US20030212265A1 - Fluorescent protein sensors for measuring the pH of a biological sample - Google Patents

Fluorescent protein sensors for measuring the pH of a biological sample Download PDF

Info

Publication number
US20030212265A1
US20030212265A1 US10/457,982 US45798203A US2003212265A1 US 20030212265 A1 US20030212265 A1 US 20030212265A1 US 45798203 A US45798203 A US 45798203A US 2003212265 A1 US2003212265 A1 US 2003212265A1
Authority
US
United States
Prior art keywords
gly
leu
amino acid
lys
acid sequence
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
US10/457,982
Inventor
Roger Tsien
Juan Llopis
Rebekka Wachter
S. Remington
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.)
Individual
Original Assignee
Individual
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
Priority claimed from US09/602,641 external-priority patent/US6608189B1/en
Application filed by Individual filed Critical Individual
Priority to US10/457,982 priority Critical patent/US20030212265A1/en
Publication of US20030212265A1 publication Critical patent/US20030212265A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43595Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from coelenteratae, e.g. medusae
    • 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
    • G01N33/84Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving inorganic compounds or pH
    • 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
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/43504Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from invertebrates
    • G01N2333/43595Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from invertebrates from coelenteratae, e.g. medusae
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/81Packaged device or kit

Definitions

  • the invention relates generally to compositions and methods for measuring the pH of a sample and more particularly to fluorescent protein sensors for measuring the pH of a biological sample.
  • the pH within various cellular compartments is regulated to provide for the optimal activity of many cellular processes.
  • posttranslational processing of secretory proteins, the cleavage of prohormones, and the retrieval of escaped luminal endoplasmic reticulum proteins are all pH-dependent.
  • a second method for measuring pH utilizes retrograde transport of fluorescein-labeled verotoxin 1B, which stains the entire Golgi complex en route to the endoplasmic reticulum.
  • This method can be used, however, only in cells bearing the receptor globotriaosyl ceramide on the plasma membrane, and it may be limited by the residence time of the verotoxin in transit through the Golgi.
  • intracellular pH has been measured using the chimeric protein CD25-TGN38, which cycles between the trans-Golgi network and the plasma membrane.
  • the CD25-motif binds extra-cellular anti-CD25 antibodies conjugated with a pH-sensitive fluorophore. Measurement of fluorescence upon return of the bound complex to the Golgi can be used to measure the pH of the organelle.
  • the invention is based on the discovery that proteins derived from the Aequora victoria green fluorescence protein (GFP) show reversible changes in fluorescence over physiological pH ranges.
  • GFP green fluorescence protein
  • the invention provides a method for determining the pH of a sample by contacting the sample with an indicator including a first fluorescent protein moiety whose emission intensity changes as the pH varies between 5 and 10, exciting the indicator, and the determining the intensity at a first wavelength.
  • the emission intensity of the first fluorescent protein moiety indicates the pH of the sample.
  • the invention provides a method for determining the pH of a region of a cell by introducing into the cell a polynucleotide encoding a polypeptide including a first fluorescent protein moiety whose emission intensity changes as the pH varies between 5 and 10, culturing the cell under conditions that permit expression of the polynucleotide, and determining the intensity at a first wavelength.
  • the emission intensity of the first fluorescent protein moiety indicates the pH of the sample.
  • the invention provides a functional engineered fluorescent protein whose amino acid sequence is substantially identical to the amino acid sequence of the 238 amino acid Aequora victoria green fluorescence protein shown in FIG. 3 of U.S. Ser. No. 08/911,825 (SEQ ID NO:______), and whose emission intensity changes as pH varies between 5 and 10.
  • the invention provides a polynucleotide encoding the functional engineered fluorescent protein.
  • the invention also includes a kit useful for the detection of pH in a sample, e.g., a region of a cell.
  • the kit includes a carrier means containing one or more containers comprising a first container containing a polynucleotide encoding a polypeptide including a first fluorescent protein moiety whose emission intensity changes as the pH varies between 5 and 10.
  • FIG. 1 is a schematic diagram depicting fluorescent protein sensors used as indicators of intracellular pH.
  • FIGS. 2A and 2B are graphs showing absorbance as a function of wavelength for the fluorescent protein pH sensor EYFP (SEQ ID NO:______) at various wavelengths (FIG. 2A), and the pH dependency of fluorescence of various GFP fluorescent protein sensors in vitro and in cells (FIG. 2B).
  • the fluorescence intensity of purified recombinant GFP mutant protein (solid symbols) as a function of pH was measured in a microplate fluorometer.
  • GT type II membrane-anchored protein galactosyltransferase
  • GT type II membrane-anchored protein galactosyltransferase
  • FIGS. 3A and 3B are graphs showing ratiometric measurements of pH G by cotransfecting HeLa cells with polynucleotides encoding GT-ECFP and GT-EYFP.
  • FIG. 3A is a graph showing single wavelength fluorescence intensities of GT-EYFP and GT-ECFP in the Golgi region of a HeLa cell.
  • FIG. 3B is a graph showing the ratio of GT-EYFP/GT-ECFP fluorescence in the same cell as a function of time.
  • FIG. 4 is a graph showing the change in mitochondrial pH of HeLa cells expressing YFP H148G.
  • FIG. 5 is a graph showing the mitochondrial pH of chick skeletal myotubes expressing YFP H148G in the mitochondrial matrix.
  • FIG. 6 is a graph showing fluorescence and pH in HeLa cells expressing YFP H148Q targeted to the mitochondrial matrix.
  • FIG. 7 is a graph showing a ratiometric measurement of mitochondrial pH following expression of YFP H148G (pH sensitive) and GFP T203I (pH insensitive) in mitochondria of HeLa cells.
  • FIG. 8 is a graph showing normalized absorbance of WT GFP and YFP in 75 mM phosphate pH 8.0, 140 mM NACl. Solid lines, WT GFP; Dashed lines, YFP.
  • FIG. 9 is a stereoview of the 2F o -F c electron density map of the YFP chromophore and the stacked Tyr203 after refinement.
  • the 2.5 ⁇ resolution map was contoured at +1 standard deviation.
  • the invention provides genes encoding fluorescent sensor proteins, or fragments thereof, whose fluorescence is sensitive to changes in pH at a range between 5 and 10.
  • the proteins of the invention are useful for measuring the pH of a sample.
  • the sample can be a biological sample and can include an intracellular region of a cell, such as the lumen of the mitochondria or golgi.
  • the pH of a sample is determined by observing the fluorescence of the fluorescent sensor protein.
  • the fluorescent protein pH sensor have a broad applicability to cells and organisms that are amenable to gene transfer. Problems associated with the use of other agents used to measure pH, e.g., problems associated with permeabilizing cells to ester-containing agents, leakage of agents, or hydrolysis of agents are avoided. With the fluorescent protein pH sensors of the invention, no leakage occurs over the course of a typical measurement, even when the measurement is made at 37° C.
  • compositions and methods described herein also avoid the need to express and purify large quantities of soluble recombinant protein, purify and label it in vitro, microinject it back into cells.
  • An important advantage of the fluorescent protein pH sensors of the invention is that they can be delivered to cells in the form of polynucleotides encoding the protein sensor fused to a targeting signal or signals.
  • the targeting signal directs the expression of the protein sensors to restricted cell locations. Thus, it is possible to measure the pH of a precisely defined cellular region or organelle.
  • the invention provides a functional engineered fluorescent protein whose amino acid sequence is substantially identical to the 238 amino acid Aequora victoria green fluorescence protein shown in FIG. 3 of U.S. Ser. No. 08/911,825 (SEQ ID NO:______).
  • fluorescent protein refers to any protein capable of emitting light when excited with appropriate electromagnetic radiation, and which has an amino acid sequence that is either natural or engineered and is derived from the amino acid sequence of Aequorea-related fluorescent protein.
  • fluorescent protein pH sensor refers to a fluorescent protein whose emitted light varies with changes in pH from 5 to 10.
  • the invention also includes functional polypeptide fragments of a fluorescent protein pH sensor.
  • functional polypeptide fragment refers to a polypeptide which possesses biological function or activity which is identified through a defined functional assay and which is associated with a particular biologic, morphologic, or phenotypic alteration in the cell.
  • functional fragments of a functional engineered fluorescent protein refers to fragments of a functional engineered protein that retain a function of the engineered fluorescent protein, e.g., the ability to fluoresce in a pH-dependent manner over the pH range 5 to 10.
  • Biologically functional fragments can vary in size from a polypeptide fragment as small as an epitope to a large polypeptide.
  • a functional engineered fluorescent protein includes amino acid sequences substantially the same as the sequence set forth in SEQ ID NO:______, and whose emission intensity changes as pH varies between 5 and 10. In some embodiments the emission intensity of the functional engineered fluorescent protein changes as pH varies between 5 and 8.5.
  • substantially identical is meant a protein or polypeptide that retains the activity of a functional engineered protein, or nucleic acid encoding the same, and which exhibits at least 80%, preferably 85%, more preferably 90%, and most preferably 95% homology to a reference amino acid or nucleic acid sequence.
  • the length of comparison sequences will generally be at least 16 amino acids, preferably at least 20 amino acids, more preferably at least 25 amino acids, and most preferably 35 amino acids.
  • the length of comparison sequences will generally be at least 50 nucleotides, preferably at least 60 nucleotides, more preferably at least 75 nucleotides, and most preferably 110 nucleotides.
  • substantially identical is meant an amino acid sequence which differs only by conservative amino acid substitutions, for example, substitution of one amino acid for another of the same class (e.g., valine for glycine, arginine for lysine, etc.) or by one or more non-conservative substitutions, deletions, or insertions located at positions of the amino acid sequence which do not destroy the function of the protein (assayed, e.g., as described herein).
  • conservative amino acid substitutions for example, substitution of one amino acid for another of the same class (e.g., valine for glycine, arginine for lysine, etc.) or by one or more non-conservative substitutions, deletions, or insertions located at positions of the amino acid sequence which do not destroy the function of the protein (assayed, e.g., as described herein).
  • such a sequence is at least 85%, more preferably 90%, more preferably 95%, more preferably 98%, and most preferably 99% identical at the amino acid level to one of the sequences of EGFP (SEQ ID NO:______), EYFP (SEQ ID NO: ______), ECFP (SEQ ID NO: ______), EYFP-V68L/Q69K (SEQ ID NO:______), YFP H148G (SEQ ID NO:______), or YFP H148Q (SEQ ID NO:_______).
  • Homology is typically measured using sequence analysis software (e.g., Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705). Such software matches similar sequences by assigning degrees of homology to various substitutions, deletions, substitutions, and other modifications. Conservative substitutions typically include substitutions within the following groups: glycine alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine.
  • sequence analysis software e.g., Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705
  • Conservative substitutions typically include substitutions within the following groups: glycine alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparag
  • the amino acid sequence of the protein includes one of the following sets of substitutions in the amino acid sequence of the Aequora green fluorescent protein (SEQ ID NO:______): F64L/S65T/H231L, referred to herein as EGFP (SEQ ID NO:______); S65G/S72A/T203Y/H231L, referred to herein as EYFP (SEQ ID NO:______); S65G/V68L/Q69K/S72A/T203Y/H231L, referred to herein as EYFP-V68L/Q69K (SEQ ID NO:______); K26R/F64L/S65T/Y66W/N146I/M153T/V163A/N164H/H231L, referred to herein as ECFP (SEQ ID NO:_______).
  • the amino acid sequences of EGFP, EYFP, ECFP, and EYFP-V68L/Q69K are shown in Tables 1-4, respectively.
  • the amino acids are numbered with the amino acid following the initiating methionine assigned the ‘1’ position.
  • F64L corresponds to a substitution of leucine for phenylalanine in the 64th amino acid following the initiating methionine.
  • the amino acid sequence of the protein is based on the sequence of the wild-type Aequora green fluorescent protein, but includes the substitution H148G (SEQ ID NO:______) or H148Q (SEQ ID NO:______).
  • these substitutions can be present along with other substitutions, e.g., the proteins can include the substitutions S65G/V68L/S72A/H148G/Q80R/T203Y (SEQ ID NO:______), which is referred to herein as the “YFP H148G mutant,” S65G/V68L/S72A/H148Q/Q80R/T203Y, which is referred to herein as the “YFP H148Q mutant” (SEQ ID NO:______), the as well as EYFP-H148G (SEQ ID NO: ______) and EFP-H148Q (SEQ ID NO: ______).
  • the amino acid sequences of these mutants are shown in Tables 5-8, respectively.
  • EYFP-H148G (SEQ ID NO:_) MVSKGEELFTGVVPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICT TGKLPVPWPTLVTTFGYGVQCFARYPDHMKQHDFFKSAMPEGYVQERTIF FKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGHKLEYNYNSGN VYIMADKQKNGIKVNFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNH YLSYQSALSKDPNEKRDHMVLLEFVTAAGITLGMDELYK
  • the protein or polypeptide is substantially purified.
  • substantially pure protein or polypeptide is meant an functional engineered fluorescent polypeptide which has been separated from components which naturally accompany it.
  • the protein or polypeptide is substantially pure when it is at least 60%, by weight, free from the proteins and naturally-occurring organic molecules with which it is naturally associated.
  • the preparation is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight, of the protein.
  • a substantially pure protein may be obtained, for example, by extraction from a natural source (e.g., a plant cell); by expression of a recombinant nucleic acid encoding a functional engineered fluorescent protein; or by chemically synthesizing the protein. Purity can be measured by any appropriate method, e.g., those described in column chromatography, polyacrylamide gel electrophoresis, or by HPLC analysis.
  • a natural source e.g., a plant cell
  • Purity can be measured by any appropriate method, e.g., those described in column chromatography, polyacrylamide gel electrophoresis, or by HPLC analysis.
  • a protein or polypeptide is substantially free of naturally associated components when it is separated from those contaminants which accompany it in its natural state.
  • a protein or polypeptide which is chemically synthesized or produced in a cellular system different from the cell from which it naturally originates will be substantially free from its naturally associated components.
  • substantially pure polypeptides include those derived from eukaryotic organisms but synthesized in E. coli or other prokaryotes.
  • the invention also provides polynucleotides encoding the functional engineered fluorescent protein described herein.
  • These polynucleotides include DNA, cDNA, and RNA sequences which encode functional engineered fluorescent proteins. It is understood that all polynucleotides encoding functional engineered fluorescent proteins are also included herein, as long as they encode a protein or polypeptide whose fluorescent emission intensity changes as pH varies between 5 and 10.
  • Such polynucleotides include naturally occurring, synthetic, and intentionally manipulated polynucleotides. For example, the polynucleotide may be subjected to site-directed mutagenesis.
  • the polynucleotides of the invention include sequences that are degenerate as a result of the genetic code. Therefore, all degenerate nucleotide sequences are included in the invention as long as the amino acid sequence of the functional engineered fluorescent protein or derivative is functionally unchanged.
  • a polynucleotide sequence encoding a functional engineered fluorescent protein that includes one of the following sets of substitutions in the amino acid sequence of the Aequora green fluorescent protein (SEQ ID NO:______): S65G/S72A/T203Y/H231L, S65G/V68L/Q69K/S72A/T203Y/H231L, or K26R/F64L/S65T/Y66W/N146I/M153T/V163A/N164H/H231L.
  • the DNA sequences encoding EGFP, EYFP, ECFP, EYFP-V68L/Q69K, YFP H148G, and YFP H148Q are those shown in Table 9-16 (SEQ ID NOs: ______ to ______), respectively.
  • nucleic acid encoding functional engineered fluorescent proteins may be reflect the codon choice in the native A. victoria coding sequence, or, alternatively, may be chosen to reflect the optimal codon frequencies used in the organism in which the proteins will be expressed.
  • nucleic acids encoding a target functional engineered protein to be expressed in a human cell may have use a codon choice that is optimized for mammals, or especially humans.
  • polynucleotide refers to a polymeric form of nucleotides of at least 10 bases in length.
  • the nucleotides can be ribonucleotides, deoxyribonucleotides, or modified forms of either type of nucleotide.
  • the term includes single and double stranded forms of DNA.
  • isolated polynucleotide is meant a polynucleotide that is not immediately contiguous with both of the coding sequences with which it is immediately contiguous (one on the 5′ end and one on the 3′ end) in the naturally occurring genome of the organism from which it is derived.
  • the term therefore includes, for example, a recombinant DNA which is incorporated into a vector, e.g., an expression vector; into an autonomously replicating plasmid or virus; or into the genomic DNA of a prokaryote or eukaryote, or which exists as a separate molecule (e.g., a cDNA) independent of other sequences.
  • a vector e.g., an expression vector
  • an autonomously replicating plasmid or virus or into the genomic DNA of a prokaryote or eukaryote, or which exists as a separate molecule (e.g., a cDNA) independent of other sequences.
  • a “substantially identical”,nucleic acid sequence codes for a substantially identical amino acid sequence as defined above.
  • the functional engineered fluorescent protein can also include a targeting sequence to direct the fluorescent protein to particular cellular sites by fusion to appropriate organellar targeting signals or localized host proteins.
  • a polynucleotide encoding a targeting sequence can be ligated to the 5′ terminus of a polynucleotide encoding the fluorescence such that the targeting peptide is located at the amino terminal end of the resulting fusion polynucleotide/polypeptide.
  • the targeting sequence can be, e.g., a signal peptide. In the case of eukaryotes, the signal peptide is believed to function to transport the fusion polypeptide across the endoplasmic reticulum.
  • the secretory protein is then transported through the Golgi apparatus, into secretory vesicles and into the extracellular space or, preferably, the external environment.
  • Signal peptides which can be utilized according to the invention include pre-pro peptides which contain a proteolytic enzyme recognition site. Other signal peptides with similar properties to pro-calcitonin described herein are known to those skilled in the art, or can be readily ascertained without undue experimentation.
  • the targeting sequence can also be a nuclear localization sequence, an endoplasmic reticulum localization sequence, a peroxisome localization sequence, a mitochondrial localization sequence, or a localized protein.
  • Targeting sequences can be targeting sequences which are described, for example, in “Protein Targeting”, chapter 35 of Stryer, L., Biochemistry (4th ed.). W. H. Freeman, 1995.
  • the localization sequence can also be a localized protein.
  • Some important targeting sequences include those targeting the nucleus (KKKRK) (SEQ ID NO: ______), mitochondrion (the 12 amino terminal amino acids of the cytochrome c oxidase subunit IV gene (SEQ ID NO:______), or the amino terminal sequence MLRTSSLFTRRVQPSLFRNILRLQST (SEQ ID NO:______), endoplasmic reticulum (KDEL (SEQ ID NO:_______) at the C-terminus, assuming a signal sequence present at N-terminus), peroxisome (SKF at C-terminus), prenylation or insertion into plasma membrane (CaaX, CC, CXC, or CCXX at C-terminus), cytoplasmic side of plasma membrane (fusion to SNAP-25), or the Golgi apparatus (fusion to the amino terminal 81 amino acids of human type II membrane-anchored protein galactosyltransferase (SEQ ID NO:__
  • Examples of targeting sequences linked to functional engineered fluorescent proteins include GT-EYFP (SEQ ID NO:______), GT-ECFP (SEQ ID NO:_____), GT-EGFP (SEQ ID NO:______), and GT-EYFP-V68L/Q69K (SEQ ID NO:_______), which are targeted to the Golgi apparatus using sequences from the GT protein; and EYFP-mito (SEQ ID NO:______) and EGFP-mito (SEQ ID NO:_______), which are targeted to the mitochondrial matrix using sequences from the amino terminal region of the cytochrome c oxidase subunit IV gene.
  • the EYFP, ECFP, EGFP, and EYFP-V68L/Q69K amino acid sequences, as well as nucleic acids encoding these polypeptides, are described above.
  • the GT-derived targeting sequence corresponds to the 81 amino terminal amino acids of the human GT sequence.
  • the GT amino acid sequences, and the polynucleotide sequences encoding the GT amino acid sequences, are described in Genbank Accession No. M70427 and Mengle-Gaw et al., Biochem. Biophys. Res. Commun. 176 (3), 1269-1276 (1991).
  • nucleic acid sequences encoding targeting sequences linked to functional engineered fluorescent proteins have the sequences shown in Tables 23-32. TABLE 17 mito-ECFP Amino Acid Sequence (SEQ ID NO:_) MLSLRQSIRFFKRSGIMVSKGEELFTGVVPILVELDGDVNGHRFSVSGEG EGDATYGKLTLKFICTTGKLPVPWPTLVTTLTWGVQCFSRYPDHMKQHDF FKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKED GNILGHKLEYNYISHNVYITADKQKNGIKAHFKIRHNIEDGSVQLADHYQ QNTPIGDGPVLLPDNHYLSTQSALSKDPNEKRDHMVLLEFVTAAGITLGM DELYK
  • the fluorescent indicators can be produced as proteins fused to other fluorescent indicators or targeting sequences by recombinant DNA technology.
  • Recombinant production of fluorescent proteins involves expressing nucleic acids having sequences that encode the proteins.
  • Nucleic acids encoding fluorescent proteins can be obtained by methods known in the art.
  • a nucleic acid encoding the protein can be isolated by polymerase chain reaction of cDNA from A. victoria using primers based on the DNA sequence of A. victoria green fluorescent protein. PCR methods are described in, for example, U.S. Pat. No. 4,683,195; Mullis, et al. Cold Spring Harbor Symp. Quant. Biol.
  • Mutant versions of fluorescent proteins can be made by site-specific mutagenesis of other nucleic acids encoding fluorescent proteins, or by random mutagenesis caused by increasing the error rate of PCR of the original polynucleotide with 0.1 mM MnCl 2 and unbalanced nucleotide concentrations. See, e.g., U.S. patent application Ser. No. 08/337,915, filed Nov. 10, 1994 or International application PCT/US95/14692, filed Nov. 10, 1995.
  • Nucleic acids used to transfect cells with sequences coding for expression of the polypeptide of interest generally will be in the form of an expression vector including expression control sequences operatively linked to a nucleotide sequence coding for expression of the polypeptide.
  • operatively linked refers to a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner.
  • a control sequence operatively linked to a coding sequence is ligated such that expression of the coding sequence is achieved under conditions compatible with the control sequences.
  • Control sequence refers to polynucleotide sequences which are necessary to effect the expression of coding and non-coding sequences to which they are ligated.
  • Control sequences generally include promoter, ribosomal binding site, and transcription termination sequence.
  • the term “control sequences” is intended to include, at a minimum, components whose presence can influence expression, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
  • nucleotide sequence coding for expression of a polypeptide refers to a sequence that, upon transcription and translation of mRNA, produces the polypeptide. This can include sequences containing, e.g., introns.
  • expression control sequences refers to nucleic acid sequences that regulate the expression of a nucleic acid sequence to which it is operatively linked. Expression control sequences are operatively linked to a nucleic acid sequence when the expression control sequences control and regulate the transcription and, as appropriate, translation of the nucleic acid sequence.
  • expression control sequences can include appropriate promoters, enhancers, transcription terminators, a start codon (i.e., ATG) in front of a protein-encoding gene, splicing signals for introns, maintenance of the correct reading frame of that gene to permit proper translation of the mRNA, and stop codons.
  • start codon i.e., ATG
  • Methods which are well known to those skilled in the art can be used to construct expression vectors containing the fluorescent indicator coding sequence and appropriate transcriptional/translational control signals. These methods include in vitro recombinant DNA techniques, synthetic techniques and in vivo recombination/genetic recombination. (See, for example, the techniques described in Maniatis, et al., Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., 1989). Transformation of a host cell with recombinant DNA may be carried out by conventional techniques as are well known to those skilled in the art. Where the host is prokaryotic, such as E.
  • competent cells which are capable of DNA uptake can be prepared from cells harvested after exponential growth phase and subsequently treated by the CaCl 2 method by procedures well known in the art. Alternatively, MgCl 2 or RbCl can be used. Transformation can also be performed after forming a protoplast of the host cell or by electroporation.
  • Eukaryotic cells can also be cotransfected with DNA sequences encoding the fusion polypeptide of the invention, and a second foreign DNA molecule encoding a selectable phenotype, such as the herpes simplex thymidine kinase gene.
  • Another method is to use a eukaryotic viral vector, such as simian virus 40 (SV40) or bovine papilloma virus, to transiently infect or transform eukaryotic cells and express the protein.
  • a eukaryotic viral vector such as simian virus 40 (SV40) or bovine papilloma virus
  • SV40 simian virus 40
  • bovine papilloma virus bovine papilloma virus
  • Techniques for the isolation and purification of polypeptides of the invention expressed in prokaryotes or eukaryotes may be by any conventional means such as, for example, preparative chromatographic separations and immunological separations such as those involving the use of monoclonal or polyclonal antibodies or antigen.
  • a variety of host-expression vector systems may be utilized to express fluorescent indicator coding sequence. These include but are not limited to microorganisms such as bacteria transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing a fluorescent indicator coding sequence; yeast transformed with recombinant yeast expression vectors containing the fluorescent indicator coding sequence; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing a fluorescent indicator coding sequence; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing a fluorescent indicator coding sequence; or animal cell systems infected with recombinant virus expression vectors (e.g., retroviruses, adenovirus, vaccinia virus) containing a fluorescent indicator coding
  • any of a number of suitable transcription and translation elements including constitutive and inducible promoters, transcription enhancer elements, transcription terminators, etc. may be used in the expression vector (see, e.g., Bitter, et al., Methods in Enzymology 153:516-544, 1987).
  • inducible promoters such as pL of bacteriophage ⁇ , plac, ptrp, ptac (ptrp-lac hybrid promoter) and the like may be used.
  • promoters derived from the genome of mammalian cells e.g., metallothionein promoter
  • mammalian viruses e.g., the retrovirus long terminal repeat; the adenovirus late promoter; the vaccinia virus 7.5K promoter
  • Promoters produced by recombinant DNA or synthetic techniques may also be used to provide for transcription of the inserted fluorescent indicator coding sequence.
  • a number of expression vectors may be advantageously selected depending upon the use intended for the fluorescent indicator expressed. For example, when large quantities of the fluorescent indicator are to be produced, vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable. Those which are engineered to contain a cleavage site to aid in recovering fluorescent indicator are preferred. In yeast, a number of vectors containing constitutive or inducible promoters may be used. For a review see, Current Protocols in Molecular Biology, Vol. 2, Ed. Ausubel, et al., Greene Publish. Assoc. & Wiley Interscience, Ch.
  • a constitutive yeast promoter such as ADH or LEU2 or an inducible promoter such as GAL may be used (Cloning in Yeast, Ch. 3, R. Rothstein In: DNA Cloning Vol. 11, A Practical Approach, Ed. D M Glover, IRL Press, Wash., D.C., 1986).
  • vectors may be used which promote integration of foreign DNA sequences into the yeast chromosome.
  • the expression of a fluorescent indicator coding sequence may be driven by any of a number of promoters.
  • viral promoters such as the 35S RNA and 19S RNA promoters of CaMV (Brisson, et al., Nature 310:511-514, 1984), or the coat protein promoter to TMV (Takamatsu, et al., EMBO J. 3:17-311, 1987) may be used; alternatively, plant promoters such as the small subunit of RUBISCO (Coruzzi, et al., 1984, EMBO J.
  • An alternative expression system which could be used to express fluorescent indicator is an insect system.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes.
  • the virus grows in Spodoptera frugiperda cells.
  • the fluorescent indicator coding sequence may be cloned into non-essential regions (for example, the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
  • Successful insertion of the fluorescent indicator coding sequence will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat coded for by the polyhedrin gene).
  • Eukaryotic systems and preferably mammalian expression systems, allow for proper post-translational modifications of expressed mammalian proteins to occur.
  • Eukaryotic cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, phosphorylation, and, advantageously secretion of the gene product should be used as host cells for the expression of fluorescent indicator.
  • host cell lines may include but are not limited to CHO, VERO, BHK, HeLa, COS, MDCK, Jurkat, HEK-293, and WI38.
  • Primary cell lines, such as neonatal rat myocytes, can also be used.
  • Mammalian cell systems which utilize recombinant viruses or viral elements to direct expression may be engineered.
  • the fluorescent indicator coding sequence may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence.
  • This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region E1 or E3) will result in a recombinant virus that is viable and capable of expressing the fluorescent indicator in infected hosts (e.g., see Logan & Shenk, Proc.
  • the vaccinia virus 7.5K promoter may-be used (e.g., see, Mackett, et al., Proc. Natl. Acad. Sci. USA, 79: 7415-7419, 1982; Mackett, et al., J. Virol. 49: 857-864,-1984; Panicali, et al., Proc. Natl. Acad. Sci. USA 79: 4927-4931, 1982).
  • vectors based on bovine papilloma virus which have the ability to replicate as extrachromosomal elements (Sarver, et al., Mol. Cell. Biol.
  • the plasmid replicates to about 100 to 200 copies per cell. Transcription of the inserted cDNA does not require integration of the plasmid into the host's chromosome, thereby yielding a high level of expression.
  • These vectors can be used for stable expression by including a selectable marker in the plasmid, such as the neo gene.
  • the retroviral genome can be modified for use as a vector capable of introducing and directing the expression of the fluorescent indicator gene in host cells (Cone & Mulligan, Proc. Natl. Acad. Sci. USA, 81:6349-6353, 1984). High level expression may also be achieved using inducible promoters, including, but not limited to, the metallothionein IIA promoter and heat shock promoters.
  • the recombinant nucleic acid can be incorporated into an expression vector including expression control sequences operatively linked to the recombinant nucleic acid.
  • the expression vector can be adapted for function in prokaryotes or eukaryotes by inclusion of appropriate promoters, replication sequences, markers, etc.
  • DNA sequences encoding the fluorescence indicator polypeptide of the invention can be expressed in vitro or in vivo by DNA transfer into a suitable recombinant host cell.
  • recombinant host cells are cells in which a vector can be propagated and its DNA expressed.
  • the term also includes any progeny of the subject host cell. It is understood that all progeny may not be identical to the parental cell since there may be mutations that occur during replication. However, such progeny are included when the term “recombinant host cell” is used. Methods of stable transfer, in other words when the foreign DNA is continuously maintained in the host, are known in the art.
  • the expression vector can be transfected into a host cell for expression of the recombinant nucleic acid.
  • Recombinant host cells can be selected for high levels of expression in order to purify the fluorescent indicator fusion protein.
  • E. coli is useful for this purpose.
  • the host cell can be a prokaryotic or eukaryotic cell selected to study the activity of an enzyme produced by the cell.
  • the linker peptide is selected to include an amino acid sequence recognized by the protease.
  • the cell can be, e.g., a cultured cell or a cell taken in vivo from a transgenic animal.
  • the invention provides a transgenic non-human animal that expresses a polynucleotide sequence which encodes a fluorescent protein pH sensor.
  • the “non-human animals” of the invention comprise any non-human animal having a polynucleotide sequence which encodes a fluorescent indicator.
  • Such non-human animals include vertebrates such as rodents, non-human primates, sheep, dog, cow, pig, amphibians, and reptiles. Preferred non-human animals are selected from the rodent family including rat and mouse, most preferably mouse.
  • the “transgenic non-human animals” of the invention are produced by introducing “transgenes” into the germline of the non-human animal. Embryonal target cells at various developmental stages can be used to introduce transgenes. Different methods are used depending on the stage of development of the embryonal target cell. The zygote is the best target for micro-injection.
  • the male pronucleus reaches the size of approximately 20 micrometers in diameter which allows reproducible injection of 1-2 pl of DNA solution.
  • the use of zygotes as a target for gene transfer has a major advantage in that in most cases the injected DNA will be incorporated into the host gene before the first cleavage (Brinster et al., Proc. Natl. Acad. Sci. USA 82:4438-4442, 1985). As a consequence, all cells of the transgenic non-human animal will carry the incorporated transgene. This will in general also be reflected in the efficient transmission of the transgene to offspring of the founder since 50% of the germ cells will harbor the transgene. Microinjection of zygotes is the preferred method for incorporating transgenes in practicing the invention.
  • transgenic is used to describe an animal which includes exogenous genetic material within all of its cells.
  • a “transgenic” animal can be produced by cross-breeding two chimeric animals which include exogenous genetic material within cells used in reproduction. Twenty-five percent of the resulting offspring will be transgenic, i.e., animals which include the exogenous genetic material within all of their cells in both alleles. 50% of the resulting animals will include the exogenous genetic material within one allele and 25% will include no exogenous genetic material.
  • Retroviral infection can also be used to introduce transgene into a non-human animal.
  • the developing non-human embryo can be cultured in vitro to the blastocyst stage.
  • the blastomeres can be targets for retro viral infection (Jaenisch, R., Proc. Natl. Acad. Sci USA 73:1260-1264, 1976).
  • Efficient infection of the blastomeres is obtained by enzymatic treatment to remove the zona pellucida (Hogan, et al. (1986) in Manipulating the Mouse Embryo, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.).
  • the viral vector system used to introduce the transgene is typically a replication-defective retrovirus carrying the transgene (Jahner, et al., Proc. Natl. Acad. Sci. USA 82:6927-6931, 1985; Van der Putten, et al., Proc. Natl. Acad. Sci USA 82:6148-6152, 1985). Transfection is easily and efficiently obtained by culturing the blastomeres on a monolayer of virus-producing cells (Van der Putten, supra; Stewart, et al., EMBO J. 6:383-388, 1987). Alternatively, infection can be performed at a later stage. Virus or virus-producing cells can be injected into the blastocoele (D.
  • ES embryonal stem cell
  • Transgenes can be efficiently introduced into the ES cells by DNA transfection or by retrovirus-mediated transduction. Such transformed ES cells can thereafter be combined with blastocysts from a nonhuman animal. The ES cells thereafter colonize the embryo and contribute to the germ line of the resulting chimeric animal. (For review see Jaenisch, R., Science 240: 1468-1474, 1988).
  • “Transformed” means a cell into which (or into an ancestor of which) has been introduced, by means of recombinant nucleic acid techniques, a heterologous polynucleotide.
  • “Heterologous” refers to a polynucleotide sequence that either originates from another species or is modified from either its original form or the form primarily expressed in the cell.
  • Transgene means any piece of DNA which is inserted by artifice into a cell, and becomes part of the genome of the organism (i.e., either stably integrated or as a stable extrachromosomal element) which develops from that-cell.
  • a transgene may include a gene which is partly or entirely heterologous (i.e., foreign) to the transgenic organism, or may represent a gene homologous to an endogenous gene of the organism. Included within this definition is a transgene created by the providing of an RNA sequence which is transcribed into DNA and then incorporated into the genome.
  • the transgenes of the invention include DNA sequences which encode the fluorescent indicator which may be expressed in a transgenic non-human animal.
  • transgenic as used herein additionally includes any organism whose genome has been altered by in vitro manipulation of the early embryo or fertilized egg or by any transgenic technology to induce a specific gene knockout.
  • gene knockout refers to the targeted disruption of a gene in vivo with complete loss of function that has been achieved by any transgenic technology familiar to those in the art.
  • transgenic animals having gene knockouts are those in which the target gene has been rendered nonfunctional by an insertion targeted to the gene to be rendered non-functional by homologous recombination.
  • transgenic includes any transgenic technology familiar to those in the art which can produce an organism carrying an introduced transgene or one in which an endogenous gene has been rendered non-functional or “knocked out.”
  • the invention provides a method for determining the pH of a sample by contacting the sample with an indicator including a first fluorescent protein moiety whose emission intensity changes as pH varies between pH 5 and 10, exciting the indicator, and then determining the intensity of light emitted by the first fluorescent protein moiety at a first wavelength.
  • the emission intensity of the first fluorescent protein moiety indicates the pH of the sample.
  • the fluorescent protein moiety can be a functional engineered protein substantially identical to the amino acid sequence of Aequora green fluorescence protein (SEQ ID NO: ______).
  • Preferred green fluorescence proteins include those having a functional engineered fluorescent protein that includes one of the following sets of substitutions in the amino acid sequence of the Aequora green fluorescent protein (SEQ ID NO:______): S65G/S72A/T203Y/H231L, S65G/V68L/Q69K/S72A/T203Y, or K26R/F64L/S65T/Y66W/N146I/M153T/V163A/N164H/H231L.
  • green fluorescence proteins include those having a functional engineered fluorescent protein that includes H148G or H148Q substitutions in the Aequora green fluorescent protein. These proteins include the YFP H148G (SEQ ID NO:______) and YFP H148Q (SEQ ID NO: ______) proteins described above.
  • the sample in which pH is to be measured can be a biological sample, e.g., a biological tissue such as an extracellular matrix, blood or lymphatic tissue, or a cell.
  • the method is particularly suitable for measuring pH in a specific region of the cell, e.g., the cytosol, or an organellar space such as the inner mitochondrial matrix, the lumen of the Golgi, cytosol, the endoplasmic reticulum, the chloroplast lumen, the lumen of lysosome, or the lumen of an endosome.
  • the first fluorescent protein moiety is linked to a targeting sequence that directs the fluorescent protein to a desired cellular compartment.
  • targeting sequences include the amino terminal 81 amino acids of human type II membrane-anchored protein galactosyltransferase (SEQ ID NO:______) for directing the fluorescent indicator protein to the Golgi and the amino terminal 12 amino acids of the presequence of subunit IV of cytochrome c oxidase (SEQ ID NO:_______) for directing a fluorescent pH indicator protein to the mitochondrial matrix.
  • the 12 amino acids of the presequence of subunit IV of cytochrome c oxidase may be linked to the pH fluorescent indicator protein through a linker sequence, e.g., Arg-Ser-Gly-Ile (SEQ ID NO:______).
  • the invention provides a method of determining the pH of a region of a cell by introducing into the cell a polynucleotide encoding a polypeptide including an indicator having a first fluorescent protein moiety whose emission intensity changes as pH varies between 5 and 10, culturing the cell under conditions that permit expression of the polynucleotide; exciting the indicator; and determining the intensity of the light emitted by the first protein moiety at a first wavelength.
  • the emission intensity of the first fluorescent protein moiety indicates the pH of the region of the cell in which the indicator is present.
  • the polynucleotide can be introduced using methods described above.
  • the method can be used to measure intracellular pH in cells cultured in vitro, e.g., HeLa cells, or alternatively in vivo, e.g., in cells of an animal carrying a transgene encoding a pH-dependent fluorescent indicator protein.
  • Fluorescence in the sample can be measured using a fluorometer.
  • excitation radiation from an excitation source having a first wavelength, passes through excitation optics.
  • the excitation optics cause the excitation radiation to excite the sample.
  • fluorescent proteins in the sample emit radiation which has a wavelength that is different from the excitation wavelength.
  • Collection optics then collect the emission from the sample.
  • the device can include a temperature controller to maintain the sample at a specific temperature while it is being scanned.
  • a multi-axis translation stage can be used to move a microtiter plate holding a plurality of samples in order to position different wells to be exposed.
  • the multi-axis translation stage, temperature controller, auto-focusing feature, and electronics associated with imaging and data collection can be managed by an appropriately programmed digital computer.
  • the computer also can transform the data collected during the assay into another format for presentation.
  • the pH can be analyzed on cells in vivo, or from samples derived from cells transfected with polynucleotides or proteins expressing the pH indicator proteins. Because fluorescent pH indicator proteins can be expressed recombinantly inside a cell, the pH in an intracellular region, e.g., an organelle, or an extracellular region of an organism can be determined simply by determining changes in fluorescence.
  • Fluorescent protein pH sensors may vary in their respective pK a , and the differences in pK a can be used to select the most suitable fluorescent protein sensor most suitable for a particular application.
  • a sensor protein should be used whose pK a is close to the pH of the sample to be measured.
  • the pK a is within 1.5 pH unit of the sample. More preferably the pK a is within 1 pH unit, and still more preferably the pK a is within 0.5 pH unit of the sample.
  • a fluorescent protein pH sensor having a pK a of about 7.1 is preferred for determining the pH of cytosolic, Golgi, and mitochondrial matrix pH areas of a cell.
  • the YFP-H148G, YFP-H148Q, EYFP-H148G and EYFP-H148Q mutants are well-suited for measuring the pH of alkaline environments, e.g., mitochondrial matrix, as they have a pKa of 7.5 and 8.0, respectively.
  • a fluorescence sensor protein having a lower pK a e.g., a pK a of about 6.1, is preferred.
  • the fluorescence of a fluorescent protein pH sensor can be compared to the fluorescence of a second sensor, e.g., a second fluorescent protein pH sensor, that is also present in the measured sample.
  • the second fluorescent protein pH sensor should have an emission spectra distinct from the first fluorescent protein pH sensor so that the emission spectra of the two sensors can be distinguished. Because experimental conditions such as focusing and cell movement will affect fluorescence of the second sensor as well as the first sensor, comparing the relative fluorescence of, the two sensors allows for the normalization of fluorescence.
  • a convenient method of comparing the samples is to compute the ratio of the fluorescence of the first fluorescent protein pH sensor to that of the second fluorescent protein pH sensor.
  • kits may comprise a carrier means being compartmentalized to receive one or more container means such as vials, tubes, and the like, each of the container means comprising one of the separate elements to be used in the method.
  • container means such as vials, tubes, and the like
  • each of the container means comprising one of the separate elements to be used in the method.
  • one of the container means may comprise a polynucleotide encoding a fluorescent protein pH sensor.
  • a second container may further comprise fluorescent protein pH sensor.
  • the constituents may be present in liquid or lyophilized form, as desired.
  • Fluorescent protein pH sensors were constructed by engineering site-specific mutations in polynucleotides encoding forms of the Aequora victoria green fluorescent protein (GFP).
  • the starting GFP variant was the polynucleotide encoding the GFP variant EGFP (for enhanced green fluorescent protein).
  • the EGFP variant had the amino acid substitutions F64L/S65T/H231L relative to the wild-type Aequora victoria GFP sequence.
  • the ECFP (enhanced cyan fluorescent protein) mutant was constructed by altering the EGFP polynucleotide sequence so that it encoded a protein having the amino acid substitutions K26R/F64L/S65T/Y66W/N146I/M153T/V163A/N164H/H231L relative to the wild-type GFP amino acid sequence.
  • a second variant, named EYFP (enhanced yellow fluorescent protein) was constructed by altering the EGFP polynucleotide to encode a protein having the amino acid substitutions S65G/S72A/T203Y/H231L relative to the amino acid sequence of GFP.
  • EYFP-V68L-Q69K was constructed by altering the EGFP polynucleotide to encode a protein having the amino acid substitutions S65G/V68L/Q69K/S72A/T203Y relative to the amino acid sequence of GFP.
  • a HindIII site and Kozak consensus sequence (GCCACCATG) was introduced at the 5′ end of the polynucleotide encoding the GFP variants, and an EcoR1 site was added at the 3′ end of the gene of each indicator, and the fragments were ultimately ligated into the HindIII/EcoR1 sites of the mammalian expression vector pcDNA3 (Invitrogen).
  • EGFP and EYFP mutant proteins with no targeting signals were used as indicators of pH in the cytosol or nucleus.
  • polynucleotides encoding the 81 N-terminal amino acids of the type II membrane-anchored protein galactosyltransferase (GT:UDP-galactose- ⁇ ,1,4-galactosyltransferase. EC 2.4.1.22) ligated to polynucleotides encoding EGFP, ECFP, or EYFP.
  • the polynucleotides encoding the resulting proteins were named GT-EGFP, GT-ECFP, and GT-EYFP, respectively.
  • Mitochondrial matrix fluorescent protein pH sensors were constructed by attaching polynucleotides encoding 12 amino acids at the amino terminus of the presequence of subunit IV of cytochrome c oxidase (Hurt et al, EMBO J. 4:2061-68 (1985) to a polynucleotide encoding the amino acid sequence Arg-Sea-Gly-Ile, which in turn was ligated to polynucleotides encoding ECFP or EYFP. These constructs were labeled ECFP-mito or EYFP-mito.
  • the solutions for cuvette titration contained 125 mM KCl, 20 mM NaCl, 0.5 mM CaCl 2 , 0.5 mM MgCl 2 , and 25 mM of one of the following buffers—acetate, Mes, Mops, Hepes, bicine, and Tris.
  • EYFP showed an acidification-dependent decrease in the absorbance peak at 514 nm and a concomitant increase in absorbance at 390 nm (FIG. 2A).
  • the fluorescence emission (527-nm peak) and excitation spectra decreased with decreasing pH, but the fluorescence excitation spectrum showed no compensating increase at 390 nm. Therefore, the species absorbing at 390 nm was nonfluorescent.
  • the apparent pKa (pK′a) of EYFP was 7.1 with a Hill coefficient (n) of 1.1 (FIG. 2B)
  • EGFP fluorescence also was quenched with decreasing pH.
  • the pK′a of EGFP was 6.15, and n was 0.7.
  • HeLa cells and AT-20 cells grown on glass coverslips were transiently lipo-transfected (LipofectinTM, GIBCO) with polynucleotide constructs encoding EYFP.
  • Cells were imaged between 2 and 4 days after transfection at 22° C. with a cooled charge-coupled device camera (Photometrices, Arlington, Ariz.) as described in Miyawaki et al., Nature 388:882, (1997).
  • the interference filters (Omega Optical and Chroma Technology, Brattleboro, Vt.) used for excitation and emission were 440 ⁇ 10 and 480 ⁇ 15 for ECFP; 480 ⁇ 15 and 535 ⁇ 22.5 for EGFP or EYFP.
  • the dichroic mirrors were 455 DCLP for ECFP and 505 DCLP for EGFP or EYFP. Regions of interest were selected manually, and pixel intensities were spatially averaged after background subtraction.
  • GT:UDP-galactose- ⁇ ,1,4-galactosyltransferase. EC 2.4.1.22 The type II membrane-anchored protein galactosyltransferase (GT:UDP-galactose- ⁇ ,1,4-galactosyltransferase. EC 2.4.1.22) has been used as a marker of the trans cisternae of the Golgi apparatus (Roth et al., J. Cell Biol. 93:223-29, (1982)). Accordingly, polynucleotide constructs encoding portions of the GT protein fused to the mutant GFP proteins were constructed as described in Example 1 in order to use the GT sequence to target the fluorescent protein pH sensor to the endoplasmic reticulum.
  • the pH of the Golgi lumen was measured by transfecting HeLa or AT-20 cells with the constructs GT-ECFP, GT-EGFP, or GT-EYFP. Bright juxtanuclear fluorescence was observed, with little increase in diffuse staining above autofluorescnce in most cells.
  • the fluorescence pattern was examined further in double-labeling experiments using rabbit polyclonal ⁇ -mannosidase II ( ⁇ -manII) antibody. Double labeling fluorescence was performed as described by McCaffery et al., Methods Enzymol. 257:259-279 (1995). The ⁇ -manII antibody was prepared as described in Velasco et al., J. Cell Biol. 122:39-51 (1993). In the double-staining experiments, it was observed that labeling of the medial trans-Golgi marker ⁇ -manII overlapped with GT-EYFP fluorescence.
  • ⁇ -manII was also fused with ECFP, and the pattern of fluorescence obtained upon transfection of the gene was indistinguishable from that of GT-EYFP by light microcopy.
  • GT-EYFP was found in the medial and trans Golgi, although endogenous GT is present in trans Golgi membranes. The difference in localization may occur as a result of overexpression of the GT-EYFP protein.
  • V-ATPase electrogenic ATP-dependent H + pump
  • the V-ATPase generates a ⁇ pH (acidic inside) and ⁇ (positive inside), which opposes further H + transport.
  • the movement of counter-ions, Cl ⁇ in (or K + out), with H + uptake would shunt the ⁇ , allowing a larger ⁇ pH to be generated.
  • bafilomycin A1 has been shown to be a potent inhibitor of vacuolar type H + ATPases (V type).
  • V type vacuolar type H + ATPases
  • bafilomycin A1 0.2 ⁇ M
  • FIG. 3A demonstrates that the fluorescence of ECFP changed less than that of EYFP during the course of the experiment. Although the ratio of EYFP to ECFP emission varied between cells, probably reflecting a different concentration of GT-EYFP and GT-ECFP in the Golgi lumen, it changed with pH as expected (FIG. 3B). Bafilomcin A1 raised the GT-EYFP/GT-ECFP emission ratio, i.e, it raised pH G .
  • the YFP H148G mutant was prepared using as a template a nucleic acid encoding the YFP mutation 10c, which includes the mutations S65G/V68L/S72A/Q80R/T203Y and is described in Ormö et al., Science 273:1392-95 (1997).
  • the YFP H148G mutant was constructed using the PCR-based QUIKCHANGETM Site-Directed Mutagenesis Kit (Stratagene, La Jolla, Calif.) following the manufacturer's instructions.
  • the YFP H148Q mutation was similarly constructed from a nucleic acid encoding the 10C mutation.
  • the pKa of the YFP H148G mutant was found to be 8.0, while the YFP H148Q mutant was found to have a pKa of 7.5.
  • the high pKa of the mutant YFP H148G allows it the to be used for the precise measurement of mitochondrial matrix pH both in cells at rest and in cells subject to manipulations that decrease mitochondrial pH.
  • FIG. 4 shows that HeLa cells transfected with YFP H148G in the mitochondrial matrix were fluorescent at an initial pH of 8.0 to 8.1 (where measurements began at t ⁇ 0 seconds). 5 ⁇ M CCCp was added at about t ⁇ 300 seconds. Although addition of 5 ⁇ m CCCP rapidly lowered the pH to 7.0, fluorescence of mito-YFP H148G was still detectable. Then a calibration was performed by perfusing the cells with extracellular medium of ph 7, 7.5, 8, and 8.35 containing the ionophores nigericin plus monensin to equilibrate mitochondrial pH and extracellular pH. Fluorescence in mitochondria increased stepwise with each change of extracellular pH.
  • FIG. 5 demonstrates that fluorescence was detectable in the mitochondrial matrix of chicken skeletal myotubes, which had a pH of 8.0-8.1 (t ⁇ 0). Fluorescence was still detectable following addition of 25 ⁇ M forskolin, which did not affect the pH, and after addition of 2 ⁇ M CCCP at t ⁇ 750 seconds, although CCCP caused the pH to rapidly drop to 6.9 at t ⁇ 1400 seconds. Thereafter fluorescence continued to be observed during calibration at ph 6.9, 7.6 and 8.0.
  • the YFP H148Q mutant has a pKa of about 7.4, which is intermediate between the pKa of EYFP and YFP mutant H148G.
  • YFP H148Q can also be used to measure mitochondrial matrix pH
  • a nucleic acid encoding mito-YFP-H148Q was transfected into HeLa cells. Fluorescence was measured over time (beginning at t ⁇ 0), including following the addition of 10 ⁇ M nigericin in high KCL titration buffer at t ⁇ 500 seconds.
  • the GFP mutant T203I was expressed in the mitochondria of HeLa cells.
  • the GFP T203I mutant can be excited with light of 400 nm, which does not appreciably excite the pH sensitive YFP mutants.
  • FIG. 7 shows the change of mitochondrial pH with oligomycin and the uncoupler CCCP as the ratio of YFP H148G emission and GFP T203I emission, with excitation of 490 and 400 nm, respectively.
  • the green fluorescent protein (GFP) from the jellyfish Aequorea victoria has been used extensively in molecular biology as a fluorescent label.
  • the structures of WT GFP (Yang et al., Nature Biotech. 14:1246-51, 1996; Brejc et al., Proc. Natl. Acad. Sci. USA. 94:2306-11, 1997) and the variant S65T were determined in 1996 (Ormö et al., Science 273:1392-95, 1996).
  • WT GFP exhibits two absorption maxima, where the major band absorbs at 398 nm and the minor band at 475 nm (Morise et al., Biochemistry 13:2656-62, 1974). Excitation of either of these bands leads to emission of green light with a maximum between 504 and 508 nm (FIG. 8).
  • GFP variants with altered spectral characteristics were identified by random mutagenesis. Some of these mutants, such as Y66H and Y66W (Tsien, Ann. Rev. Biochem. 67:509, 1998, Heim et al., Proc. Natl. Acad. Sci. (USA) 91:12501-04) result in blue-shifted absorbance and emission maxima.
  • S65T Heim et al., Nature 373: 663-64, 1995
  • T203I Heim et al., Proc. Natl. Acad. Sci. (USA) 91:12501-04, 1994.
  • S65T exhibits only one major absorption band at 489 nm, red-shifted by 14 nm from WT GFP, and is almost six times brighter (Heim et al., Nature 373: 663-64, 1995).
  • the emission spectrum is shifted by only 3 nm to 511 nm, and so cannot easily be distinguished from the wild-type emission.
  • Random mutagenesis techniques produced only one further red-shifted variant, S65T/M153A/K238E, which increases the excitation and emission wavelengths of S65T by 15 and 3 nm respectively (Heim et al., Curr. Biol. 6: 178-82, 1996).
  • crystal structures of the first set of GFP variants rationally designed based on the x-ray structure of GFP S65T (Ormö et al., Science 273:1392-95, 1996).
  • These variants termed YFPs (Yellow Fluorescent Proteins) exhibit the longest wavelength emissions of all GFPs generated by mutagenesis (FIG. 8).
  • the YFPs fluoresce around 528 nm, red-shifted by 16 nm as compared to S65T and are easily distinguishable from S65T on a fluorescence microscope.
  • the specific YFP investigated is the quadruple-mutant T203Y/S65G/V68L/S72A, where the substitution T203Y was introduced based on the structural considerations detailed below and is believed responsible for the red-shift.
  • the other three mutations have been shown to improve its brightness in live cells (Cormack et al., Gene 173:33, 1996).
  • the T203Y mutation would have been difficult to identify by random mutagenesis since this amino acid substitution requires three substitutions at the nucleotide level.
  • Thr203 is positioned close to the chromophore, it was postulated that its replacement with a tyrosine would result in ⁇ -stacking interactions between the chromophore and the highly polarizable phenol (Ormö et al., Science 273:1392-95, 1996), leading to red-shifted spectral properties.
  • the structure of S65T suggested that an aromatic amino acid introduced in position 203 would extend into the water-filled cavity adjacent to the chromophore (Ormö et al., Science 273:1392-95, 1996).
  • GFP variants were prepared as described in Example 6, above. This template incorporates the mutations T203Y/S65G/V68L/S72A, as well as the ubiquitous Q80R substitution that was accidentally introduced into the gfp cDNA early on (Ormö et al., Science 273:1392-95, 1996; Chalfie et al., Science 263:802-05, 1994). All GFP variants were expressed and purified as described (Ormö et al., Science 273:1392-95, 1996).
  • YFP H148G was concentrated to 12 mg/ml in 20 mM HEPES pH 7.9.
  • Rod-shaped crystals with approximate dimensions of 1.8 ⁇ 0.08 ⁇ 0.04 mm were grown in hanging drops containing 2 ⁇ l protein and 2 ⁇ l mother liquor at 4° C. within four days.
  • the mother liquor contained 16% PEG 4000, 50 mM sodium acetate pH 4.6, and 50 mM ammonium acetate.
  • X-ray diffraction data were collected from a single crystal at room temperature using a Xuong-Hamlin area detector (Hamlin, Methods. Enzymol. 114:416-52, 1985).
  • a model for the anionic chromophore was obtained by semi-empirical molecular orbital calculations using AM1 in the program SPARTAN version 4.1 (Wavefunction Inc., Irvine, Calif.).
  • the minimized structure which was planar, compared very favorably with a related small molecule crystallographic structure (Tinant et al., Cryst. Struct. Comm. 9:671-74, 1980), and also with the model used during refinement of GFP S65T, where a simpler modeling program had been employed (Ormö et al., Science 273:1392-95, 1996).
  • the B-factors of all chromophore atoms were set to the values obtained in the 1.9 ⁇ structure of GFP S65T (Ormö et al., Science 273:1392-95, 1996), and then refined as a group, with identical shifts for the grouped atoms.
  • l protein and 5
  • X-ray diffraction data were collected on two isomorphic crystals at room temperature using an Raxis-IV imaging plate mounted on a Rigaku RUH3 rotating anode generator equipped with mirrors.
  • Chromophore and Tyr203 densities were very clear, and both were planar.
  • the model was edited to include these groups in refinement, and solvent molecules were added where appropriate.
  • B-factors were refined using a strong correlation between neighboring atoms due to the relatively low resolution.
  • the GFP S65T structure was used as a search model for molecular replacement against a 3.0 ⁇ dataset using the program AMoRe (Navaza, Acta Crystallogr. A50:157-63, 1994), and the structure was refined. Later, the refined structure of YFP H148G (see below) was used for phasing and refinement against a 2.5 ⁇ dataset.
  • NCS Non-crystallographic symmetry
  • the refined YFP structure clearly shows that the overall fold is undisturbed, with an rms deviation from the GFP S65T structure of 0.36 ⁇ for ⁇ -carbons.
  • Three larger contact areas with adjacent molecules were identified. The largest of these covers about 722 ⁇ 2 of one monomer surface, includes a series of hydrophobic residues consisting of Ala206, Phe223, and Leu221, and also a number of hydrophilic contacts.
  • This interface is essentially identical to the dimer interface for WT GFP described by Yang et al., Nature Biotech. 14:1246-51, 1996. High salt conditions during crystallization experiments appear to favor dimerization, as has been suggested previously (Palm et al., Nat. Struct. Biol. 4:361-65, 1997).
  • YFP H148G crystallized as a monomer in the presence of polyethylene glycol and acetate at pH 4.6 in spacegroup P2 1 2 1 2 1 , isomorphous to S65T (Ormö et al., Science 273:1392-95, 1996) and the blue-emission variant BFP (Wachter et al., Biochemistry 36:9759-65, 1997).
  • S65T Ormö et al., Science 273:1392-95, 1996)
  • BFP blue-emission variant BFP
  • the rms deviation between YFP H148G and S65T a-carbons is 0.31 ⁇ , and the deviation between YFP H148G and YFP a-carbons is 0.35 ⁇ .
  • the b-strands of the two YFP variants overlay closely in all areas except around the C ⁇ of residue 148 where a movement of 1.1 ⁇ is observed. This movement has not been observed in other pH 4.6 structures grown under similar conditions and crystallizing in the same space group, such as the BFP structure (Wachter et al., Biochemistry 36:9759-65, 1997). Residue 148 and adjacent residues are not involved in crystal contacts, further indicating that the observed movement is due to the H148G substitution, not crystallization conditions.
  • Electron densities of the chromophore and the phenol ring of Tyr203 appeared to be completely planar before the atoms for these groups were added to the model.
  • the tyrosine side chain was first introduced into the model, it was modelled as co-planar to the chromophore.
  • FIG. 9 shows the electron density of the refined YFP chromophore structure together with the phenol ring of Tyr203.
  • the distance of the closest approach between atoms of the two interacting rings is 3.3 to 3.4 ⁇ , and occurs at that edge of the chromophore plane that is opposite the exo-methylene bond (FIG. 9). It appears that the phenol tilts towards this area of the chromophore since it is more open, with fewer atoms to clash with sterically.
  • the distance of largest separation between the rings is 3.5 to 3.8 ⁇ , and occurs at the opposite edge, where steric clash with the exo-methylene carbon could occur.
  • This range of plane-to-plane distances is typical for face-to-face ⁇ to ⁇ stacking interactions found in proteins, and consistent with interaction energy calculations that show a potential energy minimum for two horizontally stacked benzene molecules with a vertical separation of 3.3 ⁇ (Burley et al., J. Am. Chem. Soc. 108, 7995-8001, 1986).
  • Fluorescence measurements were carried out on a Hitachi F4500 fluorescence spectrophotometer at a constant protein concentration of approximately 0.01 mg/ml, with buffer conditions identical to those of absorbance measurements.
  • the excitation wavelength was set to the absorbance maximum of the long-wave band of the particular mutant.
  • the emission was scanned between 500 and 600 nm, and peak emission intensity was plotted as a function of pH and curve-fitted.
  • the YFPs have two absorbance maxima whose relative ratio is pH-dependent (FIG. 8 and Table 34).
  • the UV absorption peaks at 392 (YFP) or 397 nm (YFP H148G) have been ascribed to the neutral chromophore, whereas the visible absorption peaks at 514 (YFP) or 512 nm (YFP H148G) have been ascribed to the anionic chromophore (Niwa et al., Proc. Natl. Acad. Sci. (USA) 93: 13617-22, 1996).
  • the lower energy peak exhibits clear vibrational structure as indicated by the pronounced shoulder at 480-490 nm, and its mirror-image relationship with the emission band is striking (FIG. 8).
  • These features are consistent with luminescence properties of large and rigid systems in condensed phases (Barltrop et al., Principles of Photochemistry, John Wiley and Sons, New York, 1978, pp. 51-52 and 78-79), and may be more pronounced in the YFPs due to decreased chromophore flexibility in the presence of the stacked phenol. Both YFPs fluoresce intensely when excited at the longer-wavelength band, with maximum emission occurring at 528 nm (FIG. 8).
  • the chromophore pKa in the intact protein was determined to be 7.00( ⁇ 0.03) for YFP and 8.02 ( ⁇ 0.01) for YFP H148G by absorbance measurements at varying pH.
  • the pKa values determined by fluorescence were 6.95 ( ⁇ 0.03) and 7.93 ( ⁇ 0.04), respectively, for the two variants.
  • the YFP pKa is remarkably similar to that of TABLE 34 Summary of Absorption and Emission Maxima.
  • EYFP (S65G/S72A/T203Y/H231L). All titration curves gave an excellent fit to a single pKa value.
  • the Glu222 carboxy oxygen approaches the chromophore imidazolinone ring nitrogen to within 3.0 (YFP) and 3.3 ⁇ (YFP H148G), considerably closer than in WT and S65T (4.3 and 4.0 ⁇ , respectively).
  • This close approach appears to be related to the chromophore positional shift described above.
  • Distance and geometry for hydrogen bonding between Glu222 and the chromophore ring nitrogen are excellent in YFP, and somewhat less optimal in YFP H148G, where the presumed H-bond makes roughly a 45° angle with the chromophore plane.
  • the YFP structure is the first GFP structure solved that suggests H-bonding interactions of the heterocyclic ring nitrogen originating from Tyr66. The most likely interpretation in terms of charge states is a deprotonated ring nitrogen and a protonated Glu222, rendering both groups neutral, however, it is clear that they share a proton.
  • the mutation H148G was introduced into YFP to examine the effects of solvent accessibility on the fluorescent properties and the ionization constant of the chromophore.
  • the ⁇ -barrel is somewhat perturbed around the phenolic end of the chromophore.
  • the ⁇ -strand that covers the chromophore in that area bulges out around His148, so that the backbone from residue 144 to 150 is not directly hydrogen-bonded to the adjacent backbone between residues 165 and 170. Rather, they are laced together by forming H-bonds with the imidazole ring of His148 (Arg168 backbone N to His148 N ⁇ 2 in S65T and WT GFP) and several water molecules.
  • the phenolic end of the chromophore is located directly “behind” the ring of His148. It was anticipated that substitution of His with Gly would open up a solvent channel to the chromophore in the absence of other structural perturbances, or perhaps to permit the bulge to close.
  • YFP H148G contains two larger interior cavities that are in contact with the chromophore cavity and filled with some ordered waters.
  • the cavity that was largest in S65T has decreased in size from approximately 127 ⁇ 3 (S65T) to 88 ⁇ 3 (YFP H148G), because some of the space is now filled with the phenol of Tyr203. In YFP, this cavity is not accessible to a 1.4 ⁇ probe at all since several groups have moved into this space.
  • the more significant structural adjustments are C ⁇ 2 of Val224, which has moved by 1.4 ⁇ , and C ⁇ 1 of Leu42 which has moved by 2.0 ⁇ , essentially filling the cavity.
  • the second larger cavity in contact with the chromophore is nearly invariant for S65T, YFP, and YFP H148G, and is between 98 and 103 ⁇ 3 in size.
  • YFP H148G was found to be highly fluorescent, with bright greenish-yellow color under ordinary day light.
  • the light-emitting properties of the fluorophore do not appear to be changed to any extent by the introduction of a solvent channel to the chromophore, indicating that significant quenching does not occur.
  • the H148G substitution may be especially useful for allowing access of various small-molecule species to the chromophore.
  • This substitution may be introduced into other GFP variants with a larger cavity adjacent to the chromophore, such as S65T [7] or S65G, allowing for analyte binding studies where specific spectral shifts due to the interaction with small molecules or ions of interest could be monitored.
  • the highest ionization constant of all variants examined to date is found for the YFP H148G mutant with a pKa of 8.0. In this mutant, the chromophore is solvent exposed, consistent with a similarly high pKa when the protein is denatured (Nageswara et al., Biophys. J. 32:630-32, 1980).

