WO1993018153A1 - Vaccinia virus b15r used in interleukin b1-involving condition - Google Patents

Vaccinia virus b15r used in interleukin b1-involving condition Download PDF

Info

Publication number
WO1993018153A1
WO1993018153A1 PCT/GB1993/000460 GB9300460W WO9318153A1 WO 1993018153 A1 WO1993018153 A1 WO 1993018153A1 GB 9300460 W GB9300460 W GB 9300460W WO 9318153 A1 WO9318153 A1 WO 9318153A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
interleukin
receptor
binding
vaccinia virus
Prior art date
Application number
PCT/GB1993/000460
Other languages
French (fr)
Inventor
Geoffrey Lilley Smith
Original Assignee
Geoffrey Lilley Smith
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 Geoffrey Lilley Smith filed Critical Geoffrey Lilley Smith
Priority to AU36418/93A priority Critical patent/AU3641893A/en
Priority to EP93905517A priority patent/EP0656949A1/en
Publication of WO1993018153A1 publication Critical patent/WO1993018153A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • the present invention relates to vaccinia virus proteins, uses and products relating thereto.
  • IL-1 ⁇ receptor is used in the text as referring to the vaccinia virus B15R polypeptide or allele or functionally equivalent fragment, derivative or variant thereof, the word "receptor" should be
  • VV Vaccinia virus
  • the linear double-stranded genome of approximately 185,000 base pairs has the potential to encode at least 200 proteins (Moss, B.
  • vaccinia virus encodes many enzymes and protein factors necessary for transcription and replication of its genome.
  • the virus also encodes a variety of factors which modulate virus replication in the multicellular host and aid evasion of the host immune system (Moss, B. (1990a)).
  • Advances in molecular genetics have made possible the construction of recombinant vaccinia viruses that contain and express genes derived from other
  • the recombinant viruses retain their infectivity and express the foreign gene (or genes) during the normal replicative cycle of the virus. Immunisation of animals with the recombinant viruses has resulted in specific immune responses against the protein(s) expressed by the vaccinia virus, including those protein(s) expressed by the foreign gene(s) and in several cases has conferred protection against the pathogenic organism from which the foreign gene was derived.
  • Recombinant vaccinia viruses have, therefore, potential application as new live vaccines in human or veterinary medicine.
  • Advantages of this type of new vaccine include the low cost of vaccine manufacture and administration (because the virus is self-replicating), the induction of both humoral and cell-mediated immune responses, the stability of the viral vaccine without refrigeration and the practicality of inserting multiple foreign genes from different organisms into vaccinia virus, to construct polyvalent vaccines effective against multiple pathogens.
  • a disadvantage of this approach is the use of a virus vaccine that has been recognised as causing rare vaccine-related complications.
  • Vaccinia virus contains two genes designated B15R and B18R from near the right inverted terminal repeat (ITR) which are each predicted to encode a soluble glycoprotein that has three immunoglobulin-like (Ig) domains, but no transmembrane anchor sequence of
  • the product of B18R is apparently expressed on the surface of the infected cell early during infection and antibodies directed against a mixture of it and other proteins confer resistance to virus infection without directly neutralizing infectivity (Ueda, Y., Morikawa, S. and Matsuura, Y. (1990); Ueda and Tagaya, I. (1973) J. Exp. Med. 138, 1033-1043; Ikuta, K., Miyamoto, H. and Kato, S. (1980).
  • IL interleukin
  • gene product B18R does not bind interleukin-1 (IL-1) or interleukin-6 (IL-6).
  • IL-6 interleukin-6
  • gene product B15R does bind IL-1, though not IL-6.
  • this binding is specific for IL- ⁇ as IL- ⁇ and the IL-1 receptor antagonist protein (IL-1ra) do not bind to the B15R gene product.
  • one or more of the gene sequences B15R and B18R may be inactivated, or part or all of one or more of these gene sequences may be deleted from the viral genome to allow (i) greater attenuation of the virus; and/or (ii) enhancement of immunogenicity of recombinant vaccinia virus; and/or (iii) further gene sequences insertion sites, so that more foreign DNA may be included in the virus.
  • Inactivation of the gene sequences may be by mutation or the insertion of foreign DNA.
  • one or more of the gene sequences B15R and B18R may be changed to alter the function of a protein product encoded by the nucleotide sequence.
  • Mutation of the nucleotide sequence may be effected by the deletion, addition, substitution or inversion of one or more nucleotides.
  • DNA sequences encoding one or more heterologous polypeptides may be incorporated in the viral genome.
  • the DNA sequences encoding the heterologous peptides may be inserted into one or more ligation sites created by the deletion or deletions from the viral genome.
  • a heterologous peptide is one not normally coded for by wild type vaccinia virus.
  • the heterologous nucleotide sequence will encode an immunogen or a
  • polypeptide will be substantially homologous to an epitope expressed by a pathogenic organism during
  • the recombinant vaccinia viruses as described have the potential for enhanced immunogenicity. This may result from the deletion of vaccinia genes B15R and/or B18R which cause immunosuppression.
  • the recombinant vaccinia vectors as described may be used as immunogens for the production of monoclonal and polyclonal antibodies or T-cells with specificity for heterologous peptides encoded by DNA sequences ligated into the viral genome.
  • the term antibody as used above should be construed as also covering antibody fragments and derivatives of a parent antibody and which have the same specificity as the parent antibody.
  • the monoclonal antibodies, polyclonal antibodies, antisera and/or T-cells produced by the use of the recombinant virus vectors hereof can be used in the diagnostic tests and procedures, for example, in
  • detecting antigen in a clinical sample can also be used therapeutically or prophylactically for
  • Diagnostic test kits may comprise monoclonal
  • antibodies, polyclonal antibodies, antisera and/or T-cells obtained by the use of the recombinant vaccinia vectors described.
  • Vaccines and medicaments may comprise a recombinant vaccinia virus as described. These may have enhanced safety and immunogenicity over current vaccinia virus strains for the reasons indicated.
  • a method of attenuating a vaccinia virus vector may comprise: a) deleting part or all of one or more of the following nucleotide sequences from the viral genome; and/or b) inactivating one or more of said nucleotide sequences by mutating said nucleotide sequences or by inserting foreign DNA; and/or c) changing said one or more nucleotide sequences to alter the function of a protein product encoded by said nucleotide sequence;
  • nucleotide sequences are sequences designated herein as: i) B15R, ii) B18R.
  • a vaccinia virus vector as described may be used to prepare a vaccine or a medicament.
  • the translation products encoded by the nucleotide sequences B15R and B18R may have pharmaceutical utility. In particular, they may have utility as anti-inflammatory medicaments.
  • genes and/or translation products thereof may be used in processes relating to the preparation of an anti-inflammatory medicament.
  • the gene product B15R binds IL-l, though not IL-6.
  • this binding is specific for IL-1 ⁇ as IL-1 ⁇ and IL-lra do not bind to the B15R gene product.
  • IL-1 ⁇ is the IL-1 form which is released by the cell and acts on other"cell types in pathological situations (Dinarello, C.A., & Wolff S.M., 1993 New Eng. J. Med. vol.328 p106-113).
  • IL-1 ⁇ is implicated in chromic inflammatory conditions such as rheumatoid arthritis and it has the following functions in relation to inflammation: i) it acts as a T-cell activator; ii) it induces fibroblast proliferation; iii) it increases the production of mediators of the
  • fibroblasts and iv) it increases the production of collagenase from chondrocytes (Digiovine F.S. et al, 1990, Immunol. Today Vol.11 p13-20). Therefore the gene product of B15R which can bind to IL-1 ⁇ provides a means of blocking the activity of IL-1 ⁇ in order to control and/or reduce the symptoms of inflammation.
  • IL-1 ⁇ acts as a mediator in septic shock and it is a growth factor for certain malignant cells e.g. leukaemic cells. Therefore the gene product of B15R provides a means for controlling septic shock and growth of certain tumour cells.
  • IL-1 ⁇ receptor may be used to control disease mediated by excessive fever.
  • the present invention provides use of the nucleotide sequence designated herein as B15R, or of a nucleotide sequence coding for an allele of, or
  • a polypeptide which is encoded by the nucleotide sequence designated herein as B15R or of an allele of, or functionally equivalent fragment, derivative or variant of the polypeptide, to manufacture a medicament for the treatment of a condition in which IL-1 ⁇ is involved in the mediation of one or more symptoms associated with the condition.
  • An allele of, or functionally equivalent fragment, derivative or variant of the B15R polypeptide may be any polypeptide with substantial homology to part or all of the B15R polypeptide and which also has the ability to bind IL-1 ⁇ .
  • Conditions may be fever, inflammation, diseases such as rheumatoid arthritis in which inflammation is
  • cytokine IL-1 ⁇ is the primary mediator of fever.
  • the present invention particularly provides use of the nucleotide sequence designated herein as B15R, or a nucleotide sequence coding for an allele of, or functionally
  • polypeptide encoded by the B15R nucleotide sequence in a process relating to the manufacture of a medicament for the treatment of fever. Also provided is use of a polypeptide encoded by the nucleotide sequence designated herein as B15R or of an allele of, or functionally equivalent fragment, derivative or variant of the
  • polypeptide to manufacture such a medicament is also provided.
  • a pharmaceutical which is an anti-fever medicament is also provided.
  • a pharmaceutical which is an anti-fever medicament comprising a polypeptide which is encoded by the nucleotide sequence designated herein as B15R, or an allele of, or functionally equivalent fragment derivative or variant of the polypeptide.
  • a diagnostic reagent for the detection or measurement of interleukin-1 ⁇ in a sample which reagent comprises a polypeptide encoded by the nucleotide sequence designated herein as B15R, or of an allele of, or functionally equivalent fragment,
  • a diagnostic kit which comprises a diagnostic reagent as described above and one or more ancillary kit
  • the proteins may be produced in a recombinant system according to techniques well known in the art.
  • the nucleotide sequences provided herein could be inserted into a suitable expression vector (not necessarily vaccinia, for example the baculovirus system described herein).
  • a suitable expression vector not necessarily vaccinia, for example the baculovirus system described herein.
  • Such vectors can then be used to infect or transform a cell line suitable for the production of these particular proteins.
  • Reagents comprising polypeptides such as the B15R gene product, or alleles of or functionally equivalent fragments, derivatives or variants of that gene product may be used as research tools as a means to study the function of IL-1 ⁇ versus IL-1 ⁇ .
  • FIG. 1(A) HindIII restriction map of the 186kb W genome.
  • the 9.8kb SalI I fragment is expanded to show the position and direction of transcription of the genes B15R and B18R and the serpin genes (Smith, G.L., Howard, S.T. and Chan, Y.S. (1989). J. Gen. Virol. 70, 2333-2343) (this nomenclature indicates the genes are the fifteenth and eighteenth ORFs starting from the left end of the HindIII B and are transcribed rightwards towards the genomic terminus).
  • Figure 1(B) Nucleotide sequence and deduced amino acid sequence of gene B15R. Potential transcriptional control signals are underlined and a possible signal peptide at the N-terminus is boxed. Sites for the addition of N-linked carbohydrate (NXS/T) are boxed and the cys residues likely to form disulphide bonds within Ig-like domains are stippled.
  • NXS/T N-linked carbohydrate
  • Figure 1(C) Nucleotide sequence and deduced amino acid sequence of gene B18R. The three amino acid
  • FIG. 1 Amino acid alignment of the Ig domains from B15R and B18R with the Ig-like domains of the human and murine IL-1RI, the human IL-6R, the VV haemagglutinin (VV HA), domain 1 of the fasciclin II, domain 3 of myelin-associated glycoprotein and the V-domain of Ig kappa.
  • the regions predicted to form the ⁇ -strand structures of Ig-like domains are indicated above the alignment. Residues identical in 6 or more sequences are boxed. A few residues between the ⁇ -strands B and C have been omitted. Also omitted for brevity are ⁇ -strands D and, where appropriate, C' and C". Higher numbers of residues (about 30 or more) between strands C and E are indicative of the V domains.
  • FIG. 4 S1 Mapping of 5' Ends of mRNAs Coding for B15R and B18R.
  • Specific 5' radiolabeled probes (lane P), prepared as described in Experimental Procedures, were hybridized with yeast transfer RNA (lane 1) or vaccinia virus early (lane 2) or late (lane 3) RNA and digested with SI nuclease. Nuclease-resistant fragments were resolved on a sequencing gel alongside an M13 sequencing ladder (lanes A,C,G and T). Autoradiographs
  • B15R (A) and B18R (B) probes are shown. The sizes of the probes and fragments protected are indicated in bases. Indicated below the autoradiographs are the probe position relative to the ORFs (underline and asterisks), the nucleotide and the deduced amino acid sequence at the 5' end of the ORFs, the vaccinia late promoter consensus sequence (underline), and the sites of transcriptional initiation (asterisks).
  • FIG. 1 Structure of Recombinant Vaccinia Virus Genomes.
  • Vaccinia virus DNA was digested with HindIII (A) or ClaI (B), and fragments were resolved on an agarose gel and transferred to nitrocellulose. Filters were probed with fragments containing the gene and flanking sequences of B15R (lane a) or B18R (lane b), with an internal oligonucleotide to B15R (lane c) or an internal fragment to B18R (lane d). Sizes in kilobases are indicated. Schematic representations of the
  • HindIII (H) or Clal (C) fragments containing the TK gene hatchched box
  • B15R gene shading box
  • B18R gene closed box
  • B15R from vaccinia virus-infected cells.
  • BS-C-l cells were mock infected (M) or infected with WR, vB15R, or v B15R and pulse-labeled with 35 STrans-label either from 2 to 4hr after infection in the presence of cytosine arabinoside (E) or from 6 to 8 hr after infection in the absence (L) or presence (T) of tunicamycin.
  • E cytosine arabinoside
  • T tunicamycin
  • B Identification of B18R from vaccinia virus-infected cells.
  • BS-C-l cells were infected with vB18R or v B18R and pulse-labeled with 35 STrans-label from 6 to 8h after infection, the cell extracts and media were
  • (C) Expression of B15R and B18R in baculovirus-infected insects cells Sf cells infected with AcNPV, AcB15R, or AcB18R were pulse-labeled with 35 STrans-label for 2 hr after 24 hr of infection. Proteins present in cells and media were analyzed by SDS-PAGE and visualized by autoradiography. As in (A), the quantity of sample from medium was estimated to correspond to about four times the amount analyzed from cells.
  • the B15R (open arrowhead) and B18R (closed arrowhead) gene products and the molecular size markers are indicated in kilodaltons. The positions of ⁇ -galactosidase ( ⁇ gal), coexpressed with B15R and B18R in the recombinant baculoviruses, and polyhedrin (P), expressed only. in AcNPV, are shown.
  • Figure 7 Binding Assays to IL-1 ⁇ , IL-1 ⁇ and IL-6.
  • EL4 6.1 C10 cells, U266 cells, mock- or vaccinia (WR)-infected TK-143 cells, and Sf cells infected with AcNPV, AcB15R, or AcB18R were dotted onto nitrocellulose
  • IL-1 ⁇ 120 pM
  • IL-1 ⁇ 200 pM
  • IL-6 IL-6
  • FIG. 8 Binding Assay in Solution to Recombinant Viruses.
  • Binding is expressed as a
  • the percentages refer to the binding in the absence of competitor, which was 3720 cpm for 125 I-IL-1 ⁇ and EL4 6.1 C10 cells (A), 2040 cpm for 125 I-IL-1 ⁇ and EL4 6.1 C10 cells (B), and 4963 cpm for 125 I-IL-6 and U266 cells (C).
  • mice Groups of five mice were intransally infected with 3 ⁇ 10 7 (panels a), 10 7 (panels b), 10 6 (panels c), 10 5 (panels d) or 10 4 (panels e) pfu of WR (open circle) or v B15R (closed circle) and examined daily for symptoms of illness or death. The number of animals that
  • mice were intransally infected with 10 5 (panels a) of 10 4 (panels b) pfu of WR (open circle) or v B15R (closed circle). Symptoms of illness were scored from zero to four, and the mean value of each group was represented. Animals were weighed individually each day and the mean group weight was expressed as the percentage of the mean weight of that group of animals immediately prior to infection. No mortalities occurred at these doses of virus.
  • FIG. 12 Effect of Expression of the Vaccinia IL-1 ⁇ Receptor on Mice Infected with Vaccinia Virus.
  • a representative mouse 5 days after infection with 10 5 pfu of WR (a) or v B15R (b) is shown. Note the ruffled fur in (b), which correlated with accelerated weight loss (see Figure 8B, panels a).
  • Cowpox Tissue culture medium (1 ⁇ 10 5 cell equivalents) from TK-143 cells infected with the indicated viruses was incubated in a binding assay in solution with 100 pM of radioiodinated IL-1 ⁇ or mIL-1 ⁇ , expressed in femtomoles, is shown.
  • One femtomole corresponded to 935 or 535 cpm for IL-1 ⁇ or mIL-1 ⁇ , respectively.
  • FIG. 14 Kinetics of symptoms of illness and mortality in mice infected with recombinant vaccinia viruses.
  • BALB/c mice were intranasally infected with WR, v B15R or v B18R as described in Table 2.
  • recombinants were examined for illness and the number of animals with symptoms (including mortality) in each group were represented as a function of time. Symptoms scored were (a) possession of ruffled fur (b) hunching and (c) immobility.
  • the nucleotide sequence of the SalI I restriction fragment of the vaccinia virus genome were determined by established methods (Sanger, F. et al.
  • the 9.8kb SalI I fragment of vaccinia virus was isolated from cosmid 6, which contains virus DNA derived from a rifampicin resistant mutant (Baldick, C..J. & Moss, B. (1987) Virology 156, 138-145), and was cloned into SalI cut pUC13 to form plasmid pSalI I.
  • the SalI fragment was separated from plasmid sequences and self-ligated with T4 DNA ligase.
  • Circular molecules were randomly sheared by sonication, end-repaired with T4 DNA polymerase and Klenow enzyme, and fragments of greater than 300 nucleotides cloned into SmaI cut M13mp18. Single stranded DNA was prepared and sequenced using the dideoxynucleotide chain termination method (Sanger, F., Nicklen, S. & Coulson, A.R. (1977) Proc. Natl. Acad. Sci. USA. 74, 5463-5467), using
  • IL-1 a cytokine produced in response to infection and tissue injury, is involved in the regulation of the inflammatory and immune responses and in the activation of a broad spectrum of systemic effects that contribute to host defense (Dinarello 1988, 1989; Di Giovine and Duff 1990).
  • the two forms of IL-1, IL-1 ⁇ and IL-1 ⁇ produce similar biological effects that are mediated by interaction with specific receptors in different cells.
  • IL-1 receptors There are two classes of IL-1 receptors, and both bind IL-1 ⁇ and IL-1 ⁇ with similar affinities (Dower, S.K., and Urdal, D.L. (1987)).
  • the 80 kDa type I IL-1 receptor is found on T cells and fibroblasts (Bird, T.A., and Saklatvala, J. (1986) Nature 324, 263-266), while the 60 kDa type II IL-1 receptor is present in B cells and macrophages (Matsushima, K. et al (1986) J. Immunol. 136, 4496-4508).
  • IL-1ra IL-1 receptor antagonist
  • Genes B15R and B18R (Figure 1) from near the right hand inverted terminal repeat ( ITR) are predicted to encode proteins of 36.5 kDa and 40.7 kDa, respectively, that have an N-terminal hydrophobic sequence, possible attachment sites for N-linked carbohydrate and
  • the nucleotide sequence and deduced amino acid sequence around the gene designated B15R is shown in figure IB.
  • the nucleotide sequence shown is 11462-12664 nucleotides from the left end of the vaccinia virus HindIII B fragment and the coding region for B15R is at nucleotide positions 11584-12561 (or at nucleotides 815 to 1792 from the left end of the SalI I fragment).
  • nucleotide sequence and deduced amino acid sequence around the gene designated B18R is shown in figure 1c.
  • the nucleotide sequence shown is 15448-16741 nucleotides from the left end of the vaccinia virus
  • HindIII B fragment and the coding region for B18R is at nucleotide positions 15568-16621 (or at nucleotides 4799 to 5851 from the left end of the SalI I fragment).
  • the single letter code is used for the designation of amino acids.
  • B15R and B18R each possess three domains with characteristics of the immunoglobulin (Ig) superfamily (Williams, A.F. and Barclay, A.N. (1988). Ann. Rev.
  • B15R and the external region of IL-1Rs have a very similar length.
  • a glycosylation site is conserved in domain 1, ⁇ -strand F of IL-1Rs and B15R despite divergence of amino acid sequence.
  • (6) Domain 3 does not contain additional cysteines and is longer than 1 and 2 in B15R, B18R and the IL-1Rs.
  • VV proteins are related to each other (22.5% identity), to the human and murine IL-1R, the human IL-6R (Yamasaki, K., Taga, T., Hirat, Y., Yawata, H., Kawanishi, Y., Seed, B., Taniguchi, T., Hirano, T. and Kishismoto, T. (1988).
  • Ig immunoglobulin
  • VV haemagglutinin is another member of this superfamily (Jin, D., Li, Z., Jin, Q., Yuwen, H. and Hou, Y. (1989). J. Exp. Med. 170, 571-576).
  • the B18R sequence from VV strain IHD was recently
  • VV may be combatting this part of the immune response by producing proteins which mimic the receptors for IL-1 and IL-6.
  • B15R and B18R ORFs are actively transcribed, translated, and secreted to the medium during the vaccinia virus replication cycle.
  • the B15R gene product is shown to bind IL-1 ⁇ when expressed from vaccinia or from recombinant baculovirus.
  • the role of the IL-1 ⁇ binding activity in the biology of vaccinia virus was investigated by deleting the gene from the virus genome and analyzing the biological effects on infected mice. The biological effects on infected mice were also investigated for vaccinia virus without the B18R coding sequence. The presence of the binding activity in other orthopoxviruses is also presented.
  • the cell line EL4 6.1 C10 a subclone of the mouse thymoma EL4 that expresses a high number of IL-1 binding sites (MacDonald et al, 1985), was a gift of H.R.
  • Sf 21 insect cells and AcNPV were obtained from R. Possee (Natural Environmental Research Council Institute of Virology and Environmental Microbiology, Oxford) and were cultured in TC100 medium (GIBCO) containing 10% fetal calf serum (Brown and Faulkner, 1977).
  • Vaccinia virus strain WR and recombinants derived from it were grown in CV-1 or BS-C-1 cells. TK-143 and HeLa D98 cells were used for the selection of
  • the WR strain was obtained from B. Moss (National Institute of Health, Bethesda, Maryland) and cells were obtained from the American Type Culture
  • Copenhagen strain were obtained from J. Zhou (Princess Alexandra Hospital, Brisbane, Australia) and R. Drillien (University Louis Pasteur, France),
  • Radioiodinated human recombinant IL-6 was purchased from Amersham. IL-1 ⁇ had been, radioiodinated using the chloramine-T procedure to a specific activity of 70-120 ⁇ Ci/ ⁇ g. IL-1 ⁇ and IL-6 had been labeled with Bolton Hunter reagent to a specific activity of 80-180 ⁇ Ci/ ⁇ g and 800-1200 Ci/mmol, respectively. Unlabeled human recombinant IL-1 ⁇ (code 86/632), IL-1 ⁇ (code 86/680), and IL-6 (code 88/514) were obtained from the National
  • IL-1 ⁇ and IL-1 ⁇ were 10 5 U/ ⁇ g and for IL-6 was 5 ⁇ 10 3 U/ ⁇ g.
  • Unlabeled human recombinant IL-IRA specific activity 1 ⁇ 10 5 to 1.4 ⁇ 10 5 U/mg, was purchased from British Biotechnology.
  • a derivative of plasmid pUC118 (Vieira and Messing, 1987) was constructed that contained the entire B15R ORF, lacking most of the flanking regions, and with convenient restriction sites at each end. A combination of subcloning and PCR was used. The left end SalI-XbaI fragment of the SalI I fragment of vaccinia DNA
  • pAA1 containing B15R was inserted into SalI- and BamHI-cut pUC118, and the resultant plasmid was called pAA1.
  • pAA1 was digested with EcoRV and XbaI, and the largest fragment was gel purified, end filled with Klenow fragment, and self-ligated to form pAA3, which contains 348 bp of the 3' flanking region.
  • oligonucleotide that hybridizes to an internal sequence of B15R, corresponding to nucleotides 1008-1025 of the SalI I fragment (B15R-2; 5'-CCGCTCCTCGTTTTTCCC-3').
  • the fourth nucleotide of B15R in the PCR fragment was G instead of A to create a NcoI recognition sequence, giving rise to a serine to glycine substitution in the second amino acid of the protein.
  • the transfer vector used for overexpression of B15R and B18R in vaccinia virus was pRK19 (Kent, R.K. 1988), which contains the vaccinia virus 4b promoter to control the transcription of the inserted gene, flanked by sequences of the TK gene that allow insertion in the TK locus of the virus genome.
  • BamHI fragments containing the ORFs were excised from pAA4 and pAA5 and cloned into the BamHI site of pRK19, and the resulting plasmids were named pAA10 and pAA11, respectively.
  • the transfer vector for construction of baculovirus recombinants was pAcDZl, which uses the polyhedrin promoter to drive the transcription of foreign genes and coexpresses Escherichia coli ⁇ -galactosidase for
  • the genes were excised from pAA4 and ⁇ AA5 with BamHI and inserted into BamHI-cut pAcDZl, forming pAA14 and pAA15, respectively.
  • flanking sequences of B15R and B18R were excised from pAA1 or clones from a M13 library containing random subfragments of the SalII fragment of vaccinia DNA, which were used to sequence this region of the vaccinia virus genome (Smith et al, 1991a), and were cloned into pSJH7 (Hughes et al, 1991).
  • the 5' flanking region of B15R was obtained by digestion of the replicative form of the M13 clone SalII.144 with EcoRI and SphI, and the 3' flanking region was excised from pAA1 by digestion with SphI and BamHI. Both fragments were, cloned in one step into
  • the resultant plasmid contained 360 and 1316 nt of the 5' and 3' flanking sequence, including 17 and 252 nt of the coding sequence, respectively, so that 72% of the B15R coding sequence was deleted.
  • DNA fragments containing the flanking sequences of B18R were obtained by BamHI and EcoRI digestion of the replicative form of the M13 clones SalII.44 and SalII.81.
  • PCR fragment was obtained with the oligonucleotide B15R-2 (above) and the 17-mer sequencing primer (-20), using pAA1 as template.
  • the PCR product was purified, labeled with [ - 32 P]ATP and polynucleotide kinase, and
  • pAA2 was digested with EcoRI, a band of 505 bp was purified and dephosphorylated with calf intestinal alkaline
  • Both 32 P-labeled fragments specific for B15R and B18R were hybridized to 10 ⁇ g of vaccinia virus RNA obtained at 8 hr after infection from cells infected in the presence (early) or absence (late) of cycloheximide or yeast transfer RNA.
  • the hybrids were digested with SI nuclease, and the protected fragments were separated on 6% polyacrylamide sequencing gel and detected by
  • Sf cells were cotransfected with purified AcNPV DNA and pAA14 or pAA15 using the calcium phosphate precipitation technique, and the recombinant viruses were identified by staining with X-GaI as described (Zuidema et al, 1990). The insertion of foreign genes (B15R and B18R) into the baculovirus genome was confirmed by
  • Recombinant vaccinia viruses were constructed by standard procedures (Mackett et al, 1985). The genomes of viruses containing a second copy of B15R or B18R in the TK locus of the vaccinia DNA were analyzed by
  • Vaccinia virus deletion mutants were constructed by transient dominant selection as described elsewhere ( Falkner and Moss, 1990; Isaacs et al, 1990).
  • Vaccinia viruses containing deleted versions of B15R and B18R were termed vAA5 and vAA6, respectively, and are referred to here as v B15R and v B18R.
  • the B15R-specific probe containing the ORF and 348 bp of the 3' flanking region was excised from pAA4 by digestion with BamHI and used for Southern blot
  • the oligonucleotide B15R-2 was used as an internal probe for B15R ORF.
  • the internal probe for B18R was obtained by excision of a 424 bp EcoRI fragment from pAA2.
  • Biomedicals a mixture of -80% [ 35 S]methionine and -20% [ 35 S] cysteine, 1200 Ci/mmol) in methionine-free TC100 medium or methionine- and cysteine-free minimal essential medium, respectively, in the absence of serum.
  • Cytosine arabinoside 40 ⁇ g/ml
  • tunicamycin 1 ⁇ g/ml
  • Radioactive bands were detected by autoradiography or fluorography with salicilate (Harlow and Lane, 1988).
  • Sf cells and TK-143 cells grown in 175 cm 2 or 80 cm 2 flasks, were infected at a density of 1.5 ⁇ 10 5 to 2 ⁇ 10 5 cells/cm 2 with a multiplicity of infection of 5-10 pfu per cell in serum-free medium.
  • Cells and medium were harvested from vaccinia- or baculovirus-infected cells at 1 or 3 days after infection, respectively.
  • the final concentration of the supematants was 1 ⁇ 10 6 to 5 ⁇ 10 6 cell equivalents per millilitre.
  • the medium was
  • supematants made 20 mM HEPES (pH 7.4) and 0.1% sodium azide.
  • Supematants were stored at -70°C until used in binding assays in solution or concentrated and dialized against phosphate-buffered saline ( PBS ) at 4°C in a Micro-ProDiCon (Bio-Molecular Dynamics) with PA-10 ProDiMen dialysis membranes (MW 10,000) to a-final concentration of 5 ⁇ 10 7 cell equivalents per millilitre.
  • the concentrated medium was made 1% in sodium azide and stored at -70°C.
  • the supematants were harvested from cells seeded at a cellular density of 5 ⁇ 10 5 cells per
  • Sf and TK-143 cells were harvested for binding assays to intact cells by treatment with PBS containing 0.5 mM EDTA.
  • EL4 6.1 C10, U266, Sf, and TK-143 cells were washed twice in serum-free medium and resuspended in binding medium.
  • the binding medium used in the different assays was RPMI 1640 containing 20 mM HEPES ( pH 704), 1% bovine serum albumin, and 0.1% sodium azide. Solid phase binding assays on nitrocellulose were performed as described (Urdal et al, 1988). Binding to intact cells was carried out in duplicate in 150 ⁇ l of binding medium for 2 hr at 4°C, and bound 125 I-IL was determined by phthlate oil centrifugation as described (Dower et al, 1985). In the competition assays of labeled ILs to intact cells, samples were preincubated with the ILs in 125 ⁇ l for 1 hr at 4°C. Subsequently, 2.5 x 10 6 EL4 6.1 C10 or U266 cells were added in 25 ⁇ l and incubated for 2 hr at 4°C.
  • Soluble receptor binding assays were performed by precipitating the ligand-receptor complexes with
  • mice Female BALB/c mice ( 5 to 6 weeks old) were anesthetized and infected intranasally with 20 ⁇ l of the diluted virus in 1 mM Tris-HCI ( pH 9.0). Mice were weighed daily and monitored for signs of illness or death (Turner, 1967; Williamson et al, 1990). As a control, an aliquot of the dilutions of v B15R or WR used to inculate the animals was grown in TK-143 cells, and the absence or the presence of IL-1 ⁇ binding activity in the medium at 24 hr after infection was confirmed in a binding assay in solution (data not shown).
  • mice 4-6 weeks old BALB/c mice were intranasally infected with doses of WR, v B15R or v B18R as shown in Table 2. The table also shows the mortality of animals after 15 days.
  • FIG. 4A shows that the B15R-specific probe was partially protected from SI nuclease digestion by late viral RNA, and the size of the protected fragment mapped the transcriptional start site to the TAAAAT motif at the 5' end of B15R.
  • TAAAT(G) has been shown to constitute a late promoter consensus sequence for vaccinia virus, a few exceptions have been found that possess an additional A TMOSS, 1990b).
  • the B18R-specific probe was protected from S1 nucflease digestion by early viral RNA that initiated 16-18 nt upstream of the ORF ( Figure 4B).
  • Vaccinia virus recombinants overexpressing the proteins or lacking the coding regions were constructed to identify the gene products and to study the biological activity of the proteins. Overexpression of the proteins was achieved by cloning a second copy of B15R or B18R, transcribed under the control of the late 4b promoter, in the thymidine kinase (TK) locus.
  • the genomic structure of the recombinant viruses, called vB15R and vB18R was confirmed by Southern blot hybridization ( Figure 5A) and by polymerase chain reaction (PCR) using oligonucleotides specific for the 5' and 3' ends of the TK gene (data not shown).
  • cytosine arabinoside were performed in cells infected with different vaccinia virus recombinants.
  • Extracts from cells or culture supematants were obtained by:
  • the protein encoded by B18R was detected in vaccinia virus-infected cells when overexpressed at late times of infection under the strong 4b promoter but was not detected in the deletion mutant (Figure 6B). Two forms of the protein (52kd and 60-65kd) were detected in cell extracts, and only the 60-65 kd protein, presumably containing a higher degree of glycosylation, was secreted to the medium. Since translation of B18R-specific mRNA in rabbit reticuloyte lysates produces a protein of the predicted size of 40 kd (Ueda et al., 1990), the
  • carbohydrate component of the secreted B18R gene product accounts for 33%-38% of the size of the protein.
  • expressed in insect cells might not recognise that form of the protein.
  • Spodoptera frugiperda insect cells infected with Autographa califomica nuclear polyhedrosis virus (AcNPV) under the control of the polyhedrin promoter.
  • AcNPV Autographa califomica nuclear polyhedrosis virus
  • the recombinant viruses constructed were termed AcB15R and AcB18R.
  • the 45 kd protein in AcB15R-infected insect cell extracts might correspond to a glycosylated form with a signal sequence still bound to the polypeptide, possibly owing to the inability of insect cells to process properly the high amount of B15R protein expressed under the strong
  • baculovirus was not detected while these ligands bound to EL46.lC10 and U266 cells, which overexpress IL-1 and IL-6 receptors, respectively.
  • IL-1 ⁇ binding activity was clearly found in vaccinia virus-infected cells and supematants harvested at 24 hr after infection and in the baculovirus recombinant expressing B15R 3 days after infection.
  • the low binding to EL4 6.1C10 cells at this dose of 125 I-IL-1 ⁇ probably reflects a 6-fold lower affinity for the ⁇ form compared with IL-1 ⁇ , described for the type I receptor expressed in this cell line (Sims et al., 1988).
  • IL-1 ⁇ receptor The kinetics of production of soluble IL-1 ⁇ receptor from vaccinia virus-infected cells was examined by soluble binding assay and showed that no IL-1 ⁇ receptor was detected above the background attributable to virus inoculum, in the presence of cytosine arabinoside. In contrast, in the absence of the drug, IL-1 ⁇ accumulated in the supernatant and reached 80% of total by 24 hr (data not shown). These data are consistent with the transcriptional and polypeptide analyses and show that the IL-1 ⁇ receptor is expressed late during infection.
  • Figure 8 shows the binding of radioiodinated IL-1 ⁇ to medium from different recombinants using 1 ⁇ 10 4 cell equivalents, conditions that allowed a better
  • the natural competitor IL-1RA did not block the binding of labelled IL-1 ⁇ to vaccinia IL-1 receptor, even when added at higher concentrations that are required to compete the binding of 125 I-IL-1 to the type II IL-1 receptor on polymorphonuclear leukocytes or a pre-B lymphocyte line (Dripps et al., 1991; Granowitz et al., 1991; Mclntyre et al., 1991).
  • the doses of unlabelled ILs used competed the binding of the corresponding .
  • radioiodinated IL to its natural receptor on EL4 6.1 C10 or U266 cells (data not shown).
  • the receptor expressed in the baculovirus system showed similar properties.
  • equivalents of medium from AcB15R-infected cells was 94.1%, 7.4%, 99.0% and 110.1% in the presence of 10nM of IL-1 ⁇ , 10nM of IL-1 ⁇ , 100nM of IL-1ra, and 10nM of IL-6, respectively.
  • K D dissociation constant
  • Table 2 and Figure 14 show the results for the in vivo experiment in which mice were intranasally infected with doses of WR, v B15R or v B18R. The results show that groups of mice infected with a vaccinia virus unable to produce the B18R gene product had a lower incidence of mortality than equivalent groups of mice infected with either wild-type vaccinia virus or a vaccinia virus unable to produce the B15R gene product. In Figure 14, mortality and symptoms of illness are presented-as a function of days post-inoculation.
  • mice infected with either wild-type vaccinia virus or a vaccinia virus unable to produce the B15R gene product The incidence of mortality was similar for groups of mice infected with either wild-type vaccinia virus or a vaccinia virus unable to produce the B15R gene product. However, symptoms of illness appeared sooner in groups of mice infected with vaccinia virus unable to produce the B15R gene product as compared to equivalent groups of mice infected with the wild-type vaccinia virus.
  • the IL-1 ⁇ , IL-1 ⁇ , and IL-6 binding activity was investigated in a soluble receptor binding assay on supematants from cultures infected with different strains of vaccinia virus (Copenhagen, IHD-J, IHD-W, Wyeth, Lister, Tian-Tan, and Tashkent) and the related orthopoxviruses rabbitpox and cowpox compared with the WR strain.
  • the binding to labelled murine IL-1 ⁇ (mIL-1 ⁇ ) was also investigated to confirm that B15R is able to sequester IL-1 ⁇ in infected mice. No binding to human 125 I-IL-1 ⁇ or 125 I-IL-6 was detected (data not shown). However, binding to labelled IL-1 ⁇ and mIL-1 ⁇ was found in all viruses except for rabbitpox, Tashkent and
  • Copenhagen to express an IL-1 receptor is in agreement with sequencing data that showed a nonsense mutation at codon 31 of the ORF (Goebel et al., 1990). Even if translation reinitiated from the next methionine codon, which seems unlikely given its distance from the mRNA 5' end, the protein would lack a signal peptide and..
  • B15R protein is shown to suppress the temperature of the infected mouse over the first six days of infection.
  • mice infected with the Tian-Tan strain which expresses the IL-1 ⁇ receptor, have reduced temperatures following infection, while those animals infected with strains lacking the receptor
  • ORFs Two vaccinia virus ORFs, B15R and B18R, that encode proteins of the immunoglobulin superfamily related to the extracellular domains of the IL-1 and IL-6 receptors have been characterized. Both ORFs are transcribed, but at different phases of the virus replication cycle,
  • the B18R gene product Is shown not to bind IL-1 ⁇ , IL-1 ⁇ or IL-6, despite the homology with the receptors for these cytokines (McMahan et al, 1991; Smith and Chan, 1991). Two forms of the protein (52 kd and 60-65 kd) were detected, the larger of which is found in
  • B15R ORF is shown to encode an IL-1 ⁇ binding activity present in the supematants of vaccinia virus-infected cells and to represent a novel soluble IL-1 receptor.
  • the high affinity for IL-1 ⁇ binding (K D 234 pM) is similar to those reported for the cellular receptors (Sims et al, 1988, 1989; McMahan et al, 1991) and is consistent with the retention of full binding activity by the extracellular domain of the IL-1 receptor (Dower et al, 1989).
  • the size (50-60 kd ) and high carbohydrate content of the mature vaccinia IL-1 ⁇ receptor are in agreement with those reported for the truncated and complete versions of the cellular receptor, respectively (Urdal et al, 1988; Dower et al, 1989).
  • the secretion of a biologically active 40-44 kd protein from insect cells suggests that the carbohydrate is not an essential component for the IL-1 binding.
  • the vaccinia IL-1 receptor constitutes a novel receptor for IL-1 because of the specificity, for IL-1 ⁇ . This was shown in binding experiments to radioiodinated ILs and was corroborated in competition assays with unlabeled cytokines and by blocking the interaction of the ILs with the natural receptor on cells in culture.
  • B15R has a higher similarity to the cellular type II receptor than to the type I receptor (McMahan et al, 1991; Smith and Chan, 1991) and since this similarity is comparable with those found between other vaccinia virus proteins and their cellular counterparts (Smith et al, 1991a), B15R may derive from the type II IL-1 receptor or a variant thereof.
  • the comparison of the sequence of the vaccinia IL-1 ⁇ receptor with the type I and type II IL-1 receptors does not permit identification of the amino acids that confer specificity for IL-1 ⁇ since the
  • sequences are quite divergent. However, the availability of the vaccinia virus gene will allow mutagenesis studies to identify these positions. Furthermore, the sequence of B15R ORF in other vaccinia virus strains that show different affinities for the human and murine IEr-1 ⁇ may provide structural information on the binding domain. In this case, the comparison of the sequence may be more useful since genes from different orthopoxviruses are highly conserved.
  • the vaccinia IL-1 ⁇ receptor might be useful as a tool to investigate the function of IL-1 ⁇ and IL-1 ⁇ in vivo in different models.
  • the other IL-1 inhibitors available IL-IRA, a soluble truncated IL-1 receptor, and monoclonal antibodies against the receptor
  • IL-IRA a soluble truncated IL-1 receptor
  • monoclonal antibodies against the receptor block the binding of both forms of IL-1 (Fanslow et al, 1990; Gershenwald et al, 1990; Ohlsson et al, 1990;
  • the molecule may regulate responses normally controlled by IL-1 ⁇ , and, since it does not bind IL-1 ⁇ or IL-1RA, it might offer advantages over the other inhibitors.
  • the failure of the vaccinia IL-1 ⁇ receptor to bind IL-1RA illustrates the adaptation of the virus to the physiological response of the host by preventing interference with the natural antagonist.
  • the number of IL-1 ⁇ binding sites secreted from vaccinia virus-infected cells is without precedent and makes the supematants from cultures infected with vaccinia virus the most concentrated naturally occurring soluble IL-1 binding activity.
  • An excess of soluble receptors must be required to block the effects of IL-1 ⁇ in vivo, since only a few cellular IL-1 receptors need to be occupied to elicit a biological response. This was illustrated in the competition of IL-1 ⁇ binding to T- cells, which also indicates that the vaccinia IL-1 ⁇ receptor will probably block the biological effects induced in cells expressing IL-1 receptors.
  • IL-1 ⁇ in the host response to orthopoxvirus infections.
  • virulence is defined according to the number of
  • vaccinia IL-1 ⁇ receptor does play an important role in vaccinia virus infection in vivo.
  • the animals infected with v B15R developed symptoms and lost weight more rapidly than the corresponding control group.
  • the early onset of symptoms is very likely to represent systemic effects induced by circulating IL-1 ⁇ produced in response to vaccinia virus infection.
  • IL-1 is known to function as a hormone mediating multiple effects such as fever, headache, and sleep and at high doses can induce hypotension and a shocklike state (Dinarello, 1988,
  • the vaccinia IL-1 ⁇ receptor expressed in the wild-type virus, may thus limit the systemic acute phase response otherwise initiated by increased levels of IL-1 ⁇ .
  • weight loss which can be induced by IL-1 (Di Giovine and Duff, 1990) occurred earlier in animals infected with v B15R supports this view.
  • a generalized response can contribute to host defense; for example, temperature typical of fever has been reported to enhance the proliferation of T-cells that might facilitate a T-cell-dependent immune response (Duff and Durum. 1983).
  • an increased systemic reaction to infection did not affect the outcome of infection by v B15R. Second, although the absolute number of
  • B15R might function as a virulence or attenuation factor for the virus. It is unclear whether the effects of deleting B15R from the WR strain of vaccinia virus, which was selected for high
  • B15R ORF is one of a few virus genes that has been shown to increase the pathogenicity or the severity of the infection when deleted from the virus genome
  • Virus attenuation can result from deletion or
  • IL-1 ⁇ is the predominant form of IL-1 secreted from human monocytes (Hazuda et al, 1988), and the release of adrenocorticotropic hormone, one of the neuroendocrine actions of IL-1, is exclusively induced by IL-1 ⁇ (Uehara et al, 1987).
  • IL-1 ⁇ is more potent than IL-1 ⁇ in the induction of fever, and the effect is mediated through different mechanisms (Busbridge et al, 1989), which correlates with the discovery of IL-1 ⁇ (Breder et al, 1988) and receptors specific for IL-1 ⁇ (Katsuura et al, 1988) in the brain.
  • B15R is the second soluble cytokine receptor to be identified in a virus.
  • a soluble receptor for tumor necrosis factor (TNF) has been shown to be active in Leporipoxviruses and to increase the pathogenicity of the virus (Smith et al, 1991b; Upton et al, 1991).
  • the WR and Copenhagen strains of vaccinia virus contain one and two homologs, respectively, of the TNF receptor, but the presence of frameshifts and stop codons make expression of active proteins unlikely (Howard et al, 1991; Upton et al, 1991).
  • the soluble IL-1 ⁇ receptor is one of the increasing number of activities encoded by vaccinia virus that aid evasion from the host immune system (for references see Moore and Smith, 1992) and, in particular, is another viral-encoded protein that interferes with cytokine functions.
  • the TNF receptor of leporipoxvirus and the crmA protein of cowpox virus other examples found are the 14.7 kd protein of adenovirus that inhibits cytolysis by TNF (Gooding et al, 1988), the IL-10
  • the vaccinia IL-1 ⁇ receptor may be a useful tool to discriminate the physiological roles of IL-1 ⁇ and IL-1 ⁇ and might be used as an anti-inflammatory therapeutic reagent.
  • the expression of this activity by vaccinia virus and other orthopoxviruses represents a novel mechanism of virus evasion from the immune system. It is shown that the IL-1 ⁇ receptor is modulating the systemic response to infection and the severity of the disease, which suggests that IL-1 ⁇ , and not IL-1 ⁇ , is the main mediator of the endocrine effects of the IL-1 produced in response to vaccinia virus infection in mice.
  • Interleukin 1 the first interleukin. Immunol. Today 11, 13-20.
  • Blocking IL-1 interleukin 1 receptor antagonist in vivo and in vitro. Immunol. Today 12, 404-410.
  • Interleukin-1 receptor antagonist binds to the type II interleukin-1 receptor on B cells and neutrophils. J. Biol. Chem. 266, 20311-20315.
  • Verderber E., Brewer, M.T., Hannum, C.H., and Thompson, R.C. (1990). Primary structure and functional expression from complementary DNA of a human interleukin-1 receptor antagonist. Nature 343, 341-346.
  • interleukin-1 receptor J.Biol.Chem. 265, 17416-17419.
  • Interleukin-1 receptor antagonist competitively inhibits the binding of interleukin-1 to the type II interleukin-1 receptor. J. Biol. Chem. 266, 14147-14150.
  • Interleukin-1 receptor antagonist activity of a human interleukin-1 inhibitor is Interleukin-1 receptor antagonist activity of a human interleukin-1 inhibitor. Nature 343, 336-340.
  • Vaccinia virus homologues of the Shope fibroma virus inverted terminal repeat proteins and a discontinuous ORF related to the tumor necrosis factor receptor family are included in the tumor necrosis factor receptor family.
  • Vaccinia virus encodes an active thymidylate kinase that complements a cdc8 mutant of
  • a novel IL-1 receptor cloned from B cells by mammalian expression, is expressed in many cell types. EMBO J. 10, 2821-2832.
  • LIGAND a versatile computerized approach for characterization of ligand-binding systems. Anal. Biochem. 107, 220-239.
  • cowpox virus encodes an inhibitor of the interleukin-1 ⁇ converting enzyme.
  • T2 open reading frame from Shope fibroma virus encodes a soluble form of the TNF receptor. Biochem.
  • Vaccinia and cowpox viruses encode a novel secreted interleukin-1-binding protein. Cell 71, this issue.
  • a soluble binding protein specific for interleukin 1 ⁇ is produced by activated mononuclear cells. Cytokine 2, 190-198.
  • Myxoma virus expresses a secreted protein with homology to the tumor necrosis factor receptor gene family that contributes to viral virulence. Virology 184, 370-382.
  • polyhedrin-based baculovirus expression vector polyhedrin-based baculovirus expression vector.
  • CHNCAM 1 4.12 4.01 4.07 3.19 5.54 3.28
  • NB 4/5 4 deaths in a group of 5
  • mice 4-6 week old Balb/c mice were infected intranasally with indicated doses of WR, B15R-deleted (v ⁇ 15ft) or B188-del (v ⁇ B18F)virus. The mortality of animals after 15 days is show
  • Values of greater than 3.1 are significant (probability 10 -3 ), while values of 4.8, 6.0 and 7.9 indicate probabilities of 10 -6 , 10 -9 and 10 -15 , respectively.
  • the domains illustrated are from B15R amino acids 28-119 (1), 121-214 (2), 222-end (3): B18R 53-149 (1), 152-241 (2), 252-end (3); murine IL-1R precursor (Sims, J.E., March, C.J., Widmer, M.B., MacDonald, H.R., McMahan, D.J., Grubin, C.E., Wignall, J.M., Jackson, J.L., Call, S.M. Friend, D., Alpert, A.R.
  • human IL-6R Yamasaki, K., Taga, T., Hirat, Y., Yawata, H., Kawanishi, Y., Seed, B., Taniguchi, T., Hirano, T and Kishismoto, T. (1988).
  • Proc. Jpn. Acad. 64, 209-211 27-116; murine neural cell adhesion molecule L1 precursor (L1CAM) (Moos, M., Tacke, R., Scherer, H., Teplow, D., Freuth, K., Schachner, M. (1988).
  • CHNCAM chicken neural cell adhesion molecule
  • MAG myelin-associated glycoprotein
  • PDGFR platelet derived growth factor receptor
  • T cell receptor CD3 epsilon chain (TCRCD3) (Clevers, H., Duiilap, S., Saito, H., Georgopoulos, K., Wileman, T. and Terhorst, C., (1988). Proc. Natl. Acad. Sci. USA 85, 8623-8627) 1-82; leukocyte antigen receptor protein (LAR) (Streuli, M., Krueger, N.X., Hall, L.R., Schlossman, S.F. and Saito, H. (1988). J. Exp. Med.
  • LAR leukocyte antigen receptor protein

Abstract

This invention relates to proteins encoded by vaccinia virus and which function to interfere with the host's immune system. In particular, the invention provides for use of the nucleotide sequence designated herein as B15R or of a nucleotide sequence coding for an allele of, or functionally equivalent fragment, derivative or variant of the polypeptide encoded by the B15R nucleotide sequence in a process relating to the manufacture of a medicament for the treatment of a condition in which interleukin-1β is involved in the mediation of one or more symptoms associated with the condition. The invention also provides an anti-fever medicament which comprises a polypeptide which is encoded by the B15R gene product or an allele of, or functionally equivalent fragment, derivative or variant of that product.

Description

VACCINIA VIRUS B15R USED IN INTERLEUKIN B1-INVOLVING CONDITION.
The present invention relates to vaccinia virus proteins, uses and products relating thereto.
Where the term IL-1β receptor is used in the text as referring to the vaccinia virus B15R polypeptide or allele or functionally equivalent fragment, derivative or variant thereof, the word "receptor" should be
interpreted as referring to a substantially soluble binding protein.
Live vaccinia virus was used as the vaccine to immunise against, and eradicate smallpox. Vaccinia virus (VV) is the prototypical member of the poxvirus family and therefore it has been extensively studied. It is a large DNA-containing virus which replicates in the cytoplasm of the host cell. The linear double-stranded genome of approximately 185,000 base pairs has the potential to encode at least 200 proteins (Moss, B.
(1990a)). The cytoplasmic site of replication requires that vaccinia virus encodes many enzymes and protein factors necessary for transcription and replication of its genome. The virus also encodes a variety of factors which modulate virus replication in the multicellular host and aid evasion of the host immune system (Moss, B. (1990a)). Advances in molecular genetics have made possible the construction of recombinant vaccinia viruses that contain and express genes derived from other
organisms (for review see Mackett, M. & Smith G.L.
(1986), J. Gen. Virol., 6J7, 2067-2082). The recombinant viruses retain their infectivity and express the foreign gene (or genes) during the normal replicative cycle of the virus. Immunisation of animals with the recombinant viruses has resulted in specific immune responses against the protein(s) expressed by the vaccinia virus, including those protein(s) expressed by the foreign gene(s) and in several cases has conferred protection against the pathogenic organism from which the foreign gene was derived.
Recombinant vaccinia viruses have, therefore, potential application as new live vaccines in human or veterinary medicine. Advantages of this type of new vaccine include the low cost of vaccine manufacture and administration (because the virus is self-replicating), the induction of both humoral and cell-mediated immune responses, the stability of the viral vaccine without refrigeration and the practicality of inserting multiple foreign genes from different organisms into vaccinia virus, to construct polyvalent vaccines effective against multiple pathogens. A disadvantage of this approach, is the use of a virus vaccine that has been recognised as causing rare vaccine-related complications.
It would therefore be of considerable value to produce a vaccinia virus with an attenuated phenotype as a basis for the production of a new generation of live recombinant vaccinia virus vaccines.
Vaccinia virus contains two genes designated B15R and B18R from near the right inverted terminal repeat (ITR) which are each predicted to encode a soluble glycoprotein that has three immunoglobulin-like (Ig) domains, but no transmembrane anchor sequence of
cytoplasmic tail (Smith, G.L. and Chan Y.S. 1991).
The product of B18R is apparently expressed on the surface of the infected cell early during infection and antibodies directed against a mixture of it and other proteins confer resistance to virus infection without directly neutralizing infectivity (Ueda, Y., Morikawa, S. and Matsuura, Y. (1990); Ueda and Tagaya, I. (1973) J. Exp. Med. 138, 1033-1043; Ikuta, K., Miyamoto, H. and Kato, S. (1980). On the basis of studies investigating homology of these gene products with other proteins, the applicants recognised limited similarity of these gene products to receptors for certain interleukin (IL) molecules and they surmised that either of these gene products may bind ILs and prevent these cytokines
reaching their natural receptors. In consequence, the inflammatory response may be diminished and virus
replication enhanced to constitute a novel method of immune evasion. The applicants present data herein which shows that gene product B18R does not bind interleukin-1 (IL-1) or interleukin-6 (IL-6). However, the gene product B15R does bind IL-1, though not IL-6.
Surprisingly, this binding is specific for IL-β as IL-α and the IL-1 receptor antagonist protein (IL-1ra) do not bind to the B15R gene product.
The applicants propose that one or more of the gene sequences B15R and B18R may be inactivated, or part or all of one or more of these gene sequences may be deleted from the viral genome to allow (i) greater attenuation of the virus; and/or (ii) enhancement of immunogenicity of recombinant vaccinia virus; and/or (iii) further gene sequences insertion sites, so that more foreign DNA may be included in the virus. Inactivation of the gene sequences may be by mutation or the insertion of foreign DNA. Alternatively, one or more of the gene sequences B15R and B18R may be changed to alter the function of a protein product encoded by the nucleotide sequence.
Mutation of the nucleotide sequence may be effected by the deletion, addition, substitution or inversion of one or more nucleotides.
DNA sequences encoding one or more heterologous polypeptides may be incorporated in the viral genome. The DNA sequences encoding the heterologous peptides may be inserted into one or more ligation sites created by the deletion or deletions from the viral genome. A heterologous peptide is one not normally coded for by wild type vaccinia virus. Typically the heterologous nucleotide sequence will encode an immunogen or a
desirable polypeptide product. An immunogenic
polypeptide will be substantially homologous to an epitope expressed by a pathogenic organism during
infection, and which is seen by the infected individual as foreign.
The recombinant vaccinia viruses as described have the potential for enhanced immunogenicity. This may result from the deletion of vaccinia genes B15R and/or B18R which cause immunosuppression.
The recombinant vaccinia vectors as described may be used as immunogens for the production of monoclonal and polyclonal antibodies or T-cells with specificity for heterologous peptides encoded by DNA sequences ligated into the viral genome. The term antibody as used above should be construed as also covering antibody fragments and derivatives of a parent antibody and which have the same specificity as the parent antibody.
The monoclonal antibodies, polyclonal antibodies, antisera and/or T-cells produced by the use of the recombinant virus vectors hereof can be used in the diagnostic tests and procedures, for example, in
detecting antigen in a clinical sample. They can also be used therapeutically or prophylactically for
administration by way of passive immunisation.
Diagnostic test kits may comprise monoclonal
antibodies, polyclonal antibodies, antisera and/or T-cells obtained by the use of the recombinant vaccinia vectors described.
Vaccines and medicaments may comprise a recombinant vaccinia virus as described. These may have enhanced safety and immunogenicity over current vaccinia virus strains for the reasons indicated.
A method of attenuating a vaccinia virus vector may comprise: a) deleting part or all of one or more of the following nucleotide sequences from the viral genome; and/or b) inactivating one or more of said nucleotide sequences by mutating said nucleotide sequences or by inserting foreign DNA; and/or c) changing said one or more nucleotide sequences to alter the function of a protein product encoded by said nucleotide sequence;
which nucleotide sequences are sequences designated herein as: i) B15R, ii) B18R.
A vaccinia virus vector as described may be used to prepare a vaccine or a medicament. The translation products encoded by the nucleotide sequences B15R and B18R may have pharmaceutical utility. In particular, they may have utility as anti-inflammatory medicaments.
These genes and/or translation products thereof may be used in processes relating to the preparation of an anti-inflammatory medicament. As stated earlier, the gene product B15R binds IL-l, though not IL-6.
Surprisingly, this binding is specific for IL-1β as IL-1α and IL-lra do not bind to the B15R gene product.
Therefore, the B15R translation product or
functional equivalents thereof will have useful
pharmaceutical applications. Although many cell types synthesise both IL-1α and IL-1β it has been shown that most IL-1α remains in the cytosol of the cells, while much of the IL-1β is released into the extracellular space and circulation. In humans circulating IL-1β can be seen in the plasma of patients with sepsis, acute organ rejection and rheumatoid arthritis, while IL-1α is rarely detected in such patients. This evidence would support the view that IL-1β is the IL-1 form which is released by the cell and acts on other"cell types in pathological situations (Dinarello, C.A., & Wolff S.M., 1993 New Eng. J. Med. vol.328 p106-113). IL-1β is implicated in chromic inflammatory conditions such as rheumatoid arthritis and it has the following functions in relation to inflammation: i) it acts as a T-cell activator; ii) it induces fibroblast proliferation; iii) it increases the production of mediators of the
inflammatory response e.g. prostaglandin-E2 from
fibroblasts and iv) it increases the production of collagenase from chondrocytes (Digiovine F.S. et al, 1990, Immunol. Today Vol.11 p13-20). Therefore the gene product of B15R which can bind to IL-1β provides a means of blocking the activity of IL-1β in order to control and/or reduce the symptoms of inflammation.
Furthermore, IL-1β acts as a mediator in septic shock and it is a growth factor for certain malignant cells e.g. leukaemic cells. Therefore the gene product of B15R provides a means for controlling septic shock and growth of certain tumour cells.
Data presented in this application demonstrate the key role of IL-1β in controlling fever. Thus the IL-1β receptor may be used to control disease mediated by excessive fever.
Therefore, the present invention provides use of the nucleotide sequence designated herein as B15R, or of a nucleotide sequence coding for an allele of, or
functionally equivalent fragment, derivative or variant of the polypeptide encoded by the B15R nucleotide
sequence, in a process relating to the manufacture of a medicament for the treatment of a condition in which IL-1β is involved in the mediation of one or more
symptoms associated with the condition. Also provided is use of a polypeptide which is encoded by the nucleotide sequence designated herein as B15R or of an allele of, or functionally equivalent fragment, derivative or variant of the polypeptide, to manufacture a medicament for the treatment of a condition in which IL-1β is involved in the mediation of one or more symptoms associated with the condition.
An allele of, or functionally equivalent fragment, derivative or variant of the B15R polypeptide may be any polypeptide with substantial homology to part or all of the B15R polypeptide and which also has the ability to bind IL-1β.
Conditions may be fever, inflammation, diseases such as rheumatoid arthritis in which inflammation is
presented as a symptom, septic shock, and certain
leukaemias and/or cancers, inflammatory bowl disease, graft versus host disease and diabetes. The results disclosed hereafter also demonstrate that surprisingly the cytokine IL-1β is the primary mediator of fever.
Previous results have shown that a number of different cytokines including IL-1α, IL-1β, IL-6 and TNFα can cause fever (Rothwell, N.J. 1990, European Cytokine Network, Vol.1, p211-213; Kluger M.J. 1991, Physiological Reviews Vol.71 p93-127). However, in infection with vaccinia virus, inhibition of IL-1β activity alone can clearly abolish the fever response. Therefore, the present invention particularly provides use of the nucleotide sequence designated herein as B15R, or a nucleotide sequence coding for an allele of, or functionally
equivalent fragment, derivative or variant of the
polypeptide encoded by the B15R nucleotide sequence in a process relating to the manufacture of a medicament for the treatment of fever. Also provided is use of a polypeptide encoded by the nucleotide sequence designated herein as B15R or of an allele of, or functionally equivalent fragment, derivative or variant of the
polypeptide to manufacture such a medicament. Also provided is a pharmaceutical which is an anti-fever medicament. Also provided is a pharmaceutical which is an anti-fever medicament comprising a polypeptide which is encoded by the nucleotide sequence designated herein as B15R, or an allele of, or functionally equivalent fragment derivative or variant of the polypeptide.
Also provided is a diagnostic reagent for the detection or measurement of interleukin-1β in a sample which reagent comprises a polypeptide encoded by the nucleotide sequence designated herein as B15R, or of an allele of, or functionally equivalent fragment,
derivative or variant of the polypeptide. Also provided is a diagnostic kit which comprises a diagnostic reagent as described above and one or more ancillary kit
components for making the detection or measurement of interleukin-1β.
Also provided is a method of diagnosing a patient's condition in which interleukin-1β is involved in the mediation of one or more symptoms associated with the condition, which method comprises testing a sample of biological material obtained from the patient using a diagnostic reagent or kit as described above.
Also provided is a method of treating a patient suffering from a condition in which interleukin-1β is involved in the mediation of one or more symptoms
associated with the condition, which method comprises administering a medicament prepared as described above.
Also provided is a method of treating a patient for fever which comprises administering a medicament prepared as described above, or a pharmaceutical as described above.
The proteins may be produced in a recombinant system according to techniques well known in the art. Thus the nucleotide sequences provided herein could be inserted into a suitable expression vector (not necessarily vaccinia, for example the baculovirus system described herein). Such vectors can then be used to infect or transform a cell line suitable for the production of these particular proteins.
Reagents comprising polypeptides such as the B15R gene product, or alleles of or functionally equivalent fragments, derivatives or variants of that gene product may be used as research tools as a means to study the function of IL-1β versus IL-1α.
The applicants have provided herein sequence
information for B15R and B18R and identified the location of their nucleotide sequences in the viral genome.
Having done this, it is within the capacity of one skilled in the art to either inactivate these sequences in or delete these sequences from the VV genome or change them to alter the function of the encoded protein
product. All the necessary standard procedures are described in Molecular Cloning, eds. Sambrook, Fritsch and Maniatis, Cold Spring Harbour Laboratory Press 1989.
Furthermore, it would be within the normal capacity of one skilled in the art to use the sequence information provided herein to develop useful pharmaceuticals and immunogens as herein provided; to use the immunogens to produce antibodies and the like; to use the antibodies in kits and pharmaceuticals. Brief Description of the Drawings
In order that the present invention may be
understood more clearly, the identified gene sequences will be described more fully with reference to the figures listed below.
Figure 1(A). HindIII restriction map of the 186kb W genome. The 9.8kb SalI I fragment is expanded to show the position and direction of transcription of the genes B15R and B18R and the serpin genes (Smith, G.L., Howard, S.T. and Chan, Y.S. (1989). J. Gen. Virol. 70, 2333-2343) ( this nomenclature indicates the genes are the fifteenth and eighteenth ORFs starting from the left end of the HindIII B and are transcribed rightwards towards the genomic terminus).
Figure 1(B). Nucleotide sequence and deduced amino acid sequence of gene B15R. Potential transcriptional control signals are underlined and a possible signal peptide at the N-terminus is boxed. Sites for the addition of N-linked carbohydrate (NXS/T) are boxed and the cys residues likely to form disulphide bonds within Ig-like domains are stippled.
Figure 1(C). Nucleotide sequence and deduced amino acid sequence of gene B18R. The three amino acid
positions at which the sequence differs from the
published sequence of this gene from another strain of VV (Ueda, Y., Morikawa, S. and Matsuura, Y. (1990)) are shown. Other features as marked in (B).
Figure 2. Amino acid alignment of the Ig domains from B15R and B18R with the Ig-like domains of the human and murine IL-1RI, the human IL-6R, the VV haemagglutinin (VV HA), domain 1 of the fasciclin II, domain 3 of myelin-associated glycoprotein and the V-domain of Ig kappa. The regions predicted to form the β-strand structures of Ig-like domains are indicated above the alignment. Residues identical in 6 or more sequences are boxed. A few residues between the β-strands B and C have been omitted. Also omitted for brevity are β-strands D and, where appropriate, C' and C". Higher numbers of residues (about 30 or more) between strands C and E are indicative of the V domains.
Figure 3. Amino acid alignment of B15R with the external regions of the IL-lRI from human and mouse and the signal sequence and single Ig domain of human IL-6R. Gaps have been introduced to maximise the sequence alignments and are indicated by dashes. Where 4
sequences are aligned, the boxes indicate identical amino acids in three sequences, otherwise boxes indicate complete conservation in all aligned sequences.
Potential sites for addition of N-linked carbohydrate are underlined. Arrows and numbers mark the cysteines predicted to form intradomain disulphide bonds of Ig-like domains.
Figure 4. S1 Mapping of 5' Ends of mRNAs Coding for B15R and B18R. Specific 5' radiolabeled probes (lane P), prepared as described in Experimental Procedures, were hybridized with yeast transfer RNA (lane 1) or vaccinia virus early (lane 2) or late (lane 3) RNA and digested with SI nuclease. Nuclease-resistant fragments were resolved on a sequencing gel alongside an M13 sequencing ladder (lanes A,C,G and T). Autoradiographs
corresponding to B15R (A) and B18R (B) probes are shown. The sizes of the probes and fragments protected are indicated in bases. Indicated below the autoradiographs are the probe position relative to the ORFs (underline and asterisks), the nucleotide and the deduced amino acid sequence at the 5' end of the ORFs, the vaccinia late promoter consensus sequence (underline), and the sites of transcriptional initiation (asterisks).
Figure 5. Structure of Recombinant Vaccinia Virus Genomes. Vaccinia virus DNA was digested with HindIII (A) or ClaI (B), and fragments were resolved on an agarose gel and transferred to nitrocellulose. Filters were probed with fragments containing the gene and flanking sequences of B15R (lane a) or B18R (lane b), with an internal oligonucleotide to B15R (lane c) or an internal fragment to B18R (lane d). Sizes in kilobases are indicated. Schematic representations of the
structure and sizes of the relevant HindIII (H) or Clal (C) fragments containing the TK gene (hatched box), B15R gene (stippled box), or B18R gene (closed box) in the different viruses are included.
(A) Recombinant viruses overexpressing the ORFs, vB15R and vB18R, compared with WR.
(B) Deletion mutants v B15R and v B18R compared with WR. The ClaI fragments containing the genes either decreased to the expected size or were not detected in the deletion mutants when hybridized to probes that consisted of the ORF and flanking regions (lanes a and b) or probes corresponding to internal sequences (lanes c and d), respectively.
Figure 6. Identification of B15R and B18R Gene Products in Vaccinia- and Baculovirus-Infected Cells.
(A) Identification of B15R from vaccinia virus-infected cells. BS-C-l cells were mock infected (M) or infected with WR, vB15R, or v B15R and pulse-labeled with 35STrans-label either from 2 to 4hr after infection in the presence of cytosine arabinoside (E) or from 6 to 8 hr after infection in the absence (L) or presence (T) of tunicamycin. Cells and media were immunoprecipitated with rabbit antiserum raised against AcB15R-infected Sf cells, and the samples were analyzed by SDS-PAGE. A fluorograph is shown. Twice as much material was loaded onto gels from the supematants than from cells, and the fluorograph was exposed twice as long. Molecular size markers and the size of the B15R gene products (open arrow-head) are indicated in kilodaltons.
(B) Identification of B18R from vaccinia virus-infected cells. BS-C-l cells were infected with vB18R or v B18R and pulse-labeled with 35STrans-label from 6 to 8h after infection, the cell extracts and media were
immunoprecipitated with antiserum raised against B18R expressed in baculovirus-infected cells, and the samples were resolved by SDS-PAGE. A fluorograph is shown. As in (A), the quantity of sample from medium was estimated to correspond to about four times the amount analyzed from cells. Molecular size markers and the size of the B18R gene products (closed arrowhead) are indicated in kilodaltons.
(C) Expression of B15R and B18R in baculovirus-infected insects cells. Sf cells infected with AcNPV, AcB15R, or AcB18R were pulse-labeled with 35STrans-label for 2 hr after 24 hr of infection. Proteins present in cells and media were analyzed by SDS-PAGE and visualized by autoradiography. As in (A), the quantity of sample from medium was estimated to correspond to about four times the amount analyzed from cells. The B15R (open arrowhead) and B18R (closed arrowhead) gene products and the molecular size markers are indicated in kilodaltons. The positions of β-galactosidase (βgal), coexpressed with B15R and B18R in the recombinant baculoviruses, and polyhedrin (P), expressed only. in AcNPV, are shown.
Figure 7. Binding Assays to IL-1α, IL-1β and IL-6.
(A) Nitrocellulose binding assay. Triton X-100 cell extracts (C) ( 5μl ) or concentrated medium (M) ( 10μl ) from
EL4 6.1 C10 cells, U266 cells, mock- or vaccinia (WR)-infected TK-143 cells, and Sf cells infected with AcNPV, AcB15R, or AcB18R were dotted onto nitrocellulose
filters, and the membranes were incubated with
radioiodinated IL-1α (120 pM), IL-1β (200 pM) or IL-6
(120 pM). The volume added to the assay corresponded to 5 × 106 cell equivalents were used. An autoradiograph is shown.
(B) Soluble receptor binding assay. Tissue culture supematants (MEDIUM) or Triton X-100 cell extracts
(CELLS ) from the sources indicated were incubated with 100 pM of 125I-IL-1α (open box), 125I-IL-1β (closed box), or 125I-IL-6 (stippled box) in solution, and the binding was determined by the polyethylene glycol precipitation method. The amount of medium used corresponded to 1 × 105 cell equivalents. In the case of the detergentsolubilized cell extracts, 5 × 105 cell equivalents were added, except for EL4 6.1 C10 and U266 cells in which 2 × 106 cell equivalents were used. The bound radioactivity is shown.
Figure 8. Binding Assay in Solution to Recombinant Viruses. A volume of medium corresponding to 1 × 104 cell equivalents from uninfected TK-143 cells (MOCK) or from TK-143 or Sf cells infected with the indicated vaccinia virus or baculovirus recombinants, respectively, was incubated with 180 pM of 125I-IL-1β in the soluble receptor binding assay. The radioactivity bound to soluble receptor present in supematants is represented.
Figure 9. Binding Characteristics of Vaccinia IL-1β Receptor.
(A) Competition for binding of 125I-IL-1β to soluble receptor. Medium from WR-infected cells, corresponding to 4 × 104 cell equivalents, was incubated with 100 pM of 125I-IL-2β in the presence of the indicated
concentrations of unlabeled IL-1α (open circle), IL-1β (closed circle), IL-1RA (open square), or IL-6 (closed square), and the radioactivity bound to the soluble receptor was determined by the polyethylene glycol precipitation method. Binding is expressed as a
percentage of the binding occurring irr the absence of unlabeled IL (1477 cpm).
(B) Scatchard analysis of 125I-IL-1β binding to medium from vaccinia virus- or baculovirus-infected cells. Medium from cultures infected with WR ( 1.8 μl, 8 x 103 cell equivalents), vB15R (0.56 μl, 2.5 × 103 cell equivalents), or AcB15R (0.15 μl, 250 cell equivalents) was incubated with different concentrations (25-1000 pM) of radiolabeled IL-1β for 2 hr at room temperature, and the radioactivity bound was determined by the
polyethylene glycol precipitation method. Data were converted to the Scatchard co-ordinate system. Binding shown represents specific binding. The data were
analyzed by the LIGAND program.
Figure 10. Competition Experiments to EL4 6.1 C10 and U266 Cells. Different cell equivalents of medium from baculovirus-infected cells expressing B15R (AcB15R; open circle) or B18R (AcB18R; closed circle) were
incubated with 130 pM of 125I-IL-1α (A), 180 pM of
125I-IL-1β (B), or 100 pM of 125I-IL-6 (C) for 1 hr at 4°C. At the end of the incubation period, 2.5 × 106 EL4 6.1 C10 cells were added to the samples containing radiolabeled IL-1α (A) and IL-1β (B), and 2.5 × 106 U266 cells were added to those containing radioiodinated IL-6 (C). Samples were incubated at 4°C for 2 hr, and the radioactivity bound to the cells was determined by phthalate oil centrifugation. Competition of binding with unlabeled IL-1α (open square), IL-1β (closed
square), or IL-6 (open triangle), expressed in
nanomolars, was included as a control. The percentages refer to the binding in the absence of competitor, which was 3720 cpm for 125I-IL-1α and EL4 6.1 C10 cells (A), 2040 cpm for 125I-IL-1β and EL4 6.1 C10 cells (B), and 4963 cpm for 125I-IL-6 and U266 cells (C).
Figure 11. Effect of the Deletion of B15R from Vaccinia Virus on the Infection of Mice.
(A) Groups of five mice were intransally infected with 3 × 107 (panels a), 107 (panels b), 106 (panels c), 105 (panels d) or 104 (panels e) pfu of WR (open circle) or v B15R (closed circle) and examined daily for symptoms of illness or death. The number of animals that
presented strong symptoms of illness (including death) and the accumulated number of mortalities are represented for each dose of virus at different days of infection. No differences were observed between day 12 and day 17. The (f) panels summarize the onset of symptoms (left) and the number of mortalities (right) that occurred at different days after infection.
(B) Groups of 10 mice were intransally infected with 105 (panels a) of 104 (panels b) pfu of WR (open circle) or v B15R (closed circle). Symptoms of illness were scored from zero to four, and the mean value of each group was represented. Animals were weighed individually each day and the mean group weight was expressed as the percentage of the mean weight of that group of animals immediately prior to infection. No mortalities occurred at these doses of virus.
Figure 12. Effect of Expression of the Vaccinia IL-1β Receptor on Mice Infected with Vaccinia Virus. A representative mouse 5 days after infection with 105 pfu of WR (a) or v B15R (b) is shown. Note the ruffled fur in (b), which correlated with accelerated weight loss (see Figure 8B, panels a).
Figure 13. Binding of Murine and Human IL-1β to Different Strains of Vaccinia Virus, Rabbitpox, and
Cowpox. Tissue culture medium (1 × 105 cell equivalents) from TK-143 cells infected with the indicated viruses was incubated in a binding assay in solution with 100 pM of radioiodinated IL-1β or mIL-1β, expressed in femtomoles, is shown. One femtomole corresponded to 935 or 535 cpm for IL-1β or mIL-1β, respectively.
Figure 14. Kinetics of symptoms of illness and mortality in mice infected with recombinant vaccinia viruses. BALB/c mice were intranasally infected with WR, v B15R or v B18R as described in Table 2.
(A) The total number of deaths in animals infected with different recombinant viruses is represented as a function of days post-inoculation.
(B) Animals inoculated with the different
recombinants were examined for illness and the number of animals with symptoms (including mortality) in each group were represented as a function of time. Symptoms scored were (a) possession of ruffled fur (b) hunching and (c) immobility.
Figure 15. Temperature of BALB/c mice following intranasal infection with 105 pfu of either wild type vaccinia virus (strain WR: open circles), or a deletion mutant lacking the B15R ORF encoding a soluble receptor for interleukin-1β (IL-1β: closed circles), or a
revertant virus in which the B15R gene has been re-introduced into the deletion mutant virus (triangles). These data represent the mean temperature of groups of 10 animals. Figure 16. Temperatures of BALB/c mice following intranasal infection with 10^ plaque forming units of vaccinia virus strain Copenhagen (A), Tashkent (B) or Tian-Tan (C). These data represent the mean temperature of groups of 10 animals. Strain Tian-Tan expresses a soluble receptor for IL-1β while strains Copenhagen and Taskent do not. In panel D the temperature profiles of animals infected with Copenhagen or Tian-Tan are
compared. Data are expressed showing the net increase or decrease in temperature following infection.
All the genetic manipulations described below were carried out according to standard procedures (Molecular cloning, eds. Sambrook, Fritsch & Maniatis, Cold Spring Harbor Laboratory Press, 1989) and the conditions used for enzymatic reactions were as recommended by the manufacturer (GIBCO-BRL Life Technologies).
Determination of the Nucleotide and Amino Acid Sequences
The nucleotide sequence of the SalI I restriction fragment of the vaccinia virus genome (strain WR) were determined by established methods (Sanger, F. et al.
(1980), J. Mol. Biol., 143, 161-178) and Bankier, A. and Barrell, B.G. (1983) Techniques in Life Sciences B508., 1-34, Elsevier).
For example, the 9.8kb SalI I fragment of vaccinia virus (strain WR) was isolated from cosmid 6, which contains virus DNA derived from a rifampicin resistant mutant (Baldick, C..J. & Moss, B. (1987) Virology 156, 138-145), and was cloned into SalI cut pUC13 to form plasmid pSalI I. The SalI fragment was separated from plasmid sequences and self-ligated with T4 DNA ligase. Circular molecules were randomly sheared by sonication, end-repaired with T4 DNA polymerase and Klenow enzyme, and fragments of greater than 300 nucleotides cloned into SmaI cut M13mp18. Single stranded DNA was prepared and sequenced using the dideoxynucleotide chain termination method (Sanger, F., Nicklen, S. & Coulson, A.R. (1977) Proc. Natl. Acad. Sci. USA. 74, 5463-5467), using
[35S]-dATP and buffer gradient polyacrylamide gels (Biggin, M.D., Gibson, T.J. & Hong, G.F. (1983), Proc. Natl. Acad. Sci. USA, 80, 3693-3695). For further details see (Bankier, A.T., Western, K.M. & Barrell, B.G. (1987) in Wu R. (ed) Methods in Enzymology 155, 51-93. Academic Press, London). The 6.3 kb SalI L fragment was similarly treated.
Computer Analysis
Nucleotide sequence data were read from
autoradiographs by sonic digitiser and assembled into contiguous sequences using programmes DBAUTO and DBUTIL (Staden, R. (1980) Nucleic Acids Res. 8, 3673-3694;
Staden, R. (1982) Nucleic Acids Res. 10, 4731-4751) on a VAX 8350 computer. The consensus sequence was translated in 6 frames using programmes ORFFILE and DELIB (M.
Boursnell, Institute of Animal Health, Houghton, UK). Open reading frames were compared against SWISSPROT protein database verion 14 and. against the applicant's own database of vaccinia amino acid sequences using programme FASTP (Lipman, D.J. & Pearson, W.R. (1985) Science 227, 1435-1441). Alignments of multiple protein sequences were performed using programme MULTALIGN
(Barton, G.J. & Sternberg, M.J.E. (1987) J. Mol. Biol. 198, 327-337).
There follows a detailed description of the
individual gene sequences B15R and B18R and their
relationship to IL-1 receptor.
IL-1 Receptors
IL-1, a cytokine produced in response to infection and tissue injury, is involved in the regulation of the inflammatory and immune responses and in the activation of a broad spectrum of systemic effects that contribute to host defense (Dinarello 1988, 1989; Di Giovine and Duff 1990). The two forms of IL-1, IL-1α and IL-1β produce similar biological effects that are mediated by interaction with specific receptors in different cells.
There are two classes of IL-1 receptors, and both bind IL-1α and IL-1β with similar affinities (Dower, S.K., and Urdal, D.L. (1987)). The 80 kDa type I IL-1 receptor is found on T cells and fibroblasts (Bird, T.A., and Saklatvala, J. (1986) Nature 324, 263-266), while the 60 kDa type II IL-1 receptor is present in B cells and macrophages (Matsushima, K. et al (1986) J. Immunol. 136, 4496-4508). Sequence of cDNA clones of both receptors revealed that they belong to the Ig superfamily and each have three Ig-like domains followed by a transmembfane anchor sequence and a cytoplasmic tail (Sims, J.E. et al (1988) and (1989); MacMahan et al (1991)). Binding studies have shown heterogeneity in the IL-1 receptor on B cells concerning the affinity of IL-1α or IL-1β
(Benjamin, D., et al, (1990)). The existence of a secreted receptor on Raji cells, which binds only IL-1β, has been reported (Symons, J.A., et al, (1991)).
There are different natural inhibitors that modulate the biological effect of IL-1 (Larrick, J.W. (1989);
Shields, J., and Mazzei, G.J. (1991)). The best
characterized natural inhibitor is designated IL-1 receptor antagonist (IL-1ra), which competes with both IL-1α and IL-1β for binding to the receptor but cannot trigger the cellular responses of IL-1 (Hannum et al (1990); Eisenberg S.P. et al, (1990); Carter, D.B. et al, ( 1990 )). Many studies have demonstrated the ability of IL-1ra to block the biological effects of IL-1 in vitro and in vivo (Dinarello, C.A., and Thompson, R.C. (1991)). Genes B15R and B18R
Genes B15R and B18R (Figure 1) from near the right hand inverted terminal repeat ( ITR) are predicted to encode proteins of 36.5 kDa and 40.7 kDa, respectively, that have an N-terminal hydrophobic sequence, possible attachment sites for N-linked carbohydrate and
hydrophobic residues near the C-terminus. These
properties are consistent with the mature proteins being either virion, cell-surface or secretory glycoproteins.
The nucleotide sequence and deduced amino acid sequence around the gene designated B15R is shown in figure IB. the nucleotide sequence shown is 11462-12664 nucleotides from the left end of the vaccinia virus HindIII B fragment and the coding region for B15R is at nucleotide positions 11584-12561 (or at nucleotides 815 to 1792 from the left end of the SalI I fragment).
Similarly, the nucleotide sequence and deduced amino acid sequence around the gene designated B18R is shown in figure 1c. The nucleotide sequence shown is 15448-16741 nucleotides from the left end of the vaccinia virus
HindIII B fragment and the coding region for B18R is at nucleotide positions 15568-16621 (or at nucleotides 4799 to 5851 from the left end of the SalI I fragment).
The single letter code is used for the designation of amino acids.
B15R and B18R each possess three domains with characteristics of the immunoglobulin (Ig) superfamily (Williams, A.F. and Barclay, A.N. (1988). Ann. Rev.
Immunol. 6, 381-405) namely a pair of cysteines forming an intradomain disulphide bridge, sequences predicted to form β-strand structures and an invariant tryptophan in β-strand C. In B15R these cysteines are present at positions 48 and 99, 143, and 194, and 242 and 309. In B18R the corresponding cysteines are at positions 73 and 129, 172 and 221, and 272 and 333. The distance between these cysteine pairs in B15R (51, 51 and 67 residues) suggest the first two domains are C regions while the third may be a V-domain. For B18R the distances (56, 49 and 61) suggest these are C-domains. These regions are aligned with selected Ig-like domains of IL-1R (IL-1 receptor and IL-6 receptor), VV haemagglutinin and Ig kappa (Hilschman, N. and Hoppe-Seyer's, Z. (1967)
Physiol. Chem. 348, 1077-1080), fasciclin II (Harrelson, A.L. and Goodman, C.S. (1988). Science 242, 700-708), chicken neural cell adhesion molecule (NCAM) (Hemperley, J.J. Murray, B.A., Edelman, G.M. and Cunningham, B.A.
(1986). Proc. Natl. Acad. Sci. USA 83, 3037-3041) and myelin-associated glycoprotein (Salzer, J.L., Holmes,
W.P. and Colman, D.r. (1987). J. Cell Biol. 104, 957-965) (Figure 2). In this alignment the β-strands C', C" and D have been omitted for brevity. The cysteines forming the intradomain disulphide bridge and the tryptophan in β-strand C are completely conserved. The relationship between the vaccinia proteins and the Ig family was confirmed by statistical computational analysis using the programme ALIGN (Dayhoff, M.O., Barker, W.C. and Hunt, L.T. (1983). Meth. Enzymol. 91, 524-545) (Table 1).
With B15R the highest individual scores are found against the human and murine IL-1RI domains. Domain 2 of B18R also scores well against the IL-1RI domains and overall there are highly significant scores against a wide range of Ig domains.
An alignment of B15R with the extracellular regions of IL-1Rs and IL-6R (Figure 3) and the above alignment of individual domains (Figure 2 ) indicates a closer
relationship between IL-1Rs, IL-6R and the VV Ig domains than other Ig members. This is exemplified by the following observations.
(1) B15R and the external region of IL-1Rs have a very similar length.
(2) There are additional conserved cysteines in
B15R, B18R and the IL-1Rs located near the beginning of β-strands A and G in domain 1 and at similar positions in domain 2 of the IL-1Rs and B18R. These cysteines lie within the 3-dimensional structure of an Ig C domain in positions probably allowing another intradomain
disulphide bond.
(3) In B15R, both IL-1Rs and IL-6R there is a proline following the invariant cysteine in β-strand B of domains 1 and 2, an unusual residue in this position. B18R domain 1 also contains proline at this position.
(4) In β-strand F of domain 3 of B15R and both IL-1R sequences, the glycine typical of other Ig domains is absent. Moreover, the otherwise invariant tyrosine is replaced in both IL-1Rs and in B15R with phenylalanine.
(5) A glycosylation site is conserved in domain 1, β-strand F of IL-1Rs and B15R despite divergence of amino acid sequence.
(6) Domain 3 does not contain additional cysteines and is longer than 1 and 2 in B15R, B18R and the IL-1Rs.
Protein sequence comparisons show that these VV proteins are related to each other (22.5% identity), to the human and murine IL-1R, the human IL-6R (Yamasaki, K., Taga, T., Hirat, Y., Yawata, H., Kawanishi, Y., Seed, B., Taniguchi, T., Hirano, T. and Kishismoto, T. (1988). Proc. Jpn. Acad, 64, 209-211) and the immunoglobulin (Ig) superfamily (Williams, A.F. and Barclay, A.N. (1988).
Ann. Rev. Immunol. 6, 381-405). Members of this family contain varying numbers of structurally similar domains (Ig domains) and perform diverse functions, although a unifying theme is surface interations between cells or by binding cytokines. The VV haemagglutinin is another member of this superfamily (Jin, D., Li, Z., Jin, Q., Yuwen, H. and Hou, Y. (1989). J. Exp. Med. 170, 571-576). The B18R sequence from VV strain IHD was recently
reported but the relationship to interleukin receptors and the Ig superfamily was not described (Ueda, Y.,
Morikawa, S. and Matsuura, Y. (1990). Virology 177, 588-594).
Cytokines IL-1 and IL-6 by binding to their
respective natural receptors mediate immune responses against an invading pathogen and cause inflammation. VV may be combatting this part of the immune response by producing proteins which mimic the receptors for IL-1 and IL-6.
Recently the sequences of additional (type II) human and murine interleukin-1 receptors were reported (McMahan et al., (1991)). These are more closely related to the vaccinia virus B15R gene product than are the formerly described type I IL-1 receptors. Indeed vaccinia B15R is more closely related to human and murine type II IL-1 receptors than either of these are to the type I IL-1 receptors. Since both type I and II IL-1 receptors have been shown to bind IL-1, the applicants have investigated whether or not the products of either B15R and/or B18R will also bind this cytokine.
The applicants report here that both B15R and B18R ORFs are actively transcribed, translated, and secreted to the medium during the vaccinia virus replication cycle. The B15R gene product is shown to bind IL-1β when expressed from vaccinia or from recombinant baculovirus. The role of the IL-1β binding activity in the biology of vaccinia virus was investigated by deleting the gene from the virus genome and analyzing the biological effects on infected mice. The biological effects on infected mice were also investigated for vaccinia virus without the B18R coding sequence. The presence of the binding activity in other orthopoxviruses is also presented.
EXPERIMENTAL PROCEDURES
Cells and Viruses
The cell line EL4 6.1 C10, a subclone of the mouse thymoma EL4 that expresses a high number of IL-1 binding sites (MacDonald et al, 1985), was a gift of H.R.
MacDonald (Ludwig Institute for Cancer Research,
Lausanne, Switzerland). U266 cells overexpressing IL-6 receptors (Taga, et al 1987) were obtained from the Cell Bank of the Sir William Dunn School of Pathology
(University of Oxford). These cell lines were grown in suspension in RPMI 1640 medium containing 10% fetal calf serum.
Sf 21 insect cells and AcNPV were obtained from R. Possee (Natural Environmental Research Council Institute of Virology and Environmental Microbiology, Oxford) and were cultured in TC100 medium (GIBCO) containing 10% fetal calf serum (Brown and Faulkner, 1977).
Vaccinia virus strain WR and recombinants derived from it were grown in CV-1 or BS-C-1 cells. TK-143 and HeLa D98 cells were used for the selection of
recombinants. The WR strain was obtained from B. Moss (National Institute of Health, Bethesda, Maryland) and cells were obtained from the American Type Culture
Collection. Cells were grown in minimal essential medium (GIBCO) supplemented with 10% fetal calf serum. Purified virus stocks were prepared by sedimentation through a sucrose cushion (Mackett et al, 1985). The Tashkent, IHD-J, and IHD-W strains of vaccinia virus and cowpox virus were obtained from M. Mackett (Paterson Institute for Cancer Research, Manchester, England) and J.D.
Williamson ( St. Mary's Hospital Medical School, London). The New York City Board of Health vaccine strain (Wyeth) was obtained from Wyeth Laboratories, and the Lister strain was obtained from Vestric Limited. Rabbitpox virus was provided by R.W. Moyer (University of Florida, Gainesville, Florida). The Temple of Heaven strain
(Tian-Tan) and a temperature-sensitive mutant of the
Copenhagen strain were obtained from J. Zhou (Princess Alexandra Hospital, Brisbane, Australia) and R. Drillien (University Louis Pasteur, Strasbourg, France),
respectively.
Reagents
Radioiodinated human recombinant IL-6 was purchased from Amersham. IL-1α had been, radioiodinated using the chloramine-T procedure to a specific activity of 70-120 μCi/μg. IL-1β and IL-6 had been labeled with Bolton Hunter reagent to a specific activity of 80-180 μCi/μg and 800-1200 Ci/mmol, respectively. Unlabeled human recombinant IL-1α (code 86/632), IL-1β (code 86/680), and IL-6 (code 88/514) were obtained from the National
Institute for Biological Standards and Control ( South Mimms, Hertfordshire, England). The activity for IL-1α and IL-1β was 105 U/μg and for IL-6 was 5 × 103 U/μg.
Unlabeled human recombinant IL-IRA, specific activity 1 × 105 to 1.4 × 105 U/mg, was purchased from British Biotechnology.
Plasmid Constructions
Restriction endonuclease digestions, PCR, DNA ligations, and plasmid DNA preparations were performed according to standard procedures ( Sambrook et al, 1989). Cloning of B15R into pUC118
A derivative of plasmid pUC118 (Vieira and Messing, 1987) was constructed that contained the entire B15R ORF, lacking most of the flanking regions, and with convenient restriction sites at each end. A combination of subcloning and PCR was used. The left end SalI-XbaI fragment of the SalI I fragment of vaccinia DNA
containing B15R was inserted into SalI- and BamHI-cut pUC118, and the resultant plasmid was called pAA1. To remove most of the 3' flanking region of B15R, pAA1 was digested with EcoRV and XbaI, and the largest fragment was gel purified, end filled with Klenow fragment, and self-ligated to form pAA3, which contains 348 bp of the 3' flanking region. To introduce restriction sites close to the initiator methionine of B15R, a PCR copy of 5' region of the ORF was constructed, using plasmid pAA1 as template and an oligonucleotide containing the sequence of the first 21 nt of B15R and the recognition sequence for BamHI and NcoI (B15R-1;
5'-CCCGGATCCACCATGGGTATACTACCTGTTATA-3') and an
oligonucleotide that hybridizes to an internal sequence of B15R, corresponding to nucleotides 1008-1025 of the SalI I fragment (B15R-2; 5'-CCGCTCCTCGTTTTTCCC-3'). The fourth nucleotide of B15R in the PCR fragment was G instead of A to create a NcoI recognition sequence, giving rise to a serine to glycine substitution in the second amino acid of the protein. The 222 bp PCR
fragment was digested with AccI, forming a 20 bp fragment containing the BamHI and NcoI restriction sites bound to the first 8 nt of B15R, and was treated with
polynucleotide kinase. This fragment was cloned into pAA3 digested with Hindi and AccI, which removed the 5' flanking region, to render plasmid pAA4. This construct was confirmed by DNA sequencing.
Cloning of B18R into pUC4K
The XbaI-SalI right end fragment of the SalI I fragment of vaccinia DNA, which contains B18R, was cloned into XbaI- and SalI-cut pUC118 to form plasmid pAA2. A SspI fragment, containing the whole B18R ORF and 22 5' and 263 3' nucleotides, was excised from pAA2 and cloned into pUC4K (Vieira and Messing, 1982) digested with SalI and end filled with Klenow fragment to create blunt termini, and the resulting plasmid was termed pAA5. Vectors for Overexpression in Vaccinia Virus
The transfer vector used for overexpression of B15R and B18R in vaccinia virus was pRK19 (Kent, R.K. 1988), which contains the vaccinia virus 4b promoter to control the transcription of the inserted gene, flanked by sequences of the TK gene that allow insertion in the TK locus of the virus genome. BamHI fragments containing the ORFs were excised from pAA4 and pAA5 and cloned into the BamHI site of pRK19, and the resulting plasmids were named pAA10 and pAA11, respectively.
Vectors for Expression in Baculovirus
The transfer vector for construction of baculovirus recombinants was pAcDZl, which uses the polyhedrin promoter to drive the transcription of foreign genes and coexpresses Escherichia coli β-galactosidase for
selection of the recombinants (Zuidema et al, 1990).
This vector was provided by J.M. Vlak (Department of Virology, Agricultural University, Wageningen, The
Netherlands). The genes were excised from pAA4 and ρAA5 with BamHI and inserted into BamHI-cut pAcDZl, forming pAA14 and pAA15, respectively.
Vectors for Deletion of the Genes in Vaccinia Virus by Transient Dominant Selection
The flanking sequences of B15R and B18R were excised from pAA1 or clones from a M13 library containing random subfragments of the SalII fragment of vaccinia DNA, which were used to sequence this region of the vaccinia virus genome (Smith et al, 1991a), and were cloned into pSJH7 (Hughes et al, 1991). The 5' flanking region of B15R was obtained by digestion of the replicative form of the M13 clone SalII.144 with EcoRI and SphI, and the 3' flanking region was excised from pAA1 by digestion with SphI and BamHI. Both fragments were, cloned in one step into
EcoRI- and BamHI-cut pSJH7. The resultant plasmid, called pAA16, contained 360 and 1316 nt of the 5' and 3' flanking sequence, including 17 and 252 nt of the coding sequence, respectively, so that 72% of the B15R coding sequence was deleted. DNA fragments containing the flanking sequences of B18R were obtained by BamHI and EcoRI digestion of the replicative form of the M13 clones SalII.44 and SalII.81. Both fragments, of 400 and 449 bp, were cloned in one step into EcoRI-cut pSJH7, and the plasmid in which the sequence of B18R was transcribed in the same direction as the Ecogpt gene present in pSJH7 was chosen and called pAA17. This construct lacked 18 nt of the 5' flanking region and 92% of the ORF and retained only 78 nt of the 3' end of the coding region.
S1 Mapping of the 5' End of mRNAs
To prepare the 32P-labeled DNA probe used to
identify the 5' end of the mRNA coding for B15R, a PCR fragment was obtained with the oligonucleotide B15R-2 (above) and the 17-mer sequencing primer (-20), using pAA1 as template. The PCR product was purified, labeled with [ -32P]ATP and polynucleotide kinase, and
subsequently digested with SalI, giving rise to a 1024 bp fragment containing 211 nt of the 5' coding region of B15R.
To map the 5' end of the B18R transcript, pAA2 was digested with EcoRI, a band of 505 bp was purified and dephosphorylated with calf intestinal alkaline
phosphatase, and the 5' ends were labeled with [ -32P]ATP using polynucleotide kinase. After digestion with DraI, a fragment of 379 bp was isolated that contained 98 nt corresponding to the 5' coding region of B18R.
Both 32P-labeled fragments specific for B15R and B18R were hybridized to 10μg of vaccinia virus RNA obtained at 8 hr after infection from cells infected in the presence (early) or absence (late) of cycloheximide or yeast transfer RNA. The hybrids were digested with SI nuclease, and the protected fragments were separated on 6% polyacrylamide sequencing gel and detected by
autoradiography as described (Moore and Smith, 1992). An M13 sequencing ladder was used as size markers.
Transfection and Selection of Recombinant Viruses
Sf cells were cotransfected with purified AcNPV DNA and pAA14 or pAA15 using the calcium phosphate precipitation technique, and the recombinant viruses were identified by staining with X-GaI as described (Zuidema et al, 1990). The insertion of foreign genes (B15R and B18R) into the baculovirus genome was confirmed by
Southern blot hybridization of 32P-labeled specific probes on viral DNA digested with HindIII (data not shown). The recombinant viruses expressing B15R and B18R were plaque purified five times and called AcAA3 and AcAA4, respectively, and are referred to here as AcB15R and AcB18R.
Recombinant vaccinia viruses were constructed by standard procedures (Mackett et al, 1985). The genomes of viruses containing a second copy of B15R or B18R in the TK locus of the vaccinia DNA were analyzed by
Southern blotting, using viral DNA extracted from virus cores (Esposito et al, 1981). These structures were confirmed by PCR using oligonucleotides that hybridized to the 5' and 3' ends of the TK gene, which gives rise to a longer PCR product in the recombinant viruses compared with WR owing to the insertion of foreign DNA in the TK locus (data not shown). The recombinant vaccinia viruses containing a second copy of B15R and B18R were called vAA1 and vAA4, respectively, and are referred to here as vB15R and vB18R. Vaccinia virus deletion mutants were constructed by transient dominant selection as described elsewhere ( Falkner and Moss, 1990; Isaacs et al, 1990). Vaccinia viruses containing deleted versions of B15R and B18R were termed vAA5 and vAA6, respectively, and are referred to here as v B15R and v B18R.
The B15R-specific probe containing the ORF and 348 bp of the 3' flanking region was excised from pAA4 by digestion with BamHI and used for Southern blot
hybridization. The oligonucleotide B15R-2 was used as an internal probe for B15R ORF. A BamHI fragment excised from pAA5, containing B18R ORF and 263 bp of the 3' flanking region, was used as a B18R-specific probe. The internal probe for B18R was obtained by excision of a 424 bp EcoRI fragment from pAA2. Preparation of Antisera
Rabbit sera specific for B15R or B18R were obtained by immunization with AcB15R- or AcB18R-infected Sf cell extracts and concentrated medium according to standard procedures (Harlow and Lane, 1988).
Metabolic Labeling of Proteins and Immunoprecipitation
Sf or BS-C-l cells were infected with the
baculovirus or vaccinia virus recombinants, respectively, at high multiplicity of infection (20-40 pfu/cell). At the indicated times of infection, infected cells were pulse-labeled with 750 μCi/ml 35STrans-label (ICN
Biomedicals; a mixture of -80% [35S]methionine and -20% [35S] cysteine, 1200 Ci/mmol) in methionine-free TC100 medium or methionine- and cysteine-free minimal essential medium, respectively, in the absence of serum. Cytosine arabinoside (40 μg/ml) or tunicamycin (1 μg/ml) was added to the medium throughout the infection and during the pulse period when indicated.
Medium or cells were incubated in 10 mM Tris-HCl (pH 7.5), 150 mM NaCI, 1% sodium deoxycholate, 1% Nonidet P-40, 0.1% SDS, and 1 mM phenylmethylsulfonyl fluoride and immunoprecipitated by the indicated rabbit serum and protein A-Sepharose (Harlow and Lane, 1988). For the electrophoretic analysis, whole extracts or immune complexes were dissociated in sample buffer (0.4 M Tris-HCl, (pH6.3), 2.3% SDS, 10% glycerol, and 5% 2-mercaptoethanol) and analyzed by SDS-PAGE in 10% or 10-20% acrylamide gels as described (Laemmli, 1970).
Radioactive bands were detected by autoradiography or fluorography with salicilate (Harlow and Lane, 1988).
Preparation of Cell Extracts and Medium for IL-1 Binding Assays
Sf cells and TK-143 cells, grown in 175 cm2 or 80 cm2 flasks, were infected at a density of 1.5 × 105 to 2 × 105 cells/cm2 with a multiplicity of infection of 5-10 pfu per cell in serum-free medium. Cells and medium were harvested from vaccinia- or baculovirus-infected cells at 1 or 3 days after infection, respectively. The final concentration of the supematants was 1 × 106 to 5 × 106 cell equivalents per millilitre. The medium was
centrifugated at 3000 rpm for 10 min at 4°C, the pellet discarded, and supematants made 20 mM HEPES (pH 7.4) and 0.1% sodium azide. Supematants were stored at -70°C until used in binding assays in solution or concentrated and dialized against phosphate-buffered saline ( PBS ) at 4°C in a Micro-ProDiCon (Bio-Molecular Dynamics) with PA-10 ProDiMen dialysis membranes (MW 10,000) to a-final concentration of 5 × 107 cell equivalents per millilitre. The concentrated medium was made 1% in sodium azide and stored at -70°C. Cells were detached from the plastic by incubation with 0.5 mM EDTA in PBS and washed twice with PBS, and the pellet was resuspended in 1% Triton X-100 in PBS containing 1 mM phenylmethylsulfonyl fluoride to a final concentration of 1 × 108 cells per millilitre.
Samples were incubated on ice for 15 min and centrifuged at 12,000 × g for 30 min at 4°C as described (Urdal et al, 1988). The cell extracts were made 1% in sodium azide and stored at -70°C. Detergent-solubilized lysates of EL4 6.1 C10 and U266 cells were prepared in the same way to a final concentration of 4 × 108 cells per
millilitre. The supematants were harvested from cells seeded at a cellular density of 5 × 105 cells per
millilitre and grown in culture over a period of 3 days. The medium was concentrated in a Micro-ProDiCon to 5 × 107 cell equivalents per millilitre.
Sf and TK-143 cells were harvested for binding assays to intact cells by treatment with PBS containing 0.5 mM EDTA. EL4 6.1 C10, U266, Sf, and TK-143 cells were washed twice in serum-free medium and resuspended in binding medium.
Binding Assays
The binding medium used in the different assays was RPMI 1640 containing 20 mM HEPES ( pH 704), 1% bovine serum albumin, and 0.1% sodium azide. Solid phase binding assays on nitrocellulose were performed as described (Urdal et al, 1988). Binding to intact cells was carried out in duplicate in 150 μl of binding medium for 2 hr at 4°C, and bound 125I-IL was determined by phthlate oil centrifugation as described (Dower et al, 1985). In the competition assays of labeled ILs to intact cells, samples were preincubated with the ILs in 125 μl for 1 hr at 4°C. Subsequently, 2.5 x 106 EL4 6.1 C10 or U266 cells were added in 25 μl and incubated for 2 hr at 4°C.
Soluble receptor binding assays were performed by precipitating the ligand-receptor complexes with
polyethylene glycol and filtration through Whatman GF/C filters as described by Symons et al (1990).
Supematants were incubated in duplicate with labeled ILs in a final volume of 150 μl for 2 hr at room temperature. Background radioactivity precipitated in the presence of binding medium was subtracted. Kinetics experiments of 125I-IL-1β binding to vaccinia virus supematants showed that maximum binding was reached after 5 min at room temperature (data not shown). The saturation experiments to soluble receptor from WR, vB15R,a nd AcB15R were performed in a final volume of 75 μl in the same
conditions. The binding of 125I-IL-1β to supematants from v B15R or AcB18R were considered as nonspecific binding and subtracted from total binding. In previous experiments we determined that these values are similar to those obtained in the presence of 100-fold excess of cold IL-1β (data not shown). Binding data were analyzed using the LIGAND program (Munson and Rodbard, 1980).
In vivo Experiments
(1) Female BALB/c mice ( 5 to 6 weeks old) were anesthetized and infected intranasally with 20μl of the diluted virus in 1 mM Tris-HCI ( pH 9.0). Mice were weighed daily and monitored for signs of illness or death (Turner, 1967; Williamson et al, 1990). As a control, an aliquot of the dilutions of v B15R or WR used to inculate the animals was grown in TK-143 cells, and the absence or the presence of IL-1β binding activity in the medium at 24 hr after infection was confirmed in a binding assay in solution (data not shown).
(2) 4-6 weeks old BALB/c mice were intranasally infected with doses of WR, v B15R or v B18R as shown in Table 2. The table also shows the mortality of animals after 15 days.
RESULTS
Transcriptional Analysis of B15R and B18R
Transcription of genes from vaccinia virus is regulated in a temporal fashion, and genes are classified as early or late on the basis of their requirement for viral DNA synthesis (Moss, 1990b). S1 nuclease
protection experiments were performed to detect and map B15R-specific messenger RNAs ( mRNAs ) produced during infection. Figure 4A shows that the B15R-specific probe was partially protected from SI nuclease digestion by late viral RNA, and the size of the protected fragment mapped the transcriptional start site to the TAAAAT motif at the 5' end of B15R. Although the sequence TAAAT(G) has been shown to constitute a late promoter consensus sequence for vaccinia virus, a few exceptions have been found that possess an additional A TMOSS, 1990b). The B18R-specific probe was protected from S1 nucflease digestion by early viral RNA that initiated 16-18 nt upstream of the ORF (Figure 4B). This is consistent with the presence of vaccinia virus early transcriptional terminator signal TTTTTNT 13 nt downstream of the ORF (Smith and Chan, 1991) and is in agreement with results obtained by primer extension in the Lister strain (Ueda et al., 1990), although analysis of late RNA was not included in the previous report. The weak signal detected at late times probably corresponds to early transcripts still present in the late viral RNA sample since, for constitutively expressed genes, the early and late transcripts initiate at different positions (Moss, 1990b). These data show that both B15R and B18R are actively transcribed during the vaccinia virus
replication cycle from positions indicating that the first codon of each ORF is likely to be used as the translation initiation site. The transcription of the genes at different times of infection suggests that the gene products will have different functions.
Identification of Proteins Encoded by B15R and B18R
Vaccinia virus recombinants overexpressing the proteins or lacking the coding regions were constructed to identify the gene products and to study the biological activity of the proteins. Overexpression of the proteins was achieved by cloning a second copy of B15R or B18R, transcribed under the control of the late 4b promoter, in the thymidine kinase (TK) locus. The genomic structure of the recombinant viruses, called vB15R and vB18R, was confirmed by Southern blot hybridization (Figure 5A) and by polymerase chain reaction (PCR) using oligonucleotides specific for the 5' and 3' ends of the TK gene (data not shown). Deletion of 72% of B15R or 92% of B18R from the viral genome was carried out by transient dominant selection (Falkner and Moss, 1990; Isaacs et al., 1990). This method allows construction of a virus that only differs from the wild type in the deleted sequence and does not contain any selectable marker that could affect the new phenotype. The genomic structure of "the deletion mutants, named v B15R and v B18R, was confirmed by
Southern blot hybridization (Figure 5B). The isolation of deletion mutants for B15R or B18R that grow normally in tissue culture confirmed that both genes are
dispensable for virus replication in vitro (Perkus et al., 1991; Ueda et al., 1990).
To identify the gene products, labelling experiments with 35S-labelled methionine and cysteine in the presence (early) or absence (late) of an inhibitor of DNA
synthesis (cytosine arabinoside) were performed in cells infected with different vaccinia virus recombinants.
Extracts from cells or culture supematants were
immunoprecipitated and analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Specific antiserum against B15R immunoprecipitated a broad band of 50-60 kd in supematants from WR-infected cells at late times of infection (Figure 6A), confirming the transcriptional analysis. An intermediate
glycosylated form of 47 kd was detected in infected cells. As observed by immunoprecipitation (Figure 6A) and in the whole extract (data not shown), the expression of the protein either was increased or was not detected in vB15R- or v B15R-infected cells, respectively. The presence of tunicamycin inhibited the secretion,
indicating that a correct glycosylation and/or folding of the protein is required for this process, and reduced the size to 35 kd, close to the predicted molecular weight of 36,500. The other hands detected in the presence of tunicamycin were not specific for B15R since they were also immunoprecipitated by an antiserum against B18R (data not shown . Thus, a high degree of glycosylation accounts for 30%-40% of the size of the secreted protein encoded by B15R.
The protein encoded by B18R was detected in vaccinia virus-infected cells when overexpressed at late times of infection under the strong 4b promoter but was not detected in the deletion mutant (Figure 6B). Two forms of the protein (52kd and 60-65kd) were detected in cell extracts, and only the 60-65 kd protein, presumably containing a higher degree of glycosylation, was secreted to the medium. Since translation of B18R-specific mRNA in rabbit reticuloyte lysates produces a protein of the predicted size of 40 kd (Ueda et al., 1990), the
carbohydrate component of the secreted B18R gene product accounts for 33%-38% of the size of the protein.
Previous results have identified a 40 kd polypeptide immunoprecipitated from infected cells as the early surface antigen of vaccinia (Ikuta et al., 1980), later shown to be encoded by B18R (Ueda at al., 1990). The use of an antiserum against early cell surface antigens of vaccinia virus, and not antibodies specific for the B18R gene product, might explain the discrepancy.
Alternatively, the antiserum raised against B18R
expressed in insect cells might not recognise that form of the protein.
Expression of B15R and B18R in Baculovirus
To characterize further the products of genes B15R and B18R and to produce greater amounts of material for functional analysis, the proteins were expressed in
Spodoptera frugiperda (Sf) insect cells infected with Autographa califomica nuclear polyhedrosis virus (AcNPV) under the control of the polyhedrin promoter. The recombinant viruses constructed were termed AcB15R and AcB18R.
As shown in pulse-label experiments (Figure 6C), both B15R and B18R proteins were secreted to the medium by insect cells as 40-44 kd and 48 kd polypeptides, respectively. An incomplete glycosylation of the
polypeptides in insect cells (Luckow and Summers, 1988) might explain the lower size of the proteins when
compared with the vaccinia virus expression. The 45 kd protein in AcB15R-infected insect cell extracts might correspond to a glycosylated form with a signal sequence still bound to the polypeptide, possibly owing to the inability of insect cells to process properly the high amount of B15R protein expressed under the strong
polyhedrin promoter. Interestingly, two B18R proteins of 40 kd and 52 kd were detected in insect cell extracts, the smaller size corresponding to that predicted from the amino acid sequence and found in in vitro translation of specific mRNA (Ueda et al., 1990). This also correlates with two polypeptides in cells infected with vaccinia virus (vB18R), although the sizes are different, probably owing to a different posttranslational processing of the polypeptide in insect cells. Time course experiments revealed that the expression levels of both recombinant proteins reached maximum between 1 and 2 days after infection, and pulse-chase experiments showed that the proteins were stable in the medium during at least 20 hr (data not shown). IL-1 Binding Activity Expressed by Vaccinia Virus and Recombinant Baculoviruses
Since the sequences of B15R and B18R are related to IL-1 and IL-6 receptors, the presence of binding activity to human recombinant IL-1α, IL-1β, or IL-6 in detergent-solubilized cell extracts and medium from vaccinia virusor recombinant baculovirus-infected cells was examined in a solid phase binding assay (Figure 7A). Binding of radioiodinated IL-1α or IL-6 to vaccinia virus or
baculovirus was not detected while these ligands bound to EL46.lC10 and U266 cells, which overexpress IL-1 and IL-6 receptors, respectively. IL-1β binding activity was clearly found in vaccinia virus-infected cells and supematants harvested at 24 hr after infection and in the baculovirus recombinant expressing B15R 3 days after infection. The low binding to EL4 6.1C10 cells at this dose of 125I-IL-1β probably reflects a 6-fold lower affinity for the β form compared with IL-1α, described for the type I receptor expressed in this cell line (Sims et al., 1988). These results were corroborated in a more quantitative binding assay in solution, which
differentiates bound from free ligand by precfipitating the ligand-receptor complex with polyethylene glycol (Figure 7B). The finding of a higher binding activity for IL-1β in the supematants compared with the cell extracts, despite using material from 5-fold more cells, is in agreement with B15R being secreted from the cell. The cell-associated binding activity may correspond to protein present in the secretory pathway or in the plasma membrane. The first possibility was supported by
comparing 125I-IL-1β binding with detergent-solubilised cell extracts (Figure 7B) and intact cells in suspension, which showed that only 6%-9% of the cell-associated binding activity is detected on the cell surface (data not shown).
The kinetics of production of soluble IL-1β receptor from vaccinia virus-infected cells was examined by soluble binding assay and showed that no IL-1β receptor was detected above the background attributable to virus inoculum, in the presence of cytosine arabinoside. In contrast, in the absence of the drug, IL-1β accumulated in the supernatant and reached 80% of total by 24 hr (data not shown). These data are consistent with the transcriptional and polypeptide analyses and show that the IL-1β receptor is expressed late during infection.
Figure 8 shows the binding of radioiodinated IL-1β to medium from different recombinants using 1 × 104 cell equivalents, conditions that allowed a better
quantitation of the binding activity. The fact that the 125I-IL-1β binding increased in the vaccinia recombinant containing two copies of B15R (vB15R) and was absent in the mutant containing a deleted version of the gene (vB15R) demonstrates that B15R is the only gene product from vaccinia virus responsible for the IL-1β binding activity. In agreement with this, overexpression (vB18R) or deletion (v B18R) of B18R, a secreted and structurally related protein, in vaccinia virus did not affect the binding activity. The result obtained with medium from insect cells infected with AcB15R clearly reflects the high expression level of the baculovirus system.
Binding Properties of the Vaccinia IL-1β Receptor
The specific binding of only IL-1β to B15R
demonstrated a novel specificity compared with other cloned IL-1 receptors. In view of this, the applicants wanted to exclude the possibility that radioiodination of IL-1α or IL-6 has prevented their binding to the vaccinia receptor. This was determined by assaying the ability of unlabelled ILs, including IL-IRA, to compete with 125I-IL-1β for the binding to the vaccinia IL-1 receptor. As shown in Figure 9A, the interaction of radioiodinated IL-1β with the soluble receptor present in vaccinia virus supematants was competed in a dose-dependent manner by unlabelled IL-1β but not by IL-1α or IL-6. The natural competitor IL-1RA did not block the binding of labelled IL-1β to vaccinia IL-1 receptor, even when added at higher concentrations that are required to compete the binding of 125I-IL-1 to the type II IL-1 receptor on polymorphonuclear leukocytes or a pre-B lymphocyte line (Dripps et al., 1991; Granowitz et al., 1991; Mclntyre et al., 1991). As a control, the doses of unlabelled ILs used competed the binding of the corresponding .
radioiodinated IL to its natural receptor on EL4 6.1 C10 or U266 cells (data not shown). The receptor expressed in the baculovirus system showed similar properties. The binding of 100pM of 125I-IL-1β to 8 × 103 cell
equivalents of medium from AcB15R-infected cells (6610 cpm) was 94.1%, 7.4%, 99.0% and 110.1% in the presence of 10nM of IL-1α, 10nM of IL-1β, 100nM of IL-1ra, and 10nM of IL-6, respectively.
Scatchard analysis of binding of 125I-IL-1β to soluble receptor secreted from vaccinia-infected cells or insect cells infected with AcB15R was performed to estimate the affinity and the number of receptors (Figure 9B). High affinity binding sites for IL-1β were detected in supematants from WR-infected cultures, with a
dissociation constant ( KD) of 234 ± 49 pM. The estimated number of binding sites secreted after 24h per WR-infected cell was 1.1 ± 0.1 × 105. This is
extraordinarily high considering that the number of receptors in IL-1 responsive primary cultures or cell lines varies from <100 receptors per cell to a maximum of 1 × 104 receptors per cell (Dower and Urdal, 1987).
Cultures infected with vB15R secreted 3.8 ± 0.4 × 105 binding sites per cell after 24 hr with a similar
affinity ( KD 226 ± 35 pM ). In spite of a different glycosylation, the receptor secreted from infected Sf cells showed a similar affinity for IL-1β(KD 117 ± 10 pM). The number of receptors produced in insect cells after 3 days of infection (6.0 ± 0.2 × 106 sites per cell) reflects the high level of expression of the baculovirus system.
Competition of IL Binding to Cells
The specificity for IL-1β was also tested in
competition experiments of binding of labelled cytokines to their natural receptors on cell lines overexpressing IL-1 and IL-6 receptors. As shown in Figure 10, the binding of labelled IL-1α to EL4 6.1 C10 cells (A) and of IL-6 to U266 cells (C) was not competed by medium from baculovirus-infected insect cells expressing B15R or
B18R, while the binding was competed by the corresponding unlabelled ILs. In contrast, the interaction of IL-1β with EL4 6.1 C10 cells was specifically blocked by supematants containing B15R and not by B18R (Figure 10B). The percentage of competition correlated with the amount of 125I-IL-1β bound to different doses of receptor in solution (data not shown). Supematants from WR-infected cells, and not from v B15R-infected cells, competed the binding of 125I-IL-1β to cells in culture, while they did not affect binding of IL-1α to cells in culture, while they did not affect binding of 125 I-IL-1β to cells in culture, while they did not affect the binding of IL-1α or IL-6 (data not shown). Altogether, these results provide independent evidence that B15R binds specifically IL-1β and also show that the vaccinia IL-1 receptor is capable of competing with cells for IL-1β binding. This indicates that B15R might function as an inhibitor of the biological activities mediated by IL-1β.
Pathogenicity of Vaccinia Virus Lacking B15R or B18R in Mice
(1) Deletion of B15R had no effect on the growth of vaccinia virus in tissue culture. However, since the expression of an IL-1β binding activity might interfere with the inflammatory and/or immune responses in vivo, the pathogenicity of the deletion mutant v B15R was compared with that of the parent virus in a mouse model. The intranasal inoculation of the WR strain of vaccinia virus in mice produces an extensive respiratory infection followed by viraemia that leads to infection of the central nervous system and death of the animals (Turner, 1967; Williamson et al., 1990).
Figure 11A shows that, although there were not significant differences in the final number of
mortalities following infection with different doses of WR or v B15R, 70% of the mortalities on v B15R-infected animals occurred 1 day sooner than control (panels f). This unexpected and enhanced pathogenicity of v B15R was also demonstrated by the clearly accelerated onset of symptoms with doses of virus from 105 to 3 × 107 plaque-forming units (pfu) (Figure 11A, panels a-d). On day 5 after infection with v B15R, all 20 animals showed clear disease symptoms (ruffled fur, arched backs, and reduced mobility), while none of the comparable WR-infected animals did so (Figure 11A, panels f). The early onset of symptoms in v B15R-infected animals was very clear at a dose of 105 pfu (Figure 11A, panels d), where they appeared 2 or 3 days sooner than in WR-infected animals. In contrast, only the WR-infected animals developed symptoms at a low dose of virus (104 pfu; Figure 11A, panels e), suggesting a possible effect of B15R of the progression of the infection.
A second experiment was performed in which the symptoms of illness were quantified by measuring the weight of the animals, since in this animal model these symptoms parallel development of cachexia (Moore and Smith, 1992). As shown in Figures 11B and 12, an early onset of symptoms was again observed in animals infected with v B15R, and this correlated with accelerated weight loss. The apparent attenuation of v B15R compared with WR at 104pfu was not confirmed in this experiment; in contrast, it corroborated that v B15R induced earlier and, according to the weight, more severe symptoms of illness.
The earlier appearance of symptoms and weight loss probably reflect systemic effects mediated by the IL-1β induced in response to the infection that were
neutralized by an active secretion of B15R by the wild-type virus. The accelerated onset of symptoms and mortality indicated that the secreted IL-1β receptor can moderate the severity of the infection. Table 2 and Figure 14 show the results for the in vivo experiment in which mice were intranasally infected with doses of WR, v B15R or v B18R. The results show that groups of mice infected with a vaccinia virus unable to produce the B18R gene product had a lower incidence of mortality than equivalent groups of mice infected with either wild-type vaccinia virus or a vaccinia virus unable to produce the B15R gene product. In Figure 14, mortality and symptoms of illness are presented-as a function of days post-inoculation. The incidence of mortality was similar for groups of mice infected with either wild-type vaccinia virus or a vaccinia virus unable to produce the B15R gene product. However, symptoms of illness appeared sooner in groups of mice infected with vaccinia virus unable to produce the B15R gene product as compared to equivalent groups of mice infected with the wild-type vaccinia virus.
These data shown In Table 2 indicate that deletion of the B18R gene causes a lower mortality rate than deletion of the B15R gene. Therefore this discovery supports the provision of attenuates vaccinia virus by deletion of part or all of the B18R or by mutation of the B18R gene. Since removal of the B18R gene attenuates vaccinia virus and as discussed herein, the B18R gene product does not bind the cytokines discussed, the B18R gene product may be interfering with the hosts defense mechanisms in some other way.
IL-1 Binding Activity in Other Orthopoxviruses
The IL-1α, IL-1β, and IL-6 binding activity was investigated in a soluble receptor binding assay on supematants from cultures infected with different strains of vaccinia virus (Copenhagen, IHD-J, IHD-W, Wyeth, Lister, Tian-Tan, and Tashkent) and the related orthopoxviruses rabbitpox and cowpox compared with the WR strain. The binding to labelled murine IL-1β (mIL-1β) was also investigated to confirm that B15R is able to sequester IL-1β in infected mice. No binding to human 125I-IL-1α or 125I-IL-6 was detected (data not shown). However, binding to labelled IL-1β and mIL-1β was found in all viruses except for rabbitpox, Tashkent and
Copenhagen strains (Figure 13). The failure of
Copenhagen to express an IL-1 receptor is in agreement with sequencing data that showed a nonsense mutation at codon 31 of the ORF (Goebel et al., 1990). Even if translation reinitiated from the next methionine codon, which seems unlikely given its distance from the mRNA 5' end, the protein would lack a signal peptide and..
therefore would not be secreted but would probably be degraded within the cytoplasm. Binding experiments of labelled IL-1β to detergent extracts from cells infected with the different viruses gave similar results to those obtained with medium (data not shown), indicating that no intracellular receptor is produced in rabbitpox,
Tashkent, or Copenhagen strains. Interestingly, the ratios of binding of murine versus human IL-1β varied from 1.6 for the Wyeth strain to 10 for the Tian-Tan strain (Figure 13). The specific binding of 125I-mIL-1β to medium from AcB15R-infected cells indicates that the B15R protein is also responsible for binding mlL-1 (data not shown). The specific binding for IL-1β in other vaccinia virus strains and cowpox and the inability of IL-IRA to compete this binding (data not shown) indicate that the receptor encoded by other orthopoxviruses possesses similar binding properties.
Temperature Regulation by B15R
The results shown in Figures 15 and 16 demonstrate the ability of the B15R protein to regulate the
temperature of the infected animal. In Figure 15 the
B15R protein is shown to suppress the temperature of the infected mouse over the first six days of infection.
Animals infected with the wild-type ( WT ) virus (B15R +ve ) have a reduced temperature compared with those infected with the deletion mutant (B15R -ve) and re-insertion of the B15R gene into the deletion mutant virus restored the temperature profile of the infected animals to that typical of WT virus. In Figure 16 the applicants have compared the temperatures of mice infected with different strains of vaccinia virus, which had been characterised previously with respect to the expression or non-expression of the IL-1β receptor (Alcami and Smith, Cell 71, 153-167,
1992). These data show that mice infected with the Tian-Tan strain, which expresses the IL-1β receptor, have reduced temperatures following infection, while those animals infected with strains lacking the receptor
(Copenhagen and Tashkent) develop fever.
These data support the original proposals that the IL-1β receptor is functional in vivo and specifically show that it is IL-1β not IL-1α which is the predominant cytokine regulating the temperature of the infected animal.
Discussion
Two vaccinia virus ORFs, B15R and B18R, that encode proteins of the immunoglobulin superfamily related to the extracellular domains of the IL-1 and IL-6 receptors have been characterized. Both ORFs are transcribed, but at different phases of the virus replication cycle,
suggesting functional differences. The gene products are glycosylated and secreted from infected cells, in
agreement with the absence of transmembrane anchor sequences that are present in the cellular IL-1 and IL-6 receptors. The dispensability for virus replication in tissue culture, the secretion to the extracellular space, and the predicted receptor-like structure of the proteins suggest that both are involved in interference with host defense mechanisms in vivo.
The B18R gene product Is shown not to bind IL-1α, IL-1β or IL-6, despite the homology with the receptors for these cytokines (McMahan et al, 1991; Smith and Chan, 1991). Two forms of the protein (52 kd and 60-65 kd) were detected, the larger of which is found in
supematants while the smaller might represent a
membrane-associated molecule. This would be in agreement with previous reports showing immunofluorescence in the plasma membrane of infected cells using antiserum
specific for proteins secreted at early times of
infection (Ueda et al, 1972), which was attributed later to reactivity against B18R (Ueda et al, 1990).
In contrast with B18R, B15R ORF is shown to encode an IL-1β binding activity present in the supematants of vaccinia virus-infected cells and to represent a novel soluble IL-1 receptor. The high affinity for IL-1β binding (KD234 pM) is similar to those reported for the cellular receptors (Sims et al, 1988, 1989; McMahan et al, 1991) and is consistent with the retention of full binding activity by the extracellular domain of the IL-1 receptor (Dower et al, 1989). The size (50-60 kd ) and high carbohydrate content of the mature vaccinia IL-1β receptor are in agreement with those reported for the truncated and complete versions of the cellular receptor, respectively (Urdal et al, 1988; Dower et al, 1989). The secretion of a biologically active 40-44 kd protein from insect cells suggests that the carbohydrate is not an essential component for the IL-1 binding.
The vaccinia IL-1 receptor constitutes a novel receptor for IL-1 because of the specificity, for IL-1β. This was shown in binding experiments to radioiodinated ILs and was corroborated in competition assays with unlabeled cytokines and by blocking the interaction of the ILs with the natural receptor on cells in culture. A cellular receptor for IL-1β that is present in the membrane and secreted from Raji cells, which has a molecular weight similar to the type II receptor, has been described but not cloned (Benjamin et al, 1990; Giri et al, 1990; Symons and Duff, 1990; Symons et al, 1991). Since B15R has a higher similarity to the cellular type II receptor than to the type I receptor (McMahan et al, 1991; Smith and Chan, 1991) and since this similarity is comparable with those found between other vaccinia virus proteins and their cellular counterparts (Smith et al, 1991a), B15R may derive from the type II IL-1 receptor or a variant thereof. Unfortunately, the comparison of the sequence of the vaccinia IL-1β receptor with the type I and type II IL-1 receptors does not permit identification of the amino acids that confer specificity for IL-1β since the
sequences are quite divergent. However, the availability of the vaccinia virus gene will allow mutagenesis studies to identify these positions. Furthermore, the sequence of B15R ORF in other vaccinia virus strains that show different affinities for the human and murine IEr-1β may provide structural information on the binding domain. In this case, the comparison of the sequence may be more useful since genes from different orthopoxviruses are highly conserved.
The vaccinia IL-1β receptor might be useful as a tool to investigate the function of IL-1α and IL-1β in vivo in different models. In contrast, the other IL-1 inhibitors available (IL-IRA, a soluble truncated IL-1 receptor, and monoclonal antibodies against the receptor) block the binding of both forms of IL-1 (Fanslow et al, 1990; Gershenwald et al, 1990; Ohlsson et al, 1990;
Alexander et al, 1991; Mclntyre et al, 1991). As a therapeutic agent, the molecule may regulate responses normally controlled by IL-1β, and, since it does not bind IL-1α or IL-1RA, it might offer advantages over the other inhibitors. The failure of the vaccinia IL-1β receptor to bind IL-1RA illustrates the adaptation of the virus to the physiological response of the host by preventing interference with the natural antagonist.
The number of IL-1β binding sites secreted from vaccinia virus-infected cells ( about 105 receptors per cell 24 hr after infection) is without precedent and makes the supematants from cultures infected with vaccinia virus the most concentrated naturally occurring soluble IL-1 binding activity. An excess of soluble receptors must be required to block the effects of IL-1β in vivo, since only a few cellular IL-1 receptors need to be occupied to elicit a biological response. This was illustrated in the competition of IL-1β binding to T- cells, which also indicates that the vaccinia IL-1β receptor will probably block the biological effects induced in cells expressing IL-1 receptors.
The blockade of IL-1 by a virus is interesting since this cytokine orchestrates the host response to
infection, inducing a broad spectrum of systemic effects and playing an important role in initiating the
inflammatory and immune responses. But more interesting is the specificity of the blockade for IL-1β. To date, both IL-1α and IL-1β have been found to induce similar activities in a number of model systems (Dinarello, 1989). The fact that vaccinia virus secretes a protein that specifically blocks the effects of IL-1β indicates that soluble IL-1β plays a more important role than
IL-1α in the host response to orthopoxvirus infections.
The deletion of B15R ORF from the WR strain of vaccinia virus does not greatly affect virulence in intransally infected BALB/c mice, in which virus
virulence is defined according to the number of
mortalities. However, two observations revealed that the vaccinia IL-1β receptor does play an important role in vaccinia virus infection in vivo. First, the animals infected with v B15R developed symptoms and lost weight more rapidly than the corresponding control group. The early onset of symptoms is very likely to represent systemic effects induced by circulating IL-1β produced in response to vaccinia virus infection. IL-1 is known to function as a hormone mediating multiple effects such as fever, headache, and sleep and at high doses can induce hypotension and a shocklike state (Dinarello, 1988,
1989). The vaccinia IL-1β receptor, expressed in the wild-type virus, may thus limit the systemic acute phase response otherwise initiated by increased levels of IL-1β. The finding that weight loss, which can be induced by IL-1 (Di Giovine and Duff, 1990), occurred earlier in animals infected with v B15R supports this view. A generalized response can contribute to host defense; for example, temperature typical of fever has been reported to enhance the proliferation of T-cells that might facilitate a T-cell-dependent immune response (Duff and Durum. 1983). However, an increased systemic reaction to infection did not affect the outcome of infection by v B15R. Second, although the absolute number of
mortalities are indistinguishable, 70% of them occurred 1 day sooner in animals inoculated with the deletion mutant. This would be consistent with the vaccinia IL-1β receptor reducing the pathological effects mediated by excessive IL-1β production that are detrimental to the host (Dinarello, 1988, 1989) and thus moderating the severity of the disease.
The consequence of secretion of a specific inhibitor of IL-1β in the progression of vaccinia virus infection may be dual, since IL-1β triggers local and systemic effects necessary for an efficient host responnse to infection, but also contributes to the pathological process. Therefore, B15R might function as a virulence or attenuation factor for the virus. It is unclear whether the effects of deleting B15R from the WR strain of vaccinia virus, which was selected for high
neurovirulence by passage in mouse brain, are
representative of infections with other orthopoxviruses that also express the IL-1β binding activity. Other animal models for localized and systemic orthopoxvirus infections (Buller and Palumbo, 1991) may illustrate the different roles of B15R in infection. The importance of the route of virus inoculation for virus pathogenesis is illustrated by the study of Spriggs et al. (Cell 71, 145-152, 1992) in which a WR-based B15R deletion mutant had a 100-fold increase in lethal dose 50 compared with WR when administered by intracranial injection.
Interestingly, another orthopoxvirus mechanism to Inhibit specifically IL-1β action and to diminish the inflammatory response has just been reported (Ray et al, 1992). In this case, the protein encoded by the cowpox virus crmA gene (related to serine protease inhibitors) was shown to inhibit the IL-1β converting enzyme, which cleaves pro-IL-1β to generate active IL-1β. The
intracellular location and the kinetics of synthesis of this protein indicate that this mechanism is restricted to infected cells during early phases of virus
replication. The secretion of an abundant IL-1β receptor at late times of vaccinia virus infection, which we show here is also active in cowpox virus, is an ideal
complement to the inhibitory role of the crmA protein, since it is effective extracellularly and would-bind IL-1β released by both infected and uninfected cells recruited to the site of infection. However, with vaccinia virus the systemic response induced by IL-1β in infected mice seems mainly to be controlled by the soluble receptor, since the ORF corresponding to the crmA gene (B15R) is still present in the deletion mutant v B15R that induces more severe symptoms of disease.
B15R ORF is one of a few virus genes that has been shown to increase the pathogenicity or the severity of the infection when deleted from the virus genome
(Ginsberg et al, 1989; Romanczuk and Howley, 1992).
Virus attenuation can result from deletion or
inactivation of genes encoding proteins that interfere with host defense mechanisms, but as shown here, some virus proteins might also be devised to diminish effects that the infection produces in the host. This would help host survival and thereby be beneficial for the virus. Consistent with this view, in a 21.8 kb region of the genome of the highly pathogenic Harvey strain of variola major virus, 7 out of 32 ORFs are disrupted into small fragments, and 2 ORFs are partially or totally deleted, compared with vaccinia virus ( Aguado et al, 1992).
Another interesting correlation is that the vaccine strains of vaccinia virus that gave higher frequencies of postvaccinial complications (Copenhagen, Tashkent, and Tian-Tan; Fenner et al, 1988) fail to express the IL-1β binding activity or, as in Tian-Tin, recognize human IL-1β poorly. It is tempting to speculate that active expression of B15R might be to some extent responsible for the lower degree of pathogenicity of the other vaccine strains (Lister and Wyeth).
The reduction of systemic effects possibly attribute to IL-1 by neutralization of the IL-1β activity by vaccinia virus presented in this report suggests that the β form of IL-1, and not IL-1α, is mediating the
endocrine, long range effects in the host in response to vaccinia virus infection. This view is supported by previous observations. It has been reported that IL-1β is the predominant form of IL-1 secreted from human monocytes (Hazuda et al, 1988), and the release of adrenocorticotropic hormone, one of the neuroendocrine actions of IL-1, is exclusively induced by IL-1β (Uehara et al, 1987). Similarly, IL-1β is more potent than IL-1α in the induction of fever, and the effect is mediated through different mechanisms (Busbridge et al, 1989), which correlates with the discovery of IL-1β (Breder et al, 1988) and receptors specific for IL-1β (Katsuura et al, 1988) in the brain.
B15R is the second soluble cytokine receptor to be identified in a virus. A soluble receptor for tumor necrosis factor (TNF) has been shown to be active in Leporipoxviruses and to increase the pathogenicity of the virus (Smith et al, 1991b; Upton et al, 1991). The WR and Copenhagen strains of vaccinia virus contain one and two homologs, respectively, of the TNF receptor, but the presence of frameshifts and stop codons make expression of active proteins unlikely (Howard et al, 1991; Upton et al, 1991). The presence of soluble receptors for either TNF or IL-1 in different genera of poxviruses, which produce very different pattens of disease, is interesting since these cytokines share many biological properties. The soluble IL-1β receptor is one of the increasing number of activities encoded by vaccinia virus that aid evasion from the host immune system (for references see Moore and Smith, 1992) and, in particular, is another viral-encoded protein that interferes with cytokine functions. Besides the TNF receptor of leporipoxvirus and the crmA protein of cowpox virus, other examples found are the 14.7 kd protein of adenovirus that inhibits cytolysis by TNF (Gooding et al, 1988), the IL-10
activity encoded by Epstein-Barr virus (Hse et al, 1990), and the presence of IL-6 binding sites in the envelope protein of hepatitis B virus (Neurath et al, 1992).
In summary, the vaccinia IL-1β receptor may be a useful tool to discriminate the physiological roles of IL-1α and IL-1β and might be used as an anti-inflammatory therapeutic reagent. The expression of this activity by vaccinia virus and other orthopoxviruses represents a novel mechanism of virus evasion from the immune system. It is shown that the IL-1β receptor is modulating the systemic response to infection and the severity of the disease, which suggests that IL-1β, and not IL-1α, is the main mediator of the endocrine effects of the IL-1 produced in response to vaccinia virus infection in mice.
Aguado, B., Selmes, I. P., and Smith G.L. (1992).
Nucleotide sequence of 21.8 kbp of variola major virus strain Harvey and comparison with vaccinia virus.
J.Gen.Virol. 73, in press.
Alexander, H.R. Doherty, G.M., Buresh, C.M., Venson, D.J., and Norton, J.A. (1991). A recombinant human receptor antagonist to interleukin 1 improves survival after lethal endotoxemia in mice. J.Exp.Med. 173, 1029-1032.
Benjamin, D., Wormsley, S., and Dower, S.K. (1990). Heterogeneity in interleukin (IL)-1 receptors expressed on human B cell lines. J.Biol.Chem. 265, 9943-9951.
Breder, C.D., Dinarello, C.A., and Saper, C.B.
(1988). Interleukin-1 immunoreactive innervation of the human hypothalamus. Science 240, 321-324.
Brown, M., and Faulkner, P. (1977). A plaque assay for nuclear polyhedrosis using a solid overlay.
J.Gen.Virol. 36, 361-364.
Buller, R.M. and Palumbo, G.J. (1991). Poxvirus pathogenesis. Microbiol. Rev. 55, 80-122.
Busbridge, N.J., Dascombe, M.J., Tilders, F.J.H., Van Oers, J.W.A.M. Linton, E.A., and Rothwell, N.J.
(1989). Central activation of thermogenesis and fever by interleukin-1α and interleukin-1β involves different mechanisms. Biochem. Biophys. Res. Commun. 162, 591-596.
Carter, D.B., Deibel, M.R. Jr., Dunn, C.J., Tomich, C.-S.C., Laborde, A.L., Slightom, J.L. Berger, A.E.
Bienkowski, M.J., Sun, F.F., McEwan, R.N. Harris, P.K.W., Yem, A.W., Waszak, G.A., Chosay, J.G., Sieu, L.C.,
Hardee, M.M., Zurcher-Neely, H.A., Reardon, I.M.,
Heinrikson, R.L., Truesdell, S.E., Shelly, J.A., Eessalu, T.E., Taylor, B.M., and Tracey, D.E. (1990).
Purification, cloning, expression and biological
characterization of an interleukin-1 receptor antagonist protein. Nature 344, 633-638.
Di Giovine, F.S., and Duff, G.W. (1990). Interleukin 1: the first interleukin. Immunol. Today 11, 13-20.
Dinarello, C.A. (1988). Biology of interleukin 1. FASEB J.2, 108-115.
Dinarello, C.A. (1989). Interleukin-1 and its biologically related cytokines. Adv. Immunol. 44, 153-205.
Dinarello, C.A., and Thompson, R.C. (1991). Blocking IL-1: interleukin 1 receptor antagonist in vivo and in vitro. Immunol. Today 12, 404-410.
Dower, S.K., and Urdal, D.L. (1987). The
interleukin-1 receptor. Immunol. Today 8, 46-51.
Dower, S.K., Kronheim, S.R., March, C.J. Conlon, P.J., Hopp, T.P., Gillis, S., and Urdal, D.L. (1985). Detection and characterization of high affinity plasma membrane receptors for human interleukin 1. J.Exp.Med. 162, 501-515.
Dower, S.K., Wignall, J.M. Schooley, K., McMahan, C.J., Jackson, J.L., Prickett, K.S., Lupton, S., Cosman, D., and Sims, J.E. (1989). Retention of ligand binding activity by the extracellular domain of the IL-1
receptor. J. Immunol. 142, 4314-4320.
Dripps, D.J., Verderber, E., Ng, R.K., Thompson, R.C., and Eisenberg, S.P. (1991). Interleukin-1 receptor antagonist binds to the type II interleukin-1 receptor on B cells and neutrophils. J. Biol. Chem. 266, 20311-20315.
Duff, G.W., and Durum, S.K. (1983). The pyrogenic and mitogenic actions of interleukin-1 are related.
Nature 304, 449-451.
Eisenberg, S.P., Evans, R.J., Arend, W.P.,
Verderber, E., Brewer, M.T., Hannum, C.H., and Thompson, R.C. (1990). Primary structure and functional expression from complementary DNA of a human interleukin-1 receptor antagonist. Nature 343, 341-346.
Esposito, J.R., Condit, R.C., and Obijeski, J.
(1981). The preparation of orthopoxvirus DNA.
J.Virol. Meth. 2, 175-179.
Falkner, F.G., and Moss, B, (1990). Transient dominant selection of recombinant vaccinia viruses.
J.Virol. 64, 3108-3111.
Fanslow, W.C., Sims, J.E., Sassenfeld, H.,
Morrissey, P.J., Gillis, S., Dower, S.K., and Widmer, M.B. (1990). Regulation of alloreactivity in vivo by a soluble form of the interleukin-1 receptor. Science 248, 739-742. Fenner, F., Henderson, D.A. Arita, I., Jezek, Z., and Ladnyi, I.D. (1988). Smallpox and Its Eradication (Geneva: World Health Organization).
Gershenwald, J.E., Fong, Y., Fahey, T.J., III, Calvano, S.E., Chizzonite, R., Kilian, P.L., Lowry, S.F., and Moldawer, L.L. (1990). Interleukin 1 receptor
blockade attenuates the host inflammatory response.
Proc.Natl.Acad. Sci. USA 87, 4966-4970.
Ginsberg, H.S., Lundholm-Beauchamp, U., Horswood, R.L., Pernis, B., Wold, W.S., Chanock, R.M., and Prince, G.A. (1989). Role of early region 3 (E3) in pathogenesis of adenovirus disease. Proc.Natl.Acad. Sci. USA 86, 3823-3827.
Giri, J.G., Newton, R.C., and Horuk, R. (1990).
Identification of soluble interleukin-1 binding protein in cell-free supematants: evidence for soluble
interleukin-1 receptor. J.Biol.Chem. 265, 17416-17419.
Goebel, S.J., Johnson, G.P., Perkus, M.E., Davis, S.W., Winslow, J.P., and Paoletti, E. (1990). The
complete DNA sequence of vaccinia virus. Virology 179, 247-266.
Gooding, L.R., Elmore, L.W., Tollefson, A.E. Brady, H.A., and Wold, W.S.M. (1988). A 14,700 MW prptein from the E3 region of adenovirus inhibits cytolysis by tumor necrosis factor. Cell 53, 341-346.
Granowitz, E.V., Clark, B.D., Mancilla, J., and Dinarello, C.A. (1991). Interleukin-1 receptor antagonist competitively inhibits the binding of interleukin-1 to the type II interleukin-1 receptor. J. Biol. Chem. 266, 14147-14150.
Hannum, C.H., Wilcox, C.J., Arend, W.P., Joslin, F.G., Dripps, D.J., Heimdal, P.L., Armes, L.G., Sommer, A., Eisenberg, S.P., and Thompson, R.C., (1990).
Interleukin-1 receptor antagonist activity of a human interleukin-1 inhibitor. Nature 343, 336-340.
Harlow, E., and Lane, D. (1988). Antibodies: A Laboratory Manual (Cold Spring Harbor, New York: Cold Spring Harbor Laboratory Press).
Hazuda, D.J., Lee, J.C., and Young, P.R. (1988). The kinetics of interleukin 1 secretion from activated monocytes: differences between interleukin 1α and
interleukin 1β. J.Biol. Chem. 263, 8473-8479.
Howard, S.T., Chan, Y.S., and Smith, G.L. (1991). Vaccinia virus homologues of the Shope fibroma virus inverted terminal repeat proteins and a discontinuous ORF related to the tumor necrosis factor receptor family.
Virology 180, 633-647.
Hsu, D.-H., De Waal Malefyt, R., Fiorentino, D.F., Dang, M.N., Vieira, P., De Vries, J., Spits, H. Mosmann, T.R., and Moore K.W. (1990). Expression of interleukin-10 activity by Epstein-Barr virus protein BCRFl . Science 250, 830-832.
Hughes, S.J., Johnston, L.H., De Carlos, A., and Smith, G.L. (1991). Vaccinia virus encodes an active thymidylate kinase that complements a cdc8 mutant of
Saccharomyces cerevisiae. J. Biol. Chem. 266, 20103-20109.
Ikuta, K., Miyamoto, H., and Kato, S. (1980).
Biochemical studies on early cell surface antigen induced by vaccinia and cowpox viruses. J.Gen.Virol. 47, 227-232.
Isaacs, S.N., Kotwal, G.J., and Moss, B. (1990).
Reverse guanine phosphoribosyltransferase selection of recombinant vaccinia viruses. Virology 178, 626-630.
Katsuura, G., Gottschall, P.E., and Arimura, A.
(1988). Identification of a high-affinity receptor for interleukin-1 beta in rat brain. Biochem. Biophys. Res.
Commun. 156, 61-67.
Kent, R.K. (1988). Isolation and analysis of the vaccinia virus p4b gene promoter. PhD thesis, University of Cambridge, Cambridge, England.
Laemmli, U.K. (1970). Cleavage of structural
proteins during the assembly of the head of bacteriophage T4. Nature 227, 680-685.
Larrick, J.W. (1989). Native interleukin 1
inhibitors. Immunol. Today 10, 61-66.
Luckbw, V.A., and Summers, M.D. (1988). Trends in the development of baculovirus expression vectors.
Biotechnology 6, 47-55.
MacDonald, H.R., Lees, R.K., and Bron, C. (1985). Cell surface glycoproteins involved in the stimulation of interleukin 1-dependent interleukin 2 production by a subline of EL4 thymoma cells. J. Immunol. 135, 3944-3950.
Mackett, M., Smith, G.L., and Moss, B. (1985). The construction and characterization of vaccinia virus recombinants expressing foreign genes. In DNA cloning: A Practical Approach, Vol.2, D.M. Glover, ed. (Oxford: IRL Press), pp.191-211.
Mclntyre, K.W., Stepan, G.J., Kolinsky, K.D.,
Benjamin, W.R., Plocinski, J.M., Kaffka, K.L., Campen, C.A., Chizzonite, R.A., and Kilian, P.L., (1991).
Inhibition of interleukin 1 (IL-1) binding and
bioactivity in vitro and modulation of acute inflammation in vivo by IL-1 receptor antagonist and anti-IL-1
receptor monoclonal antibody. J.Exp.Med. 173, 931-939.
McMahan, C.J., Slack, J.L., Mosley, B., Cosman, D., Lupton, S.D., Brunton, L.L., Grubin, C.E., Wignall, J.M., Jenkins, N.A., Brannan, C.I., Copeland, N.G., Huebner, K., Croce, C.M., Cannizzarro, L.A., Benjamin, D., Dower, S.K., Spriggs, M.K., and Simms, J.E. (1991). A novel IL-1 receptor, cloned from B cells by mammalian expression, is expressed in many cell types. EMBO J. 10, 2821-2832.
Moore, J.B., and Smith, G.L. (1992). Steroid hormone synthesis by a vaccinia enzyme: a new type of virus virulence factor. EMBO J. 11, 1973-1980.
Moss, B. (1990a). Poxviridae and their replication. In Virology, Second Edition, B.N. Fields, D.M. Knipe, R.M. Chanock, M.S. Hirsch, J. Melnick, T.P. Monath, and B. Roizman, eds. (New York: Raven), pp.2079-2111.
Moss, B. (1990b). Regulation of vaccinia virus transcription. Annu. Rev.Biochem. 59, 661-688.
Munson, P.J., and Rodbard, D. (1980). LIGAND: a versatile computerized approach for characterization of ligand-binding systems. Anal. Biochem. 107, 220-239.
Neurath, A.R., Strick, N., and Sproul, P. (1992). Search for hepatitis B virus cell receptors reveals binding sites for interleukin 6 on the virus envelope protein. J.Exp.Med. 175, 461-469.
Ohlsson, K., Bjork, P., Bergenfeldt, M., Hageman, R., and Thompson, R.C. (1990). Interleukin-1 receptor antagonist reduces mortality from endotoxin shock. Nature 348, 550-552. Perkus, M.E., Goebel, S.J., Davis, S.W., Johnson, G.P., Norton, E.K., and Paoletti, E. (1991). Deletion of fifty five open reading frames from the termini of vaccinia virus. Virology 180, 406-410.
Ray, C.A., Black, R.A., Kronheim, S.R., Greenstreet, T.A., Sleath, P.R., Salvesen, G.S., and Pickup, D.J.
(1992). Viral inhibition of inflammation: cowpox virus encodes an inhibitor of the interleukin-1β converting enzyme. Cell 69, 597-604.
Romanczuk, H., and Howley, P.M. (1992). Disruption of either the E1 or the E2 regulatory gene of human papillomavirus type 16 increases viral immortalization capacity. Proc.Natl.Acad. Sci. USA 89, 3159-3163.
Sambrook, J., Fritsch, E.F., and Maniatis, T.
(1989). Molecular Cloning: A Laboratory Manual, Second Edition (Cold Spring Harbor, New York: Cold Spring Harbor Laboratory Press).
Shields, J., and Mazzei, G.J. (1991). Natural interleukin-1 inhibitors. Clin.Biotech. 3, 81-87.
Sims, J.E., March, C.J., Cosman, D., Widmer, M.B., MacDonald, H.R., McMahan, C.J., Grubin, C.E., Wignall, J.M., Jackson, J.L., Call, S.M., Friend, D., Alpert, A.R., Gillis, S., Urdal, D.L., and Dower, S.K. (1988). cDNA expression cloning of the IL-1 receptor, a member of the immunoglobulin superfamily. Science 241, 585-589.
Sims, J.E., Acres, R.B., Grubin, C.E., McMahan, C.J., Wignall, J.M., March, C.J., and Dower, S.K. (1989). Cloning the interleukin-1 receptor from human T cells. Proc. Natl.Acad. Sci. USA 86, 8946-8950.
Smith, G.L., and Chan, Y.S. (1991). Two vaccinia virus proteins structurally related to the interleukin-1 receptor and the immunoglobulin superfamily. J. Gen. Virol. 72, 511-518.
Smith, G.L., Chan, Y.S., and Howard, S.T. (1991a). Nucleotide sequence of 42kbp of vaccinia virus strain WR from near the right inverted terminal repeat.
J. Gen. Virol. 72, 1349-1376.
Smith, C.A., Davis, T., Wignall, J.M., Din, W.S., Farrah, T., Upton, C., McFadden, G., and Goodwin, R.G. (1991b). T2 open reading frame from Shope fibroma virus encodes a soluble form of the TNF receptor. Biochem.
Biophys.Res.Commun. 176, 335-342.
Spriggs, M.K.., Hruby, D.E., Maliszewski, C.R.,
Pickup, D.J., Sims, J.E., Buller, R.M.L., and VanSlyke, J. (1992). Vaccinia and cowpox viruses encode a novel secreted interleukin-1-binding protein. Cell 71, this issue.
Symons, J.A., and Duff, G.W. (1990). A soluble form of the interleukin-1 receptor produced by a human B cell line. FEBS Lett. 272, 133-136.
Symons, J.A., Eastgate, J.A., and Duff, G.W. (1990). A soluble binding protein specific for interleukin 1β is produced by activated mononuclear cells. Cytokine 2, 190-198.
Symons, J.A., Eastgate, J.A., and Duff, G.W. (1991). Purification and characterization of a novel soluble receptor for interleukin 1. J.Exp.Med. 174, 1251-1254.
Taga, T., Kawanishi, Y., Hardy, R.R., Hirano, T., and Kishimoto, T. (1987). Receptors for the B cell stimulatory factor 2: quantitation, specificity,
distribution, and regulation of their expression.
J.Exp.Med. 166, 967-981.
Trakman, P. (1990). The enzymology of poxvirus DNA replication. Curr. Topics Microbiol. Immunol. 163, 93-123.
Turner, G.S. (1967). Respiratory infection of mice with vaccinia virus. J. Gen.Virol. 1, 399-402.
Turner, P.C., and Moyer, R.W. (1990). The molecular pathogenesis of poxviruses. Curr. Topics Microbiol.
Immunol. 163, 125-151.
Ueda, Y., Tagaya, I., Amano, H., and Ito, M. (1972). Studies on the early antigens induced by vaccinia virus. Virology 49, 794-800.
Ueda, Y., Morikawa, S., and Matsuura, Y. (1990). Identification and nucleotide sequence of the gene encoding a surface antigen induced by vaccinia virus. Virology 177, 588-594.
Uehara, A., Gottschall, P.E., Dahl, R.R., and
Arimura, A. (1987). Stimulation of ACTH release by human interleukin-1β, but not by interleukin-1α, in conscious, freely-moving rats. Biochem.Biophys .Res. Commun. 146, 1286-1290.
Upton, C., Macen, J.L., Schreiber, M., and McFadden, G. (1991). Myxoma virus expresses a secreted protein with homology to the tumor necrosis factor receptor gene family that contributes to viral virulence. Virology 184, 370-382.
Urdal, D.L., Call, S.M., Jackson, J.L., and Dower, S.K. (1988). Affinity purification and chemical analysis of the interleukin-1 receptor. J.Biol.Chem. 263, 2870-2877.
Vieira, J., and Messing, J. (1982). The pUC
plasmids, and M13mp7-derived system for insertion, mutagenesis and sequence with systemic universal primers. Gene 19, 259-268.
Vieira, J., and Messing, J. (1987). Production of single-stranded plasmid DNA. Meth. Enzymol. 153, 3-11.
Williamson, J.D., Reith, R.W., Jeffrey, L.J.,
Arrand, J.R., and Mackett, M. (1990). Biological
characterization of recombinant vaccinia viruses in mice infected by the respiratory route. J.Gen.Virol. 71, 2761-2767.
Zuidema, D., Schouten, A., Usmany, M., Maule, A.J., Belsham, G.J., Roosien, J., Klinge-Roode, E/C, Van Lent, J.W.M., and Vlak, J.M. (1990). Expression of cauliflower mosaic virus gene I in insect cells using a novel
polyhedrin-based baculovirus expression vector.
J. Gen.Virol. 71, 2201-2209.
Table 1
B15R B18R
1 2 3 1 2 3
B15R 1
2 6.71
3 1.63 2.44
B18R 1 4.81 1.88 0.58
2 4.13 2.75 2.25 2.70
3 1.94 2.64 3.51 3.67 -0.21
IL-1R Mouse 1 7.77 4.32 3.27 3.87 4.64 2.29
2 4.70 7.62 2.20 0.39 6.42 3.11
3 2.03 1.74 2.68 2.96 1.91 3.55
IL-1R Man 1 7.62 2.37 3.21 2.53 4.60 2.78
2 4.07 8.29 1.27 0.31 5.88 3.80
3 1.07 1.29 4.64 1.73 1.65 2.44
IL-6R Man 5.87 3.98 5.28 3.24 6.30 2.10
L1CAM 6 4.40 4.13 6.44 2.79 5.43 2.41
CHNCAM 1 4.12 4.01 4.07 3.19 5.54 3.28
MAG 3 4.04 4.67 6.17 1.22 3.10 3.96
PDGFR 3 2.84 2.81 5.14 3.88 4.86 3.24
TCRCD3 5.65 4.95 1.75 2.38 4.64 2.66
LAR 2 5.33 6.52 4.46 3.26 5.32 2.07
CEA 1 3.42 3.58 3.18 1.24 6.33 4.07
RPIgR 5 3.92 4.47 4.35 1.67 1.80 3.81 Table 2
DOSE (PFU) WR vΔB15R vΔB18R
7
3×10 5/5 4/5 3/5
107 4/5 5/5 0/5
106 5/5 5/5 1/5
105 1/5 2/5 0/5
104 1/5 0/5 0/4
NB 4/5 = 4 deaths in a group of 5
4-6 week old Balb/c mice were infected intranasally with indicated doses of WR, B15R-deleted (vΔβ15ft) or B188-del (vΔB18F)virus. The mortality of animals after 15 days is show
Table 1. Similarity scores for the Ig domains of vaccinia virus proteins B15R and B18R against selected domains from other Ig superfamily members computed using the ALIGN programme (Dayhoff, M.O., Barker, W.C. and Hunt, L.T. (1983). Meth. Enzymol. 91, 524-545). The programme compares to the best alignment score for two domains with the mean score of 100 alignments of the randomly scrambled sequences. The score for the best alignment of the real sequences is presented as the number of standard deviations from the mean score of the randomised sequences. Values of greater than 3.1 are significant (probability 10-3), while values of 4.8, 6.0 and 7.9 indicate probabilities of 10-6, 10-9 and 10-15, respectively. The domains illustrated are from B15R amino acids 28-119 (1), 121-214 (2), 222-end (3): B18R 53-149 (1), 152-241 (2), 252-end (3); murine IL-1R precursor (Sims, J.E., March, C.J., Widmer, M.B., MacDonald, H.R., McMahan, D.J., Grubin, C.E., Wignall, J.M., Jackson, J.L., Call, S.M. Friend, D., Alpert, A.R. Gillis, S., Urdal, D.L. and Dower, S.K. (1988). Science 241, 585-589) 26-119 (1), 125-219 (2), 231-335 (3); human IL-1R (Sims, J.E., Acres, R.B., Grubin, C.E., McMahan, C.J., Wignall, J.M. (1989). Proc Natl. Acad. Sci USA 86, 8946-8950) 24-116 (1), 122-216 (2), 228-332 (3): human IL-6R (Yamasaki, K., Taga, T., Hirat, Y., Yawata, H., Kawanishi, Y., Seed, B., Taniguchi, T., Hirano, T and Kishismoto, T. (1988). Proc. Jpn. Acad. 64, 209-211) 27-116; murine neural cell adhesion molecule L1 precursor (L1CAM) (Moos, M., Tacke, R., Scherer, H., Teplow, D., Freuth, K., Schachner, M. (1988). Nature 334, 701-703) 518-610 (6); chicken neural cell adhesion molecule (CHNCAM) ( Hemperley, J. J., Murray, B.A., Edelman, G.M. and Cunningham, B.A. (1986). Proc. Natl. Acad. Sci. USA 83, 3037-3041) 2-97; myelin-associated glycoprotein (MAG) (Salzer, J.L., Holmes, W.P. and Colman, D.R. (1987). J. Cell Biol. 104, 957-965) 225-309; platelet derived growth factor receptor (PDGFR) (Yarden, Y., Escobedo, J.A., Kuang, W.J., Yang-Feng, T.L., Daniel, T.O., Tremble, P.M., Chen, E.Y., Ando, M.E., Harkins, R.N., Francke, U., Friend, V.A., Ullrich, A. and Williams L.T. (1986). Nature 323, 226-232) 183-279; T cell receptor CD3 epsilon chain (TCRCD3) (Clevers, H., Duiilap, S., Saito, H., Georgopoulos, K., Wileman, T. and Terhorst, C., (1988). Proc. Natl. Acad. Sci. USA 85, 8623-8627) 1-82; leukocyte antigen receptor protein (LAR) (Streuli, M., Krueger, N.X., Hall, L.R., Schlossman, S.F. and Saito, H. (1988). J. Exp. Med. 168, 1553-1562) 125-216; carcinoembryonic antigen precursor (CEA) (Oikawa, S., Nakazato, H. and Kosaki, G. (1987). Biochem. Biophys. Res. Commun. 142, 511-518) 113-201; rabbit poly-Ig receptor (RPIgR) (Mostov, K.E., Friedlander, M. and Blobel, G. (1984). Nature 308, 37-43) 458-558.

Claims

CLAIMS :
1. Use of the nucleotide sequence designated herein as B15R, or of a nucleotide sequence coding for an allele of, or functionally equivalent fragment,
derivative or variant of the polypeptide encoded by the B15R nucleotide sequence, in a process relating to the manufacture of a medicament for the treatment of a condition in which interleukin-1β is involved in the mediation of one or more symptoms associated with the condition.
2. Use of a polypeptide encoded by the nucleotide sequence designated herein as B15R, or of an allele of, or functionally equivalent fragment, derivative or variant of the polypeptide, to manufacture a medicament for the treatment of a condition in which
interleukin-1β is involved in the mediation of one or more symptoms associated with the condition.
3. Use according to claim 1 or claim 2 wherein the condition is selected from the group consisting of:
inflammation, rheumatoid arthritis, septic shock, cancer, fever; inflammatory bowl disease; graft versus host disease and diabetes.
4. Use according to claim 3 wherein the condition is fever.
5. A pharmaceutical which is an anti-fever medicament comprising a polypeptide which is encoded by the nucleotide sequence designated herein as B15R, or an allele of, or functionally equivalent fragment,
derivative or variant of said polypeptide.
6. A diagnostic reagent for the detection or measurement of interleukin-1β in a sample which reagent comprises a polypeptide encoded by the nucleotide sequence designated herein as B15R, or of an allele of, or functionally equivalent fragment, derivative or variant of the polypeptide.
7. A diagnostic kit which comprises a diagnostic reagent according to claim 6 and one or more ancillary kit components for making the detection or measurement of interleukin-1β.
8. A method of diagnosing a patient's condition in which interleukin-1β is involved in the mediation of one or more symptoms associated with the condition, which method comprises testing a sample of biological material obtained from the patient using a diagnostic reagent according to claim 6 or a diagnostic kit according to claim 7.
9. A method of treating a patient suffering from a condition in which interleukin-1β is involved in the mediation of one or more symptoms associated with the condition which method comprises administering a
medicament prepared according to claim 1 or claim 2.
10. A method of treating a patient for fever which comprises administering a medicament prepared according to claim 3 or a pharmaceutical according to claim 5.
PCT/GB1993/000460 1992-03-05 1993-03-05 Vaccinia virus b15r used in interleukin b1-involving condition WO1993018153A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU36418/93A AU3641893A (en) 1992-03-05 1993-03-05 Vaccinia virus b15r used in interleukin b1-involving condition
EP93905517A EP0656949A1 (en) 1992-03-05 1993-03-05 Vaccinia virus b15r used in interleukin b1-involving condition

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB929204780A GB9204780D0 (en) 1992-03-05 1992-03-05 Vaccinia vectors,vaccinia genes,and expression products thereof;pharmaceuticals,reagents and methods
GB9204780.2 1992-03-05

Publications (1)

Publication Number Publication Date
WO1993018153A1 true WO1993018153A1 (en) 1993-09-16

Family

ID=10711551

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB1993/000460 WO1993018153A1 (en) 1992-03-05 1993-03-05 Vaccinia virus b15r used in interleukin b1-involving condition

Country Status (3)

Country Link
EP (1) EP0656949A1 (en)
GB (1) GB9204780D0 (en)
WO (1) WO1993018153A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005014622A2 (en) * 2003-08-07 2005-02-17 F. Hoffmann-La Roche Ag Ra antigenic peptides
WO2005030971A1 (en) * 2003-09-29 2005-04-07 Gsf-Forschungszentrum Fuer Umwelt Und Gesundheit Gmbh Modified vaccinia virus ankara (mva) mutant and use thereof
WO2020011754A1 (en) * 2018-07-09 2020-01-16 Transgene Chimeric vaccinia viruses

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992007944A1 (en) * 1990-10-26 1992-05-14 Lynxvale Limited Vaccinia vectors, vaccinia genes and expression products thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992007944A1 (en) * 1990-10-26 1992-05-14 Lynxvale Limited Vaccinia vectors, vaccinia genes and expression products thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CELL vol. 71, no. 1, October 1992, CAMBRIDGE, NA US pages 153 - 167 ALCAMI, A. ET SMITH, G.L. 'A soluble receptor for interleukin-1-beta encoded by vaccinia virus a novel mechnism of virus modulation of the host response to infection' *
JOURNAL OF GENERAL VIROLOGY vol. 72, no. 3, March 1991, GB pages 511 - 518 SMITH, G.L. ET CHAN, Y. S. 'Two vaccinia virus proteins structurally related to the interleukin-1 receptor and the immunoglobulin superfamily' cited in the application *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005014622A2 (en) * 2003-08-07 2005-02-17 F. Hoffmann-La Roche Ag Ra antigenic peptides
WO2005014622A3 (en) * 2003-08-07 2005-05-06 Hoffmann La Roche Ra antigenic peptides
KR100769338B1 (en) * 2003-08-07 2007-10-24 에프. 호프만-라 로슈 아게 Ra antigenic peptides
WO2005030971A1 (en) * 2003-09-29 2005-04-07 Gsf-Forschungszentrum Fuer Umwelt Und Gesundheit Gmbh Modified vaccinia virus ankara (mva) mutant and use thereof
US7767209B2 (en) 2003-09-29 2010-08-03 Gsf-Forschungszentrum Fuer Umwelt Und Gesundheit Gmbh Modified vaccinia virus Ankara (MVA) mutant and use thereof
CN1842602B (en) * 2003-09-29 2011-06-29 德国慕尼黑亥姆霍兹研究中心健康和环境有限公司 Modified vaccinia virus ankara (MVA) mutant and use thereof
WO2020011754A1 (en) * 2018-07-09 2020-01-16 Transgene Chimeric vaccinia viruses

Also Published As

Publication number Publication date
GB9204780D0 (en) 1992-04-15
EP0656949A1 (en) 1995-06-14

Similar Documents

Publication Publication Date Title
Alcami et al. A soluble receptor for interleukin-1β encoded by vaccinia virus: a novel mechanism of virus modulation of the host response to infection
Upton et al. Myxoma virus expresses a secreted protein with homology to the tumor necrosis factor receptor gene family that contributes to viral virulence
Massung et al. Analysis of the complete genome of smallpox variola major virus strain Bangladesh-1975
US6998252B1 (en) Recombinant poxviruses having foreign DNA expressed under the control of poxvirus regulatory sequences
Whitford et al. Identification and sequence analysis of a gene encoding gp67, an abundant envelope glycoprotein of the baculovirus Autographa californica nuclear polyhedrosis virus
Guo et al. Expression in recombinant vaccinia virus of the equine herpesvirus 1 gene encoding glycoprotein gp13 and protection of immunized animals
Duncan et al. Identification and characterization of an extracellular envelope glycoprotein affecting vaccinia virus egress
US7384643B2 (en) Recombinant poxvirus
AU595190B2 (en) A transcriptional regulatory sequence from the 5&#39;flanking region of the vaccinia virus
Kettle et al. Vaccinia virus serpins B13R (SPI-2) and B22R (SPI-1) encode Mr 38.5 and 40K, intracellular polypeptides that do not affect virus virulence in a murine intranasal model
US5420026A (en) Self-assembling replication defective hybrid virus particles
Takahashi-Nishimaki et al. Regulation of plaque size and host range by a vaccinia virus gene related to complement system proteins
Barry et al. The myxoma virus M-T4 gene encodes a novel RDEL-containing protein that is retained within the endoplasmic reticulum and is important for the productive infection of lymphocytes
PT752888E (en) Nucleotide and amino acid sequences of canine herpesvirus gb, gc and gd and uses therefor
JP2007082551A (en) Recombinant poxvirus-calicivirus [rabbit hemorrhagic disease virus (rhdv)] composition and use thereof
WO1992007944A1 (en) Vaccinia vectors, vaccinia genes and expression products thereof
US5476781A (en) Entomopoxvirus spheroidin gene sequences
WO1990012101A1 (en) Vaccinia vectors, vaccinia genes and expression products thereof
Rodriguez et al. Insertional inactivation of the vaccinia virus 32-kilodalton gene is associated with attenuation in mice and reduction of viral gene expression in polarized epithelial cells
Smith et al. Host range selection of vaccinia recombinants containing insertions of foreign genes into non-coding sequences
Gong et al. A single point mutation of Ala-25 to Asp in the 14,000-Mr envelope protein of vaccinia virus induces a size change that leads to the small plaque size phenotype of the virus
Calvert et al. Identification and functional analysis of the fowlpox virus homolog of the vaccinia virus p37K major envelope antigen gene
WO1993018153A1 (en) Vaccinia virus b15r used in interleukin b1-involving condition
Strayer et al. Sequence and analysis of a portion of the genomes of Shope fibroma virus and malignant rabbit fibroma virus that is important for viral replication in lymphocytes
US6933145B1 (en) Materials and methods for delivery and expression of heterologous DNA in vertebrate cells

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AT AU BB BG BR CA CH CZ DE DK ES FI GB HU JP KP KR LK LU MG MN MW NL NO NZ PL PT RO RU SD SE SK UA US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 1993905517

Country of ref document: EP

ENP Entry into the national phase

Ref country code: US

Ref document number: 1994 295707

Date of ref document: 19941107

Kind code of ref document: A

Format of ref document f/p: F

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1993905517

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1993905517

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: CA