Process for production of Helicobacter pylori bacterioferritin

Information

  • Patent Grant
  • 7901907
  • Patent Number
    7,901,907
  • Date Filed
    Friday, August 25, 2006
    18 years ago
  • Date Issued
    Tuesday, March 8, 2011
    13 years ago
Abstract
A nucleic acid sequence encoding all or part of an 18-19 kDa Helicobater pylori protein is described to which immunoreactivity is detected in H. pylori negative individuals. A process for the production of a recombinant form of this protein and its use, particularly as a vaccine to provide immunological protection against H. pylori infection are also described.
Description
FIELD OF THE INVENTION

This invention relates to a 18-19 kDa protein or derivative or fragment thereof obtained from Helicobacter pylori, a bacterioferritin defined by sequences in previous patent applications PCT/IE95/00036 and PCT/IE95/00037, a recombinant form of this protein, methods to use this protein as a vaccine to provide immunological protection against if H. pylori infection and methods to use this protein in diagnostic assays relating to H. pylori.


The contents of previous applications PCT/IE95/00036 and PCT/IE95/00037 are incorporated herein by reference.


BACKGROUND OF THE INVENTION


Helicobacter pylori is a widely prevalent organism found on gastric biopsy in approximately 30% of the population less than 40 years old with increasing incidence thereafter. The organism is a causative agent of chronic gastritis in humans (e.g. Marshall & Warren, 1984*; Blaser, 1990*). Epidemiological studies have shown that H. pylori is most commonly found in association with gastritis. Serological investigations have demonstrated that evidence of a current or prior infection can be found in 30-50% of a randomly chosen population of blood donors. No direct causal relationship has been conclusively proven for duodenal ulcer disease. However, the organism is found in 95% of patients with duodenal ulcer. Further, eradication of the organism results in rapid ulcer healing (e.g. Rauws & Tytgat, 1990*). These data provide strong evidence that H. pylori is a dominant factor in the development of duodenal ulcer. Additional evidence for the pathogenic involvement of H. pylori in these conditions has been provided by studies with gnotobiotic piglets (Lambert et al., 1987*) and the fulfilment of Koch's postulates with human volunteers (Marshall et al., 1985*; Morris & Nicholson, 1987*/).


In addition, there is now strong circumstantial evidence implicating H. pylori in the pathogenesis of gastric carcinoma (e.g. Jiang et al., 1987*; Lambert et al, 1986*; Crabtree et al., 1992; 1993; Forman et al., 1990, 1991; Nomura et al, 1991; Parsonnet et al., 1991; Correa et al., 1990). Most recently, the Eurogast Study Group, led by Forman (1993), demonstrated a significant relationship between H. pylori seropositivity and gastric cancer mortality and incidence. Indeed, there is now a convincing body of literature implicating infection with H. pylori in a considerable proportion of upper gastrointestinal morbidity. A number of hypotheses have been suggested for the pathogenic mechanisms of H. pylori induced gastroduodenal disease, including the production of cytotoxins and mechanical disruption of the epithelium (e.g. Blaser, 1992*). Interestingly, however, many infected persons remain asymptomatic despite the persistent presence of the pathogen (Taylor & Blaser, 1991*).


Diagnosis of infections with H. pylori is based mainly on histology and culture from gastric biopsy specimens or on indirect methods based on urease activity. Various serological assays have been developed for the detection of anti-H. pylori antibodies in epidemiological studies in addition to more molecular oriented approaches such as the cloning of H. pylori species-specific antigens for use in, for example, PCR-based serological investigations (e.g. Clayton et al., 1989). However, the use of recombinant species-specific antigens has not yet received widespread use and, consequently, the majority of immunosorbent-based assays currently in use employ various subcellular fractions of H. pylori as a source of antigen. The fractions of proteins used in these assays are frequently heterogeneous in composition as are their methods of preparation. Interestingly, a number of groups have compared the inter-assay sensitivity and specificity of several commercially available ELISA kits manufactured specifically for serological studies. Not surprisingly, considerable inter-assay variation was observed. Caution, therefore, must be exercised before employing a particular preparation of protein for use in such immunosorbent assays, particularly in view of the significant genetic heterogeneity of different strains of H. pylori (e.g. Xia et al., 1994; Owen et al., 1991).


We have studied the prevalence of immuno-reactivity to H. pylori in both infected and uninfected individuals and found that un-infected individuals have a high response to H. pylori both in their B-cell and T-cell systems. In this approach, we use Western blotting to investigate antigen specificity of systemic responses to H. pylori in both healthy and H. pylori-infected individuals and show that the incidence of seropositivity in H. pylori negative individuals is much greater than has previously been demonstrated. Furthermore, we have demonstrated that antibodies to a 25 kDa protein are detectable in the majority of H. pylori negative individuals. These were detected using a technique which we have modified called Enhanced Chemiluminescence. Enhanced Chemiluminescence on Western blot analysis reveals that the majority of uninfected individuals have antibodies which are specific for H. pylori and recognize antigens which are not present on other microorganisms. Of these antigens, the most common one recognized is a 18-19 kDa protein which appears to be specific to H. pylori. Hence, these data suggest that immunization with the 18-19 kDa protein or sub-unit thereof could have the potential to confer protective immunity on individuals who are either uninfected with the organism or individuals in whom the organism has been cleared by anti-bacterial treatment. We have derived N-terminal and internal amino acid sequences from this protein.


SUMMARY OF THE INVENTION

According to the invention, there is provided a nucleic acid sequence encoding all or part of a Helicobacter pylori protein to which immunoreactivity is detected in H. pylori negative individuals.


In one embodiment of the invention, the Helicobacter pylori protein is an 18-19 kDa protein.


Preferably, the 18-19 kDa protein includes the following the N-terminal amino acid sequence:









(SEQ ID NO: 1)








Met-Lys-Thr-Phe-Glu-Ile-Leu-Lys-His-Leu-Gln-Ala-



                5                   10





Asp-Ala-Ile-Val-Leu-Phe-Met-Lys-Val-His-Asn-Phe-


        15                  20





His-Trp-Asn-Val-Lys-Gly-Thr-Asp-Phe-Phe-Asn-Val-


25                  30                  35





His-Lys-Ala-Thr-Glu-Glu-Ile-Tyr-Glu-Glu.


            40                  45







Most preferably, the nucleic acid sequence comprises the following sequence of nucleotides:









(SEQ lID NO: 2)








5′-GATCGTGTTATTTATGAAAGTGCATAACTTCCATTGGAATGTGAAAG



GCACCGATTTTTTCAAT-3′.






The invention also provides a nucleic acid sequence which is complementary to any nucleic acid sequence of the invention.


In one embodiment of the invention, the nucleic acid sequence is genomic DNA, cDNA, synthetic DNA or recombinant DNA.


The invention also provides an oligonucleotide which has a specific binding affinity for a nucleic acid sequence of the invention.


Preferably, the oligonucleotide has one of the following sequences:









(SEQ ID NO: 3)










5′-GAAGGACTTCATATGAAGACATTTG-3′;




or











(SEQ ID NO: 4)










5′-CGTGAATGGATCCTCATGCTGACTTCT-3′.







The invention further provides a vector comprising a recombinant nucleic acid sequence of the invention. Preferably, the vector is an expression vector, most preferably the expression vector pET16b.


The invention also provides a host cell transformed with a vector according to the invention. Preferably, the host cell is one of the following:—

    • E. coli XL1-blue; or
    • E. coli EL21 DE3; or
    • E. coli Novablue DE3.


The invention further provides a process for the production of a recombinant nucleic acid sequence according to the invention comprising culturing a host cell according to the invention and isolating the nucleic acid sequence therefrom.


The invention further provides a recombinant H. pylori protein or a fragment thereof whenever expressed from a vector according to the invention.


The invention also provides a process for the production of a recombinant H. pylori protein or fragment thereof according to the invention comprising culturing a host cell according to the invention and isolating the protein or protein fragment produced therefrom.


The invention further provides a vaccine including a H. pylori protein or a fragment thereof according to the invention.


The vaccine may include a pharmaceutically acceptable carrier.


The vaccine may be combined with a suitable adjuvant such as interleukin 12 or a heat shock protein or both.


The vaccine may include at least one other pharmaceutical product such as an antibiotic and/or an anti-bacterial agent such as bismuth salts. Typically the antibiotic is selected from one or more of metronidazole, amoxycillin, tetracycline, erythromycin, clarithromycin or tinidazole.


The vaccine may be in a form for oral, intranasal, intravenous or intramuscular administration.


The vaccine may include a peptide delivery system.


The vaccine is ideally for the treatment or prophylaxis of Helicobacter pylori infection or Helicobacter pylori associated disease(s)


The invention also provides a vaccine for the treatment or prophylaxis of Helicobacter pylori associated disease comprising an immunogenically effective amount of the Helicobacter pylori protein of the invention, an adjuvant such as Interleukin 12, and an antibiotic.


The vaccine may include an antibacterial agent such as bismuth salts.


The invention also includes the use of interleukin 12 in combination with any other recombinant H. pylori subunit as an adjuvant therapy.


Therefore, in another, aspect, the invention provides a vaccine against H. pylori comprising an immunogenically effective amount of a recombinant Helicobacter protein or a subunit, fragment, derivative, precursor or mutant thereof in combination with interleukin 12 as an adjuvant. Preferably, the Helicobacter is Helicobacter pylori.


In one embodiment of the invention, the vaccine includes an antibiotic and may alternatively or additionally include an antibacterial agent.


The invention also provides a process for the amplification of a nucleic acid sequence according to the invention by a polymerase chain reaction or an equivalent technique.


Preferably, the polymerase chain reaction is effected by using the oligonucleotide pair according to the invention.


The invention also provides a nucleic acid probe comprising a nucleic acid sequence or a fragment thereof according to the invention, or an oligonucleotide according to the invention.


The invention also provides a method for the treatment or prophylaxis of Helicobacter pylori associated disease in a host, comprising administering to the host an immunologically effective amount of one or more of the recombinant Helicobacter proteins of-the invention.


Preferably, the recombinant Helicobacter protein is administered in combination with at least one other pharmaceutical agent.


In a preferred embodiment, the pharmaceutical agent is an antibiotic.


Ideally, the antibiotic is selected from one or more of metronidazole, amoxycillin, tetracycline or erythromycin, clarithromycin or tinidazole.


Typically, the pharmaceutical agent includes an antibacterial agent such as bismuth salts.


In a preferred embodiment of the invention, an adjuvant is administered in combination with the recombinant Helicobacter protein. Preferably, the adjuvant is interleukin 12 or a heat shock protein or both.


The invention also provides the use of one or more Helicobacter proteins according to the invention for the preparation of a medicament for the treatment or prophylaxis of Helicobacter pylori associated disease(s).


The invention further provides monoclonal or polyclonal antibodies or fragments thereof, to the recombinant proteinaceous material of the invention and purified antibodies or serum obtained by immunization of an animal with the vaccine according to the invention.


The invention also provides the use of such serum and antibodies in the treatment or prophylaxis of Helicobacter associated disease(s) and in particular Helicobacter pylori associated disease(s).





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows Western blot analysis of the cloned 18 kDa protein. Two transformed E. coli expression hosts (BL21 DE3 and Novablue DE3) were subjected to SDS-PAGE (12.5% T) followed by Western blotting analysis. The Western blots were probed with serum obtained from children uninfected with H. pylori and developed by enhanced chemiluminescence. Two of the three sera recognized the recombinant 18 kDa protein after induction of expression of the protein with IPTG. In addition, the three sera recognized a number of E. coli proteins.





DETAILED DESCRIPTION OF THE INVENTION

We have generated DNA sequence information identifying the 18-19 kDa protein as a bacterioferritin. We have also generated a recombinant 18 kDa protein and expressed this in E. coli. This recombinant protein was found to be recognized immunologically by antisera from individuals positive for antibody to the 18 kDa helicobacter bacterioferritin. This recombinant protein will form the basis for a putative vaccine for H. pylori. FIG. 1 is a Western Blot analysis of the recombinant 18 kDa protein expressed in E. coli.


Methods Employed


Cloning and expression of the Helicobacter pylori 18 kDa gene.


Deoxyribonucleic acid (DNA) was extracted from Helicobacter pylori as described by Silhavy et al.*(1984).


Oligonucleotides (or “primers”) specific for the 5′ and 3′ termini of the 18 kDa gene were generated. The forward or 5′ oligonucleotide (designated HP18CF) was modified to incorporate an Nde 1 restriction endonuclease site. Additional modifications were made to increase the stability of the binding of the oligonucleotide to its target sequence and to prevent intramolecular secondary structure. The sequence of the HP18CF oligonucleotide is (from 5′ to 3′):









(SEQ ID NO: 3)










GAAGGACTTCATATGAAGACATTTG







The reverse or 3′ oligonucleotide (designated HP18CR) was extensively modified to incorporate a BamH1 restriction endonuclease site and a 5′ tail. The 15 3′ nucleotides of this oligonucleotide correspond to the Helicobacter pylori 18 kDa gene sequence. The sequence of the HP18CR oligonucleotide is (from 5′ to 3′):









(SEQ ID NO: 4)










CGTGAATGGATCCTCATGCTGACTTCT







These oligonucleotides were used in a polymerase chain reaction (PCR) to amplify the Helicobacter pylori 18 kDA gene sequence. The reaction conditions were as follows: Between 50 and 100 ng of Helicobacter pylori DNA was added to 75 pool of each primer, 0.4 mM of each deoxyribonucleotide triphosphate (dNTP), a final concentration of 4 mM MgSO4, 1 fold ‘ThermoPol’ (New England Biolabs) reaction buffer (composition: 10 mM KCl, 10 mM (NH4)2SO4, 20 mM Tris-HCl (pH 8.8 at 25 degrees C.), 2 mM MgSO4, 0.1% Triton X-100), and deionized water was added to bring the reaction volume to 50 ul. The reaction mixture was overlaid with 50 ul paraffin oil and placed in a Perkin-Elmer thermocycle at 90 degrees C. 1 unit vent, DNA polymerase (New England Biolabs) was then added. A ‘touchdown’ PCR procedure was utilized (Don et al. 1989)*. The cycling conditions were as follows: the DNA was denatured at 94 degrees C. for 2.5 minutes. This was followed by 2 cycles of 94 degrees for 30 seconds (denaturation step), 65 degrees for 50 seconds (annealing step), and 72 degrees C. for 20 seconds (extension step). This was followed by 2 cycles of the same conditions, with the exception that the annealing temperature was dropped 5 to 64 degrees C. After 2 cycles at 64 degrees C., the annealing temperature was reduced to 63 degrees C. for a further 2 cycles, and this pattern was followed until the annealing temperature was reduced to 60 degrees C. for 28 cycles.


The reaction products were purified on a 4% low melting point agarose gel (NuSieve GTG; FMC BioProducts). The DNA fragment was excised from the gel and the agarose was digested using β-Agarase 1 (New England Biolabs) and the DNA recovered following precipitation with isopropanol, according to the manufacturer's supplied protocol.


The purified DNA fragment corresponding to the 18 kDa protein coding gene was then digested with the restriction enzymes Nde 1 and BamN1, (Boehringer Mannheim), each of which occurs only once on the amplified fragment. 10 units of each enzyme was added to approximately 3 ug of DNA in a final concentration of 1× the manufacturer's supplied restriction buffer B in a 40 ul reaction volume. The reaction mix was incubated at 37 degrees C. for 3.5 hours.


The expression vector used was pET16b (Novagen). The 1.6 ug of the vector was digested using Nde1 and BamH 1 under the same conditions as described for the amplified fragment. The resulting 5′ phosphate groups were removed using calf intestinal alkaline phosphatase (CIAP: NewEngland Biolabs) according to the manufacturer's instructions. The enzyme was inactivated by incubating the reaction mixture in the presence of 5 mM EDTA at 65 degrees C. for 1 hour followed by a phenol/chloroform/isoamyl alcohol (25:24:1) extraction, followed by a chloroform/isoamyl alcohol (24:1) extraction.


Both the digested fragment and the digested vector were gel purified on a 3% low melting point agarose gel. (NuSieve GTG; FMC BioProducts), and. the agarose was digested using β-Agarase 1 (New England Biolabs), according to the manufacturer's instructions. The DNA fragments were allowed to remain in the resultant reaction mixture without further purification.


The amplified fragment was then ligated to the vector DNA as follows. Approximately 200 ng of vector was ligated to approximately 100 ng of the insert DNA in 1× ligation reaction buffer and 3 units of T4 DNA ligase (Boehringer Mannheim) in a reaction volume of 30 ul at 20 degrees C. for 16 hours.


The products of this reaction were used to transform competent E. coli XL 1-blue cells (Bullock at al. 1987) using a standard CaCl2 transformation procedure (Sambrook, et al., 1989). Transformed XL1-blue cells were selected on LB medium (Sambrook et al., 1989) supplemented with 50 ug per ml ampicillin and grown at 37 degrees C. Suitable colonies were picked and used to inoculate 10 ml LB broth supplemented with 50 ug per ml ampicillin and grown with shaking at 37 degrees C. The plasmids were purified from these cultures using a standard alkaline lysis plasmid preparation procedure (Sambrook, et al., 1989), and an aliquot digested with Nde 1 and KinDZZ7 according to the manufacturers instructions (Boehringer Mannheim) to verify the presence of the insert as compared to a size standard and pET16b without an insert.


Two plasmids shown to have the appropriate insert (designated pET16b-18.1 and pET16b-18.2) were then used to transform the E. coli expression hosts BL21 DE3 (Studier and Moffat, 1986) and Novablue DE3 (Novagen) using a standard CaCl2 transformation procedure (Sambrook, et al., 1989●) supplemented with 50 ug per ml ampicillin (Novablue DE3) or 50 ug per ml ampicillin and 34 ug per ml chloramphenicol (SL21 DE3) and grown at 37 degrees C. Transformed cells were selected by plating on solid LB medium. A colony of each host representing each plasmid isolate was picked after 16 hours incubation and used to inoculate 50 ml LB broth supplemented with antibiotics as described above and grown until the optical density at 600 nM was approximately 0.6. The expression of the 18 kDa protein from the expression vector was then induced by the addition of isopropyl b-D-thiogalactopyranoside (IPTG) to 15 a final concentration of 1 mM and incubation was continued for a further 2.5 hours at 37 degrees C. with shaking. The cells were then harvested by centrifugation at 4000×g for 10 minutes and resuspended in 12 ml of 50 mM Tris-HCl (ph 8.0 at 25 degrees C.) followed by a further centrifugation at 4000×g for 10 minutes.


Sequencing the Purified DNA Sequence


The purified DNA fragment corresponding to the 18 kDa protein was sequenced using forward and reverse universal sequencing primers. The DNA was sequenced in the forward and reverse orientations. Sequencing was performed using an ABI automated sequencer and a Genpak PCR based fluorescent dideoxy chain terminator termini sequencing kit.


The sequence of bases between the termini of the internal PCR primers is:









(SEQ ID NO: 2)








GATCGTGTTATTTATGAAAGTGCATAACTTCCATTGGAATGTGAAAGGCA



CCGATTTTTTCAAT.







Western Blot Analysis of the Cloned Product (18 kDa Protein)


Two transformed E. coli expression hosts (BL21 DE3 and Novablue DE3) were subjected to SDS-PAGE (12.5% T) followed by Western blotting analysis. The Western blots were probed with serum obtained from children uninfected with H. pylori and developed by enhanced chemiluminescence. As illustrated in FIG. 1, two of the three sera recognised the recombinant 18 kDa protein after induction of expression of the protein with IPTG. In addition, the three sera recognised a number of E. coli proteins.


It is understood that the recombinant proteinaceous material of the invention is used as a vaccine against H. pylori infection, and in particular as a therapeutic immunogen for eradication of H. pylori infection.


The vaccine may include the proteinaceous material according to the invention in combination with other components such as a pharmaceutically acceptable carrier, a suitable adjuvant such as interleukin 12 or a heat shock protein, an antibiotic and/or an antibacterial agent such as bismuth salts. The vaccine may be administered in a number of different ways, namely, orally, intranasally, intravenously or intramuscularly.


The invention is not limited to the embodiments hereinbefore described which may be varied in detail.


REFERENCES



  • Marshall, B. J. and Warren, J. R. (1984). Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet 1, 1311-1314.

  • Blaser M. J. (1990). Helicobacter pylori and the pathogenesis of gastroduodenal inflammation. J. Infect. Dis. 161, 626-633.

  • Rauws, E. A. J. and Tytgat, G. N. J. (1990). Eradication of Helicobacter pylori cures duodenal ulcer: Lancet 1, 1233-1235.

  • Lambert, J. R., Borromeo, M., Pinkard, K. J., Turner, H., Chapman, C. B., and Smith, M. L. (1987). Colonisation of gnotobiotic pigs with Campylobacter pylori—an animal model? J. Infect. Dis. 155, 1344.

  • Marshall, B. J. Armstrong, J. A., McGechie, D. B., and Glancy, R. J. (1985). Attempt to fulfil Koch's postulates for pyloric Campylobacter. Med. J Aust. 142, 436-439.

  • Morris, A. and Nicholson, G. (1987). Ingestion of Campylobacter pylori causes gastritis and raises fasting gastric pH. Am. J. Gastroenterol. 82, 192-199.

  • Jiang, S. J., Liu, N. Z. Zhang, D. Z., Shi, Y., Xiao, S. D., Zhang, Z. N., and Liu, D. Y. (1987). Campylobacter-like organisms in chronic gastritis, peptic ulcer and gastric carcinoma. Scand. J. Gastroenterol. 22, 553-558.

  • Lambert, J. R., Dunn, K. A, Eaves, F. R., Korman, M. G., and Hansky, J. (1986). Campylobacter pyloridis in diseases of the human upper gastrointestinal tract. Gastroenterology 90, 1509.

  • Crabtree, J. E., Figura, N., Taylor, J. D., Bugnoli, M., Armellini, D., and Tompkins, D. S. (1992). Expression of 120 kDa protein and cytotoxicity in Helicobacter pylori J. Clin. Pathol 45, 733-734.

  • Crabtree, J. E., Wyatt, J. I., Sobala, G. M., Miller, G., Tompkins, D. S., Primrose, J. N., and Morgan, A. G. (1993). Systemic and mucosal humoral responses to Helicobacter pylori in gastric cancer. Gut 34, 1339-1243.

  • Forman, D., Sitas, F., and Newell, D. G. (1990) Geographic association of Helicobacter pylori antibody prevalence and gastric cancer mortality in rural China. Int. J. Cancer, 46, 608-611.

  • Forman, D., Newell, D. G., Fullerton, F., Yarnell, J. W. G., Stacey, A. R., Wald, N., and Sitas, F. (1991). Association15 between infection with Helicobacter pylori and risk of gastric cancer evidence from. a prospective investigation. BMJ302, 1302-1305.

  • Nomura, A., Stemmermann, G. N., Chyou, P-H., Kato, I., Perez-Perez, G. Z., and Blaser, M. J. (1991). Helicobacter pylori infection and gastric carcinoma amongst Japanese Americans in Hawaii, N. Engl. J. Med. 325, 1132-1136.

  • Parsonnet, J., Friedman, G. D., Vandersteen, D. P., Chang, Y., vogelman, J. K., Orentreich, N., and Sibley, R. K. (1991). Helicobacter pylori infection and the risk of gastric carcinoma. N. Engl. J. Med. 325, 1127-1131.

  • Forman, D. (1993). An international association between Helicobacter pylori infection and gastric cancer. The EUROGAST Study Group. Lancet 341, 1359-1362.

  • Blaser, M. J. (1992). Hypothesis on the pathogenesis and natural history of Helicobacter pylori-induced inflammation. Gastroenterology 102, 720-727.

  • Taylor, D. N. and Blaser, M. J. (1991). Epidemiology of Helicobacter pylori infection. Epidemiol Rev. 13, 42-59.

  • Bullock, w. 0., Fernandez, J. M. and Short, J. M. 1987. A high efficiency plasmid transforming recA Escherichia coli strain with beta-galactosidase selection. Bio Techniques 5:376.

  • Don, R. H., Cox, P. T., Wainwright, B. J., Baker, K. and Mattick, J. S: 1989. ‘Touchdown’ PCR to circumvent spurious priming during gene amplification. Nucleic Acids Res. 19:4008.

  • Sambrook, J., Fritsch, E. F., and Maniatis, T. 1989. Molecular cloning: a laboratory manual. Second edition. Cold Spring Harbour Laboratory Press.

  • Silhavy, T. J., Berman, M. L. and Enquist, L. W. 1984. Experiments with gene fusions. Cold Spring Harbour Laboratory Press.

  • Studier, F. W. and Moffat, B. A. 1986. Use of bacteriophage T7 RNA. polymerase to direct selective high-level expression of cloned genes, J. Mol. Biol. 189:113.


Claims
  • 1. A process for the production of a recombinant nucleic acid comprising culturing a host cell comprising a vector, said vector comprising the nucleic acid sequence of SEQ ID NO:2 encoding a protein comprising an amino acid sequence of SEQ ID NO:1, and isolating the recombinant nucleic acid therefrom.
  • 2. A process for the production of a recombinant H. pylori protein comprising culturing a host cell comprising a vector, said vector comprising a nucleic acid sequence encoding a protein comprising SEQ ID NO:1, wherein said nucleic acid sequence comprises SEQ ID NO:2, and isolating the protein produced therefrom.
  • 3. A process for producing a recombinant Helicobacter pylori protein, said process comprising of expressing a Helicobacter pylori protein comprising the amino acid sequence of SEQ ID NO:1 from a vector comprising a recombinant nucleic acid sequence comprising SEQ ID NO:2.
  • 4. A process for producing a recombinant Helicobacter pylori protein, said process comprising: amplifying a nucleic acid encoding a Helicobacter pylori protein comprising a molecular weight of 18-19 kDa from Helicobacter pylori DNA using a primer comprising SEQ ID NO: 3 and a primer comprising SEQ ID NO: 4;inserting said nucleic acid into a vector; andexpressing said Helicobacter pylori protein from said vector.
  • 5. The process of claim 4 wherein said nucleic acid comprises SEQ ID NO: 2.
Priority Claims (2)
Number Date Country Kind
960004 Jan 1996 IE national
960019 Jan 1996 IE national
CROSS REFERENCE TO RELATED APPLICATIONS

This is a continuation of application Ser. No. 11/326,200, filed Jan. 5, 2006, which is a continuation of application Ser. No. 11/112,735, filed Apr. 22, 2005, which is a continuation of application Ser. No. 10/928,353, filed Aug. 27, 2004, which is a continuation of application Ser. No. 10/699,624, filed Oct. 31, 2003, which is a continuation of application Ser. No. 10/342,431, filed Jan. 13, 2003, which is a continuation of application Ser. No. 10/140,758, filed May 7, 2002, which is a continuation of application Ser. No. 09/101,158, filed Jul. 30, 1998, which is the U.S. National Stage of International Application PCT/IE97/00001, filed Jan. 3, 1997, which claims priority to Irish patent applications IE96004, filed Jan. 4, 1996, and IE960019, filed Jan. 12, 1996, each of which is incorporated herein by reference in its entirety.

US Referenced Citations (79)
Number Name Date Kind
4879213 Fox et al. Nov 1989 A
4882271 Evans et al. Nov 1989 A
4997823 Herschler Mar 1991 A
5190961 Hasegawa Mar 1993 A
5214053 Nakazawa May 1993 A
5262156 Alemohammad Nov 1993 A
5292658 Cormier et al. Mar 1994 A
5306492 Porro Apr 1994 A
5354854 Bourque et al. Oct 1994 A
5403924 Cover et al. Apr 1995 A
5434253 Thompson et al. Jul 1995 A
5527669 Resnick Jun 1996 A
5527678 Blaser et al. Jun 1996 A
5538729 Czinn et al. Jul 1996 A
5541291 Keene Jul 1996 A
5554372 Hunter Sep 1996 A
5567594 Calenoff Oct 1996 A
5571515 Scott et al. Nov 1996 A
5721349 Cover et al. Feb 1998 A
5723127 Scott et al. Mar 1998 A
5733740 Cover et al. Mar 1998 A
5814455 Pronovost et al. Sep 1998 A
5817289 Klaveness et al. Oct 1998 A
5854221 Cao et al. Dec 1998 A
5858352 Pace et al. Jan 1999 A
5861518 Dekker Jan 1999 A
5866375 Figura et al. Feb 1999 A
5869066 Pace et al. Feb 1999 A
5871749 Doidge et al. Feb 1999 A
5876943 Cover et al. Mar 1999 A
5891890 Nishino et al. Apr 1999 A
5895653 Eibl et al. Apr 1999 A
5897475 Pace et al. Apr 1999 A
5900372 Figura et al. May 1999 A
5900410 Hartmann May 1999 A
5925667 Nishino et al. Jul 1999 A
5985243 Ghiara Nov 1999 A
6001880 Nishino et al. Dec 1999 A
6005090 Doidge et al. Dec 1999 A
6013463 Cover et al. Jan 2000 A
6019982 Clements et al. Feb 2000 A
6054132 Cover et al. Apr 2000 A
6077830 Vertesy et al. Jun 2000 A
6090611 Covacci et al. Jul 2000 A
6124271 Iversen et al. Sep 2000 A
6129923 Doidge et al. Oct 2000 A
6153390 Cover et al. Nov 2000 A
6160119 Senn-Bilfinger Dec 2000 A
6231860 Fanget et al. May 2001 B1
6248329 Chandrashekar et al. Jun 2001 B1
6265415 Amin et al. Jul 2001 B1
6534064 O'Hagan Mar 2003 B1
6552047 Garvey Apr 2003 B2
6559294 Griffais et al. May 2003 B1
6649629 Bandarage Nov 2003 B2
6761894 Horwitz Jul 2004 B1
6841155 Del Giudice et al. Jan 2005 B1
6902903 Quan Jun 2005 B1
6914131 Scarlato et al. Jul 2005 B1
7038012 Grandi May 2006 B1
20010010821 Kelleher et al. Aug 2001 A1
20020064569 Hoffman May 2002 A1
20020065296 Dumas May 2002 A1
20020115078 Kleanthous Aug 2002 A1
20030105091 Riedl Jun 2003 A1
20040005667 Ratti et al. Jan 2004 A1
20040029129 Wang Feb 2004 A1
20040033234 Berinstein Feb 2004 A1
20040052799 Smith et al. Mar 2004 A1
20050074450 Giuliani et al. Apr 2005 A1
20050191316 Fraser et al. Sep 2005 A1
20060292175 Polo et al. Dec 2006 A1
20070104731 Kelleher et al. May 2007 A1
20070110765 Bryne May 2007 A1
20070243204 Covacci et al. Oct 2007 A1
20090100536 Adams Apr 2009 A1
20090103536 Basso Apr 2009 A1
20090118268 Riedl May 2009 A1
20090158452 Johnson Jun 2009 A1
Foreign Referenced Citations (85)
Number Date Country
2117490 Feb 1992 CA
2308606 May 1999 CA
2307846 Jun 1999 CA
2425303 May 2002 CA
0 329 570 Aug 1989 EP
0 367 644 May 1990 EP
0 372 501 Jun 1990 EP
0 378 881 Jul 1990 EP
0 427 347 May 1991 EP
0 471 177 Feb 1992 EP
0 477 508 Apr 1992 EP
0 761 231 Mar 1997 EP
1767214 Mar 2007 EP
2 669 929 Jun 1992 FR
2 220 221 Jan 1990 GB
183034 Mar 1997 IN
183035 Apr 1997 IN
9601063 Mar 2005 IN
240197 Apr 2010 IN
WO 8705943 Oct 1987 WO
WO 8908843 Sep 1989 WO
WO 9004030 Apr 1990 WO
WO 9006696 Jun 1990 WO
WO 9014837 Dec 1990 WO
WO 9109049 Jun 1991 WO
WO 9316723 Sep 1993 WO
WO 9317712 Sep 1993 WO
WO 9318150 Sep 1993 WO
WO 9320843 Oct 1993 WO
WO 9409823 May 1994 WO
WO 9421284 Sep 1994 WO
WO 9421292 Sep 1994 WO
WO 9426901 Nov 1994 WO
WO 9426901 Nov 1994 WO
WO 9503824 Feb 1995 WO
WO 9514093 May 1995 WO
WO 9517210 Jun 1995 WO
WO 9601272 Jan 1996 WO
WO 9601273 Jan 1996 WO
WO 9602555 Feb 1996 WO
WO 9629412 Sep 1996 WO
WO 9640893 Dec 1996 WO
WO 9701640 Jan 1997 WO
WO 9723238 Jul 1997 WO
WO 9725429 Jul 1997 WO
WO 9737044 Oct 1997 WO
WO 9804702 Feb 1998 WO
WO 9816247 Apr 1998 WO
WO 9818810 May 1998 WO
WO 9820734 May 1998 WO
WO 9827432 Jun 1998 WO
WO 9837919 Sep 1998 WO
WO 9840100 Sep 1998 WO
WO 9842721 Oct 1998 WO
WO 9843478 Oct 1998 WO
WO 9852581 Nov 1998 WO
WO 9854171 Dec 1998 WO
WO 9855495 Dec 1998 WO
WO 9856412 Dec 1998 WO
WO 9857659 Dec 1998 WO
WO 9858668 Dec 1998 WO
WO 9900380 Jan 1999 WO
WO 9911241 Mar 1999 WO
WO 9928322 Jun 1999 WO
WO 9928475 Jun 1999 WO
WO 9936544 Jul 1999 WO
WO 9944636 Sep 1999 WO
WO 9952549 Oct 1999 WO
WO 9953310 Oct 1999 WO
WO 9957278 Nov 1999 WO
WO 9957280 Nov 1999 WO
WO 0007621 Feb 2000 WO
WO 0023105 Apr 2000 WO
WO 0027994 May 2000 WO
WO 0037494 Jun 2000 WO
WO 0056360 Sep 2000 WO
WO 0056365 Sep 2000 WO
WO 0061761 Oct 2000 WO
WO 0062800 Oct 2000 WO
WO 0121152 Mar 2001 WO
WO 0121207 Mar 2001 WO
WO 0152885 Jul 2001 WO
WO 0170955 Sep 2001 WO
WO 0245741 Jun 2002 WO
WO 02077813 Oct 2002 WO
Related Publications (1)
Number Date Country
20070110765 A1 May 2007 US
Continuations (7)
Number Date Country
Parent 11326200 Jan 2006 US
Child 11510509 US
Parent 11112735 Apr 2005 US
Child 11326200 US
Parent 10928353 Aug 2004 US
Child 11112735 US
Parent 10699624 Oct 2003 US
Child 10928353 US
Parent 10342431 Jan 2003 US
Child 10699624 US
Parent 10140758 May 2002 US
Child 10342431 US
Parent 09101158 US
Child 10140758 US