Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
ACS Cent Sci ; 5(4): 629-639, 2019 Apr 24.
Article in English | MEDLINE | ID: mdl-31041382

ABSTRACT

α-Hemolysin (αHL), a ß-barrel pore-forming toxin (ßPFT), is secreted as a water-soluble monomer by Staphylococcus aureus. Upon binding to receptors on target cell membranes, αHL assembles to form heptameric membrane-spanning pores. We have previously engineered αHL to create a protease-activatable toxin that is activated by site-specific proteolysis including by tumor proteases. In this study, we redesigned αHL so that it requires 2-fold activation on target cells through (i) binding to specific receptors, and (ii) extracellular proteolytic cleavage. To assess our strategy, we constructed a fusion protein of αHL with galectin-1 (αHLG1, αHL-Galectin-1 chimera). αHLG1 was cytolytic toward cells that lack a receptor for wild-type αHL. We then constructed protease-activatable mutants of αHLG1 (PAMαHLG1s). PAMαHLG1s were activated by matrix metalloproteinase 2 (MMP-2) and had approximately 50-fold higher cytolytic activity toward MMP-2 overexpressing cells (HT-1080 cells) than toward non-overexpressing cells (HL-60 cells). Our approach provides a novel strategy for tailoring pore-forming toxins for therapeutic applications.

2.
ACS Nano ; 10(9): 8843-50, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27537396

ABSTRACT

Protein engineering has been used to remodel pores for applications in biotechnology. For example, the heptameric α-hemolysin pore (αHL) has been engineered to form a nanoreactor to study covalent chemistry at the single-molecule level. Previous work has been confined largely to the chemistry of cysteine side chains or, in one instance, to an irreversible reaction of an unnatural amino acid side chain bearing a terminal alkyne. Here, we present four different αHL pores obtained by coupling either two or three fragments by native chemical ligation (NCL). The synthetic αHL monomers were folded and incorporated into heptameric pores. The functionality of the pores was validated by hemolysis assays and by single-channel current recording. By using NCL to introduce a ketone amino acid, the nanoreactor approach was extended to an investigation of reversible covalent chemistry on an unnatural side chain at the single-molecule level.

3.
Diabetes ; 65(8): 2311-21, 2016 08.
Article in English | MEDLINE | ID: mdl-27207556

ABSTRACT

The secretion of insulin from pancreatic islet ß-cells is critical for glucose homeostasis. Disrupted insulin secretion underlies almost all forms of diabetes, including the most common form, type 2 diabetes (T2D). The control of insulin secretion is complex and affected by circulating nutrients, neuronal inputs, and local signaling. In the current study, we examined the contribution of glycine, an amino acid and neurotransmitter that activates ligand-gated Cl(-) currents, to insulin secretion from islets of human donors with and without T2D. We find that human islet ß-cells express glycine receptors (GlyR), notably the GlyRα1 subunit, and the glycine transporter (GlyT) isoforms GlyT1 and GlyT2. ß-Cells exhibit significant glycine-induced Cl(-) currents that promote membrane depolarization, Ca(2+) entry, and insulin secretion from ß-cells from donors without T2D. However, GlyRα1 expression and glycine-induced currents are reduced in ß-cells from donors with T2D. Glycine is actively cleared by the GlyT expressed within ß-cells, which store and release glycine that acts in an autocrine manner. Finally, a significant positive relationship exists between insulin and GlyR, because insulin enhances the glycine-activated current in a phosphoinositide 3-kinase-dependent manner, a positive feedback loop that we find is completely lost in ß-cells from donors with T2D.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glycine/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Receptors, Glycine/metabolism , Animals , Calcium/metabolism , Diabetes Mellitus, Type 2/physiopathology , Electrophysiology , Humans , Immunohistochemistry , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Glycine/genetics , Reverse Transcriptase Polymerase Chain Reaction
4.
Diabetologia ; 57(12): 2535-45, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25208758

