WO1999019480A2 - Human receptor proteins; related reagents and methods - Google Patents

Human receptor proteins; related reagents and methods Download PDF

Info

Publication number
WO1999019480A2
WO1999019480A2 PCT/US1998/020939 US9820939W WO9919480A2 WO 1999019480 A2 WO1999019480 A2 WO 1999019480A2 US 9820939 W US9820939 W US 9820939W WO 9919480 A2 WO9919480 A2 WO 9919480A2
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
sequence
primate
seq
cell
Prior art date
Application number
PCT/US1998/020939
Other languages
French (fr)
Other versions
WO1999019480A3 (en
Inventor
Jacqueline C. Timans
Johannes Eduard Maria Antonius Debets
Theodore R. Sana
J. Fernando Bazan
Robert A. Kastelein
Original Assignee
Schering Corporation
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 Schering Corporation filed Critical Schering Corporation
Priority to EP98952080A priority Critical patent/EP1029043A1/en
Priority to JP2000516032A priority patent/JP2004512801A/en
Priority to AU97864/98A priority patent/AU9786498A/en
Priority to CA002306455A priority patent/CA2306455A1/en
Publication of WO1999019480A2 publication Critical patent/WO1999019480A2/en
Publication of WO1999019480A3 publication Critical patent/WO1999019480A3/en

Links

Classifications

    • 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
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7155Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates to compositions and methods for affecting mammalian physiology, including, e.g., morphogenesis or immune system function.
  • it provides nucleic acids, proteins, and antibodies, e.g., which regulate development and/or the immune system along with related reagents and methods . Diagnostic and therapeutic uses of these materials are also disclosed.
  • Recombinant DNA technology refers generally to techniques of integrating genetic information from a donor source into vectors for subsequent processing, such as through introduction into a host, whereby the transferred genetic information is copied and/or expressed in the new environment.
  • the genetic information exists in the form of complementary DNA
  • cDNA derived from messenger RNA (mRNA) coding for a desired polypeptide product.
  • the carrier is frequently a plasmid having the capacity to incorporate cDNA for later replication and/or expression in a host and, in some cases, actually to control expression of the cDNA and thereby direct synthesis of the encoded product in the host.
  • Lymphokines apparently mediate cellular activities in a variety of ways . They have been shown to support the proliferation, growth, and/or differentiation of pluripotential hematopoietic stem cells into vast numbers of progenitors comprising diverse cellular lineages which make up a complex immune system. Proper and balanced interactions between the cellular components are necessary for a healthy immune response. The different cellular lineages often respond in a different manner when lymphokines are administered in conjunction with other agents .
  • B-cells which can produce and secrete immunoglobulins (proteins with the capability of recognizing and binding to foreign matter to effect its removal)
  • T-cells of various subsets that secrete lymphokines and induce or suppress the B-cells and various other cells (including other T- cells) making up the immune network.
  • B-cells can produce and secrete immunoglobulins (proteins with the capability of recognizing and binding to foreign matter to effect its removal)
  • T-cells of various subsets that secrete lymphokines and induce or suppress the B-cells and various other cells (including other T- cells) making up the immune network.
  • mast cell which has not been positively identified in all mammalian species
  • mast cell is a granule-containing connective tissue cell located proximal to capillaries throughout the body. These cells are found in especially high concentrations in the lungs, skin, and gastrointestinal and genitourinary tracts.
  • Mast cells play a central role in allergy-related disorders, particularly anaphylaxis as follows: when selected antigens crosslink one class of immunoglobulins bound to receptors on the mast cell surface, the mast cell degranulates and releases mediators, e.g., histamine, serotonin, heparin, and prostaglandins, which cause allergic reactions, e.g., anaphylaxis.
  • the interleukin-1 family of proteins includes the IL-l ⁇ , the IL-l ⁇ , the IL-1RA, and recently the IL-l ⁇ (also designated Interferon-Gamma Inducing Factor, IGIF) .
  • This related family of genes has been implicated in a broad range of biological functions . See Dinarello (1994) FASEB J. 8:1314-1325; Dinarello (1991) Blood 77:1627-1652; and Okamura, et al . (1995) Nature 378:88- 91.
  • lymphokines soluble proteins and their receptors
  • a number of degenerative or abnormal conditions directly or indirectly involve development, differentiation, or function, e.g., of the immune system and/or hematopoietic cells.
  • novel receptors for lymphokine-like molecules which enhance or potentiate the beneficial activities of other lymphokines, would be highly advantageous .
  • the present invention provides new receptors for ligands exhibiting similarity to interleukin-1 like compositions and related compounds, and methods for their use.
  • the present invention is directed to novel receptors related to IL-1 receptors and their biological activities. These receptors, e.g., primate or rodent, are designated IL-1 receptor like molecular structures, IL-1 Receptor DNAX designation 8(IL-1RD8), IL-1 Receptor DNAX designation 9(IL-1RD9) and IL-1 Receptor DNAX designation 10 (IL-IRDIO) .
  • the invention includes nucleic acids coding for the polypeptides themselves and methods for their production and use.
  • the nucleic acids of the invention are characterized, in part, by their homology to cloned complementary DNA (cDNA) sequences enclosed herein.
  • the invention provi ⁇ Tes" a composition of matter selected from the group of: an isolated or recombinant IL-1RD8 polypeptide comprising a segment of at least 12 contiguous amino acids of SEQ ID NO: 2 or 4, a natural sequence IL-1RD8 polypeptide comprising SEQ ID NO: 2 or 4, a fusion protein comprising IL-1RD8 sequence; an isolated or recombinant IL-1RD9 polypeptide comprising at least 12 contiguous amino acids of SEQ ID NO: 6, 8, 10, 12, 14, or 16; a natural sequence IL-1RD9 comprising SEQ ID NO: 6, 8, 10, 12, 14, or 16; a fusion protein comprising IL-1RD9 sequence; an isolated or recombinant IL-lRDIO polypeptide comprising at least 12 contiguous amino acids of SEQ ID NO: 18 or 20; a natural sequence IL-1RD10 comprising SEQ ID NO: 18 or 20; and a fusion protein comprising IL-1RD10 sequence.
  • the recombinant or isolated polypeptide comprises a segment identical to a corresponding portion of an IL-1RD8, as described, wherein: the number of contiguous amino acid residues is: at least 17 amino acids; at least 21 amino acids; or at least 25 amino acids; or to a corresponding portion of an IL-1RD9, as described, wherein the number of identical contiguous amino acid residues is: at least 17 amino acids; at least 21 amino acids; or at least 25 amino acids; or of an IL-lRDIO, as described, wherein the number of identical contiguous amino acid residues is: at least 17 amino acids; at least 21 amino acids; or at least 25 amino acids.
  • the invention provides a composition of matter wherein the IL-1RD8 comprises a mature sequence shown in SEQ ID NO: 2 or 4; an IL-1RD9 that comprises a mature sequence shown in SEQ ID NO: 6, 8, 10, 12, 14 or 16; an IL-1RD10 that comprises a mature sequence shown in SEQ ID NO: 18 or 20; or the IL-1RD8, IL-1RD9, or IL-lRDIO polypeptide: is from a warm blooded animal, e.g., a primate, such as a human; comprises at least one polypeptide segment of SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18, or 20; exhibits a plurality of portions having segments identical to specific sequence identifiers; is a natural allelic variant of a primate IL-1RD8; a primate or rodent IL-1RD9; or a primate IL- 1RD10; has a length at least about 30 amino acids; exhibits at least two non-overlapping epitopes that are specific for
  • compositions comprising: a sterile IL-1RD8, IL-1RD9, or IL-1RD10 polypeptide; or the IL-1RD8, IL-1RD9, or IL-1RD10 polypeptide and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration; a sterile IL-1RD8, IL-1RD9, or IL-1RD10 polypeptide; or the IL-1RD8, IL-1RD9, or IL-1RD10 polypeptide, as described, and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration.
  • the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or
  • kits comprising: mature polypeptide sequence shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 or 20; a detection or purification tag, including a FLAG, His6, or Ig sequence; or sequence of another receptor protein.
  • Kit embodiments include a kit comprising such a polypeptide, and: a compartment comprising the polypeptide; and/or instructions for use or disposal of reagents in the kit.
  • the invention provides a binding compound comprising an antigen binding site from an antibody, which specifically binds to a natural: IL-1RD8, IL-1RD9, or IL-1RD10 polypeptide, wherein: the polypeptide is a primate or rodent protein; the binding compound is an Fv, Fab, or Fab2 fragment; the binding compound is conjugated to another chemical moiety; or the antibody: is raised to a polypeptide sequence of a mature polypeptide comprising a sequence sequence shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 or 20; is raised to a mature primate or rodent IL- 1RD8; is raised to a purified human IL-1RD8; is raised to a purified mouse IL-1RD9; is immunoselected; is a polyclonal antibody; binds to a denatured IL-1RD8, IL- 1RD9, or IL-1RD10; exhibits a Kd to antigen of at least
  • IL-1RD9 protein is attached to a solid substrate, including a bead or plastic membrane; is in a sterile composition; or is detectably labeled, including a radioactive or fluorescent label; IL-1RD9 protein, wherein: the polypeptide is a primate or rodent protein; the binding compound is an Fv, Fab, or Fab2 fragment; the binding compound is conjugated to another chemical moiety; or the antibody: is raised against a polypeptide sequence of a mature polypeptide comprising a sequence sequence shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 or 20; is raised against a mature primate IL-1RD9; is raised to a purified human IL-1RD9 ; is immunoselected; is a polyclonal antibody; binds to a denatured IL-1RD9; exhibits a Kd to antigen of at least 30 ⁇ M; is attached to a solid substrate, including a bead or plastic membrane; is in a sterile composition; or is detectably
  • compositions comprising: a sterile binding compound, or the binding compound and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration.
  • Nucleic acid embodiments include an isolated or recombinant nucleic acid encoding a polypeptide or fusion protein, wherein: the IL-1RD8, IL-1RD9, or IL-lRDIO is from a mammal; said nucleic acid: encodes an antigenic polypeptide sequence sequence shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 or 20; encodes a plurality of antigenic polypeptide sequences sequence shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 or 20; exhibits at least about 30 nucleotides to a natural cDNA encoding the segment; is an expression vector; further comprises an origin of replication; is from a natural source; comprises a detectable label; comprises synthetic nucleotide sequence; is less than 6 kb, preferably less than 3 kb; is from a mammal, including a primate; comprises a natural full length coding sequence; is a hybridization probe for a gene encoding said IL-1RD8, IL- 1
  • the invention further provides a cell comprising such a recombinant nucleic acid, e.g., where the cell is: a prokaryotic cell; a eukaryotic cell; a bacterial cell; a yeast cell; an insect cell; a mammalian cell; a mouse cell; a primate cell; or a human cell.
  • Certain kit embodiments include a comprising the nucleic acid " and: a compartment comprising the nucleic acid; a compartment further comprising: a primate IL-1RD8, a primate or rodent IL-1RD9, or a primate IL-1RD10 polypeptide; and/or instructions for use or disposal of reagents in the kit.
  • the kit is capable of making a qualitative or quantitative analysis.
  • the nucleic acid is one which: hybridizes under wash conditions of 40° C and less than 2M salt to either SEQ ID NO: 1, 3, 5, 7, 9, 11, 13, 15, 17, or 19; or exhibits identity over a stretch of at least about 30 nucleotides to a primate IL- 1RD8, a primate or rodent IL-1RD9, or a primate IL-1RD10.
  • the wash conditions are: at 45° C and/or 500 mM salt; at 55° C and/or 150 mM salt; or the stretch is at least 55 nucleotides; or at least 75 nucleotides.
  • Methods of modulating physiology or development of a cell or tissue culture cells comprising contacting the cell with an agonist or antagonist of a primate IL-1RD8, a primate or rodent IL- 1RD9, or a primate IL-1RD10.
  • the cell is transformed with a nucleic acid encoding either IL-1RD8, IL-1RD9, or IL-1RD10, and another IL-1R.
  • the present invention provides the amino acid sequence and DNA sequence of mammalian, herein, e.g., primate and rodent IL-1 receptor-like molecules, these molecules IL-1 Receptor DNAX designation 8(IL-1RD8), IL-1 Receptor DNAX designation 9(IL-1RD9) and IL-1 Receptor .
  • DNAX designation 10(IL-1RD10) having particular defined properties, both structural and/or biological. These embodiments increase the number of members of the human IL-1 receptor-like family from 7 to at least 10. These receptors have been numbered internally as DNAX designations Dl, D2 , D3 , D4, D5, D6, and now D8, D9, and D10, and are referred to as IL-lRDl through D10.
  • cDNAs encoding these molecules were obtained from " primate, e.g., human, or rodent, e.g., mouse, cDNA sequence libraries. Other primate, rodent, or other mammalian counterparts would also be desired.
  • primate IL-1RD9 partial nucleotides (SEQ ID NO: 5) and corresponding amino acid sequences (SEQ ID NO: 6) of a primate IL-1RD9 coding segment are provided.
  • Supplemental primate IL-1RD9 is provided in SEQ ID NO: 7, 8, 9, and 10.
  • Rodent embodiments of IL- 1RD9 are provided in SEQ ID NO: 11, 12, with supplemental IL-1RD9 rodent sequence in SEQ ID NO: 13, 14, 15, and 16.
  • a partial nucleotide and corresponding partial amino acid sequence are provided in SEQ ID NO: 17 and 18, respectively, with supplemental human IL-1RD10 nucleotide and corresponding partial amino acid sequence provided in SEQ ID NO: 19 and 20, respectively.
  • GenBank D12763; hIL-lRD2 is GenBank X59770; mIL-lRD2 is GenBank
  • hIL-lRD5 is GenBank U43672
  • mI -lRD5 is GenBank U43673
  • mlL-lRDl is GenBank 20658, M29752
  • hI -lRDI is GenBank X16896
  • cIL-lRDl is GenBank 86325
  • hFGR4 is GenBank P22455.
  • Other species counterparts may be obtained from public sequence databases .
  • mI -lRD3 MGLL WY MSLSFYG ILQSHASERC DD LDTMR..
  • hI -lRD6 M WS LLCGLSI ALPLSVTADG CKDIFMKN..
