetramer that initiates apoptosis. By measuring activation kinetics in ovaries and single-site phosphorylation kinetics in vitro with peptides and full-length protein, we show that TAp63α phosphorylation follows a biphasic behavior. Although the first two CK1 phosphorylation events are fast, the third one, which constitutes the decisive step to form the active conformation, is slow. Structure determination of CK1 in complex with differently phosphorylated peptides reveals the structural mechanism for the difference in the kinetic behavior based on an unusual CK1/TAp63α substrate interaction in which the product of one phosphorylation step acts as an inhibitor for the following one.


Subject(s)
Apoptosis/physiology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Catalytic Domain , DNA Damage , Female , Humans , Mice , Models, Molecular , Molecular Dynamics Simulation , Oocytes , Phosphorylation , Protein Conformation , Time Factors , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
2.
Biochemistry ; 57(40): 5780-5784, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30226041

ABSTRACT

Biomembranes composed of lipids and proteins play central roles in physiological processes, and the precise balance between different lipid species is crucial for maintaining membrane function. One pathway for the biosynthesis of the abundant lipid phosphatidylcholine in eukaryotes involves a membrane-integrated phospholipid methyltransferase named Opi3 in yeast. A still unanswered question is whether Opi3 can catalyze phosphatidylcholine synthesis in trans, at membrane contact sites. While evidence for this activity was obtained from studies with complex in vitro-reconstituted systems based on endoplasmic reticulum membranes, isolated and purified Opi3 could not be analyzed. We present new insights into Opi3 activity by characterizing the in vitro-synthesized enzyme in defined hydrophobic environments. Saccharomyces cerevisiae Opi3 was cell-free synthesized and either solubilized in detergent micelles or co-translationally inserted into preformed nanodisc membranes of different lipid compositions. While detergent-solubilized Opi3 was inactive, the enzyme inserted into nanodisc membranes showed activity and stayed monomeric as revealed by native mass spectrometry. The methylation of its lipid substrate dioleoylphosphatidylmonomethylethanolamine to phosphatidylcholine was monitored by one-dimensional 31P nuclear magnetic resonance. Phosphatidylcholine formation was observed not only in nanodiscs containing inserted Opi3 but also in nanodiscs devoid of the enzyme containing the lipid substrate. This result gives a clear indication for in trans catalysis by Opi3; i.e., it acts on the substrate in juxtaposed membranes, while in cis lipid conversion may also contribute. Our established system for the characterization of pure Opi3 in defined lipid environments may be applicable to other lipid biosynthetic enzymes and help in understanding the subcellular organization of lipid synthesis.


Subject(s)
Cell Membrane/chemistry , Membrane Lipids/chemistry , Nanostructures/chemistry , Phosphatidyl-N-Methylethanolamine N-Methyltransferase/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Cell-Free System/enzymology
3.
Biophys J ; 113(6): 1331-1341, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28450130

ABSTRACT

Nanodiscs that hold a lipid bilayer surrounded by a boundary of scaffold proteins have emerged as a powerful tool for membrane protein solubilization and analysis. By combining nanodiscs and cell-free expression technologies, even completely detergent-free membrane protein characterization protocols can be designed. Nanodiscs are compatible with various techniques, and due to their bilayer environment and increased stability, they are often superior to detergent micelles or liposomes for membrane protein solubilization. However, transport assays in nanodiscs have not been conducted so far, due to limitations of the two-dimensional nature of nanodisc membranes that offers no compartmentalization. Here, we study Krokinobacter eikastus rhodopsin-2 (KR2), a microbial light-driven sodium or proton pump, with noncovalent mass-spectrometric, electrophysiological, and flash photolysis measurements after its cotranslational insertion into nanodiscs. We demonstrate the feasibility of adsorbing nanodiscs containing KR2 to an artificial bilayer. This allows us to record light-induced capacitive currents that reflect KR2's ion transport activity. The solid-supported membrane assay with nanodisc samples provides reliable control over the ionic condition and information of the relative ion activity of this promiscuous pump. Our strategy is complemented with flash photolysis data, where the lifetimes of different photointermediates were determined at different ionic conditions. The advantage of using identical samples to three complementary approaches allows for a comprehensive comparability. The cell-free synthesis in combination with nanodiscs provides a defined hydrophobic lipid environment minimizing the detergent dependence often seen in assays with membrane proteins. KR2 is a promising tool for optogenetics, thus directed engineering to modify ion selectivity can be highly beneficial. Our approach, using the fast generation of functional ion pumps incorporated into nanodiscs and their subsequent analysis by several biophysical techniques, can serve as a versatile screening and engineering platform. This may open new avenues for the study of ion pumps and similar electrogenic targets.