ABSTRACT

AIMS/HYPOTHESIS: There is evidence that ATP acts as an autocrine signal in beta cells but the receptors and pathways involved are incompletely understood. Here we investigate the receptor subtype(s) and mechanism(s) mediating the effects of ATP on human beta cells. METHODS: We examined the effects of purinergic agonists and antagonists on membrane potential, membrane currents, intracellular Ca(2+) ([Ca(2+)]i) and insulin secretion in human beta cells. RESULTS: Extracellular application of ATP evoked small inward currents (3.4 ± 0.7 pA) accompanied by depolarisation of the membrane potential (by 14.4 ± 2.4 mV) and stimulation of electrical activity at 6 mmol/l glucose. ATP increased [Ca(2+)]i by stimulating Ca(2+) influx and evoking Ca(2+) release via InsP3-receptors in the endoplasmic reticulum (ER). ATP-evoked Ca(2+) release was sufficient to trigger exocytosis in cells voltage-clamped at -70 mV. All effects of ATP were mimicked by the P2Y(1/12/13) agonist ADP and the P2Y1 agonist MRS-2365, whereas the P2X(1/3) agonist α,ß-methyleneadenosine-5-triphosphate only had a small effect. The P2Y1 antagonists MRS-2279 and MRS-2500 hyperpolarised glucose-stimulated beta cells and lowered [Ca(2+)]i in the absence of exogenously added ATP and inhibited glucose-induced insulin secretion by 35%. In voltage-clamped cells subjected to action potential-like stimulation, MRS-2279 decreased [Ca(2+)]i and exocytosis without affecting Ca(2+) influx. CONCLUSIONS/INTERPRETATION: These data demonstrate that ATP acts as a positive autocrine signal in human beta cells by activating P2Y1 receptors, stimulating electrical activity and coupling Ca(2+) influx to Ca(2+) release from ER stores.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Exocytosis/physiology , Insulin-Secreting Cells/metabolism , Receptors, Purinergic P2Y1/metabolism , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Calcium Signaling/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Exocytosis/drug effects , Humans , Insulin-Secreting Cells/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology
5.
Proc Natl Acad Sci U S A ; 110(47): E4417-26, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24194548

ABSTRACT

In stochastic sensing, the association and dissociation of analyte molecules is observed as the modulation of an ionic current flowing through a single engineered protein pore, enabling the label-free determination of rate and equilibrium constants with respect to a specific binding site. We engineered sensors based on the staphylococcal α-hemolysin pore to allow the single-molecule detection and characterization of protein kinase-peptide interactions. We enhanced this approach by using site-specific proteolysis to generate pores bearing a single peptide sensor element attached by an N-terminal peptide bond to the trans mouth of the pore. Kinetics and affinities for the Pim protein kinases (Pim-1, Pim-2, and Pim-3) and cAMP-dependent protein kinase were measured and found to be independent of membrane potential and in good agreement with previously reported data. Kinase binding exhibited a distinct current noise behavior that forms a basis for analyte discrimination. Finally, we observed unusually high association rate constants for the interaction of Pim kinases with their consensus substrate Pimtide (~10(7) to 10(8) M(-1) · s(-1)), the result of electrostatic enhancement, and propose a cellular role for this phenomenon.


Subject(s)
Peptides/metabolism , Protein Engineering/methods , Proto-Oncogene Proteins c-pim-1/isolation & purification , Proto-Oncogene Proteins c-pim-1/metabolism , Signal Transduction/genetics , Bacterial Toxins/chemistry , Hemolysin Proteins/chemistry , Kinetics , Protein Binding , Proteolysis , Static Electricity , Stochastic Processes
6.
Nat Chem ; 5(8): 651-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23881495

ABSTRACT

Capsular polysaccharides form the outermost protective layer around many Gram-negative bacteria. Antibiotics aimed directly at weakening this layer are not yet available. In pathogenic Escherichia coli E69, a protein, Wza, forms a pore in the outer membrane that transports K30 capsular polysaccharide from its site of synthesis to the outside of the cell. This therefore represents a prospective antibiotic target. Here we test a variety of grommet-like mimics of K30 capsular polysaccharide on wild-type Wza and on mutant open forms of the pore by electrical recording in planar lipid bilayers. The most effective glycomimetic was the unnatural cyclic octasaccharide octakis(6-deoxy-6-amino)cyclomaltooctaose (am8γCD), which blocks the α-helix barrel of Wza, a site that is directly accessible from the external medium. This glycomimetic inhibited K30 polysaccharide transport in live E. coli E69. With the protective outer membrane disrupted, the bacteria can be recognized and killed by the human immune system.


