USH2019H1 - Streptococcus pneumoniae gene sequence HI1648 - Google Patents

Streptococcus pneumoniae gene sequence HI1648 Download PDF

Info

Publication number
USH2019H1
USH2019H1 US09/198,284 US19828498A USH2019H US H2019 H1 USH2019 H1 US H2019H1 US 19828498 A US19828498 A US 19828498A US H2019 H USH2019 H US H2019H
Authority
US
United States
Prior art keywords
protein
dna
leu
gene
gly
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
US09/198,284
Inventor
Paul Luther Skatrud
Robert Brown Peery
Patti Jean Treadway
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
Application filed by Individual filed Critical Individual
Priority to US09/198,284 priority Critical patent/USH2019H1/en
Application granted granted Critical
Publication of USH2019H1 publication Critical patent/USH2019H1/en
Abandoned legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56911Bacteria
    • G01N33/56944Streptococcus
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/315Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Streptococcus (G), e.g. Enterococci
    • C07K14/3156Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Streptococcus (G), e.g. Enterococci from Streptococcus pneumoniae (Pneumococcus)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1003Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor
    • C12N15/1006Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor by means of a solid support carrier, e.g. particles, polymers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/74Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora
    • C12N15/746Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora for lactic acid bacteria (Streptococcus; Lactococcus; Lactobacillus; Pediococcus; Enterococcus; Leuconostoc; Propionibacterium; Bifidobacterium; Sporolactobacillus)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1247DNA-directed RNA polymerase (2.7.7.6)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01018Exo-alpha-sialidase (3.2.1.18), i.e. trans-sialidase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/195Assays involving biological materials from specific organisms or of a specific nature from bacteria
    • G01N2333/315Assays involving biological materials from specific organisms or of a specific nature from bacteria from Streptococcus (G), e.g. Enterococci
    • 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
    • Y10S435/8215Microorganisms
    • Y10S435/822Microorganisms using bacteria or actinomycetales

Definitions

  • This invention provides isolated DNA sequences, proteins encoded thereby, and methods of using said DNA and protein in a variety of applications.
  • Penicillin resistance in Streptococcus pneumoniae has been particularly problematic. This organism causes upper respiratory tract infections. Modification of a penicillin-binding protein (PBP) underlies resistance to penicillin in the majority of cases.
  • PBP penicillin-binding protein
  • Streptccoccus pneumoniae While researchers continue to develop antibiotics effective against a number of microorganisms, Streptccoccus pneumoniae has been more refractory. In part, this is because Streptococcus pneumoniae is highly recombinogenic and readily takes up exogenous DNA from its surroundings. Thus, there is a need for new antibacterial compounds and new targets for antibacterial therapy in Streptococcus pneumoniae.
  • the present invention relates to an isolated gene and encoded protein from S. pneumoniae.
  • the invention enables: (1) preparation of probes and primers for use in hybridizations and PCR amplifications, (2) production of proteins and RNAs encoded by said gene and related nucleic acids, and (3) methods to identify compounds that bind and/or inhibit said protein(s).
  • the present invention relates to an isolated nucleic acid molecule encoding HI1648 protein.
  • the invention relates to a nucleic acid molecule comprising the nucleotide sequence identified as SEQ ID NO:1 or SEQ ID NO:3.
  • the present invention relates to a nucleic acid that encodes SEQ ID NO:2.
  • the present invention relates to an isolated protein molecule, wherein said protein molecule comprises the sequence identified as SEQ ID NO:2.
  • the present invention relates to a recombinant DNA vector that incorporates the HI1648 gene in operable linkage to gene expression sequences enabling the gene to be transcribed and translated in a host cell.
  • the present invention relates to host cells that have been transformed or transfected with the cloned HI1648 gene such that said gene is expressed in the host cell.
  • This invention also provides a method of determining whether a nucleic acid sequence of the present invention, or fragment thereof, is present in a sample, comprising contacting the sample, under suitable hybridization conditions, with a nucleic acid probe of the present invention.
  • the present invention relates to a method for identifying compounds that bind and/or inhibit the HI1648 protein.
  • ORF (i.e. “open reading frame”) designates a region of genomic DNA beginning with a Met or other initiation codon and terminating with a translation stop codon, that potentially encodes a protein product.
  • Partial ORF means a portion of an ORF as disclosed herein such that the initiation codon, the stop codon, or both are not disclosed.
  • Consensus sequence refers to an amino acid or nucleotide sequence that may suggest the biological function of a protein, DNA, or RNA molecule. Consensus sequences are identified by comparing proteins, RNAs, and gene homologues from different species.
  • cleavage or “restriction” of DNA refers to the catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA (viz. sequence-specific endonucleases).
  • restriction enzymes used herein are commercially available and their reaction conditions, cofactors, and other requirements are used in the manner well known to one of ordinary skill in the art. Appropriate buffers and substrate amounts for particular restriction enzymes are specified by the manufacturer or can readily be found in the literature
  • Essential genes or “essential ORFs” or “essential proteins” refer to genomic information or the protein(s) or RNAs encoded thereby, that when disrupted by knockout mutation, or by other mutation, result in a loss of viability of cells harboring said mutation.
  • Non-essential genes or “non-essential ORFs” or “non-essential proteins” refer to genomic information or the protein(s) or RNAs encoded therefrom which when disrupted by knockout mutation, or other mutation, do not result in a loss of viability of cells harboring said mutation.
  • Minimal gene set refers to a genus comprising about 256 genes conserved among different bacteria such as M. genitalium and H. influenzae. The minimal gene set may be necessary and sufficient to sustain life. See e.g. A. Mushegian and E. Koonin, “A minimal gene set for cellular life derived by comparison of complete bacterial genomes” Proc. Nat. Acad. Sci. 93, 10268-273 (1996).
  • “Knockout mutant” or “knockout mutation” as used herein refers to an in vitro engineered disruption of a region of native chromosomal DNA, typically within a protein coding region, such that a foreign piece of DNA is inserted within the native sequence.
  • a knockout mutation occurring in a protein coding region prevents expression of the wild-type protein. This usually leads to loss of the function provided by the protein.
  • a “knockout cassette” refers to a fragment of native chromosomal DNA having cloned therein a foreign piece of DNA that may provide a selectable marker.
  • plasmid refers to an extrachromosomal genetic element.
  • the starting plasmids herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available plasmids in accordance with published procedures.
  • equivalent plasmids to those described are known in the art and will be apparent to the ordinarily skilled artisan.
  • Recombinant DNA cloning vector refers to any autonomously replicating agent, including, but not limited to, plasmids and phages, comprising a DNA molecule to which one or more additional DNA segments can or have been added.
  • recombinant DNA expression vector refers to any recombinant DNA cloning vector, for example a plasmid or phage, in which a promoter and other regulatory elements are present to enable transcription of the inserted DNA.
  • vector refers to a nucleic acid compound used for introducing exogenous DNA into host cells.
  • a vector comprises a nucleotide sequence which may encode one or more protein molecules. Plasmids, cosmids, viruses, and bacteriophages, in the natural state or which have undergone recombinant engineering, are examples of commonly used vectors.
  • complementary refers to the capacity of purine and pyrimidine nucleotides to associate through hydrogen bonding to form double stranded nucleic acid molecules.
  • the following base pairs are related by complementarity: guanine and cytosine; adenine and thymine; and adenine and uracil.
  • complementary applies to all base pairs comprising two single-stranded nucleic acid molecules. “Partially complementary” means one of two single-stranded nucleic acid molecules is shorter than the other, such that one of the molecules remains partially single-stranded.
  • Oligonucleotide refers to a short nucleotide chain comprising from about 2 to about 25 nucleotides.
  • isolated nucleic acid compound refers to any RNA or DNA sequence, however constructed or synthesized, which is locationally distinct from its natural location.
  • a “primer” is a nucleic acid fragment which functions as an initiating substrate for enzymatic or synthetic elongation of, for example, a nucleic acid molecule.
  • promoter refers to a DNA sequence which directs transcription of DNA to RNA.
  • a “probe” as used herein is a labeled nucleic acid compound which can be used to hybridize with another nucleic acid compound.
  • hybridization or “hybridize” as used herein refers to the process by which a single-stranded nucleic acid molecule joins with a complementary strand through nucleotide base pairing.
  • substantially purified means a specific isolated nucleic acid or protein, or fragment thereof, in which substantially all contaminants (i.e. substances that differ from said specific molecule) have been separated from said nucleic acid or protein.
  • a protein may, but not necessarily, be “substantially purified” by the IMAC method as described herein.
  • “Selective hybridization” refers to hybridization under conditions of high stringency.
  • the degree of hybridization between nucleic acid molecules depends upon, for example, the degree of complementarity, the string ency of hybridization, and the length of hybridizing strands.
  • stringency relates to nucleic acid hybridization conditions. High stringency conditions disfavor non-homologous base pairing. Low stringency conditions have the opposite effect. Stringency may be altered, for example, by changes in temperature and salt concentration. Typical high stringency conditions comprise hybridizing at 50° C. to 65° C. in 5 ⁇ SSPE and 50% formamide, and washing at 50° C. to 65° C. in 0.5 ⁇ SSPE; typical low stringency conditions comprise hybridizing at 35° C. to 37° C. in 5 ⁇ SSPE and 40% to 45% formamide and washing at 42° C. in 1 ⁇ -2 ⁇ SSPE.
  • SSPE denotes a hybridization and wash solution comprising sodium chloride, sodium phosphate, and EDTA, at pH 7.4.
  • a 20 ⁇ solution of SSPE is made by dissolving 174 g of NaCl, 27.6 g of NaH 2 PO 4 .H 2 O, and 7.4 g of EDTA in 800 ml of H 2 O. The pH is adjusted with NaOH and the volume brought to 1 liter.
  • SSC denotes. a hybridization and wash solution comprising sodium chloride and sodium citrate at pH 7.
  • a 20 ⁇ solution of SSC is made by dissolving 175 g of NaCl and 88 g of sodium citrate in 800 ml of H 2 O. The volume is brought to 1 liter after adjusting the pH with 10N NaOH.
  • the HI1648 gene disclosed herein (SEQ ID NO:1) and related nucleic acids (e.g. SEQ ID NO:3 and SEQ ID NO:4) encode a protein of unknown function. Since knockout mutations of the HI1648 gene produce a loss in viability, the 1648 protein product may be essential for viability.
  • the proteins categorized as “minimal gene set” counterparts are homologous to a set of highly conserved proteins found in other bacteria.
  • the minimal gene set proteins are thought to be essential for viability and are useful targets for the development of new antibacterial compounds.
  • the proteins of this invention are purified, and used in a screen to identify compounds that bind and/or inhibit the activity of said proteins.
  • a variety of suitable screens are contemplated for this purpose.
  • the protein(s) can be labeled by known techniques, such as radiolabeling or fluorescent tagging, or by labeling with biotin/avidin. Thereafter, binding of a test compound to a labeled protein can be determined by any suitable means, well known to the skilled artisan.
  • DNA molecules of this invention can be generated by general cloning methods. PCR amplification using oligonucleotide primers targeted to any suitable region of SEQ ID NO:1 is preferred. Methods for PCR amplification are widely known in the art. See e.g. PCR Protocols: A Guide to Method and Application, Ed. M. Innis et al., Academic Press (1990) or U.S. Pat. No. 4,889,818, which hereby is incorporated by reference.
  • a PCR comprises DNA, suitable enzymes, primers, and buffers, and is conveniently carried out in a DNA Thermal Cycler (Perkin Elmer Cetus, Norwalk, Conn.). A positive PCR result is determined by, for example, detecting an appropriately-sized DNA fragment following agarose gel electrophoresis.
  • the DNAs of the present invention may also be produced using synthetic methods well known in the art. (See, e.g., E. L. Brown, R. Belagaje, M. J. Ryan, and H. G. Khorana, Methods in Enzymology, 68:109-151 (1979)).
  • An apparatus such as the Applied Biosystems Model 380A or 380B DNA synthesizers (Applied Biosystems, Inc., 850 Lincoln Center Drive, Foster City, Calif. 94404) may be used to synthesize DNA.
  • Synthetic methods rely upon phosphotriester chemistry [See, e.g., M. J. Gait, ed., Oligonucleotide Synthesis, A Practical Approach, (1984)], or phosphoramidite chemistry.
  • the present invention relates further to substantially purified proteins encoded by the gene disclosed herein.
  • proteins can be synthesized by different methods, for example, chemical methods or recombinant methods, as described in U.S. Pat. No. 4,617,149, which hereby is incorporated by reference.
  • polypeptides may be synthesized by solid-phase methodology utilizing an Applied Biosystems 430A peptide synthesizer (Applied Biosystems, Foster City, Calif.) and synthesis cycles supplied by Applied Biosystems.
  • Protected amino acids such as t-butoxycarbonyl-protected amino acids, and other reagents are commercially available from many chemical supply houses.
