Compounds

Information

  • Patent Grant
  • 6348328
  • Patent Number
    6,348,328
  • Date Filed
    Wednesday, May 14, 1997
    27 years ago
  • Date Issued
    Tuesday, February 19, 2002
    22 years ago
Abstract
This invention relates to newly identified polynucleotides, polypeptides encoded by such polynucleotides, the uses of such polynucleotides and polypeptides, as well as the production of such polynucleotides and polypeptides and recombinant host cells transformed with the polynucleotides. This invention also relates to inhibiting the biosynthesis or action of such polynucleotides or polypeptides and to the use of such inhibitors in therapy.
Description




FIELD OF THE INVENTION




This invention relates to newly identified polynucleotides, polypeptides encoded by such polynucleotides, the uses of such polynucleotides and polypeptides, as well as the production of such polynucleotides and polypeptides and recombinant host cells transformed with the polynucleotides. This invention also relates to inhibiting the biosynthesis or action of such polynucleotides or polypeptides and to the use of such inhibitors in therapy. Preferred embodiments of the invention include Streptococcal polypeptides and polynucleotides, particularly those of


Streptococcus pneumoniae.






BACKGROUND OF THE INVENTION




The Streptococci make up a medically important genera of microbes known to cause several types of disease in humans, including otitis media, pneumonia and meningitis. Since its isolation more than 100 years ago,


Streptococcus pneumoniae


has been one of the more intensively studied microbes. For example, much of our early understanding that DNA is, in fact, the genetic material was predicated on the work of Griffith and of Avery, Macleod and McCarty using this microbe. Despite the vast amount of research with


S. pneumoniae


, many questions concerning the virulence of this microbe remain.




While certain Streptococcal factors associated with pathogenicity have been identified, e.g., capsule polysaccharides, peptidoglycans, pneumolysins, PspA Complement factor H binding component, autolysin, neuraminidase, peptide permeases, hydrogen peroxide, IgA1 protease, the list is certainly not complete. Further very little is known concerning the temporal expression of such genes during infection and disease progression in a mammalian host. Discovering the sets of genes the bacterium is likely to be expressing at the different stages of infection, particularly when an infection is established, provides critical information for the screening and characterization of novel antibacterials which can interrupt pathogenesis. In addition to providing a fuller understanding of known proteins, such an approach will identify previously unrecognised targets.




BRIEF DESCRIPTION OF THE INVENTION




This invention provides novel proteins, particularly those from


Streptococcus pneumoniae


, strain 0100993, characterised in that it comprises the amino acid sequences given herein or fragments, analogues or derivatives thereof.




In accordance with another aspect of the present invention, there are provided polynucleotides (DNA or RNA) which encode such polypeptides.




In particular the invention provides polynucleotides having the DNA sequences given herein.




The invention also relates to novel oligonucleotides derived from the sequences given herein which can act as PCR primers in the process herein described to determine whether or not the


Streptococcus pneumoniae


gene identified herein in whole or in part is expressed in infected tissue. It is recognised that such sequences will also have utility in diagnosis of the stage of infection and type of infection the pathogen has attained. The proteins so identified are also useful as targets in screens designed to identify antimicrobial compounds.




It is an object of the invention to provide polypeptides that have been identified as novel polypeptides by homology between the amino acid sequence set out in the Sequence Listing and a known amino acid sequence or sequences of other proteins such as the proteins identified under the heading Identity in Table 1.




It is a further object of the invention to provide polynucleotides that encode polypeptides set forth in the Sequence Listing, particularly polynucleotides that encode the polypeptide set forth in the Sequence Listing.




In a particularly preferred embodiment of the invention the polynucleotide comprises a region encoding polypeptides comprising the sequence set out in the Sequence Listing, or a variant thereof.




In another particularly preferred embodiment of the invention there is a novel protein from comprising an amino acid sequence of the Sequence Listing, or a variant thereof.




In accordance with another aspect of the invention, there is provided the use of a polynucleotide of the invention for therapeutic or prophylactic purposes, in particular genetic immunization. Among the particularly preferred embodiments of the invention are naturally occurring allelic variants the polynucleotides set forth in the Sequence Listing and polypeptides encoded thereby.




In accordance with yet another aspect of the invention, there are provided inhibitors to such polypeptides, useful as antibacterial agents, including, for example, antibodies.




In accordance with certain preferred embodiments of the invention, there are provided products, compositions and methods for assessing expression of the sequences the Sequence Listing, treating disease, for example, otitis media, conjunctivitis, pneumonia, bacteremia, meningitis, sinusitis, pleural empyema and endocarditis, and most particularly meningitis, such as for example infection of cerebrospinal fluid, assaying genetic variation, and administering a polypeptide or polynucleotide of the invention to an organism to raise an immunological response against a bacteria, especially a


Streptococcus pneumoniae


bacteria




In accordance with certain preferred embodiments of this and other aspects of the invention there are provided polynucleotides that hybridize to polynucleotide sequences of the invention, particularly under stringent conditions.




In certain preferred embodiments of the invention there are provided antibodies against polypeptides of the invention.




In other embodiments of the invention there are provided methods for identifying compounds which bind to or otherwise interact with and inhibit or activate an activity of a polypeptide or polynucleotide of the invention comprising: contacting a polypeptide or polynucleotide of the invention with a compound to be screened under conditions to permit binding to or other interaction between the compound and the polypeptide or polynucleotide to assess the binding to or other interaction with the compound, such binding or interaction being associated with a second component capable of providing a detetable signal in response to the binding or interaction of the polypeptide or polynucleotide with the compound; and determining whether the compound binds to or otherwise interacts with and activates or inhibits an activity of the polypetide or polynucleotide by detecting the presence or absence of a signal generated from the binding or interaction of the compound with the polypeptide or polynucleotide.




In accordance with yet another aspect of the invention, there are provided agonists and antagonists of the polynucleotide and/or polypeptides of the invention, preferably bacteriostatic or bacteriocidal agonists and antagonists.




In a further aspect of the invention there are provided compositions comprising a polynucleotide or a polypeptide of the invention for administration to a cell or to a multicellular organism.




Various changes and modifications within the spirit and scope of the disclosed invention will become readily apparent to those skilled in the art from reading the following descriptions and from ring the other parts of the present disclosure.




DETAILED DESCRIPTION OF THE INVENTION




Each of the DNA sequences provided herein may be used in the discovery and development of antibacterial compounds. Because each of the sequences contains an open reading frame (ORF) with appropriate initiation and termination codons, the encoded protein upon expression can be used as a target for the screening of antimicrobial drugs. Additionally, the DNA sequences encoding preferably the amino terminal regions of the encoded protein or the Shine-Delgarno region can be used to construct antisense sequences to control the expression of the coding sequence of interest. Furthermore, many of the sequences disclosed herein also provide regions upstream and downstream from the encoding sequence. These sequences are useful as a source of regulatory elements for the control of bacterial gene expression. Such sequences are conveniently isolated by restriction enzyme action or synthesized chemically and introduced, for example, into promoter identification strains. These strains contain a reporter structural gene sequence located downstream from a restriction site such that if an active promoter is inserted, the reporter gene will be expressed.




Although each of the sequences may be employed as described above, this invention also provides several means for identifying particularly useful target genes. The first of these approaches entails searching appropriate databases for sequence matches in related organisms. Thus, if a homologue exists, the Streptococcal-like form of this gene would likely play an analogous role. For example, a Streptococcal protein identified as homologous to a cell surface protein in another organism would be useful as a vaccine candidate. To the extent such homologies have been identified for the sequences disclosed herein they are reported along with the encoding sequence.




Recently techniques have become available to evaluate temporal gene expression in bacteria, particularly as it applies to viability under laboratory and infection conditions. A number of methods can be used to identify genes which are essential to survival per se, or essential to the establishment/maintenance of an infection. Identification of an ORF unknown by one of these methods yields additional information about its function and permits the selection of such an ORF for further development as a screening target. Briefly, these approaches include:




1) Signature Tagged Mutagenesis (STM)




This technique is described by Hensel et al.,


Science


269: 400-403(1995), the contents of which is incorporated by reference for background purposes. Signature tagged mutagenesis identifies genes necessary for the establishment/maintenance of infection in a given infection model.




The basis of the technique is the random mutagenesis of target organism by various means (e.g., transposons) such that unique DNA sequence tags are inserted in close proximity to the site of mutation. The tags from a mixed population of bacterial mutants and bacteria recovered from an infected hosts are detected by amplification, radiolabeling and hybridisation analysis. Mutants attenuated in virulence are revealed by absence of the tag from the pool of bacteria recovered from infected hosts.




In


Streptococcus pneumoniae


, because the transposon system is less well developed, a more efficient way of creating the tagged mutants is to use the insertion-duplication mutagenesis technique as described by Morrison et al.,


J. Bacteriol,


159:870 (1984) the contents of which is incorporated by reference for background purposes.




2) In Vivo Expression Technology (IVET)




This technique is described by Camilli et Al., i. Proc. Nat'l. Acad. Sci. USA. 91:2634-2638 (1994), the contents of which is incorporated by reference for background purposes. IVET identifies genes unregulated during infection when compared to laboratory cultivation, implying an important role in infection. ORF identified by this technique are implied to have a significant role in infection establishment/maintenance.




In this technique random chromosomal fragments of target organism are cloned upstream of a promoter-less recombinase gene in a plasmid vector. This construct is introduced into the target organism which carries an antibiotic resistance gene flanked by resolvase sites. Growth in the presence of the antibiotic removes from the population those fragments cloned into the plasmid vector capable of supporting transcription of the recombinase gene and therefore have caused loss of antibiotic resistance. The resistant pool is introduced into a host and at various times after infection bacteria may be recovered and assessed for the presence of antibiotic resistance. The chromosomal fragment carried by each antibiotic sensitive bacterium should carry a promoter or portion of a gene normally upregulated during infection. Sequencing upstream of the recombinase gene allows identification of the up regulated gene.




3) Differential Display




This technique is described by Chuang et al.,


J. Bacteriol.


175:2026-2036 (1993), the contents of which is incorporated by reference for background purposes. This method identifies those genes which are expressed in an organism by identifying mRNA present using randomly-primed RT-PCR. By comparing pre-infection and post infection profiles, genes up and down regulated during infection can be identified and the RT-PCR product sequenced and matched to ORF ‘unknowns’.




4) Generation of Conditional Lethal Mutants by Transposon Mutagenesis




This technique, described by de Lorenzo, V. et al.,


Gene


123:17-24 (1993); Neuwald, A. F. et al.,


Gene


125: 69-73(1993); and Takiff, H. E. et al.,


J. Bacteriol.


174:1544-1553(1992), the contents of which is incorporated by reference for background purposes, identifies genes whose expression are essential for cell viability.




In this technique transposons carrying controllable promoters, which provide transcription outward from the transposon in one or both directions, are generated. Random insertion of these transposons into target organisms and subsequent isolation of insertion mutants in the presence of inducer of promoter activity ensures that insertions which separate promoter from coding region of a gene whose expression is essential for cell viability will be recovered. Subsequent replica plating in the absence of inducer identifies such insertions, since they fail to survive. Sequencing of the flanking regions of the transposon allows identification of site of insertion and identification of the gene disrupted. Close monitoring of the changes in cellular processes/morphology during growth in the absence of inducer yields information on likely function of the gene. Such monitoring could include flow cytometry (cell division, lysis, redox potential, DNA replication), incorporation of radiochemically labeled precursors into DNA, RNA, protein, lipid, peptidoglycan, monitoring reporter enzyme gene fusions which respond to known cellular stresses.




5) Generation of Conditional Lethal Mutants by Chemical Mutagenesis




This technique is described by Beckwith,


J., Methods in Enzymology


204: 3-18(1991), the contents of which are incorporated herein by reference for background purposes. In this technique random chemical mutagenesis of target organism, growth at temperature other than physiological temperature (permissive temperature) and subsequent replica plating and growth at different temperature (e.g. 42° C. to identify ts, 25° C. to identify cs) are used to identify those isolates which now fail to grow (conditional mutants). As above close monitoring of the changes upon growth at the non-permissive temperature yields information on the function of the mutated gene. Complementation of conditional lethal mutation by library from target organism and sequencing of complementing gene allows matching with unknown ORF.




6) RT-PCR






Streptococcus pneumoniae


messenger RNA is isolated from bacterial infected tissue e.g. 48 hour murine lung infections, and the amount of each mRNA species assessed by reverse transcription of the RNA sample primed with random hexanucleotides followed by PCR with gene specific primer pairs. The determination of the presence and amount of a particular mRNA species by quantification of the resultant PCR product provides information on the bacterial genes which are transcribed in the infected tissue. Analysis of gene transcription can be carried out at different times of infection to gain a detailed knowledge of gene regulation in bacterial pathogenesis allowing for a clearer understanding of which gene products represent targets for screens for novel antibacterials. Because of the gene specific nature of the PCR primers employed it should be understood that the bacterial mRNA preparation need not be free of mammalian RNA. This allows the investigator to carry out a simple and quick RNA preparation from infected tissue to obtain bacterial mRNA species which are very short lived in the bacterium (in the order of 2 minute halflives). Optimally the bacterial mRNA is prepared from infected murine lung tissue by mechanical disruption in the presence of TRIzole (GIBCO-BRL) for very short periods of time, subsequent processing according to the manufacturers of TRIzole reagent and DNAase treatment to remove contaminating DNA. Preferably the process is optimised by finding those conditions which give a maximum amount of


S. pneumoniae


16S ribosomal RNA as detected by probing Northerns with a suitably labelled sequence specific oligonucleotide probe. Typically a 5′ dye labelled primer is used in each PCR primer pair in a PCR reaction which is terminated optimally between 8 and 25 cycles. The PCR products are separated on 6% polyacrylamide gels with detection and quantification using GeneScanner (manufactured by ABI).




Each of these techniques may have advantages or disadvantage depending on the particular application. The skilled artisan would choose the approach that is the most relevant with the particular end use in mind. For example, some genes might be recognised as essential for infection but in reality are only necessary for the initiation of infection and so their products would represent relatively unattractive targets for antibacterials developed to cure established and chronic infections.