Abstract

Disclosed are fluorescent protein sensors for measuring the pH of a sample, nucleic acids encoding them, and methods of use. The preferred fluorescent protein sensors are variants of the green fluorescent protein (GFP) from Aequora victoria. Also disclosed are compositions and methods for measuring the pH of a specific region of a cell, such as the mitochondrial matrix or the Golgi lumen.

Description

  • This application is a continuation-in-part of currently pending U.S. Ser. No. 09/094,359 (herein incorporated by reference), which was filed Jun. 9, 1998.[0001]
  • STATEMENT AS TO FEDERALLY SPONSORED RESEARCH
  • [0002] This invention was made with Government support under Grant No. NS27177, awarded by the National Institutes of Health. The Government has certain rights in this invention.
  • FIELD OF THE INVENTION
  • The invention relates generally to compositions and methods for measuring the pH of a sample and more particularly to fluorescent protein sensors for measuring the pH of a biological sample. [0003]
  • BACKGROUND OF THE INVENTION
  • The pH within various cellular compartments is regulated to provide for the optimal activity of many cellular processes. In the secretory pathway, posttranslational processing of secretory proteins, the cleavage of prohormones, and the retrieval of escaped luminal endoplasmic reticulum proteins are all pH-dependent. [0004]
  • Several techniques have been described for measuring intracellular pH. Commonly used synthetic pH indicators can be localized to the cytosol and nucleus, but not selectively in organelles other than those in the endocytotic pathway. In addition, some cells are resistant to loading with cell-permeant dyes because of physical barriers such as the cell wall in bacteria, yeast, and plants, or the thickness of a tissue preparation such as brain slices. [0005]
  • Several methods have been described for measuring pH in specific regions of the cell. One technique uses microinjection of fluorescent indicators enclosed in liposomes. Once inside the cell, the liposomes fuse with vesicles in the trans-Golgi, and the pH of the intracellular compartments is determined by observing the fluorescence of the indicator. This procedure can be laborious, and the fluorescence of the indicator can be diminished due to leakage of the fluorescent indicator from the Golgi, or flux of the fluorescent indicator out of the Golgi as part of the secretory traffic in the Golgi pathway. In addition, the fusion of the liposomes and components of the Golgi must take place at 37° C.; however, this temperature facilitates leakage and flux of the fluorescent indicator from the Golgi. [0006]
  • A second method for measuring pH utilizes retrograde transport of fluorescein-labeled verotoxin 1B, which stains the entire Golgi complex en route to the endoplasmic reticulum. This method can be used, however, only in cells bearing the receptor globotriaosyl ceramide on the plasma membrane, and it may be limited by the residence time of the verotoxin in transit through the Golgi. [0007]
  • In a third method, intracellular pH has been measured using the chimeric protein CD25-TGN38, which cycles between the trans-Golgi network and the plasma membrane. At the plasma membrane, the CD25-motif binds extra-cellular anti-CD25 antibodies conjugated with a pH-sensitive fluorophore. Measurement of fluorescence upon return of the bound complex to the Golgi can be used to measure the pH of the organelle. [0008]
  • SUMMARY OF THE INVENTION
  • The invention is based on the discovery that proteins derived from the [0009] Aequora victoria green fluorescence protein (GFP) show reversible changes in fluorescence over physiological pH ranges.
  • Accordingly, in one aspect, the invention provides a method for determining the pH of a sample by contacting the sample with an indicator including a first fluorescent protein moiety whose emission intensity changes as the pH varies between 5 and 10, exciting the indicator, and the determining the intensity at a first wavelength. The emission intensity of the first fluorescent protein moiety indicates the pH of the sample. [0010]
  • In another aspect, the invention provides a method for determining the pH of a region of a cell by introducing into the cell a polynucleotide encoding a polypeptide including a first fluorescent protein moiety whose emission intensity changes as the pH varies between 5 and 10, culturing the cell under conditions that permit expression of the polynucleotide, and determining the intensity at a first wavelength. The emission intensity of the first fluorescent protein moiety indicates the pH of the sample. [0011]
  • In a further aspect, the invention provides a functional engineered fluorescent protein whose amino acid sequence is substantially identical to the amino acid sequence of the 238 amino acid [0012] Aequora victoria green fluorescence protein shown in FIG. 3 of U.S. Ser. No. 08/911,825 (SEQ ID NO:______), and whose emission intensity changes as pH varies between 5 and 10.
  • In another aspect, the invention provides a polynucleotide encoding the functional engineered fluorescent protein. [0013]
  • The invention also includes a kit useful for the detection of pH in a sample, e.g., a region of a cell. The kit includes a carrier means containing one or more containers comprising a first container containing a polynucleotide encoding a polypeptide including a first fluorescent protein moiety whose emission intensity changes as the pH varies between 5 and 10.[0014]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a schematic diagram depicting fluorescent protein sensors used as indicators of intracellular pH. [0015]
  • FIGS. 2A and 2B are graphs showing absorbance as a function of wavelength for the fluorescent protein pH sensor EYFP (SEQ ID NO:______) at various wavelengths (FIG. 2A), and the pH dependency of fluorescence of various GFP fluorescent protein sensors in vitro and in cells (FIG. 2B). The fluorescence intensity of purified recombinant GFP mutant protein (solid symbols) as a function of pH was measured in a microplate fluorometer. The fluorescence of the Golgi region of HeLa cells expressing proteins having the 81 N-terminal amino acids of the type II membrane-anchored protein galactosyltransferase (GT:UDP-galactose-β,1,4-galactosyltransferase. EC 2.4.1.22) (“GT”) fused to EYFP, or EGFP, i.e., GT-EYFP or GT-EGFP (open symbols) was determined during pH titration with the ionophores monensin/nigericin in high KCL solutions. [0016]
  • FIGS. 3A and 3B are graphs showing ratiometric measurements of pH[0017] G by cotransfecting HeLa cells with polynucleotides encoding GT-ECFP and GT-EYFP. FIG. 3A is a graph showing single wavelength fluorescence intensities of GT-EYFP and GT-ECFP in the Golgi region of a HeLa cell. FIG. 3B is a graph showing the ratio of GT-EYFP/GT-ECFP fluorescence in the same cell as a function of time.
  • FIG. 4 is a graph showing the change in mitochondrial pH of HeLa cells expressing YFP H148G. [0018]
  • FIG. 5 is a graph showing the mitochondrial pH of chick skeletal myotubes expressing YFP H148G in the mitochondrial matrix. [0019]
  • FIG. 6 is a graph showing fluorescence and pH in HeLa cells expressing YFP H148Q targeted to the mitochondrial matrix. [0020]
  • FIG. 7 is a graph showing a ratiometric measurement of mitochondrial pH following expression of YFP H148G (pH sensitive) and GFP T203I (pH insensitive) in mitochondria of HeLa cells. [0021]
  • FIG. 8 is a graph showing normalized absorbance of WT GFP and YFP in 75 mM phosphate pH 8.0, 140 mM NACl. Solid lines, WT GFP; Dashed lines, YFP. [0022]
  • FIG. 9 is a stereoview of the 2F[0023] o-Fc electron density map of the YFP chromophore and the stacked Tyr203 after refinement. The 2.5 Å resolution map was contoured at +1 standard deviation.
  • DETAILED DESCRIPTION
  • The invention provides genes encoding fluorescent sensor proteins, or fragments thereof, whose fluorescence is sensitive to changes in pH at a range between 5 and 10. The proteins of the invention are useful for measuring the pH of a sample. The sample can be a biological sample and can include an intracellular region of a cell, such as the lumen of the mitochondria or golgi. The pH of a sample is determined by observing the fluorescence of the fluorescent sensor protein. [0024]
  • The fluorescent protein pH sensor have a broad applicability to cells and organisms that are amenable to gene transfer. Problems associated with the use of other agents used to measure pH, e.g., problems associated with permeabilizing cells to ester-containing agents, leakage of agents, or hydrolysis of agents are avoided. With the fluorescent protein pH sensors of the invention, no leakage occurs over the course of a typical measurement, even when the measurement is made at 37° C. [0025]
  • Compositions and methods described herein also avoid the need to express and purify large quantities of soluble recombinant protein, purify and label it in vitro, microinject it back into cells. An important advantage of the fluorescent protein pH sensors of the invention is that they can be delivered to cells in the form of polynucleotides encoding the protein sensor fused to a targeting signal or signals. The targeting signal directs the expression of the protein sensors to restricted cell locations. Thus, it is possible to measure the pH of a precisely defined cellular region or organelle. [0026]
  • Polynucleotides and Polypeptides [0027]
  • In a first aspect, the invention provides a functional engineered fluorescent protein whose amino acid sequence is substantially identical to the 238 amino acid [0028] Aequora victoria green fluorescence protein shown in FIG. 3 of U.S. Ser. No. 08/911,825 (SEQ ID NO:______). The term “fluorescent protein” refers to any protein capable of emitting light when excited with appropriate electromagnetic radiation, and which has an amino acid sequence that is either natural or engineered and is derived from the amino acid sequence of Aequorea-related fluorescent protein. The term “fluorescent protein pH sensor” refers to a fluorescent protein whose emitted light varies with changes in pH from 5 to 10.
  • The invention also includes functional polypeptide fragments of a fluorescent protein pH sensor. As used herein, the term “functional polypeptide fragment” refers to a polypeptide which possesses biological function or activity which is identified through a defined functional assay and which is associated with a particular biologic, morphologic, or phenotypic alteration in the cell. The term “functional fragments of a functional engineered fluorescent protein” refers to fragments of a functional engineered protein that retain a function of the engineered fluorescent protein, e.g., the ability to fluoresce in a pH-dependent manner over the [0029] pH range 5 to 10. Biologically functional fragments can vary in size from a polypeptide fragment as small as an epitope to a large polypeptide.
  • Minor modifications of the functional engineered fluorescent protein may result in proteins which have substantially equivalent activity as compared to the unmodified counterpart polypeptide as described herein. Such modifications may be deliberate, as by site-directed mutagenesis, or may be spontaneous. All of the polypeptides produced by these modifications are included herein as long as the pH-dependent fluorescence of the engineered protein still exists. [0030]
  • A functional engineered fluorescent protein includes amino acid sequences substantially the same as the sequence set forth in SEQ ID NO:______, and whose emission intensity changes as pH varies between 5 and 10. In some embodiments the emission intensity of the functional engineered fluorescent protein changes as pH varies between 5 and 8.5. [0031]
  • By “substantially identical” is meant a protein or polypeptide that retains the activity of a functional engineered protein, or nucleic acid encoding the same, and which exhibits at least 80%, preferably 85%, more preferably 90%, and most preferably 95% homology to a reference amino acid or nucleic acid sequence. For polypeptides, the length of comparison sequences will generally be at least 16 amino acids, preferably at least 20 amino acids, more preferably at least 25 amino acids, and most preferably 35 amino acids. For nucleic acids, the length of comparison sequences will generally be at least 50 nucleotides, preferably at least 60 nucleotides, more preferably at least 75 nucleotides, and most preferably 110 nucleotides. [0032]
  • By “substantially identical” is meant an amino acid sequence which differs only by conservative amino acid substitutions, for example, substitution of one amino acid for another of the same class (e.g., valine for glycine, arginine for lysine, etc.) or by one or more non-conservative substitutions, deletions, or insertions located at positions of the amino acid sequence which do not destroy the function of the protein (assayed, e.g., as described herein). Preferably, such a sequence is at least 85%, more preferably 90%, more preferably 95%, more preferably 98%, and most preferably 99% identical at the amino acid level to one of the sequences of EGFP (SEQ ID NO:______), EYFP (SEQ ID NO: ______), ECFP (SEQ ID NO: ______), EYFP-V68L/Q69K (SEQ ID NO:______), YFP H148G (SEQ ID NO:______), or YFP H148Q (SEQ ID NO:______). [0033]
  • Homology is typically measured using sequence analysis software (e.g., Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705). Such software matches similar sequences by assigning degrees of homology to various substitutions, deletions, substitutions, and other modifications. Conservative substitutions typically include substitutions within the following groups: glycine alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. [0034]
  • In some embodiments, the amino acid sequence of the protein includes one of the following sets of substitutions in the amino acid sequence of the Aequora green fluorescent protein (SEQ ID NO:______): F64L/S65T/H231L, referred to herein as EGFP (SEQ ID NO:______); S65G/S72A/T203Y/H231L, referred to herein as EYFP (SEQ ID NO:______); S65G/V68L/Q69K/S72A/T203Y/H231L, referred to herein as EYFP-V68L/Q69K (SEQ ID NO:______); K26R/F64L/S65T/Y66W/N146I/M153T/V163A/N164H/H231L, referred to herein as ECFP (SEQ ID NO:______). The amino acid sequences of EGFP, EYFP, ECFP, and EYFP-V68L/Q69K are shown in Tables 1-4, respectively. The amino acids are numbered with the amino acid following the initiating methionine assigned the ‘1’ position. Thus, F64L corresponds to a substitution of leucine for phenylalanine in the 64th amino acid following the initiating methionine. [0035]
    TABLE 1
    EGEP Amino Acid Sequence
    (SEQ ID NO:_)
    MVSKGEELFTGVVPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICT
    TGKLPVPWPTLVTTLTYGVQCFSRYPDHMKQHDFFKSAMPEGYVQERTIF
    FKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGHKLEYNYNSHN
    VYIMADKQKNGIKVNFKIRHNIEDGSVQLADHYOONTPIGDGPVLLPDNH
    YLSTOSALSKDPNEKRDHMVLLEFVTAAGITLGMDELYK
  • [0036]
    TABLE 2
    EYFP Amino Acid Sequence
    (SEQ ID NO:_)
    MVSKGEELFTGVVPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICT
    TGKLPVPWPTLVTTFGYGVQCFARYPDHMKQHDFFKSAMPEGYVQERTIF
    FKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGHKLEYNYNSHN
    VYIMADKQKNGIKVNFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNH
    YLSYQSALSKDPONEKRDHMVLLEFVTAAGITLGMDELYK
  • [0037]
    TABLE 3
    EYFP-V68L/Q69K Amino Acid Sequence
    (SEQ ID NO:_)
    MVSKGEELFTGVVPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICT
    TGKLPVPWPTLVTTFGYGLKCFARYPDHMKQHDFFKSAMPEGYVQERTIF
    FKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGHKLEYNYNSHN
    VYIMADKQKNGIKVNFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNH
    YLSYQSALSKDPNEKRDHMVLLEFVTAAGITLGMDELYK
  • [0038]
    TABLE 4
    ECFP Amino Acid sequence
    (SEQ ID NO:_)
    MVSKGEELFTGVVPILVELDGDVNGHRFSVSGEGEGDATYGKLTLKFICT
    TGKLPVPWPTLVTTLTWGVQCFSRYPDHMKQHDFFKSAMPEGYVQERTIF
    FKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGHKLEYNYISHN
    VYITADKQKNGIKAHFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNH
    YLSTQSALSKDPNEKRDHMVLLEFVTAAGITLGMDELYK
  • In other embodiments, the amino acid sequence of the protein is based on the sequence of the wild-type Aequora green fluorescent protein, but includes the substitution H148G (SEQ ID NO:______) or H148Q (SEQ ID NO:______). In specific embodiments, these substitutions can be present along with other substitutions, e.g., the proteins can include the substitutions S65G/V68L/S72A/H148G/Q80R/T203Y (SEQ ID NO:______), which is referred to herein as the “YFP H148G mutant,” S65G/V68L/S72A/H148Q/Q80R/T203Y, which is referred to herein as the “YFP H148Q mutant” (SEQ ID NO:______), the as well as EYFP-H148G (SEQ ID NO: ______) and EFP-H148Q (SEQ ID NO: ______). The amino acid sequences of these mutants are shown in Tables 5-8, respectively. [0039]
    TABLE 5
    Amino Acid Sequence of YFP H148G
    (SEQ ID NO:_)
    MSKGEELFTGVVPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICTT
    GKLPVPWPTLVTTFGYGLQCFARYPDHMKRHDFFKSAMPEGYVQERTIFF
    KDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGHKLEYNYNSGNV
    YIMADKQKNGIKVNFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNHY
    LSYQSALSKDPNEKRDHMVLLEFVTAAGITHGMDELYK
  • [0040]
    TABLE 6
    Amino Acid Sequence of YFP H148Q
    (SEQ ID NO:_)
    MSKGEELFTGVVPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICTT
    GKLPVPWPTLVTTFGYGLQCFARYPDHMKRHDFFKSAMPEGYVQERTIFF
    KDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGHKLEYNYNSQNV
    YIMADKQKNGIKVNFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNHY
    LSYQSALSKDPNEKRDHMVLLEFVTAAGITHGMDELYK
  • [0041]
    TABLE 7
    Amino Acid Sequence of EYFP-H148G
    (SEQ ID NO:_)
    MVSKGEELFTGVVPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICT
    TGKLPVPWPTLVTTFGYGVQCFARYPDHMKQHDFFKSAMPEGYVQERTIF
    FKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGHKLEYNYNSGN
    VYIMADKQKNGIKVNFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNH
    YLSYQSALSKDPNEKRDHMVLLEFVTAAGITLGMDELYK
  • [0042]
    TABLE 8
    Amino acid. Sequence of EYFP-H148Q
    (SEQ ID NO:_)
    MVSKGEELFTGVVPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICT
    TGKLPVPWPTLVTTFGYGVQCFARYPDHMKQHDFFKSAMPEGYVQERTIF
    FKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGHKLEYNYNSQN
    VYIMADKQKNGIKVNFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNH
    YLSYQSALSKDPNEKRDHMVLLEFVTAAGITLGMDELYK
  • In some embodiments, the protein or polypeptide is substantially purified. By “substantially pure protein or polypeptide” is meant an functional engineered fluorescent polypeptide which has been separated from components which naturally accompany it. Typically, the protein or polypeptide is substantially pure when it is at least 60%, by weight, free from the proteins and naturally-occurring organic molecules with which it is naturally associated. Preferably, the preparation is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight, of the protein. A substantially pure protein may be obtained, for example, by extraction from a natural source (e.g., a plant cell); by expression of a recombinant nucleic acid encoding a functional engineered fluorescent protein; or by chemically synthesizing the protein. Purity can be measured by any appropriate method, e.g., those described in column chromatography, polyacrylamide gel electrophoresis, or by HPLC analysis. [0043]
  • A protein or polypeptide is substantially free of naturally associated components when it is separated from those contaminants which accompany it in its natural state. Thus, a protein or polypeptide which is chemically synthesized or produced in a cellular system different from the cell from which it naturally originates will be substantially free from its naturally associated components. Accordingly, substantially pure polypeptides include those derived from eukaryotic organisms but synthesized in [0044] E. coli or other prokaryotes.
  • The invention also provides polynucleotides encoding the functional engineered fluorescent protein described herein. These polynucleotides include DNA, cDNA, and RNA sequences which encode functional engineered fluorescent proteins. It is understood that all polynucleotides encoding functional engineered fluorescent proteins are also included herein, as long as they encode a protein or polypeptide whose fluorescent emission intensity changes as pH varies between 5 and 10. Such polynucleotides include naturally occurring, synthetic, and intentionally manipulated polynucleotides. For example, the polynucleotide may be subjected to site-directed mutagenesis. The polynucleotides of the invention include sequences that are degenerate as a result of the genetic code. Therefore, all degenerate nucleotide sequences are included in the invention as long as the amino acid sequence of the functional engineered fluorescent protein or derivative is functionally unchanged. [0045]
  • Specifically disclosed herein is a polynucleotide sequence encoding a functional engineered fluorescent protein that includes one of the following sets of substitutions in the amino acid sequence of the Aequora green fluorescent protein (SEQ ID NO:______): S65G/S72A/T203Y/H231L, S65G/V68L/Q69K/S72A/T203Y/H231L, or K26R/F64L/S65T/Y66W/N146I/M153T/V163A/N164H/H231L. In specific embodiments, the DNA sequences encoding EGFP, EYFP, ECFP, EYFP-V68L/Q69K, YFP H148G, and YFP H148Q are those shown in Table 9-16 (SEQ ID NOs: ______ to ______), respectively. [0046]
  • The nucleic acid encoding functional engineered fluorescent proteins may be reflect the codon choice in the native [0047] A. victoria coding sequence, or, alternatively, may be chosen to reflect the optimal codon frequencies used in the organism in which the proteins will be expressed. Thus, nucleic acids encoding a target functional engineered protein to be expressed in a human cell may have use a codon choice that is optimized for mammals, or especially humans.
    TABLE 9
    EGFP Nucleic Acid Sequence
    ATGGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGTCGAGCTGGAC (SEQ ID NO:_)
    GGCGACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGGGCGAGGGCGATGCCACCTAC
    GGCAAGCTGACCCTGAAGTTCATCTGCACCACCGGCAAGCTGCCCGTGCCCTGGCCCACC
    CTCGTGACCACCCTGACCTACGGCGTGCAGTGCTTCAGCCCCTACCCCGACCACATGAAG
    CAGCACGACTTCTTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAGCGCACCATCTTC
    TTCAAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGGCGACACCCTG
    GTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGACGGCAACATCCTGGGGCAC
    AAGCTCGAGTACAACTACAACAGCCACAACGTCTATATCATGGCCGACAAGCAGAAGAAC
    GGCATCAAGGTGAACTTCAAGATCCGCCACAACATCGAGGACGGCAGCGTGCAGCTCGCC
    GACCACTACCAGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTGCCCGACAACCAC
    TACCTGAGCACCCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGCGATCACATGGTC
    CTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCATGGACGAGCTGTACAAGTAA
  • [0048]
    TABLE 10
    EYFP Nucleic Acid Sequence
    ATGGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGTCGAGCTGGAC (SEQ ID NO:_)
    GGCGACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGGGCGAGGGCGATGCCACCTAC
    GGCAAGCTGACCCTGAAGTTCATCTGCACCACCGGCAAGCTGCCCGTGCCCTGGCCCACC
    CTCGTGACCACCTTCGGCTACGGCGTGCAGTGCTTCGCCCGCTACCCCGACCACATGAAG
    CAGCACGACTTCTTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAGCGCACCATCTTC
    TTCAAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGGCGACACCCTG
    GTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGACGGCAACATCCTGGGGCAC
    AAGCTGGAGTACAACTACAACAGCCACAACGTCTATATCATGGCCGACAAGCAGAAGAAC
    GGCATCAAGGTGAACTTCAAGATCCGCCACAACATCGAGGACGGCAGCGTGCAGCTCGCC
    GACCACTACCAGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTGCCCGACAACCAC
    TACCTGAGCTACCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGCGATCACATGGTC
    CTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCATGGACGAGCTGTACAAGTAA
  • [0049]
    TABLE 11
    ECFP Nucleic Acid Sequence
    ATGGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGTCGAGCTGGAC (SEQ ID NO:_)
    GGCGACGTAAACGGCCACAGGTTCAGCGTGTCCGGCGAGGGCGAGGGCGATGCCACCTAC
    GGCAAGCTGACCCTGAAGTTCATCTGCACCACCGGCAAGCTGCCCGTGCCCTGGCCCACC
    CTCGTGACCACCCTGACCTGGGGCGTGCAGTGCTTCAGCCGCTACCCCGACCACATGAAG
    CAGCACGACTTCTTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAGCGTACCATCTTC
    TTCAAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGGCGACACCCTG
    GTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGACGGCAACATCCTGGGGCAC
    AAGCTGGAGTACAACTACATCAGCCACAACGTCTATATCACCGCCGACAAGCAGAAGAAC
    GGCATCAAGGCCCACTTCAAGATCCGCCACAACATCGAGGACGGCAGCGTGCAGCTCGCC
    GACCACTACCAGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTGCCCGACAACCAC
    TACCTGAGCACCCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGCGATCACATGGTC
    CTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCATGGACGAGCTGTACAAGTAA
  • [0050]
    TABLE 12
    EYFP-V68L/Q69K Nucleic Acid Sequence
    ATGGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGTCGAGCTGGAC (SEQ ID NO:_)
    GGCGACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGGGCGAGGGCGATGCCACCTAC
    GGCAAGCTGACCCTGAAGTTCATCTGCACCACCGGCAAGCTGCCCGTGCCCTGGCCCACC
    CTCGTGACCACCTTCGGCTACGGCCTGAAGTGCTTCGCCCGCTACCCCGACCACATGAAG
    CAGCACGACTTCTTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAGCGCACCATCTTC
    TTCAAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGGCGACACCCTG
    GTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGACGGCAACATCCTGGGGCAC
    GGCATCAAGGTGAACTTCAAGATCCGCCACAACATCGAGGACGGCAGCGTGCAGCTCGCC
    GACCACTACCAGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTGCCCGACAACCAC
    TACCTGAGCTACCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGCGATCACATGGTC
    CTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCATGGACGAGCTGTACAAGTAA
  • [0051]
    TABLE 13
    Nucleotide Sequence of the YFP H148G Coding Region
    ATGAGTAAAGGAGAAGAACTTTTCACTGGAGTTGTCCCAATTCTTGTTGAATTAGATGGT (SEQ ID NO:_)
    GATGTTAATGGGCACAAATTTTCTGTCAGTGGAGAGGGTGAAGGTGATGCAACATACGGA
    AAACTTACCCTTAAATTTATTTGCACTACTGGAAAACTACCTGTTCCATGGCCAACACTT
    GTCACTACTTTCGGTTATGGTCTTCAATGCTTTGCAAGATACCCAGATCATATGAAACGG
    CATGACTTTTTCAAGAGTGCCATGCCCGAAGGTTATGTTCAGGAAAGAACTATATTTTTC
    AAAGATGACGGGAACTACAAGACACGTGCTGAAGTCAAGTTTGAAGGTGATACCCTTGTT
    AATAGAATCGAGTTAAAAGGTATTGATTTTAAAGAAGATGGAAACATTCTTGGACACAAA
    TTGGAATACAACTATAACTCAGGCAATGTATACATCATGGCAGACAAACAAAAGAATGGA
    ATCAAAGTTAACTTCAAAATTAGACACAACATTGAAGATGGAAGCGTTCAACTAGCAGAC
    CATTATCAACAAAATACTCCAATTGGCGATGGCCCTGTCCTTTTACCAGACAACCATTAC
    CTGTCCTATCAATCTGCCCTTTCGAAAGATCCCAACGAAAAGAGAGACCACATGGTCCTT
    CTTGAGTTTGTAACAGCTGCTGGGATTACACATGGCATGGATGAACTATACAAA
  • [0052]
    TABLE 14
    Nucleotide Sequence of the YFP H148Q Coding Region+HZ,32
    ATGAGTAAAGGAGAAGAACTTTTCACTGGAGTTGTCCCAATTCTTGTTGA
    ATTAGATGGTGATGTTAATGGGCACAAATTTTCTGTCAGTGGAGAGGGTG
    AAGGTGATGCAACATACGGAAAACTTACCCTTAAATTTATTTGCACTACT
    GGAAAACTACCTGTTCCATGGCCAACACTTGTCACTACTTTCGGTTATGG
    TCTTCAATGCTTTGCAAGATACCCAGATCATATGAAACGGCATGACTTTT
    TCAAGAGTGCCATGCCCGAAGGTTATGTTCAGGAAAGAACTATATTTTTC
    AAAGATGACGGGAACTACAAGACACGTGCTGAAGTCAAGTTTGAAGGTGA
    TACCCTTGTTAATAGAATCGAGTTAAAAGGTATTGATTTTAAAGAAGATG
    GAAACATTCTTGGACACAAATTGGAATACAACTATAACTCAGGCAATGTA
    TACATCATGGCAGACAAACAAAAGAATGGAATCAAAGTTAACTTCAAAAT
    TAGACACAACATTGAAGATGGAAGCGTTCAACTAGCAGACCATTATCAAC
    AAAATACTCCAATTGGCGATGGCCCTGTCCTTTTACCAGACAACCATTAC
    CTGTCCTATCAATCTGCCCTTTCGAAAGATCCCAACGAAAAGAGAGACCA
    CATGGTCCTTCTTGAGTTTGTAACAGCTGCTGGGATTACACATGGCATGG
    ATGAACTATACAAA
  • [0053]
    TABLE 15
    Nucleotide Sequence of the EYFP-H148G
    Coding Region
    ATGGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGT
    CGAGCTGGACGGCGACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGG
    GCGAGGGCGATGCCACCTACGGCAAGCTGACCCTGAAGTTCATCTGCACC
    ACCGGCAAGCTGCCCGTGCCCTGGCCCACCCTCGTGACCACCTTCGGCTA
    CGGCGTGCAGTGCTTCGCCCGCTACCCCGACCACATGAAGCAGCACGACT
    TCTTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAGCGCACCATCTTC
    TTCAAGGACCACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGG
    CGACACCCTGGTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGG
    ACGGCAACATCCTGGGGCACAAGCTGGAGTACAACTACAACAGCGGCAAC
    GTCTATATCATGGCCGACAAGCAGAAGAACGGCATCAAGGTGAACTTCAA
    GATCCGCCACAACATCGAGGACGGCAGCGTGCAGCTCGCCGACCACTACC
    AGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTGCCCGACAACCAC
    TACCTGAGCTACCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGCGA
    TCACATGGTCCTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCA
    TGGACGAGCTGTACAAGTAA
  • [0054]
    TABLE 16
    Nucleotide Sequence of the EYFP-H148Q
    Coding Region
    ATGGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGT
    CGAGCTGGACGGCGACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGG
    GCGAGGGCGATGCCACCTACGGCAAGCTGACCCTGAAGTTCATCTGCACC
    ACCGGCAAGCTGCCCGTGCCCTGGCCCACCCTCGTGACCACCTTCGGCTA
    CGGCGTGCAGTGCTTCGCCCGCTACCCCGACCACATGAAGCAGCACGACT
    TCTTCAAGTCCGCCATGCCCCAAGGCTACGTCCAGGAGCGCACCATCTTC
    TTCAAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGG
    CGACACCCTGGTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGG
    ACGGCAACATCCTGGGGCACAAGCTGGAGTACAACTACAACAGCCAGAAC
    GTCTATATCATGGCCGACAAGCAGAAGAACGGCATCAAGGTGAACTTCAA
    GATCCGCCACAACATCGAGGACGGCAGCGTGCAGCTCGCCGACCACTACC
    AGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTGCCCGACAACCAC
    TACCTGAGCTACCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGCGA
    TCACATGGTCCTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCA
    TGGACGAGCTGTACAAGTAA
  • The term “polynucleotide” refers to a polymeric form of nucleotides of at least 10 bases in length. The nucleotides can be ribonucleotides, deoxyribonucleotides, or modified forms of either type of nucleotide. The term includes single and double stranded forms of DNA. By “isolated polynucleotide” is meant a polynucleotide that is not immediately contiguous with both of the coding sequences with which it is immediately contiguous (one on the 5′ end and one on the 3′ end) in the naturally occurring genome of the organism from which it is derived. The term therefore includes, for example, a recombinant DNA which is incorporated into a vector, e.g., an expression vector; into an autonomously replicating plasmid or virus; or into the genomic DNA of a prokaryote or eukaryote, or which exists as a separate molecule (e.g., a cDNA) independent of other sequences. [0055]
  • A “substantially identical”,nucleic acid sequence codes for a substantially identical amino acid sequence as defined above. [0056]
  • The functional engineered fluorescent protein can also include a targeting sequence to direct the fluorescent protein to particular cellular sites by fusion to appropriate organellar targeting signals or localized host proteins. A polynucleotide encoding a targeting sequence can be ligated to the 5′ terminus of a polynucleotide encoding the fluorescence such that the targeting peptide is located at the amino terminal end of the resulting fusion polynucleotide/polypeptide. The targeting sequence can be, e.g., a signal peptide. In the case of eukaryotes, the signal peptide is believed to function to transport the fusion polypeptide across the endoplasmic reticulum. The secretory protein is then transported through the Golgi apparatus, into secretory vesicles and into the extracellular space or, preferably, the external environment. Signal peptides which can be utilized according to the invention include pre-pro peptides which contain a proteolytic enzyme recognition site. Other signal peptides with similar properties to pro-calcitonin described herein are known to those skilled in the art, or can be readily ascertained without undue experimentation. [0057]