  • the proteins of the present invention can also be made by recombinant DNA methods. Recombinant methods are preferred if a high yield is desired. Recombinant methods involve expressing the cloned gene in a suitable host cell. The gene is introduced into the host cell by any suitable means, well known to those skilled in the art. While chromosomal integration of the cloned gene is within the scope of the present invention, it is preferred that the cloned gene be maintained extra-chromosomally, as part of a vector in which the gene is in operable-linkage to a promoter.
  • Recombinant methods can also be used to overproduce a membrane-bound or membrane-associated protein.
  • membranes prepared from recombinant cells expressing such proteins provide an enriched source of the protein.
  • Procaryotes are generally used for cloning DNA sequences and for constructing vectors.
  • Escherichia coli K12 strain 294 ATCC No. 31446
  • E. coli E. coli
  • Bacillus subtilis enterobacteriaceae
  • Salmonella typhimurium Salmonella typhimurium
  • Serratia marcescans various Pseudomonas species
  • Streptococcus and Streptocmyces are also contemplated.
  • a gene For effective recombinant protein production, a gene must be linked to a promoter sequence.
  • Suitable bacterial promoters include b-lactamase [e.g. vector pGX2907, ATCC 39344, contains a replicon and b-lactamase gene], lactose systems [Chang et al., Nature (London), 275:615. (1978); Goeddel et al., Nature (London), 281:544 (1979)], alkaline phosphatase, and the tryptophan (trp) promoter system [vector pATH1 (ATCC 37695)] designed for the expression of a trpE fusion protein.
  • Hybrid promoters such as the tac promoter (isolatable from plasmid pDR540, ATCC37282) are also suitable. Promoters for use in bacterial systems also will contain a Shine-Dalgarno sequence, operably linked to the DNA encoding the desired polypeptides. These examples are illustrative rather than limiting.
  • yeast Saccharomyces cerevisiae is commonly used.
  • Other yeasts, such as Kluyveromyces lactis are also suitable.
  • the plasmid YRp7 (ATCC-40053), for example, may be used. See, e.g., L. Stinchcomb, et al., Nature, 282:39 (1979); J. Kingsman et al., Gene, 7:141 (1979); S. Tschemper et al., Gene, 10:157 (1980).
  • Plasmid YRp7 contains the TRP1 gene, a selectable marker for a trp1 mutant.
  • An expression vector carrying a nucleic acid or gene of the present invention is transformed or transfected into a suitable host cell using standard methods.
  • Cells that contain the vector are propagated under conditions suitable for expression of a recombinant protein.
  • suitable growth conditions would incorporate the appropriate inducer.
  • the recombinantly-produced protein may be purified from cellular extracts of transformed cells by any suitable means.
  • a gene is modified at the 5′ end, or at some other position, such that the encoded protein incorporates several histidine residues (viz. “histidine tag”).
  • This “histidine tag” enables “immobilized metal ion affinity chromatography” (IMAC), a single-step protein purification method described in U.S. Pat. No. 4,569,794, which hereby is incorporated by reference.
  • IMAC immobilized metal ion affinity chromatography
  • the proteins of the invention can be encoded by a large genus of different nucleic acid sequences. This invention further comprises said genus.
  • ribonucleic acid compounds of the invention may be prepared using the polynucleotide synthetic methods discussed supra, or they may be prepared enzymatically using RNA polymerase to transcribe a DNA template.
  • RNA polymerase from the bacteriophage T7 or the bacteriophage SP6. These RNA polymerases are highly specific, requiring the insertion of bacteriophage-specific sequences at the 5′ end of a template. See, J. Sambrook, et al., supra, at 18.82-18.84.
  • This invention also provides nucleic acids that are complementary to the sequences disclosed herein.
  • the present invention also provides probes and primers, useful for a variety of molecular biology techniques including, for example, hybridization screens of genomic or subgenomic libraries, or detection and quantification of mRNA species as a means to analyze gene expression.
  • a nucleic acid compound is provided comprising any of the sequences disclosed herein, or a complementary sequence thereof, or a fragment thereof, which is at least 15 base pairs in length, and which will hybridize selectively to Streptococcus pneumoniae DNA or mRNA.
  • the 15 or more base pair compound is DNA.
  • a probe or primer length of at least 15 base pairs is dictated by theoretical and practical considerations. See e.g. B. Wallace and G. Miyada, “Oligonucleotide Probes for the Screening of Recombinant DNA Libraries,” In Methods in Enzymology, Vol. 152, 432-442, Academic Press (1987).
  • probes and primers of this invention can be prepared by methods well known to those skilled in the art (See e.g. Sambrook et al. supra). In a preferred embodiment the probes and primers are synthesized by the polymerase chain reaction (PCR).
  • PCR polymerase chain reaction
  • the present invention also relates to recombinant DNA cloning vectors and expression vectors comprising the nucleic acids of the present invention.
  • Preferred nucleic acid vectors are those that comprise DNA.
  • the skilled artisan understands that choosing the most appropriate cloning vector or expression vector depends on the availability of restriction sites, the type of host cell into which the vector is to be transfected or transformed, the purpose of transfection or transformation (e.g., stable transformation as an extrachromosomal element, or integration into a host chromosome), the presence or absence of readily assayable or selectable markers (e.g., antibiotic resistance and metabolic markers of one type and another), and the number of gene copies desired in the host cell.
  • readily assayable or selectable markers e.g., antibiotic resistance and metabolic markers of one type and another
  • Suitable vectors comprise RNA viruses, DNA viruses, lytic bacteriophages, lysogenic bacteriophages, stable bacteriophages, plasmids, viroids, and the like.
  • the most preferred vectors are plasmids.
  • Host cells harboring the nucleic acids disclosed herein are also provided by the present invention.
  • a preferred host is E. coli transfected or transformed with a vector comprising a nucleic acid of the present invention.
  • the invention also provides a host cell capable of expressing a gene described herein, said method comprising transforming or otherwise introducing into a host cell a recombinant DNA vector comprising an isolated DNA sequence that encodes said gene.
  • the preferred host cell is any strain of E. coli that can accommodate high level expression of an exogenously introduced gene.
  • Transformed host cells are cultured under conditions well known to skilled artisans, such that said gene is expressed, thereby producing the encoded protein in the recombinant host cell.
  • a method for identifying such compounds comprises contacting a suitable protein or membrane preparation with a test compound and monitoring by any suitable means an interaction and/or inhibition of a protein of this invention.
  • the instant invention provides a screen for compounds that interact with the proteins of the invention, said screen comprising:
  • the screening method of this invention may be adapted to automated procedures such as a PANDEX® (Baxter-Dade Diagnostics) system, allowing for efficient high-volume screening of compounds.
  • PANDEX® Boxter-Dade Diagnostics
  • a protein is prepared as described herein, preferably using recombinant DNA technology.
  • a test compound is introduced into a reaction vessel containing said protein.
  • the reaction/interaction of said protein and said compound is monitored by any suitable means.
  • a radioactively-labeled or. chemically-labeled compound or protein is used.
  • a specific association between the test compound and protein is monitored by any suitable means.
  • HI1648 is prepared as described herein, preferably using recombinant DNA technology.
  • a test compound is introduced into a reaction vessel containing the HI1648 protein or fragment thereof. Binding of HI1648 by a test compound is determined by any suitable means. For example, in one method radioactively-labeled or chemically-labeled test compound may be used. Binding of the protein by the compound is assessed, for example, by quantifying bound label versus unbound label using any suitable method. Binding of a test compound may also be carried out by a method disclosed in U.S. Pat. No. 5,585,277, which hereby is incorporated by reference.
  • binding of a test compound to a protein is assessed by monitoring the ratio of folded protein to unfolded protein, for example by monitoring sensitivity of said protein to a protease, or amenability to binding of said protein by a specific antibody against the folded state of the protein.
  • a ligand that binds HI1648, or related fragment thereof is identified, for example, by combining a test ligand with HI1648 under conditions that cause the protein to exist in a ratio of folded to unfolded states. If the test ligand binds the folded state of the protein, the relative amount of folded protein will be higher than in the case of a test ligand that does not bind the protein.
  • the ratio of protein in the folded versus unfolded state is easily determinable by, for example, susceptibility to digestion by a protease, or binding to a specific antibody, or binding to chaperoning protein, or binding to any suitable surface.
  • An expression vector suitable for expressing S. pneumoniae HI1648 in a variety of procaryotic host cells, such as E. coli, is easily made.
  • the vector contains an origin of replication (Ori), an ampicillin resistance gene (Amp) useful for selecting cells which have incorporated the vector following a tranformation procedure, and further comprises the T7 promoter and T7 terminator sequences in operable linkage to the HI1648 coding region.
  • Plasmid pET11A (obtained from Novogen, Madison, Wis.) is a suitable parent plasmid. pET11A is linearized by restriction with endonucleases NdeI and BamHI.
  • Linearized pET11A is ligated to a DNA fragment bearing NdeI and BamHI sticky ends and comprising the coding region of the S. pneumoniae HI1648 (SEQ ID NO:1).
  • the coding region for HI1648 is easily produced by PCR technology using suitably designed primers to the ends of the coding region specified in SEQ ID NO:1.
  • the HI1648 encoding nucleic acid used in this construct is slightly modified at the 5′ end (amino terminus of encoded protein) in order to simplify purification of the encoded protein product.
  • an oligonucleotide encoding 8 histidine residues is inserted after the ATG start codon. Placement of the histidine residues at the amino terminus of the encoded protein serves to enable the IMAC one-step protein purification procedure.
  • An expression vector that carries HI1648 from the S. pneumoniae genome as disclosed herein and which H11648 is operably-linked to an expression promoter is transformed into E. coli BL21 (DE3) (hsdS gal lcIts857 ind1Sam7nin5lacUV5-T7 gene 1) using standard methods (see Example 4). Transformants, selected for resistance to ampicillin, are chosen at random and tested for the presence of the vector by agarose gel electrophoresis using quick plasmid preparations. Colonies which contain the vector are grown in L broth and the protein product encoded by the vector-borne ORF is purified by immobilized metal ion affinity chromatography (IMAC), essentially as described in U.S. Pat. No. 4,569,794.
  • IMAC immobilized metal ion affinity chromatography
  • the IMAC column is prepared as follows.
  • a metal-free chelating resin e.g. Sepharose 6B IDA, Pharmacia
  • a suitable metal ion e.g. Ni(II), Co(II), or Cu(II)
  • the column is then ready to receive a crude cellular extract containing the recombinant protein product.
  • the bound protein is eluted in any suitable buffer at pH 4.3, or preferably with an imidizole-containing buffer at pH 7.5.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Medicinal Chemistry (AREA)
  • Plant Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Virology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pulmonology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Enzymes And Modification Thereof (AREA)

Abstract

The invention provides isolated nucleic acid compounds encoding HI1648 of Streptococcus pneumoniae. Also provided are vectors and transformed host cells for expressing the encoded protein, and a method for identifying compounds that bind and/or inhibit said protein.

Description

This application claims the benefit of divisional of Ser. No. 08/987,122, filed on Dec. 8, 1997 U.S. Provisional Application No. 60/036,281, filed Dec. 13, 1996.
BACKGROUND OF THE INVENTION
This invention provides isolated DNA sequences, proteins encoded thereby, and methods of using said DNA and protein in a variety of applications.
Widespread antibiotic resistance in common pathogenic bacterial species has justifiably alarmed the medical and research communities. Frequently, resistant organisms are co-resistant to several antibacterial agents. Penicillin resistance in Streptococcus pneumoniae has been particularly problematic. This organism causes upper respiratory tract infections. Modification of a penicillin-binding protein (PBP) underlies resistance to penicillin in the majority of cases. Combating resistance to antibiotic agents will require research into the molecular biology of pathogenic organisms. The goal of such research will be to identify new antibacterial agents.
While researchers continue to develop antibiotics effective against a number of microorganisms, Streptccoccus pneumoniae has been more refractory. In part, this is because Streptococcus pneumoniae is highly recombinogenic and readily takes up exogenous DNA from its surroundings. Thus, there is a need for new antibacterial compounds and new targets for antibacterial therapy in Streptococcus pneumoniae.
BRIEF SUMMARY OF THE INVENTION
The present invention relates to an isolated gene and encoded protein from S. pneumoniae. The invention enables: (1) preparation of probes and primers for use in hybridizations and PCR amplifications, (2) production of proteins and RNAs encoded by said gene and related nucleic acids, and (3) methods to identify compounds that bind and/or inhibit said protein(s).
In one embodiment the present invention relates to an isolated nucleic acid molecule encoding HI1648 protein.
In another embodiment, the invention relates to a nucleic acid molecule comprising the nucleotide sequence identified as SEQ ID NO:1 or SEQ ID NO:3.
In another embodiment, the present invention relates to a nucleic acid that encodes SEQ ID NO:2.
In another embodiment the present invention relates to an isolated protein molecule, wherein said protein molecule comprises the sequence identified as SEQ ID NO:2.
In yet another embodiment, the present invention relates to a recombinant DNA vector that incorporates the HI1648 gene in operable linkage to gene expression sequences enabling the gene to be transcribed and translated in a host cell.
In still another embodiment the present invention relates to host cells that have been transformed or transfected with the cloned HI1648 gene such that said gene is expressed in the host cell.
This invention also provides a method of determining whether a nucleic acid sequence of the present invention, or fragment thereof, is present in a sample, comprising contacting the sample, under suitable hybridization conditions, with a nucleic acid probe of the present invention.
In a still further embodiment, the present invention relates to a method for identifying compounds that bind and/or inhibit the HI1648 protein.
DETAILED DESCRIPTION OF THE INVENTION
“ORF” (i.e. “open reading frame”) designates a region of genomic DNA beginning with a Met or other initiation codon and terminating with a translation stop codon, that potentially encodes a protein product. “Partial ORF” means a portion of an ORF as disclosed herein such that the initiation codon, the stop codon, or both are not disclosed.
“Consensus sequence” refers to an amino acid or nucleotide sequence that may suggest the biological function of a protein, DNA, or RNA molecule. Consensus sequences are identified by comparing proteins, RNAs, and gene homologues from different species.
The terms “cleavage” or “restriction” of DNA refers to the catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA (viz. sequence-specific endonucleases). The various restriction enzymes used herein are commercially available and their reaction conditions, cofactors, and other requirements are used in the manner well known to one of ordinary skill in the art. Appropriate buffers and substrate amounts for particular restriction enzymes are specified by the manufacturer or can readily be found in the literature
“Essential genes” or “essential ORFs” or “essential proteins” refer to genomic information or the protein(s) or RNAs encoded thereby, that when disrupted by knockout mutation, or by other mutation, result in a loss of viability of cells harboring said mutation.
“Non-essential genes” or “non-essential ORFs” or “non-essential proteins” refer to genomic information or the protein(s) or RNAs encoded therefrom which when disrupted by knockout mutation, or other mutation, do not result in a loss of viability of cells harboring said mutation.
“Minimal gene set” refers to a genus comprising about 256 genes conserved among different bacteria such as M. genitalium and H. influenzae. The minimal gene set may be necessary and sufficient to sustain life. See e.g. A. Mushegian and E. Koonin, “A minimal gene set for cellular life derived by comparison of complete bacterial genomes” Proc. Nat. Acad. Sci. 93, 10268-273 (1996).
“Knockout mutant” or “knockout mutation” as used herein refers to an in vitro engineered disruption of a region of native chromosomal DNA, typically within a protein coding region, such that a foreign piece of DNA is inserted within the native sequence. A knockout mutation occurring in a protein coding region prevents expression of the wild-type protein. This usually leads to loss of the function provided by the protein. A “knockout cassette” refers to a fragment of native chromosomal DNA having cloned therein a foreign piece of DNA that may provide a selectable marker.
The term “plasmid” refers to an extrachromosomal genetic element. The starting plasmids herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available plasmids in accordance with published procedures. In addition, equivalent plasmids to those described are known in the art and will be apparent to the ordinarily skilled artisan.
“Recombinant DNA cloning vector” as used herein refers to any autonomously replicating agent, including, but not limited to, plasmids and phages, comprising a DNA molecule to which one or more additional DNA segments can or have been added.
The term “recombinant DNA expression vector” as used herein refers to any recombinant DNA cloning vector, for example a plasmid or phage, in which a promoter and other regulatory elements are present to enable transcription of the inserted DNA.
The term “vector” as used herein refers to a nucleic acid compound used for introducing exogenous DNA into host cells. A vector comprises a nucleotide sequence which may encode one or more protein molecules. Plasmids, cosmids, viruses, and bacteriophages, in the natural state or which have undergone recombinant engineering, are examples of commonly used vectors.
The terms “complementary” or “complementarity” as used herein refer to the capacity of purine and pyrimidine nucleotides to associate through hydrogen bonding to form double stranded nucleic acid molecules. The following base pairs are related by complementarity: guanine and cytosine; adenine and thymine; and adenine and uracil. As used herein, “complementary” applies to all base pairs comprising two single-stranded nucleic acid molecules. “Partially complementary” means one of two single-stranded nucleic acid molecules is shorter than the other, such that one of the molecules remains partially single-stranded.
“Oligonucleotide” refers to a short nucleotide chain comprising from about 2 to about 25 nucleotides.
“Isolated nucleic acid compound” refers to any RNA or DNA sequence, however constructed or synthesized, which is locationally distinct from its natural location.
A “primer” is a nucleic acid fragment which functions as an initiating substrate for enzymatic or synthetic elongation of, for example, a nucleic acid molecule.
The term “promoter” refers to a DNA sequence which directs transcription of DNA to RNA.
A “probe” as used herein is a labeled nucleic acid compound which can be used to hybridize with another nucleic acid compound.
The term “hybridization” or “hybridize” as used herein refers to the process by which a single-stranded nucleic acid molecule joins with a complementary strand through nucleotide base pairing.
“Substantially purified” as used herein means a specific isolated nucleic acid or protein, or fragment thereof, in which substantially all contaminants (i.e. substances that differ from said specific molecule) have been separated from said nucleic acid or protein. For example, a protein may, but not necessarily, be “substantially purified” by the IMAC method as described herein.
“Selective hybridization” refers to hybridization under conditions of high stringency. The degree of hybridization between nucleic acid molecules depends upon, for example, the degree of complementarity, the string ency of hybridization, and the length of hybridizing strands.
The term “stringency” relates to nucleic acid hybridization conditions. High stringency conditions disfavor non-homologous base pairing. Low stringency conditions have the opposite effect. Stringency may be altered, for example, by changes in temperature and salt concentration. Typical high stringency conditions comprise hybridizing at 50° C. to 65° C. in 5× SSPE and 50% formamide, and washing at 50° C. to 65° C. in 0.5× SSPE; typical low stringency conditions comprise hybridizing at 35° C. to 37° C. in 5× SSPE and 40% to 45% formamide and washing at 42° C. in 1×-2× SSPE.
“SSPE” denotes a hybridization and wash solution comprising sodium chloride, sodium phosphate, and EDTA, at pH 7.4. A 20× solution of SSPE is made by dissolving 174 g of NaCl, 27.6 g of NaH2PO4.H2O, and 7.4 g of EDTA in 800 ml of H2O. The pH is adjusted with NaOH and the volume brought to 1 liter.
“SSC” denotes. a hybridization and wash solution comprising sodium chloride and sodium citrate at pH 7. A 20× solution of SSC is made by dissolving 175 g of NaCl and 88 g of sodium citrate in 800 ml of H2O. The volume is brought to 1 liter after adjusting the pH with 10N NaOH.
The HI1648 gene disclosed herein (SEQ ID NO:1) and related nucleic acids (e.g. SEQ ID NO:3 and SEQ ID NO:4) encode a protein of unknown function. Since knockout mutations of the HI1648 gene produce a loss in viability, the 1648 protein product may be essential for viability.
The proteins categorized as “minimal gene set” counterparts are homologous to a set of highly conserved proteins found in other bacteria. The minimal gene set proteins are thought to be essential for viability and are useful targets for the development of new antibacterial compounds.
In one embodiment, the proteins of this invention are purified, and used in a screen to identify compounds that bind and/or inhibit the activity of said proteins. A variety of suitable screens are contemplated for this purpose. For example, the protein(s) can be labeled by known techniques, such as radiolabeling or fluorescent tagging, or by labeling with biotin/avidin. Thereafter, binding of a test compound to a labeled protein can be determined by any suitable means, well known to the skilled artisan.
Skilled artisans will recognize that the DNA molecules of this invention, or fragments thereof, can be generated by general cloning methods. PCR amplification using oligonucleotide primers targeted to any suitable region of SEQ ID NO:1 is preferred. Methods for PCR amplification are widely known in the art. See e.g. PCR Protocols: A Guide to Method and Application, Ed. M. Innis et al., Academic Press (1990) or U.S. Pat. No. 4,889,818, which hereby is incorporated by reference. A PCR comprises DNA, suitable enzymes, primers, and buffers, and is conveniently carried out in a DNA Thermal Cycler (Perkin Elmer Cetus, Norwalk, Conn.). A positive PCR result is determined by, for example, detecting an appropriately-sized DNA fragment following agarose gel electrophoresis.
The DNAs of the present invention may also be produced using synthetic methods well known in the art. (See, e.g., E. L. Brown, R. Belagaje, M. J. Ryan, and H. G. Khorana, Methods in Enzymology, 68:109-151 (1979)). An apparatus such as the Applied Biosystems Model 380A or 380B DNA synthesizers (Applied Biosystems, Inc., 850 Lincoln Center Drive, Foster City, Calif. 94404) may be used to synthesize DNA. Synthetic methods rely upon phosphotriester chemistry [See, e.g., M. J. Gait, ed., Oligonucleotide Synthesis, A Practical Approach, (1984)], or phosphoramidite chemistry.
Protein Production Methods
The present invention relates further to substantially purified proteins encoded by the gene disclosed herein.
Skilled artisans will recognize that proteins can be synthesized by different methods, for example, chemical methods or recombinant methods, as described in U.S. Pat. No. 4,617,149, which hereby is incorporated by reference.
The principles of solid phase chemical synthesis of polypeptides are well known in the art and may be found in general texts relating to this area. See, e.g., H. Dugas and C. Penney, Bioorganic Chemistry (1981) Springer-Verlag, N.Y., 54-92. Peptides may be synthesized by solid-phase methodology utilizing an Applied Biosystems 430A peptide synthesizer (Applied Biosystems, Foster City, Calif.) and synthesis cycles supplied by Applied Biosystems. Protected amino acids, such as t-butoxycarbonyl-protected amino acids, and other reagents are commercially available from many chemical supply houses.
The proteins of the present invention can also be made by recombinant DNA methods. Recombinant methods are preferred if a high yield is desired. Recombinant methods involve expressing the cloned gene in a suitable host cell. The gene is introduced into the host cell by any suitable means, well known to those skilled in the art. While chromosomal integration of the cloned gene is within the scope of the present invention, it is preferred that the cloned gene be maintained extra-chromosomally, as part of a vector in which the gene is in operable-linkage to a promoter.
Recombinant methods can also be used to overproduce a membrane-bound or membrane-associated protein. In some cases, membranes prepared from recombinant cells expressing such proteins provide an enriched source of the protein.
Expressing Recombinant Proteins in Procaryotic and Eucaryotic Host Cells
Procaryotes are generally used for cloning DNA sequences and for constructing vectors. For example, the Escherichia coli K12 strain 294 (ATCC No. 31446) is particularly useful for expression of foreign proteins.
Other strains of E. coli, bacilli such as Bacillus subtilis, enterobacteriaceae such as Salmonella typhimurium or Serratia marcescans, various Pseudomonas species may also be employed as host cells in cloning and expressing the recombinant proteins of this invention. Also contemplated are various strains of Streptococcus and Streptocmyces.
For effective recombinant protein production, a gene must be linked to a promoter sequence. Suitable bacterial promoters include b-lactamase [e.g. vector pGX2907, ATCC 39344, contains a replicon and b-lactamase gene], lactose systems [Chang et al., Nature (London), 275:615. (1978); Goeddel et al., Nature (London), 281:544 (1979)], alkaline phosphatase, and the tryptophan (trp) promoter system [vector pATH1 (ATCC 37695)] designed for the expression of a trpE fusion protein. Hybrid promoters such as the tac promoter (isolatable from plasmid pDR540, ATCC37282) are also suitable. Promoters for use in bacterial systems also will contain a Shine-Dalgarno sequence, operably linked to the DNA encoding the desired polypeptides. These examples are illustrative rather than limiting.
A variety of mammalian cells and yeasts are also suitable hosts. The yeast Saccharomyces cerevisiae is commonly used. Other yeasts, such as Kluyveromyces lactis, are also suitable. For expression of recombinant genes in Saccharomyces, the plasmid YRp7 (ATCC-40053), for example, may be used. See, e.g., L. Stinchcomb, et al., Nature, 282:39 (1979); J. Kingsman et al., Gene, 7:141 (1979); S. Tschemper et al., Gene, 10:157 (1980). Plasmid YRp7 contains the TRP1 gene, a selectable marker for a trp1 mutant.
Purification of Recombinantly-Produced Protein
An expression vector carrying a nucleic acid or gene of the present invention is transformed or transfected into a suitable host cell using standard methods. Cells that contain the vector are propagated under conditions suitable for expression of a recombinant protein. For example, if the gene is under the control of an inducible promoter, then suitable growth conditions would incorporate the appropriate inducer. The recombinantly-produced protein may be purified from cellular extracts of transformed cells by any suitable means.
In a preferred process for protein purification a gene is modified at the 5′ end, or at some other position, such that the encoded protein incorporates several histidine residues (viz. “histidine tag”). This “histidine tag” enables “immobilized metal ion affinity chromatography” (IMAC), a single-step protein purification method described in U.S. Pat. No. 4,569,794, which hereby is incorporated by reference. The IMAC method enables isolation of substantially pure protein starting from a crude cellular extract.
As skilled artisans will recognize, owing to the degeneracy of the code, the proteins of the invention can be encoded by a large genus of different nucleic acid sequences. This invention further comprises said genus.
The ribonucleic acid compounds of the invention may be prepared using the polynucleotide synthetic methods discussed supra, or they may be prepared enzymatically using RNA polymerase to transcribe a DNA template.
The most preferred systems for preparing the ribonucleic acids of the present invention employ the RNA polymerase from the bacteriophage T7 or the bacteriophage SP6. These RNA polymerases are highly specific, requiring the insertion of bacteriophage-specific sequences at the 5′ end of a template. See, J. Sambrook, et al., supra, at 18.82-18.84.
This invention also provides nucleic acids that are complementary to the sequences disclosed herein.
The present invention also provides probes and primers, useful for a variety of molecular biology techniques including, for example, hybridization screens of genomic or subgenomic libraries, or detection and quantification of mRNA species as a means to analyze gene expression. A nucleic acid compound is provided comprising any of the sequences disclosed herein, or a complementary sequence thereof, or a fragment thereof, which is at least 15 base pairs in length, and which will hybridize selectively to Streptococcus pneumoniae DNA or mRNA. Preferably, the 15 or more base pair compound is DNA. A probe or primer length of at least 15 base pairs is dictated by theoretical and practical considerations. See e.g. B. Wallace and G. Miyada, “Oligonucleotide Probes for the Screening of Recombinant DNA Libraries,” In Methods in Enzymology, Vol. 152, 432-442, Academic Press (1987).
The probes and primers of this invention can be prepared by methods well known to those skilled in the art (See e.g. Sambrook et al. supra). In a preferred embodiment the probes and primers are synthesized by the polymerase chain reaction (PCR).
The present invention also relates to recombinant DNA cloning vectors and expression vectors comprising the nucleic acids of the present invention. Preferred nucleic acid vectors are those that comprise DNA. The skilled artisan understands that choosing the most appropriate cloning vector or expression vector depends on the availability of restriction sites, the type of host cell into which the vector is to be transfected or transformed, the purpose of transfection or transformation (e.g., stable transformation as an extrachromosomal element, or integration into a host chromosome), the presence or absence of readily assayable or selectable markers (e.g., antibiotic resistance and metabolic markers of one type and another), and the number of gene copies desired in the host cell.
Suitable vectors comprise RNA viruses, DNA viruses, lytic bacteriophages, lysogenic bacteriophages, stable bacteriophages, plasmids, viroids, and the like. The most preferred vectors are plasmids.
Host cells harboring the nucleic acids disclosed herein are also provided by the present invention. A preferred host is E. coli transfected or transformed with a vector comprising a nucleic acid of the present invention.
The invention also provides a host cell capable of expressing a gene described herein, said method comprising transforming or otherwise introducing into a host cell a recombinant DNA vector comprising an isolated DNA sequence that encodes said gene. The preferred host cell is any strain of E. coli that can accommodate high level expression of an exogenously introduced gene. Transformed host cells are cultured under conditions well known to skilled artisans, such that said gene is expressed, thereby producing the encoded protein in the recombinant host cell.
To discover compounds having antibacterial activity, one can look for agents that inhibit cell growth and/or viability by, for example, inhibiting enzymes required for cell wall biosynthesis, and/or by identifying agents that interact with membrane proteins. A method for identifying such compounds comprises contacting a suitable protein or membrane preparation with a test compound and monitoring by any suitable means an interaction and/or inhibition of a protein of this invention.
For example, the instant invention provides a screen for compounds that interact with the proteins of the invention, said screen comprising:
a) preparing a protein, or membranes enriched in a protein;
b) exposing the protein or membranes to a test compound; and
c) detecting an interaction of a protein with said compound by any suitable means.
The screening method of this invention may be adapted to automated procedures such as a PANDEX® (Baxter-Dade Diagnostics) system, allowing for efficient high-volume screening of compounds.
In a typical screen, a protein is prepared as described herein, preferably using recombinant DNA technology. A test compound is introduced into a reaction vessel containing said protein. The reaction/interaction of said protein and said compound is monitored by any suitable means. In a preferred method, a radioactively-labeled or. chemically-labeled compound or protein is used. A specific association between the test compound and protein is monitored by any suitable means.
In such a screening protocol HI1648 is prepared as described herein, preferably using recombinant DNA technology. A test compound is introduced into a reaction vessel containing the HI1648 protein or fragment thereof. Binding of HI1648 by a test compound is determined by any suitable means. For example, in one method radioactively-labeled or chemically-labeled test compound may be used. Binding of the protein by the compound is assessed, for example, by quantifying bound label versus unbound label using any suitable method. Binding of a test compound may also be carried out by a method disclosed in U.S. Pat. No. 5,585,277, which hereby is incorporated by reference. In this method, binding of a test compound to a protein is assessed by monitoring the ratio of folded protein to unfolded protein, for example by monitoring sensitivity of said protein to a protease, or amenability to binding of said protein by a specific antibody against the folded state of the protein.
The foregoing screening methods are useful for identifying a ligand of a HI1648 protein, perhaps as a lead to a pharmaceutical compound for modulating the state of differentiation of an appropriate tissue. A ligand that binds HI1648, or related fragment thereof, is identified, for example, by combining a test ligand with HI1648 under conditions that cause the protein to exist in a ratio of folded to unfolded states. If the test ligand binds the folded state of the protein, the relative amount of folded protein will be higher than in the case of a test ligand that does not bind the protein. The ratio of protein in the folded versus unfolded state is easily determinable by, for example, susceptibility to digestion by a protease, or binding to a specific antibody, or binding to chaperoning protein, or binding to any suitable surface.
The following examples more fully describe the present invention. Those skilled in the art will recognize that the particular reagents, equipment, and procedures described are merely illustrative and are not intended to limit the present invention in any manner.
EXAMPLE 1 Production of a Vector for Expressing S. pneumoniae HI1648 in a Host Cell
An expression vector suitable for expressing S. pneumoniae HI1648 in a variety of procaryotic host cells, such as E. coli, is easily made. The vector contains an origin of replication (Ori), an ampicillin resistance gene (Amp) useful for selecting cells which have incorporated the vector following a tranformation procedure, and further comprises the T7 promoter and T7 terminator sequences in operable linkage to the HI1648 coding region. Plasmid pET11A (obtained from Novogen, Madison, Wis.) is a suitable parent plasmid. pET11A is linearized by restriction with endonucleases NdeI and BamHI. Linearized pET11A is ligated to a DNA fragment bearing NdeI and BamHI sticky ends and comprising the coding region of the S. pneumoniae HI1648 (SEQ ID NO:1). The coding region for HI1648 is easily produced by PCR technology using suitably designed primers to the ends of the coding region specified in SEQ ID NO:1.
The HI1648 encoding nucleic acid used in this construct is slightly modified at the 5′ end (amino terminus of encoded protein) in order to simplify purification of the encoded protein product. For this purpose, an oligonucleotide encoding 8 histidine residues is inserted after the ATG start codon. Placement of the histidine residues at the amino terminus of the encoded protein serves to enable the IMAC one-step protein purification procedure.
EXAMPLE 2 Recombinant Expression and Purification of a Protein Encoded by S. pneumoniae HI1648
An expression vector that carries HI1648 from the S. pneumoniae genome as disclosed herein and which H11648 is operably-linked to an expression promoter is transformed into E. coli BL21 (DE3) (hsdS gal lcIts857 ind1Sam7nin5lacUV5-T7 gene 1) using standard methods (see Example 4). Transformants, selected for resistance to ampicillin, are chosen at random and tested for the presence of the vector by agarose gel electrophoresis using quick plasmid preparations. Colonies which contain the vector are grown in L broth and the protein product encoded by the vector-borne ORF is purified by immobilized metal ion affinity chromatography (IMAC), essentially as described in U.S. Pat. No. 4,569,794.
Briefly, the IMAC column is prepared as follows. A metal-free chelating resin (e.g. Sepharose 6B IDA, Pharmacia) is washed in distilled water to remove preservative substances and infused with a suitable metal ion [e.g. Ni(II), Co(II), or Cu(II)] by adding a 5 mM metal chloride or metal sulfate aqueous solution until about 75% of the interstitial spaces of the resin are saturated with colored metal ion. The column is then ready to receive a crude cellular extract containing the recombinant protein product.
After removing unbound proteins and other materials by washing the column with any suitable buffer, pH 7.5, the bound protein is eluted in any suitable buffer at pH 4.3, or preferably with an imidizole-containing buffer at pH 7.5.
4 579 base pairs nucleic acid single linear DNA (genomic) NO NO CDS 1..579 1 ATG AAA ATC GGA ATA TTG GCC TTG CAA GGG GCC TTT GCA GAA CAT GCA 48 Met Lys Ile Gly Ile Leu Ala Leu Gln Gly Ala Phe Ala Glu His Ala 1 5 10 15 AAA GTG CTA GAT CAA TTA GGT GTC GAG AGT GTA GAA CTC AGA AAT CTA 96 Lys Val Leu Asp Gln Leu Gly Val Glu Ser Val Glu Leu Arg Asn Leu 20 25 30 GAT GAT TTT CAG CAA GAT CAG AGT GAC TTG TCG GGT TTG ATT TTG CCT 144 Asp Asp Phe Gln Gln Asp Gln Ser Asp Leu Ser Gly Leu Ile Leu Pro 35 40 45 GGT GGT GAG TCT ACA ACC ATG GGC AAG CTC TTA CGT GAC CAG AAC ATG 192 Gly Gly Glu Ser Thr Thr Met Gly Lys Leu Leu Arg Asp Gln Asn Met 50 55 60 CTA CTT CCC ATA CGA GAA GCC ATT CTA TCT GGC TTA CCA GTG TTT GGG 240 Leu Leu Pro Ile Arg Glu Ala Ile Leu Ser Gly Leu Pro Val Phe Gly 65 70 75 80 ACC TGT GCG GGC TTA ATT TTG CTG GCT AAG GAA ATC ACT TCT CAG AAA 288 Thr Cys Ala Gly Leu Ile Leu Leu Ala Lys Glu Ile Thr Ser Gln Lys 85 90 95 GAG AGT CAT CTA GGA ACT ATG GAT ATG GTG GTC GAG CGT AAT GCT TAT 336 Glu Ser His Leu Gly Thr Met Asp Met Val Val Glu Arg Asn Ala Tyr 100 105 110 GGG CGC CAA TTA GGA AGT TTC TAC ACG GAA GCA GAA TGT AAG GGA GTT 384 Gly Arg Gln Leu Gly Ser Phe Tyr Thr Glu Ala Glu Cys Lys Gly Val 115 120 125 GGC AAG ATT CCA ATG ACC TTT ATC CGT GGT CCG ATT ATC AGT AGT GTT 432 Gly Lys Ile Pro Met Thr Phe Ile Arg Gly Pro Ile Ile Ser Ser Val 130 135 140 GGT GAG GGT GTA GAA ATT TTA GCA ATA GTG AAC AAT CAA ATT GTT GCA 480 Gly Glu Gly Val Glu Ile Leu Ala Ile Val Asn Asn Gln Ile Val Ala 145 150 155 160 GCC CAA GAA AAA AAT ATG TTG GTA AGT TCT TTT CAT CCA GAA TTG ACT 528 Ala Gln Glu Lys Asn Met Leu Val Ser Ser Phe His Pro Glu Leu Thr 165 170 175 GAT GAT GTG CGC TTG CAC CAG TAC TTT ATC AAT ATG TGT AAA GAA AAA 576 Asp Asp Val Arg Leu His Gln Tyr Phe Ile Asn Met Cys Lys Glu Lys 180 185 190 AGT 579 Ser 193 amino acids amino acid linear protein 2 Met Lys Ile Gly Ile Leu Ala Leu Gln Gly Ala Phe Ala Glu His Ala 1 5 10 15 Lys Val Leu Asp Gln Leu Gly Val Glu Ser Val Glu Leu Arg Asn Leu 20 25 30 Asp Asp Phe Gln Gln Asp Gln Ser Asp Leu Ser Gly Leu Ile Leu Pro 35 40 45 Gly Gly Glu Ser Thr Thr Met Gly Lys Leu Leu Arg Asp Gln Asn Met 50 55 60 Leu Leu Pro Ile Arg Glu Ala Ile Leu Ser Gly Leu Pro Val Phe Gly 65 70 75 80 Thr Cys Ala Gly Leu Ile Leu Leu Ala Lys Glu Ile Thr Ser Gln Lys 85 90 95 Glu Ser His Leu Gly Thr Met Asp Met Val Val Glu Arg Asn Ala Tyr 100 105 110 Gly Arg Gln Leu Gly Ser Phe Tyr Thr Glu Ala Glu Cys Lys Gly Val 115 120 125 Gly Lys Ile Pro Met Thr Phe Ile Arg Gly Pro Ile Ile Ser Ser Val 130 135 140 Gly Glu Gly Val Glu Ile Leu Ala Ile Val Asn Asn Gln Ile Val Ala 145 150 155 160 Ala Gln Glu Lys Asn Met Leu Val Ser Ser Phe His Pro Glu Leu Thr 165 170 175 Asp Asp Val Arg Leu His Gln Tyr Phe Ile Asn Met Cys Lys Glu Lys 180 185 190 Ser 579 base pairs nucleic acid single linear mRNA NO NO 3 AUGAAAAUCG GAAUAUUGGC CUUGCAAGGG GCCUUUGCAG AACAUGCAAA AGUGCUAGAU 60 CAAUUAGGUG UCGAGAGUGU AGAACUCAGA AAUCUAGAUG AUUUUCAGCA AGAUCAGAGU 120 GACUUGUCGG GUUUGAUUUU GCCUGGUGGU GAGUCUACAA CCAUGGGCAA GCUCUUACGU 180 GACCAGAACA UGCUACUUCC CAUACGAGAA GCCAUUCUAU CUGGCUUACC AGUGUUUGGG 240 ACCUGUGCGG GCUUAAUUUU GCUGGCUAAG GAAAUCACUU CUCAGAAAGA GAGUCAUCUA 300 GGAACUAUGG AUAUGGUGGU CGAGCGUAAU GCUUAUGGGC GCCAAUUAGG AAGUUUCUAC 360 ACGGAAGCAG AAUGUAAGGG AGUUGGCAAG AUUCCAAUGA CCUUUAUCCG UGGUCCGAUU 420 AUCAGUAGUG UUGGUGAGGG UGUAGAAAUU UUAGCAAUAG UGAACAAUCA AAUUGUUGCA 480 GCCCAAGAAA AAAAUAUGUU GGUAAGUUCU UUUCAUCCAG AAUUGACUGA UGAUGUGCGC 540 UUGCACCAGU ACUUUAUCAA UAUGUGUAAA GAAAAAAGU 579 1863 base pairs nucleic acid single linear DNA (genomic) NO NO 4 GAGCTCGGTA CCCGGGGATC ATACTCAAGA GGAGGTAATC CAATGAACAC TAGTCTTAAA 60 CTCAGCAAAC AACTCAGTTT TGGAGAGGAG ATTGCTAATA GCGTGACCCA TGCTGTGGGT 120 GCAGTCATCA TGCTTATCTT GCTGCCTATT TCATCCATCT ATAGTTATGA AGCACACGGA 180 TTTTTATCCT CTATCGGCGT TTCCATTTTC GTCATCAGTC TCTTTCTCAT GTTCCTATCA 240 TCCACCATTT ATCACTCTAT GGCCTATGGT TCGACCCACA AATATGTTTT GCGAATCATT 300 GACCATTCTA TGATTTACGT TGCCATTGCC GGCTCATACA CGCCCGTTGT CTTGACCTTG 360 ATGAATAACT GGTTTGGCTA TCTGATTATT GTCATCCAAT GGGGAACGAC CATCTTTGGT 420 ATTCTCTATA AAATCTTTGC TAAAAAGGTC AATGAGAAAT TTAGCCTTGC TCTTTACCTG 480 ATTATGGGCT GGTTGGTTCT GGCTATCATT CCTGCCATTA TCAGTCAAAC GACACCCGTT 540 TTCTGGAGTC TCATGGTAAC TGGCGGACTC TGTTATACAG TTGGAGCTGG ATTTTATGCC 600 AAGAAAAAAC CTTATTTCCA CATGATTTGG CATCTCTTTA TCCTAGCTGC GTCCGCACTC 660 CAATACATCG CTATTGTTTA TTACATGTAA AAAAGTTGAG AAATTCAATC TCAACTTTTT 720 TCTTTACACA TATTGATAAA GTACTGGTGC AAGCGCACAT CATCAGTCAA TTCTGGATGA 780 AAAGAACTTA CCAACATATT TTTTTCTTGG GCTGCAACAA TTTGATTGTT CACTATTGCT 840 AAAATTTCTA CACCCTCACC AACACTACTG ATAATCGGAC CACGGATAAA GGTCATTGGA 900 ATCTTGCCAA CTCCCTTACA TTCTGCTTCC GTGTAGAAAC TTCCTAATTG GCGCCCATAA 960 GCATTACGCT CGACCACCAT ATCCATAGTT CCTAGATGAC TCTCTTTCTG AGAAGTGATT 1020 TCCTTAGCCA GCAAAATTAA GCCCGCACAG GTCCCAAACA CTGGTAAGCC AGATAGAATG 1080 GCTTCTCGTA TGGGAAGTAG CATGTTCTGG TCACGTAAGA GCTTGCCCAT GGTTGTAGAC 1140 TCACCACCAG GCAAAATAAA CCCGACAAGT CACTCTGATC TTGCTGAAAA CATCTAGATT 1200 TCTGAGTTCT ACACTCTCGA CACCTAATTG ATCTAGCACT TTTGCATGTT CTGCAAAGGC 1260 CCCTTGCAAG GCCAATATTC CGATTTTCAT CTATTTTCCT CGTTCAGCCA TGAGAATTTG 1320 GATTCATTTT CATTAATACC AACCATGGCT TCTCCTAAAT CTTCAGAGAT TTGAGCTAGG 1380 ATTTGAGGAT TACGGAAGTT AGTCACAGCC TTAACAATGG CACTCGCTCG TTTAACAGGA 1440 TCTCCTGACT TGAAAATACC TGAACCGACA AAGACCCCCT CTGCCCCTAA TTGCATCATT 1500 AACGCAGCAT CTGCTGGCGT TGCAACACCT CCAGCAGCGA AATTTACAAC TGGCAATTTT 1560 CCATGTTCAT GAACATATTG GACCAATTCT ACAGGGACTT GCAAATCCTT GGCAGCAACA 1620 TAAAGCTCGT CCTCACGTAA GTTTTGAATG CGGCGAATTT CCTGATTCAT CATACGCATA 1680 TGACGAACAG CTTGGACTAT ATCCCCTGTC CCTGGTTCTC CTTTAGTACG AATCATGGAA 1740 GCACCTTCAG CGATACGACG CAAGGCTTCA CCCAAATCCT TAGCACCACA GACAAAAGGA 1800 ACTTGGAATT CTTTCTTGTC CACATGGAAA CGGTCATCAG CTGGAGATAG AACTTCACTC 1860 TCG 1863

Claims (2)

We claim:
1. An isolated protein having the amino acid sequence shown in SEQ ID NO:2.
2. A method for identifying compounds that bind a protein identified herein as SEQ ID NO:2, comprising the steps of:
a) admixing a substantially purified preparation of a protein comprising SEQ ID:2 with a test compound; and
b) monitoring by any suitable means a binding interaction between said protein and said compound.
US09/198,284 1996-12-13 1998-11-23 Streptococcus pneumoniae gene sequence HI1648 Abandoned USH2019H1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US09/198,284 USH2019H1 (en) 1996-12-13 1998-11-23 Streptococcus pneumoniae gene sequence HI1648

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US3628196P 1996-12-13 1996-12-13
US08/987,122 US5910580A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence HI1648
US09/198,284 USH2019H1 (en) 1996-12-13 1998-11-23 Streptococcus pneumoniae gene sequence HI1648

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/987,122 Division US5910580A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence HI1648

Publications (1)

Publication Number Publication Date
USH2019H1 true USH2019H1 (en) 2002-04-02

Family

ID=21887710

Family Applications (15)

Application Number Title Priority Date Filing Date
US08/986,963 Expired - Fee Related US5958730A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence FtsY
US08/987,119 Abandoned USH2070H1 (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence for DNA ligase
US08/987,151 Expired - Fee Related US6162617A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence dnaG
US08/987,146 Expired - Fee Related US6350866B1 (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence FtsZ
US08/986,769 Expired - Fee Related US6074847A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence HI1146
US08/987,121 Expired - Fee Related US6268175B1 (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence GCP
US08/987,123 Expired - Fee Related US6136557A (en) 1996-12-13 1997-12-08 Strepococcus pneumoniae gene sequence FtsH
US08/986,967 Abandoned USH2023H1 (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae sequence GrpE
US08/987,122 Expired - Fee Related US5910580A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence HI1648
US08/987,152 Expired - Fee Related US5981281A (en) 1996-12-13 1997-12-08 Method for knockout mutagenesis in Streptococcus pneumoniae
US08/987,144 Expired - Fee Related US6060282A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence Dpj
US08/986,765 Expired - Fee Related US6071724A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniaegene sequence era
US08/986,768 Expired - Fee Related US6271000B1 (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence mraY
US08/987,147 Abandoned USH2071H1 (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene HI0454
US09/198,284 Abandoned USH2019H1 (en) 1996-12-13 1998-11-23 Streptococcus pneumoniae gene sequence HI1648

Family Applications Before (14)

Application Number Title Priority Date Filing Date
US08/986,963 Expired - Fee Related US5958730A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence FtsY
US08/987,119 Abandoned USH2070H1 (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence for DNA ligase
US08/987,151 Expired - Fee Related US6162617A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence dnaG
US08/987,146 Expired - Fee Related US6350866B1 (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence FtsZ
US08/986,769 Expired - Fee Related US6074847A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence HI1146
US08/987,121 Expired - Fee Related US6268175B1 (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence GCP
US08/987,123 Expired - Fee Related US6136557A (en) 1996-12-13 1997-12-08 Strepococcus pneumoniae gene sequence FtsH
US08/986,967 Abandoned USH2023H1 (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae sequence GrpE
US08/987,122 Expired - Fee Related US5910580A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence HI1648
US08/987,152 Expired - Fee Related US5981281A (en) 1996-12-13 1997-12-08 Method for knockout mutagenesis in Streptococcus pneumoniae
US08/987,144 Expired - Fee Related US6060282A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence Dpj
US08/986,765 Expired - Fee Related US6071724A (en) 1996-12-13 1997-12-08 Streptococcus pneumoniaegene sequence era
US08/986,768 Expired - Fee Related US6271000B1 (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene sequence mraY
US08/987,147 Abandoned USH2071H1 (en) 1996-12-13 1997-12-08 Streptococcus pneumoniae gene HI0454

Country Status (5)

Country Link
US (15) US5958730A (en)
EP (1) EP0950108A1 (en)
AU (1) AU5793798A (en)
CA (1) CA2274311A1 (en)
WO (1) WO1998026072A1 (en)

Families Citing this family (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2000506385A (en) * 1996-02-20 2000-05-30 スミスクライン・ビーチャム・コーポレイション New ERA
US6165989A (en) * 1996-05-14 2000-12-26 Smithkline Beecham Corporation Era of Streptococcus pneumoniae
US5681694A (en) * 1996-06-18 1997-10-28 Eli Lilly And Company Murd protein method and kit for identification of inhibitors
US5910414A (en) 1996-10-15 1999-06-08 Smithkline Beecham Corporation Topoisomerase I of streptococcus pneumoniae
US5789202A (en) * 1996-10-17 1998-08-04 Eli Lilly And Company DNA encoding a novel penicillin binding protein from streptococcus pneumoniae
US6096518A (en) 1996-10-24 2000-08-01 Smithkline Beecham Corporation DNA encoding SPO/REL polypeptides of streptococcus
CA2269663A1 (en) 1996-10-31 1998-05-07 Human Genome Sciences, Inc. Streptococcus pneumoniae antigens and vaccines
US5821335A (en) * 1996-11-19 1998-10-13 Eli Lilly And Company Biosynthetic gene murg from streptococcus pneumoniae
US6287803B1 (en) * 1996-11-27 2001-09-11 Smithkline Beecham Corporation Polynucleotides encoding a novel era polypeptide
US5948645A (en) * 1996-12-04 1999-09-07 Eli Lilly And Company Biosynthetic gene muri from Streptococcus pneumoniae
US7098023B1 (en) 1997-07-02 2006-08-29 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US6800744B1 (en) 1997-07-02 2004-10-05 Genome Therapeutics Corporation Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US5840538A (en) * 1997-08-06 1998-11-24 Smithkline Beecham Corporation Lgt
EP0896061A3 (en) * 1997-08-08 2000-07-26 Smithkline Beecham Corporation RpoA gene from Staphylococcus aureus
EP0896060A3 (en) * 1997-08-08 2000-07-19 Smithkline Beecham Corporation RpoA gene from Staphylococcus aureus
US5929045A (en) * 1997-08-12 1999-07-27 Smithkline Beecham Corporation Recombinant expression of polynucleotides encoding the UDP-N-acetylmuramoylalanine:D-glutamate ligase (MurD) of Streptococcus pneumoniae
US6197300B1 (en) * 1997-08-12 2001-03-06 Smithkline Beecham Corporation ftsZ
US5888770A (en) * 1997-08-26 1999-03-30 Smithkline Beecham Corporation Spoiiie
US6072032A (en) * 1997-08-29 2000-06-06 Smithkline Beecham Corporation FtsY polypeptides from Streptococcus pneumoniae
EP0915161A3 (en) * 1997-10-21 1999-08-25 Smithkline Beecham Corporation DnaG DNA primase of Streptococcus pneumoniae
GB9726586D0 (en) * 1997-12-16 1998-02-11 Microscience Ltd Protein and compositions containing it
AU753971B2 (en) * 1997-12-31 2002-10-31 Millennium Pharmaceuticals, Inc. Essential bacterial genes and their use
GB9808423D0 (en) * 1998-04-22 1998-06-17 Glaxo Group Ltd Bacterial polypeptide family
US6274719B1 (en) * 1998-04-24 2001-08-14 Smithkline Beecham Corporation Gcp
JP2002516078A (en) * 1998-05-28 2002-06-04 スミスクライン・ビーチャム・コーポレイション acpS
US6190881B1 (en) * 1998-06-05 2001-02-20 Smithkline Beecham Corporation Ribonucleotide diphosphate reductase, nrdF, of streptococcus pneumoniae
WO2000001801A1 (en) * 1998-07-02 2000-01-13 Smithkline Beecham Corporation Ftsz multimeric proteins and their uses
US6936252B2 (en) 1998-07-27 2005-08-30 Microbial Technics Limited Streptococcus pneumoniae proteins and nucleic acid molecules
WO2000006738A2 (en) * 1998-07-27 2000-02-10 Microbial Technics Limited NUCLEIC ACIDS AND PROTEINS FROM $i(STREPTOCOCCUS PNEUMONIAE)
EP1790730A3 (en) * 1998-07-27 2007-09-12 Sanofi Pasteur Limited Streptococcus pneumoniae proteins and nucleic acid molecules
US20030134407A1 (en) 1998-07-27 2003-07-17 Le Page Richard William Falla Nucleic acids and proteins from Streptococcus pneumoniae
US6515119B1 (en) * 1998-09-30 2003-02-04 Millennium Pharmaceuticals, Inc. Use of S-ydcB and B-ydcB, essential bacterial genes
US6537774B1 (en) 1998-10-14 2003-03-25 Smithkline Beecham Corporation UPS (undecaprenyl diphosphate synthase
US6110685A (en) * 1998-10-28 2000-08-29 Smithkline Beecham Corporation infB
WO2000025818A1 (en) * 1998-11-04 2000-05-11 Smithkline Beecham Corporation asuE/trmU
US6346395B1 (en) * 1999-01-27 2002-02-12 Smithkline Beecham Corp. Nucleic acids encoding Streptococcus pneumoniae FabG
US6110704A (en) * 1999-01-28 2000-08-29 Smithkline Beecham Corporation 3-ketoacyl-ACP-reductase (FabG) of Staphylococcus aureus
US6346396B1 (en) * 1999-01-29 2002-02-12 Jianzhong Huang MurA
JP2002542821A (en) * 1999-04-30 2002-12-17 ハイブリジェニックス・ソシエテ・アノニム Prokaryotic DNA collection for two-hybrid systems, Helicobacter pylori protein-protein interaction and uses thereof
US6548273B1 (en) * 1999-07-22 2003-04-15 Smithkline Beecham Corporation lacR from Streptococcus pneumoniae
US6168797B1 (en) * 1999-08-18 2001-01-02 Smithkline Beecham Corporation FabF
US6951729B1 (en) 1999-10-27 2005-10-04 Affinium Pharmaceuticals, Inc. High throughput screening method for biological agents affecting fatty acid biosynthesis
US6613553B1 (en) * 2000-02-04 2003-09-02 St. Jude Children's Research Hospital Enoyl reductases and methods of use thereof
GB2364054B (en) * 2000-03-24 2002-05-29 Smithkline Beecham Corp Method of amplifying quinolone-resistance-determining-regions and identifying polymorphic variants thereof
CA2424987A1 (en) * 2000-10-06 2002-04-18 Smithkline Beecham Corporation Methods of agonizing and antagonizing fabk
US7048926B2 (en) 2000-10-06 2006-05-23 Affinium Pharmaceuticals, Inc. Methods of agonizing and antagonizing FabK
US7033795B2 (en) 2000-10-06 2006-04-25 Affinium Pharmaceuticals, Inc. FabK variant
US7056697B2 (en) 2000-10-06 2006-06-06 Affinium Pharmaceuticals, Inc. FabK variant
US6821746B2 (en) 2000-10-06 2004-11-23 Affinium Pharmaceuticals, Inc. Methods of screening for FabK antagonists and agonists
JP2004515251A (en) * 2000-12-19 2004-05-27 ファゲテック,インコーポレイティド Compositions and methods related to the Staphylococcus aureus essential gene and its encoded protein STAAU_R9
EP1476568A2 (en) * 2002-02-20 2004-11-17 NCC Technology Ventures Pte Limited Materials and methods relating to cancer diagnosis
EP2311989A1 (en) * 2003-04-15 2011-04-20 Intercell AG S. pneumoniae antigens
GB0318688D0 (en) * 2003-08-08 2003-09-10 Chiron Srl Streptococcus pneumoniae knockout mutants
US20070149496A1 (en) * 2003-10-31 2007-06-28 Jack Tuszynski Water-soluble compound
WO2013043643A1 (en) * 2011-09-21 2013-03-28 St. Jude Children's Research Hospital Live, attenuated streptococcus pneumoniae strain and vaccine for protection against pneumococcal disease
EP3684922A1 (en) * 2017-07-31 2020-07-29 Enzibeta Biotech Pvt. Ltd. Modified gene sequences encoding choline oxidase and a method for preparing betaine usng the same

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4762786A (en) * 1984-09-27 1988-08-09 Eli Lilly And Company Vectors and conditions which allow genetic transformation of cephalosporium
US5747281A (en) * 1984-09-28 1998-05-05 Cornell Research Foundation, Inc. System useful for the production of proteins from recombinant DNA in single celled organisms
US4569794A (en) * 1984-12-05 1986-02-11 Eli Lilly And Company Process for purifying proteins and compounds useful in such process
RU1780542C (en) * 1985-04-22 1992-12-07 Эли Лилли Энд Компани Method for forming recombinant plasmid dna pp20 coding isopenicillin-n-synthetase, method for producing strain cephalosporium acremonium possessing activity of isopenicillin-n-synthetase
US4892819A (en) * 1985-11-25 1990-01-09 Eli Lilly And Company Recombinant DNA expression vectors and DNA compounds that encode isopenicillin N synthetase from penicillium chrysogenum
US4960707A (en) * 1987-08-17 1990-10-02 Associated Universities, Inc. Recombinant plasmids for encoding restriction enzymes DpnI and DpnII of streptococcus pneumontae
US5070020A (en) * 1988-05-09 1991-12-03 Eli Lilly And Company Recombinant dna expression vectors and dna compounds that encode deacetoxycephalosporin c synthetase
GB9100825D0 (en) * 1991-01-15 1991-02-27 Univ Guelph Pasteurella haemolytica glycoprotease gene and the purified enzyme
IL109410A0 (en) * 1993-04-30 1994-07-31 Ell Lilly & Company Fema gene of staphylococcus epidermidis, fema protein, and vectors and microorganisms comprising the fema gene
US5591839A (en) * 1994-08-18 1997-01-07 Glyko, Inc. Polynucleotides encoding α2-3 neuraminidase
JP3571378B2 (en) * 1994-09-30 2004-09-29 扶桑薬品工業株式会社 Infectious disease diagnostic probe
US5705352A (en) * 1995-02-27 1998-01-06 Eli Lilly And Company Multiple drug resistance gene of Aspergillus fumigatus
US5773214A (en) * 1995-02-27 1998-06-30 Eli Lilly And Company Multiple drug resistance gene of aspergillus flavus
US5691161A (en) * 1996-08-01 1997-11-25 Eli Lilly And Company Peptidoglycan biosynthetic mura protein from Streptococcus pneumoniae
US6197300B1 (en) * 1997-08-12 2001-03-06 Smithkline Beecham Corporation ftsZ

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Fleischmann, et al. "Whole-Genome Random Sequencing and Assembly of Haemophilus influenzae Rd." Science 269:496-512 (Jul. 28, 1995).

Also Published As

Publication number Publication date
US6271000B1 (en) 2001-08-07
US6162617A (en) 2000-12-19
US5910580A (en) 1999-06-08
US6268175B1 (en) 2001-07-31
EP0950108A1 (en) 1999-10-20
USH2071H1 (en) 2003-07-01
US5981281A (en) 1999-11-09
US6071724A (en) 2000-06-06
WO1998026072A1 (en) 1998-06-18
USH2070H1 (en) 2003-07-01
US5958730A (en) 1999-09-28
US6136557A (en) 2000-10-24
US6074847A (en) 2000-06-13
AU5793798A (en) 1998-07-03
US6060282A (en) 2000-05-09
CA2274311A1 (en) 1998-06-18
USH2023H1 (en) 2002-05-07
US6350866B1 (en) 2002-02-26

Similar Documents

Publication Publication Date Title
USH2019H1 (en) Streptococcus pneumoniae gene sequence HI1648
US5681694A (en) Murd protein method and kit for identification of inhibitors
EP0931834A2 (en) Echinocandin binding domain of 1,3-Beta-glucan synthase
US5770415A (en) Peptidoglycan biosynthetic gene mur a from Strepococcus pneumoniae
US5712108A (en) Peptidoglycan biosynthetic mure protein from streptocuccus pneumoniae
US6143868A (en) Monofunctional glycosyltransferase of Staphylococcus aureus
US5789202A (en) DNA encoding a novel penicillin binding protein from streptococcus pneumoniae
US5821335A (en) Biosynthetic gene murg from streptococcus pneumoniae
US5821100A (en) Glycosyltransferase gene gtfb from Amycolatopsis orientalis
US5948645A (en) Biosynthetic gene muri from Streptococcus pneumoniae
US5776695A (en) Biosynthetic gene ddl Streptococcus pneumoniae
WO2000034316A1 (en) Era complexes and uses thereof

Legal Events

Date Code Title Description
STCF Information on status: patent grant

Free format text: PATENTED CASE