Use of these off technologies when applied to the ORFs of the present invention enables identification of bacterial proteins expressed during infection, inhibitors of which would have utility in anti-bacterial therapy.






Streptococcus pneumoniae


, strain 0100993 has been deposited at the National Collection of Industrial and Marine Bacteria Ltd. (NCIMB), Aberdeen, Scotland under NCIMB number 40794 on Apr. 11, 1996, and a


Streptococcus pneumoniae


, strain 0100993 DNA library in


E. coli


was similarly deposited on Apr. 17, 1996 under NCIMB number




The nucleotide sequences disclosed herein can be obtained by synthetic chemical techniques known in the art or can be obtained from


Streptococcus pneumoniae


, strain 100993 by probing a DNA preparation with probes constructed from the particular sequences disclosed herein. Alternatively, oligonucleotides derived from a disclosed sequence can act as PCR primers in a process of PCR-based cloning of the sequence from a bacterial genomic source. It is recognised that such sequences will also have utility in diagnosis of the stage of infection and type of infection the pathogen has attained.




To obtain the polynucleotide encoding the protein using the DNA sequence given herein typically a library of clones of chromosomal DNA of


Streptococcus pneumoniae


, strain 0100993 in


E. coli


or some other suitable host is probed with a radiolabelled oligonucleotide, preferably a 17 mer or longer, derived from the partial sequence. Clones carrying DNA identical to that of the probe can then be distinguished using high stringency washes. By sequencing the individual clones thus identified with sequencing primers designed from the original sequence it is then possible to extend the sequence in both directions to determine the full gene sequence. Conveniently such sequencing is performed using denatured double stranded DNA prepared from a plasmid clone. Suitable techniques are described by Maniatis, T., Fritsch, E. F. and Sambrook, J. in MOLECULAR CLONING, A Laboratory Manual, 2nd edition, 1989, Cold Spring Harbor Laboratory (see: Screening By Hybridization 1.90 and Sequencing Denatured Double-Stranded DNA Templates 13.70).




A polynucleotide of the present invention may be in the form of RNA or in the form of DNA, which DNA includes cDNA, genomic DNA, and synthetic DNA. The DNA may be double-stranded or single-stranded, and if single stranded may be the coding strand or non-coding (anti-sense) strand. The coding sequence which encodes the polypeptide may be identical to the coding sequence shown or may be a different coding sequence which coding sequence, as a result of the redundancy or degeneracy of the genetic code, encoding the same polypeptide.




The present invention includes variants of the hereinabove described polynucleotides which encode fragments, analogues and derivatives of the polypeptide characterized by the deduced amino acid sequence given herein. The variant of the polynucleotide may be a naturally occurring allelic variant of the polynucleotide or a non-naturally occurring variant of the polynucleotide. In addition to the standard A, G, C, T/U representations for nucleic acid bases, the term “N” is also used. “N” means that any of the four DNA or RNA bases may appear at such a designated position in the DNA or RNA sequence, except that N cannot be a base that when taken in combination with adjacent nucleotide positions, when read in the correct reading frame, would have the effect of generating a premature termination codon in such reading frame.




Thus, the present invention includes polynucleotides encoding the same polypeptide characterized by the deduced amino acid sequence given herein as well as variants of such polynucleotides which variants encode for a fragment, derivative or analogue of the polypeptide. Such nucleotide variants include deletion variants, substitution variants and addition or insertion variants.




The polynucleotide may have a coding sequence which is a naturally occurring allelic variant of the coding sequence characterized by the DNA sequence disclosed herein. As known in the art, an allelic variant is an alternate form of a polynucleotide sequence which may have a substitution, deletion or addition of one or more nucleotides, which does not substantially alter the function of the encoded polypeptide.




The polynucleotide which encodes for the mature polypeptide, may include only the coding sequence for the mature polypeptide or the coding sequence for the mature polypeptide and additional coding sequence such as a leader or secretory sequence or a proprotein sequence.




Thus, the term “polynucleotide encoding a polypeptide” encompasses a polynucleotide which includes only coding sequence for the polypeptide as well as a polynucleotide which includes additional coding and/or non-coding sequence.




The present invention therefore includes polynucleotides, wherein the coding sequence for the mature polypeptide may be fused in the same reading frame to a polynucleotide sequence which aids in expression and secretion of a polypeptide from a host cell, for example, a leader sequence which functions as a secretory sequence for controlling transport of a polypeptide from the cell. The polypeptide having a leader sequence is a preprotein and may have the leader sequence cleaved by the host cell to form the mature form of the polypeptide. The polynucleotides may also encode for a proprotein which is the mature protein plus additional 5′ amino acid residues. A mature protein having a prosequence is a proprotein and may be an inactive form of the protein. Once the prosequence is cleaved an active mature protein remains.




Thus, for example, the polynucleotide of the present invention may code for a mature protein, or for a protein having a prosequence or for a protein having both a prosequence and a presequence (leader sequence). Further, the amino acid sequences provided herein show a methionine residue at the NH


2


-terminus. It is appreciated, however, that during post-translational modification of the peptide, this residue may be deleted. Accordingly, this invention contemplates the use of both the methionine-containing and the methionineless amino terminal variants of each protein disclosed herein.




The polynucleotides of the present invention may also have the coding sequence fused in frame to a marker sequence at either the 5′ or 3′ terminus of the gene which allows for purification of the polypeptide of the present invention. The marker sequence may be a hexa-histidine tag supplied by the pQE series of vectors (supplied commercially by Quiagen Inc.) to provide for purification of the polypeptide fused to the marker in the case of a bacterial host.




The present invention further relates to polynucleotides which hybridize to the hereinabove-described sequences if there is at least 50% and preferably at least 70% identity between the sequences. The present invention particularly relates to polynucleotides, particularly Streptococcal polynucleotides, which hybridize under stringent conditions to the hereinabove-described polynucleotides . As herein used, the term “stringent conditions” means hybridization will occur only if there is at least 95% and preferably at least 97% identity between the sequences. The polynucleotides which hybridize to the hereinabove described polynucleotides in a preferred embodiment encode polypeptides which retain substantially the same biological function or activity as the polypeptide characterised by the deduced amino acid sequence given herein. An example of stringent hybridization conditions is overnight incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing the hybridization support in 0.1×SSC at about 65° C. Hybridization and wash conditions are well known and exemplified in Sambrook, et al., Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989), particularly Chapter 11 therein.




The invention also provides a polynucleotide consisting essentially of a polynucleotide sequence obtainable by screening an appropriate library containing the complete gene for a polynucleotide sequence selected from the group consisting of the polynucleotides of the Sequence Listing under stringent hybridization conditions with a probe having the sequence of said polynucleotide sequence set forth in said polynucleotide of the Sequence Listing or a fragment thereof; and isolating said DNA sequence. Fragments useful for obtaining such a polynucleotide include, for example, probes and primers described elsewhere herein.




“Identity,” as known in the art and used herein, is a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, as determined by comparing the sequences. In the art, “identity” also means the degree of sequence relatedness between polypeptide or polynucleotide sequences, as the case may be, as determined by the match between strings of such sequences. “Identity” and “similarity” can be readily calculated by known methods, including but not limited to those described in (


Computational Molecular Biology


, Lesk, A. M., ed., Oxford University Press, New York, 1988;


Biocomputing: Informatics and Genome Projects


, Smith, D. W., ed., Academic Press, New York, 1993;


Computer Analysis of Sequence Data


, Part I, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994;


Sequence Analysis in Molecular Biology


, von Heinje, G., Academic Press, 1987; and


Sequence Analysis Primer


, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; and Carillo, H., and Lipman, D., SIAM


J. Applied Math,


48: 1073 (1988). Preferred methods to determine identity are designed to give the largest match between the sequences tested. Methods to determine identity and similarity are codified in publicly available computer programs. Preferred computer program methods to determine identity and similarity between two sequences include, but are not limited to, the GCG program package (Devereux, J., et al.,


Nucleic Acids Research


12(1): 387 (1984)), BLASTP, BLASTN, and FASTA (Atschul, S. F. et al.,


J. Molec. Biol.


215: 403-410 (1990). The BLAST X program is publicly available from NCBI and other sources (


BLAST Manual,


Altschul, S., et al., NCBI NLM NIH Bethesda, Md. 20894; Altschul, S., et al.,


J. Mol. Biol.


215: 403-410 (1990).




As an illustration, by a polynucleotide having a nucleotide sequence having at least, for example, 95% “identity” to a reference nucleotide sequence selected from the group consisting of the polynucleotide of the Sequence Listing is intended that the nucleotide sequence of the polynucleotide is identical to the reference sequence except that the polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucletide sequence. In other words, to obtain a polynucleotide having a nucleotide sequence at least 95% identical to a reference nucleotide sequence, up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence. These mutations of the reference sequence may occur at the 5 or 3 terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence.




Similarly, by a polypeptide having an amino acid sequence having at least, for example, 95% identity to a reference amino acid sequence of selected from the group consisting of the amino acids of the Sequence Listing is intended that the amino acid sequence of the polypeptide is identical to the reference sequence except that the polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the reference amino acid. In other words, to obtain a polypeptide having an amino acid sequence at least 95% identical to a reference amino acid sequence, up to 5% of the amino acid residues in the reference sequence may be deleted or substituted with another amino acid, or a number of amino acids up to 5% of the total amino acid residues in the reference sequence may be inserted into the reference sequence. These alterations of the reference sequence may occur at the amino or carboxy terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence.




The deposit referred to herein will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Micro-organisms for purposes of Patent Procedure. These deposits are provided merely as convenience to those of skill in the art and are not an admission that a deposit is required under 35 U.S.C. §112. The sequence of the polynucleotides contained in the deposited material, as well as the amino acid sequence of the polypeptides encoded thereby, are incorporated herein by reference and are controlling in the event of any conflict with any description of sequences herein. A license may be required to make, use or sell the deposited material, and no such license is hereby granted.




The terms “fragment,” “derivative” and “analogue” when referring to the polypeptide characterized by the deduced amino acid sequence herein, means a polypeptide which retains essentially the same biological function or activity as such polypeptide. Thus, an analogue includes a proprotein which can be activated by cleavage of the proprotein portion to produce an active mature polypeptide.




The polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic polypeptide, preferably a recombinant polypeptide.




The fragment, derivative or analogue of the polypeptide characterized by the deduced amino acid sequence herein may be (i) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the polypeptide, such as a leader or secretory sequence or a sequence which is employed for purification of the polypeptide or a proprotein sequence. Such fragments, derivatives and analogues are deemed to be within the scope of those skilled in the art from the teachings herein.




The polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.




The term “isolated” means that the material is removed from its original environment (e.g., the natural environment if it is naturally occurring). For example, a naturally-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated from some or all of the coexisting materials in the natural system, is isolated. Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment.




The present invention also relates to vectors which include polynucleotides of the present invention, host cells which are genetically engineered with vectors of the invention and the production of polypeptides of the invention by recombinant techniques.




In accordance with yet a further aspect of the present invention, there is therefore provided a process for producing the polypeptide of the invention by recombinant techniques by expresing a polynucleotide encoding said polypeptide in a host and recovering the expressed product. Alternatively, the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.




Host cells are genetically engineered (transduced or transformed or transfected) with the vectors of this invention which may be, for example, a cloning vector or an expression vector. The vector may be, for example, in the form of a plasmid, a cosmid, a phage, etc. The engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the genes. The culture conditions, such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.




Suitable expression vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., bacterial plasmids; phage DNA; baculovirus; yeast plasmids; vectors derived from combinations of plasmids and phage DNA. However, any other vector may be used as long as it is replicable and viable in the host.




The appropriate DNA sequence may be inserted into the vector by a variety of procedures. In general, the DNA sequence is inserted into an appropriate restriction endonuclease site(s) by procedures known in the art.




The DNA sequence in the expression vector is operatively linked to an appropriate expression control sequence(s) (promoter) to direct mRNA synthesis. As representative examples of such promoters, there may be mentioned: LTR or SV40 promoter, the


E. coli


Lac or trp, the phage lambda P


L


promoter and other promoters known to control expression of genes in eukaryotic or prokaryotic cells or their viruses. The expression vector also contains a ribosome binding site for translation initiation and a transcription terminator. The vector may also include appropriate sequences for amplifying expression.




In addition, the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in


E. coli.






The gene can be placed under the control of a promoter, ribosome binding site (for bacterial expression) and, optionally, an operator (collectively referred to herein as “control” elements), so that the DNA sequence encoding the desired protein is transcribed into RNA in the host cell transformed by a vector containing this expression construction. The coding sequence may or may not contain a signal peptide or leader sequence. The polypeptides of the present invention can be expressed using, for example, the


E. coli


tac promoter or the protein A gene (spa) promoter and signal sequence. Leader sequences can be removed by the bacterial host in post-translational processing. See, e.g., U.S. Pat. Nos. 4,431,739; 4,425,437; 4,338,397. Promoter regions can be selected from any desired gene using CAT (chloramphenicol transferase) vectors or other vectors with selectable markers. Two appropriate vectors are PKK232-8 and PCM7. Particular named bacterial promoters include lac, lacZ, T3, T7, gpt, lambda P


R


, P


L


and trp. Eukaryotic promoters include CMV immediate early, HSV thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-I. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.




In addition to control sequences, it may be desirable to add regulatory sequences which allow for regulation of the expression of the protein sequences relative to the growth of the host cell. Regulatory sequences are known to those of skill in the art, and examples include those which cause the expression of a gene to be turned on or off in response to a chemical or physical stimulus, including the presence of a regulatory compound. Other types of regulatory elements may also be present in the vector, for example, enhancer sequences.




An expression vector is constructed so that the particular coding sequence is located in the vector with the appropriate regulatory sequences, the positioning and orientation of the coding sequence with respect to the control sequences being such that the coding sequence is transcribed under the “control” of the control sequences (i.e., RNA polymerase which binds to the DNA molecule at the control sequences transcribes the coding sequence). Modification of the coding sequences may be desirable to achieve this end. For example, in some cases it may be necessary to modify the sequence so that it may be attached to the control sequences with the appropriate orientation; i.e., to maintain the reading frame. The control sequences and other regulatory sequences may be ligated to the coding sequence prior to insertion into a vector, such as the cloning vectors described above. Alternatively, the coding sequence can be cloned directly into an expression vector which already contains the control sequences and an appropriate restriction site.