Subject(s)
Carbohydrate Metabolism , Carrier Proteins/metabolism , Escherichia coli/metabolism
7.
Nat Commun ; 4: 1725, 2013.
Article in English | MEDLINE | ID: mdl-23591892

ABSTRACT

The bottom-up construction of artificial tissues is an underexplored area of synthetic biology. An important challenge is communication between constituent compartments of the engineered tissue, and between the engineered tissue and additional compartments, including extracellular fluids, further engineered tissue and living cells. Here we present a dimeric transmembrane pore that can span two adjacent lipid bilayers, and thereby allow aqueous compartments to communicate. Two heptameric staphylococcal α-hemolysin pores were covalently linked in an aligned cap-to-cap orientation. The structure of the dimer, (α7)2, was confirmed by biochemical analysis, transmission electron microscopy and single-channel electrical recording. We show that one of two ß-barrels of (α7)2 can insert into the lipid bilayer of a small unilamellar vesicle, while the other spans a planar lipid bilayer. The (α7)2 pores spanning two bilayers were also observed by transmission electron microscopy.


Subject(s)
Lipid Bilayers , Proteins/chemistry , Base Sequence , Cyclodextrins/chemistry , DNA Primers , Dimerization , Microscopy, Electron, Transmission , Molecular Dynamics Simulation , Polymerase Chain Reaction , Proteins/genetics
8.
Nano Lett ; 12(9): 4895-900, 2012 Sep 12.
Article in English | MEDLINE | ID: mdl-22849517

ABSTRACT

Nanopores have been used in label-free single-molecule studies, including investigations of chemical reactions, nucleic acid analysis, and applications in sensing. Biological nanopores generally perform better than artificial nanopores as sensors, but they have disadvantages including a fixed diameter. Here we introduce a biological nanopore ClyA that is wide enough to sample and distinguish large analyte proteins, which enter the pore lumen. Remarkably, human and bovine thrombins, despite 86% sequence identity, elicit characteristic ionic current blockades, which at -50 mV differ in their main current levels by 26 ± 1 pA. The use of DNA aptamers or hirudin as ligands further distinguished the protein analytes. Finally, we constructed ClyA nanopores decorated with covalently attached aptamers. These nanopores selectively captured and internalized cognate protein analytes but excluded noncognate analytes, in a process that resembles transport by nuclear pores.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/ultrastructure , Hemolysin Proteins/chemistry , Hemolysin Proteins/ultrastructure , Nanostructures/chemistry , Nanostructures/ultrastructure , Protein Engineering/methods , Protein Interaction Mapping/methods , Binding Sites , Particle Size , Porosity , Protein Binding
9.
Am J Physiol Endocrinol Metab ; 303(9): E1107-16, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22932785

ABSTRACT

Somatostatin-14 (SST) inhibits insulin and glucagon secretion by activating G protein-coupled somatostatin receptors (SSTRs), of which five isoforms exist (SSTR1-5). In mice, the effects on pancreatic ß-cells are mediated by SSTR5, whereas α-cells express SSTR2. In both cell types, SSTR activation results in membrane hyperpolarization and suppression of exocytosis. Here, we examined the mechanisms by which SST inhibits secretion from human ß- and α-cells and the SSTR isoforms mediating these effects. Quantitative PCR revealed high expression of SSTR2, with lower levels of SSTR1, SSTR3, and SSTR5, in human islets. Immunohistochemistry showed expression of SSTR2 in both ß- and α-cells. SST application hyperpolarized human ß-cells and inhibited action potential firing. The membrane hyperpolarization was unaffected by tolbutamide but antagonized by tertiapin-Q, a blocker of G protein-gated inwardly rectifying K⁺ channels (GIRK). The effect of SST was mimicked by an SSTR2-selective agonist, whereas a SSTR5 agonist was marginally effective. SST strongly (>70%) reduced depolarization-evoked exocytosis in both ß- and α-cells. A slightly weaker inhibition was observed in both cell types after SSTR2 activation. SSTR3- and SSTR1-selective agonists moderately reduced the exocytotic responses in ß- and α-cells, respectively, whereas SSTR4- and SSTR5-specific agonists were ineffective. SST also reduced voltage-gated P/Q-type Ca²âº currents in ß-cells, but normalization of Ca²âº influx to control levels by prolonged depolarizations only partially restored exocytosis. We conclude that SST inhibits secretion from both human ß- and α-cells by activating GIRK and suppressing electrical activity, reducing P/Q-type Ca²âº currents, and directly inhibiting exocytosis. These effects are mediated predominantly by SSTR2 in both cell types.


Subject(s)
Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Exocytosis/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Gene Expression Regulation , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/drug effects , Humans , Immunohistochemistry , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Kinetics , Membrane Potentials/drug effects , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/metabolism , Receptors, Somatostatin/agonists , Receptors, Somatostatin/genetics , Recombinant Proteins/agonists , Recombinant Proteins/metabolism , Somatostatin/agonists
10.
Biochemistry ; 50(35): 7493-502, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21815625

ABSTRACT

Styryl dyes are widely used to study synaptic vesicle (SV) recycling in neurons; vesicles are loaded with dye during endocytosis, and dye is subsequently released via exocytosis. During putative kiss-and-run exocytosis, efflux of dye from individual SVs has been proposed to occur via two sequential steps: dissociation from the membrane followed by permeation through a small fusion pore. To improve our understanding of the kinetics of efflux of dye from vesicles during kiss-and-run events, we examined the rates of efflux of different dyes through nanometer-scale pores formed in membranes by the toxins melittin and α-hemolysin; these pores approximate the size of fusion pores measured in neuroendocrine cells. We found that the axial diameter of each dye was a crucial determinant for permeation. Moreover, the two dyes with the largest cross-sectional areas were completely unable to pass through pores formed by a mutant α-hemolysin that has a slightly smaller pore than the wild-type toxin. The overall time constant for efflux (seconds) of each dye was orders of magnitude slower than the time constant for dissociation from membranes (milliseconds). Thus, the permeation step is rate-limiting, and this observation was further supported by atomistic molecular dynamics simulations. Together, the data reported here help provide a framework for interpreting dye destaining rates from secretory vesicles.


Subject(s)
Cell Membrane Permeability/physiology , Cell Membrane/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Nanotechnology/methods , Synaptic Vesicles/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Cell Membrane/microbiology , Hemolysin Proteins/chemistry , Hemolysin Proteins/metabolism , Neurotoxins/chemistry , Neurotoxins/metabolism , Pyridinium Compounds/chemistry , Pyridinium Compounds/metabolism , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/metabolism , Synaptic Vesicles/microbiology
11.
Proc Natl Acad Sci U S A ; 107(18): 8165-70, 2010 May 04.
Article in English | MEDLINE | ID: mdl-20400691

ABSTRACT

Engineered protein pores have several potential applications in biotechnology: as sensor elements in stochastic detection and ultrarapid DNA sequencing, as nanoreactors to observe single-molecule chemistry, and in the construction of nano- and micro-devices. One important class of pores contains molecular adapters, which provide internal binding sites for small molecules. Mutants of the alpha-hemolysin (alphaHL) pore that bind the adapter beta-cyclodextrin (betaCD) approximately 10(4) times more tightly than the wild type have been obtained. We now use single-channel electrical recording, protein engineering including unnatural amino acid mutagenesis, and high-resolution x-ray crystallography to provide definitive structural information on these engineered protein nanopores in unparalleled detail.


Subject(s)
Cyclodextrins/chemistry , Hemolysin Proteins/chemistry , Nanostructures/chemistry , Crystallography, X-Ray , Hemolysin Proteins/genetics , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Molecular , Mutation , Porosity , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Thermodynamics
12.
ACS Chem Biol ; 4(12): 983-5, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-20017575

ABSTRACT

An improved method for the semisynthesis of a potassium channel involving native chemical ligation allows the introduction of short sequences containing non-canonical amino acids at any position within the polypeptide chain. The work enhances the technology available for a range of fundamental investigations of membrane proteins and for applications of membrane channels and pores in biotechnology.