  • The targeting sequence can also be a nuclear localization sequence, an endoplasmic reticulum localization sequence, a peroxisome localization sequence, a mitochondrial localization sequence, or a localized protein. Targeting sequences can be targeting sequences which are described, for example, in “Protein Targeting”, chapter 35 of Stryer, L., Biochemistry (4th ed.). W. H. Freeman, 1995. The localization sequence can also be a localized protein. Some important targeting sequences include those targeting the nucleus (KKKRK) (SEQ ID NO: ______), mitochondrion (the 12 amino terminal amino acids of the cytochrome c oxidase subunit IV gene (SEQ ID NO:______), or the amino terminal sequence MLRTSSLFTRRVQPSLFRNILRLQST (SEQ ID NO:______), endoplasmic reticulum (KDEL (SEQ ID NO:______) at the C-terminus, assuming a signal sequence present at N-terminus), peroxisome (SKF at C-terminus), prenylation or insertion into plasma membrane (CaaX, CC, CXC, or CCXX at C-terminus), cytoplasmic side of plasma membrane (fusion to SNAP-25), or the Golgi apparatus (fusion to the amino terminal 81 amino acids of human type II membrane-anchored protein galactosyltransferase (SEQ ID NO:______), or fusion to furin). [0058]
  • Examples of targeting sequences linked to functional engineered fluorescent proteins include GT-EYFP (SEQ ID NO:______), GT-ECFP (SEQ ID NO:______), GT-EGFP (SEQ ID NO:______), and GT-EYFP-V68L/Q69K (SEQ ID NO:______), which are targeted to the Golgi apparatus using sequences from the GT protein; and EYFP-mito (SEQ ID NO:______) and EGFP-mito (SEQ ID NO:______), which are targeted to the mitochondrial matrix using sequences from the amino terminal region of the cytochrome c oxidase subunit IV gene. The EYFP, ECFP, EGFP, and EYFP-V68L/Q69K amino acid sequences, as well as nucleic acids encoding these polypeptides, are described above. The GT-derived targeting sequence corresponds to the 81 amino terminal amino acids of the human GT sequence. The GT amino acid sequences, and the polynucleotide sequences encoding the GT amino acid sequences, are described in Genbank Accession No. M70427 and Mengle-Gaw et al., Biochem. Biophys. Res. Commun. 176 (3), 1269-1276 (1991). [0059]
  • Amino acid sequences of mito-ECFP, mito-EYFP, GT-EGFP, and GT-EYFP, mito-YFP H148G, mito-YFP H148Q, mito-EYFP H148G, mito YFP-H148Q are shown in Tables 17-24. [0060]
  • In specific embodiments, nucleic acid sequences encoding targeting sequences linked to functional engineered fluorescent proteins have the sequences shown in Tables 23-32. [0061]
    TABLE 17
    mito-ECFP Amino Acid Sequence
    (SEQ ID NO:_)
    MLSLRQSIRFFKRSGIMVSKGEELFTGVVPILVELDGDVNGHRFSVSGEG
    EGDATYGKLTLKFICTTGKLPVPWPTLVTTLTWGVQCFSRYPDHMKQHDF
    FKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKED
    GNILGHKLEYNYISHNVYITADKQKNGIKAHFKIRHNIEDGSVQLADHYQ
    QNTPIGDGPVLLPDNHYLSTQSALSKDPNEKRDHMVLLEFVTAAGITLGM
    DELYK
  • [0062]
    TABLE 18
    mito-EYFP Amino Acid Sequence
    (SEQ ID NO:_)
    MLSLRQSIRFFKRSGIMVSKGEELFTGVVPILVELDGDVNGHKFSVSGEG
    EGDATYGKLTLKFICTTGKLPVPWPTLVTTFGYGVQCFARYPDHMKQHDF
    FKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKED
    GNILGHKLEYNYNSHNVYIMADKQKNGIKVNFKIRHNIEDGSVQLADHYQ
    QNTPIGDGPVLLPDNHYLSYQSALSKDPNEKRDHMVLLEFVTAAGITLGM
    DELYK
  • [0063]
    TABLE 19
    GT-EGFP Amino Acid Sequence
    (SEQ ID NO:_)
    MRLREPLLSGAAMPGASLQRACRLLVAVCALHLGVTLVYYLAGRDLSRLP
    QLVGVSTPLQGGSNSAAAIGQSSGELRTGGAMDPMVSKGEELFTGVVPIL
    VELDGDVNGHKFSVSGEGEGDATYGKLTLKFICTTGKLPVPWPTLVTTLT
    YGVQCFSRYPDHMKQHDFFKSAMPEGYVQERTIFFKDDGNYKTRAEVKFE
    GDTLVNRIELKGIDFKEDGNILGHKLEYNYNSHNVYIMADKQKNGIKVNF
    KIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNHYLSTQSALSKDPNEKR
    DHMVLLEFVTAAGITLGMDELYK
  • [0064]
    TABLE 20
    GT-EYFP Amino Acid Sequence
    (SEQ ID NO:_)
    MRLREPLLSGAAMPGASLQPACRLLVAVCALHLGVTLVYYTAGRDLSRLP
    QLVGVSTPLQGGSNSAAAIGQSSGELRTGGAMDPMVSKGEELFTGVVPIL
    VELDGDVNGHKFSVSGEGEGDATYGKLTLKFICTTGKLPVPWPTLVTTFG
    YGVQCFARYPDHMKQHDFFKSAMPEGYVQERTIFFKDDGNYKTRAEVKFE
    GDTLVNRIELKGIDFKEDGNILGHKLEYNYNSHNVYIMADKQKNGIKVNF
    KIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNHYLSYQSALSKDPNEKR
    DHMVLLEFVTAAGITLGMDELYK*
  • [0065]
    TABLE 21
    mito-YFP-H148G Amino Acid Sequence
    (SEQ ID NO:__)
    MLRTSSLFTRRVQPSLFRNILRLQSTSKGEELFTGVVPILVELDGDVNGH
    KFSVSGEGEGDATYGKLTLKFICTTGKLPVPWPTLVTTFGYGLQCFARYP
    DHMKRHDFFKSAMPEGYVQERTIFFKDDGNYKTPAEVKFEGDTLVNRIEL
    KGIDFKEDGNILGHKLEYNYNSGNVYIMADKQKNGIKVNFKIRHNIEDGS
    VQLADHYQQNTPIGDGPVLLPDNHYLSYQSALSKDPNEKRDHMVLLEFVT
    AAGITHGMDELYK
  • [0066]
    TABLE 22
    mito-EYFP-H148Q Amino Acid Sequence
    (SEQ ID NO:_)
    MLRTSSLFTRRVQPSLFRNILRLQSTSKGEELFTGVVPILVELDGDVNGH
    KFSVSGEGEGDATYGKLTLKFICTTGKLPVPWPTLVTTFGYGLQCFARYP
    DHMKRHDFFKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLVNPJEL
    KGIDFKEDGNILGHKLEYNYNSQNVYIMADKQKNGIKVNFKIRHNIEDGS
    VQLADHYQQNTPIGDGPVLLPDNHYLSYQSALSKDPNEKRDHMVLLEFVT
    AAGITHGMDELYK
  • [0067]
    TABLE 23
    mito-EYFP-H148G Amino Acid Sequence
    (SEQ ID NO:_)
    MLRTSSLFTRRVQPSLFRNILRLQSTMVSKGEELFTGVVPILVELDGDVN
    GHKFSVSGEGEGDATYGKLTLKFICTTGKLPVPWPTLVTTFGYGVQCFAR
    YPDHMKQHDFFKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLVNRI
    ELKGIDFKEDGNILGHKLEYNYNSGNVYIMADKQKNGIKVNFKIRHNIED
    GSVQLADHYQQNTPIGDGPVLLPDNHYLSYQSALSKDPNEKRDHMVLLEF
    VTAAGITLGMDELYK
  • [0068]
    TABLE 24
    mito-EYFP-H148Q Amino Acid Sequence
    (SEQ ID NO:_)
    MLRTSSLFTRRVQPSLFRNILRLQSTMVSKGEELFTGVVPILVELDGDVN
    GHKFSVSGEGEGDATYGKLTLKFICTTGKLPVPWPTLVTTFGYGVQCFAR
    YPDHMKQHDFFKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLVNRI
    ELKGIDFKEDGNILGHKLEYNYNSQNVYIMADKQKNGIKVNFKIRHNIED
    GSVQLADHYQQNTPIGDGPVLLPDNHYLSYQSALSKDPNEKRDHMVLLEF
    VTAAGITLGMDELYK
  • [0069]
    TABLE 25
    GT-ECFP Nucleic Acid Sequence
    ATGAGGCTTCGGGAGCCGCTCCTGAGCGGCGCCGCGATCCCAGGCGCGTCCCTACAGCGG (SEQ ID NO:_)
    GCCTGCCGCCTGCTCGTGGCCGTCTGCGCTCTGCACCTTGGCGTCACCCTCGTTTACTAC
    CTGGCTGGCCGCGACCTGAGCCGCCTGCCCCAACTGGTCGGAGTCTCCACACCGCTGCAG
    GGCGGCTCGAACAGTGCCGCCGCCATCGGGCAGTCCTCCGGGGAGCTCCGGACCGGAGGG
    GCCATGGATCCCATGGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTG
    GTCGAGCTGGACGGCGACGTAAACGGCCACAGGTTCAGCGTGTCCGGCGAGGGCGAGGGC
    GATGCCACCTACGGCAAGCTGACCCTGAAGTTCATCTGCACCACCGGCAAGCTGCCCGTG
    CCCTGGCCCACCCTCGTGACCACCCTGACCTGGGGCGTGCAGTGCTTCAGCCGCTACCCC
    GACCACATGAAGCAGCACGACTTCTTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAG
    CGTACCATCTTCTTCAAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAG
    GGCGACACCCTGGTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGACGGCAAC
    ATCCTGGGGCACAAGCTGGAGTACAACTACATCAGCCACAACGTCTATATCACCGCCGAC
    AAGCAGAAGAACGGCATCAAGGCCCACTTCAAGATCCGCCACAACATCGAGGACGGCAGC
    GTGCAGCTCGCCGACCACTACCAGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTG
    CCCGACAACCACTACCTGAGCACCCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGC
    GATCACATGGTCCTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCATGGACGAG
    CTGTACAAGTAA
  • [0070]
    TABLE 26
    mito-EYFP Nucleic Acid Secpence
    ATGCTGAGCCTGCGCCAGAGCATCCGCTTCTTCAAGCGCAGCGGCATCATGGTGAGCAAG (SEQ ID NO:_)
    GGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGTCGAGCTGGACGGCGACGTAAAC
    GGCCACAAGTTCAGCGTGTCCGGCGAGGGCGAGGGCGATGCCACCTACGGCAAGCTGACC
    CTGAAGTTCATCTGCACCACCGGCPAGCTGCCCGTGCCCTGGCCCACCCTCGTGACCACC
    TTCGGCTACGGCGTGCAGTGCTTCGCCCGCTACCCCGACCACATGAAGCAGCACGACTTC
    TTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAGCGCACCATCTTCTTCAAGGACGAC
    GGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGGCGACACCCTGGTGAACCGCATC
    GAGCTGAAGGGCATCGACTTCAAGGAGGACGGCAACATCCTGGGGCACAAGCTGGAGTAC
    AACTACAACAGCCACAACGTCTATATCATGGCCGACAAGCAGAKGAACGGCATCAAGGTG
    AACTTCAAGATCCGCCACAACATCGAGGACGGCAGCGTGCAGCTCGCCGACCACTACCAG
    CAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTGCCCGACAACCACTACCTGAGCTAC
    CAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGCGATCACATGGTCCTGCTGGAGTTC
    GTGACCGCCGCCGGGATCACTCTCGGCATGGACGAGCTGTACAAGTAA
  • [0071]
    TABLE 27
    GT-EGFP Nucleic Acid Sequence
    ATGAGGCTTCGGGAGCCGCTCCTGAGCGGCGCCGCGATGCCAGGCGCGTCCCTACAGCGG (SEQ ID NO:_)
    GCCTGCCGCCTGCTCGTGGCCGTCTGCGCTCTGCACCTTGGCGTCACCCTCGTTTACTAC
    CTGGCTGGCCGCGACCTGAGCCGCCTGCCCCAACTGGTCGGAGTCTCCACACCGCTGCAG
    GGCGGCTCGAACAGTGCCGCCGCCATCGGGCAGTCCTCCGGGGAGCTCCGGACCGGAGGG
    GCCATGGATCCCATGGTGAGCAAGCGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTG
    GTCGAGCTGGACGGCGACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGGGCGAGGGC
    GATGCCACCTACGGCAAGCTGACCCTGAAGTTCATCTGCACCACCGGCAAGCTGCCCGTG
    CCCTGGCCCACCCTCGTGACCACCCTGACCTACGGCGTGCAGTGCTTCAGCCGCTACCCC
    GACCACATGAAGCAGCACGACTTCTTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAG
    CGCACCATCTTCTTCAAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAG
    GGCGACACCCTGGTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGACGGCAAC
    ATCCTGGGGcACAAGCTGGAGTACAACTACAACAGCCACAACGTCTATATCATGGCCGAC
    AAGCAGAAGAACGGCATCAAGGTGAACTTCAAGATCCGCCACAACATCGAGGACGGCAGC
    GTGCAGCTCGCCGACCACTACCAGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTG
    CCCGACAACCACTACCTGAGCACCCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGC
    GATCACATGGTCCTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCATGGACGAG
    CTGTACAAGTAA
  • [0072]
    TABLE 28
    GT-EYFP Nucleic Acid Sequence
    ATGAGGCTTCGGGAGCCGCTCCTGAGCGGCGCCGCGATGCCAGGCGCGTCCCTACAGCGG (SEQ ID NO:_)
    GCCTGCCGCCTGCTCGTGGCCGTCTGCGCTCTGCACCTTGGCGTCACCCTCGTTTACTAC
    CTGGCTGGCCGCGACCTGAGCCGCCTGCCCCAACTGGTCGGAGTCTCCACACCGCTGCAG
    GGCGGCTCGAACAGTGCCGCCGCCATCGGGCAGTCCTCCGGGGAGCTCCGGACCGGAGGG
    GCCATGGATCCCATGGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTG
    GTCGAGCTGGACGGCGACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGGGCGAGGGC
    GATGCCACCTACGGCAAGCTGACCCTGAAGTTCATCTGCACCACCGGCAAGCTGCCCGTG
    CCCTGGCCCACCCTCGTGACCACCTTCGGCTACGGCGTGCAGTGCTTCGCCCGCTACCCC
    GACCACATGAAGCAGCACGACTTCTTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAG
    CGCACCATCTTCTTCAAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAG
    GGCGACACCCTGGTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGACGGCAAC
    ATCCTGGGGCACAAGCTGGAGTACAACTACAACAGCCACAACGTCTATATCATGGCCGAC
    AAGCAGAAGAACGGCATCAAGGTGAACTTCAAGATCCGCCACAACATCGAGGACGGCAGC
    GTGCAGCTCGCCGACCACTACCAGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTG
    CCCGACAACCACTACCTGAGCTACCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGC
    GATCACATGGTCCTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCATGGACGAG
    CTGTACAAGTAA
  • [0073]
    TABLE 29
    mito-YFP H148G Nucleic Acid Sequence
    ATGCTGAGCCTGCGCCAGAGCATCCGCTTCTTCAAGCGCAGCGGCATCATGAGTAAAGGA (SEQ ID NO:_)
    GAAGAACTTTTCACTGGAGTTGTCCCAATTCTTGTTGAATTAGATGGTGATGTTAATGGG
    CACAAATTTTCTGTCAGTGGAGAGGGTGAAGGTGATGCAACATACGGAAAACTTACCCTT
    AAATTTATTTGCACTACTGGAAAACTACCTGTTCCATGGCCAACACTTGTCACTACTTTC
    GGTTATGGTCTTCAATGCTTTGCAAGATACCCAGATCATATGAAACGGCATGACTTTTTC
    AAGAGTGCCATGCCCGAAGGTTATGTTCAGGAAAGAACTATATTTTTCAAAGATGACGGG
    AACTACAAGACACGTGCTGAAGTCAAGTTTGAAGGTGATACCCTTGTTAATAGAATCGAG
    TTAAAAGGTATTGATTTTAAAGAAGATGGAAACATTCTTGGACACAAATTGGAATACAAC
    TATAACTCAGGCAATGTATACATCATGGCAGACAAACAAAAGAATGGAATCAAAGTTAAC
    TTCAAAATTAGACACAACATTGAAGATGGAAGCGTTCAACTAGCAGACCATTATCAACAA
    AATACTCCAATTGGCGATGGCCCTGTCCTTTTACCAGACAACCATTACCTGTCCTATCAA
    TCTGCCCTTTCGAAAGATCCCAACGAAAAGAGAGACCACATGGTCCTTCTTGAGTTTGTA
    ACAGCTGCTGGGATTACACATGGCATGGATGAACTATACAAA
  • [0074]
    TABLE 30
    mito-YFP H148Q Nucleic Acid Sequence
    ATGCTGAGCCTGCGCCAGAGCATCCGCTTCTTCAAGCGCAGCGGCATCATGAGTAAAGGA (SEQ ID NO:_)
    GAAGAACTTTTCACTGGAGTTGTCCCAATTCTTGTTGAATTAGATGGTGATGTTAATGGG
    CACAAATTTTCTGTCAGTGGAGAGGGTGAAGGTGATGCAACATACGGAAAACTTACCCTT
    AAATTTATTTGCACTACTGGAAAACTACCTGTTCCATGGCCAACACTTGTCACTACTTTC
    GGTTATGGTCTTCAATGCTTTGCAAGATACCCAGATCATATGAAACGGCATGACTTTTTC
    AAGAGTGCCATGCCCGAAGGTTATGTTCAGGAAAGAACTATATTTTTCAAAGATGACGGG
    AACTACAAGACACGTGCTGAAGTCAAGTTTGAAGGTGATACCCTTGTTAATAGAATCGAG
    TTAAAAGGTATTGATTTTAAAGAAGATGGAAACATTCTTGGACACAAATTGGAATACAAC
    TATAACTCAGGCAATGTATACATCATGGCAGACAAACAAAAGAATGGAATCAAAGTTAAC
    TTCAAAATTAGACACAACATTGAAGATGGAAGCGTTCAACTAGCAGACCATTATCAACAA
    AATACTCCAATTGGCGATGGCCCTGTCCTTTTACCAGACAACCATTACCTGTCCTATCAA
    TCTGCCCTTTCGAAAGATCCCAACGAAAAGAGAGACCACATGGTCCTTCTTGAGTTTGTA
    ACAGCTGCTGGGATTACACATGGCATGGATGAACTATACAAA
  • [0075]
    TABLE 31
    mito-EYFP-H148G Nucleic Acid Sequence
    (SEQ ID NO:_)
    ATGCTGAGCCTGCGCCAGAGCATCCGCTTCTTCAAGCGCAGCGGCATCAT
    GGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGTCG
    AGCTGGACGGCGACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGGGC
    GAGGGCGATGCCACCTACGGCAAGCTGACCCTGAAGTTCATCTGCACCAC
    CGGCAAGCTGCCCGTGCCCTGGCCCACCCTCGTGACCACCTTCGGCTACG
    GCGTGCAGTGCTTCGCCCGCTACCCCGACCACATGAAGCAGCACGACTTC
    TTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAGCGCACCATCTTCTT
    CAAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGGCG
    ACACCCTGGTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGAC
    GGCAACATCCTGGGGCACAAGCTGGAGTACAACTACAACAGCGGCAACGT
    CTATATCATGGCCGACAAGCAGAAGAACGGCATCAAGGTGAACTTCAAGA
    TCCGCCACAACATCGAGGACGGCAGCGTGCAGCTCGCCGACCACTACCAG
    CAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTGCCCGACAACCACTA
    CCTGAGCTACCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGCGATC
    ACATGGTCCTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCATG
    GACGAGCTGTACAAGTAA
  • [0076]
    TABLE 32
    mito-EYFP-H148Q Nucleic Acid Sequence
    (SEQ ID NO:_)
    ATGCTGAGCCTGCGCCAGAGCATCCGCTTCTTCAAGCGCAGCGGCATCAT
    GGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGTCG
    AGCTGGACGGCGACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGGGC
    GAGGGCGATGCCACCTACGGCAAGCTGACCCTGAAGTTCATCTGCACCAC
    CGGCAAGCTGCCCGTGCCCTGGCCCACCCTCGTGACCACCTTCGGCTACG
    GCGTGCAGTGCTTCGCCCGCTACCCCGACCACATGAAGCAGCACGACTTC
    TTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAGCGCACCATCTTCTT
    CAAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGGCG
    ACACCCTGGTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGAC
    GGCAACATCCTGGGGCACAAGCTGGAGTACAACTACAACAGCCAGAACGT
    CTATATCATGGCCGACAAGCAGAAGAACGGCATCAAGGTGAACTTCAAGA
    TCCGCCACAACATCGAGGACGGCAGCGTGCAGCTCGCCGACCACTACCAG
    CAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTGCCCGACAACCACTA
    CCTGAGCTACCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGCGATC
    ACATGGTCCTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCATG
    GACGAGCTGTACAAGTAA
  • The fluorescent indicators can be produced as proteins fused to other fluorescent indicators or targeting sequences by recombinant DNA technology. Recombinant production of fluorescent proteins involves expressing nucleic acids having sequences that encode the proteins. Nucleic acids encoding fluorescent proteins can be obtained by methods known in the art. For example, a nucleic acid encoding the protein can be isolated by polymerase chain reaction of cDNA from [0077] A. victoria using primers based on the DNA sequence of A. victoria green fluorescent protein. PCR methods are described in, for example, U.S. Pat. No. 4,683,195; Mullis, et al. Cold Spring Harbor Symp. Quant. Biol. 51:263 (1987), and Erlich, ed., PCR Technology, (Stockton Press, NY, 1989). Mutant versions of fluorescent proteins can be made by site-specific mutagenesis of other nucleic acids encoding fluorescent proteins, or by random mutagenesis caused by increasing the error rate of PCR of the original polynucleotide with 0.1 mM MnCl2 and unbalanced nucleotide concentrations. See, e.g., U.S. patent application Ser. No. 08/337,915, filed Nov. 10, 1994 or International application PCT/US95/14692, filed Nov. 10, 1995.
  • The construction of expression vectors and the expression of genes in transfected cells involves the use of molecular cloning techniques also well known in the art. Sambrook et al., Molecular Cloning—A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., (1989) and Current Protocols in Molecular Biology, F. M. Ausubel et al., eds., (Current Protocols, a joint venture between Greene Publishing Associates, Inc. and John Wiley & Sons, Inc., most recent Supplement). [0078]
  • Nucleic acids used to transfect cells with sequences coding for expression of the polypeptide of interest generally will be in the form of an expression vector including expression control sequences operatively linked to a nucleotide sequence coding for expression of the polypeptide. As used herein, “operatively linked” refers to a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner. A control sequence operatively linked to a coding sequence is ligated such that expression of the coding sequence is achieved under conditions compatible with the control sequences. “Control sequence” refers to polynucleotide sequences which are necessary to effect the expression of coding and non-coding sequences to which they are ligated. Control sequences generally include promoter, ribosomal binding site, and transcription termination sequence. The term “control sequences” is intended to include, at a minimum, components whose presence can influence expression, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences. [0079]
  • As used herein, the term “nucleotide sequence coding for expression of” a polypeptide refers to a sequence that, upon transcription and translation of mRNA, produces the polypeptide. This can include sequences containing, e.g., introns. As used herein, the term “expression control sequences” refers to nucleic acid sequences that regulate the expression of a nucleic acid sequence to which it is operatively linked. Expression control sequences are operatively linked to a nucleic acid sequence when the expression control sequences control and regulate the transcription and, as appropriate, translation of the nucleic acid sequence. Thus, expression control sequences can include appropriate promoters, enhancers, transcription terminators, a start codon (i.e., ATG) in front of a protein-encoding gene, splicing signals for introns, maintenance of the correct reading frame of that gene to permit proper translation of the mRNA, and stop codons. [0080]
  • Methods which are well known to those skilled in the art can be used to construct expression vectors containing the fluorescent indicator coding sequence and appropriate transcriptional/translational control signals. These methods include in vitro recombinant DNA techniques, synthetic techniques and in vivo recombination/genetic recombination. (See, for example, the techniques described in Maniatis, et al., Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., 1989). Transformation of a host cell with recombinant DNA may be carried out by conventional techniques as are well known to those skilled in the art. Where the host is prokaryotic, such as [0081] E. coli, competent cells which are capable of DNA uptake can be prepared from cells harvested after exponential growth phase and subsequently treated by the CaCl2 method by procedures well known in the art. Alternatively, MgCl2 or RbCl can be used. Transformation can also be performed after forming a protoplast of the host cell or by electroporation.
  • When the host is a eukaryote, such methods of transfection of DNA as calcium phosphate co-precipitates, conventional mechanical procedures such as microinjection, electroporation, insertion of a plasmid encased in liposomes, or virus vectors may be used. Eukaryotic cells can also be cotransfected with DNA sequences encoding the fusion polypeptide of the invention, and a second foreign DNA molecule encoding a selectable phenotype, such as the herpes simplex thymidine kinase gene. Another method is to use a eukaryotic viral vector, such as simian virus 40 (SV40) or bovine papilloma virus, to transiently infect or transform eukaryotic cells and express the protein. (Eukaryotic Viral Vectors, Cold Spring Harbor Laboratory, Gluzman ed., 1982). [0082]
  • Techniques for the isolation and purification of polypeptides of the invention expressed in prokaryotes or eukaryotes may be by any conventional means such as, for example, preparative chromatographic separations and immunological separations such as those involving the use of monoclonal or polyclonal antibodies or antigen. [0083]
  • A variety of host-expression vector systems may be utilized to express fluorescent indicator coding sequence. These include but are not limited to microorganisms such as bacteria transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing a fluorescent indicator coding sequence; yeast transformed with recombinant yeast expression vectors containing the fluorescent indicator coding sequence; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing a fluorescent indicator coding sequence; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing a fluorescent indicator coding sequence; or animal cell systems infected with recombinant virus expression vectors (e.g., retroviruses, adenovirus, vaccinia virus) containing a fluorescent indicator coding sequence, or transformed animal cell systems engineered for stable expression. [0084]
  • Depending on the host/vector system utilized, any of a number of suitable transcription and translation elements, including constitutive and inducible promoters, transcription enhancer elements, transcription terminators, etc. may be used in the expression vector (see, e.g., Bitter, et al., Methods in Enzymology 153:516-544, 1987). For example, when cloning in bacterial systems, inducible promoters such as pL of bacteriophage λ, plac, ptrp, ptac (ptrp-lac hybrid promoter) and the like may be used. When cloning in mammalian cell systems, promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the retrovirus long terminal repeat; the adenovirus late promoter; the vaccinia virus 7.5K promoter) may be used. Promoters produced by recombinant DNA or synthetic techniques may also be used to provide for transcription of the inserted fluorescent indicator coding sequence. [0085]
  • In bacterial systems a number of expression vectors may be advantageously selected depending upon the use intended for the fluorescent indicator expressed. For example, when large quantities of the fluorescent indicator are to be produced, vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable. Those which are engineered to contain a cleavage site to aid in recovering fluorescent indicator are preferred. In yeast, a number of vectors containing constitutive or inducible promoters may be used. For a review see, Current Protocols in Molecular Biology, Vol. 2, Ed. Ausubel, et al., Greene Publish. Assoc. & Wiley Interscience, Ch. 13, 1988; Grant, et al., Expression and Secretion Vectors for Yeast, in Methods in Enzymology, Eds. Wu & Grossman, 31987, Acad. Press, N.Y., Vol. 153, pp.516-544, 1987; Glover, DNA Cloning, Vol. II, IRL Pess, Wash., D.C., Ch. 3, 1986; and Bitter, Heterologous Gene Expression in Yeast, Methods in Enzymology, Eds. Berger & Kimmel, Acad. Press, N.Y., Vol. 152, pp. 673-684, 1987; and The Molecular Biology of the Yeast Saccharomyces, Eds. Strathern et al., Cold Spring Harbor Press, Vols. I and II, 1982. A constitutive yeast promoter such as ADH or LEU2 or an inducible promoter such as GAL may be used (Cloning in Yeast, Ch. 3, R. Rothstein In: DNA Cloning Vol. 11, A Practical Approach, Ed. D M Glover, IRL Press, Wash., D.C., 1986). Alternatively, vectors may be used which promote integration of foreign DNA sequences into the yeast chromosome. [0086]
  • In cases where plant expression vectors are used, the expression of a fluorescent indicator coding sequence may be driven by any of a number of promoters. For example, viral promoters such as the 35S RNA and 19S RNA promoters of CaMV (Brisson, et al., Nature 310:511-514, 1984), or the coat protein promoter to TMV (Takamatsu, et al., EMBO J. 6:307-311, 1987) may be used; alternatively, plant promoters such as the small subunit of RUBISCO (Coruzzi, et al., 1984, EMBO J. 3:1671-1680; Broglie, et al., Science 224:838-843, 1984); or heat shock promoters, e.g., soybean hsp17.5-E or hsp17.3-B (Gurley, et al., Mol. Cell. Biol. 6:559-565, 1986) may be used. These constructs can be introduced into plant cells using Ti plasmids, Ri plasmids, plant virus vectors, direct DNA transformation, microinjection, electroporation, etc. For reviews of such techniques see, for example, Weissbach & Weissbach, Methods for Plant Molecular Biology, Academic Press, NY, Section VIII, pp. 421-463, 1988; and Grierson & Corey, Plant Molecular Biology, 2d Ed., Blackie, London, Ch. 7-9, 1988. [0087]
  • An alternative expression system which could be used to express fluorescent indicator is an insect system. In one such system, [0088] Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes. The virus grows in Spodoptera frugiperda cells. The fluorescent indicator coding sequence may be cloned into non-essential regions (for example, the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter). Successful insertion of the fluorescent indicator coding sequence will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat coded for by the polyhedrin gene). These recombinant viruses are then used to infect Spodoptera frugiperda cells in which the inserted gene is expressed, see Smith, et al., J. Viol. 46:584, 1983; Smith, U.S. Pat. No. 4,215,051.
  • Eukaryotic systems, and preferably mammalian expression systems, allow for proper post-translational modifications of expressed mammalian proteins to occur. Eukaryotic cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, phosphorylation, and, advantageously secretion of the gene product should be used as host cells for the expression of fluorescent indicator. Such host cell lines may include but are not limited to CHO, VERO, BHK, HeLa, COS, MDCK, Jurkat, HEK-293, and WI38. Primary cell lines, such as neonatal rat myocytes, can also be used. [0089]
  • Mammalian cell systems which utilize recombinant viruses or viral elements to direct expression may be engineered. For example, when using adenovirus expression vectors, the fluorescent indicator coding sequence may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region E1 or E3) will result in a recombinant virus that is viable and capable of expressing the fluorescent indicator in infected hosts (e.g., see Logan & Shenk, Proc. Natl. Acad. Sci. USA, 81: 3655-3659, 1984). Alternatively, the vaccinia virus 7.5K promoter may-be used (e.g., see, Mackett, et al., Proc. Natl. Acad. Sci. USA, 79: 7415-7419, 1982; Mackett, et al., J. Virol. 49: 857-864,-1984; Panicali, et al., Proc. Natl. Acad. Sci. USA 79: 4927-4931, 1982). Of particular interest are vectors based on bovine papilloma virus which have the ability to replicate as extrachromosomal elements (Sarver, et al., Mol. Cell. Biol. 1: 486, 1981). Shortly after entry of this DNA into mouse cells, the plasmid replicates to about 100 to 200 copies per cell. Transcription of the inserted cDNA does not require integration of the plasmid into the host's chromosome, thereby yielding a high level of expression. These vectors can be used for stable expression by including a selectable marker in the plasmid, such as the neo gene. Alternatively, the retroviral genome can be modified for use as a vector capable of introducing and directing the expression of the fluorescent indicator gene in host cells (Cone & Mulligan, Proc. Natl. Acad. Sci. USA, 81:6349-6353, 1984). High level expression may also be achieved using inducible promoters, including, but not limited to, the metallothionein IIA promoter and heat shock promoters. [0090]
  • The recombinant nucleic acid can be incorporated into an expression vector including expression control sequences operatively linked to the recombinant nucleic acid. The expression vector can be adapted for function in prokaryotes or eukaryotes by inclusion of appropriate promoters, replication sequences, markers, etc. [0091]
  • DNA sequences encoding the fluorescence indicator polypeptide of the invention can be expressed in vitro or in vivo by DNA transfer into a suitable recombinant host cell. As used herein, “recombinant host cells” are cells in which a vector can be propagated and its DNA expressed. The term also includes any progeny of the subject host cell. It is understood that all progeny may not be identical to the parental cell since there may be mutations that occur during replication. However, such progeny are included when the term “recombinant host cell” is used. Methods of stable transfer, in other words when the foreign DNA is continuously maintained in the host, are known in the art. [0092]
  • The expression vector can be transfected into a host cell for expression of the recombinant nucleic acid. Recombinant host cells can be selected for high levels of expression in order to purify the fluorescent indicator fusion protein. [0093] E. coli is useful for this purpose. Alternatively, the host cell can be a prokaryotic or eukaryotic cell selected to study the activity of an enzyme produced by the cell. In this case, the linker peptide is selected to include an amino acid sequence recognized by the protease. The cell can be, e.g., a cultured cell or a cell taken in vivo from a transgenic animal.
  • Transgenic Animals [0094]
  • In another embodiment, the invention provides a transgenic non-human animal that expresses a polynucleotide sequence which encodes a fluorescent protein pH sensor. [0095]
  • The “non-human animals” of the invention comprise any non-human animal having a polynucleotide sequence which encodes a fluorescent indicator. Such non-human animals include vertebrates such as rodents, non-human primates, sheep, dog, cow, pig, amphibians, and reptiles. Preferred non-human animals are selected from the rodent family including rat and mouse, most preferably mouse. The “transgenic non-human animals” of the invention are produced by introducing “transgenes” into the germline of the non-human animal. Embryonal target cells at various developmental stages can be used to introduce transgenes. Different methods are used depending on the stage of development of the embryonal target cell. The zygote is the best target for micro-injection. In the mouse, the male pronucleus reaches the size of approximately 20 micrometers in diameter which allows reproducible injection of 1-2 pl of DNA solution. The use of zygotes as a target for gene transfer has a major advantage in that in most cases the injected DNA will be incorporated into the host gene before the first cleavage (Brinster et al., Proc. Natl. Acad. Sci. USA 82:4438-4442, 1985). As a consequence, all cells of the transgenic non-human animal will carry the incorporated transgene. This will in general also be reflected in the efficient transmission of the transgene to offspring of the founder since 50% of the germ cells will harbor the transgene. Microinjection of zygotes is the preferred method for incorporating transgenes in practicing the invention. [0096]
  • The term “transgenic” is used to describe an animal which includes exogenous genetic material within all of its cells. A “transgenic” animal can be produced by cross-breeding two chimeric animals which include exogenous genetic material within cells used in reproduction. Twenty-five percent of the resulting offspring will be transgenic, i.e., animals which include the exogenous genetic material within all of their cells in both alleles. 50% of the resulting animals will include the exogenous genetic material within one allele and 25% will include no exogenous genetic material. [0097]
  • Retroviral infection can also be used to introduce transgene into a non-human animal. The developing non-human embryo can be cultured in vitro to the blastocyst stage. During this time, the blastomeres can be targets for retro viral infection (Jaenisch, R., Proc. Natl. Acad. Sci USA 73:1260-1264, 1976). Efficient infection of the blastomeres is obtained by enzymatic treatment to remove the zona pellucida (Hogan, et al. (1986) in Manipulating the Mouse Embryo, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.). The viral vector system used to introduce the transgene is typically a replication-defective retrovirus carrying the transgene (Jahner, et al., Proc. Natl. Acad. Sci. USA 82:6927-6931, 1985; Van der Putten, et al., Proc. Natl. Acad. Sci USA 82:6148-6152, 1985). Transfection is easily and efficiently obtained by culturing the blastomeres on a monolayer of virus-producing cells (Van der Putten, supra; Stewart, et al., EMBO J. 6:383-388, 1987). Alternatively, infection can be performed at a later stage. Virus or virus-producing cells can be injected into the blastocoele (D. Jahner et al., Nature 298:623-628, 1982). Most of the founders will be mosaic for the transgene since incorporation occurs only in a subset of the cells which formed the transgenic nonhuman animal. Further, the founder may contain various retro viral insertions of the transgene at different positions in the genome which generally will segregate in the offspring. In addition, it is also possible to introduce transgenes into the germ line, albeit with low efficiency, by intrauterine retro viral infection of the midgestation embryo (D. Jahner et al., supra). A third type of target cell for transgene introduction is the embryonal stem cell (ES). ES cells are obtained from pre-implantation embryos cultured in vitro and fused with embryos (M. J. Evans et al. Nature 292:154-156, 1981; M. O. Bradley et al., Nature 309: 255-258,1984; Gossler, et al., Proc. Natl. Acad. Sci USA 83: 9065-9069, 1986; and Robertson et al., Nature 322:445-448, 1986). Transgenes can be efficiently introduced into the ES cells by DNA transfection or by retrovirus-mediated transduction. Such transformed ES cells can thereafter be combined with blastocysts from a nonhuman animal. The ES cells thereafter colonize the embryo and contribute to the germ line of the resulting chimeric animal. (For review see Jaenisch, R., Science 240: 1468-1474, 1988). [0098]