Generally, recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e.g., the ampicillin resistance gene of


E. coli


and


S. cerevisiae


TRP


1


gene, and a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence. The heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein into the periplasmic space or extracellular medium. Optionally, the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.




The vector containing the appropriate DNA sequence as hereinabove described, as well as an appropriate promoter or control sequence, may be employed to transform an appropriate host to permit the host to express the protein.




More particularly, the present invention also includes recombinant constructs comprising one or more of the sequences as broadly described above. The constructs comprise a vector, such as a plasmid or viral vector, into which a sequence of the invention has been inserted, in a forward or reverse orientation. In a preferred aspect of this embodiment, the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters are known to those of skill in the art, and are commercially available. The following vectors are provided by way of example. Bacterial: pET-3 vectors (Stratagene), pQE70, pQE60, pQE-9 (Qiagen), pbs, pD10, phagescript, psiX174, pbluescript SK, pbsks, pNH8A, pNH16a, pNH18A, pNH46A (Stratagene); ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia). Eukaryotic: pBlueBacIII (Invitrogen), pWLNEO, pSV2CAT, pOG


44


, pXT1, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia). However, any other plasmid or vector may be used as long as they are replicable and viable in the a host.




Examples of recombinant DNA vectors for cloning and host cells which they can transform include the bacteriophage l (


E. coli


), pBR322 (


E. coli


), pACYC177 (


E. coli


), pKT230 (gram-negative bacteria), pGV1106 (gram-negative bacteria), pLAFR1 (gram-negative bacteria), pME290 (non-


E. coli


gram-negative bacteria), pHV14 (


E. coli


and


Bacillus subtilis


), pBD9 (Bacillus), pIJ61 (Streptomyces), pUC6 (Streptomyces), YIp5 (Saccharomyces), a baculovirus insect cell system, YCp19 (Saccharomyces). See, generally, “DNA Cloning”: Vols. I & II, Glover et al. ed. IRL Press Oxford (1985) (1987) and; T. Maniatis et al. (“Molecular Cloning” Cold Spring Harbor Laboratory (1982).




In some cases, it may be desirable to add sequences which cause the secretion of the polypeptide from the host organism, with subsequent cleavage of the secretory signal.




Polypeptides can be expressed in host cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al., Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989), the disclosure of which is hereby incorporated by reference.




Following transformation of a suitable host strain and growth of the host strain to an appropriate cell density, the selected promoter is induced by appropriate means (e.g., temperature shift or chemical induction) and cells are cultured for an additional period.




Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification.




Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents, such methods are well known to those skilled in the art.




Depending on the expression system and host selected, the polypeptide of the present invention may be produced by growing host cells transformed by an expression vector described above under conditions whereby the polypeptide of interest is expressed. The polypeptide is then isolated from the host cells and purified. If the expression system secretes the polypeptide into growth media, the polypeptide can be purified directly from the media. If the polypeptide is not secreted, it is isolated from cell lysates or recovered from the cell membrane fraction. Where the polypeptide is localized to the cell surface, whole cells or isolated membranes can be used as an assayable source of the desired gene product. Polypeptide expressed in bacterial hosts such as


E. coli


may require isolation from inclusion bodies and refolding. Where the mature protein has a very hydrophobic region which leads to an insoluble product of overexpression, it may be desirable to express a truncated protein in which the hydrophobic region has been deleted. The selection of the appropriate growth conditions and recovery methods are within the skill of the art.




The polypeptide can be recovered and purified from recombinant cell cultures by methods including ammonium sulphate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Protein refolding steps can be used, as necessary, in completing configuration of the mature protein. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps.




Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. Polypeptides of the invention may also include an initial methionine amino acid residue.




A “replicon” is any genetic element (e.g., plasmid, chromosome, virus) that functions as an autonomous unit of DNA replication in vivo; i.e., capable of replication under its own control.




A “vector” is a replicon, such as a plasmid, phage, or cosmid, to which another DNA segment may be attached so as to bring about the replication of the attached segment.




A “double-stranded DNA molecule” refers to the polymeric form of deoxyribonucleotides (bases adenine, guanine, thymine, or cytosine) in a double-stranded helix, both relaxed and supercoiled. This term refers only to the primary and secondary structure of the molecule, and does not limit it to any particular tertiary forms. Thus, this term includes double-stranded DNA found, inter-alia, in linear DNA molecules (e.g., restriction fragments), viruses, plasmids, and chromosomes. In discussing the structure of particular double-stranded DNA molecules, sequences may be described herein according to the normal convention of giving only the sequence in the 5′ to 3′ direction along the nontranscribed strand of DNA (i.e., the strand having the sequence homologous to the mRNA).




A DNA “coding sequence of” or a “nucleotide sequence encoding” a particular protein, is a DNA sequence which is transcribed and translated into a polypeptide when placed under the control of appropriate regulatory sequences.




A “promoter sequence” is a DNA regulatory region capable of binding RNA polymerase in a cell and initiating transcription of a downstream (3′ direction) coding sequence. For purposes of defining the present invention, the promoter sequence is bound at the 3′ terminus by a translation start codon (e.g., ATG) of a coding sequence and extends upstream (5′ direction) to include the minimum number of bases or elements necessary to initiate transcription at levels detectable above background. Within the promoter sequence will be found a transcription initiation site (conveniently defined by mapping with nuclease S1), as well as protein binding domains (consensus sequences) responsible for the binding of RNA polymerase. Eukaryotic promoters will often, but not always, contain “TATA” boxes and “CAT” boxes. Prokaryotic promoters contain Shine-Dalgarno sequences in addition to the −10 and −35 consensus sequences.




DNA “control sequences” refers collectively to promoter sequences, ribosome binding sites, polyadenylation signals, transcription termination sequences, upstream regulatory domains enhancers, and the like, which collectively provide for the expression (i.e., the transcription and translation) of a coding sequence in a host cell.




A control sequence “directs the expression” of a coding sequence in a cell when RNA polymerase will bind the promoter sequence and transcribe the coding sequence into mRNA, which is then translated into the polypeptide encoded by the coding sequence.




A “host cell” is a cell which has been transformed or transfected, or is capable of transformation or transfection by an exogenous DNA sequence.




A cell has been “transformed” by exogenous DNA when such exogenous DNA has been introduced inside the cell membrane. Exogenous DNA may or may not be integrated (covalently linked) into chromosomal DNA making up the genome of the cell. In prokaryotes and yeasts, for example, the exogenous DNA may be maintained on an episomal element, such as a plasmid. With respect to eukaryotic cells, a stably transformed or transfected cell is one in which the exogenous DNA has become integrated into the chromosome so that it is inherited by daughter cells through chromosome replication. This stability is demonstrated by the ability of the eukaryotic cell to establish cell lines or clones comprised of a population of daughter cell containing the exogenous DNA.




A “clone” is a population of cells derived from a single cell or common ancestor by mitosis. A “cell line” is a clone of a primary cell that is capable of stable growth in vitro for many generations.




A “heterologous” region of a DNA construct is an identifiable segment of DNA within or attached to another DNA molecule that is not found in association with the other molecule in nature.




This invention is also related to the use of the polynucleotides of the invention for use as diagnostic reagents. Detection of a polynucleotide or polypeptide of the invention in a eukaryote, particularly a mammal, and especially a human, will provide a diagnostic method for diagnosis of a disease. Eukaryotes (herein also “individual(s)”), particularly mammals, and especially humans, infected with an organism comprising a polynucleotide of the invention may be detected at the nucleic acid level by a variety of techniques.




Nucleic acids for diagnosis may be obtained from an infected individual's cells and tissues, such as bone, blood, muscle, cartilage, and skin. Genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR or other amplification technique prior to analysis. RNA or cDNA may also be used in the same ways. Using amplification, characterization of the species and strain of prokaryote present in an individual, may be made by an analysis of the genotype of the prokaryote gene. Deletions and insertions can be detected by a change in size of the amplified product in comparison to the genotype of a reference sequence. Point mutations can be identified by hybridizing amplified DNA to labeled polynucleotide sequences of the invention. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase digestion or by differences in melting temperatures. DNA sequence differences may also be detected by alterations in the electrophoretic mobility of the DNA fragments in gels, with or without denaturing agents, or by direct DNA sequencing. See, e.g., Myers et al.,


Science,


230: 1242 (1985). Sequence changes at specific locations also may be revealed by nuclease protection assays, such as RNase and S1 protection or a chemical cleavage method. See, e.g., Cotton et al.,


Proc. Natl. Acad. Sci


, USA, 85: 4397-4401 (1985).




Cells carrying mutations or polymorphisms in the gene of the invention may also be detected at the DNA level by a variety of techniques, to allow for serotyping, for example. For example, RT-PCR can be used to detect mutations. It is particularly preferred to used RT-PCR in conjunction with automated detection systems, such as, for example, GeneScan. RNA or cDNA may also be used for the same purpose, PCR or RT-PCR. As an example, PCR primers complementary to a nucleic acid encoding a polypeptide of the invention can be used to identify and analyze mutations. These primers may be used for, among other things, amplifying DNA isolated from a sample derived from an individual. The primers may be used to amplify the gene isolated from an infected individual such that the gene may then be subject to various techniques for elucidation of the DNA sequence. In this way, mutations in the DNA sequence may be detected and used to diagnose infection and to serotype and/or classify the infectious agent.




The invention further provides a process for diagnosing, disease, preferably bacterial infections, more preferably infections by


Streptococcus pneumoniae


, comprising determining from a sample derived from an individual a increased level of expression of polynucleotide having a sequence set forth in the Sequence Listing or a sequence of the invention. Increased or decreased expression of a polynucleotide of the invention can be measured using any on of the methods well known in the art for the quantation of polynucleotides, such as, for example, amplification, PCR, RT-PCR, RNase protection, Northern blotting and other hybridization methods.




In addition, a diagnostic assay in accordance with the invention for detecting over-expression of a polypeptide of the invention compared to normal control tissue samples may be used to detect the presence of an infection, for example. Assay techniques that can be used to determine levels of a polypeptide of the invention, in a sample derived from a host are well-known to those of-skill in the art. Such assay methods include radioimmunoassays, competitive-binding assays, Western Blot analysis and ELISA assays.




In accordance with yet a further aspect of the present invention, there is provided the use of a polypeptide of the invention for therapeutic or prophylactic purposes, for example, as an antibacterial agent or a vaccine.




In accordance with another aspect of the present invention, there is provided the use of a polynucleotide of the invention for therapeutic or prophylactic purposes, in particular genetic immunisation.




In accordance with yet another aspect of the present invention, there are provided inhibitors to such polypeptides, useful as antibacterial agents. In particular, there are provided antibodies against such polypeptides.




Polypeptides of the invention may also be used to assess the binding of small molecule substrates and ligands in, for example, cells, cell-free preparations, chemical libraries, and natural product mixtures. These substrates and ligands may be natural substrates and ligands or may be structural or functional mimetics. See, e.g., Coligan et al.,


Current Protocols in Immunology


1(2): Chapter 5 (1991).




The invention also provides a method of screening compounds to identify those which enhance (agonist) or block (antagonist) the action of polypeptides or polynucleotides of the invention, particularly those compounds that are bacteriostatic and/or bacteriocidal. The method of screening may involve high-throughput techniques. For example, to screen for agonists or antagoists, a synthetic reaction mix, a cellular compartment, such as a membrane, cell envelope or cell wall, or a preparation of any thereof, comprising a polypeptide of the invention and a labeled substrate or ligand of such polypeptide is incubated in the absence or the presence of a candidate molecule that may be an agonist or antagonist of a polypeptide of the invention. The ability of the candidate molecule to agonize or antagonize a polypeptide of the invention is reflected in decreased binding of the labeled ligand or decreased production of product from such substrate. Molecules that bind gratuitously, i.e., without inducing the effects of a polypeptide of the invention are most likely to be good antagonists. Molecules that bind well and increase the rate of product production from substrate are agonists. Detection of the rate or level of production of product from substrate may be enhanced by using a reporter system. Reporter systems that may be useful in this regard include but are not limited to colorimetric labeled substrate converted into product, a reporter gene that is responsive to changes in polynucleotide or polypeptide activity, and bindinig assays known in the art.




Another example of an assay for antagonists of a polypeptide of the invention is a competitive assay that conmbines such polypeptide and a potential antagonist with polypeptide-binding molecules, recombinant polypeptide-binding molecules, natural substrates or ligands, or substrate or ligand mimetics, under appropriate conditions for a competitive inhibition assay. Polypeptides of the invention can be labeled, such as by radioactivity or a colorimetric compound, such that the number of such polypeptide molecules bound to a binding molecule or converted to product can be determined accurately to assess the effectiveness of the potential antagonist.




Potential antagonists include small organic molecules, peptides, polypeptides and antibodies that bind to a polynucleotide or polypeptide of the invention and thereby inhibit or extinguish its activity. Potential antagonists also may be small organic molecules, a peptide, a polypeptide such as a closely related protein or antibody that binds the same sites on a binding molecule, without inducing an activities of a polypeptide of the invention, thereby preventing the action of such polypeptide by excluding it from binding.




Potential antagonists include a small molecule that binds to and occupies the binding site of the polypeptide thereby preventing binding to cellular binding molecules, such that normal biological activity is prevented. Examples of small molecules include but are not limited to small organic molecules, peptides or peptide-like molecules. Other potential antagonists include antisense molecules (see Okano,


J. Neurochem.


56: 560 (1991); OLIGODEOXYNUCLEOTIDES AS ANTISENSE INHIBITORS OF GENE EXPRESSION, CRC Press, Boca Raton, Fla. (1988), for a description of these molecules). Preferred potential antagonists include compounds related to and variants of the polypetides of the invention.




Each of the DNA sequences provided herein may be used in the discovery and development of antibacterial compounds. The encoded protein, upon expression, can be used as a target for the screening of antibacterial drugs. Additionally, the DNA sequences encoding the amino terminal regions of the encoded protein or Shine-Delgarno or other translation facilitating sequences of the respective mRNA can be used to construct antisense sequences to control the expression of the coding sequence of interest.




The invention also provides the use of the polypeptide, polynucleotide or inhibitor of the invention to interfere with the initial physical interaction between a pathogen and mammalian host responsible for sequelae of infection. In particular the molecules of the invention may be used: in the prevention of adhesion of bacteria, in particular gram positive bacteria, to mammalian extracellular matrix proteins on in-dwelling devices or to extracellular matrix proteins in wounds; to block protein-mediated mammalian cell invasion by, for example, initiating phosphorylation of mammalian tyrosine kinases (Rosenshine et al.,


Infect. Immun.


60:2211 (1992); to block bacterial adhesion between mammalian extracellular matrix proteins and bacterial proteins that mediate tissue damage and; to block the normal progression of pathogenesis in infections initiated other than by the implantation of in-dwelling devices or by other surgical techniques.




The antagonists and agonists of the invention may be employed, for instance, to inhibit and treat otitis media, conjunctivitis, pneumonia, bacteremia, meningitis, sinusitis, pleural empyema and endocarditis, and most particularly meningitis, such as for example infection of cerebrospinal fluid.




Another aspect of the invention is a pharmaceutical composition comprising the above polypeptide, polynucleotide or inhibitor of the invention and a pharmaceutically acceptable carrier.




In a particular aspect the invention provides the use of an inhibitor of the invention as an antibacterial agent.




The invention further relates to the manufacture of a medicament for such uses.




The polypeptide may be used as an antigen for vaccination of a host to produce specific antibodies which have anti-bacterial action. This invention also contemplates the use of the DNA encoding the antigen as a component in a DNA vaccine as discussed more fully below.




The polypeptides or cells expressing them can be used as an immunogen to produce antibodies thereto. These antibodies can be, for example, polyclonal or monoclonal antibodies. The term antibodies also includes chimeric, single chain, and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library. Various procedures known in the art may be used for the production of such antibodies and fragments.




Antibodies generated against the polypeptides of the present invention can be obtained by direct injection of the polypeptides into an animal or by administering the polypeptides to an animal, preferably a nonhuman. The antibody so obtained will then bind the polypeptides itself. In this manner, even a sequence encoding only a fragment of the polypeptides can be used to generate antibodies binding the whole native polypeptides. Such antibodies can then be used to isolate the polypeptide from tissue expressing that polypeptide.




Polypeptide derivatives include antigenically or immunologically equivalent derivatives which form a particular aspect of this invention.




The term ‘antigenically equivalent derivative’ as used herein encompasses a polypeptide or its equivalent which will be specifically recognised by certain antibodies which, when raised to the protein or polypeptide according to the present invention, interfere with the interaction between pathogen and mammalian host.




The term ‘immunologically equivalent derivative’ as used herein encompasses a peptide or its equivalent which when used in a suitable formulation to raise antibodies in a vertebrate, the antibodies act to interfere with the interaction between pathogen and mammalian host.




In particular derivatives which are slightly longer or slightly shorter than the native protein or polypeptide fragment of the present invention may be used. In addition, polypeptides in which one or more of the amino acid residues are modified may be used. Such peptides may, for example, be prepared by substitution, addition, or rearrangement of amino acids or by chemical modification thereof. All such substitutions and modifications are generally well known to those skilled in the art of peptide chemistry.




The polypeptide, such as an antigenically or immunologically equivalent derivative or a fusion protein thereof is used as an antigen to immunize a mouse or other animal such as a rat or chicken. The fusion protein may provide stability to the polypeptide. The antigen may be associated, for example by conjugation, with an immunogenic carrier protein for example bovine serum albumin (BSA) or keyhole limpet haemocyanin (KLH). Alternatively a multiple antigenic peptide comprising multiple copies of the protein or polypeptide, or an antigenically or immunologically equivalent polypeptide thereof may be sufficiently antigenic to improve immunogenicity so as to obviate the use of a carrier.




For preparation of monoclonal antibodies, any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler and Milstein,


Nature,


256:495-497(1975)), the trioma technique, the human B-cell hybridoma technique (Kozbor et al.,


Immunology Today


4:72(1983)), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole, et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96).




Techniques described for the production of single chain antibodies (U.S. Pat. No. 4,946,778) can be adapted to produce single chain antibodies to immunogenic polypeptide products of this invention.




Using the procedure of Kohler and Milstein (supra, (1975)), antibody-containing cells from the immunised mammal are fused with myeloma cells to create hybridoma cells secreting monoclonal antibodies.




The hybridomas are screened to select a cell line with high binding affinity and favorable cross reaction with other Streptococcal species using one or more of the original polypeptide and/or the fusion protein. The selected cell line is cultured to obtain the desired Mab.




Hybridoma cell lines secreting the monoclonal antibody are another aspect of this invention.




Alternatively phage display technology could be utilised to select antibody genes with binding activities towards the polypeptide either from repertoires of PCR amplified v-genes of lymphocytes from humans screened for possessing anti-Fbp or from naive libraries (McCafferty, J. et al.,


Nature


348:552-554(1990), and Marks, J. et al.,


Biotechnology


10:779-783(1992)). The affinity of these antibodies can also be improved by chain shuffling (Clackson, T. et al.,


Nature


352:624-628(1991)).




The antibody should be screened again for high affinity to the polypeptide and/or fusion protein.




As mentioned above, a fragment of the final antibody may be prepared.




The antibody may be either intact antibody of M


r


approx 150,000 or a derivative of it, for example a Fab fragment or a Fv fragment as described in Skerra, A and Pluckthun, A.,


Science


240:1038-1040 (1988). If two antigen binding domains are present each domain may be directed against a different epitope—termed ‘bispecific’ antibodies.




The antibody of the invention may be prepared by conventional means for example by established monoclonal antibody technology (Kohler, G. and Milstein, C. (supra (1975) or using recombinant means e.g. combinatorial libraries, for example as described in Huse, W. D. et al.,


Science


246:1275-1281 (1989).




Preferably the antibody is prepared by expression of a DNA polymer encoding said antibody in an appropriate expression system such as described above for the expression of polypeptides of the invention. The choice of vector for the expression system will be determined in part by the host, which may be a prokaryotic cell, such as


E. coli


(preferably strain B) or


Streptomyces sp


. or a eukaryotic cell, such as a mouse C127, mouse myeloma, human HeLa, Chinese hamster ovary, filamentous or unicellular fungi or insect cell. The host may also be a transgenic animal or a transgenic plant (for example, as described in Hiatt, A. et al.,


Nature


340:7678(1989). Suitable vectors include plasmids, bacteriophages, cosmids and recombinant viruses, derived from, for example, baculoviruses and vaccinia.




The Fab fragment may also be prepared from its parent monoclonal antibody by enzyme treatment, for example using papain to cleave the Fab portion from the Fc portion.




Preferably the antibody or derivative thereof is modified to make it less immunogenic in the patient. For example, if the patient is human the antibody may most preferably be ‘humanised’; where the complimentarily determining region(s) of the hybridoma-derived antibody has been transplanted into a human monoclonal antibody , for example as described in Jones, P. et al.,


Nature


321:522-525 (1986), or Tempest et al.,


Biotechnology


9:266-273 (1991).




The modification need not be restricted to one of ‘humanisation’; other primate sequences (for example Newman, R. et al.,


Biotechnology


10: 1455-1460 (1992)) may also be used.




The humanised monoclonal antibody, or its fragment having binding activity, form a particular aspect of this invention.




This invention provides a method of screening drugs to identify those which interfere with the proteins selected as targets herein, which method comprises measuring the interference of the activity of the protein by a test drug. For example if the protein selected has a catalytic activity, after suitable purification and formulation the activity of the enzyme can be followed by its ability to convert its natural substrates. By incorporating different chemically synthesised test compounds or natural products into such an assay of enzymatic activity one is able to detect those additives which compete with the natural substrate or otherwise inhibit enzymatic activity.




The invention also relates to inhibitors identified thereby.




The use of a polynucleotide of the invention in genetic immunisation will preferably employ a suitable delivery method such as direct injection of plasmid DNA into muscles (Wolff et al.,


Hum. Mol. Genet.


1:363 (1992); Manthorpe et al.,


Hum. Gene Ther.


94:419 (1963)), delivery of DNA complexed with specific protein carriers (Wu et al.,


J. Biol. Chem.


264:16985 (1989)), coprecipitation of DNA with calcium phosphate (Benvenisty & Reshef,


Proc. Nat'l Acad. Sci. USA,


83:9551 (1986)), encapsulation of DNA in various forms of liposomes (Kaneda et al.,


Science


243:375 (1989)), particle bombardment (Tang et al.,


Nature


356:152 (1992)); Eisenbraun et al.,


DNA Cell Biol.


12:791 (1993)) and in vivo infection using cloned retroviral vectors (Seeger et al.,


Proc. Nat'l. Acad. Sci. USA


81:5849 (1984)). Suitable promoters for muscle transfection include CMV, RSV, SRa, actin, MCK, alpha globin, adenovirus and dihydrofolate reductase.




In therapy or as a prophylactic, the active agent i.e., the polypeptide, polynucleotide or inhibitor of the invention, may be administered to a patient as an injectable composition, for example as a sterile aqueous dispersion, preferably isotonic.




Alternatively the composition may be formulated for topical application for example in the form of ointments, creams, lotions, eye ointments, eye drops, ear drops, mouthwash, impregnated dressings and sutures and aerosols, and may contain appropriate conventional additives, including, for example, preservatives, solvents to assist drug penetration, and emollients in ointments and creams. Such topical formulations may also contain compatible conventional carriers, for example cream or ointment bases, and ethanol or oleyl alcohol for lotions. Such carriers may constitute from about 1% to about 98% by weight of the formulation; more usually they will constitute up to about 80% by weight of the formulation.




For administration to human patients, it is expected that the daily dosage level of the active agent will be from 0.01 to 10 mg/kg, typically around 1 mg/kg. The physician in any event will determine the actual dosage which will be most suitable for an individual patient and will vary with the age, weight and response of the particular patient. The above dosages are exemplary of the average case. There can, of course, be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.




A vaccine composition is conveniently in injectable form. Conventional adjuvants may be employed to enhance the immune response.




A suitable unit dose for vaccination is 0.5-5 μg/kg of antigen, and such dose is preferably administered 1-3 times and with an interval of 1-3 weeks.




Within the indicated dosage range, no adverse toxicologicals effects are expected with the compounds of the invention which would preclude their administration to suitable patients.




The individual full length sequences given herein are summarized in the following Tables (Table 1 and Table 2). Under the column in Table 1 labeled “Identity” there is the deduced identity of each open reading frame of the invention determined using Blastp and/or MPSearch. The ORF# column indicates whether the polynucleotide encoding each ORF encodes more than one ORF. For example, SEQ ID NO: 263 has an ORF#1 but no #2; thus the polynucleotide encodes this ORF#1, but no other ORF was detected. On the other hand, SEQ ID NO:286 and 287 have ORF#s 1 and 2 respectively, indicating that they were both encoded by the same polynucleotide. This can also be seen in Table 2 where the polynucleotide of SEQ ID NO: 24 encodes the ORFs of SEQ ID NOS: 286 and 287. Table 1 also shows in the position (“POSITION”; columns) the start (“START” column) and stop (“STOP” column) codons for each ORF and their positions in the encoding polynucleotide sequence. The SEQ ID NOS of the polypeptides of this table are linked to both a deduced identity in this table and a polynucleotide sequence in Table 2 which encodes each polypeptide. The “Direction” column in Table 1 shows the direction of the ORF encoding each polypeptide in this table. “Forward” denotes the sense orientation and “Reverse” denotes the antisense orientation of the ORF. Table 1 also provides an “Assembly ID” which is a unique numerical descriptor for each polynucleotide sequence of the invention.




















TABLE 1









SEQ ID





Assembly





Codon





Position








NO:




IDENTITY




ID




ORF #




Start




Stop




Start




Stop




Direction































263.




probable transposase (insertion




3112224




1




ATG




TAA




258




746




Forward







sequence IS861)-


Streptococcus











agalactiae


(strain COH-1)






264.




Unknown




3112506




1




˜CAT




CTA˜




414




554




Reverse






265.




SUCCINYL-DIAMINOPIMELATE




3112574




1




˜CAT




TTA˜




607




990




Reverse







DESUCCINYLASE (EC 3.5.1.18)







(SDAP).-


ESCHERICHIA COLI


.






266.




Unknown




3112646




1




ATG




TGA




353




460




Forward






267.




D-alanine permease (dagA) homolog-




3112686




1




ATG




TAA




393




1235




Forward









Haemophilus influenzae


(strain Rd KW20)






268.




Unknown




3112810




1




˜CAT




TTA˜




328




468




Reverse






269.




Unknown




3112934




1




ATG




TAG




107




739




Forward






270.




3-OXOACYL-[ACYL-CARRIER




3112956




1




ATG




TGA




586




1146




Forward







PROTEIN] REDUCTASE PRECURSOR







(EC 1.1.1.100) (3-KETOACYL-ACYL







CARRIER PROTEIN REDUCTASE).-









CUPHEA LANCEOLATA


.






271.




HIGH-AFFINITY BRANCHED-CHAIN




3112994




1




ATG




TAA




307




648




Forward







AMINO ACID TRANSPORT ATP-BINDING







PROTEIN LIVG (LIV- I PROTEIN G).-









SALMONELLA TYPHIMURIUM


.






272.




Unknown




3113026




1




ATG




TGA




235




483




Forward






273.




Unknown




3113098




1




˜CAT




TTA˜




28




360




Reverse






274.




CELL DIVISION PROTEIN FTSH.-




3113274




1




ATG




TGA




125




700




Forward









BACILLUS SUBTILIS


.






275.




Unknown




3113306




1




˜CAT




CTA˜




255




509




Reverse






276.




PTS SYSTEM, FRUCTOSE-SPECIFIC




3113406




1




˜CAT




TCA˜




237




725




Reverse







IIBC COMPONENT EIIBC-FRU)







(FRUCTOSE-PERMEASE II BC







COMPONENT) (PHOSPHOTRANSFERASE







ENZYME II, BC COMPONENT)







(EC 2.7.1.69) (EII-FRU).-


XANTHOMONAS









CAMPESTRIS (PV.


CAMPESTRIS


).






277.




Unknown




3113432




1




˜CAT




CTA˜




17




310




Reverse






278.




Unknown




3113436




1




CTG




TAA




1




441




Forward






279.




HOLLIDAY JUNCTION DNA HELICASE




3113510




1




ATG




TGA




187




411




Forward







RUVB.-


ESCHERICHIA COLI


.






280.




Unknown




3113514




1




ATG




TGA




361




495




Forward






281.




Unknown




3113546




1




TTG




TAA




2




241




Forward






282.




CELL DIVISION PROTEIN FTSA.-




3113610




1




TTG




TGA




3




665




Forward









BACILLUS SUBTILIS


.






283.




Unknown




3113692




1




ATG




TAG




304




594




Forward






284.




GALACTOSE-6-PHOSPHATE ISOMERASE




3113696




1




ATG




TGA




176




661




Forward







LACB SUBUNIT (EC 5.-.-.-).-









LACTOCOCCUS LACTIS


(SUBSP.









LACTIS


) (


STREPTOCOCCUS LACTIS


).






285.




3-ISOPROPYLMALATE




3113762




1




˜CAG




TTA˜




275




508




Reverse







DEHYDROGENASE (EC 1.1.1.85)







(BETA-IPM DEHYDROGENASE) (IMDH).-









LACTOCOCCUS LACTIS


(SUBSP.









LACTIS


) (


STREPTOCOCCUS LACTIS


).






286.




PHOSPHO-2-DEHYDRO-3-




3113794




1




ATG




TAG




40




219




Forward







DEOXYHEPTONATE ALDOLASE







(EC 4.1.2.15) (PHOSPHO-2-KETO-3-







DEOXYHEPTONATE ALDOLASE) (DAHP







SYNTHETASE) (3-DEOXY-D-ARABINO-







HEPTULOSONATE 7-PHOSPHATE







SYNTHASE).-


CORYNEBACTERIUM











GLUTAMICUM


.






287.




PHOSPHO-2-DEHYDRO-3-




3113794




2




ATG




TGA




283




453




Forward







DEOXYHEPTONATE ALDOLASE,







PHE-SENSITIVE (EC 4.1.2.15)







(PHOSPHO-2-KETO-3-DEOXYHEPTONATE







ALDOLASE) (DAHP SYNTHETASE)







(3-DEOXY-D-ARABINO-







HEPTULOSONATE 7-PHOSPHATE







SYNTHASE).-


ESCHERICHIA COLI


.






288.




PYRUVATE KINASE (EC 2.7.1.40).-




3113802




1




ATG




TGA




69




260




Forward









LACTOCOCCUS LACTIS


(SUBSP.









LACTIS


) (


STREPTOCOCCUS LACTIS


).






289




Unknown




3113990




1




˜CAT




TTA˜




6




164




Reverse






290.




ADAPTIVE-RESPONSE SENSORY-KINASE




3114082




1




ATG




TAG




22




189




Forward







SASA (EC 2.7.-.-).- SYNECHOCOCCUS SP.







(STRAIN PCC 7942) (


ANACYSTIS











NIDULANS


R2).






291.




FOLYLPOLYGLUTAMATE SYNTHASE




3114096




1




CTG




TGA




1




243




Forward







(EC 6.3.2.17) (FOLYLPOLY-GAMMA-







GLUTAMATE SYNTHETASE) (FPGS).-









LACTOBACILLUS CASEI


.






292.




50S RIBOSOMAL PROTEIN L6




3174146




1




˜CAT




TCA˜




581




949




Reverse







(BL10).-


BACILLUS STEAROTHER-











MOPHILUS


.






293.




grpE protein-


Lactococcus lactis






3174148




1




˜CAT




TTA˜




256




780




Reverse






294.




Unknown




3174150




1




˜CAT




TCA˜




347




472




Reverse






295.




ribosomal protein S14 (rpS14) homolog-




3174152




1




˜CAT




TTA˜




137




391




Reverse









Haemophilus influenzae


(strain Rd KW20)






296.




50S RIBOSOMAL PROTEIN L3.-




3174154




1




ATG




TAA




441




1067




Forward









BACILLUS STEAROTHERMOPHILUS


.






297.




PHOSPHATE TRANSPORT SYSTEM




3174166




1




˜CAT




TTA˜




101




751




Reverse







REGULATORY PROTEIN.-









ESCHERICHIA COLI


.






298.




PROBABLE TRANSKETOLASE (EC 2.2.1.1)




3174184




2




˜CAT




TTA˜




895




1350




Reverse







(TK).-


STREPTOCOCCUS PNEUMONIAE


.






299.




ntpJ protein-


Enterococcus hirae






3174206




1




TTG




TAG




2




763




Forward






300.




Unknown




3174208




1




ATG




TAA




311




1054




Forward






301.




Unknown




3174210




1




˜CAT




TTA˜




290




2305




Reverse






302.




ACYL CARRIER PROTEIN.-




3174220




1




ATG




TAG




788




1021




Forward









CRYPTOMONAS PHI


.






303.




ISL2 protein-


Lactobacillus helveticus






3174224




1




˜CAT




TTA˜




447




737




Reverse






304.




ISL2 protein-


Lactobacillus helveticus






3174226




1




ATG




TAG




850




1164




Forward






305.




Unknown




3174228




1




CTG




TAA




1




240




Forward






306.




Unknown




3174228




2




ATG




TAG




278




580




Forward






307.




Possible Ca2+-




3174236




1




˜CAT




TTA˜




53




241




Reverse







transporting ATPase






308.




Possible Ca2+-




3174238




1




˜CAT




TTA˜




53




529




Reverse







transporting ATPase






309.




CATION-TRANSPORTING ATPASE




3174238




2




˜CAT




TCA˜




549




1298




Reverse







PACL (EC 3.6.1.-).- SYNECHOCOCCUS SP.







(STRAIN PCC 7 942) (


ANACYSTIS











NIDULANS


R2).






310.




PROLYL-TRNA SYNTHETASE




3174270




1




˜CAT




TTA˜




150




1646




Reverse







(EC 6.1.1.15) (PROLINE--TRNA LIGASE)







(PRORS) (GLOBAL RNA SYNTHESIS







FACTOR).-


ESCHERICHIA COLI


.






311.




Unknown




3174278




1




ATG




TGA




733




867




Forward






312.




Unknown




3174278




2




ATG




TAA




1680




1880




Forward






313.




PROLYL-TRNA SYNTHETASE




3174284




1




ATG




TAA




1009




2103




Forward







(EC 6.1.1.15) (PROLINE--TRNA LIGASE)







(PRORS) (GLOBAL RNA SYNTHESIS







FACTOR).-


ESCHERICHIA COLI


.






314.




BSCELABCD NCBI gi: 8957-




3174288




1




ATG




TAA




78




464




Forward









Bacillus subtilis


.






315.




GLUTAMINE TRANSPORT ATP-BINDING




3174294




1




ATG




TGA




370




981




Forward







PROTEIN GLNQ.-


ESCHERICHIA COLI


.






316.




Unknown




3174294




2




ATG




TAA




1044




1217




Forward






317.




GLUTAMINE TRANSPORT ATP-BINDING




3174298




1




ATG




TAA




431




976




Forward







PROTEIN GLNQ.-


BACILLUS











STEAROTHERMOPHILUS


.






318.




GLUTAMINE TRANSPORT ATP-BINDING




3174298




2




ATG




TAA




922




1179




Forward







PROTEIN GLNQ.-


BACILLUS











STEAROTHERMOPHILUS


.






319.




Unknown




3174302




1




˜CAT




TTA˜




322




1320




Reverse






320.




Unknown




3174302




2




˜CAT




CTA˜




1486




1857




Reverse






321.




Unknown




3174314




1




ATG




TGA




523




891




Forward






322.




Unknown




3174314




2




ATG




TAG




888




1370




Forward






323.




RIBOFLAVIN SYNTHASE ALPHA CHAIN




3174328




1




˜CAT




CTA˜




213




653




Reverse







(EC 2.5.1.9).-


BACILLUS SUBTILIS


.






324.




5,10-METHYLENETETRAHYDRO-




3174342




1




ATG




TAA




372




1238




Forward







FOLATE REDUCTASE (EC 1.7.99.5).-









SALMONELLA TYPHIMURIUM


.






325.




Unknown




3174356




1




˜CAA




TCA˜




584




1342




Reverse






326.




LSSAKACNP NCBI gi: 695615-




3174368




1




ATG




TAA




219




539




Forward









Lactobacillus sake


.






327.




agrB protein-


Staphylococcus aureus






3174368




2




ATG




TGA




730




1056




Forward






328.




RESD PROTEIN.-


BACILLUS SUBTILIS


.




3174372




1




CTG




TGA




1




315




Forward






329.




PROBABLE TRANSCRIPTIONAL




3174384




1




˜CAT




TCA˜




239




418




Reverse







REGULATORY PROTEIN ENDR.-









BACILLUS POLYMYXA


.






330.




Unknown




3174384




2




˜CAT




CTA˜




514




1074




Reverse






331.




30S RIBOSOMAL PROTEIN S2.-




3174390




1




ATG




TAA




597




1457




Forward









ESCHERICHIA COLI


.






332.




GLUTAMINE TRANSPORT ATP-BINDING




3174402




1




ATG




TGA




372




980




Forward







PROTEIN GLNQ.-


ESCHERICHIA COLI


.






333.




GTP-BINDING PROTEIN ERA HOMOLOG.-




3174420




1




˜CAT




TTA˜




592




1086




Reverse









STREPTOCOCCUS MUTANS


.






334.




GTP-BINDING PROTEIN ERA HOMOLOG.-




3174420




2




˜CAT




TTA˜




1022




1492




Reverse









STREPTOCOCCUS MUTANS


.






335.




SULFATE TRANSPORT ATP-BINDING




3174426




1




ATG




TGA




812




1270




Forward







PROTEIN CYSA.- SYNECHOCOCCUS SP.







(STRAIN PCC 794 2) (


ANACYSTIS











NIDULANS


R2).






336.




NITRATE TRANSPORT PROTEIN




3174426




2




ATG




TAG




1298




1543




Forward







NASD.


KLEBSIELLA PNEUMONIAE


.






337.




PROLIPOPROTEIN DIACYLGLYCERYL




3174428




1




˜CAT




TTA˜




35




835




Reverse







TRANSFERASE (EC 2.4.99.-).-









SALMONELLA TYPHIMURIUM


.






338.




Unknown




3174444




1




ATG




TAA




164




895




Forward






339.




CYSTEINYL-TRNA SYNTHETASE




3174454




1




˜CAT




TCA˜




504




1079




Reverse







(EC 6.1.1.16) (CYSTEINE--TRNA







LIGASE) (CYSRS).-


BACILLUS SUBTILIS


.






340.




Unknown




3174460




1




ATG




TAG




529




1275




Forward






341.




PRIMOSOMAL PROTEIN N′




3174462




1




TTG




TGA




2




721




Forward







(REPLICATION FACTOR Y).-









ESCHERICHIA COLI


.






342.




30S RIBOSOMAL PROTEIN S11




3174466




1




˜CAT




TTA˜




1019




1303




Reverse







(BS11).-


BACILLUS SUBTILIS


.






343.




Unknown




3174474




1




˜CAT




TTA˜




238




423




Reverse






344.




Unknown




3174476




1




˜CAT




TTA˜




241




1446




Reverse






345.




PEPTIDE CHAIN RELEASE FACTOR 3




3174490




1




˜CAT




CTA˜




668




1291




Reverse







(RF-3).-


BACTEROIDES NODOSUS









(


DICHELOBACTER NODOSUS


).






346.




mesI protein-


Leuconostoc mesenteroides






3174496




1




ATG




TAG




812




1666




Forward






347.




Unknown




3174506




1




ATG




TGA




179




352




Forward






348.




GALACTOKINASE (EC 2.7.1.6).-




3174514




1




˜CAT




TCA˜




209




340




Reverse









LACTOBACILLUS HELVETICUS


.






349.




GALACTOKINASE (EC 2.7.1.6).-




3174514




2




˜CAT




TTA˜




1177




1359




Reverse









BACILLUS SUBTILIS


.






350.




FORMATE ACETYLTRANSFERASE 1




3174524




1




˜CAT




TTA˜




19




867




Reverse







(EC 2.3.1.54) (PYRUVATE FORMATE-







LYASE 1).-


ESCHERICHIA COLI


.






351.




Unknown




3174546




1




˜CAT




CTA˜




912




1127




Reverse






352.




CACSPC NCBI gi: 899232-




3174550




1




ATG




TAG




645




956




Forward









Clostridium acetobutylicum


.






353.




Unknown




3174562




1




CTG




TAG




1




504




Forward






354.




KETOACYL REDUCTASE HETN




3174562




2




ATG




TAA




525




1280




Forward







(EC 1.3.1.-).- ANABAENA SP.







(STRAIN PCC 7120).






355.




Unknown




3174570




1




ATG




TAG




249




593




Forward






356.




ATP-DEPENDENT DNA HELICASE




3174570




2




ATG




TGA




604




1695




Forward







RECG (EC 3.6.1.-).-


ESCHERICHIA COLI


.






357.




PROTEIN DLTD PRECURSOR.-




3174580




1




ATG




TGA




3




611




Forward









BACILLUS SUBTILIS


.






358.




ALANYL-TRNA SYNTHETASE




3174582




1




ATG




TGA




537




875




Forward







(EC 6.1.1.7) (ALANINE--TRNA LIGASE)







(ALARS).-


ESCHERICHIA COLI


.






359.




PTS SYSTEM, MANNOSE-SPECIFIC IIC




3174586




1




ATG




TGA




533




865




Forward







COMPONENT (EIIC-MAN) (MANNOSE-







PERMEASE IIC COMPONENT)







(PHOSPHOTRANSFERASE ENZYME II, C







COMPONENT) (EII-P-MAN).-









ESCHERICHIA COLI


.






360.




PTS SYSTEM, FRUCTOSE-SPECIFIC IIC




3174586




2




ATG




TAA




747




1172




Forward







COMPONENT (EIIC-FRU) (FRUCTOSE-







PERMEASE IIC COMPONENT)







(PHOSPHOTRANSFERASE ENZYME II, C







COMPONENT) (P28).-


BACILLUS SUBTILIS


.






361.




URIDYLATE KINASE (EC 2.7.4.-) (UK)




3174594




1




˜CAT




TTA˜




319




579




Reverse







(URIDINE MONOPHOSPHATE KINASE)







(UMP KINASE) (SMBA PROTEIN).-









ESCHERICHIA COLI


.






362.




Unknown




3174606




1




CTG




TAA




1




153




Forward






363.




Unknown




3174608




1




ATG




TAA




336




863




Forward






364.




Unknown




3174634




1




˜CAT




CTA˜




99




308




Reverse






365.




Unknown




3174634




2




˜CAT




TCA˜




305




826




Reverse






366.




5-METHYLTETRAHYDROPTER-




3174642




1




˜CAT




TCA˜




250




1620




Reverse







OYLTRIGLUTAMATE-- HOMOCYSTEINE







METHYLTRANSFERASE (EC 2.1.1 .14)







(METHIONINE SYNTHASE, VITAMIN-B12







INDEPENDENT ISOZYME).-









ESCHERICHIA COLI


.






367.




HIGH-AFFINITY BRANCHED-CHAIN




3174644




1




ATG




TGA




283




765




Forward







AMINO ACID TRANSPORT ATP-BINDING







PROTEIN BRAF.-


PSEUDOMONAS











AERUGINOSA


.






368.




Unknown




3174652




1




ATG




TGA




913




1134




Forward






369.




Unknown




3174654




1




ATG




TGA




158




319




Forward






370.




ADENYLOSUCCINATE LYASE




3174658




1




˜CAT




TTA˜




941




1477




Reverse







(EC 4.3.2.2) (ADENYLOSUCCINASE)







(ASL).-


BACILLUS SUBTILIS


.






371.




Unknown




3174660




1




ATG




TGA




454




594




Forward






372.




Unknown




3174662




1




˜CAT




TCA˜




103




768




Reverse






373.




50S RIBOSOMAL PROTEIN L16.-




3174664




1




ATG




TAA




33




446




Forward









MYCOPLASM ACAPRICOLUM


.






374.




STAGE V SPORULATION PROTEIN E.-




3174666




1




ATG




TAG




151




816




Forward









BACILLUS SUBTILIS


.






375.




Unknown




3174676




1




˜CAT




CTA˜




24




359




Reverse






376.




Unknown




3174678




1




˜CAT




TCA˜




1




207




Reverse






377.




Unknown




3174714




1




ATG




TAA




839




1363




Forward






378.




ATP-DEPENDENT CLP PROTEASE




3174736




1




CTG




TGA




3




317




Forward







ATP-BINDING SUBUNIT CLPX.-









ESCHERICHIA COLI


.






379.




ATP-DEPENDENT CLP PROTEASE




3174736




2




ATG




TAG




411




797




Forward







ATP-BINDING SUBUNIT CLPX.-









ESCHERICHIA COLI


.






380.




Unknown




3174738




1




ATG




TAA




184




558




Forward






381.




Unknown




3174744




1




ATG




TAA




507




1082




Forward






382.




PYRROLIDONE-CARBOXYLATE




3174748




1




ATG




TAA




218




691




Forward







PEPTIDASE (EC 3.4.19.3)







(5-OXOPROLYL-PEPTIDASE).-









STREPTOCOCCUS PYOGENES


.






383.




Unknown




3174748




2




ATG




TGA




693




875




Forward






384.




Unknown




3174760




1




ATG




TGA




280




495




Forward






385.




Unknown




3174770




1




ATG




TGA




226




402




Forward






386.




RECOMBINATION PROTEIN.-




3174772




1




ATG




TAA




898




1527




Forward









BACILLUS SUBTILIS


.






387.




ALPHA-ACETOLACTATE




3174774




1




ATG




TAG




155




550




Forward







DECARBOXYLASE (EC 4.1.1.5).-









ENTEROBACTER AEROGENES









(


AEROBACTER AEROGENES


).






388.




3-ISOPROPYLMALATE DEHYDRATASE




3174784




1




ATG




TAG




291




650




Forward







(EC 4.2.1.33) (ISOPROPYLMALATE







ISOMERASE) (ALPHA- IPM ISOMERASE).-









LACTOCOCCUS LACTIS


(SUBSP.









LACTIS


) (


STREPTOCOCCUS LACTIS


).






389.




peptide chain release factor 1-




3174802




1




ATG




TAA




293




1372




Forward









Bacillus subtilis








390.




ATP-dependent Clp proteinase (EC 3.4.21.-)




3174806




1




˜CAT




CTA˜




592




1194




Reverse







chain clpL-


Lactococcus lactis


subsp.









lactis


plasmid pUCL22






391.




Unknown




3174812




1




ATG




TAA




55




933




Forward






392.




FAD synthase (EC 6.3.-.-)-


Corynebacterium






3174818




1




˜CAT




TTA˜




4




921




Reverse









ammoniagenes








393.




CYTIDINE DEAMINASE (EC 3.5.4.5)




3174826




1




ATG




TAG




25




414




Forward







(CYTIDINE AMINOHYDROLASE) (CDA).-









BACILLUS SUBTILIS


.






394.




Unknown




3174832




1




˜CAT




TTA˜




23




382




Reverse






395.




Unknown




3174838




1




ATG




TAA




184




540




Forward






380.




SARPLRPO NCBI gi: 677848NCBI gi:




3174842




1




˜CAT




TCA˜




299




574




Reverse







473748-


Staphylococcus aureus


.






381.




Unknown




3174852




1




˜CAT




TCA˜




181




540




Reverse






382.




TRK SYSTEM POTASSIUM UPTAKE




3174858




1




CTG




TGA




1




1077




Forward







PROTEIN TRKA HOMOLOG.-









METHANOSARCINA MAZEI


.






383.




Unknown




3174870




1




˜CAT




CTA˜




228




953




Reverse






384.




6-PHOSPHO-BETA-GALACTOSIDASE (EC




3174878




1




˜CAT




TCA˜




227




667




Reverse







3.2.1.85) (BETA-D-PHOSPHOGALACTO-







SIDE GALACTOHYDROLASE).-









LACTOCOCCUS LACTIS


(SUBSP.









LACTIS


)


(STREPTOCOCCUS LACTIS


).






385.




hemolytic factor-


Bacillus cereus






3174926




1




˜CAT




TCA˜




591




1142




Reverse






386.




DIPEPTIDYL PEPTIDASE IV




3174936




1




ATG




TAA




189




479




Forward







(EC 3.4.14.5) (X-PROLYL DIPEPTIDYL







AMINOPEPTIDASE IV) (X-PDAP).-









LACTOCOCCUS LACTIS


(SUBSP.









CREMORIS


) (


STREPTOCOCCUS











CREMORIS


).






387.




ARGININE HYDROXIMATE RESISTANCE




3174936




2




ATG




TGA




496




846




Forward







PROTEIN.-


BACILLUS SUBTILIS


.






388.




Unknown




3174938




1




ATG




TGA




366




587




Forward






389.




Unknown




3174946




1




˜CAT




CTA˜




111




380




Reverse






390.




Unknown




3174952




1




˜CAT




TTA˜




37




819




Reverse






391.




DnaK protein-


Lactococcus lactis






3174970




1




CTG




TGA




3




380




Forward






392.




Unknown




3174990




1




ATG




TAA




141




1058




Forward






409.




LACALS NCBI gi: 473900-


Lactococcus






3175000




1




ATG




TAA




794




1015




Forward









lactis


(strain DSM 20384,







sub-species


lactis


) DNA.






410.




Unknown




3175006




1




ATG




TGA




56




631




Forward






411.




cellobiose phosphotransferase system




3175006




2




ATG




TAA




646




963




Forward







celA-


Bacillus stearothermophilus








412.




Unknown




3175010




1




˜CAT




TTA˜




13




231




Reverse






413.




Unknown




3175014




1




ATG




TGA




58




219




Forward






414.




Unknown




3175016




1




ATG




TAA




120




503




Forward






415.




Unknown




3175032




1




ATG




TAA




364




669




Forward






416.




Unknown




3175046




1




ATG




TGA




105




401




Forward






417.




GLYCYL-TRNA SYNTHETASE ALPHA




3175074




1




˜CAT




CTA˜




107




787




Reverse







CHAIN (EC 6.1.1.14) (GLYCINE--







TRNA LIGASE ALPHA CHAIN) (GLYRS).-









ESCHERICHIA COLI


.






418.




GLYCYL-TRNA SYNTHETASE ALPHA




3175074




2




˜CAT




TCA˜




787




936




Reverse







CHAIN (EC 6.1.1.14) (GLYCINE--







TRNA LIGASE ALPHA CHAIN) (GLYRS).-









ESCHERICHIA COLI


.






419.




endopeptidase PepO (EC 3.4.-.-)-




3175092




1




˜CAT




TTA˜




47




490




Reverse









Lactococcus lactis


subsp.


lactis








420.




BSCELABCD NCBI gi: 895746-




3175094




1




˜CAT




CTA˜




303




1019




Reverse









Bacillus subtilis


.






421.




DNA LIGASE (EC 6.5.1.2) (POLYDEOXY




3175098




1




ATG




TAA




134




265




Forward







RIBONUCLEOTIDE SYNTHASE (NAD+)).-









ESCHERICHIA COLI


.






422.




Unknown




3175098




2




ATG




TAA




210




446




Forward






423.




TAGATOSE 1,6-DIPHOSPHATE




3175102




1




ATG




TGA




495




950




Forward







ALDOLASE (EC 4.1.-.-).-


LACTOCOCCUS











LACTIS


(SUBSP.


LACTIS


)







(


STREPTOCOCCUS LACTIS


).






424.




Unknown




3175104




1




CTG




TAA




1




525




Forward






425.




Unknown




3175114




1




˜CAT




TTA˜




59




508




Reverse






426.




Unknown




3175126




1




ATG




TAA




3




203




Forward






427.




Unknown




3175136




1




ATG




TGA




370




582




Forward






428.




Unknown




3175138




1




˜CAG




TCA˜




1134




1448




Reverse






429.




2-ISOPROPYLMALATE SYNTHASE




3175140




1




ATG




TAA




963




1382




Forward







(EC 4.1.3.12) (ALPHA-ISOPROPYL-







MALATE SYNTHASE) (ALPHA-IPM







SYNTHETASE).-


LACTOCOCCUS LACTIS









(SUBSP.


LACTIS


)







(


STREPTOCOCCUS LACTIS


).






430.




LPLC PROTEIN.-


BACILLUS SUBTILIS


.




3175150




1




˜CAT




TCA˜




246




827




Reverse






431.




Unknown




3175158




1




˜CAT




TTA˜




114




407




Reverse






432.




PROTEASE SYNTHASE AND




3175164




1




˜CAT




TTA˜




245




754




Reverse







SPORULATION NEGATIVE REGULATORY







PROTEIN PAI 1.-


BACILLUS SUBTILIS


.






433.




PUTRESCINE TRANSPORT ATP-BINDING




3175172




1




ATG




TAG




380




844




Forward







PROTEIN POTG.-


ESCHERICHIA COLI


.






434.




TRIOSEPHOSPHATE ISOMERASE




3175174




1




˜CAT




TTA˜




270




818




Reverse







(EC 5.3.1.1) (TIM).-


BACILLUS SUBTILIS


.






435.




Unknown




3175188




1




˜CAT




TTA˜




429




935




Reverse






436.




PNUC PROTEIN.-




3175192




1




ATG




TAG




111




536




Forward









SALMONELLA TYPHIMURIUM


.






437




recF protein-


Streptococcus pyogenes






3175228




1




˜CAT




TTA˜




144




356




Reverse






438.




3-DEHYDROQUINATE SYNTHASE




3175240




1




˜CAT




TTA˜




410




1123




Reverse







(EC 4.6.1.3).-


ESCHERICHIA COLI


.






439.




Unknown




3175256




1




˜CAT




TTA˜




77




283




Reverse






440.




Unknown




3175262




1




ATG




TAA




84




377




Forward






441.




Unknown




3175266




1




˜CAT




TTA˜




50




481




Reverse






442.




Unknown




3175288




1




ATG




TGA




430




522




Forward






443.




NAD-DEPENDENT METHANOL




3175298




1




˜CAT




TTA˜




440




658




Reverse







DEHYDROGENASE (EC 1.1.1.244)







(MEDH).-


BACILLUS METHANOLICUS


.






444.




ATP SYNTHASE ALPHA CHAIN




3175306




1




ATG




TGA




478




639




Forward







(EC 3.6.1.34).-


ENTEROCOCCUS FAECALIS









(


STREPTOCOCCUS FAECALIS


).






445.




ANTHRANILATE SYNTHASE




3175310




1




ATG




TGA




117




683




Forward







COMPONENT II (EC 4.1.3.27)







(GLUTAMINE AMIDO-TRANSFERASE).-









LACTOCOCCUS LACTIS


(SUBSP.









LACTIS


) (


STREPTOCOCCUS LACTIS


).






446.




Unknown




3175322




1




CTG




TAA




3




413




Forward






447.




ALANYL-TRNA SYNTHETASE




3175332




1




ATG




TAA




432




1250




Forward







(EC 6.1.1.7) (ALANINE--TRNA







LIGASE) (ALARS).-


ESCHERICHIA COLI


.






448.




dihydrolipoamide dehydrogenase




3175356




1




˜CAT




TTA˜




486




1241




Reverse







(EC 1.8.1.4)-


Pelobacter carbinolicus








449.




Unknown




3175366




1




CTG




TGA




1




405




Forward






450.




Unknown




3175380




1




˜CAT




TCA˜




168




413




Reverse






451.




ASPARTATE--AMMONIA LIGASE (EC




3175380




2




˜CAT




TTA˜




422




913




Reverse







6.3.1.1) (ASPARAGINE SYNTHETASE)









ESCHERICHIA COLI


.






452.




aldose 1-epimerase precursor (mutarotase)




3175406




1




˜CAA




TTA˜




6




701




Reverse







(mro) homolog-


Haemophilus influenzae









(strain Rd KW20)






453.




DIHYDROOROTATE DEHYDROGENASE




3175442




1




ATG




TAA




198




551




Forward







(EC 1.3.3.1) (DIHYDROOROTATE







OXIDASE) (DHODEHASE).-


BACILLUS











SUBTILIS


.






454.




SINGLE-STRAND BINDING PROTEIN




3175444




1




˜CAT




TCA˜




12




242




Reverse







(SSB) (HELIX-DESTABILIZING PROTEIN).-









BACILLUS SUBTILIS


.






455.




3-OXOACYL-[ACYL-CARRIER




3175444




2




˜CAT




TTA˜




319




717




Reverse







PROTEIN] REDUCTASE (EC 1.1.1.100)







(3-KETOACYL-ACYL CARRIER PROTEIN







REDUCTASE).


ESCHERICHIA COLI


.






456.




Unknown




3175446




1




˜CAT




TCA˜




432




971




Reverse






457.




Unknown




3175450




1




ATG




TAG




75




584




Forward






458.




LPLB PROTEIN.-


BACILLUS SUBTILIS


.




3175478




1




ATG




TAA




63




278




Forward






459.




Unknown




3175494




1




ATG




TGA




514




759




Forward






460.




Unknown




3175500




1




ATG




TGA




310




537




Forward






461.




30S RIBOSOMAL PROTEIN S9




3175504




1




˜CAT




TTA˜




537




866




Reverse







(BS10).-


BACILLUS STEAROTHER-











MOPHILUS


.






462.




AMINO ACID PERMEASE ROCE.-




3175512




1




ATG




TAA




319




816




Forward









BACILLUS SUBTILIS


.






463.




ARGININE HYDROXIMATE RESISTANCE




3175526




1




˜CAT




CTA˜




1




381




Reverse







PROTEIN.-


BACILLUS SUBTILIS


.






464.




Unknown




3175532




1




ATG




TAA




309




644




Forward






465.




P115 protein-


Mycoplasma hyorhinis


(SGC3)




3175536




1




ATG




TGA




132




899




Forward






466.




Unknown




3175538




1




ATG




TGA




427




507




Forward






467.




Unknown




3175552




1




ATG




TAA




76




381




Forward






468.




Unknown




3175556




1




˜CAT




TTA˜




175




597




Reverse






469.




Unknown




3175564




1




ATG




TAA




3




164




Forward






470.




30S RIBOSOMAL PROTEIN S17




3175566




1




˜CAT




TTA˜




388




648




Reverse







(BS16).-


BACILLUS SUBTILIS


.






471.




Unknown




3175600




1




˜CAT




CTA˜




366




626




Reverse






472.




LICD PROTEIN.-


HAEMOPHILUS






3175612




1




ATG




TGA




134




535




Forward









INFLUENZAE


.






473.




DNA POLYMERASE III, ALPHA CHAIN




3175632




1




TTG




TAA




3




506




Forward







(EC 2.7.7.7).-


BACILLUS SUBTILIS


.






474.




ASPARTATE CARBAMOYL




3175638




1




ATG




TGA




269




526




Forward







TRANSFERASE (EC 2.1.3.2) (ATCASE).-









BACILLUS SUBTILIS


.






475.




SPO0B-ASSOCIATED GTP-BINDING




3175640




1




˜CAT




TCA˜




21




476




Reverse







PROTEIN.-


BACILLUS SUBTILIS


.






476.




URACIL PERMEASE.-


BACILLUS






3175644




1




˜CAT




TTA˜




42




287




Reverse









CALDOLYTICUS


.






477.




Unknown




3175650




1




ATG




TAA




158




676




Forward






478.




DIHYDRODIPICOLINATE SYNTHASE




3175652




1




˜CAT




TTA˜




179




514




Reverse







(EC 4.2.1.52) (DHDPS).-









BACILLUS SUBTILIS


.






479.




Unknown




3175664




1




ATG




TGA




596




979




Forward






480.




DIACYLGLYCEROL KINASE (EC 2.7.1.107)




3175670




1




˜CAT




TTA˜




77




367




Reverse







(DAGK) (DIGLYCERIDE KINASE) (DGK).-









STREPTOCOCCUS MUTANS


.






481.




DnaK protein-


Lactococcus lactis






3175688




1




˜CAA




TTA˜




170




1093




Reverse






482.




serine transporter (sdaC) homolog-




3175698




1




ATG




TGA




289




528




Forward









Haemophilus influenzae


(strain Rd KW20)






483.




phage infection protein precursor-




3175726




1




˜CAG




TTA˜




152




892




Reverse









Lactococcus lactis


subsp.


lactis


(strain C2)






484.




SCU19250 NCBI gi: 625076-




3175744




1




ATG




TGA




3




431




Forward









Streptomyces coelicolor


.






485.




livH protein-


Escherichia coli






3175754




1




ATG




TAA




3




677




Forward






486.




URACIL PERMEASE.-


BACILLUS SUBTILIS


.




3175758




1




˜CAT




TTA˜




49




555




Reverse






487.




Unknown




3175770




1




ATG




TAG




28




171




Forward






488.




Unknown




3175770




2




ATG




TAA




141




311




Forward






489.




Unknown




3175774




1




CTG




TAA




1




408




Forward






490.




METHIONYL-TRNA SYNTHETASE




3175778




1




ATG




TGA




384




767




Forward







(EC 6.1.1.10) (METHIONINE--TRNA







LIGASE) (METRS).-


BACILLUS SUBTILIS


.






491.




STAGE III SPORULATION PROTEIN E.-




3175786




1




˜CAT




TTA˜




8




202




Reverse









BACILLUS SUBTILIS


.






492.




L-FUCOSE ISOMERASE (EC 5.3.1.-).-




3175790




1




ATG




TGA




328




534




Forward









ESCHERICHIA COLI


.






493.




L-FUCOSE ISOMERASE (EC 5.3.1.-).-




3175790




2




ATG




TGA




440




784




Forward









ESCHERICHIA COLI


.






494.




50S RIBOSOMAL PROTEIN L15.-




3175792




1




ATG




TAA




230




670




Forward









BACILLUS STEAROTHERMOPHILUS


.






495.




Unknown




3175794




1




˜CAT




TCA˜




190




381




Reverse






496.




COME OPERON PROTEIN 2.-




3175800




1




˜CAT




TTA˜




182




649




Reverse









BACILLUS SUBTILIS


.






497.




Unknown




3175804




1




CTG




TAA




1




567




Forward






498.




Unknown




3175806




1




˜CAT




TCA˜




283




657




Reverse






499.




Unknown




3175812




1




˜CAT




TCA˜




78




596




Reverse






500.




FOLYLPOLYGLUTAMATE SYNTHASE




3175836




1




ATG




TAG




60




590




Forward







(EC 6.3.2.17) (FOLYLPOLY-GAMMA-







GLUTAMATE SYNTHETASE) (FPGS).-









BACILLUS SUBTILIS


.






501.




Unknown




3175848




1




˜CAT




TCA˜




72




620




Reverse






502.




PHOSPHOSERINE PHOSPHATASE




3175854




1




ATG




TAA




347




841




Forward







(EC 3.1.3.3) (PSP) (O-PHOSPHOSERINE







PHOSPHOHYDROLASE)-









ESCHERICHIA COLI


.






503.




Unknown




3175866




1




˜CAT




CTA˜




175




534




Reverse






504.




AEROBIC GLYCEROL-3-PHOSPHATE




3175882




1




˜CAA




TTA˜




254




520




Reverse







DEHYDROGENASE (EC 1.1.99.5).-









BACILLUS SUBTILIS


.






505.




Unknown




3175896




1




˜CAT




TTA˜




64




636




Reverse






506.




Unknown




3175916




1




ATG




TGA




177




437




Forward






507.




DNA-DIRECTED RNA POLYMERASE




3175948




1




TTG




TAG




2




187




Forward







BETA CHAIN (EC 2.7.7.6)







(TRANSCRIPTASE BETA CHAIN).-









BACILLUS SUBTILIS


.






508.




Unknown




3175960




1




ATG




TGA




534




758




Forward






509.




DNA topoisomerase (ATP-hydrolyzing)




3175984




1




˜CAT




TTA˜




125




604




Reverse







(EC 5.99.1.3) chain B-









Staphylococcus aureus








510.




GLUTAMYL ENDOPEPTIDASE




3175998




1




˜CAT




TCA˜




43




540




Reverse







PRECURSOR (EC 3.4.21.19) (GLUTAMATE







SPECIFIC ENDOPEPTIDASE) (GSE).-









BACILLUS LICHENIFORMIS


.






511.




6-phosphofructokinase (EC




3176002




1




ATG




TAA




662




829




Forward







2.7.1.11)-


Lactococcus lactis








512.




Unknown




3176010




1




CTG




TAA




1




546




Forward






513.




ASPARTYL-TRNA SYNTHETASE




3176030




1




˜CAT




TCA˜




140




625




Reverse







(EC 6.1.1.12) (ASPARTATE--TRNA







LIGASE) (ASPRS).-


THERMUS AQUATICUS









(SUBSP.


THERMOPHILUS


.






514.




Unknown




3176046




1




˜CAT




TTA˜




242




454




Reverse






515.




GLYCOGEN BIOSYNTHESIS PROTEIN




3176048




1




ATG




TAG




411




704




Forward







GLGD.-


BACILLUS SUBTILIS


.






516.




S71704 NCBI gi: 560722-


Legionella






3176050




1




ATG




TGA




205




492




Forward









pneumophila


Philadelphia-1.






517.




PFS PROTEIN (P46).-


ESCHERICHIA COLI


.




3176076




1




˜CAT




TCA˜




40




234




Reverse






518.




Unknown




3176082




1




ATG




TAA




48




491




Forward






519.




SGHRDT NCBI gi: 510450-




3176086




1




˜CAG




TCA˜




23




535




Reverse









Streptomyces griseus


.






520.




Unknown




3176108




1




˜CAT




TTA˜




4




501




Reverse






521.




trsB protein-


Yersinia enterocolitica






3176112




1




ATG




TGA




127




408




Forward






522.




Unknown




3176116




1




ATG




TAA




198




515




Forward






523.




Unknown




3176120




1




˜CAA




TTA˜




295




729




Reverse






524.




SIGNAL PEPTIDASE I (EC 3.4.21.89)




3176124




1




˜CAT




TCA˜




298




570




Reverse







(SPASE I) (LEADER PEPTIDASE I).-









BACILLUS CALDOLYTICUS


.






525.




ligoendopeptidase F-


Lactococcus lactis






3176132




1




˜CAT




TCA˜




51




380




Reverse






526.




Unknown




3176134




1




˜CAT




TTA˜




299




436




Reverse






527.




Unknown




3176136




1




CTG




TAA




1




273




Forward






528.




Unknown




3176152




1




˜CAT




TCA˜




260




688




Reverse






529.




Unknown




3176158




1




ATG




TGA




273




386




Forward






530.




purine-nucleoside phosphorylase




3176172




1




˜CAT




TTA˜




89




331




Reverse







(EC 2.4.2.1)-


Bacillus subtilis


(fragment)






531.




Unknown




3176178




1




˜CAA




CTA˜




101




532




Reverse






532.




PTS SYSTEM, LACTOSE-SPECIFIC IIBC




3176182




1




˜CAT




TCA˜




43




246




Reverse







COMPONENT (EIIBC-LAC) LACTOSE-







PERMEASE IIBC COMPONENT)







PHOSPHOTRANSFERASE ENZYME II, BC







COMPONENT) (EC 2.7.1.69) (EII-LAC).-









LACTOCOCCUS LACTIS


(SUBSP.


LACTIS


).






533.




Unknown




3176184




1




ATG




TGA




61




336




Forward






534.




Unknown




3176188




1




ATG




TAG




42




248




Forward






535.




Unknown




3176208




1




ATG




TAA




41




238




Forward






536.




Unknown




3176216




1




˜CAT




TTA˜




135




335




Reverse






537.




Unknown




3176248




1




˜CAT




TTA˜




141




386




Reverse






538.




Unknown




3176260




1




ATG




TGA




158




343




Forward






539.




Unknown




3176272




1




˜CAT




TTA˜




331




465




Reverse






540.




REGULATORY PROTEIN MTRR.-




3176280




1




ATG




TAA




287




502




Forward









NEISSERIA GONORRHOEAE


.






541.




Unknown




3176288




1




˜CAA




TCA˜




5




520




Reverse






542.




Unknown




3176304




1




˜CAT




TCA˜




41




433




Reverse






543.




Unknown




3176330




1




CTG




TGA




2




298




Forward






544.




Unknown




3176330




2




ATG




TAG




271




381




Forward






545.




6-PHOSPHO-BETA-GLUCOSIDASE




3176338




1




˜CAT




TCA˜




5




130




Reverse







(EC 3.2.1.86).-


ESCHERICHIA COLI


.






546.




Unknown




3176394




1




˜CAT




TTA˜




17




223




Reverse






547.




Unknown




3176398




1




˜CAT




TTA˜




41




310




Reverse






548.




Unknown




3176420




1




˜CAA




TTA˜




382




678




Reverse






549.




possible acid phosphatase




3176446




1




ATG




TGA




113




475




Forward






550.




Unknown




3176480




1




ATG




TAG




151




417




Forward






551.




Unknown




3176542




1




ATG




TAA




129




446




Forward






552.




Unknown




3176560




1




˜CAT




TTA˜




102




374




Reverse














Table 2 shows the correlation between the SEQ ID NO of each DNA sequence of the invention with the SEQ ID NO(S) of polypeptide or polypeptides that its open reading frame(s) codes. For example, the DNA of SEQ ID NO:1 encodes one polypeptide, that of SEQ ID NO:24. Whereas, the DNA of SEQ ID NO:2 encodes two polypeptides, the polypeptides of SEQ ID NO:286 and SEQ ID NO:287.













TABLE 2









DNA




Protein (open reading frame)
























1




263






2




264






3




265






4




266






5




267






6




268






7




269






8




270






9




271






10




272






11




273






12




274






13




275






14




276






15




277






16




278






17




279






18




280






19




281






20




282






21




283






22




284






23




285






24




286, 287






25




288






26




289






27




290






28




291






29




292






30




293






31




294






32




295






33




296






34




297






35




298






36




299






37




300






38




301






39




302






40




303






41




304






42




305, 306






43




307






44




308, 309






45




310






46




311, 312






47




313






48




314






49




315, 316






50




317, 318






51




319, 320






52




321, 322






53




323






54




324






55




325






56




326, 327






57




328






58




329, 330






59




331






60




332






61




333, 334






62




335, 336






63




337






64




338






65




339






66




340






67




341






68




342






69




343






70




344






71




345






72




346






73




347






74




348, 349






75




350






76




351






77




352






78




353, 354






79




355, 356






80




357






81




358






82




359, 360






83




361






84




362






85




363






86




364, 365






87




366






88




367






89




368






90




369






91




370






92




371






93




372






94




373






95




374






96




375






97




376






98




377






99




378, 379






100




380






101




381






102




382, 383






103




384






104




385






105




386






106




387






107




388






108




389






109




390






110




391






111




392






112




393






113




394






114




395






115




396






116




397






117




398






118




399






119




400






120




401






121




402, 403






122




404






123




405






124




406






125




407






126




408






127




409






128




410, 411






129




412






130




413






131




414






132




415






133




416






134




417, 418






135




419






136




420






137




421, 422






138




423






139




424






140




425






141




426






142




427






143




428






144




429






145




430






146




431






147




432






148




433






149




434






150




435






151




436






152




437






153




438






154




439






155




440






156




441






157




442






158




443






159




444






160




445






161




446






162




447






163




448






164




449






165




450, 451






166




452






167




453






168




454, 455






169




456






170




457






171




458






172




459






173




460






174




461






175




462






176




463






177




464






178




465






179




466






180




467






181




468






182




469






183




470






184




471






185




472






186




473






187




474






188




475






189




476






190




477






191




478






192




479






193




480






194




481






195




482






196




483






197




484






198




485






199




486






200




487, 488






201




489






202




490






203




491






204




492, 493






205




494






206




495






207




496






208




497






209




498






210




499






211




500






212




501






213




502






214




503






215




504






216




505






217




506






218




507






219




508






220




509






221




510






222




511






223




512






224




513






225




514






226




515






227




516






228




517






229




518






230




519






231




520






232




521






233




522






234




523






235




524






236




525






237




526






238




527






239




528






240




529






241




530






242




531






243




532






244




533






245




534






246




535






247




536






248




537






249




538






250




539






251




540






252




541






253




542






254




543, 544






255




545






256




546






257




547






258




548






259




549






260




550






261




551






262




552





















EXAMPLES




In order to facilitate understanding of the following example certain frequently occurring methods and/or terms will be described.




“Plasmids” are designated by a lower case p preceded and/or followed by capital letters and/or numbers. The starting plasmids herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available plasmids in accord with published procedures. In addition, equivalent plasmids to those described are known in the art and will be apparent to the ordinarily skilled artisan.




“Digestion” of DNA refers to catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA. The various restriction enzymes used herein are commercially available and their reaction conditions, cofactors and other requirements were used as would be known to the ordinarily skilled artisan. For analytical purposes, typically 1 μg of plasmid or DNA fragment is used with about 2 units of enzyme in about 20 μl of buffer solution. For the purpose of isolating DNA fragments for plasmid construction, typically 5 to 50 μg of DNA are digested with 20 to 250 units of enzyme in a larger volume. Appropriate buffers and substrate amounts for particular restriction enzymes are specified by the manufacturer. Incubation times of about 1 hour at 37° C. are ordinarily used, but may vary in accordance with the supplier's instructions. After digestion the reaction is electrophoresed directly on a polyacrylamide gel to isolate the desired fragment.




Size separation of the cleaved fragments is performed using 8 percent polyacrylamide gel described by Goeddel, D. et al., (1980) Nucleic Acids Res., 8:4057.




“Oligonucleotides” refers to either a single stranded polydeoxynucleotide or two complementary polydeoxynucleotide strands which may be chemically synthesized. Such synthetic oligonucleotides have no 5′ phosphate and thus will not ligate to another oligonucleotide without adding a phosphate with an ATP in the presence of a kinase. A synthetic oligonucleotide will ligate to a fragment that has not been dephosphorylated.




“Ligation” refers to the process of forming phosphodiester bonds between two double stranded nucleic acid fragments (Maniatis, T., et al., supra., p. 146). Unless otherwise provided, ligation may be accomplished using known buffers and conditions with 10 units to T4 DNA ligase (“ligase”) per 0.5 μg of approximately equimolar amounts of the DNA fragments to be ligated.




Example 1




Isolation of DNA Coding for a Virulence Gene in


Streptococcus pneumoniae






As mentioned above each of the DNAs disclosed herein by virtue of the fact that it includes an intact open reading frame is useful to a greater or lesser extent as a screen for identifying antimicrobial compounds. A useful approach for selecting the preferred DNA sequences for screen development is evaluation by insertion-duplication mutagenesis. This system disclosed by Morrison et al.,


J. Bacteriol.


159:870 (1984), is applied as follows.




Briefly, random fragments of


Streptococcus pneumoniae


, strain 0100993 DNA are generated enzymatically (by restriction endonuclease digestion) or physically (by sonication based shearing) followed by gel fractionation and end repair employing T4 DNA polymerase. It is preferred that the DNA fragments so produced are in the range of 200-400 base pairs, a size sufficient to ensure homologous recombination and to insure a representative library in


E. coli.


The fragments are then inserted into appropriately tagged plasmids as described in Hensel et al.,


Science


269: 400-403(1995). Although a number of plasmids can be used for this purpose, a particularly useful plasmid is pJDC9 described by Pearce et al.,


Mol. Microbiol.


9:1037 (1993) which carries the erm gene facilitating erythromycin selection in either


E. coli


or


S. pneumoniae


previously modified by incorporation of DNA sequence tags into one of the polylinker cloning sites. The tagged plasmids are introduced into the appropriate


S. pneumoniae


strain selected, inter alia, on the basis of serotype and virulence in a murine model of pneumococcal pneumonia




It is appreciated that a seventeen amino acid competence factor exists (Havastein et al.,


Proc. Nat'l. Acad. Sci., USA


92:1114-44 (1995)) and may be usefully employed in this protocol to increase the transformation frequencies. A proportion of transformants are analysed to verify homologous integration and as a check on stability. Unwanted levels of reversion are minimized because the duplicated regions will be short (200-400 bp), however if significant reversion rates are encountered they may be modulated by maintaining antibiotic selection during the growth of the transformants in culture and/or during growth in the animal.




The


S. pneumoniae


transformants are pooled for inoculation into mice, eg., Swiss and/or C57B1/6. Preliminary experiments are conducted to establish the optimum complexity of the pools and level of inoculum. A particularly useful model has been described by Veber et al. (


J. Antimicrobiol. Chemother.


32:432 (1993) in which 10


5


cfu inocula sizes are introduced by mouth to the trachea. Strain differences are observed with respect to onset of disease e.g.,3-4 days for Swiss mice and 8-10 days for C57B1/16. Infection yields in the lungs approach 10


8


cfu/lung. IP administration is also possible when genes mediating blood stream infection are evaluated. Following optimization of parameters of the infection model, the mutant bank normally comprising several thousand strains is subjected to the virulence test. Mutants with attenuated virulence are identified by hybridization analysis using the labelled tags from the “input” and “recovered” pools as probes as described in Hensel et al.,


Science


269: 400-403(1995).


S. pneumoniae


DNA is colony blotted or dot blotted, DNA flanking the integrated plasmid is cloned by plasmid rescue in


E. coli


(Morrison et al.,


J. Bacteriol.


159:870 (1984)) and sequenced. Following sequencing, the DNA is compared to the nucleotide sequences given herein and the appropriate ORF is identified and function confirmed for example by knock-out studies. Expression vectors providing the selected protein are prepared and the protein is configured in an appropriate screen for the identification of anti-microbial agents. Alternatively, genomic DNA libraries are probed with restriction fragments flanking the integrated plasmid to isolate full-length cloned virulence genes whose function can be confirmed by “knock-out” studies or other methods, which are then expressed and incorporated into a screen as described above.














SEQUENCE LISTING











The patent contains a lengthy “Sequence Listing” section. A copy of the “Sequence Listing” is available in electronic form from the USPTO






web site (http://seqdata.uspto.gov/sequence.html?DocID=06348328B1). An electronic copy of the “Sequence Listing” will also be available from the






USPTO upon request and payment of the fee set forth in 37 CFR 1.19(b)(3).












Claims
  • 1. An isolated polynucleotide segment comprising: a first polynucleotide sequence, or the full complement of the entire length of the first polynucleotide sequence, wherein the first polynucleotide sequence is selected from the group consisting ofa polynucleotide that encodes a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:302, 310, 313, 390, 417, 418, 447, 472, 490 and 513.
  • 2. A vector comprising the isolated polynucleotide segment of claim 1.
  • 3. An isolated host cell transfected with the isolated polynucleotide segment of claim 1, to express the first polynucleotide sequence.
  • 4. A process for producing a polypeptide, comprising culturing the host cell of claim 3 under conditions sufficient for the production of said polypeptide, wherein said polypeptide comprises SEQ ID NO:302, 310, 313, 390, 417, 418, 447, 472, 490 or 513.
  • 5. An isolated polynucleotide segment, comprising a first polynucleotide sequence, or the full complement of the entire length of the first polynucleotide sequence, wherein the first polynucleotide sequence is selected from the group consisting of(a) a polynucleotide which encodes the same mature polypeptide expressed by a polynucleotide comprising a nucleotide sequence selected from the group consisting of SEQ ID NO:39, 45, 47, 109, 134, 162, 185, 202 and 224 in deposited strain NCIMB 40794; and, (b) a nucleotide sequence identical to the polynucleotide of (a) except that, over the entire length corresponding to the polynucleotide of (a), up to five nucleotides are substituted, deleted or inserted for every 100 nucleotides of the polynucleotide of (a) and, wherein the first polynucleotide sequence detects Streptococcus pneumoniae by hybridization.
  • 6. The isolated polynucleotide segment of claim 5, wherein the first polynucleotide sequence is a nucleotide sequence identical to the polynucleotide of (a) except that, over the entire length corresponding to the polynucleotide of (a), up to three nucleotides are substituted, deleted or inserted for every 100 nucleotides of the polynucleotide of (a).
  • 7. The isolated polynucleotide segment of claim 5, comprising the first polynucleotide sequence, or the full complement of the entire length of the first polynucleotide sequence, wherein the first polynucleotide sequence is the polynucleotide of (a).
  • 8. An isolated polynucleotide segment, comprising a first polynucleotide sequence or the full complement of the entire length of the first polynucleotide sequence, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO: 45, 47, 109, 185, 202 and 224, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65+ C.; and, wherein the first polynucleotide sequence detects Streptococcus pneumoniae by hybridization.
  • 9. The isolated polynucleotide segment of claim 8, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:45, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:45 except that, over the entire length corresponding to SEQ ID NO:45, up to five nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO:45.
  • 10. The isolated polynucleotide segment of claim 9, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:45, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 mnicrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:45 except that, over the entire length corresponding to SEQ ID NO:45, up to three nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO:45.
  • 11. The isolated polynucleotide segment of claim 8, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:47, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:47 except that, over the entire length corresponding to SEQ ID NO:47, up to five nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO:47.
  • 12. The isolated polynucleotide segment of claim 11, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:47, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:47 except that, over the entire length corresponding to SEQ ID NO:47, up to five nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO:47.
  • 13. The isolated polynucleotide segment of claim 8, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:109, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:109 except that, over the entire length corresponding to SEQ ID NO:109, up to five nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO: 109.
  • 14. The isolated polynucleotide segment of claim 13, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:109, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:109 except that, over the entire length corresponding to SEQ ID NO:109, up to five nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO: 109.
  • 15. The isolated polynucleotide segment of claim 13, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:185, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:185 except that, over the entire length corresponding to SEQ ID NO:185, up to five nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO:185.
  • 16. The isolated polynucleotide segment of claim 15, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:185, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:185 except that, over the entire length corresponding to SEQ ID NO:185, up to five nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO:185.
  • 17. The isolated polynucleotide segment of claim 8, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:202, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:202 except that, over the entire length corresponding to SEQ ID NO:202, up to five nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO:202.
  • 18. The isolated polynucleotide segment of claim 17, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:202, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:202 except that, over the entire length corresponding to SEQ ID NO:202, up to five nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO:202.
  • 19. The isolated polynucleotide segment of claim 8, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:224, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:224 except that, over the entire length corresponding to SEQ ID NO:224, up to five nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO:224.
  • 20. The isolated polynucleotide segment of claim 19, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:224, wherein the hybridization conditions include incubation at 42° C in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardts solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:224 except that, over the entire length corresponding to SEQ ID NO:224, up to five nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO:224.
  • 21. An isolated polynucleotide segment comprising a first polynucleotide sequence or the full complement of the entire length of the first polynucleotide sequence, wherein the first polynucleotide sequence hybridizes to the fall complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:39, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:39 except that, over the entire length corresponding to SEQ ID NO:39, up to five nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO:39; and wherein the first polynucleotide sequence detects Streptococcus pneumoniae by hybridization.
  • 22. The isolated polynucleotide segment of claim 21, wherein the first polynucleotide sequence hybridizes to the full complement of a nucleotide sequence selected from the group consisting of SEQ ID NO:39, wherein the hybridization conditions include incubation at 42° C. in a solution comprising: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 micrograms/ml denatured, sheared salmon sperm DNA, followed by washing in 0.1×SSC at 65° C.; and wherein the first polynucleotide sequence is identical to SEQ ID NO:39 except that, over the entire length corresponding to SEQ ID NO:39, up to three nucleotides are substituted, deleted or inserted for every 100 nucleotides of SEQ ID NO:39.
US Referenced Citations (3)
Number Name Date Kind
5753480 Lawlor May 1998 A
5756330 Lawlor May 1998 A
5863777 Lawlor Jan 1999 A
Foreign Referenced Citations (1)
Number Date Country
9610647 Apr 1996 WO
Non-Patent Literature Citations (1)
Entry
Critical Synergy: The Biotechnology Industry and Intellectual Property Protection, Biotechnology Industry Organization, Washington, D.C., 1994, pp. 75 and 100-107.