Subject(s)
Potassium Channels/chemical synthesis , Amino Acid Sequence , Amino Acids/chemical synthesis , Amino Acids/chemistry , Animals , Bacterial Proteins/chemical synthesis , Bacterial Proteins/chemistry , Models, Molecular , Potassium Channels/chemistry , Protein Conformation , Rats
13.
Nano Lett ; 9(11): 3831-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19645477

ABSTRACT

Nanopores are under development for the detection of a variety of analytes and the investigation of chemical reactions at the single molecule level. In particular, the analysis of nucleic acid molecules is under intense investigation, including the development of systems for rapid, low-cost DNA sequencing. Here, we show that DNA can be translocated through an engineered alphaHL protein pore at pH 11.7, a value at which dsDNA is denatured. Therefore, the alphaHL pore is sufficiently stable to entertain the possibility of direct nanopore sequencing of genomic dsDNA samples, which are more readily obtained and handled than ssDNA.


Subject(s)
DNA/analysis , DNA/chemistry , Nanostructures , Proteins , DNA/genetics , Hemolysin Proteins/chemistry , Hydrogen-Ion Concentration , Models, Molecular , Nanostructures/chemistry , Nanotechnology , Nucleic Acid Denaturation , Proteins/chemistry , Sequence Analysis, DNA/methods
14.
Nat Nanotechnol ; 4(7): 437-40, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19581896

ABSTRACT

Recently, we demonstrated that submicrolitre aqueous droplets submerged in an apolar liquid containing lipid can be tightly connected by means of lipid bilayers to form networks. Droplet interface bilayers have been used for rapid screening of membrane proteins and to form asymmetric bilayers with which to examine the fundamental properties of channels and pores. Networks, meanwhile, have been used to form microscale batteries and to detect light. Here, we develop an engineered protein pore with diode-like properties that can be incorporated into droplet interface bilayers in droplet networks to form devices with electrical properties including those of a current limiter, a half-wave rectifier and a full-wave rectifier. The droplet approach, which uses unsophisticated components (oil, lipid, salt water and a simple pore), can therefore be used to create multidroplet networks with collective properties that cannot be produced by droplet pairs.


Subject(s)
Bacterial Toxins/chemistry , Hemolysin Proteins/chemistry , Ion Channels/chemistry , Lipid Bilayers/chemistry , Microfluidic Analytical Techniques/instrumentation , Nanotechnology/instrumentation , Electrochemistry , Electrodes
15.
J Am Chem Soc ; 129(45): 14034-41, 2007 Nov 14.
Article in English | MEDLINE | ID: mdl-17949000

ABSTRACT

Facilitated translocation of polypeptides through a protein pore is a ubiquitous and fundamental process in biology. Several translocation systems possess various well-defined binding sites within the pore lumen, but a clear mechanistic understanding of how the interaction of the polypeptides with the binding site alters the underlying kinetics is still missing. Here, we employed rational protein design and single-channel electrical recordings to obtain detailed kinetic signatures of polypeptide translocation through the staphylococcal alpha-hemolysin (alphaHL) transmembrane pore, a robust, tractable, and versatile beta-barrel protein. Acidic binding sites composed of rings of negatively charged aspartic acid residues, engineered at strategic positions within the beta barrel, produced dramatic changes in the functional properties of the alphaHL protein, facilitating the transport of cationic polypeptides from one side of the membrane to the other. When two electrostatic binding sites were introduced, at the entry and exit of the beta barrel, both the rate constants of association and dissociation increased substantially, diminishing the free energy barrier for translocation. By contrast, more hydrophobic polypeptides exhibited a considerable decrease in the rate constant of association to the pore lumen, having to overcome a greater energetic barrier because of the hydrophilic nature of the pore interior.


Subject(s)
Bacterial Toxins/metabolism , Hemolysin Proteins/metabolism , Peptides/chemistry , Peptides/physiology , Bacterial Toxins/chemistry , Binding Sites , Catalysis , Cations/chemistry , Cell Membrane/chemistry , Cell Membrane/physiology , Hemolysin Proteins/chemistry , Kinetics , Models, Molecular , Peptides/chemical synthesis , Protein Transport/physiology , Staphylococcus/chemistry , Staphylococcus/physiology , Time Factors
17.
J Am Chem Soc ; 129(15): 4701-5, 2007 Apr 18.
Article in English | MEDLINE | ID: mdl-17375923

ABSTRACT

A robust, portable chip containing a single protein nanopore would be a significant development in the practical application of stochastic sensing technology. Here, we describe a chip in which a single alpha-hemolysin (alphaHL) pore in a planar phospholipid bilayer is sandwiched between two layers of agarose gel. These encapsulated nanopore chips remain functional after storage for weeks. The detection of the second messenger inositol 1,4,5-trisphosphate (IP3) was demonstrated with a chip containing a genetically engineered alphaHL pore as the sensor element.


Subject(s)
Nanostructures , Protein Array Analysis/instrumentation , Protein Array Analysis/methods , Proteins/analysis
18.
Chembiochem ; 7(12): 1923-7, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17068836

ABSTRACT

Stochastic sensing is an emerging approach for the detection of a wide variety of analytes at the level of individual molecules. Detection is accomplished by observing the modulation of the current that flows through a single protein pore that has been engineered to bind an analyte of interest. Previously, protein analytes have been detected by using pores to which ligands have been appended at specific sites by targeted chemical modification. Here, we report the first genetically encoded stochastic sensor element for detecting a protein. A protein kinase inhibitor peptide sequence was incorporated into the alpha-hemolysin polypeptide, which was used to form a heteroheptameric pore containing a single copy of the inhibitor sequence. With this pore, the successful detection of the catalytic subunit of protein kinase A was demonstrated. This development should greatly facilitate the detection of active kinase subunits by stochastic sensing and the rapid screening of kinase inhibitors by an approach that yields kinetic information.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/genetics , Hemolysin Proteins/genetics , Peptide Fragments/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Base Sequence , Biosensing Techniques , Cell Membrane/chemistry , Cyclic AMP-Dependent Protein Kinases/analysis , Cyclic AMP-Dependent Protein Kinases/metabolism , Electric Conductivity , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Genetic Engineering , Models, Biological , Molecular Sequence Data , Protein Binding , Recombinant Fusion Proteins/chemistry , Stochastic Processes
19.
J Am Chem Soc ; 128(33): 10684-5, 2006 Aug 23.
Article in English | MEDLINE | ID: mdl-16910655

ABSTRACT

The rapid quantification of the enantiomers of small chiral molecules is very important, notably in pharmacology. Here, we show that the enantiomers of drug molecules can be distinguished by stochastic sensing, a single-molecule detection technique. The sensing element is an engineered alpha-hemolysin protein pore, fitted with a beta-cyclodextrin adapter. By using the approach, the enantiomeric composition of samples of ibuprofen and thalidomide can be determined in less than 1 s.


Subject(s)
Hemolysin Proteins/chemistry , Hemolysin Proteins/metabolism , beta-Cyclodextrins/chemistry , Hemolysin Proteins/genetics , Ibuprofen/chemistry , Ibuprofen/metabolism , Membranes, Artificial , Molecular Structure , Mutation , Thalidomide/chemistry , Thalidomide/metabolism , beta-Cyclodextrins/metabolism
20.
Chembiochem ; 6(10): 1875-81, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16118820

ABSTRACT

Engineered versions of the transmembrane protein pore alpha-hemolysin (alphaHL) can be used as stochastic sensing elements for the identification and quantification of a wide variety of analytes at the single-molecule level. Until now, nitroaromatic analytes have eluded detection by this approach. We now report that binding sites for nitroaromatics can be built within the lumen of the alphaHL pore from simple rings of seven aromatic amino acid side chains (Phe, Tyr or Trp). By monitoring the ionic current that passes through a single pore at a fixed applied potential, various nitroaromatics can be distinguished from TNT on the basis of the amplitude and duration of individual current-blocking events. Rings of less than seven aromatics bind the analytes more weakly; this suggests that direct aromatic-aromatic interactions are involved. The engineered pores should be useful for the detection of explosives and, in combination with computational approaches and structural analysis, they could further our understanding of noncovalent interactions between aromatic molecules.


Subject(s)
Biosensing Techniques/methods , Hemolysin Proteins/chemistry , Hemolysin Proteins/genetics , Trinitrotoluene/analysis , Hemolysin Proteins/metabolism , Membrane Potentials , Models, Molecular , Mutagenesis, Insertional , Protein Engineering , Trinitrotoluene/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL