US20080166747A1 - Biosensor - Google Patents

Biosensor Download PDF

Info

Publication number
US20080166747A1
US20080166747A1 US11/785,591 US78559107A US2008166747A1 US 20080166747 A1 US20080166747 A1 US 20080166747A1 US 78559107 A US78559107 A US 78559107A US 2008166747 A1 US2008166747 A1 US 2008166747A1
Authority
US
United States
Prior art keywords
biosensor
ligand
sample
glucose
binding
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/785,591
Inventor
Homme W. Hellinga
Loren L. Looger
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Duke University
Original Assignee
Duke University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Duke University filed Critical Duke University
Priority to US11/785,591 priority Critical patent/US20080166747A1/en
Publication of US20080166747A1 publication Critical patent/US20080166747A1/en
Assigned to DUKE UNIVERSITY reassignment DUKE UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HELLINGA, HOMME W., LOOGER, LOREN L.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54366Apparatus specially adapted for solid-phase testing
    • G01N33/54373Apparatus specially adapted for solid-phase testing involving physiochemical end-point determination, e.g. wave-guides, FETS, gratings
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/001Enzyme electrodes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/536Immunoassay; Biospecific binding assay; Materials therefor with immune complex formed in liquid phase
    • G01N33/542Immunoassay; Biospecific binding assay; Materials therefor with immune complex formed in liquid phase with steric inhibition or signal modification, e.g. fluorescent quenching
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/566Immunoassay; Biospecific binding assay; Materials therefor using specific carrier or receptor proteins as ligand binding reagents where possible specific carrier or receptor proteins are classified with their target compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/195Assays involving biological materials from specific organisms or of a specific nature from bacteria

Definitions

  • the present invention relates to biosensors and to methods of making and using same.
  • Biosensors are analytical tools that can be used to measure the presence of a single molecular species in a complex mixture by combining the extraordinary molecular recognition properties of biological macromolecules with signal transduction mechanisms that couple ligand binding to readily detectable physical changes (Hall, Biosenisors , Prentice-Hall, Englewood Cliffs, N.J.; Scheller et al., Curr. Op. Biotech. 12:35-40, 2001).
  • a biosensor is reagentless and, in contrast to enzyme-based assays or competitive immunoassays, does not change composition as a consequence of making the measurement (Hellinga & Marvin, Trends Biotech. 16:183-189, 1998).
  • biosensors combine a naturally occurring macromolecule such as an enzyme or an antibody, with the identification of a suitable physical signal particular to the molecule in question, and the construction of a detector specific to that system (Meadows, Adv. Drug Deliv. Rev. 21:177-189, 1996). Recently, molecular engineering techniques have been explored to develop macromolecules that combine a wide range of binding specificities and affinities with a common signal transduction mechanism, to construct a generic detection system for many different analytes (Hellinga & Marvin, Trends Biotech. 16:183-189, 1998).
  • Escherichia coli periplasmic binding proteins are members of a protein superfamily (bacterial periplasmic binding proteins, bPBPs) (Tam & Saier, Microbiol. Rev. 57:320-346, 1993) that has been shown to be well suited for the engineering of biosensors (U.S. Pat. No. 6,277,627). These proteins comprise two domains linked by a hinge region (Quiocho & Ledvina, Molec. Microbiol. 20:17-25, 1996). The ligand-binding site is located at the interface between the two domains.
  • the proteins typically adopt two conformations: a ligand-free open form, and a ligand-bound closed form, which interconvert via a hinge-bending mechanism upon ligand binding.
  • This global, ligand-mediated conformational change has been exploited to couple ligand binding to changes in fluorescence intensity by positioning single, environmentally sensitive fluorophores in locations that undergo local conformational changes in concert with the global change (Brune et al., Biochemistry 33:8262-8271, 1994; Gilardi et al., Prot. Eng. 10:479-486, 1997; Gilardi et al., Anal. Chem. 66:3840-3847, 1994; Marvin et al., Proc. Natl. Acad. Sci.
  • Conformational coupling mechanisms can also be devised to alter the flow of current between the surface of an electrode derivatized with the engineered bPBP containing a covalently attached redox cofactor (Benson et al., Science 293: 1641-1644, 2001).
  • the present invention provides a method of utilizing bPBPs to generate biosensors for a variety of chemical classes including sugars, amino acids, dipeptides, cations, and anions. These biosensors have widespread utility including in clinical, industrial, and environmental settings.
  • the present invention relates to biosensors, making them from mutant or wildtype members of the bacterial periplasmic binding protein (bPBP) superfamily, and using them to assay for (i.e., detect and/or quantitate) ligand.
  • the tertiary structure of bPBPs is comprised of two domains linked by a hinge region with a ligand-binding pocket located at an interface between the two domains. They typically adopt two conformations: a ligand-free open form and a ligand-bound closed form, which interconvert via a hinge-bending mechanism which depends on whether ligand is bound or not at the site.
  • Biosensors are made by covalently or non-covalently attaching at least one reporter group to one or more specific positions of a bPBP. Upon binding of ligand to the biosensor, there is a change in the signal transduced by the reporter group which can be analyzed by assessing any of its observable properties (e.g., optical or electrochemical properties). Biosensors are classified according to the relationship between the attachment site of the reporter group and the binding site(s) of the ligand (i.e., allosteric, endosteric, or peristeric) or distance between those sites (i.e., distal or proximal).
  • the event of ligand binding to biosensor changes the local environment of the position-specific attached reporter group.
  • the signal of the reporter group may be generated by one or more fluorophores and/or redox cofactors.
  • the biosensor may be operated under physiological conditions without additional reagents.
  • FIG. 1 shows the 3-D structures of eleven bPBPs indicating locations of allosteric, endosteric, and peristeric sites used. Each protein is shown in the closed form, with bound ligand indicated by ball-and-stick structures. The two domains of each bPBP are oriented vertically with the first (containing the N-terminus) above the second (containing the C-terminus). A hinge segment connects the domains. The structure of histidine BP is used to represent the as yet unsolved structure of glutamate/aspartate BP.
  • Residues mutated to cysteine are indicated by differently shaded spheres, and differentiated as allosteric (heavy shading), endosteric (medium shading, in GBP only), or peristeric (light shading). Structures are grouped by cluster as defined by Tam & Saier (Microbiol. Rev. 57:320-346, 1993) according to sequence-based relationships.
  • Cluster 2 arabinose BP (ABP), glucose BP (GBP), and ribose BP (RBP).
  • Cluster 5 dipeptide BP (DPP).
  • Cluster 3 glutamine BP (QBP), histidine BP (HBP), and glutamate/aspartate BP (EBP).
  • Cluster 6 phosphate BP (PBP) and sulfate BP (SBP).
  • Cluster 1 maltose BP (MBP) and Fe(III) BP (FeBP).
  • MBP maltose BP
  • FeBP Fe(III) BP
  • FIG. 2 shows alignment of sequences of E. coli YBEJ (putative glutamate/aspartate BP), glutamine BP, and histidine BP using clustalW (Thompson et al., Nucl. Acids Res. 22:4673-4680, 1994). Numbering begins from the putative initiation codon of the open reading frame for YBEJ, including its leader sequence. The underlined methionine is the initiation codon for expression of YBEJ used in the study. Residues in each protein that were mutated to cysteine for fluorophore conjugation are in bold font. The letters “a” and “p” beneath these residues indicate their classification as allosteric or peristeric, respectively.
  • FIG. 3 shows structural formulae of thiol-reactive fluorophores. Approximate wavelengths of maximal fluorescence excitation and emission, respectively, of the protein-bound fluorophores are (in nm): pyrene (340, 390); acrylodan (390, 500); fluorescein (485, 520); NBD (490, 540); NBDE (490, 530); JPW4039 (485, 590); JPW4042 (470, 640); and JPW4045 (470, 640).
  • FIGS. 4A and 4B show a definition of fluorimetric parameters.
  • FIG. 4A shows parameters ⁇ std , I 1 and I 2 used to determine the standard intensity change ⁇ I std .
  • FIG. 4B shows parameters A 1 , A 2 , 0 A, and ⁇ A used to determine ⁇ R. Each of the areas ⁇ A encompasses the respective area 0 A.
  • FIGS. 5A and 5B show fluorimetric titration of glucose BP and glutamate/aspartate BP conjugates.
  • FIG. 5A shows titration of glucose BP W183C-acrylodan with glucose.
  • FIG. 5B Titration of glutamate/aspartate BP T129C-NBD with amino acids. Data points: ⁇ glutamic acid; + aspartic acid; ⁇ asparagine; x glutamine.
  • the lines shown are the best fit binding isotherms.
  • FIGS. 6A-6C shows occurrence of fluorimetric parameters in the set of 320 fluorescent conjugates.
  • FIG. 6A shows distribution of the shift in wavelength of maximum fluorescent intensity ( max ⁇ saturated ⁇ max ⁇ apo ).
  • FIG. 6B shows distribution of the intensity change parameter ⁇ I std .
  • FIG. 6C shows distribution of the ratiometric change parameter ⁇ R max . For each parameter, the upper bound of each interval is indicated.
  • FIG. 7 shows occurrence of changes in ligand affinity among the three classes of fluorophore attachment site.
  • endosteric sites filled bars; peristeric sites, hatched bars; allosteric sites, open bars.
  • wt K d is that of the C64A mutant, in which all conjugates were made.
  • Data for dipeptide BP and Fe(III) BP are not included.
  • the K d for Gly-Leu dipeptide in the wild-type has not been reported.
  • Fe(III) BP the K d of the unconjugated mutant E57D was not determined.
  • the upper bound is indicated.
  • the interval labeled “0” contains values of log( mut K d / wt K d )> ⁇ 1 and ⁇ 0.
  • FIGS. 8A and 8B show ratiometric titration of bPBP fluorophore conjugates using different pairs of emission wavelength bands.
  • FIG. 8A shows glucose BP-W183C conjugated to acrylodan, titrated with glucose at the following ratios of fluorescence emission (wavelengths in nm): ⁇ , F 450-459 /F 550-559 ( app K d ⁇ 5.0 mM); ⁇ , F 450-459 /F 486-495 ( app K d ⁇ 10.4 mM); ⁇ , F 472-481 /F 450-459 ( app K d ⁇ 17.4 mM). Lines show fit to equation 4.
  • FIG. 8B shows ribose BP-T135C conjugated to acrylodan, titrated with ribose at the following ratios of fluorescence emission (wavelengths in nm): ⁇ , F 501-510 /F 450-459 ( app K d ⁇ 41 ⁇ M); ⁇ , F 450-459 /F 501-510 ( app K d ⁇ 254 ⁇ M); ⁇ , F 450-459 /F 547-556 ( app K d ⁇ 461 ⁇ M).
  • the present invention relates to biosensors constructed using engineered bPBPs, for example, E. coli bPBPs.
  • conjugates are constructed that can be used to monitor binding of ligands to bPBPs.
  • Conjugates can be produced by introducing mutations into a bPBP at one or more specific positions in the protein structure where covalently attached reporter groups (e.g., fluorophores or redox cofactors) respond to a conformational change of the bPBP which occurs upon ligand binding.
  • reporter groups e.g., fluorophores or redox cofactors
  • Other methods for covalently or non-covalently attaching at least one reporter group to one or more amino acid residue positions in the primary amino acid sequence of a mutant or wildtype bPBP include: addition or substitution of any activatable crosslinkers, use of designer or non-natural tRNAs, introduction of coordination sites, etc.
  • the ligand-binding pocket may be engineered to bind ligands which are not bound by the wild-type bPBP.
  • the ligand-binding site is located at the interface between the bPBP's two domains. Mutating amino acid residues at that interface which are near (i.e., in or around) the binding site of wild-type bPBP may generate new contacts with ligand (e.g., Zn ++ for MBP) and destroy or alter binding with cognate ligand (e.g., maltose for MBP). This can be used to change the specificity of the ligand-binding pocket.
  • ligand e.g., Zn ++ for MBP
  • cognate ligand e.g., maltose for MBP
  • maltose binding protein has been mutated to specifically bind to noncognate ligand: e.g., metal Zn ++ ion, trinitrotoluene, L-lactate, and serotonin (Marvin & Hellinga, Proc. Natl. Acad. Sci. USA 98:4955-4960, 2001; Looger et al., Nature 423:185-190, 2003; Dwyer et al., Proc. Natl. Acad. Sci. USA 100:11255-11260, 2003).
  • noncognate ligand e.g., metal Zn ++ ion, trinitrotoluene, L-lactate, and serotonin
  • biosensors which bind noncognate ligand can be made by mutating amino acid residues at the interface of the two bPBP domains to generate a new ligand-binding pocket; ligand bound by such biosensors may not bind to wild-type bPBP.
  • mutations in the bPBP may be made to affect function of the biosensor: e.g., mutations may increase or decrease binding affinity or specificity; enhance or reduce signal transduction; add a new functionality by fusion with another carbohydrate, lipid, or protein domain; improve thermostability or thermolability; introduce a catalytic activity; shorten or lengthen operational life; widen or narrow the conditions for operation; or any combination thereof.
  • mutating amino acid residues at positions of the bPBP where a reporting group is not attached e.g., at least one missense mutation which is not a cysteine conjugated through a thiol bond to a fluorophore).
  • the present invention relates to a method of constructing a reagentless fluorescent biosensor.
  • the method comprises identifying sites on a bPBP that undergo a local conformational change in concert with a ligand-mediated hinge-bending motion. Cysteine residues can be introduced at one or more such sites and a fluorophore coupled thereto so that fluorescence intensity of the fluorophore changes upon ligand binding.
  • bPBPs suitable for use in the present method can be selected or designed.
  • the bPBP superfamily is well suited for the redesign of ligand-binding specificities either by computational methods or by other means or both based on the ligand to be detected (see, for example, analytes referenced in Table 1).
  • Sites on the bPBP appropriate for attachment of one or more reporters include allosteric sites, peristeric sites, and endosteric sites (a reporter can also be present at a non-signaling site for use, for example, as a reference).
  • the reporter e.g., fluorophore
  • the reporter can be placed at one or more locations distant from the ligand-binding site (i.e., distal from the ligand-binding pocket) that undergo local conformational changes upon ligand binding.
  • the reporter e.g., fluorophore
  • the reporter can be positioned on the “rim” of the binding site but not such that it directly interacts with the ligand.
  • the reporter e.g., fluorophore
  • the reporter can be present in the binding site so that it interacts directly with the ligand. The latter two examples show attachment proximal to the ligand-binding pocket.
  • Allosteric, peristeric, and endosteric sites can be designed in at least two different ways, as detailed in the Example that follows.
  • a structure-based design approach can be used in which the structures of the open and closed states (for allosteric designs) or the closed state only (for peristeric and endosteric designs) are examined.
  • a sequence-based design approach can be used wherein homology relationships can be exploited to predict the location of cysteine mutations in proteins the three-dimensional structures of which have not been determined, provided that such mutations have been characterized in proteins of known structure.
  • reporters suitable for use in the invention include, but are not limited to, fluorophores and redox cofactors.
  • fluorophores the choice is dependent, at least in part, on the nature of the location within the particular protein. While one fluorophore may function better in a certain location than another, one skilled in the art can readily select the preferred fluorophore for a particular application (see, for example, U.S. Pat. No. 6,277,627).
  • eight different fluorophores are used in the design of fluorescent sensors for:
  • ABSP Arabinose Arabinose binding protein
  • DPP Dipeptides Dipeptide binding protein Glutamate and asparate Glu/Asp binding protein
  • EBP Glutamine Glutamine binding protein
  • QBP Fe(III) Iron binding protein
  • FeBP Iron binding protein
  • HBP Maltose Maltose binding protein
  • MBP Glucose Glucose binding protein
  • GBP Phosphate Phosphate binding protein
  • SBP Sulfate Sulfate binding protein
  • Redox reporters for use in the invention can be a redox-active metal center or a redox-active organic molecule. It can be a natural organic cofactor such as NAD, NADP, FAD or a natural metal center such as Blue Copper, iron-sulfur clusters, or heme, or a synthetic center such as an organometallic compound such as a ruthenium complex, organic ligand such as a quinone, or an engineered metal center introduced into the protein or engineered organic cofactor binding site. Cofactor-binding sites can be engineered using rational design or directed evolution techniques.
  • the redox reporter can be covalently or non-covalently attached to the protein, either by site-specific or adventitious interactions between the cofactor and protein.
  • the redox reporter can be, for example, bound (e.g., covalently) at a position where the amino acid residue is on the protein's surface.
  • the redox reporter can be a metal-containing group (e.g., a transition metal-containing group) that is capable of reversibly or semi-reversibly transferring one or more electrons.
  • a metal-containing group e.g., a transition metal-containing group
  • the reporter group has a redox potential in the potential window below that subject to interference by molecular oxygen and has a functional group suitable for covalent conjugation to the protein (e.g., thiol-reactive functionalities such as maleimides or iodoacetamide for coupling to unique cysteine residues in the protein).
  • the metal of the reporter group should be substitutionally inert in either reduced or oxidized state (i.e., advantageously, exogenous groups do not form adventitious bonds with the reporter group).
  • the reporter group can be capable of undergoing an amperometric or potentiometric change in response to ligand binding.
  • the reporter group is water soluble, is capable of site-specific coupling to a protein (e.g., via a thiol-reactive functional group on the reporter group that reacts with a unique cysteine in the protein), and undergoes a potentiometric response upon ligand binding.
  • Suitable transition metals for use in the invention include, but are not limited to, copper (Cu), cobalt (Co), palladium (Pd), iron (Fe), ruthenium (Ru), rhodium (Rh), osmium (Os), rhenium (Re), platinum (Pt), scandium (Sc), titanium (Ti), vanadium (V), chromium (Cr), manganese (Mn), nickel (Ni), molybdenum (Mo), technetium (Tc), tungsten (W), and iridium (Ir).
  • the first series of transition metals the platinum metals (Ru, Rh, Pd, Os, Ir, and Pt), along with Fe, Re, W, Mo, and Tc, are preferred.
  • Particularly preferred are metals that do not change the number of coordination sites upon a change in oxidation state, including ruthenium, osmium, iron, platinum and palladium, with ruthenium being especially preferred.
  • the reporter group can be present in the biosensor as a covalent conjugate with the protein or it can be a metal center that forms part of the protein matrix (for instance, a redox center such as iron-sulfur clusters, heme, Blue copper, the electrochemical properties of which are sensitive to its local environment).
  • the reporter group can be present as a fusion between the protein and a metal binding domain (for instance, a small redox-active protein such as a cytochrome).
  • the reporter group is covalently conjugated to the protein via a maleimide functional group bound to a cysteine (thiol) on the protein.
  • the reporter group is attached to the protein so that it is located between the protein and the electrode.
  • Engineered proteins of the invention can be produced by site-specifically introducing a reporter group(s) by total synthesis, semi-synthesis, or gene fusions (see, for example, Adams et al., Nature 39:694-697, 1991; Brune et al., Biochemistry 33:8262-8271, 1994; Gilardi et al., Anal. Chem. 66:3840-3847, 1994; Godwin et al., J. Am. Chem. Soc. 118:6514-6515, 1996; Marvin et al., Proc. Natl. Acad. Sci. U.S.A. 94:4366-4371, 1997; Post et al., J. Biol. Chem.
  • Assays for ligand may be performed with the biosensor.
  • a sample is contacted with the biosensor under appropriate assay conditions.
  • Ligand present in the sample if any, may be detected by binding to the biosensor and measuring the signal transduced by ligand-bound biosensor in the assay.
  • binding does not need to be quantitative because a simple determination of whether the ligand is present or absent (within detection limits) needs to be performed. Otherwise, comparison to a series of control samples (e.g., known quantities of ligand) may be required to quantitate the amount or concentration of ligand in the sample. Given the volume of the sample, the amount (i.e., mass) of ligand and the concentration of ligand are interconvertible.
  • a blank sample containing no ligand may be used to determine background signal.
  • Standards may be used to construct a standard curve (e.g., hyperbolic) used to quantitate unknown samples.
  • homogeneous assay formats i.e., those requiring no separation of bound and non-bound ligand
  • separation in a heterogeneous assay format may be required if substances which significantly interfere with signal transduction and/or measurement are present in the sample.
  • Signal transduction preferably does not require the addition of exotic reagents so assays of body fluids may be performed with minimal sample preparation and under physiological conditions. They may even be performed in vivo if the biosensor is adapted to an implantable medical device. Alternatively, a biosensor in contact with the skin may assay interstitial fluid or perspiration. Lavage may be used to sample mucosal tissues.
  • the sample can be obtained in a laboratory setting (e.g., clinic or research institution); from an environmental source (e.g., air, aquafers and other bodies of water, animal or plant products grown on the land, soil); from an industrial source (e.g., the food, biopharmaceutical, chemical, or other manufacturing industries).
  • the analyte to be assayed is identical to the ligand, comprised of multiple copies of the ligand, chemically related to the ligand such that it is identified by a change in signal transduction (e.g., a related chemical structure is more strongly or more weakly bound by the biosensor as compared to its “correct” ligand), or any combination thereof.
  • the change in signal transduction may be correlated to the change in chemical structure such that the non-identical analyte is identified (see below description of integrative assays).
  • ligands which may be detected or quantitated include: amino acids; carbohydrates; bioactive solid and gaseous compounds which are soluble in an aqueous sample; contraband or controlled substances (i.e., substances which are illegal to use or possess, or which are highly regulated); environmental pollutants (e.g., phosphates, sulfates); explosives (e.g., TNT); food contaminants and byproducts (e.g., carcinogens, plant toxins, teratogens); lipids; metal ions (e.g., divalent cations, ferric ions); microbial toxins (e.g., toxic products of viruses, bacteria, or protozoa); neurotransmitters (e.g., serotonin); nucleosides or nucleotides (e.g., NAD, NADP, FAD
  • One or more biosensors may be covalently or noncovalently attached to a solid or porous substrate.
  • the substrate may be flat and planar (e.g., filter membrane, glass slide, semiconductor chip); cylindrical (e.g., optical fiber, plastic rod); spherical (e.g., crosslinked polymer or glass bead); or formed as a container (e.g., cell or cuvette, multiwell plate).
  • the substrate may be fabricated for analysis by instruments which measure the signal transduced by the reporter group (e.g., microscope, photometer, spectrometer).
  • biosensors may be coded by an attached marker (e.g., bar code, radio frequency or RFID, or biopolymer) which can be decoded by a reader (e.g., scanner of light-and-dark patterns, radio receiver, specific binding probe or automated sequencer) or separated by a sorter in accordance with their marker.
  • the code identifying each biosensor may be used in parallel analysis by rapidly assaying a sample for a plurality of ligands. Multiple biosensors with different ligand-binding specificities are used in the same assay to detect and/or quantitate multiple ligands at the same time. Alternatively, attaching different biosensors at particular spots on the substrate may be used to identify their ligand-binding specificities by where the signals are being produced.
  • Signals may be authenticated by repeating the assay, using multiple biosensors with the same specificity for redundant assays, or correlating the results from multiple biosensors with overlapping specificities for integrative assays. In the latter, particular reactivity patterns of the biosensors are correlated with the identity of the analyte bound by them. Analytes that are more closely related in their chemical structure to the ligand will bind more strongly to the cognate biosensor. Signals from a plurality of biosensors with overlapping, known ligand-binding specificities are integrated to deduce the identity of the analyte.
  • the invention relates, in further embodiments, to biosensors constructed using the above-described methods and to the use thereof in analyte detection in, for example, clinical, industrial, and environmental settings. Particular utilities are described in the specific Example that follows. Provided is a description of a number of sites that can be used for optical glucose sensors based on GBP (W183C conjugated to acrylodan has been used successfully in fiber-optic prototypes of a glucose sensor).
  • biosensors constructed in accordance with the present approach may be present in the public domain (e.g., may be disclosed in Marvin et al., Proc. Natl. Acad. Sci. USA 94:4366-4371, 1997 or in U.S. Pat. No. 6,277,627), such biosensors are not within the scope of the present invention.
  • PCR was used to amplify wild-type genes for bPBPs from genomic DNA of E. coli strain CSH100 (arabinose, dipeptide, histidine, ribose, sulfate, and glutamate/aspartate BP); strain W1485 (glucose and glutamine BP) and strain RU1012 (phosphate BP), or of H. influenzae strain Rd (Fe(III) BP). Amplified products were cloned into one of the protein expression vectors pAED4 (Doering, “Functional and structural studies of a small f-actin binding domain” in Ph.D.
  • N-terminal oligonucleotide primers were designed to clone only the processed periplasmic form, deleting the signal sequence.
  • Two tandem stop codons (TAATGA) follow the last His codon.
  • Maltose BP mutants were made in and expressed from plasmid pMAL-c2X (New England BioLabs). E. coli strains XL1-BLUE (Stratagene) and DH5 ⁇ (Hanahan, J. Mol. Biol. 166:557-580, 1983) were used for plasmid construction. Single amino acid substitutions were generated by overlapping PCR mutagenesis (Ho et al., Gene 77:51-59, 1989). All clones and mutations were confirmed by nucleotide sequencing.
  • Plasmids were transformed into E. coli strain BL21-DE3, grown in nutrient broth overnight at 37° C., then diluted 100-fold into fresh medium and grown further at 37° C. or 25° C. Expression was induced by the addition of isopropyl ⁇ -D-1-thiogalactopyranoside to 1 mM when the optical density of the culture at 600 nm reached 0.4. After 2 to 4 hours, cells were harvested by centrifugation, resuspended in 20 mM 3-morpholinopropanesulfonic acid (MOPS), 100 mM NaCl, pH 6.9 and stored frozen or lysed immediately for protein purification.
  • MOPS 3-morpholinopropanesulfonic acid
  • Protein was eluted with loading buffer containing 400 mM imidazole, and was collected in fractions and assessed for purity by gel electrophoresis. All preparations were at least 95% pure by this criterion. Protein-containing fractions were dialyzed exhaustively against buffer (20 mM MOPS, 100 mM NaCl, pH 6.9, or 20 mM NaH 2 PO 4 , 100 mM NaCl, pH 6.9) or desalted by gel filtration to remove bound ligand.
  • Fluorophore conjugation to cysteine-substituted bPBPs Fluorophore conjugation to cysteine-substituted bPBPs.
  • Thiol-reactive fluorophores obtained from Molecular Probes (Eugene, Oreg.) were 5-iodoacetamidofluorescein (fluorescein); N-(1-pyrene) iodoacetamide (pyrene); N,N′-dimethyl-N-(iodoacetyl)-N′-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)ethylenediamide (NBD); N-((2-(iodoacetoxy)ethyl)-N-methyl)amino-7-nitrobenz-2-oxa-1,3-diazole (NBDE); and 6-acryloyl-2-dimethylaminonaphthalene (acrylodan).
  • the styryl and naphthyl dyes JPW4039, JPW4042, and JPW4045 ( FIG. 3 ) were synthesized at the University of Connecticut. All fluorophore conjugation steps were typically carried out at room temperature. To protein at a concentration of 100 ⁇ M was added tris-(2-carboxyethyl)phosphine HCl to a five-fold molar excess to reduce intermolecular disulfide bonds. A thiol-reactive fluorophore (20 to 25 mM in acetonitrile or dimethyl sulfoxide) was added in small aliquots to a five-fold molar excess over protein.
  • Conjugation proceeded in the dark at room temperature for 4 hours, or overnight at 4° C. Separation of protein from unreacted fluorophore was achieved by exhaustive dialysis or by size-exclusion chromatography. The efficiency of reporter group attachment was assessed by determination of unreacted thiol with Ellman's reagent (Ellman, Arch. Biochem. Biophys. 74:443-450, 1958) or by measuring the ratio of fluorophore to protein from absorbance spectra of the purified conjugate.
  • Phosphate BP solutions and buffer (20 mM MOPS, 100 mM NaCl, pH 6.9) were depleted of phosphate by addition of 7-methylguanosine to 1 mM and dialyzed against bacterial nucleoside phosphorylase (1 unit ml ⁇ 1 ) (Sigma-Aldrich) partitioned in a separate dialysis tube (Brune et al., Biochem. 33:8262-8271, 1994).
  • Fluorimetry All measurements were conducted with an SLM Aminco-Bowman series 2 fluorimeter, with sample stirring at 25° C. Fluorescence emission spectra were acquired with excitation and emission slit widths of 4 and 8 nm, respectively. Photomultiplier tube potential was maintained between 400 and 800 volts. Protein concentrations were in the range of 50 to 1000 nM.
  • Fluorophore-specific excitation was at the following approximate wavelengths: tryptophan, 290 nm; acrylodan, 390 nm; fluorescein, 485 nm; pyrene, 340 nm; NBD and NBDE, 490 nm; JPW4039, 485 nm; JPW4042, 470 nm; JPW4045, 470 nm.
  • Fe(III) BP has a dissociation constant for Fe(III) on the order of 10 ⁇ 21 M (Adhikari et al., J. Biol. Chem. 270:25142-25149, 1995), hindering accurate fluorescence-based measurements of affinity at nanomolar protein concentrations.
  • Fe(III) citrate (logK ⁇ 10.25) (Martell and Smith, Critical Stability Constants , Plenum Press, New York, 1977) as the ligand in a competition assay.
  • bPBP monosaccharides
  • maltose BP maltose BP
  • three bind amino acids glutamine BP
  • one binds di- and tripeptides dipeptide BP
  • two bind oxyanions phosphate and sulfate BP
  • Most of these bPBPs bind at most two or three related ligands with high affinity (micromolar or better).
  • phosphate BP binds phosphate and arsenate but not other oxyanions (Luecke & Quiocho, Nature 347:402-406, 1990), while glucose BP binds glucose and galactose but not other monosaccharides (Anraku, J. Biol. Chem. 243:3116-3122, 1968).
  • Dipeptide BP is an exception in that it binds a wide variety of di- and tripeptides (Smith et al., Microbiology 145:2891-2901, 1999). Measured ligand dissociation constants in these proteins are typically in the range of 0.1 to 1 ⁇ M.
  • Fe(III) BP where the K d for Fe(III) (aq) is estimated to be 10 ⁇ 21 M in competition assays with Fe(III) chelates (Adhikari et al., J. Biol. Chem. 270:25142-25149, 1995).
  • Coupling between ligand binding and a change in the fluorescent signal of a covalently attached, environmentally sensitive fluorophore can be established if the local environment of the fluorophore changes as a result of formation of the complex and a linked conformational change.
  • Two mechanisms can be distinguished to establish such structural linkage relationships.
  • Direct linkage involves formation of a non-bonded contact between the bound ligand and the conjugated fluorophore.
  • Indirect linkage involves changes in the local protein structure in the immediate vicinity of the attached fluorophore, and relies on ligand-mediated conformational changes such as the hinge-bending motion observed in the bPBPs.
  • Direct linkage relationships are readily designed by replacing a residue known to form a ligand contact with a cysteine to which the fluorophore is attached (“endosteric” attachment site).
  • Indirect linkage relationships can be established in two ways. The most straightforward method relies on visual inspection of the ligand complex structure, and identifying residues that are located in the vicinity of the binding site, but do not interact directly with the ligand, and that are likely to be involved in conformational changes. In the case of the bPBPs, such are residues located at the perimeter of the inter-domain cleft that forms the ligand binding site. The environment of these “peristeric” sites changes significantly upon formation of the closed state. These are examples of positions which are proximal to the ligand-binding pocket.
  • the second approach identifies sites in the protein structure that are located some distance away from the ligand-binding site (i.e., distal to the ligand-binding pocket), and undergo a local conformational change in concert with ligand binding. If the structures of both the open and closed states are known, then such “allosteric” sites can be identified using a computational method that analyzes the conformational change (Marvin et al., Proc. Natl. Acad. Sci. USA 94:4366-4371, 1997). Alternatively, once allosteric sites have been identified in one bPBP, modeling and structural homology arguments can be invoked to identify such sites in other bPBPs in which only one state has been characterized (Marvin & Hellinga, J. Am. Chem. Soc. 120:7-11, 1998). Table 3 summarizes the designs of all three classes of sites in each of the receptors used in this study. The locations of these sites in the eleven bPBPs are shown in FIG. 1 .
  • Ligand-binding can then be determined by direct experimentation, or be inferred either by structural relationships to bPBPs of known function, or by establishing genetic linkage to other genes of known function (Pellegrini et al., Proc. Natl. Acad. Sci. USA 96:4285-4288, 1999). Subsequently sites within the homolog that undergo local conformational change, and to which reporter functions can be attached, must be identified. The selection of sites for attaching reporter functions relies on homology to bPBPs of known structure.
  • a glutamate biosensor was constructed starting from genome sequence data only.
  • the genome of E. coli K12 contains the locus ybeJ encoding a protein identified as a putative bPBP based on amino acid sequence homology with glutamine and histidine BPs (26% and 23% sequence identity; 41% and 43% sequence similarity, respectively) (Blattner et al., Science 277:1453-1474, 1997).
  • the assignment of YBEJ as an amino-acid binding protein was strengthened by the presence of conserved residues found to be associated with binding to the ⁇ -amino and ⁇ -carboxylate groups of the ligand in all bPBP amino-acid binding proteins of known structure identified in E. coli (Table 4).
  • ybeJ is located adjacent to three tandem genes (gltJ, gltK, gltL) postulated to be involved in the glutamate/aspartate transport system (Lum & Wallace, GenBank Accession Number U10981, 1995), suggesting that ybeJ encodes a glutamate/aspartate BP.
  • Putative allosteric, endosteric, and peristeric sites were identified from a structure-based sequence alignment of YBEJ with glutamine BP and histidine BP ( FIG. 2 ).
  • the protein was produced by over-expression of the processed form in the cytoplasm with an initiation methionine placed just before the N-terminus of the processed protein, under the control of a strong inducible promoter in the pAED4 (Doering, “Functional and structural studies of a small f-actin binding domain” in Ph.D. thesis, Massachusetts Institute of Technology, 1992); pET-21a (Studier et al., Meth. Enzymol. 185:60-89, 1990) (Novagen); or pKK223-3 (Blattner et al., Science 277:1453-1474, 1997) plasmids.
  • oligohistidine tag was fused to the carboxy terminus of the cloned receptor to permit facile purification by immobilized metal affinity chromatography (Hochuli et al., J. Chromatogr. A 411:177-184, 1987). In all cases, the receptors expressed well (at least 50 mg of pure protein per liter of fermentation). The molecular masses estimated by gel electrophoresis corresponded to the predicted mass of the expressed reading frame.
  • Cysteine point mutations were introduced by the PCR overlap method (Ho et al., Gene 77:51-59, 1989). Mutant proteins typically expressed as well as the wild type protein. All cysteine substitutions in arabinose BP were constructed in the C64A background to prevent interference from this endogenous cysteine (Miller et al., J. Biol. Chem. 254:7521-7528, 1979). In the case of Fe(III) BP, all mutations were constructed in the E57D background. In the crystal structure of Fe(III) BP, this glutamate is coordinated to the iron (Bruns et al., Nat. Struct. Biol. 4:919-924, 1997).
  • ⁇ I std is defined as the normalized intensity change relative to the average intensity, determined at the wavelength mid-point between the two emission maxima:
  • ⁇ ⁇ ⁇ I std ⁇ 2 ⁇ ( I 1 ⁇ ( ⁇ std ) - I 2 ⁇ ( ⁇ std ) ) I 1 ⁇ ( ⁇ std ) + I 2 ⁇ ( ⁇ std ) ⁇ ( 1 )
  • ⁇ std ( ⁇ max, unbound + ⁇ max, saturated )/2 and I 1 , I 2 are the fluorescence intensities at ⁇ std of each spectrum respectively ( FIG. 4A ).
  • ⁇ R is defined in terms of two emission bands, A 1 ([ ⁇ 1 , ⁇ 2 ]) and A 2 ([ ⁇ 3 , ⁇ 4 ]) ( FIG. 4B ):
  • ⁇ ⁇ ⁇ R ⁇ A 1 0 A 2 0 - A 1 ⁇ A 2 ⁇ ⁇ ( 2 )
  • F fluorescence at ligand concentration [S]
  • K d is the dissociation constant
  • F F , F B are the fluorescence intensities of the ligand-free and ligand-saturated states, respectively.
  • Examples of binding isotherms are shown in FIG. 5 for glucose BP and glutamate/aspartate BP.
  • eq. 3 has to be modified to account for differentially weighted contributions of the two emission bands (Lakowicz, Principles of Fluorescence Spectroscopy, 2 nd Ed. Kluwer Academic Press, New York, p. 698, 1999):
  • R is ratio A 1 /A 2
  • R B ⁇ A 1 / ⁇ A 2
  • R F 0 A 1 / 0 A 1
  • app K d is an apparent dissociation constant
  • K d app A 2 0 A 2 ⁇ ⁇ K d ( 5 )
  • the success of the fluorescent biosensor design strategy was evaluated by determining the probability of encountering an effectively responding fluorescent conjugate, and assessing how the ligand-binding affinities are affected by the fluorophore conjugate.
  • Fe(III) citrate was added to conjugates of other bPBPs and the effect on emission intensity was monitored. It was found that Fe(III) citrate quenched fluorescence in all cases, but only at concentrations much higher than those that led to the effect in Fe(III) BP.
  • the decrease in fluorescence intensity observed in all conjugates of Fe(III) BP is therefore due to a binding-specific process, and may involve relaxation of the excited state via a metal-mediated redox mechanism (Lakowicz, Principles of Fluorescence Spectroscopy, 2 nd Ed. Kluwer Academic Press, New York, p. 698, 1999).
  • the probability of encountering a conjugate that responds with a particular intensity declines with increasing magnitude of ⁇ I std ( FIG. 6B ).
  • the ratiometric response behaves similarly ( FIG. 6C ).
  • the two criteria of greatest utility for optical sensing are ⁇ I std and ⁇ R max .
  • the collection of bPBP conjugates was categorized by class of steric site, fluorophore, and protein scaffold, then, for each category, quantified according to the fraction with ⁇ I std >0.25 and with ⁇ R max >1.25.
  • the results (Tables 6 to 8) give an indication of the overall success rate for finding potentially useful fluorescent biosensor conjugates.
  • the scaffolds having the highest success rates for ⁇ I std and ⁇ R max are arabinose BP, glucose BP, ribose BP, and phosphate BP (Table 6).
  • the former three belong to cluster 2, that includes binding proteins for hexoses and pentoses, while phosphate BP, along with sulfate BP, belongs to cluster 6, that includes binding proteins for inorganic polyanions.
  • the scaffolds having the lowest success rate were dipeptide BP (cluster 5, peptide and nickel binding) and the cluster 3 (polar amino-acid binding) proteins glutamine BP, histidine BP, and Glu/Asp BP.
  • dissociation constants are determined not only by the attachment site, but also by the nature of the attached fluorophore, as illustrated for arabinose BP.
  • Dissociation constants for arabinose of the five cysteine-substitution mutants measured by tryptophan fluorescence, are 5.0 ⁇ M (F23C), 3.2 ⁇ M (L253C), 3.4 ⁇ M (D257C), 7.6 ⁇ M (L298C), and 1.6 ⁇ M (K301C).
  • the cysteine substitutions slightly perturbed affinity for arabinose (K d of C64A mutant ⁇ 2.2 ⁇ M).
  • Bioinformatics makes possible the discovery of new biochemical applications without direct experimentation.
  • individual bacterial genomes may encode scores of bPBPs that bind specific molecules to initiate transport or signal transduction (Blattner et al., Science 277:1453-1474, 1997; Quentin et al., J. Mol. Biol. 287:467-484, 1999). Few of these have been characterized, leaving a vast number untapped as scaffolds for potential biosensors. The feasibility of applying genomic information, combined with structural information from homologous proteins, to construct a biosensor of novel specificity has been demonstrated.
  • a glutamate/aspartate BP had been purified from E. coli (Barash & Halpern, Biochim. Biophys. Acta 386:168-180, 1975; Willis & Furlong, J. Biol. Chem. 250:2574-2580, 1975) and characterized.
  • YBEJ corresponds to this protein.
  • glutamate/aspartate BP was isolated from periplasmic extracts, consistent with ybeJ encoding a protein with a putative periplasmic localization signal sequence.
  • the previously determined molecular mass of glutamate/aspartate BP of 32 kDa (Barash & Halpern, Biochim. Biophys.
  • Glucose BP has the largest number of excellent conjugates. These conjugates all involve fluorophores known to be particularly environmentally sensitive (acrylodan, NBD, pyrene, and the styryl dyes). The incidence of excellent sensors is evenly distributed between allosteric and peristeric sites. All endosteric sites give rise to excellent sensors.
  • the dissociation constant of a conjugate determines the operating concentration range over which the sensor can respond accurately.
  • the operating range guaranteed to give less than a 5% error spans concentrations that fall within five-fold of the K d value (Marvin et al., Proc. Natl. Acad. Sci. USA 94:4366-4371, 1997). If the range required for accurate determination is wider than that span, then a composite biosensor can be constructed using receptors of varying affinities, as has been demonstrated for maltose BP (Marvin et al., Proc. Natl. Acad. Sci. USA 94:4366-4371, 1997). There are three factors affecting the dissociation constant: the nature of the conjugate, the choice of emission bands for a ratiometric sensor (eq. 2), and additional mutations. For particular applications, these three factors can be manipulated to construct an appropriate sensor.
  • Glucose sensor Among the analytes applicable to clinical medicine, glucose is one of the most important, particularly with regard to diagnosing and treating diabetes.
  • the normal range of glucose concentration in adult human serum is 4 to 6 mM (Burtis & Ashwood, Teitz Textbook of Clinical Chemistry, 2 nd Ed. W.B. Saunders Co., Philadelphia, Pa., 1994).
  • the ratiometric parameters the observation window is easily extended from 5.0 to 17.4 mM, allowing all clinically relevant ranges to be observed with one sensor ( FIG. 8A ).
  • the neuroexcitatory amino acid glutamate has normal serum concentrations of 20 to 220 ⁇ M (Burtis & Ashwood, Teitz Textbook of Clinical Chemistry, 2 nd Ed. W.B. Saunders Co., Philadelphia, Pa., 1994).
  • Glutamine is often measured in cerebrospinal fluid (Smith & Forman, Clin. Lab. Sci.
  • This biosensor can be used for such a purpose by mutagenesis to adjust the K d , or by sample dilution.
  • Maltose concentration is relevant to a deficiency in acid maltase, with the normal plasma concentration about 2 ⁇ M (Rozaklis et al., Clin. Chem. 48:131-139, 2002).
  • Fluorescent conjugates of maltose BP mutants having affinities in the 2 ⁇ M range have been described by Marvin et al. (Proc. Natl. Acad. Sci. USA 94:4366-4371, 1997).
  • Ratiometric sensing of ribose using a single ribose BP derivative is illustrated by the T135C-acrylodan conjugate ( FIG. 8B ).
  • emission wavelength bands in the fluorescence ratio eqs. 4, 5
  • the app K d for ribose can be adjusted over a range from 41 to 146 ⁇ M ( FIG. 8B ).
  • High concentrations of phosphate are environmentally deleterious, and could be monitored using phosphate BP conjugates, as noted above for clinical applications.
  • Iron concentration limits primary productivity in certain regions of the oceans (Martin, Iron as a Limiting Factor in Primary Productivity and Biogeochemical Cycles in the Sea . Falkowski & Woodhead, eds., pp.
  • Available ferric ion can be determined using a biosensor derived from Fe(III) BP, such as conjugate E203C-acrylodan (K d ⁇ 138 ⁇ M, ⁇ I std 0.4).

Abstract

Biosensors are made by attaching covalently or non-covalently at least one reporter group to one or more specific positions of a bacterial periplasmic binding protein (bPBP). Upon binding of ligand to the biosensor, there is a change in the signal transduced by the reporter group.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of provisional Appln. No. 60/418,359, filed Oct. 16, 2002.
  • FEDERALLY SUPPORTED RESEARCH OR DEVELOPMENT
  • The U.S. Government has certain rights in this invention as provided for by the terms of NIH-RO1-GM49871 and ONR-N00014-98-1-0110.
  • TECHNICAL FIELD
  • The present invention relates to biosensors and to methods of making and using same.
  • BACKGROUND
  • Biosensors are analytical tools that can be used to measure the presence of a single molecular species in a complex mixture by combining the exquisite molecular recognition properties of biological macromolecules with signal transduction mechanisms that couple ligand binding to readily detectable physical changes (Hall, Biosenisors, Prentice-Hall, Englewood Cliffs, N.J.; Scheller et al., Curr. Op. Biotech. 12:35-40, 2001). Ideally, a biosensor is reagentless and, in contrast to enzyme-based assays or competitive immunoassays, does not change composition as a consequence of making the measurement (Hellinga & Marvin, Trends Biotech. 16:183-189, 1998). Most biosensors combine a naturally occurring macromolecule such as an enzyme or an antibody, with the identification of a suitable physical signal particular to the molecule in question, and the construction of a detector specific to that system (Meadows, Adv. Drug Deliv. Rev. 21:177-189, 1996). Recently, molecular engineering techniques have been explored to develop macromolecules that combine a wide range of binding specificities and affinities with a common signal transduction mechanism, to construct a generic detection system for many different analytes (Hellinga & Marvin, Trends Biotech. 16:183-189, 1998).
  • Escherichia coli periplasmic binding proteins are members of a protein superfamily (bacterial periplasmic binding proteins, bPBPs) (Tam & Saier, Microbiol. Rev. 57:320-346, 1993) that has been shown to be well suited for the engineering of biosensors (U.S. Pat. No. 6,277,627). These proteins comprise two domains linked by a hinge region (Quiocho & Ledvina, Molec. Microbiol. 20:17-25, 1996). The ligand-binding site is located at the interface between the two domains. The proteins typically adopt two conformations: a ligand-free open form, and a ligand-bound closed form, which interconvert via a hinge-bending mechanism upon ligand binding. This global, ligand-mediated conformational change has been exploited to couple ligand binding to changes in fluorescence intensity by positioning single, environmentally sensitive fluorophores in locations that undergo local conformational changes in concert with the global change (Brune et al., Biochemistry 33:8262-8271, 1994; Gilardi et al., Prot. Eng. 10:479-486, 1997; Gilardi et al., Anal. Chem. 66:3840-3847, 1994; Marvin et al., Proc. Natl. Acad. Sci. USA 94:4366-4371, 1997, Marvin and Hellinga, J. Am. Chem. Soc. 120:7-11, 1998; Tolosa et al., Anal. Biochem. 267:114-120, 1999; Dattelbaum & Lakowicz, Anal. Biochem. 291:89-95, 2001; Marvin & Hellinga, Proc. Natl. Acad. Sci. USA 98:4955-4960, 2001; Salins et al., Anal. Biochem. 294:19-26, 2001). Conformational coupling mechanisms can also be devised to alter the flow of current between the surface of an electrode derivatized with the engineered bPBP containing a covalently attached redox cofactor (Benson et al., Science 293: 1641-1644, 2001).
  • The present invention provides a method of utilizing bPBPs to generate biosensors for a variety of chemical classes including sugars, amino acids, dipeptides, cations, and anions. These biosensors have widespread utility including in clinical, industrial, and environmental settings.
  • SUMMARY OF THE INVENTION
  • The present invention relates to biosensors, making them from mutant or wildtype members of the bacterial periplasmic binding protein (bPBP) superfamily, and using them to assay for (i.e., detect and/or quantitate) ligand. The tertiary structure of bPBPs is comprised of two domains linked by a hinge region with a ligand-binding pocket located at an interface between the two domains. They typically adopt two conformations: a ligand-free open form and a ligand-bound closed form, which interconvert via a hinge-bending mechanism which depends on whether ligand is bound or not at the site. Biosensors are made by covalently or non-covalently attaching at least one reporter group to one or more specific positions of a bPBP. Upon binding of ligand to the biosensor, there is a change in the signal transduced by the reporter group which can be analyzed by assessing any of its observable properties (e.g., optical or electrochemical properties). Biosensors are classified according to the relationship between the attachment site of the reporter group and the binding site(s) of the ligand (i.e., allosteric, endosteric, or peristeric) or distance between those sites (i.e., distal or proximal).
  • In accordance with the invention, the event of ligand binding to biosensor changes the local environment of the position-specific attached reporter group. The signal of the reporter group may be generated by one or more fluorophores and/or redox cofactors. The biosensor may be operated under physiological conditions without additional reagents.
  • Objects and advantages of the present invention will be clear from the description that follows.
  • BRIEF DESCRIPTIONS OF THE DRAWINGS
  • FIG. 1 shows the 3-D structures of eleven bPBPs indicating locations of allosteric, endosteric, and peristeric sites used. Each protein is shown in the closed form, with bound ligand indicated by ball-and-stick structures. The two domains of each bPBP are oriented vertically with the first (containing the N-terminus) above the second (containing the C-terminus). A hinge segment connects the domains. The structure of histidine BP is used to represent the as yet unsolved structure of glutamate/aspartate BP. Residues mutated to cysteine are indicated by differently shaded spheres, and differentiated as allosteric (heavy shading), endosteric (medium shading, in GBP only), or peristeric (light shading). Structures are grouped by cluster as defined by Tam & Saier (Microbiol. Rev. 57:320-346, 1993) according to sequence-based relationships. Cluster 2: arabinose BP (ABP), glucose BP (GBP), and ribose BP (RBP). Cluster 5: dipeptide BP (DPP). Cluster 3: glutamine BP (QBP), histidine BP (HBP), and glutamate/aspartate BP (EBP). Cluster 6: phosphate BP (PBP) and sulfate BP (SBP). Cluster 1: maltose BP (MBP) and Fe(III) BP (FeBP). Molecular graphics were rendered with Molscript (Kraulis, J. Appl. Crystallogr. 24:946-950, 1991).
  • FIG. 2, shows alignment of sequences of E. coli YBEJ (putative glutamate/aspartate BP), glutamine BP, and histidine BP using clustalW (Thompson et al., Nucl. Acids Res. 22:4673-4680, 1994). Numbering begins from the putative initiation codon of the open reading frame for YBEJ, including its leader sequence. The underlined methionine is the initiation codon for expression of YBEJ used in the study. Residues in each protein that were mutated to cysteine for fluorophore conjugation are in bold font. The letters “a” and “p” beneath these residues indicate their classification as allosteric or peristeric, respectively.
  • FIG. 3 shows structural formulae of thiol-reactive fluorophores. Approximate wavelengths of maximal fluorescence excitation and emission, respectively, of the protein-bound fluorophores are (in nm): pyrene (340, 390); acrylodan (390, 500); fluorescein (485, 520); NBD (490, 540); NBDE (490, 530); JPW4039 (485, 590); JPW4042 (470, 640); and JPW4045 (470, 640).
  • FIGS. 4A and 4B show a definition of fluorimetric parameters. FIG. 4A shows parameters λstd, I1 and I2 used to determine the standard intensity change ΔIstd. FIG. 4B shows parameters A1, A2, 0A, and A used to determine ΔR. Each of the areas A encompasses the respective area 0A.
  • FIGS. 5A and 5B show fluorimetric titration of glucose BP and glutamate/aspartate BP conjugates. FIG. 5A shows titration of glucose BP W183C-acrylodan with glucose. FIG. 5B. Titration of glutamate/aspartate BP T129C-NBD with amino acids. Data points:  glutamic acid; + aspartic acid; ♦ asparagine; x glutamine. In FIG. 5A and FIG. 5B the lines shown are the best fit binding isotherms.
  • FIGS. 6A-6C shows occurrence of fluorimetric parameters in the set of 320 fluorescent conjugates. FIG. 6A shows distribution of the shift in wavelength of maximum fluorescent intensity (maxλsaturatedmaxλapo). FIG. 6B shows distribution of the intensity change parameter ΔIstd. FIG. 6C shows distribution of the ratiometric change parameter ΔRmax. For each parameter, the upper bound of each interval is indicated.
  • FIG. 7 shows occurrence of changes in ligand affinity among the three classes of fluorophore attachment site. Legend: endosteric sites, filled bars; peristeric sites, hatched bars; allosteric sites, open bars. In the case of arabinose BP, the value for wtKd is that of the C64A mutant, in which all conjugates were made. Data for dipeptide BP and Fe(III) BP are not included. For the former, the Kd for Gly-Leu dipeptide in the wild-type has not been reported. In the case of Fe(III) BP, the Kd of the unconjugated mutant E57D was not determined. For each interval on the x-axis, the upper bound is indicated. For example, the interval labeled “0” contains values of log(mutKd/wtKd)>−1 and ≦0.
  • FIGS. 8A and 8B show ratiometric titration of bPBP fluorophore conjugates using different pairs of emission wavelength bands. FIG. 8A shows glucose BP-W183C conjugated to acrylodan, titrated with glucose at the following ratios of fluorescence emission (wavelengths in nm): ⋄, F450-459/F550-559 (appKd˜5.0 mM); □, F450-459/F486-495 (appKd˜10.4 mM); ◯, F472-481/F450-459 (appKd˜17.4 mM). Lines show fit to equation 4. The normal serum glucose range (euglycemia) of 4 to 6 mM is delimited by vertical lines. FIG. 8B shows ribose BP-T135C conjugated to acrylodan, titrated with ribose at the following ratios of fluorescence emission (wavelengths in nm): □, F501-510/F450-459 (appKd˜41 μM); ◯, F450-459/F501-510 (appKd˜254 μM); ⋄, F450-459/F547-556 (appKd˜461 μM).
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to biosensors constructed using engineered bPBPs, for example, E. coli bPBPs. In accordance with the invention, conjugates are constructed that can be used to monitor binding of ligands to bPBPs. Conjugates can be produced by introducing mutations into a bPBP at one or more specific positions in the protein structure where covalently attached reporter groups (e.g., fluorophores or redox cofactors) respond to a conformational change of the bPBP which occurs upon ligand binding. Other methods for covalently or non-covalently attaching at least one reporter group to one or more amino acid residue positions in the primary amino acid sequence of a mutant or wildtype bPBP include: addition or substitution of any activatable crosslinkers, use of designer or non-natural tRNAs, introduction of coordination sites, etc.
  • The universality of the engineered conformational coupling mechanism in bPBPs is disclosed herein. As described in the Example that follows, ten bPBPs of known structure have been used, and eight different environmentally sensitive fluorophores have been introduced at a variety of locations predicted to link local conformational changes to ligand-mediated hinge-bending motion. Bioinformatics techniques can be used to predict the location of linked sites in bPBPs the structures of which are not known, thereby making it possible to use the large number of paralogs and homologs that have recently been identified in this family by genomic sequencing studies (Blattner et al., Science 277:1453-1474, 1997; Quentin et al., J. Mol. Biol. 287:467-484, 1999). Together with the opportunities of structure-based redesign of ligand-binding specificity (Hellinga & Richards, J. Mol. Biol. 222:763-785, 1991; Marvin & Hellinga, Proc. Natl. Acad. Sci. USA 98:4955-4960, 2001), the Example provided below demonstrates the vast potential of the bPBP superfamily as the basis for a system of biosensors suited to a broad range of applications.
  • Furthermore, the ligand-binding pocket may be engineered to bind ligands which are not bound by the wild-type bPBP. The ligand-binding site is located at the interface between the bPBP's two domains. Mutating amino acid residues at that interface which are near (i.e., in or around) the binding site of wild-type bPBP may generate new contacts with ligand (e.g., Zn++ for MBP) and destroy or alter binding with cognate ligand (e.g., maltose for MBP). This can be used to change the specificity of the ligand-binding pocket. For example, maltose binding protein has been mutated to specifically bind to noncognate ligand: e.g., metal Zn++ ion, trinitrotoluene, L-lactate, and serotonin (Marvin & Hellinga, Proc. Natl. Acad. Sci. USA 98:4955-4960, 2001; Looger et al., Nature 423:185-190, 2003; Dwyer et al., Proc. Natl. Acad. Sci. USA 100:11255-11260, 2003). Thus, biosensors which bind noncognate ligand can be made by mutating amino acid residues at the interface of the two bPBP domains to generate a new ligand-binding pocket; ligand bound by such biosensors may not bind to wild-type bPBP.
  • Other mutations in the bPBP may be made to affect function of the biosensor: e.g., mutations may increase or decrease binding affinity or specificity; enhance or reduce signal transduction; add a new functionality by fusion with another carbohydrate, lipid, or protein domain; improve thermostability or thermolability; introduce a catalytic activity; shorten or lengthen operational life; widen or narrow the conditions for operation; or any combination thereof. Preferred is mutating amino acid residues at positions of the bPBP where a reporting group is not attached (e.g., at least one missense mutation which is not a cysteine conjugated through a thiol bond to a fluorophore).
  • In one embodiment, the present invention relates to a method of constructing a reagentless fluorescent biosensor. The method comprises identifying sites on a bPBP that undergo a local conformational change in concert with a ligand-mediated hinge-bending motion. Cysteine residues can be introduced at one or more such sites and a fluorophore coupled thereto so that fluorescence intensity of the fluorophore changes upon ligand binding.
  • bPBPs suitable for use in the present method can be selected or designed. The bPBP superfamily is well suited for the redesign of ligand-binding specificities either by computational methods or by other means or both based on the ligand to be detected (see, for example, analytes referenced in Table 1). Sites on the bPBP appropriate for attachment of one or more reporters (e.g., fluorophores or redox cofactors) include allosteric sites, peristeric sites, and endosteric sites (a reporter can also be present at a non-signaling site for use, for example, as a reference). In the case of an allosteric site, the reporter (e.g., fluorophore) can be placed at one or more locations distant from the ligand-binding site (i.e., distal from the ligand-binding pocket) that undergo local conformational changes upon ligand binding. In the case of a peristeric site, the reporter (e.g., fluorophore) can be positioned on the “rim” of the binding site but not such that it directly interacts with the ligand. With an endosteric site, the reporter (e.g., fluorophore) can be present in the binding site so that it interacts directly with the ligand. The latter two examples show attachment proximal to the ligand-binding pocket.
  • TABLE 1
    Potential applications of biosensors for bPBP ligands
    application
    analyte clinical industrial environmental
    arabinose Deanda et al.,
    1996
    glucose Burrin & Price, 1985 AOAC, 1995
    maltose Nelson et al., 1977 AOAC, 1995
    ribose AOAC, 1995
    glutamate Burtis & Ashwood, 1994 AOAC, 1995
    glutamine Smith and Forman, 1994
    histidine Taylor et al., 1991
    dipeptides
    phosphate Burkhardt et al., 1979 APHA, 1992
    sulfate EPA, 1999
    Fe(III) Martin, 1992
  • Allosteric, peristeric, and endosteric sites can be designed in at least two different ways, as detailed in the Example that follows. Generally, a structure-based design approach can be used in which the structures of the open and closed states (for allosteric designs) or the closed state only (for peristeric and endosteric designs) are examined. Alternatively, a sequence-based design approach can be used wherein homology relationships can be exploited to predict the location of cysteine mutations in proteins the three-dimensional structures of which have not been determined, provided that such mutations have been characterized in proteins of known structure.
  • As indicated above, reporters suitable for use in the invention include, but are not limited to, fluorophores and redox cofactors. In the case of fluorophores, the choice is dependent, at least in part, on the nature of the location within the particular protein. While one fluorophore may function better in a certain location than another, one skilled in the art can readily select the preferred fluorophore for a particular application (see, for example, U.S. Pat. No. 6,277,627). In the Example that follows, eight different fluorophores are used in the design of fluorescent sensors for:
  • Arabinose Arabinose binding protein (ABP)
    Dipeptides Dipeptide binding protein (DPP)
    Glutamate and asparate Glu/Asp binding protein (EBP)
    Glutamine Glutamine binding protein (QBP)
    Fe(III) Iron binding protein (FeBP)
    Histidine Histidine binding protein (HBP)
    Maltose Maltose binding protein (MBP)
    Glucose Glucose binding protein (GBP)
    Phosphate Phosphate binding protein (PhBP)
    Sulfate Sulfate binding protein (SBP).

    The invention, however, is in no way limited to these specific embodiments.
  • Redox reporters for use in the invention can be a redox-active metal center or a redox-active organic molecule. It can be a natural organic cofactor such as NAD, NADP, FAD or a natural metal center such as Blue Copper, iron-sulfur clusters, or heme, or a synthetic center such as an organometallic compound such as a ruthenium complex, organic ligand such as a quinone, or an engineered metal center introduced into the protein or engineered organic cofactor binding site. Cofactor-binding sites can be engineered using rational design or directed evolution techniques. The redox reporter can be covalently or non-covalently attached to the protein, either by site-specific or adventitious interactions between the cofactor and protein. It can be intrinsic to the protein such as a metal center (natural or engineered) or natural organic (NAD, NADP, FAD) or organometallic cofactor (heme), or extrinsic (such as a covalently conjugated, synthetic organometallic cluster). The redox reporter can be, for example, bound (e.g., covalently) at a position where the amino acid residue is on the protein's surface.
  • The redox reporter can be a metal-containing group (e.g., a transition metal-containing group) that is capable of reversibly or semi-reversibly transferring one or more electrons. A number of possible transition metal-containing reporter groups can be used. Advantageously, the reporter group has a redox potential in the potential window below that subject to interference by molecular oxygen and has a functional group suitable for covalent conjugation to the protein (e.g., thiol-reactive functionalities such as maleimides or iodoacetamide for coupling to unique cysteine residues in the protein). The metal of the reporter group should be substitutionally inert in either reduced or oxidized state (i.e., advantageously, exogenous groups do not form adventitious bonds with the reporter group). The reporter group can be capable of undergoing an amperometric or potentiometric change in response to ligand binding. In a preferred embodiment, the reporter group is water soluble, is capable of site-specific coupling to a protein (e.g., via a thiol-reactive functional group on the reporter group that reacts with a unique cysteine in the protein), and undergoes a potentiometric response upon ligand binding. Suitable transition metals for use in the invention include, but are not limited to, copper (Cu), cobalt (Co), palladium (Pd), iron (Fe), ruthenium (Ru), rhodium (Rh), osmium (Os), rhenium (Re), platinum (Pt), scandium (Sc), titanium (Ti), vanadium (V), chromium (Cr), manganese (Mn), nickel (Ni), molybdenum (Mo), technetium (Tc), tungsten (W), and iridium (Ir). That is, the first series of transition metals, the platinum metals (Ru, Rh, Pd, Os, Ir, and Pt), along with Fe, Re, W, Mo, and Tc, are preferred. Particularly preferred are metals that do not change the number of coordination sites upon a change in oxidation state, including ruthenium, osmium, iron, platinum and palladium, with ruthenium being especially preferred.
  • The reporter group can be present in the biosensor as a covalent conjugate with the protein or it can be a metal center that forms part of the protein matrix (for instance, a redox center such as iron-sulfur clusters, heme, Blue copper, the electrochemical properties of which are sensitive to its local environment). Alternatively, the reporter group can be present as a fusion between the protein and a metal binding domain (for instance, a small redox-active protein such as a cytochrome). Preferably, the reporter group is covalently conjugated to the protein via a maleimide functional group bound to a cysteine (thiol) on the protein. In any case, the reporter group is attached to the protein so that it is located between the protein and the electrode.
  • Engineered proteins of the invention can be produced by site-specifically introducing a reporter group(s) by total synthesis, semi-synthesis, or gene fusions (see, for example, Adams et al., Nature 39:694-697, 1991; Brune et al., Biochemistry 33:8262-8271, 1994; Gilardi et al., Anal. Chem. 66:3840-3847, 1994; Godwin et al., J. Am. Chem. Soc. 118:6514-6515, 1996; Marvin et al., Proc. Natl. Acad. Sci. U.S.A. 94:4366-4371, 1997; Post et al., J. Biol. Chem. 269:12880-12887, 1994; Romoser, J. Biol. Chem. 272:13270-13274, 1997; Thompson et al., J. Biomed. Op. 1:131-137, 1996; Walkup et al., J. Am. Chem. Soc. 119:5445-5450, 1997).
  • Assays for ligand may be performed with the biosensor. A sample is contacted with the biosensor under appropriate assay conditions. Ligand present in the sample, if any, may be detected by binding to the biosensor and measuring the signal transduced by ligand-bound biosensor in the assay. For detection purposes, binding does not need to be quantitative because a simple determination of whether the ligand is present or absent (within detection limits) needs to be performed. Otherwise, comparison to a series of control samples (e.g., known quantities of ligand) may be required to quantitate the amount or concentration of ligand in the sample. Given the volume of the sample, the amount (i.e., mass) of ligand and the concentration of ligand are interconvertible. A blank sample containing no ligand may be used to determine background signal. Standards may be used to construct a standard curve (e.g., hyperbolic) used to quantitate unknown samples. Although homogeneous assay formats (i.e., those requiring no separation of bound and non-bound ligand) are preferred, separation in a heterogeneous assay format may be required if substances which significantly interfere with signal transduction and/or measurement are present in the sample. Signal transduction preferably does not require the addition of exotic reagents so assays of body fluids may be performed with minimal sample preparation and under physiological conditions. They may even be performed in vivo if the biosensor is adapted to an implantable medical device. Alternatively, a biosensor in contact with the skin may assay interstitial fluid or perspiration. Lavage may be used to sample mucosal tissues.
  • The sample can be obtained in a laboratory setting (e.g., clinic or research institution); from an environmental source (e.g., air, aquafers and other bodies of water, animal or plant products grown on the land, soil); from an industrial source (e.g., the food, biopharmaceutical, chemical, or other manufacturing industries). The analyte to be assayed is identical to the ligand, comprised of multiple copies of the ligand, chemically related to the ligand such that it is identified by a change in signal transduction (e.g., a related chemical structure is more strongly or more weakly bound by the biosensor as compared to its “correct” ligand), or any combination thereof. The change in signal transduction may be correlated to the change in chemical structure such that the non-identical analyte is identified (see below description of integrative assays). Examples of ligands which may be detected or quantitated include: amino acids; carbohydrates; bioactive solid and gaseous compounds which are soluble in an aqueous sample; contraband or controlled substances (i.e., substances which are illegal to use or possess, or which are highly regulated); environmental pollutants (e.g., phosphates, sulfates); explosives (e.g., TNT); food contaminants and byproducts (e.g., carcinogens, plant toxins, teratogens); lipids; metal ions (e.g., divalent cations, ferric ions); microbial toxins (e.g., toxic products of viruses, bacteria, or protozoa); neurotransmitters (e.g., serotonin); nucleosides or nucleotides (e.g., NAD, NADP, FAD); peptides or steroids (e.g., growth factors, hormones, morphogenic or developmental signals); and therapeutic drugs. Objects (e.g., baggage, mail, other containers); people or vehicles passing through a checkpoint; and borders or secure areas may be inspected for biological agents, contraband, explosives, poisons, and toxins in security or military applications.
  • One or more biosensors may be covalently or noncovalently attached to a solid or porous substrate. The substrate may be flat and planar (e.g., filter membrane, glass slide, semiconductor chip); cylindrical (e.g., optical fiber, plastic rod); spherical (e.g., crosslinked polymer or glass bead); or formed as a container (e.g., cell or cuvette, multiwell plate). The substrate may be fabricated for analysis by instruments which measure the signal transduced by the reporter group (e.g., microscope, photometer, spectrometer). Individual biosensors may be coded by an attached marker (e.g., bar code, radio frequency or RFID, or biopolymer) which can be decoded by a reader (e.g., scanner of light-and-dark patterns, radio receiver, specific binding probe or automated sequencer) or separated by a sorter in accordance with their marker. The code identifying each biosensor may be used in parallel analysis by rapidly assaying a sample for a plurality of ligands. Multiple biosensors with different ligand-binding specificities are used in the same assay to detect and/or quantitate multiple ligands at the same time. Alternatively, attaching different biosensors at particular spots on the substrate may be used to identify their ligand-binding specificities by where the signals are being produced. Signals may be authenticated by repeating the assay, using multiple biosensors with the same specificity for redundant assays, or correlating the results from multiple biosensors with overlapping specificities for integrative assays. In the latter, particular reactivity patterns of the biosensors are correlated with the identity of the analyte bound by them. Analytes that are more closely related in their chemical structure to the ligand will bind more strongly to the cognate biosensor. Signals from a plurality of biosensors with overlapping, known ligand-binding specificities are integrated to deduce the identity of the analyte.
  • The invention relates, in further embodiments, to biosensors constructed using the above-described methods and to the use thereof in analyte detection in, for example, clinical, industrial, and environmental settings. Particular utilities are described in the specific Example that follows. Provided is a description of a number of sites that can be used for optical glucose sensors based on GBP (W183C conjugated to acrylodan has been used successfully in fiber-optic prototypes of a glucose sensor).
  • To the extent that specific biosensors constructed in accordance with the present approach may be present in the public domain (e.g., may be disclosed in Marvin et al., Proc. Natl. Acad. Sci. USA 94:4366-4371, 1997 or in U.S. Pat. No. 6,277,627), such biosensors are not within the scope of the present invention.
  • Certain aspects of the invention can be described in greater detail in the non-limiting Example that follows.
  • EXAMPLE Experimental Details
  • Molecular Cloning. PCR was used to amplify wild-type genes for bPBPs from genomic DNA of E. coli strain CSH100 (arabinose, dipeptide, histidine, ribose, sulfate, and glutamate/aspartate BP); strain W1485 (glucose and glutamine BP) and strain RU1012 (phosphate BP), or of H. influenzae strain Rd (Fe(III) BP). Amplified products were cloned into one of the protein expression vectors pAED4 (Doering, “Functional and structural studies of a small f-actin binding domain” in Ph.D. thesis, Massachusetts Institute of Technology, 1992); pKK223-3 (Brosius & Holy, Proc. Natl. Acad. Sci. USA 81:6929-6933, 1984); or pET vectors (Studier et al., Meth. Enzymol. 185:60-89, 1990) (Novagen). N-terminal oligonucleotide primers were designed to clone only the processed periplasmic form, deleting the signal sequence. The C-terminal primer was designed to append the sequence Gly-Ser-Gly-(His)n or Gly-Ser-(His)n, where n=5, 6, or 10. Two tandem stop codons (TAATGA) follow the last His codon. Maltose BP mutants were made in and expressed from plasmid pMAL-c2X (New England BioLabs). E. coli strains XL1-BLUE (Stratagene) and DH5α (Hanahan, J. Mol. Biol. 166:557-580, 1983) were used for plasmid construction. Single amino acid substitutions were generated by overlapping PCR mutagenesis (Ho et al., Gene 77:51-59, 1989). All clones and mutations were confirmed by nucleotide sequencing. In the case of arabinose BP, the single cysteine in the wild type sequence was replaced by alanine to eliminate the possibility of reporter group conjugation to this thiol (Miller et al., J. Biol. Chem. 254:7521-7528, 1979). Additionally the sequence of Fe(III) BP was mutated by substitution of Glu57 with Asp to raise the Ki to a concentration range conveniently measured using Fe(III) citrate.
  • Protein Expression. Plasmids were transformed into E. coli strain BL21-DE3, grown in nutrient broth overnight at 37° C., then diluted 100-fold into fresh medium and grown further at 37° C. or 25° C. Expression was induced by the addition of isopropyl β-D-1-thiogalactopyranoside to 1 mM when the optical density of the culture at 600 nm reached 0.4. After 2 to 4 hours, cells were harvested by centrifugation, resuspended in 20 mM 3-morpholinopropanesulfonic acid (MOPS), 100 mM NaCl, pH 6.9 and stored frozen or lysed immediately for protein purification.
  • Protein Purification. Cells were lysed by sonication or by passage through a French pressure cell. The lysate was treated by adding Polymin P to 0.33% (w/v), chilling on ice for 15 min, then centrifuging to pellet cell debris. The supernatant was loaded on a Ni(II)-charged column of Chelating Sepharose™ Fast Flow (Amersham Pharmacia Biotech) equilibrated with 20 mM MOPS, 500 mM NaCl, 10 mM imidazole, pH 7.5. The column was washed with loading buffer, then with the same containing 60 mM imidazole, followed by the same with 100 mM imidazole. Finally, protein was eluted with loading buffer containing 400 mM imidazole, and was collected in fractions and assessed for purity by gel electrophoresis. All preparations were at least 95% pure by this criterion. Protein-containing fractions were dialyzed exhaustively against buffer (20 mM MOPS, 100 mM NaCl, pH 6.9, or 20 mM NaH2PO4, 100 mM NaCl, pH 6.9) or desalted by gel filtration to remove bound ligand.
  • Fluorophore conjugation to cysteine-substituted bPBPs. Thiol-reactive fluorophores obtained from Molecular Probes (Eugene, Oreg.) were 5-iodoacetamidofluorescein (fluorescein); N-(1-pyrene) iodoacetamide (pyrene); N,N′-dimethyl-N-(iodoacetyl)-N′-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)ethylenediamide (NBD); N-((2-(iodoacetoxy)ethyl)-N-methyl)amino-7-nitrobenz-2-oxa-1,3-diazole (NBDE); and 6-acryloyl-2-dimethylaminonaphthalene (acrylodan). The styryl and naphthyl dyes JPW4039, JPW4042, and JPW4045 (FIG. 3) were synthesized at the University of Connecticut. All fluorophore conjugation steps were typically carried out at room temperature. To protein at a concentration of 100 μM was added tris-(2-carboxyethyl)phosphine HCl to a five-fold molar excess to reduce intermolecular disulfide bonds. A thiol-reactive fluorophore (20 to 25 mM in acetonitrile or dimethyl sulfoxide) was added in small aliquots to a five-fold molar excess over protein. Conjugation proceeded in the dark at room temperature for 4 hours, or overnight at 4° C. Separation of protein from unreacted fluorophore was achieved by exhaustive dialysis or by size-exclusion chromatography. The efficiency of reporter group attachment was assessed by determination of unreacted thiol with Ellman's reagent (Ellman, Arch. Biochem. Biophys. 74:443-450, 1958) or by measuring the ratio of fluorophore to protein from absorbance spectra of the purified conjugate.
  • Depletion of sulfate and phosphate. Solutions of sulfate BP and phosphate BP and their buffers were treated to decrease the concentration of contaminating sulfate and phosphate, respectively. Sulfate BP buffer (20 mM Tris-HCl, pH 8.0) was passed through the chloride form of Dowex 1X2-100 strongly basic anion-exchange resin. Sulfate BP solutions were treated by dialysis against treated buffer; Dowex resin held in a separate dialysis tube was also included. Phosphate BP solutions and buffer (20 mM MOPS, 100 mM NaCl, pH 6.9) were depleted of phosphate by addition of 7-methylguanosine to 1 mM and dialyzed against bacterial nucleoside phosphorylase (1 unit ml−1) (Sigma-Aldrich) partitioned in a separate dialysis tube (Brune et al., Biochem. 33:8262-8271, 1994).
  • Fluorimetry. All measurements were conducted with an SLM Aminco-Bowman series 2 fluorimeter, with sample stirring at 25° C. Fluorescence emission spectra were acquired with excitation and emission slit widths of 4 and 8 nm, respectively. Photomultiplier tube potential was maintained between 400 and 800 volts. Protein concentrations were in the range of 50 to 1000 nM. Fluorophore-specific excitation was at the following approximate wavelengths: tryptophan, 290 nm; acrylodan, 390 nm; fluorescein, 485 nm; pyrene, 340 nm; NBD and NBDE, 490 nm; JPW4039, 485 nm; JPW4042, 470 nm; JPW4045, 470 nm.
  • To measure ligand binding affinity, ligand was serially added to 3 ml of bPBP at a concentration of 50 to 1000 nM, and emission intensities recorded. Corrections were made for dilution of the protein and for background signal from buffer. Binding curves were fit to binding isotherms using equation 3 or 4, as appropriate.
  • Fe(III) BP has a dissociation constant for Fe(III) on the order of 10−21 M (Adhikari et al., J. Biol. Chem. 270:25142-25149, 1995), hindering accurate fluorescence-based measurements of affinity at nanomolar protein concentrations. Hence we used Fe(III) citrate (logK˜10.25) (Martell and Smith, Critical Stability Constants, Plenum Press, New York, 1977) as the ligand in a competition assay.
  • Results
  • Family of biosensors. A set of eleven bPBPs with widely varying ligand-binding specificities was selected for engineering biosensor function (Table 2). All were from E. coli except Fe(III) BP, which is from Haemophilis influenzae. Binding specificities and affinities of these proteins for their respective ligands have been characterized (references in Table 2). Three proteins bind monosaccharides (arabinose, glucose and ribose BP), one binds di- and trisaccharides of glucose (maltose BP), three bind amino acids (glutamate/aspartate, histidine, and glutamine BP), one binds di- and tripeptides (dipeptide BP), two bind oxyanions (phosphate and sulfate BP), and one binds a metal ion (Fe(III) BP). Most of these bPBPs bind at most two or three related ligands with high affinity (micromolar or better). For example, phosphate BP binds phosphate and arsenate but not other oxyanions (Luecke & Quiocho, Nature 347:402-406, 1990), while glucose BP binds glucose and galactose but not other monosaccharides (Anraku, J. Biol. Chem. 243:3116-3122, 1968). Dipeptide BP is an exception in that it binds a wide variety of di- and tripeptides (Smith et al., Microbiology 145:2891-2901, 1999). Measured ligand dissociation constants in these proteins are typically in the range of 0.1 to 1 μM. An exception is Fe(III) BP, where the Kd for Fe(III)(aq) is estimated to be 10−21 M in competition assays with Fe(III) chelates (Adhikari et al., J. Biol. Chem. 270:25142-25149, 1995).
  • TABLE 2
    References and PDBa files for bPBP structures, genes, and ligand binding
    crystal structure
    bPBP open form closed form DNA sequence ligand affinity
    arabinose BP Quiocho and Scripture et al., Clark et al.,
    Vyas, 1984 1ABE 1987 1982; Miller et
    al., 1983
    dipeptide BP Nickitenko et Dunten & Abouhamad et Guyer et al.,
    al., 1995 1DPE Mowbray, 1995 al., 1991 1986; Smith et
    1DPP al., 1999
    Glu/Asp BP Barash Halpern,
    1975; Willis
    Furlong, 1975
    Fe(III) BP Bruns et al., Bruns et al., 1997 Sanders et al., Adhikari et al.,
    2001 1D9V 1MRP 1994 1995
    glucose BP Vyas et al., 1988; Scholle et al., Anraku, 1968
    Vyas et al., 1994 1987
    1GLG
    histidine BP Yao et al., 1994 Joshi & Ames Miller et al.,
    1HSL 1996 1983
    maltose BP Sharff et al., Spurlino et al., Duplay et al., Schwartz et al.,
    1992 1OMP 1991; Quiocho et 1984 1976
    al, 1997 1ANF
    phosphate BP Ledvina et al., Luecke & Magota et al., Medveczky &
    1996 1OIB Quiocho, 1990 1984 Rosenberg, 1969
    1IXH
    glutamine BP Hsiao et al., Sun et al., 1998 Nohno et al., Weiner et al.,
    1996 1GGG 1WDN 1986 1971
    ribose BP Bjorkman & Mowbray & Cole, Groarke et al., Willis &
    Mowbray, 1998 1992 2DRI 1983 Furlong, 1974
    1URP
    sulfate BP Pflugrath & Hellinga & Jacobson &
    Quiocho, 1985; Evans, 1985 Quiocho, 1988
    He & Quiocho,
    1993 1SBP
    • a: Protein Data Bank (Berman et al., 2000)
    • Abouhamad et al., Molec. Microbiol. 5:1035-1047 (1991)
    • Adhikari et al., J. Biol. Chem. 270:25142-25149 (1995)
    • Anraku, J. Biol. Chem. 243:3116-3122 (1968)
    • Barash & Halpern, Biochim. Biophys. Acta 386:168-180 (1975)
    • Bjorkman & Mowbray, J. Mol. Biol. 279:651-664 (1998)
    • Bruns et al., Biochemistry 40:15631-15637 (2001)
    • Bruns et al., Nat. Struct. Biol. 4:919-924 (1997)
    • Clark et al., Biochemistry 21:2227-2233 (1982)
    • Dunten & Mowbray, Protein Sci. 4:2327-2334 (1995)
    • Duplay et al., J. Biol. Chem. 259:10606-10613 (1984)
    • Groarke et al., J. Biol. Chem. 258:12952-12956 (1983)
    • Guyer et al., J. Bacteriol. 168:775-779 (1986)
    • He & Quiocho, Protein Sci. 2:1643-1647 (1993)
    • Hellinga & Evans, Eur. J. Biochem. 149:363-373 (1985)
    • Hsiao et al., J. Mol. Biol. 262:225-242 (1996)
    • Jacobson & Quiocho, J. Mol. Biol. 204:783-787 (1988)
    • Joshi & Ames, GenBank Accession Number U47027 (1996)
    • Ledvina et al., Proc. Natl. Acad. Sci. USA 93:6786-6791 (1996)
    • Luecke & Quiocho, Nature 347:402-406 (1990)
    • Magota et al., J. Bacteriol. 157:909-917 (1984)
    • Medveczky & Rosenberg, Biochim. Biophys. Acta 192:369-371 (1969)
    • Miller et al., J. Biol. Chem. 258:13665-13672 (1983)
    • Mowbray & Cole, J. Mol. Biol. 225:155-175 (1992)
    • Nickitenko et al., Biochemistry 34:16585-16595 (1995)
    • Nohno et al., Molec. Gen. Genet. 205:260-269 (1986)
    • Pflugrath & Quiocho, Nature 314:257-260 (1985)
    • Quiocho et al., Structure 5:997-1015 (1997)
    • Quiocho & Vyas, Nature 310:381-386 (1984)
    • Sanders et al., Infect. Immun. 62:4515-4525 (1994)
    • Scholle et al., Molec. Gen. Genet. 208:247-253 (1987)
    • Scripture et al., J. Mol. Biol. 197:37-46 (1987)
    • Schwartz et al., Eur. J. Biochem. 71:167-170 (1976)
    • Sharff et al., Biochemistry 31:10657-10663 (1992)
    • Smith et al., Microbiology 145:2891-2901 (1999)
    • Spurlino et al., J. Biol. Chem. 266:5202-5219 (1991)
    • Sun et al., J. Mol. Biol. 278:219-229 (1998)
    • Vyas et al., Biochemistry 33:4762-4768 (1994)
    • Vyas et al., Science 242:1290-1295 (1988)
    • Weiner et al., Arch. Biochem. Biophys. 142:715-717 (1971)
    • Willis & Furlong, J. Biol. Chem. 249:6926-6929 (1974)
    • Willis & Furlong, J. Biol. Chem. 250:2574-2580 (1975)
    • Yao et al., Biochemistry 33:4769-4779 (1994)
  • For nine of the eleven proteins selected for this study crystal structures have been solved of the closed, ligand-bound state (Table 2). In the case of sulfate BP, the crystal structure of the E. coli protein has not been reported, so that of Salmonella typhimurium sulfate BP was adopted to model the E. coli protein. Sulfate BP from E. coli and S. typhimurium are 95% identical in amino acid sequence and hence likely to have highly similar structures, in analogy to histidine BP from these two organisms (Oh et al., J. Biol. Chem. 269:4135-4143, 1994; Yao et al., Biochemistry 33:4769-4779, 1994). Structures have been solved for the open unliganded state for six of the eleven proteins as well (Table 2).
  • Structure-based design of conformational coupling. Coupling between ligand binding and a change in the fluorescent signal of a covalently attached, environmentally sensitive fluorophore can be established if the local environment of the fluorophore changes as a result of formation of the complex and a linked conformational change. Two mechanisms can be distinguished to establish such structural linkage relationships. Direct linkage involves formation of a non-bonded contact between the bound ligand and the conjugated fluorophore. Indirect linkage involves changes in the local protein structure in the immediate vicinity of the attached fluorophore, and relies on ligand-mediated conformational changes such as the hinge-bending motion observed in the bPBPs.
  • Direct linkage relationships are readily designed by replacing a residue known to form a ligand contact with a cysteine to which the fluorophore is attached (“endosteric” attachment site). Indirect linkage relationships can be established in two ways. The most straightforward method relies on visual inspection of the ligand complex structure, and identifying residues that are located in the vicinity of the binding site, but do not interact directly with the ligand, and that are likely to be involved in conformational changes. In the case of the bPBPs, such are residues located at the perimeter of the inter-domain cleft that forms the ligand binding site. The environment of these “peristeric” sites changes significantly upon formation of the closed state. These are examples of positions which are proximal to the ligand-binding pocket. The second approach identifies sites in the protein structure that are located some distance away from the ligand-binding site (i.e., distal to the ligand-binding pocket), and undergo a local conformational change in concert with ligand binding. If the structures of both the open and closed states are known, then such “allosteric” sites can be identified using a computational method that analyzes the conformational change (Marvin et al., Proc. Natl. Acad. Sci. USA 94:4366-4371, 1997). Alternatively, once allosteric sites have been identified in one bPBP, modeling and structural homology arguments can be invoked to identify such sites in other bPBPs in which only one state has been characterized (Marvin & Hellinga, J. Am. Chem. Soc. 120:7-11, 1998). Table 3 summarizes the designs of all three classes of sites in each of the receptors used in this study. The locations of these sites in the eleven bPBPs are shown in FIG. 1.
  • TABLE 3
    Fluorophore conjugation sites
    steric design steric design
    protein mutant categorya methodb protein mutant categorya methodb
    arabinose BP D257C a 3 histidine BP E167C p 1
    F23C a 3 K229C p 1
    K301C a 3 V163C p 1
    L253C a 3 Y230C p 1
    L298C a 3 F231C p 1
    dipeptide BP D450C p 1 Y88C a 3
    K394C p 1 maltose BP D95C a 2
    R141C p 1 F92C a 2
    S111C p 1 I329C a 2
    T44C p 1 S233C p 2
    W315C p 1 phosphate BP A225C a 2
    Glu/Asp BP A207C p 4 N223C a 2
    A210C p 4 N226C a 2
    E119C p 4 S164C p 2
    F126C a 4 S39C p 2
    F131C a 4 glutamine BP N160C p 2
    F270C p 4 F221C p 2
    G211C p 4 K219C p 2
    K268C p 4 L162C p 2
    Q123C p 4 W220 p 2
    T129C a 4 Y163C p 2
    Fe(III) BP E203C p 1 Y86C a 2
    K202C p 1 ribose BP T135C p 2
    K85C a 1 D165C p 2
    V287C a 1 E192C p 2
    glucose BP Y10C e 1 A234C a 2
    N15C p 1 L236C a 2
    E93C p 1 L265C a 2
    E149C p 1 sulfate BP L65C p 1
    H152C e 1 N70C p 1
    W183C e 1 Q294C p 1
    L255C a 3 R134C p 1
    D257C a 3 W290C p 1
    V296C a 3 Y67C p 1
    aa, allosteric; e, endosteric; p, peristeric
    b1, visual inspection of the closed structure; 2, identification by automated comparison of the open and closed states; 3, structural homology; 4, sequence homology
  • Sequence-based design of conformational coupling. The number of bPBPs of known sequence greatly exceeds the number for which structures have been solved or for which functions have been assigned by genetic or biochemical characterization. To exploit this reservoir of potential biosensors, coding sequences for bPBPs must be identified and their putative ligand-binding specificities must be established. The identification of bPBPs in microbial genomes relies on finding amino acid sequence homologies to particular clusters of the bPBP family (Tam & Saier, Microbiol. Rev. 57:320-346, 1993). Ligand-binding can then be determined by direct experimentation, or be inferred either by structural relationships to bPBPs of known function, or by establishing genetic linkage to other genes of known function (Pellegrini et al., Proc. Natl. Acad. Sci. USA 96:4285-4288, 1999). Subsequently sites within the homolog that undergo local conformational change, and to which reporter functions can be attached, must be identified. The selection of sites for attaching reporter functions relies on homology to bPBPs of known structure.
  • To illustrate these principles, a glutamate biosensor was constructed starting from genome sequence data only. The genome of E. coli K12 contains the locus ybeJ encoding a protein identified as a putative bPBP based on amino acid sequence homology with glutamine and histidine BPs (26% and 23% sequence identity; 41% and 43% sequence similarity, respectively) (Blattner et al., Science 277:1453-1474, 1997). The assignment of YBEJ as an amino-acid binding protein was strengthened by the presence of conserved residues found to be associated with binding to the α-amino and α-carboxylate groups of the ligand in all bPBP amino-acid binding proteins of known structure identified in E. coli (Table 4). Of additional interest is the presence of an arginine residue in YBEJ located at a position that in the other amino acid-binding proteins interacts directly with the side chain of the bound amino acid, suggesting that YBEJ binds an amino acid bearing a negatively charged side chain. Finally, ybeJ is located adjacent to three tandem genes (gltJ, gltK, gltL) postulated to be involved in the glutamate/aspartate transport system (Lum & Wallace, GenBank Accession Number U10981, 1995), suggesting that ybeJ encodes a glutamate/aspartate BP. Putative allosteric, endosteric, and peristeric sites were identified from a structure-based sequence alignment of YBEJ with glutamine BP and histidine BP (FIG. 2).
  • TABLE 4
    Ligand interactions with residues in polar amino-acid binding proteins
    ligand group*
    sc sc sc αN αN αC sc sc αC αN
    glutamine BP D10 F13 F50 G68 T70 R75 K115 T118 G119 D157
    histidine BP D11 Y14 L52 S70 S72 R77 L117 T120 T121 D161
    lys/arg/om BP D11 Y14 F52 S70 S72 R77 L117 T120 T121 D161
    YBEJ R25 S28 S73 S91 T93 R98 T137 T140 T141 D183
    *sc: side chain, αN: α-amino, αC: α-carboxy
  • Mutagenesis and protein production All the genes for the bPBPs used in this study were cloned from E. coli or H. influenzae genomic DNA using PCR. The leader peptide sequence that directs expression into the periplasm was identified by comparison to the known N-terminus of the protein, or, in the case of YBEJ, by homology to known leader sequences (von Heijne, Nucl. Acids Res. 14:4683-4690, 1986). The protein was produced by over-expression of the processed form in the cytoplasm with an initiation methionine placed just before the N-terminus of the processed protein, under the control of a strong inducible promoter in the pAED4 (Doering, “Functional and structural studies of a small f-actin binding domain” in Ph.D. thesis, Massachusetts Institute of Technology, 1992); pET-21a (Studier et al., Meth. Enzymol. 185:60-89, 1990) (Novagen); or pKK223-3 (Blattner et al., Science 277:1453-1474, 1997) plasmids. An oligohistidine tag was fused to the carboxy terminus of the cloned receptor to permit facile purification by immobilized metal affinity chromatography (Hochuli et al., J. Chromatogr. A 411:177-184, 1987). In all cases, the receptors expressed well (at least 50 mg of pure protein per liter of fermentation). The molecular masses estimated by gel electrophoresis corresponded to the predicted mass of the expressed reading frame.
  • Cysteine point mutations were introduced by the PCR overlap method (Ho et al., Gene 77:51-59, 1989). Mutant proteins typically expressed as well as the wild type protein. All cysteine substitutions in arabinose BP were constructed in the C64A background to prevent interference from this endogenous cysteine (Miller et al., J. Biol. Chem. 254:7521-7528, 1979). In the case of Fe(III) BP, all mutations were constructed in the E57D background. In the crystal structure of Fe(III) BP, this glutamate is coordinated to the iron (Bruns et al., Nat. Struct. Biol. 4:919-924, 1997). It was found that the E57D mutation weakens the affinity of Fe(III) BP for Fe(III) from approximately 1×10−21 (Adhikari et al., J. Biol. Chem. 270:25142-25149, 1995) to approximately 3×10−8, assuming a stability constant for the 1:1 Fe(III) citrate complex of logK=10.25 (Martell & Smith, Critical Stability Constants, Plenum Press, New York, 1977). This permitted straightforward determination of Fe(III) affinity by direct titration with Fe(III) citrate at nanomolar concentrations of Fe(III) BP.
  • Signal transduction by fluorescence. To report ligand binding by the set of eleven bPBPs, fluorescent reporter groups were attached to single cysteine thiols engineered into sites that were predicted to undergo binding-dependent conformational change. Eight thiol-reactive fluorophores were examined that were chosen on the basis of the sensitivity of their emission spectra to changes in environment and spanning a wide range of emission and excitation wavelengths (FIG. 3). The results for biosensor conjugates which are illustrative of the invention are presented in Table 5 (11 receptors, 68 cysteine mutants, 320 fluorophore conjugates).
  • TABLE 5
    Spectral and binding parameters of fluorophore-conjugated bPBPs
    proteina mutant siteb fluorophore ligand λmax, apo λmax, sat ΔIstd c inc/decd ΔRmax c Kd (μM) std error
    arabinose D257C a JPW4039 arabinose 600 596 0.38 0.92 90 3
    BP Acrylodan 495 495 0.26 1.66 56 7
    Fluorescein 519 520 0.03 1.17 4.0 0.4
    NBD 538 544 0.22 + 1.15 32 2
    F23C a JPW4039 587 588 0.93 0.76 38 1
    Acrylodan 503 503 0.02 + 0.99 3.9 0.6
    Fluorescein 519 519 0.04 0.45 3.2 0.5
    NBD 543 548 0.38 0.76 5.0 0.1
    K301C a JPW4039 582 588 1.20 1.73 77 4
    Acrylodan 486 486 0.10 1.19 0.46 0.01
    Fluorescein 518 517 0.41 + 1.06 24 1
    NBD 532 538 0.08 3.15 31 1
    L253C a JPW4039 590 589 0.83 1.31 165 8
    Acrylodan 482 495 0.05 1.81 0.69 0.10
    Fluorescein 519 515 0.24 2.71 48 3
    NBD 539 539 0.41 + 1.66 775 49
    L298C a JPW4039 591 591 0.42 0.65 70 2
    Acrylodan 499 500 0.07 1.77 44 2
    Fluorescein 518 518 0.02 0.48
    NBD 543 539 0.45 + 0.41 56 4
    dipeptide D450C p JPW4039 Gly-Leu 602 604 0.20 0.29 0.91 0.20
    BP JPW4042 666 664 0.20 1.08 1.5 0.3
    JPW4045 663 666 0.23 1.18 2.0 0.5
    Acrylodan 508 521 0.06 + 1.64 11 4
    Fluorescein 520 520 0.10 + 0.04
    NBD 545 544 0.02 0.80
    K394C p JPW4039 592 598 0.37 + 1.34 30 2
    JPW4042 638 644 0.06 + 0.99 78 8
    JPW4045 631 640 0.01 + 1.07
    Acrylodan 500 500 0.23 + 0.90 23 2
    Fluorescein 522 522 0.30 + 0.21 93 6
    NBD 542 541 0.06 0.68 0.012 0.005
    R141C p JPW4039 592 596 0.06 0.69
    JPW4042 629 631 0.06 0.87
    JPW4045 610 617 0.15 1.18
    Acrylodan 502 501 0.06 0.25 2.3 1.2
    Fluorescein 522 522 0.12 0.66 38 14
    NBD 542 544 0.00 + 0.13
    S111C p JPW4039 597 598 0.24 + 0.33 34 14
    JPW4042 644 644 0.18 + 1.49 15.8 1.5
    JPW4045 634 642 0.01 1.07
    Acrylodan 499 501 0.11 + 1.61 4.8 2.3
    Fluorescein 521 521 0.07 0.18 2.6 1.9
    NBD 538 542 0.01 + 0.18
    T44C p JPW4039 594 596 0.13 0.33
    JPW4042 634 635 0.06 0.30
    JPW4045 640 636 0.13 0.82
    Acrylodan 499 501 0.01 1.52
    Fluorescein 522 522 0.05 0.21 0.64 0.38
    NBD 539 536 0.11 0.30 0.006 0.005
    W315C p JPW4039 594 593 0.26 0.45 1.00 0.19
    JPW4042 645 640 0.05 0.16
    JPW4045 640 640 0.14 0.55 3.2 1.0
    Acrylodan 503 504 0.08 0.47 0.13 0.04
    Fluorescein 521 521 0.02 0.21
    NBD 546 546 0.15 0.37 0.06 0.02
    Glu/Asp BP A207C p JPW4039 glutamate 592 593 0.05 0.35
    JPW4042 635 634 0.20 1.37
    JPW4045 637 639 0.15 1.19
    Acrylodan 498 497 0.26 + 1.61
    Fluorescein 520 520 0.12 0.25
    NBD 529 542 0.05 + 2.53 119 11
    A210C P JPW4039 593 594 0.08 0.26
    JPW4042 648 645 0.11 0.79 0.103 0.054
    JPW4045 647 650 0.09 0.71
    Acrylodan 497 496 0.09 0.40
    Fluorescein 522 522 0.02 0.14
    NBD 543 542 0.02 0.30
    E119C p JPW4039 593 594 0.12 + 0.34
    JPW4045 649 644 0.08 + 1.73
    Acrylodan 498 497 0.11 + 0.65
    Fluorescein 523 523 0.05 0.09
    NBD 544 544 0.05 0.25
    F126C a JPW4039 596 592 0.11 + 0.85
    JPW4042 642 643 0.01 + 0.40
    JPW4045 654 643 0.33 + 1.27 903 94
    Acrylodan 495 482 0.07 + 2.70 82 13
    Fluorescein 522 519 0.22 + 1.73 1.71 mM 0.13 mM
    NBD 571 572 0.03 + 0.79
    F131C a JPW4039 593 597 0.15 0.37 0.151 0.080
    JPW4042 650 643 0.06 0.68
    JPW4045 649 642 0.02 0.48
    Acrylodan 487 492 0.08 0.84
    Fluorescein 522 522 0.05 0.13
    NBD 539 541 0.01 + 0.10
    F270C p JPW4039 596 594 0.01 0.11
    JPW4042 640 645 0.08 + 0.14
    JPW4045 644 647 0.07 0.69
    Acrylodan 490 492 0.07 0.60
    Fluorescein 523 523 0.04 0.21
    NBD 572 571 0.06 + 0.31
    G211C p JPW4039 594 592 0.01 + 0.12
    JPW4042 628 631 0.09 + 0.12
    JPW4045 631 634 0.06 + 0.36
    Acrylodan 493 492 0.02 0.29
    Fluorescein 522 521 0.03 0.18
    NBD 538 538 0.07 + 0.32
    K268C p Acrylodan 496 497 0.03 0.72
    Fluorescein 522 522 0.06 0.18
    Q123C p JPW4039 592 588 0.05 + 0.75
    JPW4045 640 641 0.00 0.88
    Acrylodan 498 495 0.10 0.40
    Fluorescein 524 522 0.13 2.23 0.75 0.09
    NBD 544 542 0.01 + 0.53
    T129C a JPW4039 587 584 0.09 + 0.73 0.093 0.015
    JPW4042 649 650 0.06 0.68
    JPW4045 644 648 0.05 0.73
    Acrylodan 484 482 0.04 + 0.52
    Fluorescein 523 523 0.02 0.17
    NBD 537 538 0.09 + 0.15 0.019 0.011
    Fe(III) BP E203C p JPW4039 Fe(III) citrate 599 592 0.09 0.37
    Acrylodan 518 518 0.41 0.95 138 21
    Fluorescein 523 522 0.33 0.15 41.9 3.5
    NBD 550 548 0.31 0.21 221 31
    K202C p JPW4039 602 602 0.24 0.36 193 29
    Acrylodan 505 503 0.37 1.17 195 25
    Fluorescein 520 521 0.30 0.09 195 16
    NBD 542 543 0.23 0.14 260 36
    K85C a JPW4039 593 591 0.05 0.10
    JPW4042 638 641 0.03 0.28
    Acrylodan 503 501 0.05 0.41
    Fluorescein 519 520 0.01 0.03
    NBD 545 543 0.08 0.12
    V287C a JPW4039 596 595 0.13 0.59
    JPW4042 596 591 0.06 0.24
    Acrylodan 504 506 0.21 0.34 221 35
    Fluorescein 521 520 0.21 0.05 92.5 7.5
    NBD 551 552 0.05 0.11 0.66 0.27
    glucose BP D257C a Acrylodan glucose 505 509 0.18 1.97 0.30 0.02
    Fluorescein 523 522 0.07 + 0.41
    NBD 545 547 0.72 0.68 1.39 0.01
    Pyrene 401 402 0.06 + 0.98
    E149C p Acrylodan 525 519 0.60 + 2.26 0.90 0.03
    Fluorescein 527 518 0.32 + 3.63 253 2
    NBD 549 539
    Figure US20080166747A1-20080710-P00001
    +
    Figure US20080166747A1-20080710-P00002
    2.94 0.12
    Pyrene 385 388 0.81 + 2.60 20.2 0.3
    E93C p Acrylodan 461 462 0.44 2.81 8.74 0.08
    Fluorescein 523 521 0.10 + 0.56 0.77 0.03
    NBD 557 546 0.53 + 3.27 12.3 0.2
    Pyrene 384 385 0.11 + 0.82
    H152C e Acrylodan 527 524 0.51 + 2.97 48.1 0.5
    Fluorescein 525 519 0.40 + 2.68 33.7 0.5
    NBD 546 549 1.29 + 1.20 134 1
    Pyrene 408 389
    Figure US20080166747A1-20080710-P00003
    +
    Figure US20080166747A1-20080710-P00004
    79.3 0.4
    L255C a Acrylodan 506 509 0.57 1.98 0.494 0.004
    Fluorescein 525 523 0.23 + 1.49 0.159 0.009
    NBD 541 548 0.19 + 1.71 0.263 0.021
    Pyrene 387 385 0.90 + 0.62 0.133 0.022
    N15C e Acrylodan 522 524 0.18 0.68 0.21 0.01
    Fluorescein 521 522 0.02 + 0.07
    NBD 544 547 0.04 0.82 0.135 0.007
    Pyrene 400 408 0.51 + 2.62
    V296C a Acrylodan 501 503 0.00 0.63
    Fluorescein 522 522 0.08 0.22 0.216 0.006
    NBD 541 543 0.40 1.06 0.169 0.011
    Pyrene 388 392 0.14 + 3.40
    W183C e Acrylodan 483 504 0.73 5.57 5.98 mM 0.03 mM
    Fluorescein 525 521 0.10 + 1.16 17.6 mM 2.4 mM
    NBD 547 546 0.13 0.14 318 mM 15 mM
    Pyrene 391 390 0.06 0.95
    Y10C e Acrylodan 498 497 0.15 1.16 116 3
    Fluorescein 521 521 0.43 + 1.22 3.31 mM 0.06 mM
    NBD 540 545 0.03 + 1.28
    Pyrene 388 391 0.19 2.87
    histidine BP E167C p Acrylodan histidine 504 506 0.17 + 0.72 0.060 0.003
    Fluorescein 517 518 0.08 0.40
    NBD 539 541 0.05 + 0.42
    Pyrene 384 384 0.21 + 1.13
    K229C p Acrylodan 526 527 0.02 0.41
    Fluorescein 517 516 0.03 0.05
    NBD 532 536 0.12 + 0.31
    Pyrene 384 384 0.16 + 0.73
    V163C p JPW4042 659 654 0.82 2.44 0.25 0.02
    Acrylodan 493 500 0.03 + 2.05 0.40 0.01
    Fluorescein 520 521 0.12 0.10
    NBD 542 543 0.17 + 1.32 2.37 0.15
    Pyrene 384 384 0.08 + 0.78
    Y230C p Acrylodan 523 522 0.02 0.18
    Fluorescein 517 517 0.05 0.07
    NBD 535 534 0.09 + 0.20
    Pyrene 384 384 0.22 + 0.75
    F231C p Acrylodan 524 525 0.01 0.56
    Fluorescein 516 516 0.03 + 0.06
    NBD 545 542 0.07 + 0.19
    Y88C a Acrylodan 491 493 0.03 0.30
    Fluorescein 518 518 0.04 0.06
    NBD 532 532 0.01 0.18
    Pyrene 384 384 0.15 + 0.44
    maltose BP D95C a JPW4039 maltose 591 593 0.08 0.70
    JPW4042 663 661 0.01 0.15
    JPW4045 650 645 0.08 + 1.36 0.30 0.01
    Acrylodan 522 501 0.04 3.31
    F92C a JPW4039 577 583 0.43 1.74
    JPW4042 646 646 0.04 0.11
    Acrylodan 495 484 0.16 + 2.09
    Fluorescein 519 518 0.02 + 0.03
    NBD 531 533 0.09 + 0.27
    I329C a JPW4039 595 594 0.05 0.43
    JPW4042 660 660 0.05 + 0.60
    JPW4045 652 649 0.04 + 0.55
    Acrylodan 498 500 0.02 0.79
    Fluorescein 517 518 0.04 + 0.08
    NBD 522 523 0.37 + 1.33 0.20 0.02
    S233C p JPW4039 577 583 0.42 1.73 145 6
    JPW4042 670 652 0.87 4.00 382 16
    JPW4045 678 657 0.42 + 3.92 409 22
    Acrylodan 518 519 0.01 0.80
    Fluorescein 519 519 0.17 + 0.10
    NBD 544 544 0.76 + 0.36 9.3 0.3
    phosphate A225C a JPW4039 phosphate 591 601 0.36 + 2.86 0.038 0.019
    BP JPW4042 615 628 0.30 1.32 0.39 0.08
    JPW4045 621 633 0.02 + 0.82
    Acrylodan 503 502 0.08 1.95
    Fluorescein 522 521 0.01 0.97 0.20 0.03
    NBD 544 554 0.81 1.21 0.27 0.03
    N223C a Fluorescein 519 519 0.06 + 0.01
    N226C a JPW4039 595 571 0.26 + 2.94 0.066 0.054
    JPW4042 673 651 0.29 + 2.05 0.172 0.148
    JPW4045 675 638 0.53 + 3.83 0.277 0.169
    S164C p JPW4039 599 550
    Figure US20080166747A1-20080710-P00005
    Figure US20080166747A1-20080710-P00006
    0.66 0.03
    JPW4042 630 615 0.33 1.78 1.16 0.22
    JPW4045 645 563 0.27 2.99 0.64 0.06
    Acrylodan 505 503 0.05 + 3.53 0.22 0.06
    Fluorescein 521 520 0.07 + 0.30 0.17 0.02
    NBD 539 540 0.02 + 0.42
    S39C p JPW4039 597 551 0.36 3.15 0.42 0.06
    JPW4042 623 622 0.01 + 0.15
    JPW4045 671 647 0.18 4.13 0.23 0.04
    Acrylodan 520 520 0.10 0.80
    Fluorescein 519 518 0.03 0.21
    NBD 558 559 0.18 + 0.57 0.14 0.04
    glutamine N160C p Acrylodan glutamine 529 527 0.11 + 0.43 0.098 0.023
    BP NBD 546 543 0.09 + 0.71
    Pyrene 387 387 0.04 0.15
    F221C p JPW4042 654 652 0.18 0.70
    Acrylodan 498 498 0.04 0.40
    Fluorescein 518 518 0.02 0.10
    NBD 544 545 0.06 + 0.36 0.0099 0.0034
    NBDE 538 537 0.04 + 0.24
    K219C p Acrylodan 494 500 0.25 1.34 0.38 0.03
    NBDE 510 510 0.02 + 0.21
    L162C p Acrylodan 496 501 0.46 2.17 0.17 0.02
    Fluorescein 523 519 0.17 + 1.80 0.38 0.06
    W220 p Acrylodan 519 518 0.03 + 0.58
    Fluorescein 518 518 0.01 0.03
    NBD 538 538 0.03 0.45
    NBDE 510 510 0.00 0.28
    Pyrene 386 390 0.40 + 2.86
    Y163C p Acrylodan 503 502 0.07 + 2.52 1.40 0.12
    Fluorescein 518 518 0.04 0.04
    NBD 530 528 0.05 0.30
    Pyrene 385 385 0.01 0.07
    Y86C a JPW4042 653 653 0.11 0.83 0.338 0.038
    Acrylodan 490 484 0.41 0.49 0.052 0.003
    NBD 541 538 0.27 0.25
    NBDE 541 551 0.12 + 1.81
    ribose BP A234C a JPW4039 ribose 598 600 0.37 1.29 1.84 0.40
    JPW4042 668 654 0.06 0.99
    JPW4045 636 578
    Figure US20080166747A1-20080710-P00007
    Figure US20080166747A1-20080710-P00008
    3.76 0.38
    Acrylodan 504 522 0.01 + 1.18
    Fluorescein 517 517 0.01 0.05
    NBD 546 548 0.28 + 1.63 0.735 0.057
    D165C p JPW4039 589 593 0.13 0.36
    JPW4042 650 652 0.06 0.27
    JPW4045 646 647 0.04 0.77
    Acrylodan 501 500 0.00 0.37
    Fluorescein 522 522 0.03 0.37
    E192C p JPW4039 598 598 0.44 0.34 2.57 0.67
    JPW4042 646 679
    Figure US20080166747A1-20080710-P00009
    Figure US20080166747A1-20080710-P00010
    5.03 0.77
    JPW4045 646 666
    Figure US20080166747A1-20080710-P00011
    Figure US20080166747A1-20080710-P00012
    15.0 0.4
    Acrylodan 516 516 0.04 0.27
    Fluorescein 526 523 0.12 + 1.31 11.4 0.8
    NBD 546 540 0.00 + 1.67 2.60 0.26
    L236C a JPW4039 589 588 0.08 0.29
    JPW4042 646 670 0.55 3.58 0.62 0.22
    JPW4045 643 658 0.25 1.70 1.53 0.41
    Acrylodan 518 518 0.09 0.71
    Fluorescein 520 520 0.02 0.29
    NBD 518 525 0.11 + 1.96 0.10 0.05
    L265C a JPW4039 600 596 0.01 0.11
    JPW4042 650 654 0.91 2.13 0.26 0.06
    JPW4045 669 663 0.02 0.12
    Acrylodan 500 501 0.20 0.70
    NBD 545 540 0.01 + 0.13
    T135C p JPW4039 606 606 0.02 0.03
    JPW4042 680 674 0.02 + 0.35
    JPW4045 647 664
    Figure US20080166747A1-20080710-P00013
    Figure US20080166747A1-20080710-P00014
    >1 mM
    Acrylodan 518 498 0.31 + 6.26 0.42 mM 0.01 mM
    Fluorescein 526 523 0.18 + 1.79 2.09 mM 0.27 mM
    NBD 542 544 0.08 + 0.22
    sulfate BP L65C p JPW4042 sulfate 629 635 0.40 1.82
    Acrylodan 492 482 0.39 + 2.95
    Fluorescein 520 516 0.39 + 1.31 1.09 0.05
    NBD 522 521 0.02 0.61
    Pyrene 386 385 0.13 + 1.20
    N70C p JPW4042 522 522 0.01 + 0.18
    Acrylodan 502 502 0.01 0.10
    Fluorescein 517 517 0.01 0.01
    NBD 524 524 0.01 0.14
    Pyrene 386 386 0.01 0.13
    Q294C p JPW4042 636 630 0.27 1.17 0.83 0.08
    Acrylodan 500 500 0.04 0.13
    Fluorescein 515 514 0.00 + 0.11
    NBD 530 530 0.00 + 0.02
    Pyrene 384 384 0.01 + 0.08
    R134C p JPW4039 522 518 0.08 2.02 7.5 0.2
    JPW4042 606 608 0.52 + 0.96 29.1 1.2
    Acrylodan 493 478 0.18 2.26 4.17 0.13
    Fluorescein 512 512 0.01 0.02 0.323 0.027
    NBD 531 532 0.58 0.37 22.4 0.5
    Pyrene 382 386 0.15 + 1.30
    W290C p JPW4042 612 624 0.43 0.89 0.336 0.012
    Acrylodan 496 496 0.04 0.03
    Fluorescein 516 515 0.04 + 0.09
    NBD 538 537 0.06 0.11
    Pyrene 384 384 0.16 + 0.37
    Y67C p Acrylodan 503 502 0.00 0.12
    Fluorescein 515 515 0.01 0.04
    NBD 536 534 0.13 + 0.20
    Pyrene 383 383 0.02 + 0.48
    aAll mutants of arabinose BP were in the C64A background. All mutants in Fe(III) BP were in the E57D background.
    ba: allosteric, e: endosteric, p: peristeric
    cNumbers in bold meet the threshold criteria of sensor utility elaborated in the text. Underlined numbers indicate excellent absolute intensity or ratiometric sensors. Numbers in bold italic are excellent sensors in both parameters.
    dinc/dec, increase (+) or decrease (−) in maximum fluorescence intensity upon ligand binding.
  • Assessment of fluorescent biosensor function. Fluorescence emission spectra of bPBP-fluorophore conjugates were recorded in the absence and presence of saturating ligand concentrations. Spectral changes were characterized by four parameters: wavelength shift (the difference between the wavelengths of emission maximum in the unbound and ligand-saturated states), direction of intensity change (increase or decrease in intensity at the wavelengths of maximum emission in the two states), standard intensity change (ΔIstd), and standard ratiometric change (ΔR). ΔIstd is defined as the normalized intensity change relative to the average intensity, determined at the wavelength mid-point between the two emission maxima:
  • Δ I std = 2 ( I 1 ( λ std ) - I 2 ( λ std ) ) I 1 ( λ std ) + I 2 ( λ std ) ( 1 )
  • where λstd=(λmax, unboundmax, saturated)/2 and I1, I2 are the fluorescence intensities at λstd of each spectrum respectively (FIG. 4A). ΔR is defined in terms of two emission bands, A1 ([λ1, λ2]) and A2 ([λ3, λ4]) (FIG. 4B):
  • Δ R = A 1 0 A 2 0 - A 1 A 2 ( 2 )
  • where 0A1, A2 are the areas in the absence of ligand, and A1, A2 the areas in the presence of saturating ligand. A computer program was used to enumerate ΔR for all possible pairs of wavelength bands in the two spectra, to identify the optimal sensing condition, defined as the maximum value of ΔR. Adjustable parameters of the algorithm, and their values used for ΔRmax quantities reported here, are: step size (2 nm), step width (10 nm), minimum integration area limit (fraction of total: 0.1), and maximum integration area limit (fraction of total: 1).
  • Analyte affinity measurements. 133 bPBP-fluorophore conjugates with ΔIstd>0.1 were used to determine ligand binding affinity by fluorimetric titration (Table 5). The emission wavelength monitored was that of maximum difference in intensity between the ligand-free and bound states. For each conjugate, fluorescence intensiometric observations were fit to a hyperbolic binding isotherm for a two-state model (Marvin et al., Proc. Natl. Acad. Sci. USA 94:4366-4371, 1997):
  • F = K d F F + [ S ] F B K d + [ S ] ( 3 )
  • where F is fluorescence at ligand concentration [S], Kd is the dissociation constant, and FF, FB are the fluorescence intensities of the ligand-free and ligand-saturated states, respectively. Examples of binding isotherms are shown in FIG. 5 for glucose BP and glutamate/aspartate BP. For ratiometric observations, eq. 3 has to be modified to account for differentially weighted contributions of the two emission bands (Lakowicz, Principles of Fluorescence Spectroscopy, 2nd Ed. Kluwer Academic Press, New York, p. 698, 1999):
  • R = K d app R F + [ S ] R B K d app + [ S ] ( 4 )
  • where R is ratio A1/A2, RB=A1/A2, RF=0A1/0A1, and appKd is an apparent dissociation constant:
  • K d app = A 2 0 A 2 K d ( 5 )
  • The success of the fluorescent biosensor design strategy was evaluated by determining the probability of encountering an effectively responding fluorescent conjugate, and assessing how the ligand-binding affinities are affected by the fluorophore conjugate.
  • Assessment of ligand-mediated changes in fluorescence. Summaries of wavelength shift, ΔIstd, and ΔRmax for all conjugates (n=320) are presented as histograms in FIG. 6A. The distribution of wavelength shifts was symmetrical about zero; that is, there was no overall tendency toward either blue- or red-shifts. Of the entire collection of conjugates, 130 show increases and 190 show decreases in fluorescence intensity upon binding. A portion of this skew is due to the finding that addition of Fe(III) citrate to all Fe(III) BP conjugates caused a decreased fluorescence emission. To examine whether this was due to quenching by Fe(III) in solution, Fe(III) citrate was added to conjugates of other bPBPs and the effect on emission intensity was monitored. It was found that Fe(III) citrate quenched fluorescence in all cases, but only at concentrations much higher than those that led to the effect in Fe(III) BP. The decrease in fluorescence intensity observed in all conjugates of Fe(III) BP is therefore due to a binding-specific process, and may involve relaxation of the excited state via a metal-mediated redox mechanism (Lakowicz, Principles of Fluorescence Spectroscopy, 2nd Ed. Kluwer Academic Press, New York, p. 698, 1999). The probability of encountering a conjugate that responds with a particular intensity declines with increasing magnitude of ΔIstd (FIG. 6B). The ratiometric response behaves similarly (FIG. 6C).
  • The two criteria of greatest utility for optical sensing are ΔIstd and ΔRmax. The collection of bPBP conjugates was categorized by class of steric site, fluorophore, and protein scaffold, then, for each category, quantified according to the fraction with ΔIstd>0.25 and with ΔRmax>1.25. The results (Tables 6 to 8) give an indication of the overall success rate for finding potentially useful fluorescent biosensor conjugates. For the collection of 320 conjugates, about 24% meet the criterion for ΔIstd and about 28% the criterion for ΔRmax.
  • TABLE 6
    Signaling parameters by binding protein
    fraction fraction
    binding protein ΔIstd > 0.25 ΔRmax > 1.25 n
    arabinose BP 0.50 0.40 20
    glucose BP 0.47 0.50 36
    ribose BP 0.32 0.41 34
    dipeptide BP 0.08 0.14 36
    glutamine BP 0.20 0.24 25
    histidine BP 0.04 0.13 24
    Glu/Asp BP 0.04 0.15 54
    phosphate BP 0.45 0.55 22
    sulfate BP 0.23 0.20 30
    maltose BP 0.29 0.38 21
    Fe(III) BP 0.28 0.00 18
    aggregate 0.24 0.28 320
  • TABLE 7
    Signaling parameters by steric site
    fraction fraction
    site ΔIstd > 0.25 ΔRmax > 1.25 n
    allosteric 0.28 0.32 110
    peristeric 0.20 0.15 198
    endosteric 0.50 0.50 12
    aggregate 0.24 0.28 320
  • TABLE 8
    Signaling parameters by fluorophore
    fraction fraction
    fluorophore ΔIstd > 0.25 ΔRmax > 1.25 n
    Acrylodan 0.21 0.38 66
    Fluorescein 0.13 0.16 62
    NBD 0.25 0.20 61
    NBDE 0.00 0.25 4
    Pyrene 0.22 0.30 23
    JPW4039 0.38 0.28 39
    JPW4042 0.32 0.30 37
    JPW4045 0.29 0.39 28
    aggregate 0.24 0.28 320
  • There appears to be a correlation between signaling success rate and the sequence-related family, or cluster (Tam & Saier, Microbiol. Rev. 57:320-346, 1993), to which a scaffold belongs. The scaffolds having the highest success rates for ΔIstd and ΔRmax are arabinose BP, glucose BP, ribose BP, and phosphate BP (Table 6). The former three belong to cluster 2, that includes binding proteins for hexoses and pentoses, while phosphate BP, along with sulfate BP, belongs to cluster 6, that includes binding proteins for inorganic polyanions. The scaffolds having the lowest success rate were dipeptide BP (cluster 5, peptide and nickel binding) and the cluster 3 (polar amino-acid binding) proteins glutamine BP, histidine BP, and Glu/Asp BP.
  • Among the three classes of attachment sites the endosteric and allosteric sites have a higher chance of meeting the threshold criteria than peristeric sites (Table 7). Success rates in terms of ΔIstd varied according to the environmental sensitivity of the fluorophore, being highest with the styryl and naphthyl dyes JPW4039, JPW4042, and JPW4045. Similarly, higher success rates for ΔRmax were associated with JPW4045 and acrylodan (Table 8).
  • Assessment of changes in ligand-binding affinities. The range of dissociation constants, Kd, extracted from the binding curves for each ligand is shown in Table 9. Since there is a thermodynamic linkage between ligand binding and the interaction of the attached fluorophore with the protein, the fluorophore is expected to change the intrinsic ligand dissociation constant. The change in affinity imparted by the fluorophore is expected to be dependent on its location in the protein. The various conjugates exhibit a wide range of affinities (Table 9). The change in affinity, defined as log(mutKd/wtKd), was examined as a function of attachment site classification (endosteric, allosteric, or peristeric) among the 108 conjugates for which dissociation constants were measured and for which the dissociation constant of the unconjugated protein is known (Table 2). The results reveal that the three classes of site have different effects on affinity (FIG. 7). Fluorophore attachment at endosteric sites tends to perturb affinity the greatest, and uniformly to higher values of Kd than the wild type. Allosteric and peristeric attachment results in Kd values that are either higher or lower than the wild type, with peristeric sites exhibiting the greatest variation in effects. Interestingly, of those conjugates with higher affinity than the wild type (lower Kd), a greater proportion derives from conjugation at allosteric sites. This corroborates detailed studies in maltose BP in which affinity was increased by manipulating the volume of residues in allosteric sites (Marvin & Hellinga, Nat. Struct. Biol. 8:795-798, 2001). The differences in effects can be rationalized in terms of the likelihood that a particular conjugate will sterically interfere either directly with ligand binding (endosteric sites, and some peristeric sites), or by influencing the intrinsic equilibrium between the open and closed states (allosteric sites, peristeric sites).
  • TABLE 9
    Range of ligand affinities in bPBP fluorescent conjugates
    bPBP ligand range of Kd(μM) n
    arabinose BP arabinose  0.46-775 19
    glucose BP glucose  0.13-318000 26
    ribose BP ribose  0.1-2090 14
    dipeptide BP Gly-Leu 0.006-93 21
    glutamine BP glutamine  0.01-1.4 8
    histidine BP histidine  0.06-2.37 4
    Glu/Asp BP glutamate 0.019-1700 9
    phosphate BP phosphate 0.038-1.2 12
    sulfate BP sulfate  0.32-29 8
    maltose BP maltose  0.2-409 6
    Fe(III) BP Fe(III) citrate  0.66-260 10
  • The effect on dissociation constants is determined not only by the attachment site, but also by the nature of the attached fluorophore, as illustrated for arabinose BP. Dissociation constants for arabinose of the five cysteine-substitution mutants (all with the C64A mutation), measured by tryptophan fluorescence, are 5.0 μM (F23C), 3.2 μM (L253C), 3.4 μM (D257C), 7.6 μM (L298C), and 1.6 μM (K301C). Thus the cysteine substitutions slightly perturbed affinity for arabinose (Kd of C64A mutant ˜2.2 μM). The largest dependence on the attached fluorophore was found for the L253C mutant, for which Kd values ranged from 0.7 μM (acrylodan) to 775 μM (NBD). Similarly, the K394C mutant of dipeptide BP has affinities for Gly-Leu dipeptide ranging from 6 nM (NBD) to 93 μM (fluorescein). Most mutants did not exhibit such a wide range of fluorophore-dependent ligand affinity. For example, five different fluorophores conjugated to ribose BP E192C have affinities for ribose ranging from 2.6 μM (NBD and JPW4039) to 15 μM (JPW4045).
  • Construction of a novel biosensor using sequence information. To demonstrate that designs are not limited to those bPBPs with known structure, cysteine mutations were introduced into a paralog predicted to code for a glutamate/aspartate BP, using histidine and glutamine BPs as the structures to guide locations for likely peristeric and allosteric sites. All the ten sites that were tried yielded conjugates that exhibited glutamate and aspartate-dependent changes in fluorescence. Several sites yielded good or excellent intensiometric or ratiometric sensors. Table 10 shows that the response is specific for both aspartate and glutamate, with 50- to 500-fold weaker affinity for glutamine and asparagine. Other amino acids and sugars did not elicit ligand-mediated changes in fluorescence.
  • TABLE 10
    Binding specificity and affinity in mutants of glutamate/aspartate BP
    Kd(μM)
    mutant fluorophore Glu Asp Gln Asn
    Q123C Fluorescein 0.75 1.8 49 96
    F126C Acylodan 82 115
    F126C Fluorescein 1707 2000
    F126C JPW4045 903 1497
    T129C NBD 0.019 0.061 12.1 5.4
    T129C JPW4039 0.093 0.035 23
    F131C JPW4039 0.15
    A207C NBD 119 454
    A210C JPW4042 0.10
  • Bioinformatics makes possible the discovery of new biochemical applications without direct experimentation. In the case of biosensors, individual bacterial genomes may encode scores of bPBPs that bind specific molecules to initiate transport or signal transduction (Blattner et al., Science 277:1453-1474, 1997; Quentin et al., J. Mol. Biol. 287:467-484, 1999). Few of these have been characterized, leaving a vast number untapped as scaffolds for potential biosensors. The feasibility of applying genomic information, combined with structural information from homologous proteins, to construct a biosensor of novel specificity has been demonstrated.
  • Previously, a glutamate/aspartate BP had been purified from E. coli (Barash & Halpern, Biochim. Biophys. Acta 386:168-180, 1975; Willis & Furlong, J. Biol. Chem. 250:2574-2580, 1975) and characterized. Several pieces of evidence suggest that YBEJ corresponds to this protein. First, glutamate/aspartate BP was isolated from periplasmic extracts, consistent with ybeJ encoding a protein with a putative periplasmic localization signal sequence. Second, the previously determined molecular mass of glutamate/aspartate BP of 32 kDa (Barash & Halpern, Biochim. Biophys. Acta 386:168-180, 1975) or 31 kDa (Willis & Furlong, J. Biol. Chem. 250:2574-2580, 1975) match the mass of 32.5 kDa predicted for the processed ybeJ product, and the mass of 30 kDa found by gel electrophoresis in the present study. Third, the amino acid compositions determined previously (Barash & Halpern, Biochim. Biophys. Acta 386:168-180, 1975; Willis & Furlong, J. Biol. Chem. 250:2574-2580, 1975) are similar to that predicted from the gene sequence, with some deviations due likely to inherent inaccuracy in analysis of protein acid hydrolyzates. Finally, the reported Kd values for glutamate (0.8 μM), aspartate (1.2 μM), as well as the relatively lower affinity for glutamine and asparagine (Willis & Furlong, J. Biol. Chem. 250:2574-2580, 1975) are similar to those determined here, and comparable to the Q123C-fluorescein conjugate (Table 10). Hence, ybeJ likely encodes the glutamate/aspartate BP previously characterized.
  • Effective sensor designs. The utility of a conjugate is determined by the absolute change in signal intensity, the ratiometric change, and the operating concentration range over which the sensor can respond accurately. Of the two observable parameters, ratiometric change is preferable to absolute intensities, since it is independent of probe concentration.
  • Although usable conjugates can be defined as having ΔIstd>0.25 and ΔRmax>1.25, “excellent” sensors can be defined as having ΔIstd>0.9 and ΔRmax>2.5. The magnitudes of the changes in the excellent sensors are likely to be sufficiently large to permit robust measurements in “real-world” applications in complex fluids such as blood. Based on these criteria there are only thirteen excellent absolute intensity-based sensors (4% of total), but 36 excellent ratiometric sensors (11% of total); there are seven conjugates that are both excellent absolute intensity and excellent ratiometric sensors (Table 5). With the exception of dipeptide BP, Fe(III) BP, and histidine BP, all the proteins have at least one excellent ratiometric and intensity-based conjugate. Glucose BP has the largest number of excellent conjugates. These conjugates all involve fluorophores known to be particularly environmentally sensitive (acrylodan, NBD, pyrene, and the styryl dyes). The incidence of excellent sensors is evenly distributed between allosteric and peristeric sites. All endosteric sites give rise to excellent sensors.
  • The dissociation constant of a conjugate determines the operating concentration range over which the sensor can respond accurately. The operating range guaranteed to give less than a 5% error spans concentrations that fall within five-fold of the Kd value (Marvin et al., Proc. Natl. Acad. Sci. USA 94:4366-4371, 1997). If the range required for accurate determination is wider than that span, then a composite biosensor can be constructed using receptors of varying affinities, as has been demonstrated for maltose BP (Marvin et al., Proc. Natl. Acad. Sci. USA 94:4366-4371, 1997). There are three factors affecting the dissociation constant: the nature of the conjugate, the choice of emission bands for a ratiometric sensor (eq. 2), and additional mutations. For particular applications, these three factors can be manipulated to construct an appropriate sensor.
  • Glucose sensor. Among the analytes applicable to clinical medicine, glucose is one of the most important, particularly with regard to diagnosing and treating diabetes. The normal range of glucose concentration in adult human serum is 4 to 6 mM (Burtis & Ashwood, Teitz Textbook of Clinical Chemistry, 2nd Ed. W.B. Saunders Co., Philadelphia, Pa., 1994). The acrylodan conjugate of the endosteric site W183C in glucose BP has an excellent ratiometric response (ΔRmax=5.57) and a dissociation constant of 5.98 mM, and is therefore a good candidate for detecting glucose fluctuations in the physiological range by ratiometry (FIG. 8A). Furthermore, by adjusting the ratiometric parameters, the observation window is easily extended from 5.0 to 17.4 mM, allowing all clinically relevant ranges to be observed with one sensor (FIG. 8A).
  • Other sensors for clinical chemistry. Amino acids are also commonly assayed in clinical tests as indicators of disease states. Histidine is an indicator of histidase deficiency (Taylor et al., Molec. Biol. Med. 8: 101-116, 1991). The best signaling histidine BP conjugate, V163C-JPW4042, has a Kd of 0.25 μM, below the normal range in serum of about 48 to 125 μM. However, with sample dilution this conjugate could function effectively. Alternatively the Kd can be adjusted by mutagenesis as was done for maltose BP (Marvin & Hellinga, Nat. Struct. Biol. 8:795-798, 2001) and Fe(III) BP with the E57D mutation. The neuroexcitatory amino acid glutamate has normal serum concentrations of 20 to 220 μM (Burtis & Ashwood, Teitz Textbook of Clinical Chemistry, 2nd Ed. W.B. Saunders Co., Philadelphia, Pa., 1994). The best-suited biosensor is glutamate/aspartate BP F126C-acrylodan, which has a Kd˜80 μM and ΔRmax=2.70. Glutamine is often measured in cerebrospinal fluid (Smith & Forman, Clin. Lab. Sci. 7:32-38, 1994) in which its normal range is 120 to 360 μM, considerably higher than the Kd (˜1.4 μM) of the best-signaling glutamine BP conjugate, Y163C-acrylodan. This biosensor can be used for such a purpose by mutagenesis to adjust the Kd, or by sample dilution.
  • Phosphate concentrations in serum and urine are clinically relevant (Burkhardt et al., Am. J. Clin. Pathol. 72:326-329, 1979). Several phosphate BP conjugates signal well, the best being S39C-JPW4045, and their Kd values are all less than 2 μM. Inorganic phosphate in serum is typically 1 to 3 mM (Burtis & Ashwood, Teitz Textbook of Clinical Chemistry, 2nd Ed. W.B. Saunders Co., Philadelphia, Pa. 1994), requiring adjustment of the Kd or sample dilution for accurate measurements with these sensors.
  • Maltose concentration is relevant to a deficiency in acid maltase, with the normal plasma concentration about 2 μM (Rozaklis et al., Clin. Chem. 48:131-139, 2002). The best maltose sensors in the present work are maltose BP conjugates S233C-JPW4042 (ΔRmax=4.0) and S233C-JPW4045 (ΔRmax=3.9), both with similar affinities (Kd˜400 μM). Fluorescent conjugates of maltose BP mutants having affinities in the 2 μM range have been described by Marvin et al. (Proc. Natl. Acad. Sci. USA 94:4366-4371, 1997).
  • Industrial and environmental applications. bPBP conjugates can function as sensors for industrial and environmental analytes. Arabinose is relevant to improving the efficiency of ethanol production from corn (Deanda et al., Appl. Environ. Microbiol. 62:4465-4470, 1996). Of the arabinose BP conjugates, the best signalers are K301C-NBD (Kd˜31 μM, ΔRmax=3.2) and L253C-fluorescein, (Kd˜48 μM, ΔRmax=2.7). Ribose concentration, assayed in foods and beverages (AOAC, Official Methods of Analysis of AOAC International, 16th Ed. AOAC International, Arlington, Va., 1995), can be measured by ribose BP conjugates T135C-acrylodan (Kd˜0.4 mM, ΔRmax=6.3) and A234C-JPW4045 (Kd˜3.8 μM, ΔRmax=4.1). Ratiometric sensing of ribose using a single ribose BP derivative is illustrated by the T135C-acrylodan conjugate (FIG. 8B). By varying emission wavelength bands in the fluorescence ratio (eqs. 4, 5) the appKd for ribose can be adjusted over a range from 41 to 146 μM (FIG. 8B). Sulfate concentrations in drinking water are of concern (U.S. EPA, Health Effects From Exposure to High Levels of Sulfate in Drinking Water, pp. 1-25, Office of Drinking Water and Ground Water, 1999), and can be analyzed by sulfate BP conjugate R134C-acrylodan (Kd˜4 μM, ΔRmax=2.3). High concentrations of phosphate are environmentally deleterious, and could be monitored using phosphate BP conjugates, as noted above for clinical applications. Iron concentration limits primary productivity in certain regions of the oceans (Martin, Iron as a Limiting Factor in Primary Productivity and Biogeochemical Cycles in the Sea. Falkowski & Woodhead, eds., pp. 123-137, Plenum Press, New York). Available ferric ion can be determined using a biosensor derived from Fe(III) BP, such as conjugate E203C-acrylodan (Kd˜138 μM, ΔIstd 0.4).
  • All documents cited above are hereby incorporated in their entirety by reference. Also incorporated by reference for their disclosure of electronic devices containing bioelectronic sensors are U.S. application Ser. No. 10/229,286 (published as US 2003/0129622) and Int'l Appln. No. PCT/US02/27279 (WO 03/021247).

Claims (22)

1. A biosensor for ligand, which comprises a glucose binding protein (GBP) and at least one reporter group attached at position 183 of said GBP, wherein binding of glucose in a glucose-binding pocket of said biosensor causes a change in signaling by said reporter group.
2. The biosensor according to claim 1, wherein said GBP is a W183C mutant.
3-6. (canceled)
7. The biosensor according to claim 1, wherein said reporter group is covalently attached.
8. The biosensor according to claim 1, wherein said reporter group is noncovalently attached.
9. The biosensor according to claim 1, wherein said reporter group is a redox cofactor.
10. The biosensor according to claim 1, wherein said reporter group is a fluorophore.
11. The biosensor according to claim 1, wherein said biosensor's standard intensity change (ΔIstd) upon binding of ligand is greater than 0.25.
12. The biosensor according to claim 11, wherein said ΔIstd is greater than 0.9.
13. The biosensor according to claim 1, wherein said biosensor's maximum value of standard ratiometric change (ΔRmax) upon binding of ligand is greater than 1.25.
14. The biosensor according to claim 13, wherein said ΔRmax is greater than 2.5.
15. (canceled)
16. A method of detecting presence or absence of glucose in a sample, which comprises: contacting a biosensor according to claim 1 with said sample under conditions such that said biosensor is able to bind to glucose present in said sample; comparing the signal transduced by said reporter group when said biosensor is contacted with said sample with the signal(s) transduced by said reporter group when said biosensor is contacted with at least one control sample containing a known quantity of glucose; and determining the presence or absence of glucose in said sample from said comparison.
17. A method of quantitating amount or concentration of glucose in a sample, which comprises: contacting a biosensor according to claim 1 with said sample under conditions such that said biosensor is able to bind to glucose present in said sample; comparing the signal transduced by said reporter group when said biosensor is contacted with said sample against signals transduced by a series of control samples containing known quantities of glucose; and calculating the quantity of glucose in said sample from said comparison.
18. A method of assaying for glucose in a sample, which comprises:
(a) contacting a biosensor according to claim 1;
(b) measuring a ratiometric change (ΔR) for the signal transduced by said reporter group; and
(c) at least detecting or quantitating ligand present in said sample.
19. The method of claim 18, wherein said sample is comprised of a physiological fluid.
20. The method of claim 19, wherein said physiological fluid is selected from the group consisting of blood, interstitial fluid, lavage, perspiration, plasma, saliva, serum, and urine.
21-22. (canceled)
23. The method of claim 16, wherein said sample is comprised of a physiological fluid.
24. The method of claim 23, wherein said physiological fluid is selected from the group consisting of blood, interstitial fluid, lavage, perspiration, plasma, saliva, serum, and urine.
25. The method of claim 17, wherein said sample is comprised of a physiological fluid.
26. The method of claim 25, wherein said physiological fluid is selected from the group consisting of blood, interstitial fluid, lavage, perspiration, plasma, saliva, serum, and urine.
US11/785,591 2002-10-16 2007-04-18 Biosensor Abandoned US20080166747A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/785,591 US20080166747A1 (en) 2002-10-16 2007-04-18 Biosensor

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US41835902P 2002-10-16 2002-10-16
US10/686,529 US9625458B2 (en) 2002-10-16 2003-10-16 Biosensor
US11/785,591 US20080166747A1 (en) 2002-10-16 2007-04-18 Biosensor

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/686,529 Continuation US9625458B2 (en) 2002-10-16 2003-10-16 Biosensor

Publications (1)

Publication Number Publication Date
US20080166747A1 true US20080166747A1 (en) 2008-07-10

Family

ID=32107920

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/686,529 Active 2025-06-24 US9625458B2 (en) 2002-10-16 2003-10-16 Biosensor
US11/785,591 Abandoned US20080166747A1 (en) 2002-10-16 2007-04-18 Biosensor
US15/487,665 Expired - Lifetime US10712341B2 (en) 2002-10-16 2017-04-14 Biosensor

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/686,529 Active 2025-06-24 US9625458B2 (en) 2002-10-16 2003-10-16 Biosensor

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/487,665 Expired - Lifetime US10712341B2 (en) 2002-10-16 2017-04-14 Biosensor

Country Status (13)

Country Link
US (3) US9625458B2 (en)
EP (1) EP1552303B1 (en)
JP (1) JP4571499B2 (en)
KR (1) KR101107630B1 (en)
CN (1) CN100492010C (en)
AT (1) ATE423318T1 (en)
AU (1) AU2003277377B2 (en)
CA (1) CA2502272C (en)
DE (1) DE60326256D1 (en)
DK (1) DK1552303T3 (en)
ES (1) ES2321931T3 (en)
HK (1) HK1080937A1 (en)
WO (1) WO2004036176A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8115635B2 (en) 2005-02-08 2012-02-14 Abbott Diabetes Care Inc. RF tag on test strips, test strip vials and boxes
WO2014120647A1 (en) * 2013-01-30 2014-08-07 Siemens Healthcare Diagnostics Inc. Signal ratio in assay calibrators
WO2017087915A1 (en) * 2015-11-20 2017-05-26 Duke University Lactate biosensors and uses thereof
US9829491B2 (en) 2009-10-09 2017-11-28 The Research Foundation For The State University Of New York pH-insensitive glucose indicator protein
US10712341B2 (en) 2002-10-16 2020-07-14 Duke University Biosensor
US11906524B2 (en) 2015-11-20 2024-02-20 Duke University Urea biosensors and uses thereof

Families Citing this family (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100627206B1 (en) * 1998-07-30 2006-09-25 유니챰 가부시키가이샤 Thin-article container
EP1620548A2 (en) * 2003-05-07 2006-02-01 Duke University Protein design for receptor-ligand recognition and binding
US8506775B2 (en) * 2003-06-20 2013-08-13 Roche Diagnostics Operations, Inc. Devices and methods relating to electrochemical biosensors
US8679853B2 (en) 2003-06-20 2014-03-25 Roche Diagnostics Operations, Inc. Biosensor with laser-sealed capillary space and method of making
US8148164B2 (en) 2003-06-20 2012-04-03 Roche Diagnostics Operations, Inc. System and method for determining the concentration of an analyte in a sample fluid
JP2007524816A (en) 2003-06-20 2007-08-30 エフ ホフマン−ラ ロッシュ アクチェン ゲゼルシャフト Method for producing thin uniform reagent strip and its reagent
US20050112685A1 (en) * 2003-11-26 2005-05-26 Amiss Terry J. Compositions and methods for measuring analyte concentrations
US20050181141A1 (en) * 2004-02-18 2005-08-18 Aiden Flanagan Laser-induced explosive vaporization coating method, associated system, and device made by the method
US20050181116A1 (en) * 2004-02-18 2005-08-18 Rob Worsham Method for coating a medical device using a matrix assisted pulsed-laser evaporation technique and associated system and medical device
US20050188921A1 (en) * 2004-02-27 2005-09-01 Anthony Malone Matrix assisted pulsed-laser evaporation technique for coating a medical device and associated system and medical device
US7563891B2 (en) * 2004-05-21 2009-07-21 Becton, Dickinson & Company Long wavelength thiol-reactive fluorophores
AU2005295655A1 (en) 2004-10-14 2006-04-27 Carnegie Institution Of Washington Neurotransmitter sensors and methods of using the same
CA2584108A1 (en) * 2004-10-14 2006-04-27 Carnegie Institution Of Washington Development of sensitive fret sensors and methods of using the same
WO2006096213A1 (en) * 2005-03-03 2006-09-14 Carnegie Institution Of Washington Polyamine sensors and methods of using the same
WO2006096214A1 (en) * 2005-03-04 2006-09-14 Carnegie Institution Of Washington Environmentally stable sensors and methods of using the same
US20070154947A1 (en) * 2005-08-31 2007-07-05 The Trustees Of Princeton University Chemical biodiscriminator
ATE481412T1 (en) * 2005-10-14 2010-10-15 Carnegie Inst Of Washington PHOSPHATE BIOSENSORS AND METHOD FOR USE THEREOF
CA2625165A1 (en) * 2005-10-14 2007-04-26 Carnegie Institution Of Washington Sucrose biosensors and methods of using the same
JP2009520462A (en) * 2005-11-16 2009-05-28 カーネギー インスチチューション オブ ワシントン Multimeric biosensor and method using the same
US8241567B2 (en) 2007-04-20 2012-08-14 Becton, Dickinson And Company Hydrogel compositions
EP2150586B1 (en) * 2007-05-22 2018-03-14 Becton, Dickinson and Company Dyes having ratiometric fluorescence response for detecting metabolites
US8129532B2 (en) * 2007-08-03 2012-03-06 The University Of Connecticut Amino(oligo)thiophene dyes, preparation thereof, and optical methods of use
US8465981B2 (en) * 2007-08-06 2013-06-18 University Of Kentucky Research Foundation Polypeptides, systems, and methods useful for detecting glucose
US10684282B2 (en) 2011-10-07 2020-06-16 Howard Hughes Medical Institute Genetically encoded biosensors
US9719992B2 (en) 2011-10-07 2017-08-01 Howard Hughes Medical Institute Genetically encoded biosensors
US10466247B2 (en) 2012-11-20 2019-11-05 Becton, Dickinson And Company System and method for diagnosing sensor performance using analyte-independent ratiometric signals
KR101455244B1 (en) 2013-02-04 2014-10-31 경희대학교 산학협력단 A quantitative evaluation system and a method for measuring cytotoxicity using AFM and probe-type sensor
CN105683210B (en) * 2013-08-23 2020-07-14 贝林格尔·英格海姆Rcv两合公司 Microparticles for cell disruption and/or recovery of biomolecules
US10379125B2 (en) 2013-12-27 2019-08-13 Becton, Dickinson And Company System and method for dynamically calibrating and measuring analyte concentration in diabetes management monitors
WO2017066441A1 (en) 2015-10-13 2017-04-20 The Research Foundation For The State University Of New York Cleavable fusion tag for protein overexpression and purification
CA3005890A1 (en) 2015-11-20 2017-05-26 Duke University Thermostable glucose biosensors and uses thereof
EP3377643A4 (en) 2015-11-20 2019-10-02 Duke University Glucose biosensors and uses thereof
EP3377899A4 (en) 2015-11-20 2019-10-02 Duke University Bicarbonate biosensors, calcium biosensors, and uses thereof
GB201714478D0 (en) * 2017-09-08 2017-10-25 Univ Bristol Sensor
EP4007767A4 (en) * 2019-08-01 2023-08-02 The Trustees of Indiana University Glyphosate biosensor
CN113980992B (en) * 2021-12-29 2022-03-25 中国科学院天津工业生物技术研究所 L-cysteine biosensor and application thereof

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6096497A (en) * 1998-06-15 2000-08-01 Biosensor Systems Design, Inc. Electrostatic enzyme biosensor
US6130037A (en) * 1996-04-25 2000-10-10 Pence And Mcgill University Biosensor device and method
US6231733B1 (en) * 1993-04-17 2001-05-15 Kurt Nilsson Immobilized carbohydrate biosensor
US6248229B1 (en) * 1997-06-12 2001-06-19 Clinical Micro Sensors, Inc. Detection of analytes using reorganization energy
US6277627B1 (en) * 1997-12-31 2001-08-21 Duke University Biosensor
US20030129622A1 (en) * 2001-08-28 2003-07-10 Hellinga Homme W. Biosensor
US20030134346A1 (en) * 2002-01-04 2003-07-17 Amiss Terry J. Binding protein as biosensors
US6663862B1 (en) * 1999-06-04 2003-12-16 Duke University Reagents for detection and purification of antibody fragments
US20040118681A1 (en) * 2002-10-16 2004-06-24 Hellinga Homme W. Biosensor
US20040229290A1 (en) * 2003-05-07 2004-11-18 Duke University Protein design for receptor-ligand recognition and binding
US20080305489A1 (en) * 2007-06-06 2008-12-11 Becton, Dickinson And Company Near-infrared dyes as surface enhanced raman scattering reporters
US20080311675A1 (en) * 2007-05-22 2008-12-18 Becton, Dickinson And Company Dyes having ratiometric fluorescence response for detecting metabolites
US20090104714A1 (en) * 2007-10-18 2009-04-23 Becton, Dickinson And Company Visual glucose sensor and methods of use thereof

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0844246A4 (en) 1996-03-15 1999-06-02 Ss Pharmaceutical Co Reagents for labeling sh groups, process for the preparation of them, and method for labeling with them
SE9602545L (en) 1996-06-25 1997-12-26 Michael Mecklenburg Method of discriminating complex biological samples
AU7715898A (en) 1997-05-27 1998-12-30 Duke University Synthetic metalloproteins and method of preparation thereof
AUPO714897A0 (en) * 1997-06-03 1997-06-26 Australian Membrane And Biotechnology Research Institute Model membrane systems
CA2327400A1 (en) 1998-04-06 1999-10-14 Bunsen Rush Laboratories, Inc. Directed evolution biosensors
DE69941085D1 (en) * 1998-07-17 2009-08-20 Univ Maryland MANIPULATED PROTEINS FOR ANALYSIS PROOF
GB9923146D0 (en) * 1999-09-30 1999-12-01 Imperial College Detector array

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6231733B1 (en) * 1993-04-17 2001-05-15 Kurt Nilsson Immobilized carbohydrate biosensor
US6130037A (en) * 1996-04-25 2000-10-10 Pence And Mcgill University Biosensor device and method
US6248229B1 (en) * 1997-06-12 2001-06-19 Clinical Micro Sensors, Inc. Detection of analytes using reorganization energy
US6277627B1 (en) * 1997-12-31 2001-08-21 Duke University Biosensor
US20020004217A1 (en) * 1997-12-31 2002-01-10 Duke University Biosensor
US6521446B2 (en) * 1997-12-31 2003-02-18 Duke University Biosensor
US6096497A (en) * 1998-06-15 2000-08-01 Biosensor Systems Design, Inc. Electrostatic enzyme biosensor
US6663862B1 (en) * 1999-06-04 2003-12-16 Duke University Reagents for detection and purification of antibody fragments
US6977180B2 (en) * 2001-08-28 2005-12-20 Duke University Biosensor
US20030129622A1 (en) * 2001-08-28 2003-07-10 Hellinga Homme W. Biosensor
US20030134346A1 (en) * 2002-01-04 2003-07-17 Amiss Terry J. Binding protein as biosensors
US6855556B2 (en) * 2002-01-04 2005-02-15 Becton, Dickinson And Company Binding protein as biosensors
US20040118681A1 (en) * 2002-10-16 2004-06-24 Hellinga Homme W. Biosensor
US20040229290A1 (en) * 2003-05-07 2004-11-18 Duke University Protein design for receptor-ligand recognition and binding
US20080311675A1 (en) * 2007-05-22 2008-12-18 Becton, Dickinson And Company Dyes having ratiometric fluorescence response for detecting metabolites
US20080305489A1 (en) * 2007-06-06 2008-12-11 Becton, Dickinson And Company Near-infrared dyes as surface enhanced raman scattering reporters
US20090104714A1 (en) * 2007-10-18 2009-04-23 Becton, Dickinson And Company Visual glucose sensor and methods of use thereof

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10712341B2 (en) 2002-10-16 2020-07-14 Duke University Biosensor
US8115635B2 (en) 2005-02-08 2012-02-14 Abbott Diabetes Care Inc. RF tag on test strips, test strip vials and boxes
US8223021B2 (en) 2005-02-08 2012-07-17 Abbott Diabetes Care Inc. RF tag on test strips, test strip vials and boxes
US8358210B2 (en) 2005-02-08 2013-01-22 Abbott Diabetes Care Inc. RF tag on test strips, test strip vials and boxes
US8390455B2 (en) 2005-02-08 2013-03-05 Abbott Diabetes Care Inc. RF tag on test strips, test strip vials and boxes
US8542122B2 (en) 2005-02-08 2013-09-24 Abbott Diabetes Care Inc. Glucose measurement device and methods using RFID
US9829491B2 (en) 2009-10-09 2017-11-28 The Research Foundation For The State University Of New York pH-insensitive glucose indicator protein
WO2014120647A1 (en) * 2013-01-30 2014-08-07 Siemens Healthcare Diagnostics Inc. Signal ratio in assay calibrators
WO2017087915A1 (en) * 2015-11-20 2017-05-26 Duke University Lactate biosensors and uses thereof
US11099176B2 (en) 2015-11-20 2021-08-24 Duke University Lactate biosensors and uses thereof
US11906524B2 (en) 2015-11-20 2024-02-20 Duke University Urea biosensors and uses thereof

Also Published As

Publication number Publication date
US20040118681A1 (en) 2004-06-24
DK1552303T3 (en) 2009-05-25
US10712341B2 (en) 2020-07-14
CN100492010C (en) 2009-05-27
US9625458B2 (en) 2017-04-18
AU2003277377B2 (en) 2009-05-21
EP1552303B1 (en) 2009-02-18
WO2004036176A2 (en) 2004-04-29
JP4571499B2 (en) 2010-10-27
ES2321931T3 (en) 2009-06-15
ATE423318T1 (en) 2009-03-15
KR101107630B1 (en) 2012-01-25
AU2003277377A1 (en) 2004-05-04
CN1705882A (en) 2005-12-07
DE60326256D1 (en) 2009-04-02
JP2006503293A (en) 2006-01-26
WO2004036176A3 (en) 2004-07-29
CA2502272A1 (en) 2004-04-29
EP1552303A4 (en) 2006-12-27
CA2502272C (en) 2011-10-11
EP1552303A2 (en) 2005-07-13
KR20050083772A (en) 2005-08-26
US20170322209A1 (en) 2017-11-09
HK1080937A1 (en) 2006-05-04

Similar Documents

Publication Publication Date Title
US10712341B2 (en) Biosensor
De Lorimier et al. Construction of a fluorescent biosensor family
CA2316199C (en) Biosensor
Marvin et al. Engineering biosensors by introducing fluorescent allosteric signal transducers: construction of a novel glucose sensor
Salins et al. A novel reagentless sensing system for measuring glucose based on the galactose/glucose-binding protein
Khan et al. Fluorescence intensity-and lifetime-based glucose sensing using an engineered high-Kd mutant of glucose/galactose-binding protein
Mohsin et al. Genetically encoded FRET-based nanosensor for in vivo measurement of leucine
EP2426500B1 (en) Method for detecting a ligand using fret biosensor
US11099176B2 (en) Lactate biosensors and uses thereof
Tian et al. Structure‐based design of robust glucose biosensors using a Thermotoga maritima periplasmic glucose‐binding protein
Sakaguchi-Mikami et al. Engineering of ligand specificity of periplasmic binding protein for glucose sensing
Constantinou et al. Opportunities for bioprocess monitoring using FRET biosensors
Vercillo et al. Analysis of ligand binding to a ribose biosensor using site‐directed mutagenesis and fluorescence spectroscopy
Hellinga et al. Biosensor
Hellinga et al. Biosensor
Grünewald Periplasmic binding proteins in biosensing applications
Soleja et al. Enhanced sensitivity and detection range of FRET-based vitamin B 12 nanosensor
Yu et al. Highly Sensitive and Selective Detection of Inorganic Phosphates in the Water Environment by Biosensors Based on Bioluminescence Resonance Energy Transfer
Hsieh et al. Fluorescence Glucose Sensing, Part I: Fluorescence Resonance Energy Transfer Glucose Sensor from Site-Specific Dual Labeling of Glucose/Galactose Binding Protein Using Ligand Protection
Shetty et al. Recombinant methods in protein and whole-cell biosensing
Tian Protein engineering and design of thermostable biosensors
EP1908771A1 (en) Thermophilic sulfate-binding protein, its use and a biosensor comprising the protein

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- AFTER EXAMINER'S ANSWER OR BOARD OF APPEALS DECISION

AS Assignment

Owner name: DUKE UNIVERSITY, NORTH CAROLINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HELLINGA, HOMME W.;LOOGER, LOREN L.;REEL/FRAME:039661/0555

Effective date: 20031114