Subject(s)
Membranes, Artificial , Optical Imaging , Rhodopsins, Microbial/chemistry , Chromatography, Gel , Escherichia coli , Feasibility Studies , Flavobacteriaceae , Ion Transport , Mass Spectrometry , Membrane Potentials , Nanostructures , Optogenetics , Photolysis , Rhodopsins, Microbial/isolation & purification
4.
J Biol Chem ; 291(5): 2535-46, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26620564

ABSTRACT

Screening of new compounds directed against key protein targets must continually keep pace with emerging antibiotic resistances. Although periplasmic enzymes of bacterial cell wall biosynthesis have been among the first drug targets, compounds directed against the membrane-integrated catalysts are hardly available. A promising future target is the integral membrane protein MraY catalyzing the first membrane associated step within the cytoplasmic pathway of bacterial peptidoglycan biosynthesis. However, the expression of most MraY homologues in cellular expression systems is challenging and limits biochemical analysis. We report the efficient production of MraY homologues from various human pathogens by synthetic cell-free expression approaches and their subsequent characterization. MraY homologues originating from Bordetella pertussis, Helicobacter pylori, Chlamydia pneumoniae, Borrelia burgdorferi, and Escherichia coli as well as Bacillus subtilis were co-translationally solubilized using either detergent micelles or preformed nanodiscs assembled with defined membranes. All MraY enzymes originating from Gram-negative bacteria were sensitive to detergents and required nanodiscs containing negatively charged lipids for obtaining a stable and functionally folded conformation. In contrast, the Gram-positive B. subtilis MraY not only tolerates detergent but is also less specific for its lipid environment. The MraY·nanodisc complexes were able to reconstitute a complete in vitro lipid I and lipid II forming pipeline in combination with the cell-free expressed soluble enzymes MurA-F and with the membrane-associated protein MurG. As a proof of principle for future screening platforms, we demonstrate the inhibition of the in vitro lipid II biosynthesis with the specific inhibitors fosfomycin, feglymycin, and tunicamycin.


Subject(s)
Bacterial Proteins/chemistry , Monosaccharides/biosynthesis , Oligopeptides/biosynthesis , Transferases/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Bacillus subtilis/enzymology , Biosynthetic Pathways , Bordetella pertussis/enzymology , Borrelia burgdorferi/enzymology , Cell Wall/chemistry , Cell-Free System , Chlamydophila pneumoniae/enzymology , Cytoplasm/chemistry , DNA/chemistry , Detergents/chemistry , Escherichia coli/enzymology , Fosfomycin/chemistry , Helicobacter pylori/enzymology , Micelles , Peptides/chemistry , Peptidoglycan/chemistry , Proteins/chemistry , Recombinant Proteins/chemistry , Transferases (Other Substituted Phosphate Groups) , Tunicamycin/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/biosynthesis
5.
J Neurochem ; 140(2): 280-293, 2017 01.
Article in English | MEDLINE | ID: mdl-27917477

ABSTRACT

The integral synaptic vesicle protein SV31 has been shown to bind divalent cations. Here, we demonstrate that SV31 protein synthesized within a cell-free system binds Zn2+ and to a lower extent Ni2+ and Cu2+ ions. Expression with Zn2+ stabilized the protein and increased solubility. SV31 was preferentially monomeric in detergent and revealed specific binding of Zn2+ . When co-translationally inserted into defined nanodisc bilayers, SV31 assembled into dimeric complexes, resulting in increased binding of Zn2+ . Putative Zn2+ -binding motifs within SV31 comprise aspartic acid and histidine residues. Site-directed mutagenesis of two conserved aspartic acid residues leads to a potent decrease in Zn2+ binding but did not affect dimerization. Chemical modification of histidine residues abolished some of the Zn2+ -binding capacity. We demonstrate proton-dependent transport of Zn2+ as by accumulation of fluorescent FluoZin-1 inside of SV31-containing proteoliposomes. Transport activity has a Km value of 44.3 µM and required external Zn2+ and internal acidic pH. Our results demonstrate that the synaptic vesicle-integral protein SV31 functions as a proton-dependent Zn2+ transporter. SV31 may attribute specific and yet undiscovered functions to subsets of synapses.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Synaptic Vesicles/metabolism , Zinc/metabolism , Animals , Biological Transport , Cations, Divalent/metabolism , Membrane Transport Proteins/metabolism , Mutagenesis, Site-Directed/methods , Protein Multimerization , Rats
6.
Anal Chem ; 89(22): 12314-12318, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29039652

ABSTRACT

Cotranslational insertion of membrane proteins into defined nanoparticle membranes has been developed as an efficient process to produce highly soluble samples in native-like environments and to study lipid-dependent effects on protein structure and function. Numerous examples of the structural and functional characterization of transporters, ion channels, or G-protein-coupled receptors in cotranslationally formed nanodisc complexes demonstrate the versatility of this approach, although the basic underlying mechanisms of membrane insertion are mainly unknown. We have revealed the first aspects of the insertion of proteins into nanodiscs by combining cell-free expression, noncovalent mass spectrometry, and NMR spectroscopy. We provide evidence of cooperative insertion of homo-oligomeric complexes and demonstrate the possibility to modulate their stoichiometry by modifying reaction conditions. Additionally, we show that significant amounts of lipid are released from the nanodiscs upon insertion of larger protein complexes.


Subject(s)
Lasers , Membrane Proteins/analysis , Nuclear Magnetic Resonance, Biomolecular , Spectrometry, Mass, Electrospray Ionization
7.
Biochim Biophys Acta ; 1818(12): 3098-106, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22960287

ABSTRACT

Nanodiscs (NDs) enable the analysis of membrane proteins (MP) in natural lipid bilayer environments. In combination with cell-free (CF) expression, they could be used for the co-translational insertion of MPs into defined membranes. This new approach allows the characterization of MPs without detergent contact and it could help to identify effects of particular lipids on catalytic activities. Association of MPs with different ND types, quality of the resulting MP/ND complexes as well as optimization parameters are still poorly analyzed. This study describes procedures to systematically improve CF expression protocols for the production of high quality MP/ND complexes. In order to reveal target dependent variations, the co-translational ND complex formation with the bacterial proton pump proteorhodopsin (PR), with the small multidrug resistance transporters SugE and EmrE, as well as with the Escherichia coli MraY translocase was studied. Parameters which modulate the efficiency of MP/ND complex formation have been identified and in particular effects of different lipid compositions of the ND membranes have been analyzed. Recorded force distance pattern as well as characteristic photocycle dynamics indicated the integration of functionally folded PR into NDs. Efficient complex formation of the E. coli MraY translocase was dependent on the ND size and on the lipid composition of the ND membranes. Active MraY protein could only be obtained with ND containing anionic lipids, thus providing new details for the in vitro analysis of this pharmaceutically important protein.


Subject(s)
Bacterial Proteins/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Rhodopsin/chemistry , Transferases/chemistry , Antiporters/chemistry , Antiporters/metabolism , Bacterial Proteins/metabolism , Escherichia coli Proteins/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Rhodopsin/metabolism , Rhodopsins, Microbial , Transferases/metabolism , Transferases (Other Substituted Phosphate Groups)
8.
J Magn Reson ; 302: 50-63, 2019 05.
Article in English | MEDLINE | ID: mdl-30959416

ABSTRACT

Combinatorial selective isotope labeling is a valuable tool to facilitate polypeptide backbone resonance assignment in cases of low sensitivity or extensive chemical shift degeneracy. It involves recording of 15N-HSQC and 2D HN-projections of triple-resonance spectra on a limited set of samples containing different combinations of labeled and unlabeled amino acid types. Using labeling schemes in which the three backbone heteronuclei (amide nitrogen, α-carbon and carbonyl carbon) are enriched in 15N or 13C isotopes - individually as well as simultaneously - usually yields abundant amino-acid type information of consecutive residues i and i - 1. Although this results in a large number of anchor points that can be used in the sequential assignment process, for most amide signals the exact positioning of the corresponding residue the polypeptide sequence still relies on matching intra- and interresidual 13C chemical shifts obtained from 3D spectra. An obvious way to obtain more sequence-specific assignments directly with combinatorial labeling would be to increase the number of samples. This is, however, undesirable because of increased sample preparation efforts and costs. Irrespective of the number of samples, unambiguous assignments cannot be accomplished for i - 1/i pairs that are not unique in the sequence. Here we show that the ambiguity for non-unique pairs can be resolved by including information about the labeling state of residues i + 1 and i - 2. Application to a 35-residue peptide resulted in complete assignments of all detectable signals in the 15N HSQC which, due to its repetitive sequence and 13C chemical shift degeneracies, was difficult to achieve by other means. For a medium-sized protein (165 residues, rotational correlation time 8.2 ns) the improved protocol allowed the extent of backbone amide assignment to be expanded to 88% solely using a suite of 2D 1H-15N correlated spectra.


Subject(s)
Combinatorial Chemistry Techniques , Nitrogen/chemistry , Peptides/chemistry , Amides/chemistry , Amino Acids/chemistry , Carbon Isotopes , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Protein Folding , Reactive Nitrogen Species
9.
J Am Soc Mass Spectrom ; 30(1): 181-191, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30225732

ABSTRACT

Native mass spectrometry is applied for the investigation of proteins and protein complexes worldwide. The challenge in native mass spectrometry is maintaining the features of the proteins of interest, such as oligomeric state, bound ligands, or the conformation of the protein complex, during transfer from solution to gas phase. This is an essential prerequisite to allow conclusions about the solution state protein complex, based on the gas phase measurements. Therefore, soft ionization techniques are required. Widely used for the analysis of protein complexes are nanoelectro spray ionization (nESI) mass spectrometers. A newer ionization method is laser induced liquid bead ion desorption (LILBID), which is based on the release of protein complexes from solution phase via infrared (IR) laser desorption. We use both methods in our lab, depending on the requirements of the biological system we are interested in. Here we benchmark the performance of our LILBID mass spectrometer in comparison to a nESI instrument, regarding sample conditions, buffer and additive tolerances, dissociation mechanism and applicability towards soluble and membrane protein complexes. Graphical Abstract ᅟ.


Subject(s)
Mass Spectrometry/instrumentation , Mass Spectrometry/methods , Membrane Proteins/chemistry , Antiporters/analysis , Antiporters/chemistry , Avidin/analysis , Avidin/chemistry , Bacterial Proteins/analysis , Bacterial Proteins/chemistry , Buffers , Detergents/chemistry , Escherichia coli Proteins/analysis , Escherichia coli Proteins/chemistry , Glycerol/chemistry , Lasers , Membrane Proteins/analysis , Potassium Channels/analysis , Potassium Channels/chemistry , Spectrometry, Mass, Electrospray Ionization/instrumentation
10.
Methods Enzymol ; 614: 143-185, 2019.
Article in English | MEDLINE | ID: mdl-30611423

ABSTRACT

Although membrane proteins are in the focus of biochemical research for many decades the general knowledge of this important class is far behind soluble proteins. Despite several recent technical developments, the most challenging feature still is the generation of high-quality samples in environments suitable for the selected application. Reconstitution of membrane proteins into lipid bilayers will generate the most native-like environment and is therefore commonly desired. However, it poses tremendous problems to solution-state NMR analysis due to the dramatic increase in particle size resulting in high rotational correlation times. Nevertheless, a few promising strategies for the solution NMR analysis of membrane inserted proteins are emerging and will be discussed in this chapter. We focus on the generation of membrane protein samples in nanodisc membranes by cell-free systems and will describe the characteristic advantages of that platform in providing tailored protein expression and folding environments. We indicate frequent problems that have to be overcome in cell-free synthesis, nanodisc preparation, and customization for samples dedicated for solution-state NMR. Detailed instructions for sample preparation are given, and solution NMR approaches suitable for membrane proteins in bilayers are compiled. We further discuss the current strategies applied for signal detection from such difficult samples and describe the type of information that can be extracted from the various experiments. In summary, a comprehensive guideline for the analysis of membrane proteins in native-like membrane environments by solution-state NMR techniques will be provided.


Subject(s)
Escherichia coli/chemistry , Lipid Bilayers/chemistry , Membrane Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Synthetic Biology/methods , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Lipid Bilayers/metabolism , Membrane Proteins/metabolism , Nanostructures/chemistry , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/chemistry , Phosphorylcholine/metabolism , Protein Folding , Sodium Cholate/chemistry , Sodium Cholate/metabolism , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
11.
N Biotechnol ; 40(Pt B): 245-260, 2018 Jan 25.
Article in English | MEDLINE | ID: mdl-28943390

ABSTRACT

Protein production using processed cell lysates is a core technology in synthetic biology and these systems are excellent to produce difficult toxins or membrane proteins. However, the composition of the central lysate of cell-free systems is still a "black box". Escherichia coli lysates are most productive for cell-free expression, yielding several mgs of protein per ml of reaction. Their preparation implies proteome fractionation, resulting in strongly biased and yet unknown lysate compositions. Many metabolic pathways are expected to be truncated or completely removed. The lack of knowledge of basic cell-free lysate proteomes is a major bottleneck for directed lysate engineering approaches as well as for assay design using non-purified reaction mixtures. This study is starting to close this gap by providing a blueprint of the S30 lysate proteome derived from the commonly used E. coli strain A19. S30 lysates are frequently used for cell-free protein production and represent the basis of most commercial E. coli cell-free expression systems. A fraction of 821 proteins was identified as the core proteome in S30 lysates, representing approximately a quarter of the known E. coli proteome. Its classification into functional groups relevant for transcription/translation, folding, stability and metabolic processes will build the framework for tailored cell-free reactions. As an example, we show that SOS response induction during cultivation results in tuned S30 lysate with better folding capacity, and improved solubility and activity of synthesized proteins. The presented data and protocols can serve as a platform for the generation of customized cell-free systems and product analysis.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/chemistry , Proteome/chemistry , Proteome/metabolism , Chromatography, Liquid , Escherichia coli/metabolism , Green Fluorescent Proteins/chemistry , Mass Spectrometry , Protein Folding , Solubility , Spectrometry, Fluorescence
12.
Methods Mol Biol ; 1586: 291-312, 2017.
Article in English | MEDLINE | ID: mdl-28470613

ABSTRACT

Cell-free expression allows to synthesize membrane proteins in completely new formats that can relatively easily be customized for particular applications. Amphiphilic superstructures such as micelles, lipomicelles, or nanodiscs can be provided as nano-devices for the solubilization of membrane proteins. Defined empty bilayers in the form of nanodiscs offer native like environments for membrane proteins, supporting functional folding, proper oligomeric assembly as well as stability. Even very difficult and detergent-sensitive membrane proteins can be addressed by the combination of nanodisc technology with efficient cell-free expression systems as the direct co-translational insertion of nascent membrane proteins into supplied preassembled nanodiscs is possible. This chapter provides updated protocols for the synthesis of membrane proteins in presence of preassembled nanodiscs suitable for emerging applications such as screening of lipid effects on membrane protein function and the modulation of oligomeric complex formation.


Subject(s)
Cell-Free System/metabolism , Escherichia coli/genetics , Lipid Bilayers/chemistry , Membrane Proteins/genetics , Nanostructures/chemistry , Synthetic Biology/methods , Detergents/chemistry , Gene Expression , Lipids/chemistry , Membrane Proteins/chemistry , Membrane Proteins/isolation & purification , Protein Folding , Solubility
13.
Elife ; 62017 01 09.
Article in English | MEDLINE | ID: mdl-28067619

ABSTRACT

Membrane proteins frequently assemble into higher order homo- or hetero-oligomers within their natural lipid environment. This complex formation can modulate their folding, activity as well as substrate selectivity. Non-disruptive methods avoiding critical steps, such as membrane disintegration, transfer into artificial environments or chemical modifications are therefore essential to analyze molecular mechanisms of native membrane protein assemblies. The combination of cell-free synthetic biology, nanodisc-technology and non-covalent mass spectrometry provides excellent synergies for the analysis of membrane protein oligomerization within defined membranes. We exemplify our strategy by oligomeric state characterization of various membrane proteins including ion channels, transporters and membrane-integrated enzymes assembling up to hexameric complexes. We further indicate a lipid-dependent dimer formation of MraY translocase correlating with the enzymatic activity. The detergent-free synthesis of membrane protein/nanodisc samples and the analysis by LILBID mass spectrometry provide a versatile platform for the analysis of membrane proteins in a native environment.


Subject(s)
Membrane Proteins/metabolism , Protein Multimerization , Mass Spectrometry , Protein Binding , Protein Folding , Synthetic Biology
14.
Methods Mol Biol ; 1432: 1-21, 2016.
Article in English | MEDLINE | ID: mdl-27485326

ABSTRACT

The complexity of membrane protein synthesis is largely reduced in cell-free systems and it results into high success rates of target expression. Protocols for the preparation of bacterial lysates have been optimized in order to ensure reliable efficiencies in membrane protein production that are even sufficient for structural applications. The open accessibility of the semisynthetic cell-free expression reactions allows to adjust membrane protein solubilization conditions according to the optimal folding requirements of individual targets. Two basic strategies will be exemplified. The post-translational solubilization of membrane proteins in detergent micelles is most straightforward for crystallization approaches. The co-translational integration of membrane proteins into preformed nanodiscs will enable their functional characterization in a variety of natural lipid environments.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/chemistry , Membrane Proteins/metabolism , Cell-Free System , Protein Folding , Solubility , Synthetic Biology
15.
Structure ; 24(10): 1830-1841, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27618661

ABSTRACT

Nanodiscs and isotropic bicelles are promising membrane mimetics in the field of solution nuclear magnetic resonance (NMR) spectroscopy of integral membrane proteins (IMPs). Despite varied challenges to solution NMR studies of IMPs, we attribute the paucity of solution NMR structures in these environments to the inability of diverse IMPs to withstand detergent treatment during standard nanodisc and bicelle preparations. Here, we present a strategy that creates small isotropic bicelles from IMPs co-translationally embedded in large nanodiscs using cell-free expression. Our results demonstrate appreciable gains in NMR spectral quality while preserving lipid-IMP contacts. We validate the approach on the detergent-sensitive LspA, which finally allowed us to perform high-quality triple-resonance NMR experiments for structural studies. Our strategy of producing bicelles from nanodiscs comprehensively avoids detergent during expression and preparation and is suitable for solution NMR spectroscopy of lipid-IMP complexes.


Subject(s)
Membrane Proteins/chemistry , Nanostructures/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Lipid Bilayers/chemistry , Solutions
16.
Methods Enzymol ; 556: 351-69, 2015.
Article in English | MEDLINE | ID: mdl-25857790

ABSTRACT

Cell-free (CF) protein expression has emerged as one of the most efficient production platforms for membrane proteins. Central bottlenecks prevalent in conventional cell-based expression systems such as mistargeting, inclusion body formation, degradation as well as product toxicity can be addressed by taking advantage of the reduced complexity of CF expression systems. However, the open accessibility of CF reactions offers the possibility to design customized artificial expression environments by supplying synthetic hydrophobic compounds such as micelles or membranes of defined composition. The open nature of CF systems therefore generally allows systematic screening approaches for the identification of efficient cotranslational solubilization environments of membrane proteins. Synergies exist in particular with the recently developed nanodisc (ND) technology enabling the synthesis of stable and highly soluble particles containing membrane discs of defined composition. Specific types of lipids frequently modulate folding, stability, and activity of integrated membrane proteins. One recently reported example are phospho-MurNAc-pentapeptide (MraY) translocases that catalyze a crucial step in bacterial peptidoglycan biosynthesis making them interesting as future drug targets. Production of functionally active MraY homologues from most human pathogens in conventional cellular production systems was so far not successful due to their obviously strict lipid dependency for functionally folding. We demonstrate that the combination of CF expression with ND technologies is an efficient strategy for the production of folded MraY translocases, and we present a general protocol for the rapid screening of lipid specificities of membrane proteins.


Subject(s)
Cell-Free System/metabolism , Escherichia coli/genetics , Lipid Bilayers/chemistry , Liposomes/chemistry , Membrane Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Escherichia coli/cytology , Escherichia coli/growth & development , Gene Expression , Humans , Lipids/chemistry , Membrane Proteins/chemistry , Protein Folding , Transferases/chemistry , Transferases/genetics , Transferases (Other Substituted Phosphate Groups)
17.
FEBS Lett ; 589(15): 1713-22, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-25937121

ABSTRACT

Cell-free protein production has become a core technology in the rapidly spreading field of synthetic biology. In particular the synthesis of membrane proteins, highly problematic proteins in conventional cellular production systems, is an ideal application for cell-free expression. A large variety of artificial as well as natural environments for the optimal co-translational folding and stabilization of membrane proteins can rationally be designed. The high success rate of cell-free membrane protein production allows to focus on individually selected targets and to modulate their functional and structural properties with appropriate supplements. The efficiency and robustness of lysates from Escherichia coli strains allow a wide diversity of applications and we summarize current strategies for the successful production of high quality membrane protein samples.


Subject(s)
Escherichia coli/genetics , Membrane Proteins/biosynthesis , Cell-Free System , Membrane Proteins/chemistry , Membrane Proteins/genetics , Protein Biosynthesis , Protein Folding , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
18.
Methods Mol Biol ; 1258: 125-43, 2015.
Article in English | MEDLINE | ID: mdl-25447862

ABSTRACT

Precipitation, aggregation, and inclusion body (IB) formation are frequently observed problems upon overexpression of recombinant proteins. The open accessibility of cell-free reactions allows addressing such critical steps by the addition of protein stabilizers such as chemical chaperones or detergents directly into the expression reactions. This approach could therefore reduce or even prevent initial protein precipitation already in the translation environment. The strategy might be considered to generally improve protein sample quality and to rescue proteins that are difficult to refold from IBs or from aggregated precipitates. We describe a protocol for the co-translational stabilization of difficult proteins by their expression in the presence of supplements such as alcohols, poly-ions, or detergents. We compile potentially useful compounds together with their recommended stock and working concentrations. Examples of screening experiments in order to systematically identify compounds or compound mixtures that stabilize particular proteins of interest are given. The method can primarily be considered for the production of unstable soluble proteins or of membrane proteins containing larger soluble domains.


Subject(s)
Cell-Free System/metabolism , Protein Biosynthesis/physiology , Recombinant Proteins/metabolism , Animals , Humans , Membrane Proteins/metabolism , Protein Folding , Protein Stability
19.
Chem Biol ; 21(4): 552-562, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24704508

ABSTRACT

Phosphopantetheine transferases represent a class of enzymes found throughout all forms of life. From a structural point of view, they are subdivided into three groups, with transferases from group II being the most widespread. They are required for the posttranslational modification of carrier proteins involved in diverse metabolic pathways. We determined the crystal structure of the group II phosphopantetheine transferase Sfp from Bacillus in complex with a substrate carrier protein in the presence of coenzyme A and magnesium, and observed two protein-protein interaction sites. Mutational analysis showed that only the hydrophobic contacts between the carrier protein's second helix and the C-terminal domain of Sfp are essential for their productive interaction. Comparison with a similar structure of a complex of human proteins suggests that the mode of interaction is highly conserved in all domains of life.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Protein Processing, Post-Translational , Transferases (Other Substituted Phosphate Groups)/chemistry , Transferases (Other Substituted Phosphate Groups)/metabolism , Acyl Carrier Protein/chemistry , Acyl Carrier Protein/metabolism , Amino Acid Sequence , Bacillus subtilis/enzymology , Bacterial Proteins/genetics , Carrier Proteins/genetics , Crystallography, X-Ray , Genetic Variation/genetics , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Sequence Data , Protein Conformation , Transferases (Other Substituted Phosphate Groups)/genetics
SELECTION OF CITATIONS
SEARCH DETAIL