  • “Transformed” means a cell into which (or into an ancestor of which) has been introduced, by means of recombinant nucleic acid techniques, a heterologous polynucleotide. “Heterologous” refers to a polynucleotide sequence that either originates from another species or is modified from either its original form or the form primarily expressed in the cell. [0099]
  • “Transgene” means any piece of DNA which is inserted by artifice into a cell, and becomes part of the genome of the organism (i.e., either stably integrated or as a stable extrachromosomal element) which develops from that-cell. Such a transgene may include a gene which is partly or entirely heterologous (i.e., foreign) to the transgenic organism, or may represent a gene homologous to an endogenous gene of the organism. Included within this definition is a transgene created by the providing of an RNA sequence which is transcribed into DNA and then incorporated into the genome. The transgenes of the invention include DNA sequences which encode the fluorescent indicator which may be expressed in a transgenic non-human animal. The term “transgenic” as used herein additionally includes any organism whose genome has been altered by in vitro manipulation of the early embryo or fertilized egg or by any transgenic technology to induce a specific gene knockout. The term “gene knockout” as used herein, refers to the targeted disruption of a gene in vivo with complete loss of function that has been achieved by any transgenic technology familiar to those in the art. In one embodiment, transgenic animals having gene knockouts are those in which the target gene has been rendered nonfunctional by an insertion targeted to the gene to be rendered non-functional by homologous recombination. As used herein, the term “transgenic” includes any transgenic technology familiar to those in the art which can produce an organism carrying an introduced transgene or one in which an endogenous gene has been rendered non-functional or “knocked out.”[0100]
  • Detection of pH Using Fluorescent Indicator Proteins [0101]
  • In another embodiment, the invention provides a method for determining the pH of a sample by contacting the sample with an indicator including a first fluorescent protein moiety whose emission intensity changes as pH varies between [0102] pH 5 and 10, exciting the indicator, and then determining the intensity of light emitted by the first fluorescent protein moiety at a first wavelength. The emission intensity of the first fluorescent protein moiety indicates the pH of the sample.
  • The fluorescent protein moiety can be a functional engineered protein substantially identical to the amino acid sequence of Aequora green fluorescence protein (SEQ ID NO: ______). Preferred green fluorescence proteins include those having a functional engineered fluorescent protein that includes one of the following sets of substitutions in the amino acid sequence of the Aequora green fluorescent protein (SEQ ID NO:______): S65G/S72A/T203Y/H231L, S65G/V68L/Q69K/S72A/T203Y, or K26R/F64L/S65T/Y66W/N146I/M153T/V163A/N164H/H231L. Other preferred green fluorescence proteins include those having a functional engineered fluorescent protein that includes H148G or H148Q substitutions in the Aequora green fluorescent protein. These proteins include the YFP H148G (SEQ ID NO:______) and YFP H148Q (SEQ ID NO: ______) proteins described above. [0103]
  • The sample in which pH is to be measured can be a biological sample, e.g., a biological tissue such as an extracellular matrix, blood or lymphatic tissue, or a cell. The method is particularly suitable for measuring pH in a specific region of the cell, e.g., the cytosol, or an organellar space such as the inner mitochondrial matrix, the lumen of the Golgi, cytosol, the endoplasmic reticulum, the chloroplast lumen, the lumen of lysosome, or the lumen of an endosome. [0104]
  • In some embodiments, the first fluorescent protein moiety is linked to a targeting sequence that directs the fluorescent protein to a desired cellular compartment. Examples of targeting sequences include the amino terminal 81 amino acids of human type II membrane-anchored protein galactosyltransferase (SEQ ID NO:______) for directing the fluorescent indicator protein to the Golgi and the amino terminal 12 amino acids of the presequence of subunit IV of cytochrome c oxidase (SEQ ID NO:______) for directing a fluorescent pH indicator protein to the mitochondrial matrix. The 12 amino acids of the presequence of subunit IV of cytochrome c oxidase (SEQ ID NO:______) may be linked to the pH fluorescent indicator protein through a linker sequence, e.g., Arg-Ser-Gly-Ile (SEQ ID NO:______). [0105]
  • In another embodiment, the invention provides a method of determining the pH of a region of a cell by introducing into the cell a polynucleotide encoding a polypeptide including an indicator having a first fluorescent protein moiety whose emission intensity changes as pH varies between 5 and 10, culturing the cell under conditions that permit expression of the polynucleotide; exciting the indicator; and determining the intensity of the light emitted by the first protein moiety at a first wavelength. The emission intensity of the first fluorescent protein moiety indicates the pH of the region of the cell in which the indicator is present. [0106]
  • The polynucleotide can be introduced using methods described above. Thus, the method can be used to measure intracellular pH in cells cultured in vitro, e.g., HeLa cells, or alternatively in vivo, e.g., in cells of an animal carrying a transgene encoding a pH-dependent fluorescent indicator protein. [0107]
  • Fluorescence in the sample can be measured using a fluorometer. In general, excitation radiation, from an excitation source having a first wavelength, passes through excitation optics. The excitation optics cause the excitation radiation to excite the sample. In response, fluorescent proteins in the sample emit radiation which has a wavelength that is different from the excitation wavelength. Collection optics then collect the emission from the sample. The device can include a temperature controller to maintain the sample at a specific temperature while it is being scanned. If desired, a multi-axis translation stage can be used to move a microtiter plate holding a plurality of samples in order to position different wells to be exposed. The multi-axis translation stage, temperature controller, auto-focusing feature, and electronics associated with imaging and data collection can be managed by an appropriately programmed digital computer. The computer also can transform the data collected during the assay into another format for presentation. [0108]
  • Methods of performing assays on fluorescent materials are well known in the art and are described in, e.g., Lakowicz, J. R., [0109] Principles of Fluorescence Spectroscopy, New York:Plenum Press (1983); Herman, B., Resonance energy transfer microscopy, in: Fluorescence Microscopy of Living Cells in Culture, Part B, Methods in Cell Biology, vol. 30, ed. Taylor, D. L. & Wang, Y.-L., San Diego: Academic Press (1989), pp. 219-243; Turro, N. J., Modern Molecular Photochemistry, Menlo Park: Benjamin/Cummings Publishing Col, Inc. (1978), pp. 296-361.
  • The pH can be analyzed on cells in vivo, or from samples derived from cells transfected with polynucleotides or proteins expressing the pH indicator proteins. Because fluorescent pH indicator proteins can be expressed recombinantly inside a cell, the pH in an intracellular region, e.g., an organelle, or an extracellular region of an organism can be determined simply by determining changes in fluorescence. [0110]
  • Fluorescent protein pH sensors may vary in their respective pK[0111] a, and the differences in pKa can be used to select the most suitable fluorescent protein sensor most suitable for a particular application. In general, a sensor protein should be used whose pKa is close to the pH of the sample to be measured. Preferably the pKa is within 1.5 pH unit of the sample. More preferably the pKa is within 1 pH unit, and still more preferably the pKa is within 0.5 pH unit of the sample.
  • Thus, a fluorescent protein pH sensor having a pK[0112] a of about 7.1, e.g., the EYFP mutant described below, is preferred for determining the pH of cytosolic, Golgi, and mitochondrial matrix pH areas of a cell. The YFP-H148G, YFP-H148Q, EYFP-H148G and EYFP-H148Q mutants are well-suited for measuring the pH of alkaline environments, e.g., mitochondrial matrix, as they have a pKa of 7.5 and 8.0, respectively.
  • For more acidic organelles, a fluorescence sensor protein having a lower pK[0113] a, e.g., a pKa of about 6.1, is preferred.
  • To minimize artefactually low fluorescence measurements that occur due to cell movement or focusing, the fluorescence of a fluorescent protein pH sensor can be compared to the fluorescence of a second sensor, e.g., a second fluorescent protein pH sensor, that is also present in the measured sample. The second fluorescent protein pH sensor should have an emission spectra distinct from the first fluorescent protein pH sensor so that the emission spectra of the two sensors can be distinguished. Because experimental conditions such as focusing and cell movement will affect fluorescence of the second sensor as well as the first sensor, comparing the relative fluorescence of, the two sensors allows for the normalization of fluorescence. [0114]
  • A convenient method of comparing the samples is to compute the ratio of the fluorescence of the first fluorescent protein pH sensor to that of the second fluorescent protein pH sensor. [0115]
  • Kits [0116]
  • The materials and components described for use in the methods of the invention are ideally suited for the preparation of a kit. Such a kit may comprise a carrier means being compartmentalized to receive one or more container means such as vials, tubes, and the like, each of the container means comprising one of the separate elements to be used in the method. For example, one of the container means may comprise a polynucleotide encoding a fluorescent protein pH sensor. A second container may further comprise fluorescent protein pH sensor. The constituents may be present in liquid or lyophilized form, as desired. [0117]
  • Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to limit the scope of the invention described in the claims. [0118]
  • EXAMPLES Example 1 Construction of Fluorescent Protein pH Sensors
  • Fluorescent protein pH sensors were constructed by engineering site-specific mutations in polynucleotides encoding forms of the [0119] Aequora victoria green fluorescent protein (GFP). The starting GFP variant was the polynucleotide encoding the GFP variant EGFP (for enhanced green fluorescent protein). The EGFP variant had the amino acid substitutions F64L/S65T/H231L relative to the wild-type Aequora victoria GFP sequence.
  • The ECFP (enhanced cyan fluorescent protein) mutant was constructed by altering the EGFP polynucleotide sequence so that it encoded a protein having the amino acid substitutions K26R/F64L/S65T/Y66W/N146I/M153T/V163A/N164H/H231L relative to the wild-type GFP amino acid sequence. A second variant, named EYFP (enhanced yellow fluorescent protein) was constructed by altering the EGFP polynucleotide to encode a protein having the amino acid substitutions S65G/S72A/T203Y/H231L relative to the amino acid sequence of GFP. A third variant, named EYFP-V68L-Q69K, was constructed by altering the EGFP polynucleotide to encode a protein having the amino acid substitutions S65G/V68L/Q69K/S72A/T203Y relative to the amino acid sequence of GFP. [0120]
  • A HindIII site and Kozak consensus sequence (GCCACCATG) was introduced at the 5′ end of the polynucleotide encoding the GFP variants, and an EcoR1 site was added at the 3′ end of the gene of each indicator, and the fragments were ultimately ligated into the HindIII/EcoR1 sites of the mammalian expression vector pcDNA3 (Invitrogen). EGFP and EYFP mutant proteins with no targeting signals were used as indicators of pH in the cytosol or nucleus. [0121]
  • To construct fluorescent protein pH sensors to use as pH indicators in the Golgi, polynucleotides encoding the 81 N-terminal amino acids of the type II membrane-anchored protein galactosyltransferase (GT:UDP-galactose-β,1,4-galactosyltransferase. EC 2.4.1.22) ligated to polynucleotides encoding EGFP, ECFP, or EYFP. The polynucleotides encoding the resulting proteins were named GT-EGFP, GT-ECFP, and GT-EYFP, respectively. [0122]
  • Mitochondrial matrix fluorescent protein pH sensors were constructed by attaching polynucleotides encoding 12 amino acids at the amino terminus of the presequence of subunit IV of cytochrome c oxidase (Hurt et al, EMBO J. 4:2061-68 (1985) to a polynucleotide encoding the amino acid sequence Arg-Sea-Gly-Ile, which in turn was ligated to polynucleotides encoding ECFP or EYFP. These constructs were labeled ECFP-mito or EYFP-mito. [0123]
  • The constructs used to examine intracellular pH are summarized in FIG. 1. [0124]
  • Example 2 pH Titration of Fluorescent Sensor Proteins in Vitro
  • The pH sensitivity of the fluorescence of the proteins ECFP, EGFP, EYFP, GT-EGFP, and GT-EYFP was first examined. [0125]
  • Absorbance spectra were obtained in a Cary 3E spectrophotometer (Varian). For pH titration, a monochromator-equipped fluorometer (Spex Industries, NJ) and a 96-well microplate fluorometer (Cambridge Technology) were used. In the latter case the filters used for excitation were 482±10 (460±18 for ECFP) and for emission were 532±14. Filters were named as the center wavelength±the half-bandwidth, both in nm. The solutions for cuvette titration contained 125 mM KCl, 20 mM NaCl, 0.5 mM CaCl[0126] 2, 0.5 mM MgCl2, and 25 mM of one of the following buffers—acetate, Mes, Mops, Hepes, bicine, and Tris.
  • EYFP showed an acidification-dependent decrease in the absorbance peak at 514 nm and a concomitant increase in absorbance at 390 nm (FIG. 2A). The fluorescence emission (527-nm peak) and excitation spectra decreased with decreasing pH, but the fluorescence excitation spectrum showed no compensating increase at 390 nm. Therefore, the species absorbing at 390 nm was nonfluorescent. The apparent pKa (pK′a) of EYFP was 7.1 with a Hill coefficient (n) of 1.1 (FIG. 2B) [0127]
  • EGFP fluorescence also was quenched with decreasing pH. The pK′a of EGFP was 6.15, and n was 0.7. [0128]
  • The change in fluorescence of ECFP (Tyr66→Trp in the chromophore) with pH was smaller than that of EGFP or EYFP (pK′a 6.4, n, 0.6) (FIG. 2B) The fluorescence change was reversible in the pH range 5-8.5 for all three proteins, which covers the pH range of most subcellular compartments. These results demonstrate that the GFP variants EGFP, EYFP, and ECFP can be used as fluorescent protein pH sensors. [0129]
  • Example 3 Measurements of pH in the Cytosol and Nucleus Using Fluorescent Protein pH Sensors
  • HeLa cells and AT-20 cells grown on glass coverslips were transiently lipo-transfected (Lipofectin™, GIBCO) with polynucleotide constructs encoding EYFP. [0130]
  • Cells were imaged between 2 and 4 days after transfection at 22° C. with a cooled charge-coupled device camera (Photometrices, Tucson, Ariz.) as described in Miyawaki et al., Nature 388:882, (1997). The interference filters (Omega Optical and Chroma Technology, Brattleboro, Vt.) used for excitation and emission were 440±10 and 480±15 for ECFP; 480±15 and 535±22.5 for EGFP or EYFP. The dichroic mirrors were 455 DCLP for ECFP and 505 DCLP for EGFP or EYFP. Regions of interest were selected manually, and pixel intensities were spatially averaged after background subtraction. A binning of 2 was used to improve signal/noise and minimize photodamage and photoisomerization of EYFP. High KCl buffer plus 5 μM each of the ionophores nigericin (Fluka) and monensin (Calbiochem) was used for in situ titrations in living cells. Cells were loaded with cytosolic pH indicators by incubation with 3 μM carboxy-SNARF/AM or BCECF/AM (Molecular Probes) for 45 minutes, then washed for 30 minutes, all at 22° C. [0131]
  • Fluorescence of HeLa cells transfected with the gene encoding EYFP was diffusely distributed in the cytosol and nucleus. This was expected for a protein of the size of GFP (27 kDa), which is small enough to pass through nuclear pores. [0132]
  • The fluorescence observed with EYFP was reversible. Perfusion with NH[0133] 4Cl caused an increase in fluorescence (rise in pH), which reversed upon washing out the NH4Cl. Conversely, perfusion of lactate, which lowers pH, induced a decrease in fluorescence. The decrease in fluorescence was also reversible on wash-out.
  • Calibration of fluorescence intensity with pH in situ was accomplished with a mix of the alkali cation/H+ ionophores nigericin and monensin in bath solutions of defined pH and high K+. Fluorescence equilibrated within 1-4 minutes after each exchange of solution. These results demonstrate that EYFP, when present intracellularly, can report pH in the physiological range. [0134]
  • Example 4 Measurement of pH in the Mitochondrial Matrix Using Fluorescent Protein pH Sensors
  • To measure pH in the mitochondrial matrix using mutant GFP sensor proteins, HeLa cells and neonatal rat cardiomyocytes were transfected with the fluorescent protein pH sensor EYFP-mito. A Bio-Rad MRC-1000 confocal microscope was used for analysis of the targeted protein. Microscopy analysis revealed that the transfected cells showed a fluorescence pattern indistinguishable from that of the conventional mitochondrial dye rhodamine 123. [0135]
  • In situ pH titration was performed with nigericin/monensin as described in Example 3. Subsequent addition of the protonophore carbonylcyanide m-chlorophenylhydrazone (CCCP) did not change the fluorescence intensity of the cells. This demonstrates that the nigericin/monensin treatment effectively collapsed the pH gradient (ΔpH) in the mitochondria. [0136]
  • The estimated pHm was 7.98±0.07 in HeLa cells (n=17 cells, from six experiments). Similar pH values were obtained in a HeLa cell line stably expressing EYFP-mito. Resting pH did not change by superfusion of cells with medium 10 mM glucose, which would provide cells with an oxidizable substrate, but 10 mM lactate plus 1 mM pyruvate caused an acidification, which reversed on washout. This can be accounted for by diffusion of protonated acid or by cotransport of pyruvate[0137] /H+ through the inner mitochondrial membrane. The protonophore CCCP rapidly induced an acidification of mitochondria to about pH 7.
  • Example 5 Measurement of pH in the Golgi Lumen Using Fluorescent Protein pH Sensors
  • The type II membrane-anchored protein galactosyltransferase (GT:UDP-galactose-β,1,4-galactosyltransferase. EC 2.4.1.22) has been used as a marker of the trans cisternae of the Golgi apparatus (Roth et al., J. Cell Biol. 93:223-29, (1982)). Accordingly, polynucleotide constructs encoding portions of the GT protein fused to the mutant GFP proteins were constructed as described in Example 1 in order to use the GT sequence to target the fluorescent protein pH sensor to the endoplasmic reticulum. [0138]
  • The pH of the Golgi lumen was measured by transfecting HeLa or AT-20 cells with the constructs GT-ECFP, GT-EGFP, or GT-EYFP. Bright juxtanuclear fluorescence was observed, with little increase in diffuse staining above autofluorescnce in most cells. [0139]
  • The fluorescence pattern was examined further in double-labeling experiments using rabbit polyclonal α-mannosidase II (α-manII) antibody. Double labeling fluorescence was performed as described by McCaffery et al., Methods Enzymol. 257:259-279 (1995). The α-manII antibody was prepared as described in Velasco et al., J. Cell Biol. 122:39-51 (1993). In the double-staining experiments, it was observed that labeling of the medial trans-Golgi marker α-manII overlapped with GT-EYFP fluorescence. [0140]
  • α-manII was also fused with ECFP, and the pattern of fluorescence obtained upon transfection of the gene was indistinguishable from that of GT-EYFP by light microcopy. [0141]
  • To identify the subcellular localization of GT-EYFP at higher resolution, immunogold electron microscopy was performed on ultra-thin cryosections by using antibodies against GFP. Immunogold labeling of ultra-thin sections was performed as described by McCaffery et al., supra, using rabbit polyclonal anti-GFP antibody or a monoclonal anti-TGN38 antibody. [0142]
  • In double-labeling experiments, GT-EYFP was found in the medial and trans Golgi, although endogenous GT is present in trans Golgi membranes. The difference in localization may occur as a result of overexpression of the GT-EYFP protein. [0143]
  • When protein TGN38 was used as a trans-Golgi network (TGN) marker, its immunogold localization pattern was found to overlap with that of GT-EYFP in the medial/trans-Golgi membranes. The localization data demonstrate that GT-EYFP labels the medial/trans Golgi. Thus, GT-EYFP can be used to identify the pH of this organelle. [0144]
  • The pH titration of GT-EYFP fluorescence in the Golgi region of the cells after treatment with nigericin/monensin was in good agreement with that of EYFP in vitro (see Example 2). Resting pH in HeLa cells was on average 6.58 (range 6.4-6.81, n=30 cells, 9 experiments). These results also demonstrate that-neither fusion with GT nor the composition of the Golgi lumen affects the pH sensitivity of EYFP. Thus, Golgi-targeted EYFP can be used as a local pH indicator. [0145]
  • The effect of various treatments on the pH of the Golgi was next examined using Golgi-targeted EYFP. [0146]
  • The pH gradient across the Golgi membrane is maintained by the electrogenic ATP-dependent H[0147] + pump (V-ATPase). The V-ATPase generates a ΔpH (acidic inside) and Δψ (positive inside), which opposes further H+ transport. The movement of counter-ions, Cl in (or K+ out), with H+ uptake would shunt the Δψ, allowing a larger ΔpH to be generated. These mechanisms were investigated in intact single HeLa cells transfected with GT-EYFP.
  • The macrolide antibiotic bafilomycin A1 has been shown to be a potent inhibitor of vacuolar type H[0148] + ATPases (V type). In Hela cells expressing GT-EYFP, bafilomycin A1 (0.2 μM) increased pHG by about 0.6 units, to pH 7.16 (range 7.02-7.37, n=12 cells. This suggests that the H+ pump compensates for a positive H+ efflux or leak. The initial rate of Golgi alkalinization by bafilomycin A1 was 0.52 pH units per minute (range 0.3-0.77, n=12 cells), faster than that reported for other acidic compartments such as macrophage phagosomes (0.09 pH/min). Similar results regarding resting pHG and alkalinization by bafilomycin Al were obtained when HeLa cells were transfected with GT-EGFP. Calibration of GT-EGFP in situ also mirrored its in vitro titration (FIG. 1B). Thus, both EGFP and EYFP are suitable Golgi pH indicators.
  • Example 6 Measuring Intracellular pH with Two Fluorescent Protein Sensors
  • Quantitative measurements of fluorescence with nonratiometric indicators can suffer from artifacts as a result of cell movement or focusing. To correct for these effects, the cyan-emitting mutant GT-ECFP was co-transfected into cells along with GT-EYFP. ECFP has excitation and emission peaks that can be separated from those of EYFP by appropriate filters. In addition, ECFP is less pH-sensitive than EYFP (see FIG. 2B). [0149]
  • FIG. 3A demonstrates that the fluorescence of ECFP changed less than that of EYFP during the course of the experiment. Although the ratio of EYFP to ECFP emission varied between cells, probably reflecting a different concentration of GT-EYFP and GT-ECFP in the Golgi lumen, it changed with pH as expected (FIG. 3B). Bafilomcin A1 raised the GT-EYFP/GT-ECFP emission ratio, i.e, it raised pH[0150] G.
  • Example 7 Construction of YFP THR H148G and YFP H1480 Mutants
  • The YFP H148G mutant was prepared using as a template a nucleic acid encoding the YFP mutation 10c, which includes the mutations S65G/V68L/S72A/Q80R/T203Y and is described in Ormö et al., [0151] Science 273:1392-95 (1997). The YFP H148G mutant was constructed using the PCR-based QUIKCHANGE™ Site-Directed Mutagenesis Kit (Stratagene, La Jolla, Calif.) following the manufacturer's instructions. The YFP H148Q mutation was similarly constructed from a nucleic acid encoding the 10C mutation.
  • The pKa of the YFP H148G mutant was found to be 8.0, while the YFP H148Q mutant was found to have a pKa of 7.5. [0152]
  • Example 8 Expression of Mito-YFP H148G in the Mitochondrial Matrix at a pH Range of 7.0 to 8.4
  • The high pKa of the mutant YFP H148G allows it the to be used for the precise measurement of mitochondrial matrix pH both in cells at rest and in cells subject to manipulations that decrease mitochondrial pH. [0153]
  • This was demonstrated directly by transfecting a nucleic acid encoding mito-YFP H148G into HeLa cells using the procedures described in Example 4. YFP H148G expression was monitored by observing fluorescence over time. Mitochondrial pH was also monitored by pH titration as described in Example 3 using nigericin and monensin. [0154]
  • FIG. 4 shows that HeLa cells transfected with YFP H148G in the mitochondrial matrix were fluorescent at an initial pH of 8.0 to 8.1 (where measurements began at t≈0 seconds). 5 μM CCCp was added at about t≈300 seconds. Although addition of 5 μm CCCP rapidly lowered the pH to 7.0, fluorescence of mito-YFP H148G was still detectable. Then a calibration was performed by perfusing the cells with extracellular medium of [0155] ph 7, 7.5, 8, and 8.35 containing the ionophores nigericin plus monensin to equilibrate mitochondrial pH and extracellular pH. Fluorescence in mitochondria increased stepwise with each change of extracellular pH.
  • Fluorescence was also examined, and pH measured, in primary cultures of chick skeletal myotubes transfected with the mito-YFP H148G mutant. FIG. 5 demonstrates that fluorescence was detectable in the mitochondrial matrix of chicken skeletal myotubes, which had a pH of 8.0-8.1 (t≈0). Fluorescence was still detectable following addition of 25 μM forskolin, which did not affect the pH, and after addition of 2 μM CCCP at t≈750 seconds, although CCCP caused the pH to rapidly drop to 6.9 at t≈1400 seconds. Thereafter fluorescence continued to be observed during calibration at ph 6.9, 7.6 and 8.0. [0156]
  • These results demonstrate mito-YFP H148G fluorescence is detectable in the mitochondrial matrix over the pH range of 7.0 to 8.4 in both established cell lines (HeLa cells) and primary cultures (chick skeletal myotubes). [0157]
  • Example 9 Expression of Mito-YFP H148Q in Response to PH Changes
  • The YFP H148Q mutant has a pKa of about 7.4, which is intermediate between the pKa of EYFP and YFP mutant H148G. To demonstrate that YFP H148Q can also be used to measure mitochondrial matrix pH, a nucleic acid encoding mito-YFP-H148Q was transfected into HeLa cells. Fluorescence was measured over time (beginning at t≈0), including following the addition of 10 μM nigericin in high KCL titration buffer at t≈500 seconds. [0158]
  • FIG. 6 reveals the effect of changing mitochondrial pH to 6.9, 7.5, 8, and 8.4 with the ionophore nigericin on fluorescence intensity. Fluorescence decreased to about 175 units at t=1000 seconds by addition of nigericin, which lowered the pH to about 6.9. Fluorescence then returned stepwise to 400 units with each change of the extracellular medium. These results demonstrate that the fluorescence of the mito-YFP H148Q mutant can be used to measure the pH of the inner mitochondrial matrix. [0159]
  • Example 10 Measuring Intracellular pH by Coexpression of YFP H148G and a Second PH-Insensitive Sensor
  • As is discussed above in Example 6, for quantitative measurements it is desirable to compute the fluorescence of the sensor used to measure pH with the fluorescence of a second sensor molecule whose fluorescence does not change over the pH range being tested. A ratiometric measurement is useful to correct for movement or focusing artifacts that may occur during live cell imaging experiments. [0160]
  • To identify a GFP sensor protein suitable for use as a reference protein for measuring mitochondrial matrix pH, the GFP mutant T203I was expressed in the mitochondria of HeLa cells. The GFP T203I mutant can be excited with light of 400 nm, which does not appreciably excite the pH sensitive YFP mutants. [0161]
  • Fluorescence of HeLa cells transfected with the GFP T2031 mutant was monitored for about 400 seconds using an excitation ratio of 480 nm/400 nm. 10 μM CCCP was then added to the cells, and fluorescence was monitored for an additional 250 seconds. Addition of CCCP did not affect fluorescence. In control experiments, it was observed that addition of CCCP corresponded to a drop in pH of about 1 unit. Thus, the GFP T203I mutant is suitable for use as a reference, pH-insensitive mutant. [0162]
  • HeLa cells were then transfected with the GFP T203I mutant and YFP H148G. FIG. 7 shows the change of mitochondrial pH with oligomycin and the uncoupler CCCP as the ratio of YFP H148G emission and GFP T203I emission, with excitation of 490 and 400 nm, respectively. [0163]
  • Example 11 Structural Characterization of YFP T203Y/S65G/V68L/S72A/H148G
  • The green fluorescent protein (GFP) from the jellyfish [0164] Aequorea victoria has been used extensively in molecular biology as a fluorescent label. The structures of WT GFP (Yang et al., Nature Biotech. 14:1246-51, 1996; Brejc et al., Proc. Natl. Acad. Sci. USA. 94:2306-11, 1997) and the variant S65T were determined in 1996 (Ormö et al., Science 273:1392-95, 1996).
  • A large number of mutants have been identified that exhibit broadly varying absorption and emission maxima (Heim et al. above; Heim et al., Curr. Biol. 6: 178-82, 1996). The yellow fluorescent protein (YFP) mutant is of particular interest since its spectrum is shifted enough to render it readily distinguishable from the spectrum of Cyan Fluorescent Protein (CFP) for FRET measurements (Tsien, Ann. Rev. Biochem. 67:509, 1998; Miawaki et al., Nature 388:882-87, 1997). WT GFP exhibits two absorption maxima, where the major band absorbs at 398 nm and the minor band at 475 nm (Morise et al., Biochemistry 13:2656-62, 1974). Excitation of either of these bands leads to emission of green light with a maximum between 504 and 508 nm (FIG. 8). Before a structure was available, GFP variants with altered spectral characteristics were identified by random mutagenesis. Some of these mutants, such as Y66H and Y66W (Tsien, Ann. Rev. Biochem. 67:509, 1998, Heim et al., Proc. Natl. Acad. Sci. (USA) 91:12501-04) result in blue-shifted absorbance and emission maxima. Others focus on changes in the immediate environment of the chromophore π system, such as S65T (Heim et al., Nature 373: 663-64, 1995) and T203I (Heim et al., Proc. Natl. Acad. Sci. (USA) 91:12501-04, 1994). At physiological pH, S65T exhibits only one major absorption band at 489 nm, red-shifted by 14 nm from WT GFP, and is almost six times brighter (Heim et al., Nature 373: 663-64, 1995). Yet, the emission spectrum is shifted by only 3 nm to 511 nm, and so cannot easily be distinguished from the wild-type emission. Random mutagenesis techniques produced only one further red-shifted variant, S65T/M153A/K238E, which increases the excitation and emission wavelengths of S65T by 15 and 3 nm respectively (Heim et al., Curr. Biol. 6: 178-82, 1996). Here is described crystal structures of the first set of GFP variants rationally designed based on the x-ray structure of GFP S65T (Ormö et al., Science 273:1392-95, 1996). These variants, termed YFPs (Yellow Fluorescent Proteins), exhibit the longest wavelength emissions of all GFPs generated by mutagenesis (FIG. 8). The YFPs fluoresce around 528 nm, red-shifted by 16 nm as compared to S65T and are easily distinguishable from S65T on a fluorescence microscope. [0165]
  • The specific YFP investigated is the quadruple-mutant T203Y/S65G/V68L/S72A, where the substitution T203Y was introduced based on the structural considerations detailed below and is believed responsible for the red-shift. The other three mutations have been shown to improve its brightness in live cells (Cormack et al., Gene 173:33, 1996). The T203Y mutation would have been difficult to identify by random mutagenesis since this amino acid substitution requires three substitutions at the nucleotide level. Since Thr203 is positioned close to the chromophore, it was postulated that its replacement with a tyrosine would result in π-stacking interactions between the chromophore and the highly polarizable phenol (Ormö et al., Science 273:1392-95, 1996), leading to red-shifted spectral properties. The structure of S65T suggested that an aromatic amino acid introduced in position 203 would extend into the water-filled cavity adjacent to the chromophore (Ormö et al., Science 273:1392-95, 1996). Replacement of Thr203 with any of the aromatic amino acids His, Trp,Tyr, or Phe was found to lead to the desired spectral shifts (Ormö et al., Science 273:1392-95, 1996, Dickson et al., Nature 388:355-58, 1997). The most dramatic red-shift was observed for the T203Y substitution, therefore this variant has been termed YFP. [0166]
  • In order to determine the role of His148 in modulating the pKa of the chromophore or its spectral properties, an additional mutation, H148G, was introduced into the YFP background. The x-ray structures of YFP and YFP H148G were analyzed in order to better correlate structural changes with spectral properties. GFP variants were prepared as described in Example 6, above. This template incorporates the mutations T203Y/S65G/V68L/S72A, as well as the ubiquitous Q80R substitution that was accidentally introduced into the gfp cDNA early on (Ormö et al., Science 273:1392-95, 1996; Chalfie et al., Science 263:802-05, 1994). All GFP variants were expressed and purified as described (Ormö et al., Science 273:1392-95, 1996). [0167]
  • Structural Determination of YFP H148G [0168]
  • YFP H148G was concentrated to 12 mg/ml in 20 mM HEPES pH 7.9. Rod-shaped crystals with approximate dimensions of 1.8×0.08×0.04 mm were grown in hanging drops containing 2 μl protein and 2 μl mother liquor at 4° C. within four days. The mother liquor contained 16[0169] % PEG 4000, 50 mM sodium acetate pH 4.6, and 50 mM ammonium acetate. X-ray diffraction data were collected from a single crystal at room temperature using a Xuong-Hamlin area detector (Hamlin, Methods. Enzymol. 114:416-52, 1985). Data were collected in space group P212121, to 99% completeness at 2.6 Å resolution, and reduced using the supplied software (Howard et al., Methods Enzymol. 114:452-71, 1985). Unit cell parameters were a=52.0, b=62.7, and c=69.9. The GFP S65T coordinate file (Ormö et al., Science 273:1392-95, 1996) which-served as a model for phasing was edited to reflect the mutations, with the introduced residues Tyr203 and Leu68 initially modeled as alanines to prevent model bias. A model for the anionic chromophore was obtained by semi-empirical molecular orbital calculations using AM1 in the program SPARTAN version 4.1 (Wavefunction Inc., Irvine, Calif.). The minimized structure, which was planar, compared very favorably with a related small molecule crystallographic structure (Tinant et al., Cryst. Struct. Comm. 9:671-74, 1980), and also with the model used during refinement of GFP S65T, where a simpler modeling program had been employed (Ormö et al., Science 273:1392-95, 1996).
  • Using the program TNT (Tronrud et al., Acta Crystallogr. Sect. A 43:489, 1987), rigid body refinement was carried out to position the isomorphous model in the unit cell of YFP H148G. Initial positional refinement was carried out using the data to 4.0 Å, then to 3.5, 3.0, and finally to 2.6 Å. Electron density maps (2Fo-Fc and Fo-Fc) were inspected using O (Tronrud et el., above), and solvent molecules were added if consistent with Fo-Fc features, and only when in proximity of hydrogen bond partners. B-factors were refined using a strong correlation between neighboring atoms due to the relatively low resolution. Since no B-factor library is available for the chromophore itself, the B-factors of all chromophore atoms were set to the values obtained in the 1.9 Å structure of GFP S65T (Ormö et al., Science 273:1392-95, 1996), and then refined as a group, with identical shifts for the grouped atoms. [0170]
  • Structure Determination of YFP [0171]
  • YFP was concentrated to 10 mg/ml in 50 mM HEPES pH 7.5. After 2 weeks crystals grew to a size of 0.03×0.12×0.8 mm at 15° C. in hanging drops containing 5=|l protein and 5=|l well solution, which contained 2.2 M sodium/potassium phosphate pH 6.9. These crystals belong to space group P2[0172] 1212 and have the unit cell dimensions a=77.1, b=117.4, w and c=62.7. X-ray diffraction data were collected on two isomorphic crystals at room temperature using an Raxis-IV imaging plate mounted on a Rigaku RUH3 rotating anode generator equipped with mirrors. The data were processed with Denzo and scaled using ScalePack (Otwinowski et al., Methods Enzymol. 276:307-26, 1997). The YFP structure was solved by molecular replacement using the program AMoRe (Navaza, Acta Crystallogr. A50:157-63, 1994), with the 1.9 Å GFP S65T coordinate file as the search model (Ormö et al., Science 273:1392-95, 1996). Two solutions were identified, consistent with two molecules per asymmetric unit.
  • For refinement, the 2.6 Å structure of YFP H148G was chosen as the initial model, which was edited to reflect the mutations present in YFP. To avoid model bias, the occupancies of the Tyr203 side chain atoms and all chromophore atoms were set to zero during the first several rounds of refinement. Constrained NCS averaging over the A and B chains in the asymmetric unit was applied, initial refinement was carried out to 3.5 Å only, and the electron density maps (2Fo-Fc and Fo-Fc) were averaged. These maps were then inspected, and the model adjusted using O (Jones et al., Acta Crystallogr. Sect. A 47:110, 1991), followed by additional positional refinement to 2.5 Å. Chromophore and Tyr203 densities were very clear, and both were planar. The model was edited to include these groups in refinement, and solvent molecules were added where appropriate. B-factors were refined using a strong correlation between neighboring atoms due to the relatively low resolution. [0173]
  • Comparison of the Structure of YFP and YFP H148G [0174]
  • YFP crystallized in 2.2 M Na/K phosphate at pH 6.9 in spacegroup P2[0175] 1212, with 2 molecules per asymmetric unit (chains A and B). The GFP S65T structure was used as a search model for molecular replacement against a 3.0 Å dataset using the program AMoRe (Navaza, Acta Crystallogr. A50:157-63, 1994), and the structure was refined. Later, the refined structure of YFP H148G (see below) was used for phasing and refinement against a 2.5 Ådataset. Even though the introduced Tyr203 and the chromophore itself were not modeled during early cycles of refinement, clear electron density for a planar chromophore and a stacked Tyr203 phenol was immediately apparent. Non-crystallographic symmetry (NCS) constraints were employed throughout refinement of the model using TNT, and maps were averaged. At the end of refinement, non-averaged maps for the A-and B-chain in the asymmetric unit were calculated and compared to each other. No obvious features were identifiable that would suggest significant differences between the two chains. A test run of refinement without any NCS constraints confirmed that the differences would be smaller than the rms error of a 2.5 Å structure. Therefore, the NCS constraints were not relaxed or eliminated. Data collection and atomic model statistics are shown in Table 33. The final R-factor of the YFP model was 19.2% for all data between 20 and 2.5 Å resolution.
    TABLE 33
    Data collection and atomic model statistics of
    YFP and YFP H148G.
    YFP
    YFP H148G
    Total observations 53,039 29,904
    Unique reflections 18,916 7,373
    Completenessa 92% 99%
    Completeness (shellb) 94% 97%
    Number of crystals 2 1
    Rmergec (%) 8.0% 6.5%
    Resolution 2.5 Å 2.6 Å
    Atomic model statistics:
    Spacegroup P212121
    P212121
    Molecules per asymm, unit 2 1
    Crystallographic R-factor 0.192 0.159
    Protein atoms 1,810 1,810
    Solvent atoms per asymmm, unit 130 30
    Bond length deviations (Å) 0.013 0.012
    Bond angle deviations (°) 1.76 2.07
    Thermal parameter restraints (Å2) 4.53 3.82
  • The refined YFP structure clearly shows that the overall fold is undisturbed, with an rms deviation from the GFP S65T structure of 0.36 Å for α-carbons. Three larger contact areas with adjacent molecules were identified. The largest of these covers about 722 Å2 of one monomer surface, includes a series of hydrophobic residues consisting of Ala206, Phe223, and Leu221, and also a number of hydrophilic contacts. This interface is essentially identical to the dimer interface for WT GFP described by Yang et al., Nature Biotech. 14:1246-51, 1996. High salt conditions during crystallization experiments appear to favor dimerization, as has been suggested previously (Palm et al., Nat. Struct. Biol. 4:361-65, 1997). [0176]
  • YFP H148G crystallized as a monomer in the presence of polyethylene glycol and acetate at pH 4.6 in spacegroup P2[0177] 12121, isomorphous to S65T (Ormö et al., Science 273:1392-95, 1996) and the blue-emission variant BFP (Wachter et al., Biochemistry 36:9759-65, 1997). Molecular replacement using the S65T structure for phasing and refinement gave a final model with an R-factor of 15.9% for all data between 24.0 and 2.6 Å (Table 33). As with YFP, electron density for a stacked phenol was clearly visible even before the Tyr203 ring was modeled. The rms deviation between YFP H148G and S65T a-carbons is 0.31 Å, and the deviation between YFP H148G and YFP a-carbons is 0.35 Å. The b-strands of the two YFP variants overlay closely in all areas except around the Cα of residue 148 where a movement of 1.1 Å is observed. This movement has not been observed in other pH 4.6 structures grown under similar conditions and crystallizing in the same space group, such as the BFP structure (Wachter et al., Biochemistry 36:9759-65, 1997). Residue 148 and adjacent residues are not involved in crystal contacts, further indicating that the observed movement is due to the H148G substitution, not crystallization conditions.
  • π-Stacking of the Introduced Phenol [0178]
  • Electron densities of the chromophore and the phenol ring of Tyr203 appeared to be completely planar before the atoms for these groups were added to the model. When the tyrosine side chain was first introduced into the model, it was modelled as co-planar to the chromophore. Refinement consistently rotated the phenol ring by 12 with respect to the chromophore plane in both YFP and YFP H148G. FIG. 9 shows the electron density of the refined YFP chromophore structure together with the phenol ring of Tyr203. The distance of the closest approach between atoms of the two interacting rings is 3.3 to 3.4 Å, and occurs at that edge of the chromophore plane that is opposite the exo-methylene bond (FIG. 9). It appears that the phenol tilts towards this area of the chromophore since it is more open, with fewer atoms to clash with sterically. [0179]
  • The distance of largest separation between the rings is 3.5 to 3.8 Å, and occurs at the opposite edge, where steric clash with the exo-methylene carbon could occur. This range of plane-to-plane distances is typical for face-to-face π to π stacking interactions found in proteins, and consistent with interaction energy calculations that show a potential energy minimum for two horizontally stacked benzene molecules with a vertical separation of 3.3 Å (Burley et al., J. Am. Chem. Soc. 108, 7995-8001, 1986). A recent analysis of protein structures has led to the conclusion that aromatic ring interactions in an off-centered parallel orientation have an energetically favorable, stabilizing effect, and in fact are the preferred interactions (McGaughey et al., J. Biol. Chem. 273, 15458-63, 1998). [0180]
  • Positional Shift of the Chromophore [0181]
  • The entire chromophore ring system of YFP has moved out towards the protein surface by about 0.9 Å when compared to S65T or WT GFP. The chromophore of YFP H148G has Moved in the same direction but to a lesser extent, about 0.5 Å. Overlay of all α-carbons shows that this shift is very much a local effect, only involving residues 65 to 68. The overlay suggests that this shift may be due to the compensating effects of the V68L and S65G substitutions. The Leu68 Cδ1 occupies the same space as the original Val68 Cγ1, whereas the Leu68 backbone is displaced so that the chromophore is pushed further out towards the protein surface. As part of the same movement, the Cα of Gly65 is pushed into the position of the wild-type C[0182] β of Ser65. The V68L and S65G substitutions had been previously found to significantly increase the brightness of GFP-expressing cells (Cormack et al., Gene 173:33, 1996) in a WT background, and were suggested to either improve folding at 37° or increase the rate of chromophore formation. It is unclear at this point why the chromophore is not shifted to the same extent in the YFP and YFP H148G structures, though both of them incorporate the V68L and S65G mutations.
  • Even though the imidazolinone ring of the YFPs is not in the same position as in WT GFP (Brejc et al., Proc. Natl. Acad. Sci. USA. 94:2306-11, 1997), S65T (Ormö et al. Science 273:1392-95, 1996), and blue-fluorescent protein BFP (Wachter et al., Biochemistry 36:9759-65, 1997), no electron density consistent with partially formed or unformed chromophore is observed. This indicates that the machinery to generate the chromophore is not only intact, but more flexible than previously thought. Apparently, the exact positions of the backbone atoms of residues 65 and 67 that undergo the cyclization reaction is not as crucial as was previously suggested, based on the nearly exact superposition of the imidazolinone rings observed in WT GFP, S65T, and BFP (Yang et al., Nature Biotech. 14:1246-51, 1996; Brejc, K. et al., Proc. Natl. Acad. Sci. USA 94:2306-2311, 1997; Ormö et al., Science 273:1392-95, 1996; Palm et al., Nat. Struct. Biol. 4:361-65, 1997; Wachter et al., Biochemistry 36:9759-65, 1997). [0183]
  • Chromophore Spectral Properties, Charge State and Hydrogen Bonding Interactions [0184]
  • The spectral properties of the YFPs were examined. Small aliquots of protein (16 mg/ml) were diluted 48-fold into 75 mM buffer (acetate, phosphate, Tris, or CHES), 140 mM NaCl, and then scanned for absorbance between 250 and 600 nm (Shimadzu 2101 spectrophotometer at medium scan rate and room temperature). The optical density at 514 or 512 nm was plotted as a function of pH and computer-fitted to a Nitration curve (Kaleidagraph™, SynergySoftware). [0185]
  • Fluorescence measurements were carried out on a Hitachi F4500 fluorescence spectrophotometer at a constant protein concentration of approximately 0.01 mg/ml, with buffer conditions identical to those of absorbance measurements. The excitation wavelength was set to the absorbance maximum of the long-wave band of the particular mutant. The emission was scanned between 500 and 600 nm, and peak emission intensity was plotted as a function of pH and curve-fitted. [0186]
  • Like S65T (Kneen et al., Biophys. J. 74:1591-99, 1998), the YFPs have two absorbance maxima whose relative ratio is pH-dependent (FIG. 8 and Table 34). The UV absorption peaks at 392 (YFP) or 397 nm (YFP H148G) have been ascribed to the neutral chromophore, whereas the visible absorption peaks at 514 (YFP) or 512 nm (YFP H148G) have been ascribed to the anionic chromophore (Niwa et al., Proc. Natl. Acad. Sci. (USA) 93: 13617-22, 1996). The lower energy peak exhibits clear vibrational structure as indicated by the pronounced shoulder at 480-490 nm, and its mirror-image relationship with the emission band is striking (FIG. 8). These features are consistent with luminescence properties of large and rigid systems in condensed phases (Barltrop et al., Principles of Photochemistry, John Wiley and Sons, New York, 1978, pp. 51-52 and 78-79), and may be more pronounced in the YFPs due to decreased chromophore flexibility in the presence of the stacked phenol. Both YFPs fluoresce intensely when excited at the longer-wavelength band, with maximum emission occurring at 528 nm (FIG. 8). Fluorescence is extremely weak when the excitation occurs at the shorter-wavelength band (Table 34), even if the experiment is carried out at a pH where this peak dominates. The chromophore pKa in the intact protein was determined to be 7.00(±0.03) for YFP and 8.02 (±0.01) for YFP H148G by absorbance measurements at varying pH. The pKa values determined by fluorescence were 6.95 (±0.03) and 7.93 (±0.04), respectively, for the two variants. The YFP pKa is remarkably similar to that of [0187]
    TABLE 34
    Summary of Absorption and Emission Maxima.
    absorbance absorbance emissiona emissiona
    band #1 band #2 band band
    WT GFP 398 475 460/598 504
    S65T 394 489 (weak) 511
    YFP 392 514 (weak) 528
    HFP H148G 397 51122 (weak) 528
  • EYFP (S65G/S72A/T203Y/H231L). All titration curves gave an excellent fit to a single pKa value. [0188]
  • It is likely that the charge state of the chromophore is mixed in the YFP crystals which were grown at [0189] pH 7, and which is the chromophore pKa. In YFP, His148 is directly hydrogen-bonded to the phenolic end of the chromophore. Its electron density is well-defined, suggesting that the imidazole ring does not change position when the chromophore ionizes. It is therefore unlikely that structural rearrangements in the immediate chromophore environment occur in response to changes in chromophore charge state. In both the YFP and YFP H148G structures, the phenolic end of the chromophore is nearly in H-bonding contact with bulk solvent via two ordered waters, and therefore may not be as tightly embedded in the protein as in WT and S65T (Brejc et al., Proc. Natl. Acad. Sci. USA. 94:2306-11, 1997, Ormö et al., Science 273:1392-95, 1996]. Structural readjustments to accommodate the anion may only affect solvent molecules.
  • The strong hydrogen bond to Arg96 that has been suggested to play a role in the chemistry of backbone cyclization (Ormö et al., above) is maintained in both structures. The carbonyl oxygen of the chromophore imidazolinone ring interacts with two hydrogen bond donors, Arg96 and Gln69 in YFP, and Arg96 and Gln94 in YFP H148G. This compares to similar interactions with Arg96 and Gln94 in WT and S65T. The Glu222 carboxy oxygen approaches the chromophore imidazolinone ring nitrogen to within 3.0 (YFP) and 3.3 Å (YFP H148G), considerably closer than in WT and S65T (4.3 and 4.0 Å, respectively). This close approach appears to be related to the chromophore positional shift described above. Distance and geometry for hydrogen bonding between Glu222 and the chromophore ring nitrogen are excellent in YFP, and somewhat less optimal in YFP H148G, where the presumed H-bond makes roughly a 45° angle with the chromophore plane. The YFP structure is the first GFP structure solved that suggests H-bonding interactions of the heterocyclic ring nitrogen originating from Tyr66. The most likely interpretation in terms of charge states is a deprotonated ring nitrogen and a protonated Glu222, rendering both groups neutral, however, it is clear that they share a proton. [0190]
  • Solvent-Accessible Surface and Cavities [0191]
  • The mutation H148G was introduced into YFP to examine the effects of solvent accessibility on the fluorescent properties and the ionization constant of the chromophore. In all GFP structures examined to date, the β-barrel is somewhat perturbed around the phenolic end of the chromophore. The β-strand that covers the chromophore in that area bulges out around His148, so that the backbone from residue 144 to 150 is not directly hydrogen-bonded to the adjacent backbone between residues 165 and 170. Rather, they are laced together by forming H-bonds with the imidazole ring of His148 (Arg168 backbone N to His148 N[0192] ε2 in S65T and WT GFP) and several water molecules. The phenolic end of the chromophore is located directly “behind” the ring of His148. It was anticipated that substitution of His with Gly would open up a solvent channel to the chromophore in the absence of other structural perturbances, or perhaps to permit the bulge to close.
  • The crystal structure clearly shows this anticipated solvent channel as an invagination of the protein surface with no ordered solvent molecules within. Elimination of the imidazole ring in the H148G substitution only leads to minor structural rearrangements of protein groups. The β-strands do not close up to form a directly H-bonded sheet between [0193] residues 144 and 150. Instead, the Cα of residue 148 has actually moved in the opposite direction by 1.1 Å, causing an even larger strand separation between the backbones of residues 148 and 168. The side chain of Ile167 has moved by 1.1 Å towards the space previously occupied by the imidazole ring. Nevertheless, direct solvent access to the phenolic end of the chromophore is greatly improved. Calculation of the solvent-accessible area of the chromophore using a probe sphere of radius of 1.4 Å (Connolly, Science 221:709-13, 1983), as implemented by UCSF MidasPlus
    Figure US20030212265A1-20031113-P00900
    (UCSF MidasPlus®, Computer Graphics Laboratory, University of San Francisco, Calif. 94143), shows that 22% of the chromophore surface is solvent-accessible only the phenolic end of the chromophore is exposed to exterior solvent, though, due to the opening in the protein wall. The phenolic oxygen of the chromophore is also hydrogen-bonded to a water molecule that is near H-bonding distance to a surface water, though a 1.4 Å probe cannot access the chromophore via this path. If both the solvent channel as well as this hydrogen bond are included, 8% of the chromophore surface is accessible to exterior solvent, entirely at the phenolic end, and 14% is accessible to interior solvent due to contact with internal cavities.
  • YFP H148G contains two larger interior cavities that are in contact with the chromophore cavity and filled with some ordered waters. The cavity that was largest in S65T has decreased in size from approximately 127 Å3 (S65T) to 88 Å3 (YFP H148G), because some of the space is now filled with the phenol of Tyr203. In YFP, this cavity is not accessible to a 1.4 Å probe at all since several groups have moved into this space. The more significant structural adjustments are C[0194] γ2 of Val224, which has moved by 1.4 Å, and Cδ1 of Leu42 which has moved by 2.0 Å, essentially filling the cavity. The second larger cavity in contact with the chromophore is nearly invariant for S65T, YFP, and YFP H148G, and is between 98 and 103 Å3 in size.
  • Solvent Accessibility to the Chromophore [0195]
  • YFP H148G was found to be highly fluorescent, with bright greenish-yellow color under ordinary day light. The light-emitting properties of the fluorophore do not appear to be changed to any extent by the introduction of a solvent channel to the chromophore, indicating that significant quenching does not occur. [0196]
  • Since the protein fold is entirely intact in YFP H148G in spite of the generation of an opening in the β-barrel, the H148G substitution may be especially useful for allowing access of various small-molecule species to the chromophore. This substitution may be introduced into other GFP variants with a larger cavity adjacent to the chromophore, such as S65T [7] or S65G, allowing for analyte binding studies where specific spectral shifts due to the interaction with small molecules or ions of interest could be monitored. The highest ionization constant of all variants examined to date is found for the YFP H148G mutant with a pKa of 8.0. In this mutant, the chromophore is solvent exposed, consistent with a similarly high pKa when the protein is denatured (Nageswara et al., Biophys. J. 32:630-32, 1980). [0197]
  • Other Embodiments
  • It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims. [0198]
  • 1 38 1 716 DNA Aequorea victoria CDS (1)...(714) Green fluorescent protein 1 atg agt aaa gga gaa gaa ctt ttc act gga gtt gtc cca att ctt gtt 48 Met Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu Val 1 5 10 15 gaa tta gat ggt gat gtt aat ggg cac aaa ttt tct gtc agt gga gag 96 Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly Glu 20 25 30 ggt gaa ggt gat gca aca tac gga aaa ctt acc ctt aaa ttt att tgc 144 Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile Cys 35 40 45 act act gga aaa cta cct gtt cca tgg cca aca ctt gtc act act ttc 192 Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr Phe 50 55 60 tct tat ggt gtt caa tgc ttt tca aga tac cca gat cat atg aaa cgg 240 Ser Tyr Gly Val Gln Cys Phe Ser Arg Tyr Pro Asp His Met Lys Arg 65 70 75 80 cat gac ttt ttc aag agt gcc atg ccc gaa ggt tat gta cag gaa aga 288 His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu Arg 85 90 95 act ata ttt ttc aaa gat gac ggg aac tac aag aca cgt gct gaa gtc 336 Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu Val 100 105 110 aag ttt gaa ggt gat acc ctt gtt aat aga atc gag tta aaa ggt att 384 Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly Ile 115 120 125 gat ttt aaa gaa gat gga aac att ctt gga cac aaa ttg gaa tac aac 432 Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr Asn 130 135 140 tat aac tca cac aat gta tac atc atg gca gac aaa caa aag aat gga 480 Tyr Asn Ser His Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn Gly 145 150 155 160 atc aaa gtt aac ttc aaa att aga cac aac att gaa gat gga agc gtt 528 Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser Val 165 170 175 caa cta gca gac cat tat caa caa aat act cca att ggc gat ggc cct 576 Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly Pro 180 185 190 gtc ctt tta cca gac aac cat tac ctg tcc aca caa tct gcc ctt tcg 624 Val Leu Leu Pro Asp Asn His Tyr Leu Ser Thr Gln Ser Ala Leu Ser 195 200 205 aaa gat ccc aac gaa aag aga gac cac atg gtc ctt ctt gag ttt gta 672 Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe Val 210 215 220 aca gct gct ggg att aca cat ggc atg gat gaa cta tac aaa 714 Thr Ala Ala Gly Ile Thr His Gly Met Asp Glu Leu Tyr Lys 225 230 235 ta 716 2 238 PRT Aequorea victoria 2 Met Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu Val 1 5 10 15 Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly Glu 20 25 30 Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile Cys 35 40 45 Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr Phe 50 55 60 Ser Tyr Gly Val Gln Cys Phe Ser Arg Tyr Pro Asp His Met Lys Arg 65 70 75 80 His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu Arg 85 90 95 Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu Val 100 105 110 Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly Ile 115 120 125 Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr Asn 130 135 140 Tyr Asn Ser His Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn Gly 145 150 155 160 Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser Val 165 170 175 Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly Pro 180 185 190 Val Leu Leu Pro Asp Asn His Tyr Leu Ser Thr Gln Ser Ala Leu Ser 195 200 205 Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe Val 210 215 220 Thr Ala Ala Gly Ile Thr His Gly Met Asp Glu Leu Tyr Lys 225 230 235 3 239 PRT Aequorea victoria VARIANT (0)...(0) EGFP 3 Met Val Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu 1 5 10 15 Val Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly 20 25 30 Glu Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile 35 40 45 Cys Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr 50 55 60 Leu Thr Tyr Gly Val Gln Cys Phe Ser Arg Tyr Pro Asp His Met Lys 65 70 75 80 Gln His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu 85 90 95 Arg Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu 100 105 110 Val Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly 115 120 125 Ile Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr 130 135 140 Asn Tyr Asn Ser His Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn 145 150 155 160 Gly Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser 165 170 175 Val Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly 180 185 190 Pro Val Leu Leu Pro Asp Asn His Tyr Leu Ser Thr Gln Ser Ala Leu 195 200 205 Ser Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe 210 215 220 Val Thr Ala Ala Gly Ile Thr Leu Gly Met Asp Glu Leu Tyr Lys 225 230 235 4 239 PRT Aequorea victoria VARIANT (0)...(0) EYFP 4 Met Val Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu 1 5 10 15 Val Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly 20 25 30 Glu Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile 35 40 45 Cys Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr 50 55 60 Phe Gly Tyr Gly Val Gln Cys Phe Ala Arg Tyr Pro Asp His Met Lys 65 70 75 80 Gln His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu 85 90 95 Arg Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu 100 105 110 Val Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly 115 120 125 Ile Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr 130 135 140 Asn Tyr Asn Ser His Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn 145 150 155 160 Gly Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser 165 170 175 Val Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly 180 185 190 Pro Val Leu Leu Pro Asp Asn His Tyr Leu Ser Tyr Gln Ser Ala Leu 195 200 205 Ser Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe 210 215 220 Val Thr Ala Ala Gly Ile Thr Leu Gly Met Asp Glu Leu Tyr Lys 225 230 235 5 239 PRT Aequorea victoria VARIANT (0)...(0) EYFP-V68L/Q69K 5 Met Val Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu 1 5 10 15 Val Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly 20 25 30 Glu Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile 35 40 45 Cys Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr 50 55 60 Phe Gly Tyr Gly Leu Lys Cys Phe Ala Arg Tyr Pro Asp His Met Lys 65 70 75 80 Gln His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu 85 90 95 Arg Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu 100 105 110 Val Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly 115 120 125 Ile Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr 130 135 140 Asn Tyr Asn Ser His Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn 145 150 155 160 Gly Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser 165 170 175 Val Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly 180 185 190 Pro Val Leu Leu Pro Asp Asn His Tyr Leu Ser Tyr Gln Ser Ala Leu 195 200 205 Ser Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe 210 215 220 Val Thr Ala Ala Gly Ile Thr Leu Gly Met Asp Glu Leu Tyr Lys 225 230 235 6 239 PRT Aequorea victoria VARIANT (0)...(0) ECFP 6 Met Val Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu 1 5 10 15 Val Glu Leu Asp Gly Asp Val Asn Gly His Arg Phe Ser Val Ser Gly 20 25 30 Glu Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile 35 40 45 Cys Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr 50 55 60 Leu Thr Trp Gly Val Gln Cys Phe Ser Arg Tyr Pro Asp His Met Lys 65 70 75 80 Gln His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu 85 90 95 Arg Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu 100 105 110 Val Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly 115 120 125 Ile Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr 130 135 140 Asn Tyr Ile Ser His Asn Val Tyr Ile Thr Ala Asp Lys Gln Lys Asn 145 150 155 160 Gly Ile Lys Ala His Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser 165 170 175 Val Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly 180 185 190 Pro Val Leu Leu Pro Asp Asn His Tyr Leu Ser Thr Gln Ser Ala Leu 195 200 205 Ser Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe 210 215 220 Val Thr Ala Ala Gly Ile Thr Leu Gly Met Asp Glu Leu Tyr Lys 225 230 235 7 238 PRT Aequorea victoria VARIANT (0)...(0) YFP H148G 7 Met Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu Val 1 5 10 15 Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly Glu 20 25 30 Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile Cys 35 40 45 Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr Phe 50 55 60 Gly Tyr Gly Leu Gln Cys Phe Ala Arg Tyr Pro Asp His Met Lys Arg 65 70 75 80 His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu Arg 85 90 95 Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu Val 100 105 110 Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly Ile 115 120 125 Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr Asn 130 135 140 Tyr Asn Ser Gly Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn Gly 145 150 155 160 Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser Val 165 170 175 Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly Pro 180 185 190 Val Leu Leu Pro Asp Asn His Tyr Leu Ser Tyr Gln Ser Ala Leu Ser 195 200 205 Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe Val 210 215 220 Thr Ala Ala Gly Ile Thr His Gly Met Asp Glu Leu Tyr Lys 225 230 235 8 238 PRT Aequorea victoria VARIANT (0)...(0) YFP H148Q 8 Met Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu Val 1 5 10 15 Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly Glu 20 25 30 Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile Cys 35 40 45 Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr Phe 50 55 60 Gly Tyr Gly Leu Gln Cys Phe Ala Arg Tyr Pro Asp His Met Lys Arg 65 70 75 80 His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu Arg 85 90 95 Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu Val 100 105 110 Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly Ile 115 120 125 Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr Asn 130 135 140 Tyr Asn Ser Gln Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn Gly 145 150 155 160 Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser Val 165 170 175 Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly Pro 180 185 190 Val Leu Leu Pro Asp Asn His Tyr Leu Ser Tyr Gln Ser Ala Leu Ser 195 200 205 Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe Val 210 215 220 Thr Ala Ala Gly Ile Thr His Gly Met Asp Glu Leu Tyr Lys 225 230 235 9 239 PRT Aequorea victoria VARIANT (0)...(0) EYFP-H148G 9 Met Val Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu 1 5 10 15 Val Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly 20 25 30 Glu Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile 35 40 45 Cys Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr 50 55 60 Phe Gly Tyr Gly Val Gln Cys Phe Ala Arg Tyr Pro Asp His Met Lys 65 70 75 80 Gln His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu 85 90 95 Arg Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu 100 105 110 Val Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly 115 120 125 Ile Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr 130 135 140 Asn Tyr Asn Ser Gly Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn 145 150 155 160 Gly Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser 165 170 175 Val Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly 180 185 190 Pro Val Leu Leu Pro Asp Asn His Tyr Leu Ser Tyr Gln Ser Ala Leu 195 200 205 Ser Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe 210 215 220 Val Thr Ala Ala Gly Ile Thr Leu Gly Met Asp Glu Leu Tyr Lys 225 230 235 10 239 PRT Aequorea victoria VARIANT (0)...(0) EYFP-H148Q 10 Met Val Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu 1 5 10 15 Val Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly 20 25 30 Glu Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile 35 40 45 Cys Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr 50 55 60 Phe Gly Tyr Gly Val Gln Cys Phe Ala Arg Tyr Pro Asp His Met Lys 65 70 75 80 Gln His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu 85 90 95 Arg Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu 100 105 110 Val Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly 115 120 125 Ile Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr 130 135 140 Asn Tyr Asn Ser Gln Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn 145 150 155 160 Gly Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser 165 170 175 Val Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly 180 185 190 Pro Val Leu Leu Pro Asp Asn His Tyr Leu Ser Tyr Gln Ser Ala Leu 195 200 205 Ser Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe 210 215 220 Val Thr Ala Ala Gly Ile Thr Leu Gly Met Asp Glu Leu Tyr Lys 225 230 235 11 720 DNA Aequorea victoria misc_feature (0)...(0) EGFP 11 atggtgagca agggcgagga gctgttcacc ggggtggtgc ccatcctggt cgagctggac 60 ggcgacgtaa acggccacaa gttcagcgtg tccggcgagg gcgagggcga tgccacctac 120 ggcaagctga ccctgaagtt catctgcacc accggcaagc tgcccgtgcc ctggcccacc 180 ctcgtgacca ccctgaccta cggcgtgcag tgcttcagcc gctaccccga ccacatgaag 240 cagcacgact tcttcaagtc cgccatgccc gaaggctacg tccaggagcg caccatcttc 300 ttcaaggacg acggcaacta caagacccgc gccgaggtga agttcgaggg cgacaccctg 360 gtgaaccgca tcgagctgaa gggcatcgac ttcaaggagg acggcaacat cctggggcac 420 aagctggagt acaactacaa cagccacaac gtctatatca tggccgacaa gcagaagaac 480 ggcatcaagg tgaacttcaa gatccgccac aacatcgagg acggcagcgt gcagctcgcc 540 gaccactacc agcagaacac ccccatcggc gacggccccg tgctgctgcc cgacaaccac 600 tacctgagca cccagtccgc cctgagcaaa gaccccaacg agaagcgcga tcacatggtc 660 ctgctggagt tcgtgaccgc cgccgggatc actctcggca tggacgagct gtacaagtaa 720 12 720 DNA Aequorea victoria misc_feature (0)...(0) EYFP 12 atggtgagca agggcgagga gctgttcacc ggggtggtgc ccatcctggt cgagctggac 60 ggcgacgtaa acggccacaa gttcagcgtg tccggcgagg gcgagggcga tgccacctac 120 ggcaagctga ccctgaagtt catctgcacc accggcaagc tgcccgtgcc ctggcccacc 180 ctcgtgacca ccttcggcta cggcgtgcag tgcttcgccc gctaccccga ccacatgaag 240 cagcacgact tcttcaagtc cgccatgccc gaaggctacg tccaggagcg caccatcttc 300 ttcaaggacg acggcaacta caagacccgc gccgaggtga agttcgaggg cgacaccctg 360 gtgaaccgca tcgagctgaa gggcatcgac ttcaaggagg acggcaacat cctggggcac 420 aagctggagt acaactacaa cagccacaac gtctatatca tggccgacaa gcagaagaac 480 ggcatcaagg tgaacttcaa gatccgccac aacatcgagg acggcagcgt gcagctcgcc 540 gaccactacc agcagaacac ccccatcggc gacggccccg tgctgctgcc cgacaaccac 600 tacctgagct accagtccgc cctgagcaaa gaccccaacg agaagcgcga tcacatggtc 660 ctgctggagt tcgtgaccgc cgccgggatc actctcggca tggacgagct gtacaagtaa 720 13 720 DNA Aequorea victoria misc_feature (0)...(0) ECFP 13 atggtgagca agggcgagga gctgttcacc ggggtggtgc ccatcctggt cgagctggac 60 ggcgacgtaa acggccacag gttcagcgtg tccggcgagg gcgagggcga tgccacctac 120 ggcaagctga ccctgaagtt catctgcacc accggcaagc tgcccgtgcc ctggcccacc 180 ctcgtgacca ccctgacctg gggcgtgcag tgcttcagcc gctaccccga ccacatgaag 240 cagcacgact tcttcaagtc cgccatgccc gaaggctacg tccaggagcg taccatcttc 300 ttcaaggacg acggcaacta caagacccgc gccgaggtga agttcgaggg cgacaccctg 360 gtgaaccgca tcgagctgaa gggcatcgac ttcaaggagg acggcaacat cctggggcac 420 aagctggagt acaactacat cagccacaac gtctatatca ccgccgacaa gcagaagaac 480 ggcatcaagg cccacttcaa gatccgccac aacatcgagg acggcagcgt gcagctcgcc 540 gaccactacc agcagaacac ccccatcggc gacggccccg tgctgctgcc cgacaaccac 600 tacctgagca cccagtccgc cctgagcaaa gaccccaacg agaagcgcga tcacatggtc 660 ctgctggagt tcgtgaccgc cgccgggatc actctcggca tggacgagct gtacaagtaa 720 14 720 DNA Aequorea victoria misc_feature (0)...(0) EYFP-V68L/Q69K 14 atggtgagca agggcgagga gctgttcacc ggggtggtgc ccatcctggt cgagctggac 60 ggcgacgtaa acggccacaa gttcagcgtg tccggcgagg gcgagggcga tgccacctac 120 ggcaagctga ccctgaagtt catctgcacc accggcaagc tgcccgtgcc ctggcccacc 180 ctcgtgacca ccttcggcta cggcctgaag tgcttcgccc gctaccccga ccacatgaag 240 cagcacgact tcttcaagtc cgccatgccc gaaggctacg tccaggagcg caccatcttc 300 ttcaaggacg acggcaacta caagacccgc gccgaggtga agttcgaggg cgacaccctg 360 gtgaaccgca tcgagctgaa gggcatcgac ttcaaggagg acggcaacat cctggggcac 420 aagctggagt acaactacaa cagccacaac gtctatatca tggccgacaa gcagaagaac 480 ggcatcaagg tgaacttcaa gatccgccac aacatcgagg acggcagcgt gcagctcgcc 540 gaccactacc agcagaacac ccccatcggc gacggccccg tgctgctgcc cgacaaccac 600 tacctgagct accagtccgc cctgagcaaa gaccccaacg agaagcgcga tcacatggtc 660 ctgctggagt tcgtgaccgc cgccgggatc actctcggca tggacgagct gtacaagtaa 720 15 714 DNA Aequorea victoria misc_feature (0)...(0) YFP H148G 15 atgagtaaag gagaagaact tttcactgga gttgtcccaa ttcttgttga attagatggt 60 gatgttaatg ggcacaaatt ttctgtcagt ggagagggtg aaggtgatgc aacatacgga 120 aaacttaccc ttaaatttat ttgcactact ggaaaactac ctgttccatg gccaacactt 180 gtcactactt tcggttatgg tcttcaatgc tttgcaagat acccagatca tatgaaacgg 240 catgactttt tcaagagtgc catgcccgaa ggttatgttc aggaaagaac tatatttttc 300 aaagatgacg ggaactacaa gacacgtgct gaagtcaagt ttgaaggtga tacccttgtt 360 aatagaatcg agttaaaagg tattgatttt aaagaagatg gaaacattct tggacacaaa 420 ttggaataca actataactc aggcaatgta tacatcatgg cagacaaaca aaagaatgga 480 atcaaagtta acttcaaaat tagacacaac attgaagatg gaagcgttca actagcagac 540 cattatcaac aaaatactcc aattggcgat ggccctgtcc ttttaccaga caaccattac 600 ctgtcctatc aatctgccct ttcgaaagat cccaacgaaa agagagacca catggtcctt 660 cttgagtttg taacagctgc tgggattaca catggcatgg atgaactata caaa 714 16 714 DNA Aequorea victoria misc_feature (0)...(0) YFP H148Q 16 atgagtaaag gagaagaact tttcactgga gttgtcccaa ttcttgttga attagatggt 60 gatgttaatg ggcacaaatt ttctgtcagt ggagagggtg aaggtgatgc aacatacgga 120 aaacttaccc ttaaatttat ttgcactact ggaaaactac ctgttccatg gccaacactt 180 gtcactactt tcggttatgg tcttcaatgc tttgcaagat acccagatca tatgaaacgg 240 catgactttt tcaagagtgc catgcccgaa ggttatgttc aggaaagaac tatatttttc 300 aaagatgacg ggaactacaa gacacgtgct gaagtcaagt ttgaaggtga tacccttgtt 360 aatagaatcg agttaaaagg tattgatttt aaagaagatg gaaacattct tggacacaaa 420 ttggaataca actataactc aggcaatgta tacatcatgg cagacaaaca aaagaatgga 480 atcaaagtta acttcaaaat tagacacaac attgaagatg gaagcgttca actagcagac 540 cattatcaac aaaatactcc aattggcgat ggccctgtcc ttttaccaga caaccattac 600 ctgtcctatc aatctgccct ttcgaaagat cccaacgaaa agagagacca catggtcctt 660 cttgagtttg taacagctgc tgggattaca catggcatgg atgaactata caaa 714 17 720 DNA Aequorea victoria misc_feature (0)...(0) EYFP-H148G 17 atggtgagca agggcgagga gctgttcacc ggggtggtgc ccatcctggt cgagctggac 60 ggcgacgtaa acggccacaa gttcagcgtg tccggcgagg gcgagggcga tgccacctac 120 ggcaagctga ccctgaagtt catctgcacc accggcaagc tgcccgtgcc ctggcccacc 180 ctcgtgacca ccttcggcta cggcgtgcag tgcttcgccc gctaccccga ccacatgaag 240 cagcacgact tcttcaagtc cgccatgccc gaaggctacg tccaggagcg caccatcttc 300 ttcaaggacg acggcaacta caagacccgc gccgaggtga agttcgaggg cgacaccctg 360 gtgaaccgca tcgagctgaa gggcatcgac ttcaaggagg acggcaacat cctggggcac 420 aagctggagt acaactacaa cagcggcaac gtctatatca tggccgacaa gcagaagaac 480 ggcatcaagg tgaacttcaa gatccgccac aacatcgagg acggcagcgt gcagctcgcc 540 gaccactacc agcagaacac ccccatcggc gacggccccg tgctgctgcc cgacaaccac 600 tacctgagct accagtccgc cctgagcaaa gaccccaacg agaagcgcga tcacatggtc 660 ctgctggagt tcgtgaccgc cgccgggatc actctcggca tggacgagct gtacaagtaa 720 18 720 DNA Aequorea victoria misc_feature (0)...(0) EYFP-H148Q 18 atggtgagca agggcgagga gctgttcacc ggggtggtgc ccatcctggt cgagctggac 60 ggcgacgtaa acggccacaa gttcagcgtg tccggcgagg gcgagggcga tgccacctac 120 ggcaagctga ccctgaagtt catctgcacc accggcaagc tgcccgtgcc ctggcccacc 180 ctcgtgacca ccttcggcta cggcgtgcag tgcttcgccc gctaccccga ccacatgaag 240 cagcacgact tcttcaagtc cgccatgccc gaaggctacg tccaggagcg caccatcttc 300 ttcaaggacg acggcaacta caagacccgc gccgaggtga agttcgaggg cgacaccctg 360 gtgaaccgca tcgagctgaa gggcatcgac ttcaaggagg acggcaacat cctggggcac 420 aagctggagt acaactacaa cagccagaac gtctatatca tggccgacaa gcagaagaac 480 ggcatcaagg tgaacttcaa gatccgccac aacatcgagg acggcagcgt gcagctcgcc 540 gaccactacc agcagaacac ccccatcggc gacggccccg tgctgctgcc cgacaaccac 600 tacctgagct accagtccgc cctgagcaaa gaccccaacg agaagcgcga tcacatggtc 660 ctgctggagt tcgtgaccgc cgccgggatc actctcggca tggacgagct gtacaagtaa 720 19 255 PRT Aequorea victoria VARIANT (0)...(0) mito-ECFP 19 Met Leu Ser Leu Arg Gln Ser Ile Arg Phe Phe Lys Arg Ser Gly Ile 1 5 10 15 Met Val Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu 20 25 30 Val Glu Leu Asp Gly Asp Val Asn Gly His Arg Phe Ser Val Ser Gly 35 40 45 Glu Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile 50 55 60 Cys Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr 65 70 75 80 Leu Thr Trp Gly Val Gln Cys Phe Ser Arg Tyr Pro Asp His Met Lys 85 90 95 Gln His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu 100 105 110 Arg Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu 115 120 125 Val Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly 130 135 140 Ile Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr 145 150 155 160 Asn Tyr Ile Ser His Asn Val Tyr Ile Thr Ala Asp Lys Gln Lys Asn 165 170 175 Gly Ile Lys Ala His Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser 180 185 190 Val Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly 195 200 205 Pro Val Leu Leu Pro Asp Asn His Tyr Leu Ser Thr Gln Ser Ala Leu 210 215 220 Ser Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe 225 230 235 240 Val Thr Ala Ala Gly Ile Thr Leu Gly Met Asp Glu Leu Tyr Lys 245 250 255 20 255 PRT Aequorea victoria VARIANT (0)...(0) mito-EYFP 20 Met Leu Ser Leu Arg Gln Ser Ile Arg Phe Phe Lys Arg Ser Gly Ile 1 5 10 15 Met Val Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu 20 25 30 Val Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly 35 40 45 Glu Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile 50 55 60 Cys Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr 65 70 75 80 Phe Gly Tyr Gly Val Gln Cys Phe Ala Arg Tyr Pro Asp His Met Lys 85 90 95 Gln His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu 100 105 110 Arg Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu 115 120 125 Val Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly 130 135 140 Ile Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr 145 150 155 160 Asn Tyr Asn Ser His Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn 165 170 175 Gly Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser 180 185 190 Val Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly 195 200 205 Pro Val Leu Leu Pro Asp Asn His Tyr Leu Ser Tyr Gln Ser Ala Leu 210 215 220 Ser Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe 225 230 235 240 Val Thr Ala Ala Gly Ile Thr Leu Gly Met Asp Glu Leu Tyr Lys 245 250 255 21 323 PRT Aequorea victoria VARIANT (0)...(0) GT-EGFP 21 Met Arg Leu Arg Glu Pro Leu Leu Ser Gly Ala Ala Met Pro Gly Ala 1 5 10 15 Ser Leu Gln Arg Ala Cys Arg Leu Leu Val Ala Val Cys Ala Leu His 20 25 30 Leu Gly Val Thr Leu Val Tyr Tyr Leu Ala Gly Arg Asp Leu Ser Arg 35 40 45 Leu Pro Gln Leu Val Gly Val Ser Thr Pro Leu Gln Gly Gly Ser Asn 50 55 60 Ser Ala Ala Ala Ile Gly Gln Ser Ser Gly Glu Leu Arg Thr Gly Gly 65 70 75 80 Ala Met Asp Pro Met Val Ser Lys Gly Glu Glu Leu Phe Thr Gly Val 85 90 95 Val Pro Ile Leu Val Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe 100 105 110 Ser Val Ser Gly Glu Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr 115 120 125 Leu Lys Phe Ile Cys Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr 130 135 140 Leu Val Thr Thr Leu Thr Tyr Gly Val Gln Cys Phe Ser Arg Tyr Pro 145 150 155 160 Asp His Met Lys Gln His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly 165 170 175 Tyr Val Gln Glu Arg Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys 180 185 190 Thr Arg Ala Glu Val Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile 195 200 205 Glu Leu Lys Gly Ile Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His 210 215 220 Lys Leu Glu Tyr Asn Tyr Asn Ser His Asn Val Tyr Ile Met Ala Asp 225 230 235 240 Lys Gln Lys Asn Gly Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile 245 250 255 Glu Asp Gly Ser Val Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro 260 265 270 Ile Gly Asp Gly Pro Val Leu Leu Pro Asp Asn His Tyr Leu Ser Thr 275 280 285 Gln Ser Ala Leu Ser Lys Asp Pro Asn Glu Lys Arg Asp His Met Val 290 295 300 Leu Leu Glu Phe Val Thr Ala Ala Gly Ile Thr Leu Gly Met Asp Glu 305 310 315 320 Leu Tyr Lys 22 323 PRT Aequorea victoria VARIANT (0)...(0) GT-EYFP 22 Met Arg Leu Arg Glu Pro Leu Leu Ser Gly Ala Ala Met Pro Gly Ala 1 5 10 15 Ser Leu Gln Arg Ala Cys Arg Leu Leu Val Ala Val Cys Ala Leu His 20 25 30 Leu Gly Val Thr Leu Val Tyr Tyr Leu Ala Gly Arg Asp Leu Ser Arg 35 40 45 Leu Pro Gln Leu Val Gly Val Ser Thr Pro Leu Gln Gly Gly Ser Asn 50 55 60 Ser Ala Ala Ala Ile Gly Gln Ser Ser Gly Glu Leu Arg Thr Gly Gly 65 70 75 80 Ala Met Asp Pro Met Val Ser Lys Gly Glu Glu Leu Phe Thr Gly Val 85 90 95 Val Pro Ile Leu Val Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe 100 105 110 Ser Val Ser Gly Glu Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr 115 120 125 Leu Lys Phe Ile Cys Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr 130 135 140 Leu Val Thr Thr Phe Gly Tyr Gly Val Gln Cys Phe Ala Arg Tyr Pro 145 150 155 160 Asp His Met Lys Gln His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly 165 170 175 Tyr Val Gln Glu Arg Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys 180 185 190 Thr Arg Ala Glu Val Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile 195 200 205 Glu Leu Lys Gly Ile Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His 210 215 220 Lys Leu Glu Tyr Asn Tyr Asn Ser His Asn Val Tyr Ile Met Ala Asp 225 230 235 240 Lys Gln Lys Asn Gly Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile 245 250 255 Glu Asp Gly Ser Val Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro 260 265 270 Ile Gly Asp Gly Pro Val Leu Leu Pro Asp Asn His Tyr Leu Ser Tyr 275 280 285 Gln Ser Ala Leu Ser Lys Asp Pro Asn Glu Lys Arg Asp His Met Val 290 295 300 Leu Leu Glu Phe Val Thr Ala Ala Gly Ile Thr Leu Gly Met Asp Glu 305 310 315 320 Leu Tyr Lys 23 263 PRT Aequorea victoria VARIANT (0)...(0) mito-YFP H148G 23 Met Leu Arg Thr Ser Ser Leu Phe Thr Arg Arg Val Gln Pro Ser Leu 1 5 10 15 Phe Arg Asn Ile Leu Arg Leu Gln Ser Thr Ser Lys Gly Glu Glu Leu 20 25 30 Phe Thr Gly Val Val Pro Ile Leu Val Glu Leu Asp Gly Asp Val Asn 35 40 45 Gly His Lys Phe Ser Val Ser Gly Glu Gly Glu Gly Asp Ala Thr Tyr 50 55 60 Gly Lys Leu Thr Leu Lys Phe Ile Cys Thr Thr Gly Lys Leu Pro Val 65 70 75 80 Pro Trp Pro Thr Leu Val Thr Thr Phe Gly Tyr Gly Leu Gln Cys Phe 85 90 95 Ala Arg Tyr Pro Asp His Met Lys Arg His Asp Phe Phe Lys Ser Ala 100 105 110 Met Pro Glu Gly Tyr Val Gln Glu Arg Thr Ile Phe Phe Lys Asp Asp 115 120 125 Gly Asn Tyr Lys Thr Arg Ala Glu Val Lys Phe Glu Gly Asp Thr Leu 130 135 140 Val Asn Arg Ile Glu Leu Lys Gly Ile Asp Phe Lys Glu Asp Gly Asn 145 150 155 160 Ile Leu Gly His Lys Leu Glu Tyr Asn Tyr Asn Ser Gly Asn Val Tyr 165 170 175 Ile Met Ala Asp Lys Gln Lys Asn Gly Ile Lys Val Asn Phe Lys Ile 180 185 190 Arg His Asn Ile Glu Asp Gly Ser Val Gln Leu Ala Asp His Tyr Gln 195 200 205 Gln Asn Thr Pro Ile Gly Asp Gly Pro Val Leu Leu Pro Asp Asn His 210 215 220 Tyr Leu Ser Tyr Gln Ser Ala Leu Ser Lys Asp Pro Asn Glu Lys Arg 225 230 235 240 Asp His Met Val Leu Leu Glu Phe Val Thr Ala Ala Gly Ile Thr His 245 250 255 Gly Met Asp Glu Leu Tyr Lys 260 24 263 PRT Aequorea victoria VARIANT (0)...(0) mito-YFP H148Q 24 Met Leu Arg Thr Ser Ser Leu Phe Thr Arg Arg Val Gln Pro Ser Leu 1 5 10 15 Phe Arg Asn Ile Leu Arg Leu Gln Ser Thr Ser Lys Gly Glu Glu Leu 20 25 30 Phe Thr Gly Val Val Pro Ile Leu Val Glu Leu Asp Gly Asp Val Asn 35 40 45 Gly His Lys Phe Ser Val Ser Gly Glu Gly Glu Gly Asp Ala Thr Tyr 50 55 60 Gly Lys Leu Thr Leu Lys Phe Ile Cys Thr Thr Gly Lys Leu Pro Val 65 70 75 80 Pro Trp Pro Thr Leu Val Thr Thr Phe Gly Tyr Gly Leu Gln Cys Phe 85 90 95 Ala Arg Tyr Pro Asp His Met Lys Arg His Asp Phe Phe Lys Ser Ala 100 105 110 Met Pro Glu Gly Tyr Val Gln Glu Arg Thr Ile Phe Phe Lys Asp Asp 115 120 125 Gly Asn Tyr Lys Thr Arg Ala Glu Val Lys Phe Glu Gly Asp Thr Leu 130 135 140 Val Asn Arg Ile Glu Leu Lys Gly Ile Asp Phe Lys Glu Asp Gly Asn 145 150 155 160 Ile Leu Gly His Lys Leu Glu Tyr Asn Tyr Asn Ser Gln Asn Val Tyr 165 170 175 Ile Met Ala Asp Lys Gln Lys Asn Gly Ile Lys Val Asn Phe Lys Ile 180 185 190 Arg His Asn Ile Glu Asp Gly Ser Val Gln Leu Ala Asp His Tyr Gln 195 200 205 Gln Asn Thr Pro Ile Gly Asp Gly Pro Val Leu Leu Pro Asp Asn His 210 215 220 Tyr Leu Ser Tyr Gln Ser Ala Leu Ser Lys Asp Pro Asn Glu Lys Arg 225 230 235 240 Asp His Met Val Leu Leu Glu Phe Val Thr Ala Ala Gly Ile Thr His 245 250 255 Gly Met Asp Glu Leu Tyr Lys 260 25 265 PRT Aequorea victoria VARIANT (0)...(0) mito-EYFP-H148G 25 Met Leu Arg Thr Ser Ser Leu Phe Thr Arg Arg Val Gln Pro Ser Leu 1 5 10 15 Phe Arg Asn Ile Leu Arg Leu Gln Ser Thr Met Val Ser Lys Gly Glu 20 25 30 Glu Leu Phe Thr Gly Val Val Pro Ile Leu Val Glu Leu Asp Gly Asp 35 40 45 Val Asn Gly His Lys Phe Ser Val Ser Gly Glu Gly Glu Gly Asp Ala 50 55 60 Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile Cys Thr Thr Gly Lys Leu 65 70 75 80 Pro Val Pro Trp Pro Thr Leu Val Thr Thr Phe Gly Tyr Gly Val Gln 85 90 95 Cys Phe Ala Arg Tyr Pro Asp His Met Lys Gln His Asp Phe Phe Lys 100 105 110 Ser Ala Met Pro Glu Gly Tyr Val Gln Glu Arg Thr Ile Phe Phe Lys 115 120 125 Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu Val Lys Phe Glu Gly Asp 130 135 140 Thr Leu Val Asn Arg Ile Glu Leu Lys Gly Ile Asp Phe Lys Glu Asp 145 150 155 160 Gly Asn Ile Leu Gly His Lys Leu Glu Tyr Asn Tyr Asn Ser Gly Asn 165 170 175 Val Tyr Ile Met Ala Asp Lys Gln Lys Asn Gly Ile Lys Val Asn Phe 180 185 190 Lys Ile Arg His Asn Ile Glu Asp Gly Ser Val Gln Leu Ala Asp His 195 200 205 Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly Pro Val Leu Leu Pro Asp 210 215 220 Asn His Tyr Leu Ser Tyr Gln Ser Ala Leu Ser Lys Asp Pro Asn Glu 225 230 235 240 Lys Arg Asp His Met Val Leu Leu Glu Phe Val Thr Ala Ala Gly Ile 245 250 255 Thr Leu Gly Met Asp Glu Leu Tyr Lys 260 265 26 265 PRT Aequorea victoria VARIANT (0)...(0) mito-EYFP-H148Q 26 Met Leu Arg Thr Ser Ser Leu Phe Thr Arg Arg Val Gln Pro Ser Leu 1 5 10 15 Phe Arg Asn Ile Leu Arg Leu Gln Ser Thr Met Val Ser Lys Gly Glu 20 25 30 Glu Leu Phe Thr Gly Val Val Pro Ile Leu Val Glu Leu Asp Gly Asp 35 40 45 Val Asn Gly His Lys Phe Ser Val Ser Gly Glu Gly Glu Gly Asp Ala 50 55 60 Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile Cys Thr Thr Gly Lys Leu 65 70 75 80 Pro Val Pro Trp Pro Thr Leu Val Thr Thr Phe Gly Tyr Gly Val Gln 85 90 95 Cys Phe Ala Arg Tyr Pro Asp His Met Lys Gln His Asp Phe Phe Lys 100 105 110 Ser Ala Met Pro Glu Gly Tyr Val Gln Glu Arg Thr Ile Phe Phe Lys 115 120 125 Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu Val Lys Phe Glu Gly Asp 130 135 140 Thr Leu Val Asn Arg Ile Glu Leu Lys Gly Ile Asp Phe Lys Glu Asp 145 150 155 160 Gly Asn Ile Leu Gly His Lys Leu Glu Tyr Asn Tyr Asn Ser Gln Asn 165 170 175 Val Tyr Ile Met Ala Asp Lys Gln Lys Asn Gly Ile Lys Val Asn Phe 180 185 190 Lys Ile Arg His Asn Ile Glu Asp Gly Ser Val Gln Leu Ala Asp His 195 200 205 Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly Pro Val Leu Leu Pro Asp 210 215 220 Asn His Tyr Leu Ser Tyr Gln Ser Ala Leu Ser Lys Asp Pro Asn Glu 225 230 235 240 Lys Arg Asp His Met Val Leu Leu Glu Phe Val Thr Ala Ala Gly Ile 245 250 255 Thr Leu Gly Met Asp Glu Leu Tyr Lys 260 265 27 972 DNA Aequorea victoria misc_feature (0)...(0) GT-ECFP 27 atgaggcttc gggagccgct cctgagcggc gccgcgatgc caggcgcgtc cctacagcgg 60 gcctgccgcc tgctcgtggc cgtctgcgct ctgcaccttg gcgtcaccct cgtttactac 120 ctggctggcc gcgacctgag ccgcctgccc caactggtcg gagtctccac accgctgcag 180 ggcggctcga acagtgccgc cgccatcggg cagtcctccg gggagctccg gaccggaggg 240 gccatggatc ccatggtgag caagggcgag gagctgttca ccggggtggt gcccatcctg 300 gtcgagctgg acggcgacgt aaacggccac aggttcagcg tgtccggcga gggcgagggc 360 gatgccacct acggcaagct gaccctgaag ttcatctgca ccaccggcaa gctgcccgtg 420 ccctggccca ccctcgtgac caccctgacc tggggcgtgc agtgcttcag ccgctacccc 480 gaccacatga agcagcacga cttcttcaag tccgccatgc ccgaaggcta cgtccaggag 540 cgtaccatct tcttcaagga cgacggcaac tacaagaccc gcgccgaggt gaagttcgag 600 ggcgacaccc tggtgaaccg catcgagctg aagggcatcg acttcaagga ggacggcaac 660 atcctggggc acaagctgga gtacaactac atcagccaca acgtctatat caccgccgac 720 aagcagaaga acggcatcaa ggcccacttc aagatccgcc acaacatcga ggacggcagc 780 gtgcagctcg ccgaccacta ccagcagaac acccccatcg gcgacggccc cgtgctgctg 840 cccgacaacc actacctgag cacccagtcc gccctgagca aagaccccaa cgagaagcgc 900 gatcacatgg tcctgctgga gttcgtgacc gccgccggga tcactctcgg catggacgag 960 ctgtacaagt aa 972 28 768 DNA Aequorea victoria misc_feature (0)...(0) mito-EYFP 28 atgctgagcc tgcgccagag catccgcttc ttcaagcgca gcggcatcat ggtgagcaag 60 ggcgaggagc tgttcaccgg ggtggtgccc atcctggtcg agctggacgg cgacgtaaac 120 ggccacaagt tcagcgtgtc cggcgagggc gagggcgatg ccacctacgg caagctgacc 180 ctgaagttca tctgcaccac cggcaagctg cccgtgccct ggcccaccct cgtgaccacc 240 ttcggctacg gcgtgcagtg cttcgcccgc taccccgacc acatgaagca gcacgacttc 300 ttcaagtccg ccatgcccga aggctacgtc caggagcgca ccatcttctt caaggacgac 360 ggcaactaca agacccgcgc cgaggtgaag ttcgagggcg acaccctggt gaaccgcatc 420 gagctgaagg gcatcgactt caaggaggac ggcaacatcc tggggcacaa gctggagtac 480 aactacaaca gccacaacgt ctatatcatg gccgacaagc agaagaacgg catcaaggtg 540 aacttcaaga tccgccacaa catcgaggac ggcagcgtgc agctcgccga ccactaccag 600 cagaacaccc ccatcggcga cggccccgtg ctgctgcccg acaaccacta cctgagctac 660 cagtccgccc tgagcaaaga ccccaacgag aagcgcgatc acatggtcct gctggagttc 720 gtgaccgccg ccgggatcac tctcggcatg gacgagctgt acaagtaa 768 29 972 DNA Aequorea victoria misc_feature (0)...(0) GT-EGFP 29 atgaggcttc gggagccgct cctgagcggc gccgcgatgc caggcgcgtc cctacagcgg 60 gcctgccgcc tgctcgtggc cgtctgcgct ctgcaccttg gcgtcaccct cgtttactac 120 ctggctggcc gcgacctgag ccgcctgccc caactggtcg gagtctccac accgctgcag 180 ggcggctcga acagtgccgc cgccatcggg cagtcctccg gggagctccg gaccggaggg 240 gccatggatc ccatggtgag caagggcgag gagctgttca ccggggtggt gcccatcctg 300 gtcgagctgg acggcgacgt aaacggccac aagttcagcg tgtccggcga gggcgagggc 360 gatgccacct acggcaagct gaccctgaag ttcatctgca ccaccggcaa gctgcccgtg 420 ccctggccca ccctcgtgac caccctgacc tacggcgtgc agtgcttcag ccgctacccc 480 gaccacatga agcagcacga cttcttcaag tccgccatgc ccgaaggcta cgtccaggag 540 cgcaccatct tcttcaagga cgacggcaac tacaagaccc gcgccgaggt gaagttcgag 600 ggcgacaccc tggtgaaccg catcgagctg aagggcatcg acttcaagga ggacggcaac 660 atcctggggc acaagctgga gtacaactac aacagccaca acgtctatat catggccgac 720 aagcagaaga acggcatcaa ggtgaacttc aagatccgcc acaacatcga ggacggcagc 780 gtgcagctcg ccgaccacta ccagcagaac acccccatcg gcgacggccc cgtgctgctg 840 cccgacaacc actacctgag cacccagtcc gccctgagca aagaccccaa cgagaagcgc 900 gatcacatgg tcctgctgga gttcgtgacc gccgccggga tcactctcgg catggacgag 960 ctgtacaagt aa 972 30 972 DNA Aequorea victoria misc_feature (0)...(0) GT-EYFP 30 atgaggcttc gggagccgct cctgagcggc gccgcgatgc caggcgcgtc cctacagcgg 60 gcctgccgcc tgctcgtggc cgtctgcgct ctgcaccttg gcgtcaccct cgtttactac 120 ctggctggcc gcgacctgag ccgcctgccc caactggtcg gagtctccac accgctgcag 180 ggcggctcga acagtgccgc cgccatcggg cagtcctccg gggagctccg gaccggaggg 240 gccatggatc ccatggtgag caagggcgag gagctgttca ccggggtggt gcccatcctg 300 gtcgagctgg acggcgacgt aaacggccac aagttcagcg tgtccggcga gggcgagggc 360 gatgccacct acggcaagct gaccctgaag ttcatctgca ccaccggcaa gctgcccgtg 420 ccctggccca ccctcgtgac caccttcggc tacggcgtgc agtgcttcgc ccgctacccc 480 gaccacatga agcagcacga cttcttcaag tccgccatgc ccgaaggcta cgtccaggag 540 cgcaccatct tcttcaagga cgacggcaac tacaagaccc gcgccgaggt gaagttcgag 600 ggcgacaccc tggtgaaccg catcgagctg aagggcatcg acttcaagga ggacggcaac 660 atcctggggc acaagctgga gtacaactac aacagccaca acgtctatat catggccgac 720 aagcagaaga acggcatcaa ggtgaacttc aagatccgcc acaacatcga ggacggcagc 780 gtgcagctcg ccgaccacta ccagcagaac acccccatcg gcgacggccc cgtgctgctg 840 cccgacaacc actacctgag ctaccagtcc gccctgagca aagaccccaa cgagaagcgc 900 gatcacatgg tcctgctgga gttcgtgacc gccgccggga tcactctcgg catggacgag 960 ctgtacaagt aa 972 31 762 DNA Aequorea victoria misc_feature (0)...(0) mito-YFP H148G 31 atgctgagcc tgcgccagag catccgcttc ttcaagcgca gcggcatcat gagtaaagga 60 gaagaacttt tcactggagt tgtcccaatt cttgttgaat tagatggtga tgttaatggg 120 cacaaatttt ctgtcagtgg agagggtgaa ggtgatgcaa catacggaaa acttaccctt 180 aaatttattt gcactactgg aaaactacct gttccatggc caacacttgt cactactttc 240 ggttatggtc ttcaatgctt tgcaagatac ccagatcata tgaaacggca tgactttttc 300 aagagtgcca tgcccgaagg ttatgttcag gaaagaacta tatttttcaa agatgacggg 360 aactacaaga cacgtgctga agtcaagttt gaaggtgata cccttgttaa tagaatcgag 420 ttaaaaggta ttgattttaa agaagatgga aacattcttg gacacaaatt ggaatacaac 480 tataactcag gcaatgtata catcatggca gacaaacaaa agaatggaat caaagttaac 540 ttcaaaatta gacacaacat tgaagatgga agcgttcaac tagcagacca ttatcaacaa 600 aatactccaa ttggcgatgg ccctgtcctt ttaccagaca accattacct gtcctatcaa 660 tctgcccttt cgaaagatcc caacgaaaag agagaccaca tggtccttct tgagtttgta 720 acagctgctg ggattacaca tggcatggat gaactataca aa 762 32 762 DNA Aequorea victoria misc_feature (0)...(0) mito-YFP H148Q 32 atgctgagcc tgcgccagag catccgcttc ttcaagcgca gcggcatcat gagtaaagga 60 gaagaacttt tcactggagt tgtcccaatt cttgttgaat tagatggtga tgttaatggg 120 cacaaatttt ctgtcagtgg agagggtgaa ggtgatgcaa catacggaaa acttaccctt 180 aaatttattt gcactactgg aaaactacct gttccatggc caacacttgt cactactttc 240 ggttatggtc ttcaatgctt tgcaagatac ccagatcata tgaaacggca tgactttttc 300 aagagtgcca tgcccgaagg ttatgttcag gaaagaacta tatttttcaa agatgacggg 360 aactacaaga cacgtgctga agtcaagttt gaaggtgata cccttgttaa tagaatcgag 420 ttaaaaggta ttgattttaa agaagatgga aacattcttg gacacaaatt ggaatacaac 480 tataactcag gcaatgtata catcatggca gacaaacaaa agaatggaat caaagttaac 540 ttcaaaatta gacacaacat tgaagatgga agcgttcaac tagcagacca ttatcaacaa 600 aatactccaa ttggcgatgg ccctgtcctt ttaccagaca accattacct gtcctatcaa 660 tctgcccttt cgaaagatcc caacgaaaag agagaccaca tggtccttct tgagtttgta 720 acagctgctg ggattacaca tggcatggat gaactataca aa 762 33 768 DNA Aequorea victoria misc_feature (0)...(0) mito-EYFP-H148G 33 atgctgagcc tgcgccagag catccgcttc ttcaagcgca gcggcatcat ggtgagcaag 60 ggcgaggagc tgttcaccgg ggtggtgccc atcctggtcg agctggacgg cgacgtaaac 120 ggccacaagt tcagcgtgtc cggcgagggc gagggcgatg ccacctacgg caagctgacc 180 ctgaagttca tctgcaccac cggcaagctg cccgtgccct ggcccaccct cgtgaccacc 240 ttcggctacg gcgtgcagtg cttcgcccgc taccccgacc acatgaagca gcacgacttc 300 ttcaagtccg ccatgcccga aggctacgtc caggagcgca ccatcttctt caaggacgac 360 ggcaactaca agacccgcgc cgaggtgaag ttcgagggcg acaccctggt gaaccgcatc 420 gagctgaagg gcatcgactt caaggaggac ggcaacatcc tggggcacaa gctggagtac 480 aactacaaca gcggcaacgt ctatatcatg gccgacaagc agaagaacgg catcaaggtg 540 aacttcaaga tccgccacaa catcgaggac ggcagcgtgc agctcgccga ccactaccag 600 cagaacaccc ccatcggcga cggccccgtg ctgctgcccg acaaccacta cctgagctac 660 cagtccgccc tgagcaaaga ccccaacgag aagcgcgatc acatggtcct gctggagttc 720 gtgaccgccg ccgggatcac tctcggcatg gacgagctgt acaagtaa 768 34 768 DNA Aequorea victoria misc_feature (0)...(0) mito-EYFP-H148Q 34 atgctgagcc tgcgccagag catccgcttc ttcaagcgca gcggcatcat ggtgagcaag 60 ggcgaggagc tgttcaccgg ggtggtgccc atcctggtcg agctggacgg cgacgtaaac 120 ggccacaagt tcagcgtgtc cggcgagggc gagggcgatg ccacctacgg caagctgacc 180 ctgaagttca tctgcaccac cggcaagctg cccgtgccct ggcccaccct cgtgaccacc 240 ttcggctacg gcgtgcagtg cttcgcccgc taccccgacc acatgaagca gcacgacttc 300 ttcaagtccg ccatgcccga aggctacgtc caggagcgca ccatcttctt caaggacgac 360 ggcaactaca agacccgcgc cgaggtgaag ttcgagggcg acaccctggt gaaccgcatc 420 gagctgaagg gcatcgactt caaggaggac ggcaacatcc tggggcacaa gctggagtac 480 aactacaaca gccagaacgt ctatatcatg gccgacaagc agaagaacgg catcaaggtg 540 aacttcaaga tccgccacaa catcgaggac ggcagcgtgc agctcgccga ccactaccag 600 cagaacaccc ccatcggcga cggccccgtg ctgctgcccg acaaccacta cctgagctac 660 cagtccgccc tgagcaaaga ccccaacgag aagcgcgatc acatggtcct gctggagttc 720 gtgaccgccg ccgggatcac tctcggcatg gacgagctgt acaagtaa 768 35 5 PRT Homo sapiens 35 Lys Lys Lys Arg Lys 1 5 36 26 PRT Homo sapiens 36 Met Leu Arg Thr Ser Ser Leu Phe Thr Arg Arg Val Gln Pro Ser Leu 1 5 10 15 Phe Arg Asn Ile Leu Arg Leu Gln Ser Thr 20 25 37 4 PRT Homo sapiens 37 Lys Asp Glu Leu 1 38 4 PRT Aequorea victoria VARIANT (0)...(0) Linker sequence 38 Arg Ser Gly Ile 1

Claims (25)

What is claimed is:
1. A polynucleotide comprising a nucleotide sequence encoding a functional engineered fluorescent protein whose amino acid sequence is substantially identical to the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2) and whose emission intensity changes as pH varies between 5 and 10.
2. The polynucleotide of claim 1, wherein the amino acid sequence of the protein includes the substitutions S65G/S72A/T203Y/H231L in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
3. The polynucleotide of claim 1, wherein the amino acid sequence of the protein includes the substitutions S65G/V68L/Q69K/S72A/T203Y/H231L in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
4. The polynucleotide of claim 1, wherein the amino aid sequence of the protein includes the substitutions K26R/F64L/S65T/Y66W/N146I/M153T/V163A/N164H/H231L in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
5. The polynucleotide of claim 1, wherein the amino acid sequence of the protein includes the substitution H148G in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
6. The polynucleotide of claim 1, wherein the amino acid sequence of the protein includes the substitution H148Q in the amino acid of Aequorea green fluorescent protein (SEQ ID NO:2).
7. An expression vector comprising expression control sequences operatively linked to a polynucleotide molecule comprising a nucleotide sequence encoding a functional engineered fluorescent protein whose amino acid sequence is substantially identical to the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2) and whose emission intensity changes as pH varies between 5 and 10.
8. A recombinant host cell comprising the expression vector of claim 7.
9. The recombinant host cell of claim 8, wherein the recombinant host cell is a prokaryotic cell.
10. The recombinant host cell of claim 8, wherein the recombinant host cell is a eukaryotic cell.
11. A method of determining the pH of a region of a cell comprising:
introducing into the cell a polynucleotide encoding a polypeptide including an indicator having a first fluorescent protein moiety whose emission intensity changes as pH varies between 5 and 10;
culturing the cell under conditions that permit expression of the polynucleotide;
exciting the indicator; and
determining the intensity of the light emitted by the first protein moiety at a first wavelength, wherein the emission intensity of the first fluorescent protein moiety indicates the pH of the region of the cell in which the indicator is present.
12. The method of claim 11, wherein the polypeptide encoded by the polynucleotide further includes a targeting sequence linked by a peptide bond to the indicator.
13. The method of claim 11, wherein the amino acid sequence of the first fluorescent protein moiety includes the substitutions S65G/S72A/T203Y/H231L in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
14. The method of claim 11, wherein the amino acid sequence of the first fluorescent protein moiety includes the substitutions S65G/V68L/Q69K/S72A/T203Y/H231L in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
15. The method of claim 11, wherein the amino acid sequence of the first fluorescent protein moiety includes the substitutions K26R/F64L/S65T/Y66W/N146I/M153T/V163A/N164H/H231L in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
16. The method of claim 11, wherein the amino acid sequence of the first fluorescent protein moiety includes the substitution H148G in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
17. The method of claim 11, wherein the amino acid sequence of the first fluorescent protein moiety includes the substitution H148Q in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
18. The method of claim 12, wherein the amino acid of the targeting sequence comprises the amino terminal 81 amino acids of human type II membrane-anchored protein galactosyltransferase.
19. The method of claim 12, wherein the targeting sequence comprises the amino terminal 12 amino acids of the presequence of subunit IV of cytochrome c oxidase.
20. A kit useful for the detection of the pH in a sample, the kit comprising carrier means containing one or more containers comprising a first container containing a polynucleotide comprising a nucleotide sequence encoding a functional engineered fluorescent protein whose amino acid sequence is substantially identical to the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2) and whose emission intensity changes as pH varies between 5 and 10.
21. The kit of claim 20, wherein the amino acid sequence of the protein includes the substitutions S65G/S72A/T203Y/H231L in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
22. The kit of claim 20, wherein the amino acid sequence of the protein includes the substitutions S65G/V68L/Q69K/S72A/T203Y/H231L in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
23. The kit of claim 20, wherein the amino acid sequence of the protein includes the substitutions K26R/F64L/S65T/Y66W/N146I/M153T/V163A/N164H/H231L in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
24. The kit of claim 20, wherein the amino acid sequence of the protein includes the substitution H148G in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
25. The kit of claim 20, wherein the amino acid sequence of the protein includes the substitution H148Q in the amino acid sequence of Aequorea green fluorescent protein (SEQ ID NO:2).
US10/457,982 1998-06-09 2003-06-09 Fluorescent protein sensors for measuring the pH of a biological sample Abandoned US20030212265A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/457,982 US20030212265A1 (en) 1998-06-09 2003-06-09 Fluorescent protein sensors for measuring the pH of a biological sample

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/094,359 US6140132A (en) 1998-06-09 1998-06-09 Fluorescent protein sensors for measuring the pH of a biological sample
US09/602,641 US6608189B1 (en) 1998-06-09 2000-06-22 Fluorescent protein sensors for measuring the pH of a biological sample
US10/457,982 US20030212265A1 (en) 1998-06-09 2003-06-09 Fluorescent protein sensors for measuring the pH of a biological sample

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/602,641 Continuation US6608189B1 (en) 1998-06-09 2000-06-22 Fluorescent protein sensors for measuring the pH of a biological sample

Publications (1)

Publication Number Publication Date
US20030212265A1 true US20030212265A1 (en) 2003-11-13

Family

ID=22244686

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/094,359 Expired - Fee Related US6140132A (en) 1998-06-09 1998-06-09 Fluorescent protein sensors for measuring the pH of a biological sample
US09/704,463 Expired - Fee Related US6627449B1 (en) 1998-06-09 2000-10-31 Fluorescent protein sensors for measuring the pH of a biological sample
US10/457,982 Abandoned US20030212265A1 (en) 1998-06-09 2003-06-09 Fluorescent protein sensors for measuring the pH of a biological sample

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/094,359 Expired - Fee Related US6140132A (en) 1998-06-09 1998-06-09 Fluorescent protein sensors for measuring the pH of a biological sample
US09/704,463 Expired - Fee Related US6627449B1 (en) 1998-06-09 2000-10-31 Fluorescent protein sensors for measuring the pH of a biological sample

Country Status (1)

Country Link
US (3) US6140132A (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020164674A1 (en) * 1996-01-31 2002-11-07 The Regents Of The University Of California Tandem fluorescent protein constructs
US20080299565A1 (en) * 2005-12-12 2008-12-04 Schneider Thomas D Probe for Nucleic Acid Sequencing and Methods of Use
US7544776B2 (en) 1996-08-16 2009-06-09 The University Of Oregon Long wavelength engineered fluorescent proteins
US7560287B2 (en) 1996-08-16 2009-07-14 The Regents Of The University Of California Long wavelength engineered fluorescent proteins
US8703734B2 (en) 2005-12-12 2014-04-22 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Nanoprobes for detection or modification of molecules
US9829491B2 (en) 2009-10-09 2017-11-28 The Research Foundation For The State University Of New York pH-insensitive glucose indicator protein
WO2020018946A1 (en) * 2018-07-19 2020-01-23 University Of Washington Systems and methods for accurate optical ph sensing of biofilms

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5625048A (en) * 1994-11-10 1997-04-29 The Regents Of The University Of California Modified green fluorescent proteins
US6140132A (en) * 1998-06-09 2000-10-31 The Regents Of The University Of California Fluorescent protein sensors for measuring the pH of a biological sample
US7687614B2 (en) 2001-02-26 2010-03-30 The Regents Of The University Of California Monomeric and dimeric fluorescent protein variants and methods for making same
US7022826B2 (en) * 2001-02-26 2006-04-04 The Regents Of The University Of California Non-oligomerizing fluorescent proteins
WO2002077011A2 (en) * 2001-03-12 2002-10-03 The State Of Oregon Acting By And Through The State Board Of Higher Education On Behalf Of The University Of Oregon Oxidation-reduction sensitive green fluorescent protein variants
JP3829252B2 (en) * 2001-06-08 2006-10-04 独立行政法人理化学研究所 Fluorescent protein
US20050181464A1 (en) * 2002-04-04 2005-08-18 Affinium Pharmaceuticals, Inc. Novel purified polypeptides from bacteria
WO2004009788A2 (en) * 2002-07-19 2004-01-29 Diversa Corporation Fluorescent proteins, nucleic acids encoding them and methods for making and using them
US9453251B2 (en) 2002-10-08 2016-09-27 Pfenex Inc. Expression of mammalian proteins in Pseudomonas fluorescens
ATE527280T1 (en) * 2003-06-16 2011-10-15 Riken FLUORESCENT PROTEIN AND CHROMEPROTEIN
US20050037364A1 (en) * 2003-08-13 2005-02-17 International Business Machines Corporation Techniques for recording signals
EP1514922A1 (en) * 2003-09-11 2005-03-16 Institut Pasteur Development of an anionic sensor for living cells: application to the human glycine receptor
US8603824B2 (en) 2004-07-26 2013-12-10 Pfenex, Inc. Process for improved protein expression by strain engineering
DK1920248T3 (en) 2005-04-05 2010-12-13 Allergan Inc Clostridial toxin activity assays
US7588907B2 (en) * 2005-06-02 2009-09-15 The Regents Of The University Of Michigan Ubiquitin mediated fluorescence complementation assay
CA2617930A1 (en) 2005-08-09 2007-03-29 Revivicor, Inc. Transgenic ungulates expressing ctla4-ig and uses thereof
CA2671431C (en) * 2006-12-14 2019-03-26 Georgia State University Research Foundation, Inc. Analyte sensors, methods for preparing and using such sensors, and methods of detecting analyte activity
US9580719B2 (en) 2007-04-27 2017-02-28 Pfenex, Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
BRPI0810120A2 (en) 2007-04-27 2014-11-11 Dow Global Technologies Inc PROCESS TO QUICKLY SELECT MICROBIAN HOST FOR THE IDENTIFICATION OF CERTAIN BETTER YIELDS AND / OR QUALITY IN EXPRESSION OF HETEROLOGICAL PROTEINS
WO2011135040A1 (en) 2010-04-30 2011-11-03 F. Hoffmann-La Roche Ag Fluorescent antibody fusion protein, its production and use
WO2013130808A1 (en) 2012-02-29 2013-09-06 D.E. Shaw Research, Llc Methods for screening voltage gated proteins
CA2879103A1 (en) * 2012-07-13 2014-01-16 Jordan L. Holtzman Cellular and animal models for screening therapeutic agents for the treatment of alzheimer's disease
AU2016321298A1 (en) 2015-09-09 2018-03-29 The Trustees Of Columbia University In The City Of New York Reduction of ER-MAM localized APP-C99 and methods of treating Alzheimer's Disease
US10845305B1 (en) 2015-11-20 2020-11-24 Verily Life Sciences Llc Method for increasing sensor resolution by spectrally stacking responsive dyes
WO2022087260A1 (en) * 2020-10-22 2022-04-28 The Regents Of The University Of California Devices and methods for evaluating the viability of embryos

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5625048A (en) * 1994-11-10 1997-04-29 The Regents Of The University Of California Modified green fluorescent proteins
US5777079A (en) * 1994-11-10 1998-07-07 The Regents Of The University Of California Modified green fluorescent proteins
US6054321A (en) * 1996-08-16 2000-04-25 The Regents Of The University Of California Long wavelength engineered fluorescent proteins
US6141132A (en) * 1999-02-02 2000-10-31 Silkroad, Inc. Method of modulating a coherent light source with an electronic digital data stream
US6150176A (en) * 1998-06-09 2000-11-21 The Regents Of The University Of California Fluorescent protein sensors for measuring the pH of a biological sample
US6469154B1 (en) * 1999-05-21 2002-10-22 The Regents Of The University Of California Fluorescent protein indicators
US6627449B1 (en) * 1998-06-09 2003-09-30 The Regents Of The University Of California Fluorescent protein sensors for measuring the pH of a biological sample
US6670449B1 (en) * 1997-02-13 2003-12-30 Memorial Sloan-Kettering Cancer Center Hybrid molecules and their use for optically detecting changes in cellular microenvironments

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5625048A (en) * 1994-11-10 1997-04-29 The Regents Of The University Of California Modified green fluorescent proteins
US5777079A (en) * 1994-11-10 1998-07-07 The Regents Of The University Of California Modified green fluorescent proteins
US6800733B2 (en) * 1994-11-10 2004-10-05 The Regents Of The University Of California Modified green fluorescent proteins
US6066476A (en) * 1994-11-10 2000-05-23 The Regents Of The University Of California Modified green fluorescent proteins
US20020123113A1 (en) * 1994-11-10 2002-09-05 The Regents Of The University Of California Modified green fluorescent proteins
US6403374B1 (en) * 1996-08-16 2002-06-11 The Regents Of The University Of California Long wavelength engineered fluorescent proteins
US6124128A (en) * 1996-08-16 2000-09-26 The Regents Of The University Of California Long wavelength engineered fluorescent proteins
US6077707A (en) * 1996-08-16 2000-06-20 The Regents Of The University Of California Long wavelength engineered fluorescent proteins
US20030036178A1 (en) * 1996-08-16 2003-02-20 The Regents Of The University Of California Long wavelength engineered fluorescent proteins
US6780975B2 (en) * 1996-08-16 2004-08-24 The Regents Of The University Of California Long wavelength engineered fluorescent proteins
US6054321A (en) * 1996-08-16 2000-04-25 The Regents Of The University Of California Long wavelength engineered fluorescent proteins
US20050079525A1 (en) * 1996-08-16 2005-04-14 The Regents Of The University Of California Long wavelength engineered fluorescent proteins
US6670449B1 (en) * 1997-02-13 2003-12-30 Memorial Sloan-Kettering Cancer Center Hybrid molecules and their use for optically detecting changes in cellular microenvironments
US6150176A (en) * 1998-06-09 2000-11-21 The Regents Of The University Of California Fluorescent protein sensors for measuring the pH of a biological sample
US6627449B1 (en) * 1998-06-09 2003-09-30 The Regents Of The University Of California Fluorescent protein sensors for measuring the pH of a biological sample
US6141132A (en) * 1999-02-02 2000-10-31 Silkroad, Inc. Method of modulating a coherent light source with an electronic digital data stream
US6469154B1 (en) * 1999-05-21 2002-10-22 The Regents Of The University Of California Fluorescent protein indicators

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020164674A1 (en) * 1996-01-31 2002-11-07 The Regents Of The University Of California Tandem fluorescent protein constructs
US8679742B2 (en) 1996-01-31 2014-03-25 The Regents Of The University Of California Method for determining the presence of an enzyme in a sample
US20100233726A1 (en) * 1996-01-31 2010-09-16 The Regents Of The University Of California Tandem fluorescent protein constructs
US7544776B2 (en) 1996-08-16 2009-06-09 The University Of Oregon Long wavelength engineered fluorescent proteins
US7560287B2 (en) 1996-08-16 2009-07-14 The Regents Of The University Of California Long wavelength engineered fluorescent proteins
US20100145024A1 (en) * 1996-08-16 2010-06-10 The State Of Oregon, Acting By And Through The State Board Of Higher Education On Behalf Of The Long wavelength engineered fluorescent proteins
US8263412B2 (en) 1996-08-16 2012-09-11 The State Of Oregon, Acting By And Through The State Board Of Higher Education On Behalf Of The University Of Oregon Long wavelength engineered fluorescent proteins
US20110111975A1 (en) * 2005-12-12 2011-05-12 The Government of the U.S.A as represented by the Secretary of the Dept. of Health & Human Services Probe for nucleic acid sequencing and methods of use
US7871777B2 (en) 2005-12-12 2011-01-18 The United States Of America As Represented By The Department Of Health And Human Services Probe for nucleic acid sequencing and methods of use
US20100227913A1 (en) * 2005-12-12 2010-09-09 The Govt. of the U.S.A. as represented by the Sec. of the Deparment of Health and Human Services Nanoprobes for detection or modification of molecules
US8344121B2 (en) 2005-12-12 2013-01-01 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Nanoprobes for detection or modification of molecules
US20080299565A1 (en) * 2005-12-12 2008-12-04 Schneider Thomas D Probe for Nucleic Acid Sequencing and Methods of Use
US8703734B2 (en) 2005-12-12 2014-04-22 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Nanoprobes for detection or modification of molecules
US9829491B2 (en) 2009-10-09 2017-11-28 The Research Foundation For The State University Of New York pH-insensitive glucose indicator protein
WO2020018946A1 (en) * 2018-07-19 2020-01-23 University Of Washington Systems and methods for accurate optical ph sensing of biofilms
US20210263051A1 (en) * 2018-07-19 2021-08-26 University Of Washington Systems and methods for accurate optical ph sensing of biofilms

Also Published As

Publication number Publication date
US6627449B1 (en) 2003-09-30
US6140132A (en) 2000-10-31

Similar Documents

Publication Publication Date Title
US6608189B1 (en) Fluorescent protein sensors for measuring the pH of a biological sample
US20030212265A1 (en) Fluorescent protein sensors for measuring the pH of a biological sample
US6469154B1 (en) Fluorescent protein indicators
JP4232992B2 (en) Long wavelength engineered fluorescent protein
US7060869B2 (en) Fluorescent protein sensors for detection of analytes
JP4215512B2 (en) Non-oligomerized tandem fluorescent protein
CA2283629C (en) Fluorescent protein sensors for detection of analytes
US8263412B2 (en) Long wavelength engineered fluorescent proteins
US7687614B2 (en) Monomeric and dimeric fluorescent protein variants and methods for making same
US7060793B2 (en) Circularly permuted fluorescent protein indicators
WO2000071565A9 (en) Fluorescent protein indicators
US7022826B2 (en) Non-oligomerizing fluorescent proteins
US6699687B1 (en) Circularly permuted fluorescent protein indicators
MXPA98002972A (en) Long wavelength engineered fluorescent proteins
AU2004200425A8 (en) Long wavelength engineered fluorescent proteins

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION