US20040096973A1 - Environmentally regulated genes of Streptococcus suis - Google Patents

Environmentally regulated genes of Streptococcus suis Download PDF

Info

Publication number
US20040096973A1
US20040096973A1 US10/632,117 US63211703A US2004096973A1 US 20040096973 A1 US20040096973 A1 US 20040096973A1 US 63211703 A US63211703 A US 63211703A US 2004096973 A1 US2004096973 A1 US 2004096973A1
Authority
US
United States
Prior art keywords
suis
streptococcus
fbps
gene
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US10/632,117
Other versions
US7927605B2 (en
Inventor
Hilda Smith
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Stichting Dienst Landbouwkundig Onderzoek DLO
Original Assignee
ID Lelystad Instituut voor Dierhouderij en Diergezondheid BV
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by ID Lelystad Instituut voor Dierhouderij en Diergezondheid BV filed Critical ID Lelystad Instituut voor Dierhouderij en Diergezondheid BV
Assigned to ID-LELYSTAD, INSTITUUT VOOR DIERHOURDERIJ EN DIERGEZONDHEID B.V. reassignment ID-LELYSTAD, INSTITUUT VOOR DIERHOURDERIJ EN DIERGEZONDHEID B.V. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SMITH, HILDA ELIZABETH
Publication of US20040096973A1 publication Critical patent/US20040096973A1/en
Assigned to ID-LELYSTAD, INSTITUUT VOOR DIERHOUDERIJ EN DIERGEZONDHEID B.V. reassignment ID-LELYSTAD, INSTITUUT VOOR DIERHOUDERIJ EN DIERGEZONDHEID B.V. CORRECTIVE ASSIGNMENT TO CORRECT ASSIGNEE ADDRESS, PREVIOUSLY RECORDED ON REEL 014818 FRAME 0220. Assignors: SMITH, HILDA ELIZABETH
Priority to US11/982,192 priority Critical patent/US8324354B2/en
Assigned to STICHTING DIENST LANDBOUWKUNDIG ONDERZOEK reassignment STICHTING DIENST LANDBOUWKUNDIG ONDERZOEK ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ID-LELYSTAD, INSTITUUT VOOR DIERHOUDERIJ EN DIERGEZONDHEID B.V.
Application granted granted Critical
Publication of US7927605B2 publication Critical patent/US7927605B2/en
Adjusted expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/315Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Streptococcus (G), e.g. Enterococci
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies

Definitions

  • the invention relates to the field of the diagnosis of and vaccination against Streptococcal infections, and to the detection of virulence markers of Streptococci.
  • Streptococcus species of which there are a large variety of that cause infections in domestic animals and man, are often grouped according to Lancefield's groups. Typing according to Lancefield occurs on the basis of serological determinants or antigens that are, among others, present in the capsule of the bacterium and only allows for an approximate determination. Often, bacteria from a different group show cross-reactivity with each other, while other Streptococci cannot be assigned a group-determinant at all. Within groups, further differentiation is often possible on the basis of serotyping. These serotypes further contribute to the large antigenic variability of Streptococci, a fact that creates an array of difficulties within diagnosis of and vaccination against Streptococcal infections.
  • Lancefield group C infections such as those with S. equi, S. zooepidemicus, S. dysgalactiae , and others are mainly seen associated with horses, cattle and pigs, but can also cross the species barrier to humans.
  • Lancefield group D S. bovis ) infections are found with all mammals and some birds, sometimes resulting in endocarditis or septicemia.
  • S. suis is found and is an important cause of meningitis, septicemia, arthritis and sudden death in young pigs. Incidentally, S. suis can also cause meningitis in man.
  • Ungrouped Streptococcus species such as S. mutans , causes carries with humans.
  • S. uberis causes mastitis with cattle
  • S. pneumonia causes major infections in humans
  • Enterococcus faecilalis and E. faecium further contribute to the large group of Streptococci.
  • Streptococcus pneumoniae (the pneumococcus) is a human pathogen causing invasive diseases, such as pneumonia, bacteremia, and meningitis.
  • Streptococcus suis is an important cause of meningitis, septicemia, arthritis and sudden death in young pigs. It can also cause meningitis in man. Attempts to control the disease are hampered by the lack of sufficient knowledge about the pathogenesis of the disease and the lack of effective vaccines and sensitive diagnostic methods.
  • S. suis serotype 2 is the most frequently isolated serotype.
  • S. suis serotype 2 pathogenic, weak-pathogenic and non-pathogenic strains can be found.
  • the pathogenic strains cause severe clinical signs of disease in pigs and large numbers of bacteria can be re-isolated from the central nervous system (CNS) and the joints after experimental infection.
  • the weak-pathogenic strains cause only mild clinical signs of disease and infrequently are bacteria re-isolated from the CNS and the joints after experimental infection.
  • the non-pathogenic strains are completely avirulent in young pigs after experimental infection.
  • MRP muramidase-related protein
  • EF 110-kDa extracellular factor
  • the invention discloses a method for modulating virulence of a Streptococcus comprising modifying a genomic fragment of Streptococcus.
  • the genomic fragment comprises at least a functional part of a fragment identifiable by hybridization in Streptococcus suis to a nucleic acid or fragment thereof as shown in SEQ ID NOS: 8 through 45 and obtaining a clone including the modified genomic fragment.
  • the genomic fragment comprises at least a functional part of a gene, the expression of which can be environmentally regulated in S. suis by iron-restricted conditions.
  • the genomic fragment comprises at least a functional part of a gene which is expressed in a wild-type S. suis infected pig (in vivo).
  • selection under iron-restricted conditions is combined with selection in vivo.
  • the gene encodes a fibronectin/fibrinogen-binding protein. The method disclosed herein is useful for modulating virulence of Streptococcus suis and comprises functionally deleting the expression of at least the functional part of the gene by Streptococcus.
  • the phrase “functionally deleting” as used herein refers to any technique known in the art (such as allowing for a deletion, insertion, mutation or the occurrence of a frame-shift in the open-reading frame of the respective gene) that is instrumental in hampering or inhibiting the expression of a gene-product (be it mRNA and/or protein) of the gene.
  • the invention discloses a clone of a Streptococcus obtained or obtainable by a method according to the invention.
  • the invention describes the identification of environmentally regulated genes of Streptococcus suis by iron-restricted conditions and by experimental infection of piglets. Eighteen unique iron-restricted induced (iri) genes and 22 unique in vivo selected (ivs) genes of S. suis were found. None of the ivs genes was exclusively expressed in vivo. Four iri genes were substantially identical to four ivs genes selected in piglets. Two ivs genes were similar to genes for putative virulence factors.
  • One of these ivs genes was substantially identical to the epf gene of virulent S. suis serotype 2 strains and the other ivs gene showed homology to a gene encoding a fibronectin-binding protein of Streptococcus gordonii.
  • the invention discloses a study of the characteristics of fibronectin- and fibrinogen-binding protein of Streptococcus suis (FBPS) and its gene as identified herein.
  • FBPS Streptococcus suis
  • the gene encoding FBPS was identified using an in vivo selection system in pigs as described herein, thus, showing an important role of the protein in vivo. This finding was supported by the observation that isogenic FBPS mutants, herein also disclosed, of S.
  • fibronectin-binding proteins are large cell surface proteins with predicted sizes of 60-100 kDa and have similar structural organizations.
  • the proteins contain an N-terminal signal sequence as well as the cell wall signaling sequence (LPXTGE) (SEQ ID NO: 1).
  • LXTGE cell wall signaling sequence
  • the Fn-binding sites include 30-42 amino acid long motifs, repeated 3-4 times.
  • the first fibronectin- and fibrinogen-binding protein of S. suis is disclosed herein.
  • FBPS The gene encoding FBPS was cloned and sequenced and FBPS was purified. Binding of FBPS to human fn and fgn was shown. FBPS was shown to be involved in the colonization of the organs specific for an S. suis infection in piglets, but not in the colonization of S. suis on the tonsils of piglets.
  • the mutant strain 10 ⁇ FBPS may be used in the vaccine, which mutant is possibly further attenuated by deleting one or more virulence factors as described herein.
  • this vaccine is based on purified FBPS protein or an antigenic part thereof with a suitable adjuvant.
  • FBPS fibronectin-/fibrinogen-binding protein
  • FBPS mutant unable to produce FBPS was constructed and the virulence of the wild-type and mutant strains was compared in a competitive infection model in young piglets. Organ cultures showed that FBPS was not required for colonization of the tonsils, but that FBPS played a role in the colonization of the specific organs involved in an S. suis infection. Therefore, the FBPS mutant was considered as an attenuated mutant which is useful in a vaccine.
  • a vaccine is used that mainly includes the FBPS protein or at least of an antigenic part thereof, such that an FBPS-specific antibody or T-cell response in pigs is developed after vaccination with the FBPS or antigenic part thereof.
  • the invention also discloses a method for assaying virulence of a Streptococcus comprising assaying a genomic fragment of Streptococcus, wherein the genomic fragment comprises at least a functional part of a fragment identifiable by hybridization in Streptococcus suis to a nucleic acid or fragment thereof as described herein.
  • the invention also discloses a vector comprising a nucleic acid according to the invention and a host cell comprising a nucleic acid or a vector according to the invention.
  • a host cell comprises an easily modifiable organism such as E. coli .
  • other host cells such as a recombinant Streptococcus comprising a vector. or nucleic acid according to the invention are also disclosed herein.
  • the invention additionally discloses a vaccine comprising a nucleic acid, a vector or a host cell according to the invention, and use of such a vaccine in the prevention and/or treatment of Streptococcal infections.
  • a protein or fragment thereof encoded by a nucleic acid according to the invention such as a protein encoded by a nucleic acid or fragment thereof disclosed herein or functional, i.e., antigenic fragment thereof.
  • the invention also discloses an antibody directed against a protein or fragment thereof according to the invention and an antigen reactive with such an antibody, for example comprising a protein or fragment.
  • Such a protein or fragment thereof need not be obtained by recombinant means. Synthesis of peptides, according to their amino acid sequence, is as well equally possible.
  • Such antigens and antibodies as described herein can be used in a diagnostic test comprising an antibody according to the invention, or within a vaccine or diagnostic test comprising an antigen according to the invention.
  • Such vaccines and diagnostic tests can be used in the field of the diagnosis of and vaccination against Streptococcal infections and for the detection of virulence markers of Streptococci.
  • FIG. 1 is a schematic presentation of the procedure used to clone the fbps gene of S. suis serotype 2 and the construction of an insertional knock-out mutant in S. suis serotype 2.
  • a 5 kb EcoRI fragment was cloned in pGEM7Zf(+), yielding pFBPS7-46.
  • pFBPS7-47 the 382 bp SalI-SalI fragment of pFBPS7-46 was replaced by 1.2 kb spectinomycin-resistance gene, after the vector was made blunt to obtain an insertional knock-out of fbps.
  • Ivs-31 in vivo selected gene 31.
  • FIG. 2 shows purity and immunogenicity of FBPS purified under native conditions.
  • Convalescent serum raised against S. suis strain 10 was used to test immunogenicity of FPBS present in 4:1 of E.
  • coli M15 pQE-30-pREP4-FBPS lysate and 0.5 ⁇ g of purified FBPS (Panel C, lanes 1 and 2). Arrowhead, 64 kDa FPBS; Mw, molecular weight marker.
  • FIG. 3 depicts the binding studies with purified FBPS.
  • Panels A and B were probed with 5 ⁇ g/ml of fn (A) or fgn (B).
  • Lanes 1 contain 500 ng of purified FBPS
  • lanes 2 contain 500 ng of BSA.
  • Panels C and D lanes 3 and 4 contain 500 ng of purified FBPS.
  • Lanes 3 were probed with 20 ⁇ g/ml of fn (C) or fgn (D)
  • lanes 4 were incubated with conjugate without fn or fgn.
  • Panels E and F were probed with 20 ⁇ g/ml of fn (E) or fgn (F).
  • Lanes 5 contain 1.8 ⁇ g of purified FBPS digested with enterokinase, lanes 6 contain 500 ng of purified FBPS.
  • the closed arrowhead indicates 64 kDa FBPS; the open arrowhead indicates approximately 55 kDa FBPS without 6 ⁇ HIS.
  • FIG. 4 shows the distribution of fbps among various S. suis serotypes. 1 ⁇ g of chromosomal DNA was spotted onto nitrocellulose membrane and hybridized with a 32 P-labelled fbps probe. Serotypes were spotted as indicated. S10: S. suis serotype 2, MRP + EF + ; T15 : S. suis serotype 2 MRP ⁇ EF ⁇ ; S17 :S. suis serotype 2 MRP + EF*.
  • FIG. 5 illustrates the efficiency of colonization of wild-type and mutant bacteria on various organs of infected pigs.
  • Panel A depicts colonization of the wild-type strain 10 and the mutant strain 10 ⁇ FBPS of the tonsils.
  • a closed diamond symbol is tonsil pig no. 4664; ⁇ tonsil pig no. 4665; ⁇ tonsil pig no 4666; • tonsil pig no. 4668.
  • Panel B depicts colonization of the specific organs. Open and closed diamond symbols are pus from joints pig no. 4664; ⁇ pus from joint pig no. 4666; • CNS pig no. 4668.
  • Each dot represents the numbers of wild-type or mutant bacteria isolated from one particular organ, from one piglet.
  • Streptococcus suis is an important cause of meningitis, septicemia, arthritis and sudden death in young pigs (Clifton-Haclley, 1983; Vecht et al., 1985). It can also cause meningitis in man (Arends and Zanen, 1988). Attempts to control the disease are still hampered by the lack of sufficient knowledge about the pathogenesis of the disease, the lack of effective vaccines and sensitive diagnostic methods. To meet these shortages, it is necessary to identify the genes that are involved in the pathogenic process. So far, only a limited number of S.
  • the present invention identifies virulence genes of S. suis by selecting environmentally regulated genes by experimental infections of piglets and by the use of iron-restricted conditions in vitro. For this purpose, chromosomal DNA fragments of S. suis were cloned in a plasmid in front of a promoterless erythromycin-resistance gene. Subsequently, the library was used for the selection of bacteria in which erythromycin resistance was induced under iron-restricted conditions. In addition, erythromycin-resistant bacteria were selected after infection of piglets with the library and treatment of the piglets with erythromycin. Pigs were used instead of mice for these experiments since it was recently shown that virulence of S.
  • S. suis strains were grown in Todd-Hewitt broth (Oxoid), and plated on Columbia agar (Oxoid) containing 6% (v/v) horse blood.
  • S. suis cells were plated on agar plates containing Todd-Hewitt medium, 5% (w/v) yeast extract and 75 ⁇ M deferoxamine mesylate (Sigma). Control plates were supplemented with 38 ⁇ M FeSO 4 .7H 2 O (Sigma).
  • antibiotics were added at the following concentrations: 100 ⁇ g spectinomycin ml ⁇ 1 and 1 ⁇ g erythromycin ml ⁇ 1 .
  • E. coli strains were grown in Luria broth (Miller, 1972) and plated on Luria broth containing 1.5% (w/v) agar. If required, 50 ⁇ g ampicillin ml ⁇ 1 or 50 ⁇ g spectinomycin ml ⁇ 1 was added.
  • the IVS selection vector used in this study comprises a spectinomycin-resistance gene, a promoterless erythromycin-resistance gene and the origin of replication of the plasmid pWVO1 (Van der Vossen et al., 1987).
  • the spectinomycin-resistance gene was amplified from pKUN19-spc (Konings et al., 1987; Smith et al., 1995).
  • the primers 5′-TGCATGCATGGATCCATCGA TTTTCGTTCG-3′ (SEQ ID NO: 2) and 5′-CGAGCTCGGTACCTGATTACCAATTAGAAT-3′ (SEQ ID NO: 3), which contained NsiI and SacI restriction sites at their respective 5′-ends were used.
  • the obtained PCR product was digested with NsiI and SacI and ligated into pGKV210 (Van der Vossen et al., 1987) that had been digested with SacI (partially) and NsiI.
  • the resulting plasmid was designated pGKV210-spc.
  • pE194 (Horinouchi and Weisblum, 1982) was used as a template for the amplification of a promoterless erythromycin-resistance gene.
  • the primers 5′-GGGTCGACCCTATAACCAAATTAAAGAGGG-3′ (SEQ ID NO: 4) and 5′-CCCAAGCTTGGGCAGTTTATGCATCCCTTAAC-3′ (SEQ ID NO: 5) were used in a PCR reaction. These primers contained SalI and HindIII restriction sites at their respective 5′-ends. The amplified fragment was digested with SalI and HindIII and the fragment was ligated into pGKV210-spc that had been digested with SalI and HindIII.
  • the resulting plasmid was designated pIVS-E.
  • the promoter region of the mrp gene was inserted into pIVS-E 5′ to the promoterless erythromycin-resistance gene.
  • the promoter region of the mrp gene was amplified by PCR from pMRP11 (Smith et al., 1992) using the primers 5′-CCCAAGCTTGGGAATTCATAATGTTTTTTTGAGG-3′ (SEQ ID NO: 6) and 5′-GCGTCGACATCTACGCATAAAAAATCCCCC-3′ (SEQ ID NO: 7). These primers contained EcoRI and SalI sites at their respective 5′-ends. Amplified DNA was digested with EcoRI and SalI and the resulting fragment was ligated into EcoRI and SalI-digested pIVS-E.
  • PCR reaction mixtures (50 ⁇ l) contained 10 mM Tris-HCl, pH 8.3, 1.5 mM MgCl 2 , 50 mM KCl, 0.2 mM of each of the four deoxynucleotide triphosphates, 1 ⁇ M of each of the primers and 1 U of AmpliTaq Gold DNA polymerase (Perkin Elmer Applied Biosystems). DNA amplification was carried out in a Perkin Elmer 9600 thermal cycler and the program included an incubation for ten minutes at 95° C. and 30 cycles of one minute at 95° C., two minutes at 56° C. and two minutes at 72° C.
  • SPF pathogen-free piglets
  • erythromycin stearate Abbott, 20 or 40 mg body weight kg ⁇ 1
  • Erythrocin 200 Sanofi Sante, 20 or 40 mg body weight kg ⁇ 1
  • Blood samples were collected 3 hours, 6 hours or 24 hours after the administration of the antibiotics to determine erythromycin levels.
  • Gnotobiotic Great Yorkshire and Dutch Landrace crossed piglets were obtained from sows by cesarian section. The surgery was performed in sterile flexible film isolators. The piglets were allotted to groups, each having 4 piglets, and were housed in sterile stainless steel incubators. Housing conditions and feeding regimens were as described (Vecht et al., 1989; Vecht et al., 1992). One-week-old piglets were inoculated intravenously with S. suis strain 10 (pIVS-E), 10 (pIVS-PE) or 10 (pIVS-RE) as described (Vecht et al., 1989; Vecht et al., 1992, Table 3).
  • S. suis strain 10 pIVS-E
  • pIVS-PE 10
  • pIVS-RE 10
  • the pigs were injected intramuscularly with erythromycin for the first time and thereafter received erythromycin twice a day: once intramuscularly (Erythrocin, 40 mg body weight kg ⁇ 1 ) and once orally (erythromycin stearate, 40 mg body weight kg ⁇ 1 ).
  • Piglets were monitored twice a day for clinical signs of disease, such as fever, nervous signs and lameness. Blood samples were collected three times a week from each pig. Leukocyte concentrations were determined using a conducting counter (Contraves A. G., Switzerland). To monitor infection with S. suis and to check for absence of contaminants, swabs of the nasopharynx and of feces were collected daily. The swabs were directly plated onto Columbia agar containing 6% (v/v) horse blood. After the piglets were killed, they were examined for gross pathological changes. Tissue specimens were collected from the central nervous system, serosae, joints, lungs, heart and tonsils. The tissues were homogenized in the presence of Todd-Hewitt medium using an Ultra-Turrax tissuemizer (Omni International) and frozen at ⁇ 80° C. in the presence of 15% (v/v) glycerol.
  • Omni International Ultra-Turrax tissuemizer
  • the plasmid pIVS-E was constructed to allow introduction of S. suis DNA fragments into a number of unique restriction sites in front of a promoterless erythromycin-resistance resistance gene.
  • the plasmid carries the origin of replication of pWVO1, which functions in E. coli and in S. suis (Smith et al., 1995).
  • S. suis strain 10 cells containing pIVS-E were sensitive to 1 ⁇ g erythromycin ml ⁇ 1 on agar plates.
  • pIVS-PE the promoter of the mrp gene of S. suis (Smith et al., 1992), which is highly expressed in vivo as well as in vitro, drives expression of the erythromycin-resistance gene.
  • S. suis strain 10 cells containing pIVS-PE were resistant to high concentrations of erythromycin (>256 ⁇ g erythromycin ml ⁇ 1 ) on agar plates.
  • a S. suis DNA library in pIVS-E (pIVS-RE) was constructed and 30,000 individual S. suis clones or mutants were obtained. As determined by analysis of 24 randomly selected transformants, more than 80% of these clones or mutants contained an insert (results not shown). Moreover, 2% of the clones were resistant to 1 ⁇ g erythromycin ml ⁇ 1 on agar plates indicating the presence of some promoter sequences that were functional in vitro.
  • the inserts of the 24 selected iri clones were amplified by PCR using primers complementary to the 5′ ends of the erythromycin- and spectinomycin-resistance genes and the nucleotide sequences of these fragments were determined.
  • the sequence data showed that the 24 clones contained 18 unique sequences.
  • the 18 sequences were analyzed for similarity to known genes by comparison with the sequences in the GenBank/EMBL and SWISSPROT databases.
  • One sequence, iri3I was identical to cps2A, a previously identified S. suis gene putatively involved in the regulation of capsule expression (Smith et al., 1999). Fourteen iri sequences were similar to sequences of known, non- S. suis , genes.
  • iri2 SEQ ID NO: 15
  • iri1, 6 and 22 SEQ ID NO: 8
  • iri34 SEQ ID NO: 21
  • V. cholerae Camilli and Mekalanos, 1995
  • S. aureus S. aureus
  • P. aeruginosa Wang et al., 1996)
  • One, contained in iri1, 6, and 22 was similar to the agrA gene of Staphylococcus aureus , a key locus involved in the regulation of numerous virulence proteins.
  • Three iri sequences had no significant similarity to any sequences in the databases (Table 2).
  • piglets were treated with different concentrations of erythromycin once a day.
  • the erythromycin was administered either orally or intramuscularly.
  • Levels of erythromycin in sera were determined 3, 6 or 24 hours after treatment over one week. High erythromycin levels were detected three hours and six hours after both treatments (results not shown). However, 24 hours after the treatments, the levels decreased dramatically. Based on these data, we hypothesized that for efficient promoter selection, it was necessary to treat the animals twice a day with erythromycin (40 mg kg ⁇ 1 ), once intramuscularly (at 9 a.m.) and once orally (at 4 p.m.).
  • pigs were inoculated either with S. suis strain 10 (pIVS-PE) or with strain 10 (pIVS-E).
  • pIVS-PE the promoter of the mrp gene of S. suis (Smith et al., 1992), which is highly expressed in vivo as well as in vitro, drives expression of the erythromycin-resistance gene.
  • the control plasmid, pIVS-E does not contain a promoter in front of the erythromycin-resistance gene.
  • the strains were inoculated intravenously or intranasally. All pigs infected with strain 10 (pIVS-PE) showed specific S.
  • Piglets were inoculated intravenously with different doses (5 ⁇ 10 5 to 5 ⁇ 10 8 c.f.u.) of the S. suis library (Table 3) and treated with erythromycin as described herein. Specific signs of disease developed in all animals three to eight days after infection (Table 3). High numbers of bacteria were recovered from tissues (central nervous system, joints, serosae, lung, liver, spleen, heart and kidney) of the individual piglets. Analysis of the recovered bacteria showed that a limited number of different clones were present in each of the bacterial samples isolated from the diseased pigs. For example, 30 randomly selected clones from the joints of one pig all possessed identical DNA inserts as assessed by PCR and DNA sequence analysis (results not shown).
  • Two-hundred forty-five clones were analyzed by PCR and partial sequence analysis. Among these, 22 unique ivs clones were found. The 22 sequences were analyzed for similarity to sequences of known genes by comparison with the GenBank/EMBL and SWISSPROT databases (Table 4). The sequences of two genes showed similarity to genes encoding putative virulence factors: ivs21, 26 and 30 which was identical to the epf gene, a previously identified S. suis gene, putatively involved in virulence (Smith et al., 1993; Smith et al., 1996); and ivs31 (SEQ ID NO: 37), which was similar to the fibronectin-binding protein of S.
  • This system differs from the antibiotic-based IVET system described for S. typhimurium (Mahan et al., 1995) in two ways.
  • One is that the lacZ reporter gene fusion is omitted in our vector constructions because inclusion of the lacZ gene resulted in structural instability of the vector.
  • the other difference is that a plasmid system was used rather than a chromosomal integration system.
  • a plasmid system was used because the low transformation efficiency of S. suis (Smith et al., 1995) might prevent the generation of a complete gene library using a chromosomal integration system.
  • Ivs21, 26 and 30 were shown to be identical to the epf gene of S. suis (Smith et al., 1993), which is found in virulent strains of S. suis serotypes 1 and 2 (Stockhofe-Zurwieden et al., 1996; Vecht et al., 1991; Vecht et al., 1992).
  • Ivs31 SEQ ID NO: 37
  • Ivs31 showed similarity to the fibronectin/fibrinogen-binding protein of S. gordonii (accession no.
  • Ivs25 (SEQ ID NO: 34) showed significant similarity to the sapR gene of S. mutans (accession no. P72485) and Lactobacillus sake Lb706 (Axelsson and Holck, 1995) as well as to the agrA gene of S. aureus (Projan and Novick, 1997), both of which encode response regulator proteins of bacterial two-component signal-transduction systems, thus mediating the response to an environmental signal (Projan and Novick, 1997).
  • Use of an IVET selection system for S. aureus in mice selected the region preceding the agrA gene, suggesting induction of agrA expression under in vivo conditions (Lowe et al., 1998).
  • the agr locus was shown to play an important role in altering the expression of a considerable number of virulence factors in response to cell density (Projan and Novick, 1997).
  • Clones ivs6, 7, 13 and 14 showed similarity to a gene, iviVI, previously identified by IVET selection in V. cholerae (Camilli and Mekalanos, 1995). The function of iviVI is unknown. However, the genes showed similarity to members of the ATP-binding cassette family of transporters.
  • the sequenced portion of ivs6, 7, 13 and 14 included an N-terminal ATP-binding Walker A box motif, which is highly conserved in this transporter family.
  • the oxyR gene is the central regulator of oxidative stress response in E. coli (Demple, 1999) and approximately ten genes are under the control of the OxyR protein.
  • the second gene, ivs2, 4 and 28 (SEQ ID NO: 31), which is identical to iri10 and 20 (SEQ ID NO: 9), showed similarity to the yoaE gene of E. coli (accession no. P76262), a putative ABC transporter protein.
  • the third and the fourth genes, ivs25 (SEQ ID NO: 34) and ivs6, 7, 13 and 14 (SEQ ID NO: 43) were identical to iri1, 6 and 22 (SEQ ID NO: 8) and iri2 (SEQ ID NO: 15), respectively. These genes also showed similarity to ivi genes selected using IVET in other bacterial species.
  • S. suis ivs genes Two of the S. suis ivs genes, ivs1 (SEQ ID NO: 25) and ivs8 (SEQ ID NO: 44), showed similarity to transposon sequences. Moreover, one S. suis ivs gene, ivs2, 4 and 28 (SEQ ID NO: 31), had a GC% that was considerably higher than the composition of the rest of the selected genes. It is striking that in S. typhimurium , several of the ivi clones that are required for full virulence have been found to be associated with mobile elements. Their atypical base composition and codon usage has led to the suggestion that they have been acquired from other bacterial species by horizontal transfer (Conner et al., 1998).
  • iri7 SEQ ID NO: 23
  • SEQ ID NO: 23 Another iri gene, iri7 (SEQ ID NO: 23), showed similarity to the rpgG gene of S. mutans . This gene was shown to be required for the biosynthesis of rhamnose-glucose polysaccharide (Yamashita et al., 1999). Because rhamnose is part of the polysaccharide capsule in S. suis serotype 2 (Elliott and Tai, 1978), a role of the iri7 (SEQ ID NO: 23) gene in capsule biosynthesis can be proposed.
  • Iri34 (SEQ ID NO: 21) showed similarity to the np 16 gene, previously identified using IVET selection in P. aeruginosa and suspected to encode threonine dehydratase activity (Wang et al., 1996). Together with the observation that 4 iri genes could be selected by the in vivo approach, these data show that the iri genes encode important virulence factors for S. suis.
  • Streptococcus suis causes severe infections, such as meningitis, septicemia, and arthritis, in piglets. The animals often do not survive the infection (6, 28). Occasionally, S. suis causes septicemia and meningitis in humans (3). The pathogenesis of an S. suis infection is rarely understood. Sows are symptomless carriers of S. suis on their tonsils and pass the bacteria on to their piglets. The piglets cannot cope with the bacteria and subsequently develop the specific symptoms of an S. suis infection. Until now, 35 capsular serotypes of S. suis have been described (26), but serotype 2 strains are most often isolated from diseased piglets.
  • the capsule is an important virulence factor since piglets infected with an acapsular mutant of S. suis serotype 2 strains do not develop any clinical symptoms (22).
  • Bacterial proteins have been suggested to play a role in the pathogenesis as well (2, 26).
  • the expression of murimidase-released protein (MRP), extracellular factor (EF) and suilysin was shown to be strongly associated with pathogenic strains of S. suis serotype 2 (1, 29, 30). Since isogenic mutants lacking MRP and EF and isogenic mutants lacking suilysin were still pathogenic in young piglets, these proteins are not absolutely required for virulence (2, 23).
  • MRP murimidase-released protein
  • EF extracellular factor
  • suilysin was not absolutely required for virulence (2, 23).
  • Recently, a new virulence factor was identified (21) by using a complementation approach. The function of this virulence factor in the pathogenesis has to be further investigated.
  • the other (ivs-31) (SEQ ID NO: 37) showed homology to genes encoding fibronectin-/fibrinogen-binding proteins of Streptococcus gordonii (GenBank accession no. X65164) and Streptococcus pyogenes FBP54 (8).
  • a considerable number of fibronectin-binding proteins of various bacterial species have been shown to be important virulence factors (12).
  • FBP54 was shown to be expressed in the human host and to preferentially mediate adherence to human buccal epithelial cells (7). It was shown that the FBP54 protein induces protective immunity against S. pyogenes challenge in mice (13).
  • FBPS fibronectin-/fibrinogen-binding protein of S. suis
  • S. suis strains were grown in Todd-Hewitt broth (code CM 189; Oxoid, Ltd.) and plated on Columbia blood base agar plates (code CM331; Oxoid, Ltd., London, United Kingdom), containing 6% (vol/vol) horse blood.
  • E. coli strains were grown in Luria Broth (17) and plated on Luria Broth containing 1.5% (wt/vol) agar. If required, antibiotics were added at the following concentrations: 50 ⁇ g/ml of spectinomycin (Sigma, St. Louis, Mo.) for E. coli and 100 ⁇ g/ml for S. suis, 100 ⁇ g/ml of ampicillin (Boehringer, Mannheim, Germany) for E. coli and 25 ⁇ g/ml of kanamycin (Boehringer) for E. coli.
  • DNA sequences were determined on a 373A DNA Sequencing System (Applied Biosystems, Warrington, Great Britain). Samples were prepared by use of an ABI Prism dye terminator cycle sequencing ready reaction kit (Applied Biosystems). Sequencing data were assembled and analyzed using the Lasergene program (DNASTAR). The BLAST software package was used to search for protein sequences homologous to the deduced amino acid sequences in the GenBank/EMBL databases.
  • Chromosomal DNA was isolated as described by Sambrook et al. (19). DNA fragments were separated on 0.8% agarose gels and transferred to GeneScreen Plus hybridization transfer membrane (NENTM Life Science Products, Boston, USA) as described by Sambrook et al. (19). DNA probes of the fbps and spc genes were labeled with ( ⁇ - 32 P)dCTP (3,000 Ci/mmol; Amersham Life Science, Buckinghamshire, Great Britain) by use of a random primed DNA labeling kit (Boehringer). The DNA on the blots was pre-hybridized for at least 30 minutes at 65° C. and subsequently hybridized for 16 hours at 65° C.
  • the pathogenic strain 10 27, 29 of S. suis serotype 2 was electrotransformed (24) with the plasmid pFBPS7-47.
  • the fbps gene was inactivated by the insertion of a spectinomycin-resistance gene.
  • pFBPS7-47 FIG. 1
  • the 382 bp SalI-SalI fragment of pFBPS7-46 was replaced by the 1.2 kb EcoRV-SmaI fragment of pIC-Spc, containing the spectinomycin resistance gene, after the SalI sites of the vector were made blunt (FIG. 1).
  • FBPS expression plasmid To construct an FBPS expression plasmid, the QIAexpress Kit (Qiagen GmbH, Hilden, Germany) was used. The primers corresponded to positions 250 to 272 and from 1911 to 1892 of the fbps gene. The sequences of these primers were 5′(GCGGATCCGATGACGATGACAAATCTTTTGACGGATTTTTTTTAC)3′ (SEQ ID NO: 46) and 5′(CCCAAGCTTGGGCATGAACTAGATTTTCATGG)3′ (SEQ ID NO: 47). The primers contained restriction sites for BamHI and HindIII, respectively, to amplify the fbps gene from pFBPS7-47.
  • the amplified PCR product was digested with BamHI and HindIII and the 1.8 kb fbps gene was cloned into pQE-30 digested with BamHI and HindIII, yielding pQE-30-FBPS.
  • pQE-30-FBPS was transformed to M15 (pREP4).
  • M15 (pREP4) (pQE-30-FBPS) was used to express and purify the FBPS using the QIAexpressionistTM (Qiagen).
  • M15 (pREP4) (pQE-30FBPS) cells were grown exponentially; 1 mM IPTG was added and the cells were allowed to grow another four hours at 37° C. Subsequently, cells were harvested and lysed. The cleared supernatants were loaded onto Ni 2+ -NTA agarose columns. FBPS containing a 6 ⁇ HIS tag was bound to the Ni 2++ -column. The columns were washed and the protein was eluted. Different buffers were used for native and for denaturing purification.
  • FBPS purified under denaturing conditions was renaturated on a Ni 2+ -NTA column by using a linear 6 M-1 M urea gradient in 500 mM NaCl, 20% glycerol and 20 mM Tris-HCl (pH7.4), containing protease inhibitors (25 ⁇ g/ml of pefabloc, 0.7 ⁇ g/ml of pepstatin, 1 ⁇ g/ml of aprotinin, 0.5 ⁇ /ml of leupeptin). All procedures were performed according to the manufacturer's recommendations.
  • the 6 ⁇ HIS tag was removed from the protein by incubating purified FBPS in 20 mM Tris-HCl (pH 7.4), 50 mM NaCl, 2 mM CaCl 2 and 0.5 U of light chain enterokinase (New England Biolabs, Beverly, Mass.) for 16 hours at RT.
  • FBPS Purified and renaturated FBPS was used to immunize two rabbits. To remove urea, the protein was dialyzed against phosphate buffered saline (136 mM NaCl; 2.68 mM KCl; 8.1 mM Na 2 HPO 4 ; 2.79 mM KH 2 PO 4 (pH 7.2)) over night at 4° C. Seven days before immunization, blood was collected from the rabbits to determine the natural titers against FBPS.
  • phosphate buffered saline 136 mM NaCl; 2.68 mM KCl; 8.1 mM Na 2 HPO 4 ; 2.79 mM KH 2 PO 4 (pH 7.2)
  • those rabbits with negative anti-FBPS titers were immunized intramuscularly with two times 0.5 ml of 100 ⁇ g/ml of FBPS in a water-in-oil emulsion (Specol; ID-Lelystad).
  • rabbits were immunized for the second time using the same amount of protein and the same route of immunization.
  • the rabbits were sacrificed and blood was collected. The blood was coagulated and serum was collected and used for immunodetection of FBPS.
  • Proteins were separated by sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis (PAGE) by standard procedures (19). Proteins in the gel were visualized using SYPRO-orange (Molecular Probes, Sunnyvale, Calif.) staining according to the manufacturer's recommendations. Signals were detected on a phosphor imager (Storm; Molecular Dynamics). A known bovine serum albumin concentration range was used as a standard to calculate the amounts of protein present in the gel. The Molecular Dynamics program was used for the calculations.
  • Proteins were transferred to a nitrocellulose membrane by standard procedures (19). The membranes were blocked in Blotto: Tris-buffered saline (TBS) (50 mM Tris-HCl (pH 7.5), 150 mM NaCl) containing 4% skimmed milk and 0.05% Tween 20 at room temperature (RT) for one hour.
  • TBS Tris-buffered saline
  • RT room temperature
  • FBPS recombinant purified FBPS
  • membranes were incubated with a monoclonal antibody against the 6 ⁇ HIS tag (Clontech, Palo Alto, Calif.) in a 1:10,000 dilution in Blotto-TBS (1:1) at RT for one hour, followed by an incubation with alkaline phosphatase-conjugated anti-mouse antibody in a 1:1,000 dilution in Blotto-TBS (1:1) at RT for one hour.
  • Reactivity of purified FBPS was tested by using a convalescent serum of a pig that had survived an S. suis infection.
  • Nitrocellulose membranes were incubated with the polyclonal pig serum in a 1:200 dilution in Blotto-TBS (1:1) at RT for one hour, followed by incubation at RT for one hour with alkaline phosphatase-conjugated anti-swine antibody in a 1:2,000 dilution in Blotto-TBS (1:1).
  • Nitro Blue Tetrazolium Merck, Darmstadt, Germany
  • bromochloroindolyl phosphate Sigma
  • Binding studies were performed by indirect Western blotting. Proteins were separated by SDS-PAGE and transferred to a nitrocellulose membrane as described herein. The membranes were blocked in MPBS: PBS containing 4% skimmed milk and 0.05% Tween 20. Subsequently, the membrane was incubated with 5 ⁇ g/ml of human fibronectin (fn) (Sigma) or 5 ⁇ g/ml of human fibrinogen (fgn) (Sigma) in PBS containing 5% fetal calf serum, 2% NaCl, and 0.05% Tween 80 at RT for one hour.
  • fn human fibronectin
  • fgn human fibrinogen
  • the membranes were incubated with horse-radish peroxidase-conjugated anti-fibronectin (DAKO) or anti-fibrinogen (DAKO) antibodies in a 1:1,000 dilution in PBS containing 5% fetal calf serum, 2% NaCl, and 0.05% Tween 80 at RT for one hour.
  • the signal was visualized by using ECL + (Amersham Pharmacia Biotech, N.J.) according to the manufacturer's recommendations. Signals were detected on a phosphor imager (Storm; Molecular Dynamics). All washing steps were performed in MPBS-PBS (1:1).
  • Germ-free piglets cross-breeds of Great Yorkshire and Dutch Landrace, were obtained from sows by cesarean section. The surgery was performed in sterile flexible film isolators. Piglets were allotted to groups of four and were housed in sterile stainless steel incubators. Housing conditions and feeding regimens were as described (27, 29). Six-day-old piglets were inoculated intranasally with about 10 7 cfu of Bordetella bronchiseptica 92932 to predispose the piglets to infection with S. suis . Two days later, the piglets were inoculated intranasally with 10 6 cfu of S. suis strain 10 plus 10 6 cfu of S.
  • LD 50 values should be determined. To do this, large numbers of piglets are required. For ethical reasons, this is not acceptable. To circumvent this problem, co-colonization studies were performed.
  • swabs taken from the nasopharynx and the feces were cultured three times a week.
  • the swabs were plated directly onto Columbia agar containing 6% horse blood or grown for 48 hours in Todd-Hewitt broth and subsequently plated onto Columbia agar containing 6% horse blood.
  • Pigs were monitored twice a day for clinical signs and symptoms, such as fever, nervous signs, and lameness. Blood samples from each pig were collected three times a week. Leukocytes were counted with a cell counter. The piglets were killed when specific signs of an S.
  • Tissue specimens from heart, lung, liver, kidney, spleen, and tonsil, and from the organs specifically involved in an S. suis infection were sliced with a scalpel or a tissuenizer. Tissue slices from each organ or site were resuspended in 2-25 ml of Todd-Hewitt containing 15% glycerol depending on the size of the tissue slice. The suspension was centrifuged at 3,000 rpm for five minutes.
  • the supernatant was collected and serial dilutions were plated on Columbia agar containing 6% horse blood, as well as on Columbia agar plates containing 6% horse blood and 100 ⁇ g/ml of spectinomycin to quantitate the number of wild-type and mutant bacteria present.
  • the number of mutant strain 10 ⁇ FBPS cells was determined by counting the number of CFU on the appropriate serial dilution on the selective plates; the number of wild-type strain 10 cells was determined. by counting the number of CFU on the appropriate serial dilution on the Columbia Agar blood plates of which the number of CFU counted on the selective plates was subtracted.
  • the ratio of wild-type and mutant strain was determined again by toothpicking about 100 individual colonies onto both Columbia Agar plates and onto Columbia Agar plates containing 100 ⁇ g/ml spectinomycin.
  • nucleotide sequence data fbps have been submitted to GenBank, in which the sequence is listed under accession no. AF438158.
  • ivs-31 SEQ ID NO: 37 (20) showed homology to the 5′ part of genes encoding for FlpA and FBP54, fibronectin-binding proteins (FnBP) of Streptococcus gordonii (GenBank accession no. X65164) and Streptococcus pyogenes (8), respectively.
  • FnBP fibronectin-binding proteins
  • FBPS was substantially homologous to FlpA of S. gordonii (76%) and also showed homology to FnBP's of other organisms, like Streptococcus pneumoniae (73%), S. pyogenes (69%), Lactococcus lactis (59%), and Bacillus subtilis (41%).
  • FBP54 FBPS has a longer N-terminus with 76 additional amino acids. This longer N-terminus was also seen in other organisms like S. gordonii, S. pneumoniae and B. subtilis .
  • the primary fibronectin-/fibrinogen-binding domain was localized to its N-terminal part, to the first 89 amino acids (8). Over this region, the homology of FBPS to FBP54 is very high (80%), suggesting that FBPS can bind both fibronectin and fibrinogen.
  • FBPS was purified under native conditions.
  • a protein expression construct which expresses FBPS with a 6 ⁇ HIS tag fused to the N-terminus, was used for purification.
  • Four hundred ⁇ g of FBPS was purified from 50 ml of exponential-phase E. coli cells after induction with IPTG. The purity of this FPBS was determined with SDS-PAGE and Western blotting (FIG. 2).
  • the induced E. coli lysate contained a broad range of proteins, among which the 64 kDa protein FBPS was present (panel A, lane 1).
  • highly purified FBPS with 6 ⁇ HIS tag was obtained (panel A, lane 2).
  • FBPS was the only protein that was detected (panel B).
  • FIG. 3 panels E and F, show that FBPS without the 6 ⁇ HIS tag still efficiently bound to fn and fgn. Therefore, it appears that FBPS can specifically bind to fn and fgn.
  • the mutant strain 10 ⁇ FBPS was used in a competition challenge experiment with the wild-type strain to determine the relative attenuation of the mutant strain.
  • the growth rates of the wild-type and mutant strain in Todd-Hewitt medium were found to be essentially identical (data not shown).
  • Wild-type and mutant strain were inoculated at an actual ratio of 0.65 (1.63 ⁇ 10 6 cfu of wild-type bacteria ml ⁇ 1 and 3.09 ⁇ 10 6 CfU of mutant bacteria ml ⁇ 1 ).
  • piglets that developed specific S. suis symptoms meningitis, arthritis, or mortal illness
  • Piglets that did not develop these symptoms were killed at the end of the experiment. From all piglets, the ratio of wild-type and mutant strain in various organs was determined.
  • subtilis YvyD P28368) sigma-54 modulator homologue 44 (SEQ ID NO: 16) Metabolic functions iri 7 800 39 S. mutans RgpG (Q9XDW8) rhamnose-glucose biosynthesis 76 (SEQ ID NO: 23) iri 11 700 34 L. lactis NrdD (Q9ZAX6) anaerobic ribonucleotide reductase 51 (SEQ ID NO: 10) iri 14 500 38 S. pneumoniae SulB (Q54614) dihydrofolate synthetase 41 (SEQ ID NO: 12) iri 16 850 48 B.
  • subtilis TrmU (O35020) RNA methyltransferase 62 (SEQ ID NO: 13) iri 32 300 41 C. histolyticum RuvB (O9ZNJ5) hypoxanthine-guanine 55 (SEQ ID NO: 20) phosphoribosyl transferase iri 34 1000 44 L. lactis IlvA (U92974) probable threonine dehydratase 56 (SEQ ID NO: 21) P. aeruginosa Pn16 Transporter functions iri 2 750 36 B. subtilis YloD (O34328) putative guanylate kinase 50 (SEQ ID NO: 15) S.
  • gordonii ComYA (U81957) putative ABC transporter 37 Vibrio cholerae IviVI (Q56605) putative ABC transporter 47 iri 10, 20 1350 51 E. coli YoaE (P76262) putative transport protein 94 (SEQ ID NO: 9) Unknown iri 13, 15, 27 800 34 M. tuberculosis MTCY336_33 unknown 38 (SEQ ID NO: 11) hypothetical protein (O06593) iri 29 850 36 S. aureus Yp15 (P13977) unknown 39 (SEQ ID NO: 18) hypothetical protein iri 18 800 39 S.
  • S. suis genes selected in pigs. Sites of Insert Data base homology Function % Clone isolation (bp) GC % (accession no.) of homolog Identity Putative virulence factors ivs 31 CNS 200 47 S. gordonii FlpA (X65164) fibronectin/fibrinogen binding 70 (SEQ ID NO: 37) Regulatory functions ivs 25 joint 800 34 S. mutans SapR (P72485) response regulator protein 49 (SEQ ID NO: 34) S. aureus AgrA (X52543) response regulator protein 51 S. suis Iri 1, 6, 22 100 ivs 23, 24 other 850 38 S.
  • agalactiae CpsY (CAB36982) regulation capsule expression 46 (SEQ ID NO: 33) E. coli OxyR (P11721) oxidative stress regulator 51 S. suis Iri 24 100 ivs 16 CNS 800 43 S. epidermidis AltR (U71377) putative transcriptional regulator 26 (SEQ ID NO: 28) ivs 20 lung 800 41 L. lactis AldR (O34133) putative regulator AldR 64 (SEQ ID NO: 32) Metabolic functions ivs 33 CNS 570 36 E. coli ThrC (P00934) threonine synthase 41 (SEQ ID NO: 39) ivs 5, 10, CNS, joint 900 36 S.
  • gordonii Tdk P47848 thymidine kinase 87 12, 22 (SEQ ID NO: 42) ivs 18 lung 730 32 S. mutans NADH oxidase (JC4541) NADH oxidase 80 (SEQ ID NO: 29) Transporter functions ivs 2, 4, 28 CNS, joint 1350 51 E. coli YoaE (P76262) putative transport protein 94 (SEQ ID NO: 31) S. suis iri 10, 20 100 ivs 3 joint 1000 42 S. mutans OrfU (AF267498) putative ABC transporter (permease) 33 (SEQ ID NO: 36) ivs 6, 7, CNS, joint 1350 36 B.
  • subtilis Ylo D (O34328) putative guanylate kinase 50 13, 14 (SEQ ID NO: 43) S. gordonii ComYa (U81957) putative ABC transporter 37 V. cholera IviVI (Q56605) putative ABC transporter 47 S. suis Iri 2 100 Transposases ivs 8 CNS 600 41 S. pneumoniae transposase transposase 70 (SEQ ID NO: 44) (Z86112) ivs 1 joint 1600 39 C. perfringens (X71844) putative transposase 56 (SEQ ID NO: 25) Miscellaneous ivs 32, 35 CNS 500 38 S.
  • typhimurium FliF flagellar M-protein precursor 36 (SEQ ID NO: 38) ivs 9, 17 joint, CNS 800 36 B. subtilis ComE ORF2 (P32393) competence development 37 (SEQ ID NO: 45) ivs 11 serosea 800 44 P. syringae TabA (P31851) diaminopimelate decarboxylase/ 53 (SEQ ID NO: 26) tabtoxin Unknown ivs 15 CNS 750 42 B. subtilis conserved hypothetical unknown 43 (SEQ ID NO: 27) protein YdiB (D88802) ivs 29 joint 800 38 S.
  • salivarius hypothetical protein unknown 79 SEQ ID NO: 35
  • AF130465 ivs 34 CNS 600
  • B. subtilis conserved hypothetical unknown 61 SEQ ID NO: 40
  • protein YRRK O34634
  • YqeG P54452
  • S. cristatus hypothetical protein unknown 86 SEQ ID NO: 30
  • Polissi, A., A. Pontiggia, G. Feger, M. Altieri, H. Mottl, L. Ferrari and D. Simon. (1998). Large-scale identification of virulence genes from Streptococcus pneumoniae. Infect. Immun. 66, 5620-5629.

Abstract

The invention relates to the field of the diagnosis of and vaccination against Streptococcal infections, and to the detection of virulence markers of Streptococci. The invention discloses a method for modulating virulence of a Streptococcus comprising modifying a genomic fragment of the Streptococcus, wherein the genomic fragment comprises at least a functional part of a fragment identifiable by hybridization in Streptococcus suis to a nucleic acid or fragment thereof.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of PCT/NL02/00073, filed Jan. 31, 2002, designating the United States of America, corresponding to PCT International Publication WO 02/061070 (published in English on Aug. 8, 2002), the contents of which are incorporated herein in its entirety.[0001]
  • TECHNICAL FIELD
  • The invention relates to the field of the diagnosis of and vaccination against Streptococcal infections, and to the detection of virulence markers of Streptococci. [0002]
  • BACKGROUND
  • Streptococcus species, of which there are a large variety of that cause infections in domestic animals and man, are often grouped according to Lancefield's groups. Typing according to Lancefield occurs on the basis of serological determinants or antigens that are, among others, present in the capsule of the bacterium and only allows for an approximate determination. Often, bacteria from a different group show cross-reactivity with each other, while other Streptococci cannot be assigned a group-determinant at all. Within groups, further differentiation is often possible on the basis of serotyping. These serotypes further contribute to the large antigenic variability of Streptococci, a fact that creates an array of difficulties within diagnosis of and vaccination against Streptococcal infections. [0003]
  • Lancefield group A Streptococcus (GAS, [0004] Streptococcus pyogenes) are common with children and cause nasopharyngeal infections and complications thereof. Animals, such as cattle, are susceptible to GAS, wherein mastitis is often found associated with the cattle.
  • Lancefield group B Streptococcus (GBS) are most often seen with cattle and cause mastitis. However, human infants are susceptible as well, often with fatal consequences. Group B Streptococci (GBS) constitute a major cause of bacterial sepsis and meningitis among human neonates born in the United States and Western Europe and are emerging as significant neonatal pathogens in developing countries. [0005]
  • Lancefield group C infections, such as those with [0006] S. equi, S. zooepidemicus, S. dysgalactiae, and others are mainly seen associated with horses, cattle and pigs, but can also cross the species barrier to humans.
  • Lancefield group D ([0007] S. bovis) infections are found with all mammals and some birds, sometimes resulting in endocarditis or septicemia.
  • Lancefield groups E, G, L, P, U and V ([0008] S. porcinus, S. canis, S. dysgalactiae) are found with various hosts and cause neonatal infections, nasopharyngeal infections or mastitis.
  • Within Lancefield groups R, S, and T (and with ungrouped types), [0009] S. suis is found and is an important cause of meningitis, septicemia, arthritis and sudden death in young pigs. Incidentally, S. suis can also cause meningitis in man.
  • Ungrouped Streptococcus species, such as [0010] S. mutans, causes carries with humans. S. uberis causes mastitis with cattle, S. pneumonia causes major infections in humans, and Enterococcus faecilalis and E. faecium further contribute to the large group of Streptococci. Streptococcus pneumoniae (the pneumococcus) is a human pathogen causing invasive diseases, such as pneumonia, bacteremia, and meningitis.
  • Little is known about the pathogenesis of the disease caused by Streptococci. Various cellular components, such as muramidase-released protein (MRP), extracellular factor (EF) and cell-membrane-associated proteins including fimbriae, hemagglutinins, and hemolysin have been suggested as virulence factors. However, the precise role of these protein components in the pathogenesis of the disease remains unclear. It is known and generally accepted that the polysaccharidic capsule of various Streptococci and other gram-positive bacteria play an important role in pathogenesis. The capsule enables these microorganisms to resist phagocytosis and is, therefore, regarded as an important virulence factor or marker. [0011]
  • In particular, [0012] Streptococcus suis is an important cause of meningitis, septicemia, arthritis and sudden death in young pigs. It can also cause meningitis in man. Attempts to control the disease are hampered by the lack of sufficient knowledge about the pathogenesis of the disease and the lack of effective vaccines and sensitive diagnostic methods.
  • So far, 35 serotypes of [0013] S. suis are described. Virulence of S. suis can differ within and among serotypes. Worldwide, S. suis serotype 2 is the most frequently isolated serotype. Within S. suis serotype 2, pathogenic, weak-pathogenic and non-pathogenic strains can be found. The pathogenic strains cause severe clinical signs of disease in pigs and large numbers of bacteria can be re-isolated from the central nervous system (CNS) and the joints after experimental infection. The weak-pathogenic strains cause only mild clinical signs of disease and infrequently are bacteria re-isolated from the CNS and the joints after experimental infection. The non-pathogenic strains are completely avirulent in young pigs after experimental infection.
  • The 136-kDa muramidase-related protein (MRP) and the 110-kDa extracellular factor (EF) are generally considered as important virulence markers for [0014] S. suis serotype 2 strains isolated in Europe and the United States. However, differences in virulence between pathogenic, weak-pathogenic and non-pathogenic strains cannot exclusively be explained by differences in their MRP and EF expression patterns. In addition, it is known that the capsule of Streptococcus suis serotype 2 is an important virulence factor. However, since pathogenic, weak-pathogenic and non-pathogenic strains seem to be fully encapsulated after growth in vitro and in vivo, it is not likely that the level of encapsulation of these fully encapsulated strains is associated with their difference in virulence.
  • SUMMARY OF THE INVENTION
  • The invention discloses a method for modulating virulence of a Streptococcus comprising modifying a genomic fragment of Streptococcus. The genomic fragment comprises at least a functional part of a fragment identifiable by hybridization in [0015] Streptococcus suis to a nucleic acid or fragment thereof as shown in SEQ ID NOS: 8 through 45 and obtaining a clone including the modified genomic fragment. In one exemplary embodiment, the genomic fragment comprises at least a functional part of a gene, the expression of which can be environmentally regulated in S. suis by iron-restricted conditions. In another exemplary embodiment, the genomic fragment comprises at least a functional part of a gene which is expressed in a wild-type S. suis infected pig (in vivo). In a further exemplary embodiment, selection under iron-restricted conditions is combined with selection in vivo. In one embodiment, the gene encodes a fibronectin/fibrinogen-binding protein. The method disclosed herein is useful for modulating virulence of Streptococcus suis and comprises functionally deleting the expression of at least the functional part of the gene by Streptococcus.
  • The phrase “functionally deleting” as used herein refers to any technique known in the art (such as allowing for a deletion, insertion, mutation or the occurrence of a frame-shift in the open-reading frame of the respective gene) that is instrumental in hampering or inhibiting the expression of a gene-product (be it mRNA and/or protein) of the gene. Thus, the invention discloses a clone of a Streptococcus obtained or obtainable by a method according to the invention. [0016]
  • To get insight in the differences between pathogenic, weak-pathogenic and nonpathogenic strains or clones that are determined by their difference in virulence, the invention describes the identification of environmentally regulated genes of [0017] Streptococcus suis by iron-restricted conditions and by experimental infection of piglets. Eighteen unique iron-restricted induced (iri) genes and 22 unique in vivo selected (ivs) genes of S. suis were found. None of the ivs genes was exclusively expressed in vivo. Four iri genes were substantially identical to four ivs genes selected in piglets. Two ivs genes were similar to genes for putative virulence factors. One of these ivs genes was substantially identical to the epf gene of virulent S. suis serotype 2 strains and the other ivs gene showed homology to a gene encoding a fibronectin-binding protein of Streptococcus gordonii.
  • In yet another embodiment, the invention discloses a study of the characteristics of fibronectin- and fibrinogen-binding protein of [0018] Streptococcus suis (FBPS) and its gene as identified herein. The ability to bind fibronectin, either in fluid phase or immobilized onto a surface, is a property of S. suis and is one of the mechanisms S. suis uses for attachment to and invasion of host cells. Therefore, FBPS is an important virulence factor. The gene encoding FBPS was identified using an in vivo selection system in pigs as described herein, thus, showing an important role of the protein in vivo. This finding was supported by the observation that isogenic FBPS mutants, herein also disclosed, of S. suis are attenuated in pigs. Surprisingly, FBPS bound to fibronectin, as well as to fibrinogen, but did not show the structural characteristics of the fibronectin-binding proteins most commonly described and explains why FBPS has not been found earlier. Most fibronectin-binding proteins described to date are large cell surface proteins with predicted sizes of 60-100 kDa and have similar structural organizations. The proteins contain an N-terminal signal sequence as well as the cell wall signaling sequence (LPXTGE) (SEQ ID NO: 1). The Fn-binding sites include 30-42 amino acid long motifs, repeated 3-4 times. In particular, the first fibronectin- and fibrinogen-binding protein of S. suis is disclosed herein. The gene encoding FBPS was cloned and sequenced and FBPS was purified. Binding of FBPS to human fn and fgn was shown. FBPS was shown to be involved in the colonization of the organs specific for an S. suis infection in piglets, but not in the colonization of S. suis on the tonsils of piglets.
  • Many Streptococci and Staphylococci have several different fibronectin- and/or fibrinogen-binding proteins, most of which are very large, about 130 kDa. Until now, [0019] S. pyogenes was the only organism to have a large, as well as a smaller (54 kDa), FnBP. The existence of more than one FnBP explains why in some organisms, isogenic mutants defective in only one of the FnBPs can still bind to fn and/or fgn can be further attenuated in vivo in relation to fibronectin binding.
  • The role of FBPS in the pathogenesis of [0020] S. suis was studied in an experimental infection model in piglets. Since we were unable to determine a LD50 values for the mutant clones because no lethal dose could be established using normally used numbers of bacteria, it was decided to compare the virulence of the isogenic FBPS clone to the wild-type S. suis in a competitive infection assay in piglets. This kind of co-colonization experiment has been successfully applied to determine the virulence of mutants of Actinobacillus pleuropneumoniae in piglets. The data showed that the mutant clone was capable of colonizing the tonsil as efficiently as the wild-type. This strongly indicates that FBPS is not involved in the colonization of the tonsil. The data also indicated that FBPS does play a role in the colonization of specific organs, since in the competition assay, joints and the CNS were more efficiently colonized by wild-type than by mutant bacteria.
  • In addition, higher numbers of wild-type bacteria were re-isolated from the specific organs compared to the numbers of mutant bacteria, indicating that the mutant clone is attenuated in vivo. Although the number of pigs used for this experiment was low, the data indicates that the FBPS mutant is less virulent than the wild-type strain. It was demonstrated that FBPS reacted with a convalescent serum of a pig that survived an [0021] S. suis infection. Therefore, FBPS is immunogenic in pigs and this finding demonstrates that FBPS of S. suis is expressed under in vivo conditions.
  • It is also shown that the fbps gene was present in all known serotypes of [0022] S. suis (except for two), as well as in all three phenotypes of serotype 2. This suggests that the fbps gene is present among most serotypes. However, the expression of FBPS in all serotypes and phenotypes was not studied. Therefore, it is possible that although all strains, except for serotypes 32 and 34, possess the fbps gene, not all strains express FBPS. Based on the facts that FBPS is immunogenic in pigs and that the fbps gene is present in all prevailing S. suis serotypes, (except for two), FBPS is an attractive candidate for a cross-protective vaccine against essentially all serotypes. In one embodiment, the mutant strain 10ΔFBPS may be used in the vaccine, which mutant is possibly further attenuated by deleting one or more virulence factors as described herein. In another embodiment, this vaccine is based on purified FBPS protein or an antigenic part thereof with a suitable adjuvant.
  • To further validate a method for identifying a virulence factor, the role of the fibronectin-/fibrinogen-binding protein (FBPS) in the pathogenesis of [0023] S. suis serotype 2 was investigated in piglets as described herein. The complete gene encoding FBPS from S. suis serotype 2 was cloned in E. coli and sequenced. The occurrence of the gene in various serotypes was analyzed by hybridization studies. The FBPS protein was expressed in E. coli, purified and binding to human fibronectin and fibrinogen was demonstrated. The induction of antibodies in piglets was studied upon infection. An isogenic mutant unable to produce FBPS was constructed and the virulence of the wild-type and mutant strains was compared in a competitive infection model in young piglets. Organ cultures showed that FBPS was not required for colonization of the tonsils, but that FBPS played a role in the colonization of the specific organs involved in an S. suis infection. Therefore, the FBPS mutant was considered as an attenuated mutant which is useful in a vaccine. Alternatively, a vaccine is used that mainly includes the FBPS protein or at least of an antigenic part thereof, such that an FBPS-specific antibody or T-cell response in pigs is developed after vaccination with the FBPS or antigenic part thereof.
  • Two additional ivs genes showed homology to environmentally regulated genes previously identified by using an in vivo expression technology (IVET) selection in other bacterial species. One of these showed similarity to the agrA gene of [0024] Staphylococcus aureus, a key locus involved in the regulation of numerous virulence proteins.
  • Thus, the invention also discloses a method for assaying virulence of a Streptococcus comprising assaying a genomic fragment of Streptococcus, wherein the genomic fragment comprises at least a functional part of a fragment identifiable by hybridization in [0025] Streptococcus suis to a nucleic acid or fragment thereof as described herein.
  • The invention also discloses a vector comprising a nucleic acid according to the invention and a host cell comprising a nucleic acid or a vector according to the invention. Such a host cell comprises an easily modifiable organism such as [0026] E. coli. However, other host cells, such as a recombinant Streptococcus comprising a vector. or nucleic acid according to the invention are also disclosed herein.
  • The invention additionally discloses a vaccine comprising a nucleic acid, a vector or a host cell according to the invention, and use of such a vaccine in the prevention and/or treatment of Streptococcal infections. [0027]
  • Also disclosed is a protein or fragment thereof encoded by a nucleic acid according to the invention, such as a protein encoded by a nucleic acid or fragment thereof disclosed herein or functional, i.e., antigenic fragment thereof. The invention also discloses an antibody directed against a protein or fragment thereof according to the invention and an antigen reactive with such an antibody, for example comprising a protein or fragment. Such a protein or fragment thereof need not be obtained by recombinant means. Synthesis of peptides, according to their amino acid sequence, is as well equally possible. Such antigens and antibodies as described herein can be used in a diagnostic test comprising an antibody according to the invention, or within a vaccine or diagnostic test comprising an antigen according to the invention. Such vaccines and diagnostic tests can be used in the field of the diagnosis of and vaccination against Streptococcal infections and for the detection of virulence markers of Streptococci.[0028]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a schematic presentation of the procedure used to clone the fbps gene of [0029] S. suis serotype 2 and the construction of an insertional knock-out mutant in S. suis serotype 2. A 5 kb EcoRI fragment was cloned in pGEM7Zf(+), yielding pFBPS7-46. In pFBPS7-47, the 382 bp SalI-SalI fragment of pFBPS7-46 was replaced by 1.2 kb spectinomycin-resistance gene, after the vector was made blunt to obtain an insertional knock-out of fbps. Ivs-31: in vivo selected gene 31.
  • FIG. 2 shows purity and immunogenicity of FBPS purified under native conditions. SDS-PAGE analysis with SYPRO orange, a non-specific protein-staining (panel A) and Western blot analysis with a monoclonal antibody against the 6×HIS tag (panel B) of 4:1 of [0030] E. coli M15 (pQE-3O-pREP4-FBPS) lysate (lanes 1) and 165 ng of purified FBPS (lanes 2). Convalescent serum raised against S. suis strain 10 was used to test immunogenicity of FPBS present in 4:1 of E. coli M15 (pQE-30-pREP4-FBPS) lysate and 0.5 μg of purified FBPS (Panel C, lanes 1 and 2). Arrowhead, 64 kDa FPBS; Mw, molecular weight marker.
  • FIG. 3 depicts the binding studies with purified FBPS. Panels A and B were probed with 5 μg/ml of fn (A) or fgn (B). [0031] Lanes 1 contain 500 ng of purified FBPS, lanes 2 contain 500 ng of BSA. Panels C and D, lanes 3 and 4 contain 500 ng of purified FBPS. Lanes 3 were probed with 20 μg/ml of fn (C) or fgn (D), lanes 4 were incubated with conjugate without fn or fgn. Panels E and F were probed with 20 μg/ml of fn (E) or fgn (F). Lanes 5 contain 1.8 μg of purified FBPS digested with enterokinase, lanes 6 contain 500 ng of purified FBPS. The closed arrowhead indicates 64 kDa FBPS; the open arrowhead indicates approximately 55 kDa FBPS without 6×HIS.
  • FIG. 4 shows the distribution of fbps among various [0032] S. suis serotypes. 1 μg of chromosomal DNA was spotted onto nitrocellulose membrane and hybridized with a 32P-labelled fbps probe. Serotypes were spotted as indicated. S10: S. suis serotype 2, MRP+EF+; T15: S. suis serotype 2 MRPEF; S17:S. suis serotype 2 MRP+EF*.
  • FIG. 5 illustrates the efficiency of colonization of wild-type and mutant bacteria on various organs of infected pigs. Panel A depicts colonization of the wild-[0033] type strain 10 and the mutant strain 10ΔFBPS of the tonsils. A closed diamond symbol is tonsil pig no. 4664; ▪ tonsil pig no. 4665; ▴ tonsil pig no 4666; • tonsil pig no. 4668. Panel B depicts colonization of the specific organs. Open and closed diamond symbols are pus from joints pig no. 4664; ▴ pus from joint pig no. 4666; • CNS pig no. 4668. Each dot represents the numbers of wild-type or mutant bacteria isolated from one particular organ, from one piglet.
  • DETAILED DESCRIPTION
  • [0034] Streptococcus suis is an important cause of meningitis, septicemia, arthritis and sudden death in young pigs (Clifton-Haclley, 1983; Vecht et al., 1985). It can also cause meningitis in man (Arends and Zanen, 1988). Attempts to control the disease are still hampered by the lack of sufficient knowledge about the pathogenesis of the disease, the lack of effective vaccines and sensitive diagnostic methods. To meet these shortages, it is necessary to identify the genes that are involved in the pathogenic process. So far, only a limited number of S. suis genes are known (Smith et al., 1992; Smith et al., 1993; Serhir et al., 1997; Segers et al., 1998; Smith et al., 1999; and accession nos. AF106927, Z95920 and A57222) and of these, only a few are putatively involved in virulence (Smith et al., 1992; Smith et al., 1993; Jacobs et al., 1994; Gottschalk et al., 1995; Segers et al., 1998; Smith et al., 1999). Previously, putative virulence factors have been identified after growth of the bacteria in standard laboratory media. However, it is known that many important virulence factors are environmentally regulated and are induced at specific stages of the infection process (Mahan et al., 1993).
  • Recently, several approaches have been reported that allow the identification of genes that are specifically expressed in the host. Examples are signature-tagged mutagenesis (STM) and in vivo expression technology (IVET; Mahan et al., 1993; Carnilli and Mekalanos, 1995; Hensel et al., 1995; Mahan et al., 1995; Mei et al., 1997; Young and Miller, 1997; Chiang and Mekalanos, 1998; Coulter et al., 1998; Lowe et al., 1998; Polissi et al., 1998; Camacho et al., 1999; Darwin et al., 1999; Edeistein et al., 1999; Fuller et al., 1999; Zhao et al., 1999). In addition, important virulence proteins could also be identified by the selection of genes specifically expressed under conditions mimicking in vivo conditions, for example by growth in iron-restricted conditions (Litwin and Calderwood, 1993; Martinez et al., 1990). [0035]
  • The present invention identifies virulence genes of [0036] S. suis by selecting environmentally regulated genes by experimental infections of piglets and by the use of iron-restricted conditions in vitro. For this purpose, chromosomal DNA fragments of S. suis were cloned in a plasmid in front of a promoterless erythromycin-resistance gene. Subsequently, the library was used for the selection of bacteria in which erythromycin resistance was induced under iron-restricted conditions. In addition, erythromycin-resistant bacteria were selected after infection of piglets with the library and treatment of the piglets with erythromycin. Pigs were used instead of mice for these experiments since it was recently shown that virulence of S. suis is different in these two animal species (Vecht et al., 1997). Using this approach, 18 unique iron-restriction-induced (iri) genes, as well as 22 unique in vivo selected (ivs) genes, were identified, several of which are putatively involved in virulence (Smith et al., 1993; Smith et al., 1996).
  • Methods. [0037]
  • Bacterial strains and growth conditions. The bacterial strains and plasmids used in this study are listed in Table 1. [0038] S. suis strains were grown in Todd-Hewitt broth (Oxoid), and plated on Columbia agar (Oxoid) containing 6% (v/v) horse blood. For the selection of genes induced in iron-limited conditions, S. suis cells. were plated on agar plates containing Todd-Hewitt medium, 5% (w/v) yeast extract and 75 μM deferoxamine mesylate (Sigma). Control plates were supplemented with 38 μM FeSO4.7H2O (Sigma). If required, antibiotics were added at the following concentrations: 100 μg spectinomycin ml−1and 1 μg erythromycin ml−1 . E. coli strains were grown in Luria broth (Miller, 1972) and plated on Luria broth containing 1.5% (w/v) agar. If required, 50 μg ampicillin ml−1 or 50 μg spectinomycin ml−1 was added.
  • Construction of pIVS-E. [0039]
  • The IVS selection vector used in this study comprises a spectinomycin-resistance gene, a promoterless erythromycin-resistance gene and the origin of replication of the plasmid pWVO1 (Van der Vossen et al., 1987). To construct this pIVS-E, the spectinomycin-resistance gene was amplified from pKUN19-spc (Konings et al., 1987; Smith et al., 1995). In a PCR reaction, the [0040] primers 5′-TGCATGCATGGATCCATCGA TTTTCGTTCG-3′ (SEQ ID NO: 2) and 5′-CGAGCTCGGTACCTGATTACCAATTAGAAT-3′ (SEQ ID NO: 3), which contained NsiI and SacI restriction sites at their respective 5′-ends were used. The obtained PCR product was digested with NsiI and SacI and ligated into pGKV210 (Van der Vossen et al., 1987) that had been digested with SacI (partially) and NsiI. The resulting plasmid was designated pGKV210-spc. pE194 (Horinouchi and Weisblum, 1982) was used as a template for the amplification of a promoterless erythromycin-resistance gene. To do this, the primers 5′-GGGTCGACCCTATAACCAAATTAAAGAGGG-3′ (SEQ ID NO: 4) and 5′-CCCAAGCTTGGGCAGTTTATGCATCCCTTAAC-3′ (SEQ ID NO: 5) were used in a PCR reaction. These primers contained SalI and HindIII restriction sites at their respective 5′-ends. The amplified fragment was digested with SalI and HindIII and the fragment was ligated into pGKV210-spc that had been digested with SalI and HindIII. The resulting plasmid was designated pIVS-E. To construct pIVS-PE, the promoter region of the mrp gene was inserted into pIVS-E 5′ to the promoterless erythromycin-resistance gene. The promoter region of the mrp gene was amplified by PCR from pMRP11 (Smith et al., 1992) using the primers 5′-CCCAAGCTTGGGAATTCATAATGTTTTTTTGAGG-3′ (SEQ ID NO: 6) and 5′-GCGTCGACATCTACGCATAAAAAATCCCCC-3′ (SEQ ID NO: 7). These primers contained EcoRI and SalI sites at their respective 5′-ends. Amplified DNA was digested with EcoRI and SalI and the resulting fragment was ligated into EcoRI and SalI-digested pIVS-E.
  • Construction of Genomic [0041] S. suis Libraries in pIVS-E.
  • Alu I partial digests of [0042] S. suis serotype 2 strain 10 DNA were size fractionated (500-1000 bp) on a 0.8% (w/v) agarose gel. The purified fragments were ligated to SmaI and calf intestinal phosphatase digested pIVS-E and the ligation mixtures were transformed to E. coli XL2-blue cells. Spectinomycin-resistant colonies were selected. Analysis of the transformants by PCR showed that more then 80% contained an insert. From 15 pools of about 2000-3000 independent E. coli transformants, plasmid DNA was isolated. This plasmid DNA was subsequently used for the electrotransformation of S. suis strain 10 (Smith et al., 1995). This resulted in approximately 30,000 independent S. suis transformants. The transformants were pooled and stored at −80° C.
  • DNA Techniques. [0043]
  • Routine DNA manipulations and PCR reactions were performed as described by Sambrook et al. (1989). DNA sequences were determined on a 373A DNA Sequencing System (Applied Biosystems). Samples were prepared by using the ABI/PRISM dye terminator cycle sequencing ready reaction kit (Applied Biosystems). Custom-made sequencing primers were purchased from Life Technologies. Sequencing data were assembled and analyzed using the McMollyTetra software package. The BLAST program was used to search for protein sequences similar to the deduced amino acid sequences. [0044]
  • PCR reaction mixtures (50 μl) contained 10 mM Tris-HCl, pH 8.3, 1.5 mM MgCl[0045] 2, 50 mM KCl, 0.2 mM of each of the four deoxynucleotide triphosphates, 1 μM of each of the primers and 1 U of AmpliTaq Gold DNA polymerase (Perkin Elmer Applied Biosystems). DNA amplification was carried out in a Perkin Elmer 9600 thermal cycler and the program included an incubation for ten minutes at 95° C. and 30 cycles of one minute at 95° C., two minutes at 56° C. and two minutes at 72° C.
  • Assessment of Erythromycin Levels in Treated Piglets. [0046]
  • One-week-old specific pathogen-free (SPF) piglets were treated orally with erythromycin stearate (Abbott, 20 or 40 mg body weight kg[0047] −1) or intramuscularly with erythromycin (Erythrocin 200; Sanofi Sante, 20 or 40 mg body weight kg−1). Blood samples were collected 3 hours, 6 hours or 24 hours after the administration of the antibiotics to determine erythromycin levels.
  • Experimental Infections. [0048]
  • Gnotobiotic Great Yorkshire and Dutch Landrace crossed piglets were obtained from sows by cesarian section. The surgery was performed in sterile flexible film isolators. The piglets were allotted to groups, each having 4 piglets, and were housed in sterile stainless steel incubators. Housing conditions and feeding regimens were as described (Vecht et al., 1989; Vecht et al., 1992). One-week-old piglets were inoculated intravenously with [0049] S. suis strain 10 (pIVS-E), 10 (pIVS-PE) or 10 (pIVS-RE) as described (Vecht et al., 1989; Vecht et al., 1992, Table 3). Two hours after infection, the pigs were injected intramuscularly with erythromycin for the first time and thereafter received erythromycin twice a day: once intramuscularly (Erythrocin, 40 mg body weight kg−1) and once orally (erythromycin stearate, 40 mg body weight kg−1).
  • Piglets were monitored twice a day for clinical signs of disease, such as fever, nervous signs and lameness. Blood samples were collected three times a week from each pig. Leukocyte concentrations were determined using a conducting counter (Contraves A. G., Switzerland). To monitor infection with [0050] S. suis and to check for absence of contaminants, swabs of the nasopharynx and of feces were collected daily. The swabs were directly plated onto Columbia agar containing 6% (v/v) horse blood. After the piglets were killed, they were examined for gross pathological changes. Tissue specimens were collected from the central nervous system, serosae, joints, lungs, heart and tonsils. The tissues were homogenized in the presence of Todd-Hewitt medium using an Ultra-Turrax tissuemizer (Omni International) and frozen at −80° C. in the presence of 15% (v/v) glycerol.
  • Results. [0051]
  • Promoter Selection System. [0052]
  • The plasmid pIVS-E was constructed to allow introduction of [0053] S. suis DNA fragments into a number of unique restriction sites in front of a promoterless erythromycin-resistance resistance gene. The plasmid carries the origin of replication of pWVO1, which functions in E. coli and in S. suis (Smith et al., 1995). S. suis strain 10 cells containing pIVS-E were sensitive to 1 μg erythromycin ml−1 on agar plates. In pIVS-PE the promoter of the mrp gene of S. suis (Smith et al., 1992), which is highly expressed in vivo as well as in vitro, drives expression of the erythromycin-resistance gene. S. suis strain 10 cells containing pIVS-PE were resistant to high concentrations of erythromycin (>256 μg erythromycin ml−1) on agar plates. A S. suis DNA library in pIVS-E (pIVS-RE) was constructed and 30,000 individual S. suis clones or mutants were obtained. As determined by analysis of 24 randomly selected transformants, more than 80% of these clones or mutants contained an insert (results not shown). Moreover, 2% of the clones were resistant to 1 μg erythromycin ml−1 on agar plates indicating the presence of some promoter sequences that were functional in vitro.
  • Selection of Promoters Induced Under Iron-Restricted Conditions. [0054]
  • Gene sequences that were specifically induced on agar plates under iron-restricted conditions were selected. For this purpose, about 96,000 c.f.u. were plated under iron-limiting conditions on agar plates containing deferoxamine mesylate and erythromycin. The 1500 colonies that grew on these plates were inoculated onto plates containing erythromycin, deferoxamine mesylate and FeSO[0055] 4. Twenty-four clones showed reduced growth in the presence of FeSO4. The inserts of the 24 selected iri clones were amplified by PCR using primers complementary to the 5′ ends of the erythromycin- and spectinomycin-resistance genes and the nucleotide sequences of these fragments were determined. The sequence data showed that the 24 clones contained 18 unique sequences. The 18 sequences were analyzed for similarity to known genes by comparison with the sequences in the GenBank/EMBL and SWISSPROT databases. One sequence, iri3I, was identical to cps2A, a previously identified S. suis gene putatively involved in the regulation of capsule expression (Smith et al., 1999). Fourteen iri sequences were similar to sequences of known, non-S. suis, genes. Three of these sequences (iri2 (SEQ ID NO: 15), iri1, 6 and 22 (SEQ ID NO: 8), and iri34 (SEQ ID NO: 21) were similar to sequences of environmentally regulated genes previously selected by applying the IVET to V. cholerae (Camilli and Mekalanos, 1995), S. aureus (Lowe et al., 1998) and P. aeruginosa (Wang et al., 1996), respectively. One, contained in iri1, 6, and 22 (SEQ ID NO: 8), was similar to the agrA gene of Staphylococcus aureus, a key locus involved in the regulation of numerous virulence proteins. Three iri sequences had no significant similarity to any sequences in the databases (Table 2).
  • Conditions for Selection of Promoter Sequences in Piglets. [0056]
  • To determine the antibiotic treatment regime required for a successful selection of in vivo-expressed promoter sequences, piglets were treated with different concentrations of erythromycin once a day. The erythromycin was administered either orally or intramuscularly. Levels of erythromycin in sera were determined 3, 6 or 24 hours after treatment over one week. High erythromycin levels were detected three hours and six hours after both treatments (results not shown). However, 24 hours after the treatments, the levels decreased dramatically. Based on these data, we hypothesized that for efficient promoter selection, it was necessary to treat the animals twice a day with erythromycin (40 mg kg[0057] −1), once intramuscularly (at 9 a.m.) and once orally (at 4 p.m.).
  • To test this hypothesis, pigs were inoculated either with [0058] S. suis strain 10 (pIVS-PE) or with strain 10 (pIVS-E). In pIVS-PE, the promoter of the mrp gene of S. suis (Smith et al., 1992), which is highly expressed in vivo as well as in vitro, drives expression of the erythromycin-resistance gene. The control plasmid, pIVS-E, does not contain a promoter in front of the erythromycin-resistance gene. The strains were inoculated intravenously or intranasally. All pigs infected with strain 10 (pIVS-PE) showed specific S. suis symptoms (Table 3) and, except for one, all pigs died in the course of the experiment. Moreover, high numbers of bacteria were isolated from the central nervous system, the serosae and the joints. In contrast, none of the pigs inoculated with strain 10 (pIVS-E) showed specific clinical signs of disease and all survived the infection until the end of the experiment. Moreover, bacteria were not isolated from the central nervous system, the serosae or the joints of these animals. These data demonstrated that in vivo-expressed sequences could be selected from pigs using the applied antibiotic treatment regimen.
  • Selection of Gene Sequences Expressed in Vivo in Piglets. [0059]
  • Piglets were inoculated intravenously with different doses (5×10[0060] 5 to 5×108 c.f.u.) of the S. suis library (Table 3) and treated with erythromycin as described herein. Specific signs of disease developed in all animals three to eight days after infection (Table 3). High numbers of bacteria were recovered from tissues (central nervous system, joints, serosae, lung, liver, spleen, heart and kidney) of the individual piglets. Analysis of the recovered bacteria showed that a limited number of different clones were present in each of the bacterial samples isolated from the diseased pigs. For example, 30 randomly selected clones from the joints of one pig all possessed identical DNA inserts as assessed by PCR and DNA sequence analysis (results not shown). In addition, at 80% of the 62 sample sites analyzed, four randomly selected clones were identical. However, from different tissues of a single animal, different clones or mutants could be isolated. On the other hand, identical clones could be isolated from different, as well as from corresponding, tissues of different animals. These findings indicated that a limited number of clones had been selected in vivo and were greatly enriched in the affected tissues. The observed selection was not tissue specific. Further, none of the selected clones failed to grow on agar plates that contained 1 μg erythromycin ml−1.
  • Two-hundred forty-five clones were analyzed by PCR and partial sequence analysis. Among these, 22 unique ivs clones were found. The 22 sequences were analyzed for similarity to sequences of known genes by comparison with the GenBank/EMBL and SWISSPROT databases (Table 4). The sequences of two genes showed similarity to genes encoding putative virulence factors: ivs21, 26 and 30 which was identical to the epf gene, a previously identified [0061] S. suis gene, putatively involved in virulence (Smith et al., 1993; Smith et al., 1996); and ivs31 (SEQ ID NO: 37), which was similar to the fibronectin-binding protein of S. gordonii. Moreover, the sequences of two ivs genes (ivs25 (SEQ ID NO: 24) and ivs6, 7, 13 and 14 (SEQ ID NO: 43)) were homologous to two environmentally regulated ivi genes, previously identified using IVET selection in other bacterial species (Camilli and Mekalanos, 1995; Lowe et al., 1998). Four ivs sequences (ivs25 (SEQ ID NO: 34); ivs23 and 24 (SEQ ID NO: 33), ivs2, 4 and 28 (SEQ ID NO: 31); and ivs6, 7, 13 and 14 (SEQ ID NO: 43)) were also found when the library was selected using iron-restricted conditions. The remainder of the sequences showed similarity to sequences of known, non-S. suis genes, including two genes showing similarity to mobile elements and five genes showing similarity to genes of unknown function.
  • The identification of environmentally regulated genes of [0062] S. suis serotype 2 by the use of iron-restricted conditions and by experimental infection of piglets is described. Eighteen unique iri genes and 22 unique ivs genes were found. None of the ivs genes was exclusively expressed in vivo. Four iri genes were identical to four clones selected in vivo. The selected gene sequences encode for potential virulence factors, expand our knowledge about the pathogenesis of S. suis infections in pigs and are of value in control of the disease either by the development of effective vaccines or by the development of new diagnostic methods. A promoter trap was used to identify environmentally regulated S. suis genes expressed under specific conditions, i.e., during iron-restriction or during experimental infection. This system differs from the antibiotic-based IVET system described for S. typhimurium (Mahan et al., 1995) in two ways. One is that the lacZ reporter gene fusion is omitted in our vector constructions because inclusion of the lacZ gene resulted in structural instability of the vector. The other difference is that a plasmid system was used rather than a chromosomal integration system. A plasmid system was used because the low transformation efficiency of S. suis (Smith et al., 1995) might prevent the generation of a complete gene library using a chromosomal integration system.
  • From the data, it is evident that a number of inducible and environmentally regulated sequences were selected. Four iri genes were identical to four ivs genes. Because most bacteria require iron for their growth and because there is a limited amount of free iron available within the host (Payne, 1993), it might be expected that the expression of some ivs genes is regulated by iron. With the in vivo selection system, tissue-specific colonization was not observed: clones isolated from one piglet were also isolated from other piglets from corresponding as well as from different tissues. This might be due to the mechanisms involved in the molecular pathogenesis of [0063] S. suis infections in pigs. Furthermore, it was striking and different from the observations made with IVET systems that only a limited number of clones could be selected. In addition, we were not able to demonstrate that we selected for gene sequences that are exclusively expressed in vivo. This could be explained either by the absence of promoter sequences exclusively expressed in vivo among the 22 identified ivs genes, and/or by the inability of this plasmid-based system to identify such sequences due to gene dose effects.
  • A number of interesting genes were selected. Two ivs genes showed similarity to genes encoding putative virulence factors. Ivs21, 26 and 30 were shown to be identical to the epf gene of [0064] S. suis (Smith et al., 1993), which is found in virulent strains of S. suis serotypes 1 and 2 (Stockhofe-Zurwieden et al., 1996; Vecht et al., 1991; Vecht et al., 1992). Ivs31 (SEQ ID NO: 37) showed similarity to the fibronectin/fibrinogen-binding protein of S. gordonii (accession no. X65164) and group A Streptococci (Courtney et al., 1994). In Streptococci, fibronectin/fibrinogen-binding proteins play an important role in adhesion to host cells and are considered to be important virulence factors. The selection of these two ivs genes demonstrated the selectivity of the system and might be indicative for the relevance of the other ivs genes in the pathogenesis of S. suis infections in pigs. The performance of the system was further demonstrated by the observation that two ivs genes, ivs25 (SEQ ID NO: 34) and ivs6, 7, 13 and 14 (SEQ ID NO: 43) showed similarity to environmentally regulated genes previously identified using an IVET selection system in other bacterial species.
  • Ivs25 (SEQ ID NO: 34) showed significant similarity to the sapR gene of [0065] S. mutans (accession no. P72485) and Lactobacillus sake Lb706 (Axelsson and Holck, 1995) as well as to the agrA gene of S. aureus (Projan and Novick, 1997), both of which encode response regulator proteins of bacterial two-component signal-transduction systems, thus mediating the response to an environmental signal (Projan and Novick, 1997). Use of an IVET selection system for S. aureus in mice selected the region preceding the agrA gene, suggesting induction of agrA expression under in vivo conditions (Lowe et al., 1998). Moreover, in S. aureus, the agr locus was shown to play an important role in altering the expression of a considerable number of virulence factors in response to cell density (Projan and Novick, 1997).
  • Clones ivs6, 7, 13 and 14 (SEQ ID NO: 43) showed similarity to a gene, iviVI, previously identified by IVET selection in [0066] V. cholerae (Camilli and Mekalanos, 1995). The function of iviVI is unknown. However, the genes showed similarity to members of the ATP-binding cassette family of transporters. The sequenced portion of ivs6, 7, 13 and 14 (SEQ ID NO: 43) included an N-terminal ATP-binding Walker A box motif, which is highly conserved in this transporter family.
  • Four ivs genes were identical to four iri genes. The first gene, ivs23 and 24 (SEQ ID NO: 33), which is identical to iri24 (SEQ ID NO: 17), showed similarity to cpsY of [0067] S. agalactiae (Koskiniemi et al., 1998) and to oxyR of various organisms (Demple, 1999). CpsY of S. agalactiae is involved in the regulation of capsule expression and environmental induction of expression of the cpsY gene has been suggested by Koskiniemi et al. (1998). In S. suis, ivs23 and 24 (SEQ ID NO: 33).and iri24 (SEQ ID NO: 17) are not linked to the capsular locus (Smith et al., 1999). The oxyR gene is the central regulator of oxidative stress response in E. coli (Demple, 1999) and approximately ten genes are under the control of the OxyR protein. The second gene, ivs2, 4 and 28 (SEQ ID NO: 31), which is identical to iri10 and 20 (SEQ ID NO: 9), showed similarity to the yoaE gene of E. coli (accession no. P76262), a putative ABC transporter protein. The third and the fourth genes, ivs25 (SEQ ID NO: 34) and ivs6, 7, 13 and 14 (SEQ ID NO: 43) were identical to iri1, 6 and 22 (SEQ ID NO: 8) and iri2 (SEQ ID NO: 15), respectively. These genes also showed similarity to ivi genes selected using IVET in other bacterial species.
  • Based on data presented by Niven et al. (1999), selection of iri genes of [0068] S. suis is not expected. The authors described that S. suis does not require iron for growth. However, in their studies the authors used media reduced from iron by using ethylenediamine di-o-hydroxyphenylacetic acid (EDDA). Therefore, the different conditions used in vitro may explain the different results obtained.
  • Two of the [0069] S. suis ivs genes, ivs1 (SEQ ID NO: 25) and ivs8 (SEQ ID NO: 44), showed similarity to transposon sequences. Moreover, one S. suis ivs gene, ivs2, 4 and 28 (SEQ ID NO: 31), had a GC% that was considerably higher than the composition of the rest of the selected genes. It is striking that in S. typhimurium, several of the ivi clones that are required for full virulence have been found to be associated with mobile elements. Their atypical base composition and codon usage has led to the suggestion that they have been acquired from other bacterial species by horizontal transfer (Conner et al., 1998).
  • Our screen also identified five ivs genes that showed similarity to sequences encoding proteins of unknown function. These genes are not standard housekeeping or metabolic genes. [0070]
  • Besides the four ivs/iri genes, a considerable number of other iri genes have been selected in this study by plating the library under iron-restricted conditions. Interestingly, one of the selected iri genes, iri31, is identical to the cps2A gene of [0071] S. suis. This gene was previously isolated as a part of the capsular locus of S. suis serotype 2 (Smith et al., 1999) and was implicated in the regulation of capsular polysaccharide biosynthesis (Kolkman et al., 1997; Smith et al., 1999). Moreover, because the capsule of S. suis is expressed in larger size after in vivo growth when compared to growth in vitro (Quessy et al., 1994), regulated expression of cps2A might be expected. Another iri gene, iri7 (SEQ ID NO: 23), showed similarity to the rpgG gene of S. mutans. This gene was shown to be required for the biosynthesis of rhamnose-glucose polysaccharide (Yamashita et al., 1999). Because rhamnose is part of the polysaccharide capsule in S. suis serotype 2 (Elliott and Tai, 1978), a role of the iri7 (SEQ ID NO: 23) gene in capsule biosynthesis can be proposed. Iri34 (SEQ ID NO: 21) showed similarity to the np 16 gene, previously identified using IVET selection in P. aeruginosa and suspected to encode threonine dehydratase activity (Wang et al., 1996). Together with the observation that 4 iri genes could be selected by the in vivo approach, these data show that the iri genes encode important virulence factors for S. suis.
  • Contribution of Fibronectin-Binding Protein to Pathogenesis of [0072] Streptococcus suis Serotype 2.
  • [0073] Streptococcus suis causes severe infections, such as meningitis, septicemia, and arthritis, in piglets. The animals often do not survive the infection (6, 28). Occasionally, S. suis causes septicemia and meningitis in humans (3). The pathogenesis of an S. suis infection is rarely understood. Sows are symptomless carriers of S. suis on their tonsils and pass the bacteria on to their piglets. The piglets cannot cope with the bacteria and subsequently develop the specific symptoms of an S. suis infection. Until now, 35 capsular serotypes of S. suis have been described (26), but serotype 2 strains are most often isolated from diseased piglets. The capsule is an important virulence factor since piglets infected with an acapsular mutant of S. suis serotype 2 strains do not develop any clinical symptoms (22). Bacterial proteins have been suggested to play a role in the pathogenesis as well (2, 26). The expression of murimidase-released protein (MRP), extracellular factor (EF) and suilysin was shown to be strongly associated with pathogenic strains of S. suis serotype 2 (1, 29, 30). Since isogenic mutants lacking MRP and EF and isogenic mutants lacking suilysin were still pathogenic in young piglets, these proteins are not absolutely required for virulence (2, 23). Recently, a new virulence factor was identified (21) by using a complementation approach. The function of this virulence factor in the pathogenesis has to be further investigated.
  • Many important virulence factors are environmentally regulated and are induced at specific stages of the infection process (15). To identify these genes in [0074] S. suis, promoters and their downstream sequences that are “on” during experimental S. suis infection of piglets (20) were cloned. Twenty-two in vivo selected (ivs) genes were found. Two of the ivs genes were directly linked to virulence since homology was found to genes in the database that encode for known virulence factors. One of these ivs genes (ivs-21) was identical to the epf gene of virulent S. suis serotype 2 strains (30). The other (ivs-31) (SEQ ID NO: 37) showed homology to genes encoding fibronectin-/fibrinogen-binding proteins of Streptococcus gordonii (GenBank accession no. X65164) and Streptococcus pyogenes FBP54 (8). A considerable number of fibronectin-binding proteins of various bacterial species have been shown to be important virulence factors (12). In S. pyogenes, FBP54 was shown to be expressed in the human host and to preferentially mediate adherence to human buccal epithelial cells (7). It was shown that the FBP54 protein induces protective immunity against S. pyogenes challenge in mice (13).
  • A fibronectin-/fibrinogen-binding protein of [0075] S. suis (FBPS) is described herein and the sequence of fbps was determined. Binding studies showed that purified FBPS bound fibronectin and fibrinogen. A contribution of FBPS to the pathogenesis of S. suis serotype 2 was found.
  • Materials and Methods. [0076]
  • Bacterial Strains and Growth Conditions. [0077]
  • The bacterial strains and plasmids used in this study are listed in Table 5. [0078] S. suis strains were grown in Todd-Hewitt broth (code CM 189; Oxoid, Ltd.) and plated on Columbia blood base agar plates (code CM331; Oxoid, Ltd., London, United Kingdom), containing 6% (vol/vol) horse blood. E. coli strains were grown in Luria Broth (17) and plated on Luria Broth containing 1.5% (wt/vol) agar. If required, antibiotics were added at the following concentrations: 50 μg/ml of spectinomycin (Sigma, St. Louis, Mo.) for E. coli and 100 μg/ml for S. suis, 100 μg/ml of ampicillin (Boehringer, Mannheim, Germany) for E. coli and 25 μg/ml of kanamycin (Boehringer) for E. coli.
  • DNA Techniques and Sequence Analysis. [0079]
  • Routine DNA manipulations were performed as described by Sambrook et al. (19). DNA sequences were determined on a 373A DNA Sequencing System (Applied Biosystems, Warrington, Great Britain). Samples were prepared by use of an ABI Prism dye terminator cycle sequencing ready reaction kit (Applied Biosystems). Sequencing data were assembled and analyzed using the Lasergene program (DNASTAR). The BLAST software package was used to search for protein sequences homologous to the deduced amino acid sequences in the GenBank/EMBL databases. [0080]
  • Southern Blotting and Hybridization. [0081]
  • Chromosomal DNA was isolated as described by Sambrook et al. (19). DNA fragments were separated on 0.8% agarose gels and transferred to GeneScreen Plus hybridization transfer membrane (NEN™ Life Science Products, Boston, USA) as described by Sambrook et al. (19). DNA probes of the fbps and spc genes were labeled with (α-[0082] 32P)dCTP (3,000 Ci/mmol; Amersham Life Science, Buckinghamshire, Great Britain) by use of a random primed DNA labeling kit (Boehringer). The DNA on the blots was pre-hybridized for at least 30 minutes at 65° C. and subsequently hybridized for 16 hours at 65° C. with the appropriate DNA probes in a buffer containing 0.5 M sodium phosphate (pH 7.2), 1 mM EDTA and 7% sodium dodecyl sulphate. After hybridization, the membranes were washed twice with a buffer containing 40 mM sodium phosphate (pH 7.2), 1 mM EDTA and 5% sodium dodecyl sulphate for 30 minutes at 65° C. and twice with a buffer containing 40 mM sodium phosphate (pH 7.2), 1 mM EDTA and 1% sodium dodecyl sulphate for 30 minutes at 65° C. The signal was detected on a phosphor-imager (Storm; Molecular Dynamics, Sunnyvale, Calif.).
  • Construction of a fbps Knock-Out Mutant. [0083]
  • To construct the mutant strain 10ΔFBPS, the pathogenic strain 10 (27, 29) of [0084] S. suis serotype 2 was electrotransformed (24) with the plasmid pFBPS7-47. In this plasmid, the fbps gene was inactivated by the insertion of a spectinomycin-resistance gene. To create pFBPS7-47 (FIG. 1), the 382 bp SalI-SalI fragment of pFBPS7-46 was replaced by the 1.2 kb EcoRV-SmaI fragment of pIC-Spc, containing the spectinomycin resistance gene, after the SalI sites of the vector were made blunt (FIG. 1). After electrotransformation of strain 10 with pFBPS7-47, spectinomycin-resistant colonies were selected on Columbia agar plates containing 100 μg/ml of spectinomycin. Southern blotting and hybridization experiments were used to select for double cross-over integration events (data not shown).
  • FBPS Expression Construct. [0085]
  • To construct an FBPS expression plasmid, the QIAexpress Kit (Qiagen GmbH, Hilden, Germany) was used. The primers corresponded to positions 250 to 272 and from 1911 to 1892 of the fbps gene. The sequences of these primers were 5′(GCGGATCCGATGACGATGACAAATCTTTTGACGGATTTTTTTTAC)3′ (SEQ ID NO: 46) and 5′(CCCAAGCTTGGGCATGAACTAGATTTTCATGG)3′ (SEQ ID NO: 47). The primers contained restriction sites for BamHI and HindIII, respectively, to amplify the fbps gene from pFBPS7-47. The amplified PCR product was digested with BamHI and HindIII and the 1.8 kb fbps gene was cloned into pQE-30 digested with BamHI and HindIII, yielding pQE-30-FBPS. pQE-30-FBPS was transformed to M15 (pREP4). [0086]
  • Purification of FBPS. [0087]
  • M15 (pREP4) (pQE-30-FBPS) was used to express and purify the FBPS using the QIAexpressionist™ (Qiagen). In short, M15 (pREP4) (pQE-30FBPS) cells were grown exponentially; 1 mM IPTG was added and the cells were allowed to grow another four hours at 37° C. Subsequently, cells were harvested and lysed. The cleared supernatants were loaded onto Ni[0088] 2+-NTA agarose columns. FBPS containing a 6×HIS tag was bound to the Ni2++-column. The columns were washed and the protein was eluted. Different buffers were used for native and for denaturing purification. FBPS purified under denaturing conditions was renaturated on a Ni2+-NTA column by using a linear 6 M-1 M urea gradient in 500 mM NaCl, 20% glycerol and 20 mM Tris-HCl (pH7.4), containing protease inhibitors (25 μg/ml of pefabloc, 0.7 μg/ml of pepstatin, 1 μg/ml of aprotinin, 0.5 μ/ml of leupeptin). All procedures were performed according to the manufacturer's recommendations. The 6×HIS tag was removed from the protein by incubating purified FBPS in 20 mM Tris-HCl (pH 7.4), 50 mM NaCl, 2 mM CaCl2 and 0.5 U of light chain enterokinase (New England Biolabs, Beverly, Mass.) for 16 hours at RT.
  • Immunization of Rabbits with FBPS. [0089]
  • Purified and renaturated FBPS was used to immunize two rabbits. To remove urea, the protein was dialyzed against phosphate buffered saline (136 mM NaCl; 2.68 mM KCl; 8.1 mM Na[0090] 2HPO4; 2.79 mM KH2PO4 (pH 7.2)) over night at 4° C. Seven days before immunization, blood was collected from the rabbits to determine the natural titers against FBPS. At day one, those rabbits with negative anti-FBPS titers were immunized intramuscularly with two times 0.5 ml of 100 μg/ml of FBPS in a water-in-oil emulsion (Specol; ID-Lelystad). At day 28, rabbits were immunized for the second time using the same amount of protein and the same route of immunization. Three weeks after the second immunization, the rabbits were sacrificed and blood was collected. The blood was coagulated and serum was collected and used for immunodetection of FBPS.
  • Immunodetection of FBPS. [0091]
  • Proteins were separated by sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis (PAGE) by standard procedures (19). Proteins in the gel were visualized using SYPRO-orange (Molecular Probes, Sunnyvale, Calif.) staining according to the manufacturer's recommendations. Signals were detected on a phosphor imager (Storm; Molecular Dynamics). A known bovine serum albumin concentration range was used as a standard to calculate the amounts of protein present in the gel. The Molecular Dynamics program was used for the calculations. [0092]
  • Proteins were transferred to a nitrocellulose membrane by standard procedures (19). The membranes were blocked in Blotto: Tris-buffered saline (TBS) (50 mM Tris-HCl (pH 7.5), 150 mM NaCl) containing 4% skimmed milk and 0.05[0093] % Tween 20 at room temperature (RT) for one hour. To detect recombinant purified FBPS, membranes were incubated with a monoclonal antibody against the 6×HIS tag (Clontech, Palo Alto, Calif.) in a 1:10,000 dilution in Blotto-TBS (1:1) at RT for one hour, followed by an incubation with alkaline phosphatase-conjugated anti-mouse antibody in a 1:1,000 dilution in Blotto-TBS (1:1) at RT for one hour. Reactivity of purified FBPS was tested by using a convalescent serum of a pig that had survived an S. suis infection. Nitrocellulose membranes were incubated with the polyclonal pig serum in a 1:200 dilution in Blotto-TBS (1:1) at RT for one hour, followed by incubation at RT for one hour with alkaline phosphatase-conjugated anti-swine antibody in a 1:2,000 dilution in Blotto-TBS (1:1). As a substrate, Nitro Blue Tetrazolium (Merck, Darmstadt, Germany) bromochloroindolyl phosphate (Sigma) was used. All washing steps were performed in Blotto-TBS (1:1).
  • Fibronectin and Fibrinogen Binding. [0094]
  • Binding studies were performed by indirect Western blotting. Proteins were separated by SDS-PAGE and transferred to a nitrocellulose membrane as described herein. The membranes were blocked in MPBS: PBS containing 4% skimmed milk and 0.05[0095] % Tween 20. Subsequently, the membrane was incubated with 5 μg/ml of human fibronectin (fn) (Sigma) or 5 μg/ml of human fibrinogen (fgn) (Sigma) in PBS containing 5% fetal calf serum, 2% NaCl, and 0.05% Tween 80 at RT for one hour. To detect bound fibronectin and fibrinogen, the membranes were incubated with horse-radish peroxidase-conjugated anti-fibronectin (DAKO) or anti-fibrinogen (DAKO) antibodies in a 1:1,000 dilution in PBS containing 5% fetal calf serum, 2% NaCl, and 0.05% Tween 80 at RT for one hour. The signal was visualized by using ECL+ (Amersham Pharmacia Biotech, N.J.) according to the manufacturer's recommendations. Signals were detected on a phosphor imager (Storm; Molecular Dynamics). All washing steps were performed in MPBS-PBS (1:1).
  • Experimental Infections. [0096]
  • Germ-free piglets, cross-breeds of Great Yorkshire and Dutch Landrace, were obtained from sows by cesarean section. The surgery was performed in sterile flexible film isolators. Piglets were allotted to groups of four and were housed in sterile stainless steel incubators. Housing conditions and feeding regimens were as described (27, 29). Six-day-old piglets were inoculated intranasally with about 10[0097] 7 cfu of Bordetella bronchiseptica 92932 to predispose the piglets to infection with S. suis. Two days later, the piglets were inoculated intranasally with 106 cfu of S. suis strain 10 plus 106 cfu of S. suis strain 10ΔFBPS. To determine differences in virulence between wild-type and mutant strains, LD50 values should be determined. To do this, large numbers of piglets are required. For ethical reasons, this is not acceptable. To circumvent this problem, co-colonization studies were performed.
  • To monitor for the presence of [0098] S. suis and B. bronchiseptica and to check for absence of contaminants, swabs taken from the nasopharynx and the feces were cultured three times a week. The swabs were plated directly onto Columbia agar containing 6% horse blood or grown for 48 hours in Todd-Hewitt broth and subsequently plated onto Columbia agar containing 6% horse blood. Pigs were monitored twice a day for clinical signs and symptoms, such as fever, nervous signs, and lameness. Blood samples from each pig were collected three times a week. Leukocytes were counted with a cell counter. The piglets were killed when specific signs of an S. suis infection were observed, such as arthritis or meningitis, or when the pigs became mortally ill. The other piglets were killed two weeks after inoculation with S. suis and examined the same way as the piglets that were killed based on their clinical symptoms. All piglets were examined for pathological changes.
  • Tissue specimens from heart, lung, liver, kidney, spleen, and tonsil, and from the organs specifically involved in an [0099] S. suis infection (central nervous system (CNS), serosae, and joints) were sliced with a scalpel or a tissuenizer. Tissue slices from each organ or site were resuspended in 2-25 ml of Todd-Hewitt containing 15% glycerol depending on the size of the tissue slice. The suspension was centrifuged at 3,000 rpm for five minutes. The supernatant was collected and serial dilutions were plated on Columbia agar containing 6% horse blood, as well as on Columbia agar plates containing 6% horse blood and 100 μg/ml of spectinomycin to quantitate the number of wild-type and mutant bacteria present. The number of mutant strain 10ΔFBPS cells was determined by counting the number of CFU on the appropriate serial dilution on the selective plates; the number of wild-type strain 10 cells was determined. by counting the number of CFU on the appropriate serial dilution on the Columbia Agar blood plates of which the number of CFU counted on the selective plates was subtracted. When wild-type and mutant bacteria were found in tissues, the ratio of wild-type and mutant strain was determined again by toothpicking about 100 individual colonies onto both Columbia Agar plates and onto Columbia Agar plates containing 100 μg/ml spectinomycin.
  • All animal experiments were approved by the ethical committee of the Institute for Animal Science and Health in accordance with the Dutch law on animal experiments. [0100]
  • Nucleotide Sequence Accession Number. [0101]
  • The nucleotide sequence data fbps have been submitted to GenBank, in which the sequence is listed under accession no. AF438158. [0102]
  • Results. [0103]
  • Cloning of the [0104] S. suis fbps Gene.
  • One of the in vivo selected genes (ivs-31) (SEQ ID NO: 37) (20) showed homology to the 5′ part of genes encoding for FlpA and FBP54, fibronectin-binding proteins (FnBP) of [0105] Streptococcus gordonii (GenBank accession no. X65164) and Streptococcus pyogenes (8), respectively. To clone the entire fbps gene of S. suis, ivs31 (SEQ ID NO: 37) was used as a probe to identify a chromosomal DNA fragment of S. suis serotype 2 containing flanking fbps sequences. A 5 kb EcoRI fragment was identified and cloned in pGEM7Zf(+) yielding pFBPS7-46 (FIG. 1). Sequence analysis revealed that this fragment contained the entire fbps gene of S. suis serotype 2.
  • An open reading frame of 1659 bp coding for a polypeptide of 553 amino acids was found. The putative ATG start codon is preceded by a sequence similar to ribosome binding sites of gram-positive bacteria. Further upstream, two putative promoter sequences could be identified. Upstream of these promoter sequences of fbps, a direct repeat was found that could serve as a transcription terminator of the gene located 5′ of fbps. Downstream of fbps, a gene that showed homology to an alpha-acetolactate decarboxylase was found. This gene is transcribed in the opposite direction of fbps. The deduced amino acid sequence was aligned with that of several previously identified FnBPs from other bacteria. As expected, FBPS was substantially homologous to FlpA of [0106] S. gordonii (76%) and also showed homology to FnBP's of other organisms, like Streptococcus pneumoniae (73%), S. pyogenes (69%), Lactococcus lactis (59%), and Bacillus subtilis (41%). Compared to the sequence of FBP54, FBPS has a longer N-terminus with 76 additional amino acids. This longer N-terminus was also seen in other organisms like S. gordonii, S. pneumoniae and B. subtilis. In FBP54, the primary fibronectin-/fibrinogen-binding domain was localized to its N-terminal part, to the first 89 amino acids (8). Over this region, the homology of FBPS to FBP54 is very high (80%), suggesting that FBPS can bind both fibronectin and fibrinogen.
  • Binding of FBPS to Fibronectin and Fibrinogen. [0107]
  • To confirm the binding of FBPS of [0108] S. suis to fibronectin (fn) and fibrinogen (fgn), FBPS was purified under native conditions. A protein expression construct, which expresses FBPS with a 6×HIS tag fused to the N-terminus, was used for purification. Four hundred μg of FBPS was purified from 50 ml of exponential-phase E. coli cells after induction with IPTG. The purity of this FPBS was determined with SDS-PAGE and Western blotting (FIG. 2). The induced E. coli lysate contained a broad range of proteins, among which the 64 kDa protein FBPS was present (panel A, lane 1). After purification, highly purified FBPS with 6×HIS tag was obtained (panel A, lane 2). When both samples were incubated with a monoclonal antibody against the 6×HIS tag, FBPS was the only protein that was detected (panel B).
  • To determine whether FBPS binds fn and fgn, a Western blot containing purified FBPS was incubated with soluble human fn and human fgn (FIG. 3, panels A and B). Specific binding of fn and fgn to FBPS was detected. No binding of fn and fgn to BSA, a negative control protein, was observed. To exclude possible background signals due to immunoglobulin-binding of FBPS, the same experiment was performed without addition of fibronectin or fibrinogen. No binding was found (FIG. 3, panels C and D) indicating that the binding was specific for fibronectin and fibrinogen. To control whether the binding of fn and fgn to FBPS was not mediated by the 6×HIS tag, the tag was removed by an enterokinase treatment. FIG. 3, panels E and F, show that FBPS without the 6×HIS tag still efficiently bound to fn and fgn. Therefore, it appears that FBPS can specifically bind to fn and fgn. [0109]
  • Immunogenicity of FBPS. [0110]
  • Since it was shown that FBP54 induced a protective immune response in mice against a lethal dose of [0111] S. pyogenes (13), it was determined whether purified FBPS was recognized by convalescent serum of a pig that survived an S. suis infection. As shown in FIG. 2 panel C, the FBPS reacted with this anti-serum. When the same experiment was performed with non-immune serum of an SPF piglet, no band of the size of FBPS was detected (data not shown). These findings indicate that FBPS is expressed in vivo and that the protein is indeed immunogenic in young pigs.
  • Distribution of the fbps Gene Among the 35 [0112] S. suis Serotypes.
  • Since we were interested in a cross-protective vaccine candidate, the presence of the fbps gene among the various [0113] S. suis serotypes was analyzed. Ivs-31 (SEQ ID NO: 37), the clone containing the promoter and the 5′-part of the fbps gene, was radiolabeled and chromosomal DNA of the reference strains of the 35 different S. suis serotypes was hybridized with this probe. The three different phenotypes of S. suis serotype 2, a pathogenic, a non-pathogenic and a weak pathogenic strain, were included in this study. The fbps gene was present in all S. suis serotypes and phenotypes except for serotypes 32 and 34 (FIG. 4).
  • Role of FBPS in Pathogenesis. [0114]
  • To test the role of FBPS in the pathogenesis of [0115] S. suis, an isogenic knock-out mutant of FBPS was constructed in strain 10, strain 10ΔFBPS. Since upstream of fbps a direct repeat was found that could serve as a transcription terminator and downstream of fbps a gene showing homology to an alpha-acetolactate decarboxylase was found that is transcribed in the opposite direction, polar effects to genes upstream or downstream of fbps are not expected. To verify that the mutant strain 10ΔFBPS did not produce FBPS, protoplasts of strain 10 and strain 10ΔFBPS were subjected to SDS-PAGE and Western blotting. FBPS was detected using a polyclonal antiserum raised against purified FBPS. It was shown that strain 10ΔFBPS expressed no FBPS, while strain 10 did express FBPS (data not shown). Subsequently, the virulence of this mutant strain was tested in an experimental infection in piglets.
  • The mutant strain 10ΔFBPS was used in a competition challenge experiment with the wild-type strain to determine the relative attenuation of the mutant strain. Using in vitro conditions, the growth rates of the wild-type and mutant strain in Todd-Hewitt medium were found to be essentially identical (data not shown). Wild-type and mutant strain were inoculated at an actual ratio of 0.65 (1.63×10[0116] 6 cfu of wild-type bacteria ml−1 and 3.09×106 CfU of mutant bacteria ml−1). During the experiment, piglets that developed specific S. suis symptoms (meningitis, arthritis, or mortal illness) were killed. Piglets that did not develop these symptoms were killed at the end of the experiment. From all piglets, the ratio of wild-type and mutant strain in various organs was determined.
  • As shown in FIG. 5, panel A, similar numbers of wild-type and mutant bacteria were re-isolated from tonsils. The ratio was similar to the input ratio (ratio varied from 0.33-0.85, average 0.61). This indicates that the efficiency of colonization of wild-type and mutant strain on tonsils was essentially identical. Apparently, FBPS is not strictly required for colonization of the tonsils of the piglets. Three out of four piglets developed clinical signs specific for an [0117] S. suis infection. Two piglets (4664 and 4666) showed clinical signs of arthritis and one piglet (4668) showed clear central nervous signs. The fourth piglet did not develop any clinical signs. These observations coincided with pathomorphological abnormalities of the specific organs of an S. suis infection in post-mortem sections.
  • As shown in FIG. 5, panel B, exclusively wild-type bacteria were re-isolated from the joints of piglet 4664 and from the CNS of piglet 4668. The numbers of CFU of wild-type bacteria that were re-isolated from these specific organs were very high, while no mutant bacteria were found. From the joints of pig 4666, low numbers of both wild-type and mutant bacteria were re-isolated in a ratio of 0.84 (1.0×10[0118] 2 CFU of wild-type bacteria and 5.2×102 CFU of mutant bacteria), a ratio essentially identical to the input ratio (FIG. 5, panel B). Southern blot experiments using the fbps and the spc genes as probes, confirmed that the mutant bacteria isolated from the joint of pig 4666 were indeed identical to the input mutant bacteria. Taken together, these data indicate, that the FBPS mutant is capable of reaching and colonizing the specific S. suis organs (at least the joints), but that the mutant is far less efficiently recovered from organs than the wild type.
    TABLE 1
    Bacterial strains and plasmids.
    Strain/plasmid relevant characteristics* source/reference
    Strain
    E. coli
    XL2 blue Stratagene
    S. suis
    10 virulent serotype 2 strain (Vecht et al.,
    1992)
    Plasmid
    pKUN19-spc replication functions pUC, AmpR, (Konings et al.,
    SpcR 1987, Smith et
    al., 1995)
    pGKV210 replication functions pWVO1, CmR, (Van der Vossen
    EmR et al., 1987)
    pE194 EmR (Horinouchi &
    Weisblum, 1987)
    pMR11 pKUN19 containing S. suis mrp gene (Smith et al.,
    1992)
    pIVS-E replication functions pWVO1, SpcR, this work
    promoterless emR gene of pE194
    pIVS-PE pIVS-E containing promoter of mrp this work
    preceding the promoterless emR gene
    pIVS-RE pIVS-E containing random S. suis this work
    sequences preceding the
    promoterless emR gene
  • [0119]
    TABLE 2
    Iron-restriction induced S. suis genes.
    Insert Data base homology %
    Clone (bp) GC % (accession no.) Function of homolog Identity
    Regulatory functions
    iri 1, 6, 22 800 34 S. mutans SapR (U75483) response regulator protein 44
    (SEQ ID NO: 8)
    S. aureus AgrA (X52543) response regulator protein 51
    S. aureus Ivi2
    iri 24 850 38 S. agalactiae CpsY (CAB36982) regulation capsule expression 46
    (SEQ ID NO: 17)
    E. coli OxyR (P11721) oxidative stress regulator 51
    iri 23 1000 38 B. subtilis YvyD (P28368) sigma-54 modulator homologue 44
    (SEQ ID NO: 16)
    Metabolic functions
    iri 7 800 39 S. mutans RgpG (Q9XDW8) rhamnose-glucose biosynthesis 76
    (SEQ ID NO: 23)
    iri 11 700 34 L. lactis NrdD (Q9ZAX6) anaerobic ribonucleotide reductase 51
    (SEQ ID NO: 10)
    iri 14 500 38 S. pneumoniae SulB (Q54614) dihydrofolate synthetase 41
    (SEQ ID NO: 12)
    iri 16 850 48 B. subtilis TrmU (O35020) RNA methyltransferase 62
    (SEQ ID NO: 13)
    iri 32 300 41 C. histolyticum RuvB (O9ZNJ5) hypoxanthine-guanine 55
    (SEQ ID NO: 20) phosphoribosyl transferase
    iri 34 1000 44 L. lactis IlvA (U92974) probable threonine dehydratase 56
    (SEQ ID NO: 21)
    P. aeruginosa Pn16
    Transporter functions
    iri 2 750 36 B. subtilis YloD (O34328) putative guanylate kinase 50
    (SEQ ID NO: 15)
    S. gordonii ComYA (U81957) putative ABC transporter 37
    Vibrio cholerae IviVI (Q56605) putative ABC transporter 47
    iri 10, 20 1350 51 E. coli YoaE (P76262) putative transport protein 94
    (SEQ ID NO: 9)
    Unknown
    iri 13, 15, 27 800 34 M. tuberculosis MTCY336_33 unknown 38
    (SEQ ID NO: 11)
    hypothetical protein (O06593)
    iri 29 850 36 S. aureus Yp15 (P13977) unknown 39
    (SEQ ID NO: 18) hypothetical protein
    iri 18 800 39 S. crista hypothetical protein (AAF61316) unknown 82
    (SEQ ID NO: 14)
    iri 3 700 36 no homology found
    (SEQ ID NO: 19)
    iri 4 700 36 no homology found
    (SEQ ID NO: 22)
    iri 8, 26 900 35 no homology found
    (SEQ ID NO: 24)
  • [0120]
    TABLE 3
    Virulence of S. suis 10 (pIVET-E), 10 (pIVET-PE) and 10 (pIVET-RE) in gnotobiotic piglets.
    Clinical index
    of the group ¦
    Non- Fever No. of pigs from which
    Strains/ No. of Dose (route Mortality Morbidity Specific specific index Leukocyte S. suis was isolated
    library piglets of infection) * (%) ¥ (%)
    Figure US20040096973A1-20040520-P00801
    à index £ CNS Serosae Joints
    10 (pIVS-E) 4 106 (i.n.) 0 0 0 6 9 75 0 0 0
    10 (pIVS-E) 4 106 (i.v.) 0 0 6 12 31 0 0 0 0
    10 (pIVS-PE) 4 106 (i.n.) 100 100 30 40 35 100 3 0 2
    10 (pIVS-PE) 4 106 (i.v.) 75 100 50 42 43 50 3 3 4
    10 (pIVS-RE) 4 5 × 105 (i.v.) 100 100 56 75 44 83 2 2 4
    10 (pIVS-RE) 4 5 × 106 (i.v.) 100 100 43 73 43 60 3 0 4
    10 (pIVS-RE) 4 5 × 107 (i.v.) 100 100 60 74 48 75 4 1 4
    10 (pIVS-RE) 4 5 × 108 (i.v.) 100 100 49 70 37 50 3 3 4
  • [0121]
    TABLE 4
    S. suis genes selected in pigs.
    Sites of Insert Data base homology Function %
    Clone isolation (bp) GC % (accession no.) of homolog Identity
    Putative virulence factors
    ivs 31 CNS 200 47 S. gordonii FlpA (X65164) fibronectin/fibrinogen binding 70
    (SEQ ID NO: 37)
    Regulatory functions
    ivs 25 joint 800 34 S. mutans SapR (P72485) response regulator protein 49
    (SEQ ID NO: 34)
    S. aureus AgrA (X52543) response regulator protein 51
    S. suis Iri 1, 6, 22 100
    ivs 23, 24 other 850 38 S. agalactiae CpsY (CAB36982) regulation capsule expression 46
    (SEQ ID NO: 33)
    E. coli OxyR (P11721) oxidative stress regulator 51
    S. suis Iri 24 100
    ivs 16 CNS 800 43 S. epidermidis AltR (U71377) putative transcriptional regulator 26
    (SEQ ID NO: 28)
    ivs 20 lung 800 41 L. lactis AldR (O34133) putative regulator AldR 64
    (SEQ ID NO: 32)
    Metabolic functions
    ivs 33 CNS 570 36 E. coli ThrC (P00934) threonine synthase 41
    (SEQ ID NO: 39)
    ivs 5, 10, CNS, joint 900 36 S. gordonii Tdk (P47848) thymidine kinase 87
    12, 22
    (SEQ ID NO: 42)
    ivs 18 lung 730 32 S. mutans NADH oxidase (JC4541) NADH oxidase 80
    (SEQ ID NO: 29)
    Transporter functions
    ivs 2, 4, 28 CNS, joint 1350 51 E. coli YoaE (P76262) putative transport protein 94
    (SEQ ID NO: 31)
    S. suis iri 10, 20 100
    ivs 3 joint 1000 42 S. mutans OrfU (AF267498) putative ABC transporter (permease) 33
    (SEQ ID NO: 36)
    ivs 6, 7, CNS, joint 1350 36 B. subtilis Ylo D (O34328) putative guanylate kinase 50
    13, 14
    (SEQ ID NO: 43)
    S. gordonii ComYa (U81957) putative ABC transporter 37
    V. cholera IviVI (Q56605) putative ABC transporter 47
    S. suis Iri 2 100
    Transposases
    ivs 8 CNS 600 41 S. pneumoniae transposase transposase 70
    (SEQ ID NO: 44) (Z86112)
    ivs 1 joint 1600 39 C. perfringens (X71844) putative transposase 56
    (SEQ ID NO: 25)
    Miscellaneous
    ivs 32, 35 CNS 500 38 S. typhimurium FliF (P15928) flagellar M-protein precursor 36
    (SEQ ID NO: 38)
    ivs 9, 17 joint, CNS 800 36 B. subtilis ComE ORF2 (P32393) competence development 37
    (SEQ ID NO: 45)
    ivs 11 serosea 800 44 P. syringae TabA (P31851) diaminopimelate decarboxylase/ 53
    (SEQ ID NO: 26) tabtoxin
    Unknown
    ivs 15 CNS 750 42 B. subtilis conserved hypothetical unknown 43
    (SEQ ID NO: 27) protein YdiB (D88802)
    ivs 29 joint 800 38 S. salivarius hypothetical protein unknown 79
    (SEQ ID NO: 35) (AF130465)
    ivs 34 CNS 600 43 B. subtilis conserved hypothetical unknown 61
    (SEQ ID NO: 40) protein YRRK (O34634)
    ivs 36 joint 830 42 B. subtilis hypothetical protein unknown 35
    (SEQ ID NO: 41) YqeG (P54452)
    ivs 19 lung 950 34 S. cristatus hypothetical protein unknown 86
    (SEQ ID NO: 30) (U96166)
  • [0122]
    TABLE 5
    Bacterial strains and plasmids.
    Strain/plasmid Relevant Characteristicsa Source/reference
    Strains
    E. coli
    XL2-Blue recA1 endA1 gyrA96 thi-1 hsdR17 supE44 relA1 Stratagene
    lac (Fl proAB lacIq Z)M15 TN10 (TetR) amy CmR)
    M15 NalS StrS RifS ThiLacAra+ Gal+ MtlFRecA+ Uvr+ Lon+ Qiagen
    S. suis
    10 Virulent serotype 2 strain Vecht et al. (29)
    10ΔFBPS Isogenic fbps mutant of strain 10 This work
    Plasmids
    pGEM7Zf(+) Replication functions pUC, AmpR Promega Corp.
    pKUN19 Replication functions pUC, AmpR Konings et al. (14)
    pIC19R Replication functions pUC, AmpR Marsh et al. (16)
    pDL282 Replication functions of pBR322 and pVT736-1, AmpR, SpcR Sreenivasan et al. (25)
    pIC-spc pIC19R containing SpcR gene of pDL282 Lab collection
    pQE-30 Replication functions pBR322, AmpR, expression vector, 6x HIS tag Qiagen
    pQE-30-FBPS pQE-30 containing the 1.8 kb fbps gene This work
    pREP4 Replication functions pACYC, KanR, lacI gene Qiagen
    pE194 EmR Horinouchi and Weisblum (11)
    pIVS-E Replication functions of pWVO1, SpcR, promoterless erm gene of pE194 Smith et al. (20)
    pIVS-31 pIVS-E containing 200 bp showing homology to Streptococcus gordonii Smith et al. (20)
    flpa
    pFBPS7-46 pGEM7Zf(+) containing EcoRI-EcoRI fragment of fbps This work
    pFBPS7-47 pFBPS7-46 in which 382 bp SalI-SalI fragment is replaced by 1.2 kb SpcR This work
    from pIC-spc
  • [0123]
    TABLE 6
    Numbers of re-isolated wild-type (strain 10) and mutant (strain 10ΔFBPS) bacteria
    from organs of infected piglets (mean actual inoculation ratio 65% mutant strain).
    Pig number
    4664 4665 4666 4667
    perc.c perc.c perc.c perc.c
    Organ w.t.a mut.b mut. w.t.a mut.b mut. w.t.a mut.b mut. w.t.a mut.b mut.
    Tonsil 1.77e5 3.29e5 65 4.35e5 2.42e6 85 5.34e4 8.73e4 61 7.94e5 3.96e5 33
    pus joint 1 6.75e4 <10 0 1.02e2 5.2e2 84
    pus joint 2 5.15e4 <10 0
    CNS 1.88e5 <10 0
  • REFERENCES
  • Arends, J. P. and H. C. Zanen. (1988). Meningitis caused by [0124] Streptococcus suis in humans. Rev. Infect. Dis. 10, 131-137.
  • Axelsson, L. and A. Holck. (1995). The genes involved in production of and immunity to sakacin A, a bacteriocin from [0125] Lactobacillus sake Lb706. J. Bacteriol. 177, 2125-2137.
  • Camacho, L. R., D. Ensergueix, E. Perez, B. Gicquel and C. Guilhot. (1999). Identification of a gene cluster of [0126] Mycobacterium tuberculosis by signature-tagged transposon mutagenesis. Mol. Microbiol. 34, 257-267.
  • Camilli, A. and J. J. Mekalanos. (1995). Use of recombinase gene fusions to identify [0127] Vibrio cholerae genes induced during infection. Mol. Microbiol. 18, 671-683.
  • Chiang, S. L. and J. J. Mekalanos. (1998). Use of signature-tagged transposon mutagenesis to identify [0128] Vibrio cholerae genes critical for colonization. Mol. Microbiol. 27, 797-805.
  • Clifton-Hadley, F. A. (1983). [0129] Streptococcus suis type 2 infections. Br. Vet. J. 139, 1-5.
  • Conner, C. P., D. M. Heithoff, S. M. Julio, R. L. Sinsheimer and M. J. Mahan. (1998). Differential patterns of acquired virulence genes distinguish Salmonella strains. [0130] Proc. Natl. Acad. Sci. USA 95, 4641-4645.
  • Coulter, S. N., W. R. Schwan, E. Y. W. Ng, M. H. Langhorne, H. D. Ritchie, S. Westbrock-Wadman, W. O. Hufnagle, K. R. Folger, A. S. Bayer and C. K. Stover. (1998). [0131] Staphylococcus aureus genetic loci impacting growth and survival in multiple infection environments. Mol. Microbiol. 30, 393-404.
  • Courtney, H. S., J. B. Dale, Y. Li and D. L. Hasty. (1994). Cloning, sequencing, and expression of a fibronectin/fibrinogen-binding protein from group A Streptococci. [0132] Infect. Immun. 62, 3937-3946.
  • Darwin, A. J. and V. L. Miller. (1999). Identification of [0133] Yersinia enterocolitica genes affecting survival in an animal host using signature-tagged transposon mutagenesis. Mol. Microbiol. 32, 51-62.
  • Demple, B. (1999). Radical ideas: genetic responses to oxidative stress. [0134] Clin. Exp. Pharm. Phys. 26, 64-68.
  • Edelstein, P. H., M. A. C. Edelstein, F. Higa and S. Falkow. (1999). Discovery of virulence genes of [0135] Legionella pneumophila by using signature tagged mutagenesis in a guinea pig pneumonia model. Proc. Natl. Acad. Sci. USA 96, 8190-8195.
  • Elliott, S. D. and J. Y. Tai. (1978). The type specific polysaccharide of [0136] Streptococcus suis. J. Exp. Med. 148, 1699-1704.
  • Fuller, T. E., R. J. Shea, B. J. Thacker and M. H. Mulks. (1999). Identification of in vivo induced genes in [0137] Actinobacillus pleuropneumoniae. Microb. Path. 27, 311-327.
  • Gottschalk, M., S. Lacouture and J. D. Dubreuil. (1995). Characterization of [0138] Streptococcus suis type 2 hemolysin. Microbiology 141, 189-195.
  • Hensel, M., J. E. Shea, C. Gleeson, M. D. Jones, E. Dalton and D. W. Holden. (1995). Simultaneous identification of bacterial virulence genes by negative selection. [0139] Science 269, 400-403.
  • Horinouchi, S. and B. Weisblum. (1982). Nucleotide sequence and functional map of pE194, a plasmid that specifies inducible resistance to macrolide, lincosamide and streptogramin type B antibiotics. [0140] J. Bacteriol. 150, 804-814.
  • Jacobs, A. A., P. L. W. Loeffen, A. J. G. van den Berg and P. K. Storm. (1994). Identification, purification and characterization of a thiol-activated hemolysin (suilysin) of [0141] Streptococcus suis. Infect. Immun. 62, 1742-1748.
  • Kolkman, M. A. B., W. Wakarchuk, P. J. M. Nuijten and B. A. M. van der Zeijst. (1997). Capsular polysaccharide synthesis in [0142] Streptococcus pneumoniae serotype 14: molecular analysis of the complete cps locus and identification of genes encoding glycosyltransferases required for the biosynthesis of the tetrasaccharide subunit. Mol. Microbiol. 26, 197-208.
  • Konings, R. N. H., E. J. M. Verhoeven and B. P. H. Peeters. (1987). pKUN vectors for the separate production of both DNA strands of recombinant plasmids. [0143] Methods Enzymol. 153, 12-34.
  • Koskiniemi, S., M. Sellin and M. Norgren. (1998). Identification of two genes, cpsX and cpsY, with putative regulatory function on capsule expression in group B Streptococci. [0144] FEMS Immun. Med. Microbiol. 21, 159-168.
  • Litwin, C. M. and S. B. Calderwood. (1993). Role of iron regulation of virulence genes. [0145] Clin. Microbiol. Rev. 6, 137-149.
  • Lowe, A. M., D. T. Beattie and R. L. Deresiewics. (1998). Identification of novel staphylococcal virulence genes by in vivo expression technology. [0146] Mol. Microbiol. 27, 967-976.
  • Mahan, M. J., J. M. Slauch and J. J. Mekalanos. (1993). Selection of bacterial virulence genes that are specifically induced in host tissues. [0147] Science 259, 686-688.
  • Mahan, M. J., T. W. Tobias, J. M. Slauch, P. C. Hanna, R. J. Collier and J. J. Mekalanos. (1995). Antibiotic-based selection for bacterial genes that are specifically induced during infection of a host. [0148] Proc. Natl. Acad. Sci. USA 92, 669-673.
  • Martinez, J. L., A. Delgado-Iribarren and F. Basquero. (1990). Mechanisms of iron acquisition and bacterial virulence. [0149] FEMS Microbiol. 75, 45-56.
  • Mei, J-M., F. Nourbakhsh, C. W. Ford and D. W. Holden. (1997). Identification of [0150] Staphylococcus aureus virulence genes in a murine model of bacteremia using signature-tagged mutagenesis. Mol. Microbiol. 26, 399-407.
  • Miller, J. (1972). [0151] Experiments in Molecular Genetics. Cold Spring Harbor, N.Y.: Cold Spring Harbor Laboratory.
  • Niven, D. F., A. Ekins and A. A.-W Al-Samaurai. (1999). Effects of iron and manganese availability on growth and production of superoxide dismutase by [0152] Streptococcus suis. Can. J. Microbiol. 45, 1027-1032.
  • Payne, S. M. (1993). Iron acquisition in microbial pathogenesis. [0153] Trends Microbiol. 1, 66-69.
  • Polissi, A., A. Pontiggia, G. Feger, M. Altieri, H. Mottl, L. Ferrari and D. Simon. (1998). Large-scale identification of virulence genes from [0154] Streptococcus pneumoniae. Infect. Immun. 66, 5620-5629.
  • Projan, S. J. and R. P. Novick. (1997). The molecular basis of pathogenicity. [0155] The Staphylococci in Human Disease, pp 55-81. Edited by K. B. Crossley and G. L. Archer. Churchill Livingstone, NY.
  • Quessy, S., J. D. Dubreuil, M. Jacques, F. Malouin and R. Higgins. (1994). Increase in capsular material thickness following in vivo growth of virulent [0156] Streptococcus suis serotype 2 strains. FEMS Microbiol. Lett. 115, 19-26.
  • Sambrook, J., E. F. Fritsch and T. Maniatis. (1989). [0157] Molecular Cloning: a Laboratory Manual, 2nd edn. Cold Spring Harbor, N.Y.: Cold Spring Harbor Laboratory.
  • Segers, R. P. A. M., T. Kenter, L. A. M. de Haan and A. A. C. Jacobs. (1998). Characterization of the gene encoding suilysin from [0158] Streptococcus suis and expression in field strains. FEMS Microbiol. Lett. 167, 255-261.
  • Serhir, B., D. Dugourd, M. Jacques, R. Higgins and J. Harel. (1997). Cloning and characterization of a dextranase gene (dexS) from [0159] Streptococcus suis. Gene 19, 257-261.
  • Smith, H. E., U. Vecht, A. L. J. Gielkens and M. A. Smits. (1992). Cloning and nucleotide sequence of the gene encoding the 136-kilodalton surface protein (muramidase-released protein) of [0160] Streptococcus suis type 2. Infect. Immun. 60, 2361-2367.
  • Smith, H. E., F. H. Reek, U. Vecht, A. L. J. Gielkens and M. A. Smits. (1993). Repeats in an extracellular protein of weakly pathogenic strains of [0161] Streptococcus suis type 2 are absent in pathogenic strains. Infect. Immun. 61, 3318-3326.
  • Smith, H. E., H. J. Wisselink, U. Vecht, A. L. J. Gielkens and M. A. Smits. (1995). High-efficiency transformation and gene inactivation in [0162] Streptococcus suis type 2. Microbiol. 141, 181-188.
  • Smith, H. E., U. Vecht, H. J. Wisselink, N. Stockhofe-Zurwieden, Y. Biermann and M. A. Smits. (1996). Mutants of [0163] Streptococcus suis types 1 and 2 impaired in expression of muranidase-released protein and extracellular protein induce disease in newborn germ-free pigs. Infect. Immun. 64, 4409-4412.
  • Smith, H. E., M. Damman, J. van der Velde, F. Wagenaar, H. J. Wisselink, N. Stockhofe-Zurwieden and M. A. Smits. (1999). Identification and characterization of the cps locus of [0164] Streptococcus suis serotype 2: the capsule protects against phagocytosis and is an important virulence factor. Infect. Immun. 67, 1750-1756.
  • Stockhofe-Zurwieden, N., U. Vecht, H. J. Wisselink, H. van Lieshout and H. E. Smith. (1996). Comparative studies on the pathogenicity of different [0165] Streptococcus suis serotype 1 strains. In Proceedings of the 14th International Pig Veterinary Society Congress, pp 299. Edited by P. G. Monetti and G. Vignola. Bologna, Italy.
  • Van der Vossen, J. M. B. M., D. van der Lelie and G. Venema. (1987). Isolation and characterization of [0166] Lactococcus lactis subsp. cremoris Wg2-specific promoters. Appl. Environ. Microbiol. 52, 2452-2457.
  • Vecht, U., L. A. M. G. van Leengoed and E. R. M. Verheyden. (1985). [0167] Streptococcus suis infections in pigs in The Netherlands (part one). Vet. Quart. 7: 315-321.
  • Vecht, U., J. P. Arends, E. J. van der Molen and L. A. M. G. van Leengoed. (1989). Difference in virulence between two strains of [0168] Streptococcus suis type 2 after experimentally induced infection of newborn germ-free pigs. Am. J. Vet. Res. 50, 1037-1043.
  • Vecht, U., H. J. Wisselink, M. L. Jellema and H. E. Smith. (1991). Identification of two proteins associated with virulence of [0169] Streptococcus suis type 2. Infect. Immun. 59, 3156-3162.
  • Vecht, U., H. J. Wisselink, J. E. van Dijk and H. E. Smith. (1992). Virulence of [0170] Streptococcus suis type 2 strains in newborn germ-free pigs depends on phenotype. Infect. Immun. 60, 550-556.
  • Vecht, U., N. Stockhofe-Zurwieden, B. J. Tetenburg, H. J. Wisselink and H. E. Smith. (1997). Virulence of [0171] Streptococcus suis type 2 for mice and pigs appeared host-specific. Vet. Microbiol. 58, 53-60.
  • Wang, J., A. Mushegian, S. Lory and S. Jin. (1996). Large-scale isolation of candidate virulence genes of [0172] Pseudomonas aeruginosa by in vivo selection. Proc. Natl. Acad. Sci. USA 93, 10434-10439.
  • Yamashita, Y., Y. Shibata, Y. Nakano, H. Tsuda, N. Kido, M. Ohta and T. Koga. (1999). A novel gene required for rhamnose-glucose polysaccharide synthesis in [0173] Streptococcus mutans. J. Bacteriol. 181, 6556-6559.
  • Young, G. M. and V. L. Miller. (1997). Identification of novel chromosomal loci affecting [0174] Yersinia enterocolitica pathogenesis. Mol. Microbiol. 25, 319-328.
  • Zhao, H., X. Li, D. E. Johnson and H. L. T. Mobley. (1999). Identification of protease and rpoN-associated genes of uropathogenic [0175] Proteus mirabilis by negative selection in a mouse model of ascending urinary tract infection. Microbiol. 145, 185-195.
  • OTHER REFERENCES
  • 1. Allgaier A. R. G., H. J. Wisselink, H. E. Smith, and P. Valentin-Weigand. 2001. Relatedness of [0176] Streptococcus suis isolates of various serotypes and clinical backgrounds as evaluated by macrorestriction analysis and expression of potential virulence traits. J. Clin. Microbiol. 39:445-453.
  • 2. Allen, A. G., S. Bolitho, H. Lindsay, S. Khan, C. Bryant, P. Norton, P. Ward, J. Leigh, J. Morgan, H. Riches, S. Eastty, D. Maskell. 2001. Generation and characterization of a defined mutant of [0177] Streptococcus suis lacking suilysin. Infect. Immun. 69:2732-2735.
  • 3. Arends, J. P., and H. C. Zanen. 1988. Meningitis caused by [0178] Streptococcus suis in humans. Rev. Infect. Dis. 10:131-137.
  • 4. Charland, N., V. Nizet, C. Rubens, K. Kim, S. Lacouture, and M. Gottschalk. 2000. [0179] Streptococcus suis interactions with human brain microvascular endothelial cells. Infect. Immun. 68:637-643.
  • 5. Chia, J. S., C. Y. Yeh, and J. Y. Chen. 2000. Identification of a fibronectin binding protein from [0180] Streptococcus mutans. Infect. Immun. 68:1864-1870.
  • 6. Clifton-Hadley, F. A. 1983. [0181] Streptococcus suis type 2 infections. Br. Vet. J. 139:1-5.
  • 7. Courtney, H. S., J. B. Dale, and D. L. Hasty. 1996. Differential effects of the Streptococcal fibronectin-binding protein, FBP54, on adhesion of group A Streptococci to human buccal cells and Hep-2 tissue culture cells. [0182] Infect. Immun. 64:2415-2419.
  • 8. Courtney, H. S., Y. Li, J. B. Dale, and D. L. Hasty. 1994. Cloning, sequencing, and expression of a fibronectin/fibrinogen-binding protein from group A [0183] Streptococci. Infect. Immun. 62:3937-3946.
  • 9. Fuller, T. E., S. Martin, J. F. Teel, G. R. Alaniz, M. J. Kennedy, and D. E. Lowery. 2000. Identification of [0184] Actinobacillus pleuropneumoniae virulence genes using signature-tagged mutagenesis in a swine infection model. Microb. Pathog. 29:39-51.
  • 10. Greene, C., D. McDevitt, P. Francois, P. Vaudaux, and T. J. Foster. 1995. Adhesion properties of mutants of [0185] Staphylococcus aureus defective in fibronectin-binding proteins and studies on the expression of fnb genes. Mol. Microbiol. 17:1065-1076.
  • 11. Horinouchi, S., and B. Weisblum. 1982. Nucleotide sequence and functional map of pE194, a plasmid that specifies inducible resistance to macrolide, lincosamide and streptogramin type B antibiotics. [0186] J. Bacteriol. 150:804-815.
  • 12. Joh, D., E. R. W. Wann, B. Kreikemeyer, P. Speziale, and M. Höök. 1999. Role of fibronectin-binding MSCRAMMS in bacterial adherence and entry into mammalian cells. [0187] Matrix Biol. 18:211-223.
  • 13. Kawabata, S., E. Kunitomo, Y. Terao, I. Nagakawa, K. Kiruchi, K.-I.. Totsuka, and S. Hamada. 2001. Systemic and mucosal immunizations with fibronectin-binding protein FBP54 induce protective immune responses against [0188] Streptococcus pyogenes challenge in mice. Infect. Immun. 69:924-930.
  • 14. Konings, R. N. H., E. J. M. Verhoeven, and B. P. H. Peeters. 1987. pKUN vectors for the separate production of both DNA strands of recombinant plasmids. [0189] Methods Enzymol. 153:12-34.
  • 15. Mahan, M. J., J. M. Slauch, and J. J. Mekalanos. 1993. Selection of bacterial virulence genes that are specifically induced in host tissues. [0190] Science 259:686-688.
  • 16. Marsh, J. L., M. Erfle, and E. J. Wykes. 1984. The pIC plasmid and phage vectors with versatile cloning sites for recombinant selection by insertional inactivation. [0191] Gene 32:481-485.
  • 17. Miller, J. 1972. Experiments in molecular genetics. Cold Spring Harbor Laboratory. Cold Spring-Harbor, N.Y. [0192]
  • 18. Peacock, S. J., T. J. Foster, B. J. Cameron, and A. R. Berendt. 1999. Bacterial fibronectin-binding proteins and endothelial cell surface fibronectin mediate adherence of [0193] Staphylococcus aureus to resting human endothelial cells. Microbiology 145:3477-3486.
  • 19. Sambrook, J., E. F. Fritsch, and T. Maniatis. 1989. Molecular cloning: a laboratory manual, 2nd ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor. N.Y. [0194]
  • 20. Smith, H. E., H. Buijs, R. de Vries, H. J. Wisselink, N. Stockhofe-Zurwieden, and M. A. Smits. 2001. Environmentally regulated genes of [0195] Streptococcus suis: identification by the use of iron-restricted conditions in vitro and by experimental infection of piglets. Microbiol. 147:271-280.
  • 21. Smith, H. E., H. Buijs, H. J. Wisselink, N. Stockhofe-Zurwieden, and M. A. Smits. 2001. Selection of virulence-associated determinants of [0196] Streptococcus suis serotype 2 by in vivo complementation. Infect. Immun. 69:1961-1966.
  • 22. Smith, H. E., M. Damman, J. van der Velde, F. Wagenaar, H. J. Wisselink, N. Stockhofe-Zurwieden, and M. A. Smits. 1999. Identification and characterization of the cps locus of [0197] Streptococcus suis serotype 2: the capsule protects against phagocytosis and is an important virulence factor. Infect. Immun. 60:1750-1756.
  • 23. Smith, H. E., U. Vecht, H. J. Wisselink, N. Stockhofe-Zurwieden, Y. Biermann, and M. A. Smits. 1996. Mutants of [0198] Streptococcus suis types 1 and 2 impaired in expression of murimidase-released protein and extracellular protein induce disease in newborn germ-free pigs. Infect. Immun. 64:4409-4412.
  • 24. Smith, H. E., H. J. Wisselink, U. Vecht, A. L. J. Gielkens, and M. A. Smits. 1995. High-efficiency transformation and gene inactivation in [0199] Streptococcus suis type 2. Microbiology 141:181-188.
  • 25. Sreenivasan, P. K., D. L. LeBlanc, L. N. Lee, and P. Fives-Taylor. 1991. Transformation of [0200] Actinobacillus actinomycetemcomitans by electroporation, utilizing constructed shuttle plasmids. Infect. Immun. 59:4621-4627.
  • 26. Staats, J. J., I. Feder, O. Okwumabua, and M. M. Cheganppa. 1995. [0201] Streptococcus suis: past and present. Vet. Res. Comm. 21:381-407.
  • 27. Vecht, U., J. P. Arends, E. J. van der Molen, and L. A. M. G. van Leengoed. 1989. Differences in virulence between two strains of [0202] Streptococcus suis type 2 after experimentally induced infection of newborn germ-free pigs. Am. J. Vet. Res. 50:1037-1043.
  • 28. Vecht, U., L. A. M. G. van Leengoed, and E. R. M. Verheyen. 1985. [0203] Streptococcus suis infections in pigs in The Netherlands (part I). Vet. Q. 7:315-321.
  • 29. Vecht, U., H. J. Wisselink, J. E. van Dijk, and H. E. Smith. 1992. Virulence of [0204] Streptococcus suis type 2 strains in newborn germ-free pigs depend on phenotype. Infect. Immun. 60:550-556.
  • 30. Vecht, U., H. J. Wisselink, M. L. Jellema, and H. E. Smith. 1991. Identification of two proteins associated with virulence of [0205] Streptococcus suis type 2. Infect. Immun. 59:3156-3162.
  • 1 47 1 6 PRT Artificial Sequence Cell wall signalling sequence 1 Leu Pro Xaa Thr Gly Glu 1 5 2 30 DNA artificial sequence PCR primer containing NsiI and SacI restriction sites at the 5′ end 2 tgcatgcatg gatccatcga ttttcgttcg 30 3 30 DNA artificial sequence PCR primer containing NsiI and SacI restriction sites at the 5′-end 3 cgagctcggt acctgattac caattagaat 30 4 30 DNA artificial sequence PCR primer containing SalI and HindIII restriction sites at the 5′-end 4 gggtcgaccc tataaccaaa ttaaagaggg 30 5 32 DNA artificial sequence PCR primer containing SalI and HindIII restriction sites at the 5′-end 5 cccaagcttg ggcagtttat gcatccctta ac 32 6 34 DNA artificial sequence PCR primer containing EcoRI and SalI restriction sites at the 5′-end 6 cccaagcttg ggaattcata atgttttttt gagg 34 7 30 DNA artificial sequence PCR primer containing EcoRI and SalI restriction sites at the 5′-end 7 gcgtcgacat ctacgcataa aaaatccccc 30 8 335 DNA Streptococcus suis iri 1, 6, 22 8 gaaacgtcag taaggtataa attcctagaa gttnttggta aaccaaatca atnattggaa 60 tcaattgggg aagcagggaa tcatcaattt ttctttttag atattgaaat aaaaggagaa 120 gaaaagaaag gaatggaaat cgctaaagaa atccgggctc gagatcctta tgctgctatt 180 gtctttgtaa caactcactc agaattnatg ccagtaacat atcgttatca ggtttctgct 240 ttagatttta tagataaagg cctggaggat cgtgactttc aaaaggcagt atcagangtc 300 ttagtgcatg cttttgaaaa tatcgatcat actnt 335 9 347 DNA Streptococcus suis iri 10, 20 9 gntnggcggt tttccagccc gttnatgcag ttnggttgtt gctnngaaca gcaagaatat 60 cccccngaac aacataatca ggtcgngtcc ggagaaggag aaatcccatg acggtaaatn 120 gcggtttggt cagngtgacc atccatgaaa tcagcgacag cagccccaga cgcataatca 180 gcgccagtga taaccccagc aaacgcgctt tatcgngttg ttttggcggc agtttgtcag 240 caagaatggc gatgaagacc aggttataga tacccagcac aatttcgaga acaaccagcg 300 tgagtagccc cgcccaaatt gaggggtcca ttaagncnta cgaaant 347 10 339 DNA Streptococcus suis iri 11 10 ttcatcctng tcgagngggg aaatggggca cagttgtttt ccaattgata gaatttttaa 60 gaacctatat atanaaacat ggataggttg tttaatattt tttnacacaa gatattgatt 120 tttgttttgt gaagtgctac actaatagaa gtgaagaatt ttggaggttt gtcagatgaa 180 tgtgcaagaa aatgtcttac cgagtataga attattggtt ttgaaacgtg atggacggac 240 agtatccttt gaccangata agattttttn tgctcttcag cgggcaaacc aagaattaga 300 acatcctgtt tcagnggcag gtttaaaaat tgtattaga 339 11 270 DNA Streptococcus suis iri 13, 15, 27 11 ggaggnggtt anacnggcat ttgcagatgc cgaatttatc taatgatttt gtagaagagt 60 ggtngcatng aacaaccctc tttatcntta agaaaatgnt aggatagtcg gtcaatctan 120 gctatactag aaacgttatt aagtcccgaa aaggtagttt atagactagt taatatttgc 180 agaaacactt gnaacacaat taaagaaact ggtantattg aatagtaagc gtaaaaactt 240 tactacactt cagtcactat tttacntcaa 270 12 288 DNA Streptococcus suis iri 14 12 tattnntagt gtactataga aaaaactaac ntaccacaan acgtgatagg ttagttaant 60 taatgacatt gggctntttg cccagcntct tttttnttat attagacagt atgtaggagg 120 tggntangtt agaaaattgg ttaaacacca aacaaggtca ggtgtttcat tacaagatgg 180 aaaagattga gtatgcccta gaactgctag ggaatcccca gttngcagtt ccggtcattc 240 atgtcgctgg aactaatggc aagggatcga ccattgcctt tatgcgca 288 13 342 DNA Streptococcus suis iri 16 13 gtacggcaac cggaagataa caagggatnt ggcngcagtt ggcagntcaa atcggcattc 60 cttagtantc tgtcaacttt gaaaaagagt actgggaccg cgtatttgag tactttttag 120 cagagtatcg ggntggtcgc acgcccnatc cagatgtcan gtgtaacaag ganatcaagt 180 tcaaggcttt nttggantac gntatgaact tgggtgcgga ctatgtggcg acagggcact 240 acgctcaggt nacccgcgac gangacggca ccgttcatat gctgcgtggg gcagataatg 300 gtaaggacca gacctatttc cncagccaac tctcacgnna ac 342 14 340 DNA Streptococcus suis iri 18 14 ctgtctaacc accctaacat ngcatantcc tcctttttta tctattttat caaaaaatcg 60 gngcttttct accatttgtc aagttcatca aggtatttga cgaaaaatan tnngtgtctc 120 gtcatccaaa taaggaaatt gttttatttt ggactaaagt tacgtgtaaa aagngcatac 180 aaaaccaaca ccttntgttg naattttttg ataaggtgtt acaatgatag agcataaaca 240 gttttaccga ttttgggtng aagcgtaatc gtnaaatttg ttatgcntaa tgaggtaata 300 cattgtccga atgagacgat gtatggaggc gatcgnangn 340 15 355 DNA Streptococcus suis iri 2 15 ancctttcnn ggcccnatgg atgtttcgng gagaaaattg gaaaaataat cgggattatc 60 cagggaaatn tcaaaatcac agttatccgt gaaacaagag caacngangt tgcnaagtaa 120 atgtagaaga gtccgaaggg ctctttttnt actggctcaa agttcgttan gggttgggaa 180 tagaaaatag aaaatatttt aatcgtattt aaaagcagtt gaaattcatg ctaaattttg 240 ttacactaga atgaaagatt taaaaggaga tatcatgaaa gagcgaggct tactcatngt 300 cttttctggt ccatctggtg cnggaaaagg aacagttcga aaggnaattt ntgaa 355 16 337 DNA Streptococcus suis iri 23 16 gntctngtag tagataagat tgaaacgcca aattcntcgt aacaaaacca agattgaacg 60 aaagcntcgt caaaaagtgg caactggtca agtctttaca gatgaacttg ttgngcaaac 120 aggcgaggaa gtaaaagtgg ttcgtactaa gcaagtagac ttgaaaccaa tggatatgga 180 agaagcagtc ctccaattgg agttgctcgg acatgatttc tttatctata cagatgctaa 240 tgacggtaca acaaatgtat tgtatagacg cgaagatgga gatttgggtc ttctagagnt 300 acgtcaataa agataataaa acagcncnan cgnannn 337 17 350 DNA Streptococcus suis iri 24 17 agaagagaaa tgggggaacc tggnagttct acaagaaatt agttttgaag aacaggacgg 60 ggctagtcta tttgcgaaaa cgcaagagtt cagcaaattc ttgctttttt tgatataatg 120 gtagaagcag ttttaagagg tatccaggta tgaatattca acaattacgc tacgttgtag 180 ccattgcaaa cagtggtaca tttcgagagg cggctgagaa aatgtangtg tcccagccta 240 gntngtccat ttccattcgt gatttggaaa aagagttagg ttttcaaatt tttngccgaa 300 ctagttcagg aacttttttg acacaaaaag ggatggnact cacggagata 350 18 333 DNA Streptococcus suis iri 29 18 gcgtatgggg gaaattgccc aagatgttan ccggtgacaa aatcacagat gcagcccgta 60 atcangcaaa agaattagta gaaaaaggtt nggcgactag cctttttgct catgtccaaa 120 gtttattttt tagttaaaat ttgttataat agatggcaaa attgaagaga attgtaggnt 180 gaaatatgtc aaagattaag attgttacgg attcaagtac gactatcgaa cccagtttgg 240 tcgaagaatt gaatataaca gttgttccnt tatctgtaat ggttgacgga gtcgtatact 300 ctgacaacga tttaaaagaa ggcgaantcn tag 333 19 341 DNA Streptococcus suis iri 3 19 aggntgctan gaaaaaattg gctcacaaat catttctttt anttgacgat tgcctttctt 60 tngatttngg tgatttactt tagtggaata gataaacgtt ggattatttt ggcaagtttt 120 cttnacttca ttccatcgca gattttatac cgtcgtcgcc taagagagcg actccaagaa 180 gaccagccca agnaggcngg ttttttgatg tgtaaattgg actacaattc tttattaact 240 gtgctataat agtttttgca gaaaagtaaa gacggnggct ctaatttctg aaaggtaggt 300 ggtgtctatg ggcaaatcat cnaaatctna cagaaaggag n 341 20 264 DNA Streptococcus suis iri 32 20 gaatcgaatt ggagntcgcc cctcaaacgg ctggcatatc ttttcaatcc ttatctntna 60 gtcgcaagcg acaagganta gggatnatat aatctcctga gaatactgga ctcactgagt 120 ctggtatttt cattttatgc tataatggtt tcatgacaaa tcgaatttta gatatggaac 180 aaatgcagga cgaggaatat gtcgngcgta ccctgcgtcc ncagaaatta aacgaataca 240 tcggtcagga caaggttaag gacc 264 21 338 DNA Streptococcus suis iri 34 21 acagtagcct atgaantctt ggaagnaagc gggaagaagc aaaccattag tttcgaccaa 60 attttagttc ccataggagg aggtggtctg gttgcaggcg tttcggccta tntgaaagaa 120 catgcacctg aaattangat tgttggtgtt gaagcaagtg gggcacggtc aatgaaagcg 180 gctttngata aaggtcgtcc ggttnaatta gaccaaattg ataaatttgc tgacggtatt 240 gcggtacaga aagtcggtaa gtcgacctac gaagtggctc ggaaatacgt agatcgcctg 300 attngtntgg atgaagggtg gatttccggg antatttt 338 22 351 DNA Streptococcus suis iri 4 22 aaaatggcag gggggaccca agggaantct tttctgatat caagggacaa cctggtcagt 60 cagntggntc aantacaagc cttaccactn gaacaaatan tcgaaaaccg ttatcaacgc 120 tttagaaaat antaggaaga cctagnattt ttttgataga tttgatacaa tggataaaat 180 aatttcagga ggttttccat gttagtaaaa gcagatctat caaacgcagn agaattgcta 240 cntattcagc nccgagcatt tgcggcttta tataaaacct atcaggacca gtacaaccct 300 gccattgnaa ctatggacta tttccaatca cgctttgcac gaccaaattg t 351 23 362 DNA Streptococcus suis iri 7 23 gaaattgatg ggcatctttg gtattaatag gaactccatg gctcaatctt cttcggttta 60 ttggtaatag tagttaccgt cangaanaaa tcgcaaagta taaaaagtgc tgtgaagaga 120 aaaaaagaaa ataagaatct ttctaaacaa gataagagcc gtcaggctct tttttngata 180 taatatagtg gatatggtta attaaaattg tcagaaaaga ctattttana gattaacact 240 ctctgaaaat cntcattaac aagaaaagag gcngggctca agccccgcat cacntctcaa 300 agttagcgtc aacatctcag cgcagtagtg gtngattggg tttaacagtc cagtggagtg 360 tc 362 24 362 DNA Streptococcus suis iri 8, 26 24 ttcggaatcc ttctntctcc attggaacag ggatacaaag ggacgttaag gaaatccgta 60 ngaaaatagg aaattgacgc agtgtgctan acacacaggg aagtttatct ttttccacta 120 ggattttagt ccgtgttcaa ctaagatacg agatatgttc tggtttacca gaaatttcng 180 nagaaaatta ggagactgac gctgagtgtt aacactcaag gaaggctatc tatttttcta 240 agaaattaat ctcgagttca atttcttntg attagtaaat aaatgaattg tatctatttt 300 ttggggtatc gccaagcggt aaggcaaggg actttgactc cctcatgcgc cggttngcat 360 cc 362 25 405 DNA Streptococcus suis ivs 1 25 aatgatgttt gataaacacg ccaatctcaa atacaaattt ggtaatcgtc atttctgggc 60 agagggatat tatgtaagta cggttggact aaatgaagcc acaattaaga aatatataca 120 agaacaggaa aaacatgata tagcacttga taagttgagt gtaaaagagt atgaagatcc 180 ctttagggat aatggcaagt agtacgaatg cctctttaag aggctagtga cgagtcaaaa 240 gcagtgaggc ttgaacaaag tgaaagccag cgtctttagg cgctggctgg tgatgtgggc 300 ttatagccct tgttcaaacc acccgtttga cgggtggtca tgattttttt tgaatatttt 360 tcactatttt gttttacaaa ctagccacct tgtgttagac tatag 405 26 410 DNA Streptococcus suis ivs 11 26 taccaccata tcaccaatat cacgcgccca gatgcgccaa tcgaagtggt ggatgtggct 60 ggttcccttt gtgaaaacaa cgacaagttt gcggtcaatc gtgaattacc acgggtagaa 120 gtaggagaca ccttggtcat tcatgacagt ggggcccacg gcttctccat gggctacaac 180 tacaacggtc gtctgcgttc ttctgaaatc cttttgcagg aagatggcac agcgcggatg 240 attcgtcgtg ctgaaacacc agaagactat ttcgcaacta tttacggttt tgattttgac 300 aggtaagtct tggaaaagac tagggaattt ggtataatag ggttattgaa agattgttaa 360 aaacaatcag aagtatactt tttagaagag tcaggagatt gacagatgaa 410 27 412 DNA Streptococcus suis ivs 15 27 gggctatggt ataattaaaa gacatgtata gtcagaatga aaatgaattg attgccattg 60 gtgagagaat tggaaaggcc tgtaagccaa atcaagttct agtattatca ggggatttgg 120 gtgctgggaa aacaactctg accaagggtt tggccaaggg gttaaaaatt gaacagatga 180 ttaagagtcc tacttatacg attgttcgag agtatgaggg ggccatgccg ctctatcact 240 tagatgttta tcgaattgga gatgaccctg actcgattga tttggatgat tttctctatg 300 gaggaggtct aacggttatc gagtggggag aattactgga tgtcagtcta tttgatgact 360 atttgctcat tcgtatagag aaagagggag atggtcgacg attgacagtc ga 412 28 449 DNA Streptococcus suis ivs 16 28 gaaaattgtt gttgttttgg aacactagta gaccagaggc ttctagtaag gtagttgtgc 60 tcactgagga gggggaagga tgatggaagt tgagaaaagg agtaaggatt atgctcgtat 120 gtttgaccag caagtcggtc tttatgaaga ctatgctcgt ggacatggac tcaatgcaaa 180 atgtttatcc attctcatgt ggatttatta taatcccgga ggtgtgacgc aaaactgggt 240 cagtaagaag acctattcaa gcaaacaagt tgtcaatgct actgtaaaga aatttttgga 300 tggaggcctg gtagttctag aggagaatcc agcagataag cgacataaga aaattaaatt 360 gacagaggag gggcaacaat ttgctagtcg cattttggat cccttagagg aggcggaaaa 420 taaggcgctg tctcaactca gtcaggagg 449 29 410 DNA Streptococcus suis ivs 18 29 gcgttttgga acaagtacgt taagagaaac ctagagaaat ctagggtttt tgcttttata 60 tatctttaca ttgtttaaag aaaatagcat ttcaaaaact ttttgaaaaa aatgtgatat 120 tctgagcata ttttttgaaa tcggtaacat ttatattgta taatatagtt cgtaaaaaaa 180 tatattttcg aaagtgagat tttacattat ggctaaaatc gttgttgtcg gtgctaacca 240 tgctggtact gccgcaatca aaactatgtt gacaaattat ggtcaagaaa atgaaatcgt 300 tgtatttgac caaaactcac atatttcatt cttgggttgt ggtatggctt tgtggatcgg 360 tgagcaaatt ggcggtcctg aaggactctt ctactcaaac aaagaagagt 410 30 437 DNA Streptococcus suis ivs 19 30 tcgttccatt tgctggtgaa atgcccagca atacgcttcn tagcaataga agaaccaaat 60 agatggcact caatttcatg aggaagaaca gaagagtaaa aagcctgtct aaccacccta 120 acatagnata ttcctccttt ttcatctatt ttatcaaaaa atcggtgctt ttctaccatt 180 tgtcaagttc atcaaggtat ttgacgaaaa atattttgtg tctcgtcatc caaataagga 240 aattgtttta ttttggacta aagttacgtg taaaaagtgc atacaaaacc aacaccttat 300 gttgaaattt tttgataagg tgttacaatg atagagcata aacagtttta ccgattttgg 360 gttgaagcgt aatcgtaaaa tttgttatgc ataatgaggt aatacattgt ccgaatgaga 420 cgatgtatgg aggcaat 437 31 417 DNA Streptococcus suis ivs 2, 4, 28 31 aagacggcgt caaggatgac aatcttgtgg tgacgaccac ccagaaactg gcgtagcntt 60 taccgtggcc ggaatcatga tcgcggtttt ccagccgttc atgcagttcg gttgttgctt 120 tgaacagcaa gaatatcccc ccgaacaaca taatcaggtc gcgtccggag aaggagaaat 180 ccatgacggt aaaatagcgg tttggtcagc gtgaccatcc atgaaatcag cgacagcagc 240 cccagacgca taatcagcgc cagtgataac cccagcaaac gcgctttatc gcgttgtttt 300 ggcggcagtt tgtcagcaag aatggcgatg aagaccaggt tatcgatacc cagcacaatt 360 tcgagaacaa caagcgtgag tagccccgcc caaattgagg ggtccattaa gaattcc 417 32 444 DNA Streptococcus suis ivs 20 32 agttcagatg ttcggtttag gaattgccgg cgtctggctg tcgattttga tggacctgct 60 cttgcgagcg atttttctga cttggaggtt tattgtgcaa acacgaaaac tggctgaata 120 ggctagtttt ttggtataat atcagtagaa tgataaaaag gagataatca gatgaaaacc 180 attcacacag ataaggcacc tgcagcaatt ggcccatacg ttcaagggaa ggttgttgga 240 aatttcctat ttgcctctgg tcaagttcct ttgtcacctg aaactggtga agtggttggt 300 gaaaccattc aggagcagac tgagcaagtc ttgaaaaata tcgcagcaat tttatcagaa 360 gcaggaacag actttgacca tgtggtgaag acgacttgtt tcctaaaaga tatgaatgat 420 tttgtagcct ttaatgaagt ttat 444 33 480 DNA Streptococcus suis ivs 23, 24 33 tctgcactgt tgcgctgcct ataagttcta cgttcagtag tagatgaaat gttcagagga 60 agtggtatgg gttccaactt agtaaaatta gtcattgatg atttggcgaa cagaaattcc 120 aaagcctttc aaatcgcagt tgaagaagag aaattgggaa cctggaagtt ctacaagaaa 180 ttagttttga agaacaggac gggctagtct atttgcgaaa acgcaagagt tcagcaaatt 240 cttgcttttt ttgatataat ggtagaagca gttttaagag gtatcaggta tgaatattca 300 acaattacgc tacgttgtag ccattgcaaa cagtggtaca tttcgagagg cggctgagaa 360 aatgtatgtg tcccagccta gtttgtccat ttccattcgt gatttggaaa aagagttagg 420 ttttcaaatt tttagccgaa ctagttcagg aacttttttg acacaaaaag ggatggaatt 480 34 418 DNA Streptococcus suis ivs 25 34 ggagatagca atgcttaata tttttgtatt agaagatgat ttttttcagc agagcaggtt 60 agaaaatgct attaggcagt gtgttgaaga aacgtcagta aggtataaat tcctagaagt 120 ttttggtaaa ccaaatcaat tattggaatc aattgaggaa gcagggaatc atcaattttt 180 ctttttagat attgaaataa aaggagaaga aaagaaagga atggaaatcg ctaaagaaat 240 ccgggctcga gatccttatg ctgctattgt ctttgtaaca actcactcag aatttatgcc 300 agtaacatat cgttatcagg tttctgcttt agattttata gataaaggcc tggaggatng 360 tgactttcaa aaggcagtat cagatgtctt agtgcatgct tttgaaaata ttgatcat 418 35 446 DNA Streptococcus suis ivs 29 35 ggcaagggtg ggtaaatttc taattggtga caaggcactt gaattctacc cagatagcaa 60 cgttgaacgc tatatccaga ttccttggtc agaaatgact agcattggcg caaaacgttt 120 ctggcaaagc aatcagccgt cattttgaaa tttatacaga gaaaagtcga tttcttgttg 180 gcatctaaag attctggtaa gattcttaaa attgcccgtg agcatatcgg caatgaaaaa 240 gttgtgaaat taccgactct tatgcaaaca atcggcagaa aaatttcgaa tctatttgcc 300 aaaaaataaa aattcaagta taatagtaga aacggataag tagcatctgg ctccttccag 360 aaagtctgcg gtcgctgtga gcagatagga aaaagttgtg aaattctacc gttatgaaat 420 tatcaaaata caatcaagtg cacaga 446 36 416 DNA Streptococcus suis ivs 3 36 ggattatcta ctataagcag tattcagaag ggcatgagga caagaaatcc tacaagattc 60 tacaagaagt aggcatgagc cagaaggctg tcaagaaaac aattaactcc caaacactta 120 cggtcttctt tatgcctttg gtcatggcga ccctacactt tgtcatcgcc cttatcatgc 180 tcaagcaaat gctactaagt tttggtgtta cctcatcact aatgatttac acagtcagtg 240 gcatcaccct actggcagtc actctgattt actttgtcat ttacaagtgg actagtcgca 300 cttattatcg cattattgaa cggtagcaga agtctcgcct tgtgcgagat ttcttgcttt 360 ttcagggaaa tggtgttaca atggtaatac caaaggaata ctcgaagagg tgagaa 416 37 263 DNA Streptococcus suis ivs 31 37 acgaaaatdg atggatccat gcataaactg catcccttaa cttgtttttc gtgtgcctat 60 tttttgtgaa tcgaattcga gctcgcccct cctgaccacc tatntgcatc aagtgccaaa 120 tgaccagtcg agtgtgcggt tagacaacta ctatacgggc aaggaactgg agattgagtt 180 ggatgtggct ttgactccta gccaaaatgc ccagcggtac ttcaagaagt accagaaact 240 caaggaggcg gtcaagcacc tga 263 38 403 DNA Streptococcus suis ivs 32, 35 38 atatttgctc tcctgctctt taggggacaa tggaaaaagt agtctgtatc caacatttta 60 caaagtagga ttttttctat aaaatagatt gtatatgaca ttcaaatcca ttctcaaaca 120 actcaaacta tttgattata tcttaatcgg attcacccta gttttatcct ttcttccagc 180 aatttttacc tacacacaac tgacaacaga tgcaaatgag gcaaaaacaa ttgcctatgt 240 ccgcatcaat ggtgaggtgg tcgaccaatt tgaattatca aaggacacac cccgtcaaga 300 aaagacctac tatcccaatg aagggcaata caatatcatt gaagttgatg gcgaacgcat 360 tcgtgtcaag gaagacaata gcccagacca aatcgccgtt atg 403 39 401 DNA Streptococcus suis ivs 33 39 actcagttga acggagtagg atttataggt aaattgcctc caaatatcgt aagacaatcc 60 tctattgaaa aataggggat tgtttgttta gaaataatgg tggagattct gtaaaaagcg 120 aaagtggttg gaaagttagg gtttagccga gaaaaagaga cttttctatc tatctttcac 180 aattttctgt caatttgtgg tagaatagaa aaaatagatt ttttatgagg gataccatga 240 cattagtata tcaatcaaca cgcgatgcta aaaatactgt atcggctagt caagcgattt 300 tgcagggctt ggcgaccgac ggtggtttgt ttacaccgct ttctattcca acagttgact 360 tggatttttc tgttttgaaa gatgcttctt atcaagacgt t 401 40 404 DNA Streptococcus suis ivs 34 40 gtttatcgtt cgctggagga aaagggctat aatccgatta accaaatcat tggctatgta 60 ttaagtgggg accctgctta tattcctcgc tataatgatg cccgcaatca gattcgtaag 120 catgaacgag atgaaatcat tgaagaattg gtgcgctact atttgaaagg gaatgggatt 180 gacctctaat gagaataatg ggattagacg tcggttccaa gacagttggt gtagccattt 240 cagatccgtt aggtttcacg gcccaagggt tggaaatcat cccaatcgat gaagaaaagg 300 gcgaattcgg tctggagcgt ttgaccgaac ttgtagaaca gtacaaggtt gataaatttg 360 ttgtaggctt gccgaagaat atgaataata ctagtggtcc acgt 404 41 384 DNA Streptococcus suis ivs 36 41 ggtataatta tctgataaaa aactttggag acgacagtga gtttagaaaa ttacatgccg 60 gattttgcct tggaaaaggc ttatgacgtg accgtcgaaa gcttgaaaaa acatggcata 120 aaagtagtgt ttgttgactt ggataatacc ttgattgctt ggaataatcc cgatggtacg 180 ccagagatgc gccagtggtt acatgatttg caggacgcag gtattcctgt tgtggtggtg 240 tctaacaata aatacgaacg tgtcaaacgg gcggttgaaa aatttgggat tgaatttgaa 300 gccttcgctc tcaagccttt cacctttggg attaaccgtg ctttgaaacg ctttgatgtc 360 cagccgtatg aggtaattat gatt 384 42 413 DNA Streptococcus suis ivs 5, 10, 12, 22 42 acgcacttgc tcgcgtagtc gatgaattag atgtacccgt tatggctttc ggtcttaaaa 60 atgatttccg aaatgaacta tttgaaggtt cccaacattt gctcttattg gctgataaat 120 tagatgaaat caaaacaatc tgccaatatt gttctaaaaa agcgacaatg gttttgagaa 180 cacaggatgg aaaacctact tatgaaggag aacaaatcca aattggtggc aatgaaacct 240 acattcctgt ctgtcgcaaa cattattttt caccagaaat taaagattta ccctaatttt 300 tgaaaatgaa atgagaagca actgtaaact gagcaactat atagaactga atttgcctat 360 gactctgtgc caattttcat aacttacata ctacggcaaa ggaattgaac acg 413 43 428 DNA Streptococcus suis ivs 6, 7, 13, 14 43 gaagggatta aacaatccta tgctattcag gctgttcgtg aaattcggat tatcgttcat 60 cctaacaagg tcactgatga tcagattacc atcttggccc atgatgttcg tgagaaaatt 120 gaaaataatc tggattatcc aggaaatatc aaaatcacag ttatccgtga aacaagagca 180 acagatgttg ctaagtaaat gtagaagagt ccgaagggct ctttttctac tggctcaaag 240 ttcgttttgg gttgggaata gaaaatagaa aatattttaa tcgtatttaa aagcagttga 300 aattcatgct aaattttgtt acactagaat gaaagattta aaaggagata tcatgaaaga 360 gcgaggctta ctcattgtct tttctggtcc atctggtgcc ggaaaaggaa cagttcgaaa 420 ggaaattt 428 44 383 DNA Streptococcus suis ivs 8 44 cttcaaagga ccccaggacc tttgaattct caaatacgca tcatgttgac agttgccaca 60 cctacaccaa aatcaaatgc caacaagcgt tgagtcgggt aatagcgtaa gtagcgcaag 120 gtcatgataa gctgctcttc catacttaga cggcgtgggc gtcctccttt tcggtgttgc 180 tcttgataag cgtcagtgag acaatcaagc atcagatgaa acgtcgcttt tttacaccta 240 tcaacaattt gaaattctct gagtttaatt ttaagacttt ttcgtatgtt gtttccatac 300 ctttagtata ccgcctttga gttaccgaac aagtctattg ctaaacttga tgaaggttgt 360 attgtctgtt ataatattgg ata 383 45 415 DNA Streptococcus suis ivs 9, 17 45 gcctatgaga ctcattttcc ctgtctcaac tgctctaagc aattgttaca ggttggttgt 60 aagcgggttg tctatatcaa tgaataccgc atggatgact atgctcagta cttgtataaa 120 gaaaagggct gtgagttggt tcatttgcct ctagaggtgg ttaaacaggc atttgcagat 180 gccgaattta tctaatgatt ttgtagaaga gtggttgcat agaacaaccc tctttatctt 240 taagaaaatg ctaggatagt cggtcaatct atgctatact agaaacgtta ttaagtcccg 300 aaaaggtagt ttatagacta gttaatattt gcagaaacac ttgaaacaca attaaagaaa 360 ctggtaatat tgaatagtaa gcgtaaaaac tttactacac ttcagtcact atttt 415 46 45 DNA artificial sequence PCR primer corresponding to positions 250 to 273 of the fbps gene 46 gcggatccga tgacgatgac aaatcttttg acggattttt tttac 45 47 32 DNA artificial sequence PCR primer corresponding to positions1911 to 1892 of the fbps gene 47 cccaagcttg ggcatgaact agattttcat gg 32

Claims (20)

What is claimed is:
1. A process for modulating virulence of a Streptococcus comprising:
modifying a genomic fragment of the Streptococcus;
wherein at least part of the genomic fragment is capable of hydridizing to a nucleotide sequence selected from the group of nucleotide sequences consisting of any one of SEQ ID NOS: 8-45 or fragments thereof; and
generating a clone having the modified genomic fragment.
2. The process according to claim 2, wherein the genomic fragment comprises a functional part of a gene, the expression of which can be environmentally regulated by iron-restricted conditions in Streptococcus suis.
3. The process according to claim 1 or 2, wherein the genomic fragment comprises a functional part of a wild-type Streptococcus suis gene expressed in a pig infected with wild-type Streptococcus suis.
4. The process according to claim 3, wherein the wild-type Streptococcus suis gene encodes a fibronectin/fibrinogen-binding protein.
5. The process according to any one of claims 1 to 4, wherein the Streptococcus is Streptococcus suis.
6. The process according to any one of claims 1 to 5, wherein modifying the genomic fragment comprises functionally deleting the at least part of the genomic fragment capable of hydridizing to the nucleotide sequence.
7. A clone of a Streptococcus, obtained by the process according to any one of claims 1 to 6.
8. The process according to claim 1, wherein the genomic fragment encodes a fibronectin/fibrinogen-binding protein.
9. A process for assaying virulence of a Streptococcus comprising:
assaying an ability of the Streptococcus to infect a subject;
wherein the Streptococcus comprises a genomic fragment associated with a virulence factor to infect a subject; and
wherein at least part of the genomic fragment is capable of hydridizing to a nucleotide sequence selected from the group of nucleotide sequences consisting of any one of SEQ ID NOS: 8-45 or fragments thereof.
10. The process according to claim 9, wherein the genomic fragment encodes a fibronectin/fibrinogen-binding protein.
11. An isolated or recombinant nucleic acid molecule of a Streptococcus origin comprising a nucleotide sequence capable of hybridizing to a nucleotide sequence selected from the group of nucleotide sequences consisting of any one of SEQ ID NOS: 8-45 or fragments thereof.
12. A vector comprising the isolated or recombinant nucleic acid molecule of claim 11.
13. A host cell comprising the isolated or recombinant nucleic acid molecule of claim 11 or the vector of claim 12.
14. The host cell of claim 13, wherein the host cell is of a Streptococcus origin.
15. A vaccine comprising the clone of claim 7, the isolated or recombinant nucleic acid molecule of claim 11, the vector of claim 12 or the host cell of claim 13 or 14.
16. A protein or fragment thereof, encoded by the isolated or recombinant nucleic acid molecule of claim 11.
17. An antibody directed against the protein or fragment thereof of claim 16.
18. An antigen comprising the protein or fragment thereof of claim 16.
19. A diagnostic test comprising the antibody of claim 17.
20. A vaccine or diagnostic test comprising the antigen of claim 18.
US10/632,117 2001-02-02 2003-07-31 Environmentally regulated genes of Streptococcus suis Expired - Fee Related US7927605B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/982,192 US8324354B2 (en) 2001-02-02 2007-10-31 Environmentally regulated genes of Streptococcus suis

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP01200419 2001-02-02
EP01200419 2001-02-02
EP01200419.8 2001-02-02
PCT/NL2002/000073 WO2002061070A2 (en) 2001-02-02 2002-01-31 Environmentally regulated genes, involved in the virulence of streptococcus suis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2002/000073 Continuation WO2002061070A2 (en) 2001-02-02 2002-01-31 Environmentally regulated genes, involved in the virulence of streptococcus suis

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/982,192 Division US8324354B2 (en) 2001-02-02 2007-10-31 Environmentally regulated genes of Streptococcus suis

Publications (2)

Publication Number Publication Date
US20040096973A1 true US20040096973A1 (en) 2004-05-20
US7927605B2 US7927605B2 (en) 2011-04-19

Family

ID=8179856

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/632,117 Expired - Fee Related US7927605B2 (en) 2001-02-02 2003-07-31 Environmentally regulated genes of Streptococcus suis
US11/982,192 Expired - Fee Related US8324354B2 (en) 2001-02-02 2007-10-31 Environmentally regulated genes of Streptococcus suis

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/982,192 Expired - Fee Related US8324354B2 (en) 2001-02-02 2007-10-31 Environmentally regulated genes of Streptococcus suis

Country Status (8)

Country Link
US (2) US7927605B2 (en)
EP (1) EP1368373B1 (en)
JP (1) JP4275413B2 (en)
AT (1) ATE507238T1 (en)
AU (1) AU2002230275A1 (en)
CA (1) CA2436530C (en)
DE (1) DE60239858D1 (en)
WO (1) WO2002061070A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080241181A1 (en) * 2001-02-02 2008-10-02 Id-Lelystad, Instituut Voor Dierhouderij En Diergezondheid B.V. Environmentally regulated genes of Streptococcus suis
CN100449005C (en) * 2006-06-29 2009-01-07 中国检验检疫科学研究院动植物检疫研究所 Multiple fluorescence PCR detection reagent for detecting pathogenicity of streptococcus suis serotype 2 and method
CN100449006C (en) * 2006-08-10 2009-01-07 中国检验检疫科学研究院动植物检疫研究所 Primer and probe sequence for pig streptococcus 2 type ectofactor (EF) fluorescence PCR detection
US20100297183A1 (en) * 2007-08-06 2010-11-25 Elizabert Hilda Smith Immunogenic streptococcus proteins
US20110171252A1 (en) * 1998-07-22 2011-07-14 Stichting Dienst Landbouwkundig Onderzoek Streptococcus suis vaccines and diagnostic tests
CN112368373A (en) * 2018-06-04 2021-02-12 尹特荣生物科技株式会社 Novel streptococcus suis bacteriophage STR-SUP-3 and application thereof in inhibiting streptococcus suis bacterial proliferation

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1205552A1 (en) 2000-11-09 2002-05-15 ID-Lelystad, Instituut voor Dierhouderij en Diergezondheid B.V. Virulence of streptococci, involving ORFs from Streptococcus suis
WO2006048757A2 (en) * 2004-11-05 2006-05-11 Pharmacia & Upjohn Company Llc Anti-bacterial vaccine compositions
CN109771446B (en) * 2019-02-28 2020-11-17 北京大学人民医院(北京大学第二临床医学院) Application of streptococcus salivarius K12 in preparation of medicines for preventing and/or treating autoimmune diseases

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5610011A (en) * 1991-03-21 1997-03-11 Centraal Diergeneeskundig Instituut Virulence-encoding DNA sequences of Strepococcus suis and related products and methods
US6660852B1 (en) * 1994-09-30 2003-12-09 Fuso Pharmaceutical Industries, Ltd. Probe for diagnosing infectious diseases
US7109006B2 (en) * 2000-11-09 2006-09-19 Id-Lelystad, Instituut Voor Dierhouderij En Diergezondheid B.V. Virulence of Streptococci
US7125548B2 (en) * 1998-07-22 2006-10-24 Stichting Dienst Landbouwkundig Onderzoek Streptococcus suis vaccines and diagnostic tests
US20080241181A1 (en) * 2001-02-02 2008-10-02 Id-Lelystad, Instituut Voor Dierhouderij En Diergezondheid B.V. Environmentally regulated genes of Streptococcus suis
US7670548B2 (en) * 2007-06-18 2010-03-02 Mk Electron Co., Ltd. Lead free solder containing Sn, Ag and Bi

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5542712A (en) 1991-12-18 1996-08-06 Itt Corporation Quick connector housing with elongated barb design
FR2688515B1 (en) 1992-03-13 1995-03-31 Institut Rech Agronomique THERMOSENSITIVE PLASMID.
US5928900A (en) 1993-09-01 1999-07-27 The Rockefeller University Bacterial exported proteins and acellular vaccines based thereon
EP0804582A1 (en) 1994-05-16 1997-11-05 The Uab Research Foundation $i(STREPTOCOCCUS PNEUMONIAE) CAPSULAR POLYSACCHARIDE GENES AND FLANKING REGIONS
WO1996021485A1 (en) 1995-01-11 1996-07-18 Giltech Limited Heating of physiological fluids
US5681570A (en) 1995-01-12 1997-10-28 Connaught Laboratories Limited Immunogenic conjugate molecules
AU4431196A (en) 1995-01-23 1996-08-14 Novo Nordisk A/S Dna integration by transposition
EP0750042A1 (en) 1995-06-20 1996-12-27 Societe Des Produits Nestle S.A. Exopolysaccarides-producing lactic acid bacteria
EP0750043B1 (en) 1995-06-20 2001-05-23 Societe Des Produits Nestle S.A. Exopolysaccharides-producing lactic acid bacteria
WO1998019689A1 (en) 1996-11-01 1998-05-14 Smithkline Beecham Corporation Novel coding sequences
US6582706B1 (en) 1998-12-21 2003-06-24 Medimmune, Inc. Vaccine compositions comprising Streptococcus pneumoniae polypeptides having selected structural MOTIFS
EP2023143A1 (en) * 2007-08-06 2009-02-11 Boehringer Ingelheim Vetmedica Gmbh Immunogenic streptococcus proteins

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5610011A (en) * 1991-03-21 1997-03-11 Centraal Diergeneeskundig Instituut Virulence-encoding DNA sequences of Strepococcus suis and related products and methods
US6660852B1 (en) * 1994-09-30 2003-12-09 Fuso Pharmaceutical Industries, Ltd. Probe for diagnosing infectious diseases
US7125548B2 (en) * 1998-07-22 2006-10-24 Stichting Dienst Landbouwkundig Onderzoek Streptococcus suis vaccines and diagnostic tests
US20070111236A1 (en) * 1998-07-22 2007-05-17 Id-Lelystad, Instituut Voor Dierhouderij En Deirgezondheid B.V. Streptococcus suis vaccines and diagnostic tests
US7109006B2 (en) * 2000-11-09 2006-09-19 Id-Lelystad, Instituut Voor Dierhouderij En Diergezondheid B.V. Virulence of Streptococci
US20080241181A1 (en) * 2001-02-02 2008-10-02 Id-Lelystad, Instituut Voor Dierhouderij En Diergezondheid B.V. Environmentally regulated genes of Streptococcus suis
US7670548B2 (en) * 2007-06-18 2010-03-02 Mk Electron Co., Ltd. Lead free solder containing Sn, Ag and Bi

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110171252A1 (en) * 1998-07-22 2011-07-14 Stichting Dienst Landbouwkundig Onderzoek Streptococcus suis vaccines and diagnostic tests
US8728490B2 (en) 1998-07-22 2014-05-20 Stichting Dienst Landbouwkundig Onderzoek Streptococcus suis vaccines and diagnostic tests
US20080241181A1 (en) * 2001-02-02 2008-10-02 Id-Lelystad, Instituut Voor Dierhouderij En Diergezondheid B.V. Environmentally regulated genes of Streptococcus suis
US8324354B2 (en) 2001-02-02 2012-12-04 Stichting Dienst Landbouwkundig Onderzoek Environmentally regulated genes of Streptococcus suis
CN100449005C (en) * 2006-06-29 2009-01-07 中国检验检疫科学研究院动植物检疫研究所 Multiple fluorescence PCR detection reagent for detecting pathogenicity of streptococcus suis serotype 2 and method
CN100449006C (en) * 2006-08-10 2009-01-07 中国检验检疫科学研究院动植物检疫研究所 Primer and probe sequence for pig streptococcus 2 type ectofactor (EF) fluorescence PCR detection
US20100297183A1 (en) * 2007-08-06 2010-11-25 Elizabert Hilda Smith Immunogenic streptococcus proteins
CN112368373A (en) * 2018-06-04 2021-02-12 尹特荣生物科技株式会社 Novel streptococcus suis bacteriophage STR-SUP-3 and application thereof in inhibiting streptococcus suis bacterial proliferation

Also Published As

Publication number Publication date
EP1368373A2 (en) 2003-12-10
US7927605B2 (en) 2011-04-19
CA2436530A1 (en) 2002-08-08
AU2002230275A1 (en) 2002-08-12
ATE507238T1 (en) 2011-05-15
WO2002061070A2 (en) 2002-08-08
US8324354B2 (en) 2012-12-04
CA2436530C (en) 2013-10-01
WO2002061070A3 (en) 2003-04-17
DE60239858D1 (en) 2011-06-09
WO2002061070A8 (en) 2003-11-06
JP4275413B2 (en) 2009-06-10
EP1368373B1 (en) 2011-04-27
US20080241181A1 (en) 2008-10-02
JP2004528826A (en) 2004-09-24

Similar Documents

Publication Publication Date Title
de Greeff et al. Contribution of fibronectin-binding protein to pathogenesis of Streptococcus suis serotype 2
US8324354B2 (en) Environmentally regulated genes of Streptococcus suis
US8071111B2 (en) Virulence of Streptococci
JP2008022856A (en) Nucleic acid and protein derived from streptococcus pneumoniae
JP2011097938A (en) Antibacterial vaccine composition
PL204863B1 (en) Streptococcus suis
CA2105057C (en) Dna sequences which code for virulence characteristics of streptococcus suis and part thereof polypeptides and antibodies derived therefrom and the use thereof for the diagnosis of and protection against infection by s. suis in mammals, including man
US6669940B2 (en) Recombinant fusobacterium necrophorum leukotoxin vaccine and preparation thereof
WO2004020609A9 (en) Streptococcus pneumoniae antigens for diagnosis, treatment and prevention of active infection
AU2903700A (en) Vaccine
AU2010201539C1 (en) Anti-bacterial vaccine compositions
AU2013201267B2 (en) Anti-bacterial vaccine compositions
Abeyta et al. Genetic Alteration of Capsule Type but Not
AU2007242960A1 (en) Anti-bacterial vaccine compositions

Legal Events

Date Code Title Description
AS Assignment

Owner name: ID-LELYSTAD, INSTITUUT VOOR DIERHOURDERIJ EN DIERG

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:SMITH, HILDA ELIZABETH;REEL/FRAME:014818/0220

Effective date: 20030804

AS Assignment

Owner name: ID-LELYSTAD, INSTITUUT VOOR DIERHOUDERIJ EN DIERGE

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT ASSIGNEE ADDRESS, PREVIOUSLY RECORDED ON REEL 014818 FRAME 0220;ASSIGNOR:SMITH, HILDA ELIZABETH;REEL/FRAME:015601/0906

Effective date: 20031223

AS Assignment

Owner name: STICHTING DIENST LANDBOUWKUNDIG ONDERZOEK,NETHERLA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ID-LELYSTAD, INSTITUUT VOOR DIERHOUDERIJ EN DIERGEZONDHEID B.V.;REEL/FRAME:023998/0642

Effective date: 20091207

Owner name: STICHTING DIENST LANDBOUWKUNDIG ONDERZOEK, NETHERL

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ID-LELYSTAD, INSTITUUT VOOR DIERHOUDERIJ EN DIERGEZONDHEID B.V.;REEL/FRAME:023998/0642

Effective date: 20091207

FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20150419