  • rIL-lRD6 MG PPL FCWVSF V P FVAAGN CTDVYMH3H.. mIL-lRD4 MI DRQRMGL AL AILTLPMY T VTEGSKSS .. hIL-lRD4 MG FWILAI TI MYSTAAKFSK QS hIL-lRD2 MLRLYV LVMGVSAFTL QPAAHTGAAR SCRFRGRHYK mIL-lRD2 MFI LV VTG VSAFTTPTW HTGKVSESPI TSEKPTVHGD NCQFRGREFK hlL-lRDlO hI -lRD5 MNCRE LPLT WVLIS VSTAESCTSR PHITWE...
  • MFPVITNPPY hIL-lRD4 VMTEDAGDYT CKFIHNENGA NYSVTATRSF TVKDEQGFS .
  • LF V3t,GAPAQ hIL-lRD2 VALEDAGYYR CVLTFAHEGQ QYNITRSIEL RIKKK..KEE TIPVIISP..
  • mIL-lRD2 TSMDDAGYYR CVMTFTYNGQ EYNITRNIEL RVKGT..TTE
  • PIPVIISP.. hIL-lRDIO EFG... S CEL .. KYGGF V..VRRTTEL
  • TVTAPLTDKP PKLLYPMESK hIL-lRD5 ...
  • ...VSTAYII rIL-lRD6 QEGIETNLSL RNHILYTVNI TFLEVKMEDY GHPFTCHAA.
  • hIL-lRD9 is SEQ ID NO: 8; mIL-lRD9 is SEQ ID NO: 14; hIL-lRDI isTM GenBank
  • hIL-lRD6 is GenBank U49065
  • mIL-lRD3 is GenBank X85999
  • huIL-lRD8 is SEQ ID NO: 3
  • mIL-lRD4 is GenBank Y07519.
  • HuIL- -1RD1 GRDDYV.GED IVEVINENVK KSRRLIIILV RETSGFSWLG GSSEEQIAMY HuIL--1RD6 GRDEFP .
  • HuIL- -1RD1 NALVQDGIKV VLLELEKIQ DYEKM PESIKFIKQK HGAIRWSGDF HuIL--1RD6 SALIQDGMKV ILIELEKIE DYTVM PESIQYIKQK HGAIRWHGDF MoIL--1RD3 NMASRGNINV ILVQYKAVK. ...DMKVKEL KRAKTVLT .. ..VIKWKGEK HuIL--1RD8 NMLVSGEIKV ILIECTELKG KVNCQEVESL KRSIKLLS .. ..
  • HuIL- -1RD1 TQGPQSAKTR FWKNVRYHMP VQRRSPSSKH HuIL-•1RD6 TEQSQCMKTK FWKTVRYHMP PRRCRPFLRS MoIL--1RD3 SKYPQ ...GR FWKQLQVAMP VKKSPRWSSN HuIL-•1RD8 SSKLN.
  • SK FWKHLVYEMP IKKKEMLPRC HuIL--1RD5 SLSYN.
  • SR FWAKMRYHMP VKNSQGFTWN MoIL--1RD4 VADKQSLSSK FWKHVRYQMP VPERASKTAS
  • Ig domains there are characteristic Ig domains, and" " subdomains therein.
  • the corresponding regions of the IL- 1RD10 are about: f2 to gly7; g2 from vallO to thr23; a3 from leu30 to met33; a3 ' from thr38 to gln40; b3 from ala48 to ala54; c3 from pro64 to lys70; c3 ' from glu72 to phe74; d3 from val83 to lys92; e3 from gln98 to vall06; and f3 from tyr117 to trpl26.
  • the corresponding regions of the IL- 1RD9 are about: Igl domain from glyl ⁇ to prol27, with cysl05 probably linked to cys52 (or possibly cys48) ; Ig2 domain from glyl28 to pro229, with cysl53 probably linked to cysl99; and the Ig3 domain from glu230 to lys333, with cys251 probably linked to cys315; transmembrane segment from val336 to tyr360; THD domain from gly381 to val539; conserved trp residues probably correspond to residues 64, 169, and 267.
  • Alignment of the IL-1RD9 embodiments is shown in Table 4.
  • Table 4 There are characteristic beta strand sections, and alpha helical structures, as described above for IL- 1RD10.
  • the corresponding segments of the human IL-1RD9 sequence (SEQ ID NO: 6, 8, and 10) are roughly: ⁇ B from gly3 to val13; ⁇ 2 from prol5 to lys28; ⁇ c from ser30 to ser46; ⁇ 3 from ile47 to gln61; ⁇ D from lys64 to glu75; ⁇ 4 from glu77 to leu87; ⁇ E from val93 to leu98; and ⁇ 5 from argl06 to valll7.
  • mice IL-1RD9 sequence (SEQ ID NO: 12, 14, and 16) are roughly: ⁇ 3 to glnlO; ⁇ D from lys13 to glu24; ⁇ 4 from glu26 to leu36; ⁇ E from va42 to leu47; and ⁇ 5 from arg55 to val66.
  • IL-1 like receptor D8 IL- 1RD8
  • IL-1RD9 IL-1RD9
  • IL-1 like receptor D10 IL-lRDIO
  • the invention also includes a polypeptide variation" of the respective IL-1RD8, IL-1RD9, IL-1RD10 alleles whose sequences are provided, e.g., a mutein or soluble extracellular or intracellular construct.
  • such agonists or antagonists will exhibit less than about 10% sequence differences, and thus will often have between 1- and 11-fold substitutions, e.g., 2-, 3-, 5-, 7-fold, and others. It also encompasses allelic and other variants, e.g., natural polymorphic, of the polypeptide described. Typically, it will bind to its corresponding biological ligand, perhaps in a dimerized state with an alpha receptor subunit, with high affinity, e.g., at least about 100 nM, usually better than about 30 nM, preferably better than about 10 nM, and more preferably at better than about 3 nM.
  • the term shall also be used herein to refer to related naturally occurring forms, e.g., alleles, polymorphic variants, and metabolic variants of the mammalian protein.
  • This invention also encompasses polypeptides having substantial amino acid sequence identity with the amino acid sequences shown in SEQ ID NO: 2, 4, 6, 8, 10, 12,
  • IL-1RD4 forms in association with an alpha subunit, e.g., an IL-1RD4, IL-1RD5, or IL-1RD6.
  • a substantial polypeptide "fragment”, or “segment” is a stretch of amino acid residues of at least about 8 contiguous amino acids, generally at least 10 contiguous amino acids, more generally at least 12 contiguous amino acids, often at least 14 contiguous amino acids, more often at least 16 contiguous amino acids, typically at least 18 contiguous amino acids, more typically at least 20 contiguous amino acids, usually at least 22 contiguous amino acids, more usually at least 24 contiguous amino acids, preferably at least 26 contiguous amino acids, more preferably at least 28 contiguous amino acids," and, in particularly preferred embodiments, at least about 30 or more contiguous amino acids, usually 40, 50, 70, 90, 110, etc.
  • Sequences of segments of different polypeptides can be compared to one another over appropriate length stretches.
  • the matching will involve a plurality of distinct, e.g., nonoverlapping, segments of the specified length.
  • the plurality will be at least two, more usually at least three, and preferably 5, 7, or even more. While the length minima are provided, longer lengths, of various sizes, may be appropriate, e.g., one of length 7, and two of length 12. Similar features apply to segments of nucleic acid.
  • Amino acid sequence homology, or sequence identity is determined by optimizing residue matches, if necessary, by introducing gaps as required. See, e.g., Needleham, et al . (1970) J. Mol. Biol. 48:443-453; Sankoff, et al . (1983) chapter one in Time Warps , String Edits, and Macromolecules : The Theory and Practice of Sequence Comparison, Addison-Wesley, Reading, MA; and software packages from IntelliGenetics, Mountain View, CA; and the University of Wisconsin Genetics Computer Group (GCG) , Madison, WI; each of which is incorporated herein by reference. This changes when considering conservative substitutions as matches.
  • Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine.
  • Homologous amino acid sequences are intended to include natural allelic and interspecies variations in the cytokine sequence. Typical homologous polypeptides will have from 50-100% homology (if gaps can be introduced) , to 60-100% homology (if conservative substitutions are included) with an amino acid sequence segment shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 and 20.
  • Homology measures will be at least about 70%, generally at least 76%, more generally at least 81%, often at least 85%, more often at least 88%, typically at least 90%, more typically at least 92%, usually at least 94%, more usually at least 95%, preferably at least 96%, and more preferably at least 97%, and in particularly preferred embodiments, at least 98% or more.
  • the degree of homology will vary with the length of the compared segments.
  • Homologous polypeptides, such as the allelic variants, will share most biological activities with the embodiments described in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 and 20.
  • biological activity is used to describe, without limitation, effects on inflammatory responses, innate immunity, and/or morphogenic development by respective ligands .
  • these receptors should, like IL-1 receptors, mediate phosphatase or phosphorylase activities, which activities are easily measured by standard procedures. See, e.g., Hardie, et al. (eds. 1995) The Protein Kinase FactBook vols . I and II, Academic Press, San Diego, CA; Hanks, et al . (1991) Meth. Enzvmol . 200:38-62; Hunter, et al.
  • the receptors exhibit biological activities much like regulatable enzymes, regulated by ligand binding. However, the enzyme turnover number is more close to an enzyme than a receptor complex. Moreover, the numbers of occupied receptors necessary to induce such enzymatic activity is less than most receptor systems, and may number closer to dozens per cell, in contrast to most receptors which will trigger at numbers in the thousands per cell.
  • the receptors, or portions thereof, may be useful as phosphate labeling enzymes to label general or specific substrates.
  • the terms ligand, agonist, antagonist, and analog of, e.g., an IL-1RD8, IL-1RD9, or IL-lRDIO, include molecules that modulate the characteristic cellular responses to IL-1 ligand proteins, as well as molecules possessing the more standard structural binding competition features of ligand-receptor interactions, e.g., where the receptor is a natural receptor or an antibody.
  • the cellular responses likely are mediated through binding of various IL-1 ligands to cellular receptors related to, but possibly distinct from, the type I or type II IL-1 receptors. See, e.g., Belvin and Anderson (1996) Ann. Rev.
  • a ligand is a molecule which serves either as a natural ligand to which said receptor, or an analog thereof, binds, or a molecule which is a functional analog of the natural ligand.
  • the functional analog may be a ligand with structural modifications, or may be a wholly unrelated molecule which has a molecular shape which interacts with the appropriate ligand binding determinants.
  • the ligands may serve as agonists or antagonists, see, e.g., Goodman, et al . (eds. 1990) Goodman & Gilman's: The Pharmacological Bases of Therapeutics , Pergamon Press, New York.
  • Rational drug design may also be based upon structural studies of the molecular shapes of a receptor or antibody and other effectors or ligands. Effectors may be other proteins which mediate other functions in response to ligand binding, or other proteins which normally interact with the receptor.
  • One means for determining which sites interact with specific other proteins is a physical structure determination, e.g., x- ray crystallography or 2 dimensional NMR techniques . These will provide guidance as to which amino acid residues form molecular contact regions.
  • x- ray crystallography or 2 dimensional NMR techniques .
  • the IL-1 receptor-like polypeptides will have " a number of different biological activities, e.g., in phosphate metabolism, being added to or removed from specific substrates, typically proteins. Such will generally result in modulation of an inflammatory function, other innate immunity response, or a morphological effect.
  • a human IL-1RD9 gene coding sequence probably has about 60-80% identity with the nucleotide coding sequence of mouse IL-1RD9. At the amino acid level, there is also likely to be reasonable identity.
  • the receptors will also exhibit immunogenic activity, e.g., in being capable of eliciting a selective immune response.
  • Antiserum or antibodies resulting therefrom will exhibit both selectivity and affinity of binding.
  • the polypeptides will also be antigenic, in binding antibodies raised thereto, in the native state, or in denatured.
  • the biological activities of the IL-lRDs will generally be related to addition or removal of phosphate moieties to substrates, typically in a specific manner, but occasionally in a non specific manner. Substrates may be identified, or conditions for enzymatic activity may be assayed by standard methods, e.g., as described in Hardie, et al. (eds. 1995) The Protein Kinase FactBook vols. I and II, Academic Press, San Diego, CA; Hanks, et al. (1991) Meth. Enzvmol . 200:38-62; Hunter, et al. (1992) Cell 70:375-388; Lewin (1990) Cell 61:743-752; Pines, et al . (1991) Cold Spring Harbor Svmp. Quant. Biol. 56:449-463; and Parker, et al . (1993) Nature 363:736-738.
  • This invention contemplates use of isolated nucleic acid or fragments, e.g., which encode these or closely related proteins, or fragments thereof, e.g., to encode a corresponding polypeptide, preferably one which is biologically active.
  • this invention covers isolated or recombinant DNA which encodes such polypeptides or polypeptides having characteristic " sequences of the respective IL-lRDs, individually or as a group.
  • the nucleic acid is capable of hybridizing, under appropriate conditions, with a nucleic acid coding sequence segment shown in SEQ ID NO: 1, 3, 5, 7, 9, 11, 13, 15, 17 or 19 but preferably not with a ' corresponding segment of other receptors.
  • Said biologically active polypeptide can be a full length polypeptide, or fragment, and will typically have a segment of amino acid sequence highly homologous to one shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 and 20. Further, this invention covers the use of isolated or recombinant nucleic acid, or fragments thereof, which encode polypeptides having fragments which are equivalent to the IL-1RD9 proteins.
  • the isolated nucleic acids can have the respective regulatory sequences in the 5' and 3' flanks, e.g., promoters, enhancers, poly-A addition signals, and others from the natural gene.
  • an "isolated" nucleic acid is a nucleic acid, e.g., an RNA, DNA, or a mixed polymer, which is substantially pure, e.g., separated from other components which naturally accompany a native sequence, e.g., ribosomes, polymerases, and flanking genomic sequences from the originating species .
  • the term embraces a nucleic acid sequence which has been removed from its naturally occurring environment, and includes recombinant or cloned DNA isolates, which are thereby distinguishable from naturally occurring compositions, and chemically synthesized analogs or analogs biologically synthesized by heterologous systems.
  • a substantially pure molecule includes isolated forms of the molecule, either completely or substantially pure.
  • An isolated nucleic acid will generally be a homogeneous composition of molecules, but will, in some embodiments, contain heterogeneity, preferably minor. This heterogeneity is typically found at the polymer ends or portions not critical to a desired biological function or activity.
  • a "recombinant" nucleic acid is typically defined either by its method of production or its structure" In reference to its method of production, e.g., a product made by a process, the process is use of recombinant nucleic acid techniques, e.g., involving human intervention in the nucleotide sequence. Typically this intervention involves in vitro manipulation, although under certain circumstances it may involve more classical animal breeding techniques. Alternatively, it can be a nucleic acid made by generating a sequence comprising fusion of two fragments which are not naturally contiguous to each other, but is meant to exclude products of nature, e.g., naturally occurring mutants as found in their natural state.
  • nucleic acids comprising sequence derived using any synthetic oligonucleotide process .
  • Such a process is often done to replace a codon with a redundant codon encoding the same or a conservative amino acid, while typically introducing or removing a restriction enzyme sequence recognition site.
  • the process is performed to join together nucleic acid segments of desired functions to generate a single genetic entity comprising a desired combination of functions not found in the commonly available natural forms, e.g., encoding a fusion protein.
  • Restriction enzyme recognition sites are often the target of such artificial manipulations, but other site specific targets, e.g., promoters, DNA replication sites, regulation sequences, control sequences, or other useful features may be incorporated by design.
  • site specific targets e.g., promoters, DNA replication sites, regulation sequences, control sequences, or other useful features may be incorporated by design.
  • a similar concept is intended for a recombinant, e.g., fusion, polypeptide. This will include a dimeric repeat.
  • synthetic nucleic acids which, by genetic code redundancy, encode equivalent polypeptides to fragments of IL-1RD9 and fusions of sequences from various different related molecules, e.g., other IL-1 receptor family members.
  • a "fragment" in a nucleic acid context is a contiguous segment of at least about 17 contiguous nucleotides, generally at least 21 contiguous " " nucleotides, more generally at least 25 contiguous nucleotides, ordinarily at least 30 contiguous nucleotides, more ordinarily at least 35 contiguous nucleotides, often at least 39 contiguous nucleotides, more often at least 45 contiguous nucleotides, typically at least 50 contiguous nucleotides, more typically at least 55 contiguous nucleotides, usually at least 60 contiguous nucleotides, more usually at least 66 contiguous nucleotides, preferably at least 72 contiguous nucleotides, more preferably at least 79 contiguous nucleotides, and in particularly preferred embodiments will be at least 85 or more contiguous nucleotides, e.g., 100, 120, 140, etc.
  • fragments of different genetic sequences can be
  • a nucleic acid which codes for an IL-1RD8, IL-1RD9, or IL-1RD10 will be particularly useful to identify genes, mRNA, and cDNA species which code for itself or closely related proteins, as well as DNAs which code for polymorphic, allelic, or other genetic variants, e.g., from different individuals or related species.
  • Preferred probes for such screens are those regions of the interleukin which are conserved between different polymorphic variants or which contain nucleotides which lack specificity, and will preferably be full length or nearly so. In other situations, polymorphic variant specific sequences will be more useful.
  • This invention further covers recombinant nucleic acid molecules and fragments having a nucleic acid sequence identical to or highly homologous to the isolated DNA set forth herein.
  • the sequences will often be operably linked to DNA segments which control transcription, translation, and DNA replication. These additional segments typically assist in expression of the desired nucleic acid segment.
  • Homologous, or highly identical, nucleic acid sequences when compared to one another, e.g., IL-1RD9 sequences, exhibit significant similarity.
  • the standards for homology in nucleic acids are either measures for homology generally used in the art by sequence comparison or based upon hybridization conditions . Comparative hybridization conditions are described in greater detail below.
  • Substantial identity in the nucleic acid sequence comparison context means either that the segments, or their complementary strands, when compared, are identical when optimally aligned, with appropriate nucleotide insertions or deletions, in at least about 60% of the nucleotides, generally at least 66%, ordinarily at least 71%, often at least 76%, more often at least 80%, usually at least 84%, more usually at least 88%, typically at least 91%, more typically at least about 93%, preferably at least about 95%, more preferably at least about 96 to 98% or more, and in particular embodiments, as high at about 99% or more of the nucleotides, including, e.g., segments encoding structural domains such as the segments described below.
  • segment will hybridize under selective hybridization conditions, to a strand or its complement, typically using a sequence derived from SEQ ID NO: 1, 3, 5, 7, 9, 11, 13, 15, 17 or 19.
  • selective hybridization will occur when there is at least about 55% homology over a stretch of at least about 14 nucleotides, more typically at least about 65%, preferably at least about 75%, and more preferably at least about 90%. See, Kanehisa (1984) Nuc . Acids Res . 12:203-213, which is incorporated herein by reference.
  • the length of homology comparison may be over longer stretches, and in certain embodiments will be over a stretch of at least about 17 nucleotides, generally at least about 20 nucleotides, ordinarily at least about 24 nucleotides, usually at least about 28 nucleotides, typically at least about 32 nucleotides, more typically at least about 40 nucleotides, preferably at least about 50 nucleotides, and more preferably at least about 75 to 100 or more nucleotides.
  • Stringent conditions in referring to homology" in the hybridization context, will be stringent combined conditions of salt, temperature, organic solvents, and other parameters typically controlled in hybridization reactions .
  • Stringent temperature conditions will usually include temperatures in excess of about 30° C, more usually in excess of about 37 * C, typically in excess of about 45' C, more typically in excess of about 55" C, preferably in excess of about 65° C, and more preferably in excess of about 70° C.
  • Stringent salt conditions will ordinarily be less than about 500 mM, usually less than about 400 mM, more usually less than about 300 mM, typically less than about 200 mM, preferably less than about 100 mM, and more preferably less than about 80 mM, even down to less than about 20 mM.
  • the combination of parameters is much more important than the measure of any single parameter. See, e.g., Wet ur and Davidson (1968) J. Mol. Biol. 31:349-370, which is hereby incorporated herein by reference.
  • the signal should be at least 2X over background, generally at least 5-10X over background, and preferably even more.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are input into a computer, subsequent coordinates are designated, if necessary, and sequence algorithm program parameters are designated.
  • sequence comparison algorithm then calculates the percent sequence identity for the test sequence (s) relative to the reference sequence, based on the designated program parameters.
  • Optical alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1981) Adv. APPI . Math. 2:482, by the homology alignment algorithm of Needleman and Wunsch (1970) CT . Mol. Biol. 48:443, by the search for similarity method of Pearson and Lipman (1988) Proc . Nat ' 1 Acad. Sci . USA 85:2444, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Com uter Group, 575 Science Dr., Madison, WI) , or by visual inspection (see generally Ausubel et al., supra).
  • PILEUP creates a multiple sequence alignment from a group of related sequences using progressive, pairwise alignments to show relationship and percent sequence identity. It also plots a tree or dendrogram showing the clustering relationships used to create the alignment. PILEUP uses a simplification of the progressive alignment method of Feng and Doolittle (1987) J. Mol. Evol. 35:351-360. The method used is similar to the method described by Higgins and Sharp (1989) CABIOS 5:151-153. The program can align up to 300 sequences, each of a maximum length of 5,000 nucleotides or amino acids. The multiple alignment procedure begins with the pairwise alignment of the two most similar sequences, producing a cluster of two aligned sequences .
  • This cluster is then aligned to the next most related sequence or cluster of aligned sequences.
  • Two clusters of sequences are aligned by a simple extension of the pairwise alignment of two individual sequences .
  • the final alignment is achieved by a series of progressive, pairwise alignments.
  • the program is run by designating specific sequences and their amino acid or nucleotide coordinates for regions of sequence comparison and by designating the program parameters. For example, a reference sequence can be compared to other test sequences to determine the percent sequence identity relationship using the following parameters: default gap weight (3.00), default gap length weight (0.10) , and weighted end gaps.
  • HSPs high scoring sequence pairs
  • initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them.
  • the word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
  • the BLAST program uses as defaults a wordlength (W) of 11, the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1989) Proc. Nat'l Acad. Sci. USA 89:10915) alignments (B) of
  • the BLAST algorithm In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Nat'l Acad. Sci. USA 90:5873-5787) .
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • P(N) the smallest sum probability
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.
  • a further indication that two nucleic acid sequences of polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is " " immunologically cross reactive with the polypeptide encoded by the second nucleic acid, as described below.
  • a polypeptide is typically substantially identical to a second polypeptide, e.g., where the two peptides differ only by conservative substitutions.
  • Another indication that two nucleic acid sequences are substantially identical is that the two molecules hybridize to each other under stringent conditions, as described below.
  • the isolated DNA can be readily modified by nucleotide substitutions, nucleotide deletions, nucleotide insertions, and inversions of nucleotide stretches.
  • mutant IL-1RD9 encompasses a polypeptide otherwise falling within the homology definition of the IL-1R9 as set forth above, but having an amino acid sequence which differs from that of other IL-lRD-like polypeptides as found in nature, whether by way of deletion, substitution, or insertion.
  • site specific mutant IL-1RD9 encompasses a polypeptide having substantial homology with a polypeptide of SEQ ID NO: 6, 8, 10, 12, 14 or 14, and typically shares most of the biological activities or effects of the forms disclosed herein. Although site specific mutation sites are predetermined, mutants need not be site specific. Mammalian IL-1RD9 mutagenesis can be achieved by making amino acid insertions or deletions in the gene, coupled with expression. Substitutions, deletions, insertions, or many combinations may be generated to arrive at a final construct. Insertions include amino- or carboxy- terminal fusions.
  • Random mutagenesis can be conducted at a target codon and the expressed mammalian IL-1RD9 mutants can then be screened for the desired activity, providing some aspect of a structure-activity relationship.
  • Methods for making substitution mutations at predetermined sites in DNA having a known sequence are well known in the art, e.g., by M13 primer mutagenesis. See also Sambrook, et al . (1989) and Ausubel, et al . (1987 and periodic Supplements) .
  • the mutations in the DNA normally should not place coding sequences out of reading frames and preferably will not create complementary regions that could hybridize to produce secondary mRNA structure such as loops or hairpins .
  • the present invention encompasses primate IL-1RD8, primate or rodent IL-1RD9, and primate IL-lRD10,e.g. , whose sequences are disclosed e.g., in Tables 1-3, and described herein. Descriptions of features of IL-1RD9 are applicable in most cases, with appropriate modifications, also to IL-1RD8 and/or to IL- 1RD10. Allelic and other variants are also contemplated, including, e.g., fusion proteins combining portions of such sequences with others, including epitope tags and functional domains. Particularly interesting constructs will be intact extracellular or intracellular domains .
  • the present invention also provides recombinant polypeptides, e.g., heterologous fusion proteins using segments from these rodent proteins.
  • a heterologous fusion protein is a fusion of proteins or segments which are naturally not normally fused in the same manner.
  • the fusion product of, e.g., an IL-1RD9 with another IL-1 receptor is a continuous protein molecule having sequences fused in a typical polypeptide linkage, typically made as a single translation product and exhibiting properties, e.g., sequence or antigenicity, derived from each source peptide.
  • a similar concept applies to heterologous nucleic acid sequences.
  • new constructs may be made from combining similar functional or structural domains from other related proteins, e.g., IL-1 receptors or Toll-like receptors, including species variants.
  • ligand-binding or other segments may be "swapped" between different new fusion polypeptides or fragments. See, e.g., Cunningham, et al . (1989) Science 243:1330-1336; and O'Dowd, et al. (1988) J. Biol. Chem. 263:15985-15992, each of which is incorporated herein by reference.
  • new chimeric polypeptides exhibiting new combinations of specificities will result from the functional linkage of receptor-binding specificities.
  • a fusion protein may include a targeting domain which may serve to provide sequestering of the fusion protein to a particular subcellular organelle.
  • Candidate fusion partners and sequences can be selected from various sequence data bases, e.g., GenBank, c/o NCBI, and BCG, University of Wisconsin Biotechnology Computing Group, Madison, WI, which are each incorporated herein by reference.
  • the present invention particularly provides muteins which bind IL-1-like ligands, and/or which are affected in signal transduction.
  • Structural alignment of human IL-1RD9 with other members of the IL-1R family show conserved features/residues. See Tables 1-4. Alignment of the human IL-1RD9 sequence with other members of the IL-1R family indicates various structural and functionally shared features. See also, Bazan, et al . (1996) Nature 379:591; Lodi, et al . (1994) Science 263:1762-1766; Sayle and Milner-White (1995) TIBS 20:374- 376; and Gronenberg, et al . (1991) Protein Engineering
  • the IL-l ⁇ and IL-l ⁇ ligands bind an IL-1 receptor type I (IL-1RD1) as the primary receptor and this complex then forms a high affinity receptor complex with the IL-1 receptor type III (IL-1RD3) .
  • IL-1RD1 IL-1 receptor type I
  • IL-1RD3 IL-1 receptor type III
  • Such receptor subunits are probably shared with the receptors for the new IL-1 ligand family members. See, e.g., USSN 60/044,165 and USSN 60/055,111. It is likely that the IL-l ⁇ ligand signals through a receptor comprising the association of IL-1RD9 (alpha component) with IL-1RD5 (beta component) .
  • the IL-l ⁇ and IL-l ⁇ ligands each probably signal through a receptor comprising the association of one of IL-1RD4, IL-1RD6, or IL-1RD9 (alpha components) with one of IL- 1RD3, IL-1RD5, IL-1RD7, IL-1RD8, or IL-1RD10 (beta components) .
  • “Derivatives” of the primate or mouse IL-1RD9 include amino acid sequence mutants, glycosylation variants, metabolic derivatives and covalent or aggregative conjugates with other chemical moieties.
  • Covalent derivatives can be prepared by linkage of functionalities to groups which are found in the IL-1RD9 amino acid side chains or at the N- or C- termini, e.g., by means which are well known in the art.
  • These derivatives can include, without limitation, aliphatic esters or amides of the carboxyl terminus, or of residues containing carboxyl side chains, O-acyl derivatives of hydroxyl group-containing residues, and N-acyl derivatives of the amino terminal amino acid or amino-group containing residues, e.g., lysine or arginine.
  • Acyl groups are selected from the group of alkyl-moieties including C3 to C18 normal alkyl, thereby forming alkanoyl aroyl species.
  • glycosylation alterations are included, e.g., made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing, or in further processing steps. Particularly preferred means for accomplishing this are by exposing the polypeptide to glycosylating enzymes derived from cells which normally provide such processing, e.g., mammalian glycosylation enzymes. Deglycosylation enzymes are also contemplated. Also embraced are versions of the same primary amino acid sequence which have other minor modifications, including phosphorylated amino acid residues, e.g., phosphotyrosine, phosphoserine, or phosphothreonine .
  • a major group of derivatives are covalent conjugates of the receptors or fragments thereof with other polypeptides.
  • These derivatives can be synthesized in recombinant culture such as N- or C-terminal fusions or by the use of agents known in the art for their " " usefulness in cross-linking proteins through reactive side groups.
  • Preferred derivatization sites with cross-linking agents are at free amino groups, carbohydrate moieties, and cysteine residues. Fusion polypeptides between the receptors and other homologous or heterologous proteins are also provided.
  • Homologous polypeptides may be fusions between different receptors, resulting in, for instance, a hybrid protein exhibiting binding specificity for multiple different IL- 1 ligands, or a receptor which may have broadened or weakened specificity of substrate effect.
  • heterologous fusions may be constructed which would exhibit a combination of properties or activities of the derivative proteins.
  • Typical examples are fusions of a reporter polypeptide, e.g., luciferase, with a segment or domain of a receptor, e.g., a ligand-binding segment, so that the presence or location of a desired ligand may be easily determined. See, e.g., Dull, et al . , U.S. Patent No.
  • GST glutathione-S-transferase
  • bacterial ⁇ - galactosidase bacterial ⁇ - galactosidase
  • trpE bacterial ⁇ - galactosidase
  • Protein A ⁇ -lactamase
  • alpha amylase alpha amylase
  • alcohol dehydrogenase yeast alpha mating factor
  • a double stranded fragment will often be obtained either by synthesizing the complementary strand and annealing the strand together under appropriate conditions or by adding the complementary strand using DNA polymerase with an appropriate primer sequence.
  • Such polypeptides may also have amino acid residues which have been chemically modified by phosphorylation, sulfonation, biotinylation, or the addition or removal of other moieties, particularly those which have molecular shapes similar to phosphate groups.
  • the modifications will be useful labeling reagents, or serve as purification targets, e.g., affinity ligands.
  • Fusion proteins will typically be made by either recombinant nucleic acid methods or by synthetic polypeptide methods. Techniques for nucleic acid manipulation and expression are described generally, e.g., in Sambrook, et al . (1989) Molecular Cloning: A Laboratory Manual (2d ed. ) , Vols. 1-3, Cold Spring Harbor Laboratory, and Ausubel, et al . (eds. 1987 and periodic supplements) Current Protocols in Molecular Biology. Greene/Wiley, New York, which are each incorporated herein by reference.
  • covalent or aggregative derivatives are useful as immunogens, as reagents in immunoassays, or in purification methods such as for affinity purification of a receptor or other binding molecule, e.g., an antibody.
  • an IL-1 ligand can be immobilized by covalent bonding to a solid support such as cyanogen bromide-activated Sepharose, by methods which are well known in the art, or adsorbed onto polyolefin surfaces, with or without glutaraldehyde cross-linking, for use in the assay or purification of an IL-1 receptor, antibodies, or other similar molecules.
  • the ligand can also be labeled with a detectable group, e.g., " " radioiodinated by the chloramine T procedure, covalently bound to rare earth chelates, or conjugated to another fluorescent moiety for use in diagnostic assays.
  • An IL-1RD8, IL-1RD9, or IL-1RD10 of this invention can be used as an immunogen for the production of antisera or antibodies specific, e.g., capable of distinguishing between other IL-1 receptor family members, for the IL-1RD8, IL-1RD9, or IL-lRDIO or various fragments thereof.
  • the purified IL-1RD8, IL-1RD9, or IL- 1RD10 can be used to screen monoclonal antibodies or antigen-binding fragments prepared by immunization with various forms of impure preparations containing the protein.
  • the term "antibodies" also encompasses antigen binding fragments of natural antibodies, e.g., Fab, Fab2, Fv, etc.
  • the purified IL- 1RD9 can also be used as a reagent to detect antibodies generated in response to the presence of elevated levels of expression, or immunological disorders which lead to antibody production to the endogenous receptor.
  • IL-1RD8, IL-1RD9, or IL-lRDIO fragments may also serve as immunogens to produce the antibodies of the present invention, as described immediately below.
  • this invention contemplates antibodies having binding affinity to or being raised against the amino acid sequences shown, e.g., in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 and 20, fragments thereof, or various homologous peptides.
  • this invention contemplates antibodies having binding affinity to, or having been raised against, specific fragments which are predicted to be, or actually are, exposed at the exterior polypeptide surface of the native IL-1RD8, IL-1RD9, or IL-1RD10.
  • Various preparations of desired selectivity in binding can be prepared by appropriate cross absorptions, etc.
  • the blocking of physiological response to the receptor ligands may result from the inhibition of binding of the ligand to the receptor, likely through competitive inhibition.
  • in vitro assays of the present invention will often use antibodies or antigen binding segments of these antibodies, or fragments attached to solid phase substrates. These assays will also allow for the diagnostic determination of the effects of either ligand binding region mutations and modifications, or other mutations and modifications, e.g., which affect signaling or enzymatic function.
  • This invention also contemplates the use of competitive drug screening assays, e.g., where neutralizing antibodies to the receptor or fragments compete with a test compound for binding to a ligand or other antibody.
  • the neutralizing antibodies or fragments can be used to detect the presence of a polypeptide which shares one or more binding sites to a receptor and can also be used to occupy binding sites on a receptor that might otherwise bind a ligand.
  • DNA which encodes the polypeptides or fragments thereof can be obtained by chemical synthesis, screening cDNA libraries, or by screening genomic libraries prepared from a wide variety of cell lines or tissue samples. Natural sequences can be isolated using standard methods and the sequences provided herein, e.g., in Tables 1-3. Other species counterparts can be identified by hybridization techniques, or by various PCR techniques, combined with or by searching in sequence databases , e.g., GenBank.
  • This DNA can be expressed in a wide variety of host cells for the synthesis of a full-length receptor or fragments which can in turn, e.g., be used to generate polyclonal or monoclonal antibodies; for binding studies; for construction and expression of modified ligand binding or kinase/phosphatase domains; and for structure/function studies.
  • Variants or fragments can be expressed in host cells that are transformed or transfected with appropriate expression vectors .
  • These molecules can be substantially free of protein or cellular contaminants, other than those derived from the recombinant host, and therefore are particularly useful in pharmaceutical compositions when combined with a pharmaceutically acceptable carrier and/or diluent .
  • the protein, or portions thereof, may be expressed as fusions with other proteins .
  • Expression vectors are typically self-replicating DNA or RNA constructs containing the desired receptor gene or its fragments, usually operably linked to suitable genetic control elements that are recognized in a suitable host cell. These control elements are capable of effecting expression within a suitable host. The specific type of control elements necessary to effect expression will depend upon the eventual host cell used.
  • the genetic control elements can include a prokaryotic promoter system or a eukaryotic promoter expression control system, and typically include a transcriptional promoter, an optional operator to control the onset of transcription, transcription enhancers to elevate the level of mRNA expression, a sequence that encodes a suitable ribosome binding site, and sequences that terminate transcription and translation.
  • Expression vectors also usually contain an origin of replication that allows the vector to replicate independently of the host cell.
  • the vectors of this invention include those which contain DNA which encodes a protein, as described, or a fragment thereof encoding a biologically active equivalent polypeptide.
  • the DNA can be under the control of a viral promoter and can encode a selection marker.
  • This invention further contemplates use of such expression vectors which are capable of expressing eukaryotic cDNA coding for such a polypeptide in a prokaryotic or eukaryotic host, where the vector is compatible with the host and where the eukaryotic cDNA coding for the receptor is inserted into the vector such that growth of the host containing the vector expresses the cDNA in question.
  • expression vectors are designed for stable replication in their host cells or for amplification to greatly increase the total number of copies of the desirable gene per cell . It is not always necessary to require that an expression vector replicate in a host cell, e.g., it is possible to effect transient expression of the polypeptide or its fragments in various hosts using vectors that do not contain a replication origin that is recognized by the host cell. It is also possible to use vectors that cause integration of the polypeptide encoding portion or its fragments into the host DNA by recombination. Vectors, as used herein, comprise plasmids, viruses, bacteriophage, integratable DNA fragments, and other vehicles which enable the integration of DNA fragments into the genome of the host.
  • Expression vectors are specialized vectors which contain genetic control elements that effect expression of operably linked genes. Plasmids are the most commonly used form of vector but all other forms of vectors which serve an equivalent function and which are, or become, known in the art are suitable for use herein. See, e.g., Pouwels, et al . (1985 and Supplements) Cloning Vectors : A Laboratory Manual , Elsevier, N.Y. , and Rodriquez, et al . (eds.) Vectors : A Survey of Molecular Cloning Vectors and Their Uses, Buttersworth, Boston, 1988, which are incorporated herein by reference.
  • Transformed cells are cells, preferably mammalian, that have been transformed or transfected with receptor vectors constructed using recombinant DNA techniques.
  • Transformed host cells usually express the desired polypeptide or its fragments, but for purposes of cloning, amplifying, and manipulating its DNA, do not need to express the subject protein.
  • This invention further contemplates culturing transformed cells in a nutrient medium, thus permitting the receptor to accumulate in the cell membrane.
  • the polypeptide can be recovered, either from the culture or, in certain instances, from the culture medium.
  • nucleic sequences are operably linked when they are functionally related to each other.
  • DNA for a presequence or secretory leader is operably linked to a polypeptide if it is expressed as a preprotein or participates in directing the polypeptide to the cell membrane or in secretion of the polypeptide.
  • a promoter is operably linked to a coding sequence if it controls the transcription of the polypeptide;
  • a ribosome binding site is operably linked to a coding sequence if it is positioned to permit translation.
  • operably linked means contiguous and in reading frame, however, certain genetic elements such as repressor genes are not contiguously linked but still bind to operator sequences that in turn control expression.
  • Suitable host cells include prokaryotes, lower eukaryotes, and higher eukaryotes .
  • Prokaryotes include both gram negative and gram positive organisms, e.g., E. coli and B. subtilis.
  • Lower eukaryotes include yeasts, e.g., S. cerevisiae and Pichia, and species of the genus Dictvostelium.
  • Higher eukaryotes include established tissue culture cell lines from animal cells, both of non-mammalian origin, e.g., insect cells, and birds, and of mammalian origin, e.g., human, primates, and rodents.
  • Prokaryotic host-vector systems include a wide variety of vectors for many different species. As used herein, E. coli and its vectors will be used generically to include equivalent vectors used in other prokaryotes.
  • a representative vector for amplifying DNA is pBR322 or many of its derivatives .
  • Vectors that can be used to express the receptor or its fragments include, but are not limited to, such vectors as those containing the lac promoter (pUC-series) ; trp promoter (pBR322-trp) ; Ipp promoter (the pIN-series) ; lambda-pP or pR promoters (pOTS) ; or hybrid promoters such as ptac (pDR540) .
  • yeast vectors typically consist of a replication origin (unless of the integrating type) , a selection gene, a promoter, DNA encoding the receptor or its fragments, and sequences for translation termination, polyadenylation, and transcription termination.
  • Suitable expression vectors for yeast include such constitutive promoters as 3-phosphoglycerate kinase and various other glycolytic enzyme gene promoters or such inducible promoters as the alcohol dehydrogenase 2 promoter or metallothionine promoter.
  • Suitable vectors include derivatives of the following types: self-replicating low copy number (such as the YRp-series) , self-replicating high copy number (such as the YEp-series) ; integrating types (such as the Yip-series) , or mini-chromosomes (such as the YCp-series) .
  • Higher eukaryotic tissue culture cells are normally the preferred host cells for expression of the functionally active interleukin protein.
  • many higher eukaryotic tissue culture cell lines are workable, e.g., insect baculovirus expression systems, whether from an invertebrate or vertebrate source.
  • mammalian cells are preferred. Transformation or transfection and propagation of such cells has become a routine procedure.
  • useful cell lines include HeLa cells, Chinese hamster ovary (CHO) cell lines, baby rat kidney (BRK) cell lines, insect cell lines, bird cell lines, and monkey (COS) cell lines.
  • Expression vectors for such cell lines usually include an origin of replication, a promoter, a translation initiation site, RNA splice sites (if genomic DNA is used) , a polyadenylation site, and a transcription termination site. These vectors also usually contain a selection gene or amplification gene. Suitable expression vectors may be plasmids, viruses, or retroviruses carrying promoters derived, e.g., from such sources as from adenovirus, SV40, parvoviruses , vaccinia virus, or cytomegalovirus . Representative examples of suitable expression vectors include pCDNAl; pCD, see Okayama, et al. (1985) Mol. Cell Biol. 5:1136-1142; pMClneo PolyA, see Thomas, et al . (1987) Cell 51:503-512; and a baculovirus vector such as pAC 373 or pAC 610.
  • an open reading frame usually encodes a polypeptide that consists of a mature or secreted product covalently linked at its N-terminus to a signal peptide.
  • the signal peptide is cleaved prior to secretion of the mature, or active, polypeptide.
  • the cleavage site can be predicted with a high degree of accuracy from empirical rules, e.g., von-Heijne (1986) Nucleic Acids Research 14:4683-4690 and Nielsen, et al . (1997) Protein Enq. 10:1-12, and the precise amino acid composition of the signal peptide often does not appear to be critical to its function, e.g., Randall, et al. (1989) Science 243:1156-1159; Kaiser, et al. (1987) Science 235:312-317.
  • polypeptides it will often be desired to express these polypeptides in a system which provides a specific or defined glycosylation pattern.
  • the usual pattern will be that provided naturally by the expression system.
  • the pattern will be modifiable by exposing the polypeptide, e.g., an unglycosylated form, to appropriate glycosylating proteins introduced into a heterologous expression system.
  • the receptor gene may be co-transformed with one or more genes encoding mammalian or other glycosylating enzymes . Using this approach, certain mammalian glycosylation patterns will be achievable in prokaryote or other cells.
  • the source of IL-1RD8, IL-1RD9, or IL-lRDIO can be a eukaryotic or prokaryotic host expressing recombinant IL- 1RD8, IL-1RD9, or IL-lRDIO such as is described above.
  • the source can also be a cell line such as mouse Swiss 3T3 fibroblasts, but other mammalian cell lines are also contemplated by this invention, with the preferred cell line being from the human species.
  • the primate IL- lRs, fragments, or derivatives thereof can be prepared by conventional processes for synthesizing peptides. These include processes such as are described in Stewart and Young (1984) Solid Phase Peptide Synthesis. Pierce Chemical Co., Rockford, IL; Bodanszky and Bodanszky (1984) The Practice of Peptide Synthesis, Springer-Verlag, New York; and Bodanszky (1984) The
  • an azide process for example, an acid chloride process, an acid anhydride process, a mixed anhydride process, an active ester process (e.g., p-nitrophenyl ester, N-hydroxysuccinimide ester, or cyanomethyl ester), a carbodiimidazole process, an oxidative-reductive process, or a dicyclohexylcarbodiimide (DCCD) /additive process
  • Solid phase and solution phase syntheses are both applicable to the foregoing processes. Similar techniques can be used with partial IL-1RD9 sequences.
  • the IL-1RD8, IL-1RD9, or IL-1RD10 proteins, polypeptides, fragments, or derivatives are suitably prepared in accordance with the above processes as typically employed in peptide synthesis, generally either by a so-called stepwise process which comprises condensing an amino acid to the terminal amino acid, one by one in sequence, or by coupling peptide fragments to the terminal amino acid. Amino groups that are not being used in the coupling reaction typically must be protected to prevent coupling at an incorrect location.
  • the C-terminal amino acid is bound to an insoluble carrier or support through its carboxyl group.
  • the insoluble carrier is not particularly limited as long as it has a binding capability to a reactive carboxyl group.
  • examples of such insoluble carriers include halomethyl resins, such as chloromethyl resin or bromomethyl resin, hydroxymethyl resins, phenol resins, tert-alkyloxycarbonylhydrazidated resins, and the like.
  • An amino group-protected amino acid is bound in sequence through condensation of its activated carboxyl group and the reactive amino group of the previously formed peptide or chain, to synthesize the peptide step by step. After synthesizing the complete sequence, the peptide is split off from the insoluble carrier to produce the peptide. This solid-phase approach is generally described by Merrifield, et al . (1963) in J.
  • the prepared protein and fragments thereof can be isolated and purified from the reaction mixture by means of peptide separation, e.g., by extraction, precipitation, electrophoresis, various forms of chromatography, and the like.
  • the receptors of this invention can be obtained in varying degrees of purity depending upon desired uses. Purification can be accomplished by use of the protein purification techniques disclosed herein, see below, or by the use of the antibodies herein described in methods of immunoabsorbant affinity chromatography.
  • This immunoabsorbant affinity chromatography is carried out by first linking the antibodies to a solid support and then contacting the linked antibodies with solubilized lysates of appropriate cells, lysates of other cells expressing the receptor, or lysates or supernatants of cells producing the polypeptide as a result of DNA techniques, see below.
  • the purified protein will be at least .. about 40% pure, ordinarily at least about 50% pure, usually at least about 60% pure, typically at least about 70% pure, more typically at least about 80% pure, preferable at least about 90% pure and more preferably at least about 95% pure, and in particular embodiments, 97%- 99% or more.
  • Purity will usually be on a weight basis, but can also be on a molar basis. Different assays will be applied as appropriate. Similar concepts apply to polynucleotides and antibodies .
  • Antibodies can be raised to the various mammalian IL-1RD8, IL-1RD9, or IL-lRDIO described herein, e.g. ⁇ primate IL-1RD9 polypeptides and fragments thereof, both in naturally occurring native forms and in their recombinant forms, the difference being that antibodies to the active receptor are more likely to recognize epitopes which are only present in the native conformations .
  • Denatured antigen detection can also be useful in, e.g., Western analysis.
  • Anti-idiotypic antibodies are also contemplated, which would be useful as agonists or antagonists of a natural receptor or an antibody.
  • Antibodies, including binding fragments and single chain versions, against predetermined fragments of the polypeptide can be raised by immunization of animals with conjugates of the fragments with immunogenic proteins.
  • Monoclonal antibodies are prepared from cells secreting the desired antibody. These antibodies can be screened for binding to normal or defective protein, or screened for agonistic or antagonistic activity. These monoclonal antibodies will usually bind with at least a K ⁇ , of about 1 mM, more usually at least about 300 ⁇ M, typically at least about 100 ⁇ M, more typically at least about 30 ⁇ M, preferably at least about 10 ⁇ M, and more preferably at least about 3 ⁇ M or better.
  • the antibodies, including antigen binding fragments, of this invention can have significant diagnostic or therapeutic value. They can be potent antagonists that bind to the receptor and inhibit binding to ligand or inhibit the ability of the receptor to elicit a biological response, e.g., act on its substrate. They also can be useful as non-neutralizing antibodies and can be coupled to toxins or radionuclides to bind producing cells, or cells localized to the source of the interleukin. Further, these antibodies can be conjugated to drugs or other therapeutic agents, either directly or indirectly by means of a linker.
  • the antibodies of this invention can also be useful in diagnostic applications. As capture or non-neutralizing antibodies, they might bind to the receptor without inhibiting ligand or substrate binding. As neutralizing antibodies, they can be useful in competitive binding assays. They will also be useful in detecting or quantifying ligand. They may be used as reagents for Western blot analysis, or for immunoprecipitation or immunopurification of the respective protein.
  • Protein fragments may be joined to other materials, particularly polypeptides, as fused or covalently joined polypeptides to be used as immunogens .
  • Mammalian IL-IRs and fragments may be fused or covalently linked to a variety of immunogens, such as keyhole limpet hemocyanin, bovine serum albumin, tetanus toxoid, etc. See Microbiology, Hoeber Medical Division, Harper and Row, 1969; Landsteiner (1962) Specificity of Serological Reactions , Dover Publications, New York; and Williams, et al . (1967) Methods in Immunology and Immunochemistry, Vol.
  • a typical method involves hyperimmunization of an animal with an antigen. The blood of the animal is then collected shortly after the repeated immunizations and the gamma globulin is isolated.
  • monoclonal antibodies from various mammalian hosts, such as mice, rodents, primates, humans, etc.
  • Description of techniques for preparing such monoclonal antibodies may be found in, e.g., Stites, et al . (eds.) Basic and Clinical Immunology (4th ed. ) , Lange Medical Publications, Los Altos, CA, and references cited therein; Harlow and Lane (1988) Antibodies : A Laboratory Manual , CSH Press; Goding (1986) Monoclonal Antibodies: Principles and Practice (2d ed.
  • the polypeptides and antibodies will be labeled by joining, either covalently or non-covalently, a substance which provides for a detectable signal .
  • labels and conjugation techniques are known and are reported extensively in both the scientific and patent literature. Suitable labels include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent moieties, chemiluminescent moieties, magnetic particles, and the like. Patents, teaching the use of such labels include U.S. Patent Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241.
  • recombinant or chimeric immunoglobulins may be produced, see Cabilly, U.S. Patent No. 4,816,567; or made in transgenic mice, see Mendez, et al. (1997) Nature Genetics 15:146-156. These references are incorporated herein by reference .
  • the antibodies of this invention can also be used for affinity chromatography in isolating the IL-lRs.
  • Columns can be prepared where the antibodies are linked to a solid support, e.g., particles, such as agarose, Sephadex, or the like, where a cell lysate may be passed through the column, the column washed, followed by increasing concentrations of a mild denaturant, whereby the purified protein will be released.
  • the protein may be used to purify antibody.
  • the antibodies may also be used to screen expression libraries for particular expression products .
  • the antibodies used in such a procedure will be labeled with a moiety allowing easy detection of presence of antigen by antibody binding.
  • Antibodies raised against an IL-1R will also be used to raise anti-idiotypic antibodies. These will be useful in detecting or diagnosing various immunological conditions related to expression of the protein or cells which express the protein. They also will be useful as agonists or antagonists of the ligand, which may be competitive inhibitors or substitutes for naturally occurring ligands.
  • An IL-1R polypeptide that specifically binds to or that is specifically immunoreactive with an antibody generated against a defined immunogen is typically determined in an immunoassay.
  • the immunoassay typically uses a polyclonal antiserum which was raised, e.g., to a polypeptide of SEQ ID NO: 4, 10, or 20. This antiserum is selected to have low crossreactivity against other IL-1R family members, e.g., IL-lRs Dl through D8, preferably from the same species, and any such crossreactivity is removed by immunoabsorption prior to use in the immunoassay.
  • polypeptide of, e.g., SEQ ID NO: 4, 10, or 20, is " * isolated as described herein.
  • recombinant polypeptide may be produced in a mammalian cell line.
  • An appropriate host e.g., an inbred strain of mice such as Balb/c, is immunized with the selected protein, typically using a standard adjuvant, such as Freund's adjuvant, and a standard mouse immunization protocol (see Harlow and Lane, supra) .
  • a synthetic peptide derived from the sequences disclosed herein and conjugated to a carrier polypeptide can be used an immunogen.
  • Polyclonal sera are collected and titered against the immunogen polypeptide in an immunoassay, e.g., a solid phase immunoassay with the immunogen immobilized on a solid support.
  • Polyclonal antisera with a titer of 10 ⁇ or greater are selected and tested for their cross reactivity against other IL-1R family members, e.g., IL- 1RD1 through IL-1RD6, using a competitive binding immunoassay such as the one described in Harlow and Lane, supra, at pages 570-573.
  • IL-1R family members e.g., IL- 1RD1 through IL-1RD6
  • a competitive binding immunoassay such as the one described in Harlow and Lane, supra, at pages 570-573.
  • at least two IL-1R family members are used in this determination.
  • These IL- 1R family members can be produced as recombinant polypeptides and isolated using standard molecular biology and protein chemistry techniques as described herein.
  • Immunoassays in the competitive binding format can be used for the crossreactivity determinations.
  • the polypeptide of SEQ ID NO: 4, 10, or 20 can be immobilized to a solid support.
  • Polypeptides added to the assay compete with the binding of the antisera to the immobilized antigen.
  • the ability of the above polypeptides to compete with the binding of the antisera to the immobilized polypeptide is compared to the polypeptides of IL-1RD1 through IL-1RD6.
  • the percent crossreactivity for the above polypeptides is calculated, using standard calculations. Those antisera with less than 10% crossreactivity with each of the polypeptides listed above are selected and pooled.
  • the cross-reacting antibodies are then removed from the pooled antisera by immunoabsorption with the above-listed proteins .
  • the immunoabsorbed and pooled antisera are then used in a competitive binding immunoassay as described above to compare a second polypeptide to the immunogen polypeptide (e.g., the IL-1RD8, IL-1RD9, or IL-1RD10 like polypeptide of SEQ ID NO: 4, 10, or 20) .
  • the two polypeptides are each assayed at a wide range of concentrations and the amount of each polypeptide required to inhibit 50% of the binding of the antisera to the immobilized polypeptide is determined. If the amount of the second polypeptide required is less than twice the amount of the polypeptide of the selected polypeptide or polypeptides that is required, then the second polypeptide is said to specifically bind to an antibody generated to the immunogen.
  • these IL-1R polypeptides are members of a family of homologous polypeptides that comprise at least 7 genes previously identified.
  • the term refers not only to the amino acid sequences disclosed herein, but also to other polypeptides that are allelic, non-allelic, or species variants.
  • the terms include nonnatural mutations introduced by deliberate mutation using conventional recombinant technology such as single site mutation, or by excising short sections of DNA encoding the respective proteins, or by substituting new amino acids, or adding new amino acids . Such minor alterations typically will substantially maintain the immunoidentity of the original molecule and/or its biological activity.
  • these alterations include polypeptides that are specifically immunoreactive with a designated naturally occurring IL- 1RD8, IL-1RD9, or IL-1RD10 protein.
  • the biological properties of the altered polypeptides can be determined by expressing the polypeptide in an appropriate cell line and measuring the appropriate effect, e.g., upon transfected lymphocytes .
  • Particular polypeptide modifications considered minor would include conservative substitution of amino acids with similar chemical properties, as described above for the IL-1R family as a whole.
  • kits and assay methods Both naturally occurring and recombinant forms of the IL-1R like molecules of this invention are particularly useful in kits and assay methods. For example, these methods would also be applied to screening for binding activity, e.g., ligands for these proteins.
  • ligands for these proteins.
  • automating assays have been developed in recent years so as to permit screening of tens of thousands of compounds per year. See, e.g., a BIOMEK automated workstation, Beckman Instruments, Palo Alto,
  • kits and methods for detecting the presence of the protein or its ligand Alternatively, or additionally, antibodies against the molecules may be incorporated into the kits and methods.
  • the kit will have a compartment containing, e.g., either an IL-1RD9 peptide or gene segment or a reagent which recognizes one or the other.
  • recognition reagents in the case of peptide, would be a ligand or antibody, or in the case of a gene segment, would usually be a hybridization probe.
  • a preferred kit for determining the concentration of IL-1RD8, IL-1RD9, or IL-lRDlO in a sample would typically comprise a labeled compound, e.g., ligand or antibody, having known binding affinity for IL-1RD9, a source of IL-1RD9 (naturally occurring or recombinant) as a positive control, and a means for separating the bound from free labeled compound, for example a solid phase for immobilizing the IL-1RD9 in the test sample. Compartments containing reagents, and instructions, will normally be provided.
  • Antibodies including antigen binding fragments, specific for mammalian IL-1RD8 or a peptide fragment, or receptor fragments are useful in diagnostic applications to detect the presence of elevated levels of ligand and/or its fragments. Diagnostic assays may be homogeneous (without a separation step between free reagent and antibody-antigen complex) or heterogeneous (with a separation step) .
  • Various commercial assays exist, such as radioimmunoassay (RIA) , enzyme-linked immunosorbent assay (ELISA) , enzyme immunoassay (EIA) , enzyme-multiplied immunoassay technique (EMIT) , substrate-labeled fluorescent immunoassay (SLFIA) and the like.
  • unlabeled antibodies can be employed by using a second antibody which is labeled and which recognizes the antibody to an IL-1R or to a particular fragment thereof .
  • a second antibody which is labeled and which recognizes the antibody to an IL-1R or to a particular fragment thereof .
  • Anti-idiotypic antibodies may have similar use to serve as agonists or antagonists of IL-IRs . These should be useful as therapeutic reagents under appropriate circumstances . Frequently, the reagents for diagnostic assays *" are supplied in kits, so as to optimize the sensitivity of the assay. For the subject invention, depending upon the nature of the assay, the protocol, and the label, either labeled or unlabeled antibody, or labeled ligand is provided.
  • the kit will also contain instructions for proper use and disposal of the contents after use.
  • the kit has compartments for each useful reagent, and will contain instructions for proper use and disposal of reagents.
  • the reagents are provided as a dry lyophilized powder, where the reagents may be reconstituted in an aqueous medium having appropriate concentrations for performing the assay.
  • labeling may be achieved by covalently or non-covalently joining a moiety which directly or indirectly provides a detectable signal.
  • a test compound, IL-1R, or antibodies thereto can be labeled either directly or indirectly.
  • Possibilities for direct labeling include label groups: radiolabels such as ⁇ - ⁇ 1 , enzymes (U.S. Pat. No. 3,645,090) such as peroxidase and alkaline phosphatase, and fluorescent labels (U.S. Pat. No.
  • Possibilities for indirect labeling include biotinylation of one constituent followed by binding to avidin coupled to one of the above label groups .
  • the IL-1R can be immobilized on various matrixes followed by washing. Suitable matrices include plastic such as an ELISA plate, filters, and beads. Methods of immobilizing the receptor to a matrix include, without limitation, direct adhesion to plastic, use of a capture antibody, chemical coupling, and biotin-avidin.
  • the last step in this approach involves the precipitation of antibody/antigen complex by any of several methods including those utilizing, e.g., an organic solvent such as polyethylene glycol or a salt such as ammonium sulfate.
  • organic solvent such as polyethylene glycol
  • salt such as ammonium sulfate.
  • suitable separation techniques include, without limitation, the fluorescein antibody magnetizable particle method described in
  • Another diagnostic aspect of this invention involves use of oligonucleotide or polynucleotide sequences taken from the sequence of an IL-1R. These sequences can be used as probes for detecting levels of the respective IL- 1R in patients suspected of having an immunological disorder.
  • the preparation of both RNA and DNA nucleotide sequences, the labeling of the sequences, and the preferred size of the sequences has received ample description and discussion in the literature.
  • an oligonucleotide probe should have at least about 14 nucleotides, usually at least about 18 nucleotides, and the polynucleotide probes may be up to several kilobases .
  • Various labels may be employed, most commonly radionuclides, particularly 32p.
  • biotin modified nucleotides for introduction into a polynucleotide.
  • the biotin then serves as the site for binding to avidin or antibodies, which may be labeled with a wide variety of labels, such as radionuclides, fluorescers, enzymes, or the like.
  • antibodies may be employed which can recognize specific duplexes, including DNA duplexes, RNA duplexes, DNA-RNA hybrid duplexes, or DNA-protein duplexes.
  • the antibodies in turn may be labeled and the assay carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected.
  • probes to the novel anti-sense RNA may be carried out in conventional techniques such as nucleic acid hybridization, plus and minus screening, recombinational probing, hybrid released translation (HRT) , and hybrid arrested translation (HART) .
  • This also includes amplification techniques such as polymerase chain reaction (PCR) .
  • kits which also test for the qualitative or quantitative presence of other markers are also contemplated. Diagnosis or prognosis may depend on the combination of multiple indications used as markers . Thus, kits may test for combinations of markers. See, e.g., Viallet, et al. (1989) Progress in Growth Factor Res. 1:89-97.
  • the IL-IRs naturally occurring or recombinant
  • fragments thereof, mutein receptors, and antibodies should be useful in the treatment of conditions exhibiting abnormal expression of the receptors of their ligands. Such abnormality will typically be manifested by immunological disorders.
  • this invention should provide therapeutic value in various disease ' s or disorders associated with abnormal expression or abnormal triggering of response to the ligand.
  • the IL-1 ligands have been suggested to be involved in morphologic development, e.g., dorso-ventral polarity determination, and immune responses, particularly the primitive innate responses. See, e.g., Sun, et al . (1991) Eur . J . Biochem. 196:247-254; Hultmark (1994) Nature 367:116-117.
  • Recombinant IL-IRs, muteins, agonist or antagonist antibodies thereto, or antibodies can be purified and then administered to a patient.
  • These reagents can be combined for therapeutic use with additional active ingredients, e.g. , in conventional pharmaceutically acceptable carriers or diluents, along with physiologically innocuous stabilizers and excipients.
  • additional active ingredients e.g. , in conventional pharmaceutically acceptable carriers or diluents, along with physiologically innocuous stabilizers and excipients.
  • These combinations can be sterile, e.g., filtered, and placed into dosage forms as by lyophilization in dosage vials or storage in stabilized aqueous preparations.
  • This invention also contemplates use of antibodies or binding fragments thereof which are not complement binding.
  • Ligand screening using IL-1R or fragments thereof can be performed to identify molecules having binding affinity to the receptors. Subsequent biological assays can then be utilized to determine if a putative ligand can provide competitive binding, which can block intrinsic stimulating activity. Receptor fragments can be used as a blocker or antagonist in that it blocks the activity of ligand. Likewise, a compound having intrinsic stimulating activity can activate the receptor and is thus an agonist in that it simulates the activity of ligand, e.g., inducing signaling. This invention further contemplates the therapeutic use of antibodies to IL-IRs as antagonists.
  • reagents necessary for effective therapy will depend upon many different factors, including means of administration, target site, reagent physiological life, pharmacological life, physiological state of the patient, and other medicants administered.
  • treatment dosages should be titrated to optimize safety and efficacy.
  • dosages used in vitro may provide useful guidance in the amounts useful for in situ administration of these reagents.
  • Animal testing of effective doses for treatment of particular disorders will provide further predictive indication of human dosage.
  • Various considerations are described, e.g., in Gilman, et al . (eds., 1990) Goodman and Gilman's: The Pharmacological Bases of Therapeutics, 8th Ed.
  • Pharmaceutically acceptable carriers will include water, saline, buffers, and other compounds described, e.g., in the Merck Index, Merck & Co., Rahway, New Jersey. Because of the likely high affinity binding, or turnover numbers, between a putative ligand and its receptors, low dosages of these reagents would be initially expected to be effective. And the signaling pathway suggests extremely low amounts of ligand may have effect. Thus, dosage ranges would ordinarily be expected to be in amounts lower than 1 mM concentrations, typically less than about 10 ⁇ M concentrations, usually less than about 100 nM, preferably less than about 10 pM (picomolar) , and most preferably less than about 1 fM (femtomolar) , with an appropriate carrier. Slow release formulations, or slow release apparatus will often be utilized for continuous administration.
  • IL-IRs may be administered directly to the host to be treated or, depending on the size of the compounds, it may be desirable to conjugate them to carrier proteins such as ovalbumin or serum albumin prior to their administration.
  • Therapeutic formulations may be administered in many conventional dosage formulations . While it is possible for the active ingredient to be administered alone, it is preferable to present it as a pharmaceutical formulation.
  • Formulations comprise at least one active ingredient, as defined above, together with one or more acceptable carriers thereof. Each carrier must be both pharmaceutically and physiologically acceptable in the sense of being compatible with the other ingredients and not injurious to the patient.
  • Formulations include those suitable for oral, rectal, nasal, or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by methods well known in the art of pharmacy. See, e.g., Gilman, et al . (eds. 1990) Goodman and Gilman's: The Pharmacological Bases of Therapeutics , 8th Ed. , Pergamon Press; and Remington's Pharmaceutical Sciences, 17th ed. (1990), Mack Publishing Co., Easton, Perm.; Avis, et al . (eds. 1993) Pharmaceutical Dosage Forms : Parenteral Medications Dekker, NY; Lieberman, et al .
  • compositions comprising Tablets Dekker, NY; and Lieberman, et al. (eds. 1990) Pharmaceutical Dosage Forms : Disperse Systems Dekker, NY.
  • the therapy of this invention may be combined with or used in association with other therapeutic agents, particularly agonists or antagonists of other IL-1 family members.
  • IL-1 receptors herein provide means to identify ligands, as described above. Such ligand should bind specifically to the respective receptor with reasonably high affinity. Typical ligand receptor binding constants will be at least about 30 mM, e.g., generally at least about 3 mM, more generally at least about 300 ⁇ M, typically at least about 30 ⁇ M, 3 ⁇ M, 300 nM, 30 nM, etc. Various constructs are made available which allow either labeling of the receptor to detect its ligand. For example, directly labeling IL-1R, fusing onto it markers for secondary labeling, e.g., FLAG or other epitope tags, etc., will allow detection of receptor.
  • IL-IRs will be analogously applicable to individual specific embodiments directed to IL-1RD8, IL-1RD9, OR IL-lRDIO reagents and compositions .
  • Methods for protein purification include such methods as ammonium sulfate precipitation, column chromatography, electrophoresis, centrifugation, crystallization, and others. See, e.g., Ausubel, et al. (1987 and periodic supplements); Coligan, et al. (ed. 1996 and periodic supplements) Current Protocols In Protein Science Greene/Wiley, New York; Deutscher (1990) "Guide to Protein Purification” in Methods in Enzvmology, vol. 182, and other volumes in this series; and manufacturer's literature on use of protein purification products, e.g., Pharmacia, Piscataway, N.J., or Bio-Rad, Richmond, CA.
  • OIAexpress The High Level Expression & Protein Purification System QUIAGEN, Inc., Chatsworth, CA.
  • GenBank Computer sequence analysis is performed, e.g., using available software programs, including those from the GCG (U. Wisconsin) and GenBank sources. Public sequence databases were also used, e.g., from GenBank, NCBI, SWISSPROT, and others.
  • IL-10 receptors may be applied to IL-IRs, as described, e.g., in USSN 08/110,683 (IL-10 receptor) , which is incorporated herein by reference for all purposes. Also, while many of the techniques described are directed to the IL-1RD9 reagents, corresponding methods will typically be applicable with the IL-1RD8, and IL-1RD10 reagents. See also, USSN 60/065,776, filed November 17, 1997, and USSN 60/078,008, filed March 12, 1998, both of which are incorporated herein by reference.
  • IL-IRs Human sequences related to IL-IRs were identified from various EST databases using, e.g., the BLAST server (Altschul, et al . (1994) Nature Genet. 6:119-129). More sensitive pattern- and profile-based methods (Bork and Gibson (1996) Meth. Enzvmol. 266:162-184) were used to identify a fragment of a gene which exhibited certain homology to the IL-IRs .
  • Full length cDNAs for human IL-1RD9 are cloned, e.g., by DNA hybridization screening of ⁇ gtlO phage. PCR reactions were conducted using T. aquaticus Taqplus DNA polymerase (Stratagene) under appropriate conditions.
  • IL-1RD9 Message for genes encoding, e.g., IL-1RD9 will be assayed by appropriate technology, e.g., PCR, immunoassay, hybridization, or otherwise. Tissue and organ cDNA preparations are available, e.g., from Clontech, Mountain View, CA. Identification of sources of natural expression are useful, as described. And the identification of functional receptor subunit pairings will allow for prediction of what cells express the combination of receptor subunits which will result in a physiological responsiveness to each of the IL-1 ligands.
  • the message for IL-1RD9 is quite rare, as it is not found with a degree of frequency in the available sequence databases. This suggests, e.g., a very rare message, or a highly restricted distribution. IL-1R9 is expressed predominantly on T cells, NK cells, monocytes and dendritic cells . Southern Analysis on cDNA libraries can be performed:
  • DNA (5 ⁇ g) from a primary amplified cDNA library is digested with appropriate restriction enzymes to release the inserts, run on a 1% agarose gel and transferred to a nylon membrane ' (Schleicher and Schuell, Keene, NH) .
  • Samples for human mRNA isolation may include, e.g.
  • peripheral blood mononuclear cells peripheral blood mononuclear cells (monocytes, T cells, NK cells, granulocytes, B cells), resting (T100); peripheral blood mononuclear cells, activated with anti- CD3 for 2, 6, 12 h pooled (T101) ; T cell, THO clone Mot 72, resting (T102) ; T cell, THO clone Mot 72, activated with anti-CD28 and anti-CD3 for 3, 6, 12 h pooled (T103); T cell, THO clone Mot 72, anergic treated with specific peptide for 2, 7, 12 h pooled (T104) ; T cell, THl clone HY06, resting (T107) ; T cell, THl clone HY06, activated with anti-CD28 and anti-CD3 for 3, 6, 12 h pooled (T108) ; T cell, THl clone HY06, anergic treated with specific
  • IL-10 for 1, 2, 6, 12, 24 h pooled M103
  • elutriated monocytes, activated with LPS, IFN ⁇ , anti-IL-10 for 4, 16 h pooled M106
  • elutriated monocytes activated with LPS, IFN ⁇ , IL-10 for 4, 16 h pooled
  • M107 elutriated monocytes, activated LPS for 1 h
  • M108 elutriated monocytes, activated LPS for 6 h (M109)
  • DC 70% CDla+ from CD34+ GM-CSF, TNF ⁇ 12 days, resting (D101)
  • DC 70% CDla+ from CD34+ GM-CSF, TNF ⁇ 12 days, activated with PMA and ionomycin for 1 hr (D102)
  • DC 70% CDla+ from CD34+ GM-CSF, TNF ⁇ 12 days, activated with PMA and ionomycin for 6 hr (D103);
  • DC 9 DC 9
  • IL-1RD8 message is described below in Table 5.
  • fetal and transformed tissues express high levels, whereas normal, adult tissues express low levels (with the exception of skeletal muscle) .
  • Further insights into this phenomenon will need further experiments.
  • Message for genes encoding IL-1RD8 will be assayed by appropriate technology, e.g., PCR, immunoassay, hybridization, or otherwise.
  • Tissue and organ cDNA preparations are available, e.g., from Clontech, Mountain View, CA. Identification of sources of natural expression are useful, as described. And the identification of functional receptor subunit pairings will allow for prediction of what cells express the combination of receptor subunits which will result in a physiological responsiveness to each of the IL-1 ligands.
  • An appropriate, e.g., GST, fusion construct is engineered for expression, e.g., in E. coli.
  • a mouse IGIF pGex plasmid is constructed and transformed into E. coli.
  • Freshly transformed cells are grown, e.g.., in LB medium containing 50 ⁇ g/ml ampicillin and induced with IPTG (Sigma, St. Louis, MO). After overnight induction, the bacteria are harvested and the pellets containing, e.g., the IL-1R8 polypeptide are isolated.
  • the pellets are homogenized, e.g., in TE buffer (50 mM Tris-base pH 8.0, 10 mM EDTA and 2 mM pefabloc) in 2 liters. This material is passed through a microfluidizer (Microfluidics, Newton, MA) three times.
  • TE buffer 50 mM Tris-base pH 8.0, 10 mM EDTA and 2 mM pefabloc
  • the fluidized supernatant is spun down on a Sorvall GS-3 rotor for 1 h at 13,000 rpm.
  • the resulting supernatant containing the IL-1R polypeptide is filtered and passed over a glutathione-SEPHAROSE column equilibrated in 50 mM Tris-base pH 8.0.
  • the fractions containing the IL-1RD9- GST fusion protein are pooled and cleaved, e.g., with thrombin (Enzyme Research Laboratories, Inc., South Bend, IN) .
  • the cleaved pool is then passed over a Q-SEPHAROSE column equilibrated in 50 mM Tris-base.
  • Fractions containing IL-1RD9 are pooled and diluted in cold distilled H2O, to lower the conductivity, and passed back over a fresh Q-Sepharose column, alone or in succession with an immunoaffinity antibody column.. Fractions containing the IL-1RD9 polypeptide are pooled, aliquoted, and stored in the -70° C freezer.
  • the IL-l ⁇ and IL-l ⁇ ligands bind an IL-1RD1 as the primary receptor and this complex then forms a high affinity receptor complex with the IL-1RD3.
  • Such receptor subunits are probably shared with the receptors for the new IL-1 ligand family members. See, e.g., USSN 60/044,165 and USSN 60/055,111.
  • Combination of the IL- 1RD9 ( ⁇ subunit type, based upon sequence analysis) will combine with the IL-1RD5 ( ⁇ subunit type, based upon sequence analysis) to form a heterodimer receptor.
  • the IL-l ⁇ and IL-l ⁇ ligands each probably signal through a receptor comprising the association of IL-1RD4, IL-1RD6, or IL-1RD9 (alpha components) with IL-1RD3 , IL-1RD8, or IL-1RD10 (beta components) .
  • IL-1RD4, IL-1RD6, or IL-1RD9 alpha components
  • IL-1RD9, IL-1RD8, or IL-1RD10 beta components
  • IL-1RD7 IL-1RD7
  • IL-1RD8 IL-1RD8
  • the IL-1RD10 is most similar to the IL-1RD8 , suggesting that it may also be a beta receptor subunit.
  • Combinatorial transfections of transformations can make cells expressing defined subunits, which can be tested for response to each of the IL-1 ligands.
  • Appropriate cell types can be used, e.g., 293 T cells, Jurkat cells, with, e.g., a nuclear kappa B (NFKb) -controlled luciferase reporter construct such as described e.g., in Otieno et al.,(1997) Am J Physiol 273-xxx.
  • NFKb nuclear kappa B
  • IL-1RD3, IL-1RD5, IL-1RD8, and IL-lRDIO may functionally substitute for each other when in combination with IL-l ⁇ + IL-l ⁇ + IL-1RD1.
  • IL-l ⁇ + IL-1RD9 + IL-1RD8 -
  • Bio assays will generally be directed to the ligand binding feature of the protein or to the kinase/phosphatase activity of the receptor.
  • the activity will typically be reversible, as are many other enzyme actions mediate phosphatase or phosphorylase activities, which activities are easily measured by standard procedures. See, e.g., Hardie, et al . (eds. 1995) The Protein Kinase FactBook vols. I and II, Academic Press, San Diego, CA; Hanks, et al . (1991) Meth. Enzvmol . 200:38-62; Hunter, et al.
  • the family of interleukins 1 contains molecules, each of which is an important mediator of inflammatory disease.
  • interleukins 1 contains molecules, each of which is an important mediator of inflammatory disease.
  • Dinarello (1996) "Biologic basis for interleukin-1 in disease” Blood 87:2095-2147.
  • the various IL-1 ligands may play important roles in the initiation of disease, particularly inflammatory responses .
  • novel polypeptides related to the IL-1 family furthers the identification of molecules that provide the molecular basis for initiation of disease and allow for the development of therapeutic strategies of increased range and efficacy.
  • Balb/c mice are immunized intraperitoneally with recombinant forms of the polypeptide, e.g., purified IL-1RD8, IL-1RD9, and IL-1RD10, or stable transfected
  • NIH-3T3 cells Animals are boosted at appropriate time points with protein, with or without additional adjuvant, to further stimulate antibody production. Serum is collected, or hybridomas produced with harvested spleens. Alternatively, Balb/c mice are immunized with cells transformed with the gene or fragments thereof, either endogenous or exogenous cells, or with isolated membranes enriched for expression of the antigen. Serum is collected at the appropriate time, typically after numerous further administrations. Various gene therapy techniques may be useful, e.g., in producing protein in situ, for generating an immune response.
  • Monoclonal antibodies may be made. For example, splenocytes are fused with an appropriate fusion partner and hybridomas are selected in growth medium by standard procedures . Hybridoma supernatants are screened for the presence of antibodies which bind to the desired IL-IR, e.g., by ELISA or other assay. Antibodies which selectively recognize specific IL-IR embodiments may also be selected or prepared.
  • binding reagent is either labeled as described above, e.g., fluorescence or otherwise, or immobilized to a substrate for panning methods.
  • Nucleic acids may also be introduced into cells in an animal to produce the antigen, which serves to elicit an immune response. See, e.g., Wang, et al . (1993) Proc. Nat'l. Acad. Sci.
  • antibodies which may be useful to determine the combination of the IL-1RD8, IL-1RD9, or IL- 1RD10 with a functional beta subunit may be generated.
  • epitopes characteristic of a particular functional alpha/beta combination may be identified with appropriate antibodies .
  • IL-IRs Various fusion constructs are made with IL-IRs.
  • a portion of the appropriate gene is fused to an epitope tag, e.g., a FLAG tag, or to a two hybrid system construct. See, e.g., Fields and Song (1989) Nature 340:245-246.
  • the epitope tag may be used in an expression cloning procedure with detection with anti-FLAG antibodies to detect a binding partner, e.g., ligand for the respective IL-IR.
  • the two hybrid system may also be used to isolate proteins which specifically bind, e.g., to IL-1RD9. X. Structure activity relationship
  • Standard mutagenesis analysis is performed, e.g., by generating many different variants at determined positions, e.g., at the positions identified above, and evaluating biological activities of the variants. This may be performed to the extent of determining positions which modify activity, or to focus on specific positions to determine the residues which can be substituted to either retain, block, or modulate biological activity.
  • analysis of natural variants can indicate what positions tolerate natural mutations . This may result from population analysis of variation among individuals, or across strains or species. Samples from selected individuals are analyzed, e.g., by PCR analysis and sequencing. This allows evaluation of population polymorphisms .
  • An IL-IR can be used as a specific binding reagent to identify its binding partner, by taking advantage of its specificity of binding, much like an antibody would be used.
  • the binding receptor is a heterodimer of receptor subunits.
  • a binding reagent is either labeled as described above, e.g., fluorescence or otherwise, or immobilized to a substrate for panning methods .
  • the binding composition is used to screen an expression library made from a cell line which expresses a binding partner, i.e., ligand, preferably membrane associated.
  • a binding partner i.e., ligand, preferably membrane associated.
  • Standard staining techniques are used to detect or sort surface expressed ligand, or surface expressing transformed cells are screened by panning. Screening of intracellular expression is performed by various staining or immunofluorescence procedures. See also McMahan, et al . (1991) EMBO J. 10:2821-2832.
  • HBSS HBSS.
  • the slides may be stored at -80° C after all liquid is repoved.
  • 0.5 ml incubations are performed as follows. Add HBSS/saponin (0.1%) with 32 ⁇ l/ml of 1 M NaN3 for 20 min. Cells are then washed with HBSS/saponin IX. Add appropriate IL-IR or IL- lR/antibody complex to cells and incubate for 30 min. Wash cells twice with HBSS/saponin. If appropriate, add first antibody for 30 min. Add second antibody, e.g., Vector anti-mouse antibody, at 1/200 dilution, and incubate for 30 min.
  • second antibody e.g., Vector anti-mouse antibody
  • ELISA solution e.g., Vector Elite ABC horseradish peroxidase solution, and preincubate for 30 min.
  • Use e.g., 1 drop of solution A (avidin) and 1 drop solution B (biotin) per 2.5 ml HBSS/saponin. Wash cells twice with HBSS/saponin.
  • IL-IR reagents are used to affinity purify or sort out cells expressing a putative ligand. See, e.g., Sambrook, et al . or Ausubel, et al .
  • Another strategy is to screen for a membrane bound receptor by panning.
  • the receptor cDNA is constructed as described above.
  • the ligand can be immobilized and used to immobilize expressing cells. Immobilization may be achieved by use of appropriate antibodies which recognize, e.g., a FLAG sequence of an IL-IR fusion construct, or by use of antibodies raised against the first antibodies. Recursive cycles of selection and amplification lead to enrichment of appropriate clones and eventual isolation of receptor expressing clones.
  • Phage expression libraries can be screened by mammalian IL-IRs. Appropriate label techniques, e.g., anti-FLAG antibodies, will allow specific labeling of appropriate clones.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Cell Biology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

Nucleic acids encoding mammalian, e.g., human receptors, purified receptor proteins and fragments thereof. Antibodies, both polyclonal and monoclonal, are also provided. Methods of using the compositions for both diagnostic and therapeutic utilities are provided.

Description

HUMAN RECEPTOR PROTEINS; RELATED REAGENTS AND METHODS
FIELD OF THE INVENTION
The present invention relates to compositions and methods for affecting mammalian physiology, including, e.g., morphogenesis or immune system function. In particular, it provides nucleic acids, proteins, and antibodies, e.g., which regulate development and/or the immune system along with related reagents and methods . Diagnostic and therapeutic uses of these materials are also disclosed.
BACKGROUND OF THE INVENTION
Recombinant DNA technology refers generally to techniques of integrating genetic information from a donor source into vectors for subsequent processing, such as through introduction into a host, whereby the transferred genetic information is copied and/or expressed in the new environment. Commonly, the genetic information exists in the form of complementary DNA
(cDNA) derived from messenger RNA (mRNA) coding for a desired polypeptide product. The carrier is frequently a plasmid having the capacity to incorporate cDNA for later replication and/or expression in a host and, in some cases, actually to control expression of the cDNA and thereby direct synthesis of the encoded product in the host.
For some time, it has been known that the mammalian immune response is based on a series of complex cellular interactions, called the "immune network". Recent research has provided new insights into the inner workings of this network. While it remains clear that much of the immune response does, in fact, revolve around the network-like interactions of lymphocytes, macrophages, granulocytes, and other cells, immunologists now generally hold the opinion that soluble proteins, known as lymphokines, cytokines, or monokines, play critical roles in controlling these cellular interactions. Thus, there is considerable interest in the isolation, characterization, and mechanisms of action of cell modulatory factors, an understanding of which will lead to significant advancements in the diagnosis and therapy of numerous medical abnormalities, e.g., immune system disorders .
Lymphokines apparently mediate cellular activities in a variety of ways . They have been shown to support the proliferation, growth, and/or differentiation of pluripotential hematopoietic stem cells into vast numbers of progenitors comprising diverse cellular lineages which make up a complex immune system. Proper and balanced interactions between the cellular components are necessary for a healthy immune response. The different cellular lineages often respond in a different manner when lymphokines are administered in conjunction with other agents .
Cell lineages especially important to the immune response include two classes of lymphocytes: B-cells, which can produce and secrete immunoglobulins (proteins with the capability of recognizing and binding to foreign matter to effect its removal) , and T-cells of various subsets that secrete lymphokines and induce or suppress the B-cells and various other cells (including other T- cells) making up the immune network. These lymphocytes interact with many other cell types.
Another important cell lineage is the mast cell (which has not been positively identified in all mammalian species) , which is a granule-containing connective tissue cell located proximal to capillaries throughout the body. These cells are found in especially high concentrations in the lungs, skin, and gastrointestinal and genitourinary tracts. Mast cells play a central role in allergy-related disorders, particularly anaphylaxis as follows: when selected antigens crosslink one class of immunoglobulins bound to receptors on the mast cell surface, the mast cell degranulates and releases mediators, e.g., histamine, serotonin, heparin, and prostaglandins, which cause allergic reactions, e.g., anaphylaxis. Research to better understand and treat various immune disorders has been hampered by the general inability to maintain cells of the immune system in vitro. Immunologists have discovered that culturing many of these cells can be accomplished through the use of T- cell and other cell supernatants , which contain various growth factors, including many of the lymphokines.
The interleukin-1 family of proteins includes the IL-lα, the IL-lβ, the IL-1RA, and recently the IL-lγ (also designated Interferon-Gamma Inducing Factor, IGIF) . This related family of genes has been implicated in a broad range of biological functions . See Dinarello (1994) FASEB J. 8:1314-1325; Dinarello (1991) Blood 77:1627-1652; and Okamura, et al . (1995) Nature 378:88- 91.
From the foregoing, it is evident that the discovery and development of new soluble proteins and their receptors, including ones similar to lymphokines, should contribute to new therapies . A number of degenerative or abnormal conditions directly or indirectly involve development, differentiation, or function, e.g., of the immune system and/or hematopoietic cells. In particular, the discovery and understanding of novel receptors for lymphokine-like molecules which enhance or potentiate the beneficial activities of other lymphokines, would be highly advantageous . The present invention provides new receptors for ligands exhibiting similarity to interleukin-1 like compositions and related compounds, and methods for their use.
SUMMARY OF THE INVENTION
The present invention is directed to novel receptors related to IL-1 receptors and their biological activities. These receptors, e.g., primate or rodent, are designated IL-1 receptor like molecular structures, IL-1 Receptor DNAX designation 8(IL-1RD8), IL-1 Receptor DNAX designation 9(IL-1RD9) and IL-1 Receptor DNAX designation 10 (IL-IRDIO) . The invention includes nucleic acids coding for the polypeptides themselves and methods for their production and use. The nucleic acids of the invention are characterized, in part, by their homology to cloned complementary DNA (cDNA) sequences enclosed herein. In certain embodiments, the invention proviόTes" a composition of matter selected from the group of: an isolated or recombinant IL-1RD8 polypeptide comprising a segment of at least 12 contiguous amino acids of SEQ ID NO: 2 or 4, a natural sequence IL-1RD8 polypeptide comprising SEQ ID NO: 2 or 4, a fusion protein comprising IL-1RD8 sequence; an isolated or recombinant IL-1RD9 polypeptide comprising at least 12 contiguous amino acids of SEQ ID NO: 6, 8, 10, 12, 14, or 16; a natural sequence IL-1RD9 comprising SEQ ID NO: 6, 8, 10, 12, 14, or 16; a fusion protein comprising IL-1RD9 sequence; an isolated or recombinant IL-lRDIO polypeptide comprising at least 12 contiguous amino acids of SEQ ID NO: 18 or 20; a natural sequence IL-1RD10 comprising SEQ ID NO: 18 or 20; and a fusion protein comprising IL-1RD10 sequence. In various embodiments, the recombinant or isolated polypeptide comprises a segment identical to a corresponding portion of an IL-1RD8, as described, wherein: the number of contiguous amino acid residues is: at least 17 amino acids; at least 21 amino acids; or at least 25 amino acids; or to a corresponding portion of an IL-1RD9, as described, wherein the number of identical contiguous amino acid residues is: at least 17 amino acids; at least 21 amino acids; or at least 25 amino acids; or of an IL-lRDIO, as described, wherein the number of identical contiguous amino acid residues is: at least 17 amino acids; at least 21 amino acids; or at least 25 amino acids.
In polypeptide embodiments, the invention provides a composition of matter wherein the IL-1RD8 comprises a mature sequence shown in SEQ ID NO: 2 or 4; an IL-1RD9 that comprises a mature sequence shown in SEQ ID NO: 6, 8, 10, 12, 14 or 16; an IL-1RD10 that comprises a mature sequence shown in SEQ ID NO: 18 or 20; or the IL-1RD8, IL-1RD9, or IL-lRDIO polypeptide: is from a warm blooded animal, e.g., a primate, such as a human; comprises at least one polypeptide segment of SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18, or 20; exhibits a plurality of portions having segments identical to specific sequence identifiers; is a natural allelic variant of a primate IL-1RD8; a primate or rodent IL-1RD9; or a primate IL- 1RD10; has a length at least about 30 amino acids; exhibits at least two non-overlapping epitopes that are specific for: a primate IL-1RD8, a primate or rodent IL- 1RD9, or primate IL-1RD10; exhibits a sequence identity over a length of at least about 20 amino acids to: a primate IL-1RD8, a primate or rodent IL-1RD9, or a primate IL-1RD10; has a molecular weight of at least 100 kD with natural glycosylation; is a synthetic polypeptide; is attached to a solid substrate; is conjugated to another chemical moiety; is a 5-fold or less substitution from natural sequence; or is a deletion or insertion variant from a natural sequence. Certain preferred embodiments include compositions comprising: a sterile IL-1RD8, IL-1RD9, or IL-1RD10 polypeptide; or the IL-1RD8, IL-1RD9, or IL-1RD10 polypeptide and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration; a sterile IL-1RD8, IL-1RD9, or IL-1RD10 polypeptide; or the IL-1RD8, IL-1RD9, or IL-1RD10 polypeptide, as described, and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration.
Certain fusion proteins are provided, e.g., comprising: mature polypeptide sequence shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 or 20; a detection or purification tag, including a FLAG, His6, or Ig sequence; or sequence of another receptor protein. Kit embodiments include a kit comprising such a polypeptide, and: a compartment comprising the polypeptide; and/or instructions for use or disposal of reagents in the kit. In binding compound embodiments, the invention provides a binding compound comprising an antigen binding site from an antibody, which specifically binds to a natural: IL-1RD8, IL-1RD9, or IL-1RD10 polypeptide, wherein: the polypeptide is a primate or rodent protein; the binding compound is an Fv, Fab, or Fab2 fragment; the binding compound is conjugated to another chemical moiety; or the antibody: is raised to a polypeptide sequence of a mature polypeptide comprising a sequence sequence shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 or 20; is raised to a mature primate or rodent IL- 1RD8; is raised to a purified human IL-1RD8; is raised to a purified mouse IL-1RD9; is immunoselected; is a polyclonal antibody; binds to a denatured IL-1RD8, IL- 1RD9, or IL-1RD10; exhibits a Kd to antigen of at least
30 UM; is attached to a solid substrate, including a bead or plastic membrane; is in a sterile composition; or is detectably labeled, including a radioactive or fluorescent label; IL-1RD9 protein, wherein: the polypeptide is a primate or rodent protein; the binding compound is an Fv, Fab, or Fab2 fragment; the binding compound is conjugated to another chemical moiety; or the antibody: is raised against a polypeptide sequence of a mature polypeptide comprising a sequence sequence shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 or 20; is raised against a mature primate IL-1RD9; is raised to a purified human IL-1RD9 ; is immunoselected; is a polyclonal antibody; binds to a denatured IL-1RD9; exhibits a Kd to antigen of at least 30 μM; is attached to a solid substrate, including a bead or plastic membrane; is in a sterile composition; or is detectably labeled, including a radioactive or fluorescent label; IL-1RD10 protein, wherein: the polypeptide is a primate or rodent protein; the binding compound is an Fv, Fab, or Fab2 fragment; the binding compound is conjugated to another chemical moiety; or the antibody: is raised against a polypeptide sequence of a mature polypeptide comprising a sequence sequence shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 or 20; is raised against a mature primate IL-1RD10; is raised to a purified human IL-1RD10; is immunoselected; is a polyclonal antibody; binds to a denatured IL-lRDIO; exhibits a Kd to antigen of at least 30 μM; is attached to a solid substrate, including a bead or plastic membrane; is in a sterile composition; or is detectably labeled, including a radioactive or fluorescent label. Kits are provided, e.g., those comprising the binding compound, and: a compartment comprising the binding compound; and/or instructions for use or disposal of reagents in the kit. Preferably, the kit is capable of making a qualitative or quantitative analysis.
Other embodiments include a composition comprising: a sterile binding compound, or the binding compound and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration.
Nucleic acid embodiments include an isolated or recombinant nucleic acid encoding a polypeptide or fusion protein, wherein: the IL-1RD8, IL-1RD9, or IL-lRDIO is from a mammal; said nucleic acid: encodes an antigenic polypeptide sequence sequence shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 or 20; encodes a plurality of antigenic polypeptide sequences sequence shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 or 20; exhibits at least about 30 nucleotides to a natural cDNA encoding the segment; is an expression vector; further comprises an origin of replication; is from a natural source; comprises a detectable label; comprises synthetic nucleotide sequence; is less than 6 kb, preferably less than 3 kb; is from a mammal, including a primate; comprises a natural full length coding sequence; is a hybridization probe for a gene encoding said IL-1RD8, IL- 1RD9, or IL-lRDIO; comprises a plurality of nonoverlapping segments of at least 15, 18, 21, or 25 nucleotides shown in SEQ ID NO: 1, 3, 5, 7, 9 11, 13, 15, 17, 19; or is a PCR primer, PCR product, or mutagenesis primer. The invention further provides a cell comprising such a recombinant nucleic acid, e.g., where the cell is: a prokaryotic cell; a eukaryotic cell; a bacterial cell; a yeast cell; an insect cell; a mammalian cell; a mouse cell; a primate cell; or a human cell. Certain kit embodiments include a comprising the nucleic acid" and: a compartment comprising the nucleic acid; a compartment further comprising: a primate IL-1RD8, a primate or rodent IL-1RD9, or a primate IL-1RD10 polypeptide; and/or instructions for use or disposal of reagents in the kit. Preferably, the kit is capable of making a qualitative or quantitative analysis.
In other nucleic acid embodiments, the nucleic acid is one which: hybridizes under wash conditions of 40° C and less than 2M salt to either SEQ ID NO: 1, 3, 5, 7, 9, 11, 13, 15, 17, or 19; or exhibits identity over a stretch of at least about 30 nucleotides to a primate IL- 1RD8, a primate or rodent IL-1RD9, or a primate IL-1RD10. In various preferred embodiments : the wash conditions are: at 45° C and/or 500 mM salt; at 55° C and/or 150 mM salt; or the stretch is at least 55 nucleotides; or at least 75 nucleotides.
Methods of modulating physiology or development of a cell or tissue culture cells are provided, e.g., comprising contacting the cell with an agonist or antagonist of a primate IL-1RD8, a primate or rodent IL- 1RD9, or a primate IL-1RD10. Preferably, the cell is transformed with a nucleic acid encoding either IL-1RD8, IL-1RD9, or IL-1RD10, and another IL-1R.
DETAILED DESCRIPTION OF THE INVENTION I . General
The present invention provides the amino acid sequence and DNA sequence of mammalian, herein, e.g., primate and rodent IL-1 receptor-like molecules, these molecules IL-1 Receptor DNAX designation 8(IL-1RD8), IL-1 Receptor DNAX designation 9(IL-1RD9) and IL-1 Receptor. DNAX designation 10(IL-1RD10) having particular defined properties, both structural and/or biological. These embodiments increase the number of members of the human IL-1 receptor-like family from 7 to at least 10. These receptors have been numbered internally as DNAX designations Dl, D2 , D3 , D4, D5, D6, and now D8, D9, and D10, and are referred to as IL-lRDl through D10. Various cDNAs encoding these molecules were obtained from" primate, e.g., human, or rodent, e.g., mouse, cDNA sequence libraries. Other primate, rodent, or other mammalian counterparts would also be desired.
Some of the standard methods applicable are described or referenced, e.g., in Maniatis, et al. (1982) Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor Press; Sambrook, et al. (1989) Molecular Cloning: A Laboratory Manual. (2d ed. ) , vols. 1-3, CSH Press, NY; Ausubel, et al . Biology, Greene Publishing Associates, Brooklyn, NY; or Ausubel, et al. (1987 and periodic supplements) Current Protocols in Molecular Biology, Greene/Wiley, New York; each of which is incorporated herein by reference.
A partial nucleotide and corresponding amino acid sequence of a human IL-1RD8 coding segment is shown in
SEQ ID NO: 1 and 2, respectively. Supplemental human IL- 1RD8 nucleotide and corresponding sequence is provided in SEQ ID NO: 3 and 4, respectively.
Similarly for primate IL-1RD9, partial nucleotides (SEQ ID NO: 5) and corresponding amino acid sequences (SEQ ID NO: 6) of a primate IL-1RD9 coding segment are provided. Supplemental primate IL-1RD9 is provided in SEQ ID NO: 7, 8, 9, and 10. Rodent embodiments of IL- 1RD9 are provided in SEQ ID NO: 11, 12, with supplemental IL-1RD9 rodent sequence in SEQ ID NO: 13, 14, 15, and 16.
For an embodiment of human IL-1RD10, a partial nucleotide and corresponding partial amino acid sequence are provided in SEQ ID NO: 17 and 18, respectively, with supplemental human IL-1RD10 nucleotide and corresponding partial amino acid sequence provided in SEQ ID NO: 19 and 20, respectively.
Some sequences provided lack some portions of these receptors, as suggested by alignment of sequences shown in Tables 1-4) . Note the alignment of IL-1RD10 with IL- 1RD8 and D3s, which are alpha type receptor subunits . Table 4 exhibits alignment of primate and rodent IL-1RD9.
It is to be understood that this invention is not limited to the particular methods, compositions and receptors specifically embodied herein, as such met-ods, compositions and receptors may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which is only limited by the appended claims .
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by a person of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety including all figures, graphs, and drawings.
Table 1
Alignment of the extracellular domains of various IL-l s. hl -
1RD10 is SEQ ID NO: 20; hIL-lRD8 is SEQ ID NO: 3; mI -lRD3 is
GenBank X85999; hIL-lRD6 is GenBank U49065; rI -lRD6 is GenBank U49066; mIL-lRD4 is GenBank Y07519 and GenBank D13695; hI -lRD4 is
GenBank D12763; hIL-lRD2 is GenBank X59770; mIL-lRD2 is GenBank
X59769; hIL-lRD5 is GenBank U43672; mI -lRD5 is GenBank U43673; mlL-lRDl is GenBank 20658, M29752; hI -lRDI is GenBank X16896; cIL-lRDl is GenBank 86325; and hFGR4 is GenBank P22455. Other species counterparts may be obtained from public sequence databases . mI -lRD3 MGLL WY MSLSFYG ILQSHASERC DD LDTMR.. hI -lRD6 M WS LLCGLSI ALPLSVTADG CKDIFMKN.. rIL-lRD6 MG PPL FCWVSF V P FVAAGN CTDVYMH3H.. mIL-lRD4 MI DRQRMGL AL AILTLPMY T VTEGSKSS .. hIL-lRD4 MG FWILAI TI MYSTAAKFSK QS hIL-lRD2 MLRLYV LVMGVSAFTL QPAAHTGAAR SCRFRGRHYK mIL-lRD2 MFI LV VTG VSAFTTPTW HTGKVSESPI TSEKPTVHGD NCQFRGREFK hlL-lRDlO hI -lRD5 MNCRE LPLT WVLIS VSTAESCTSR PHITWE... mIL-lRD5 MHHEE LILT CI IV KSASKSCIHR SQIHWE... mlL-lRDl MENMK V LG IC MV PL SLEIDVC TEYPNQIVL.F hIL-lRDI MK V LRLICFIA ISSLEADK CKEREEKII cIL-lRDl MHKMT STFLLIGHLI LLIPLFSAEE CVICNYFVLV hIL-lRD8 M KPPFL ALW CSWSTN KM VSKRNSVDGC IDWSVDLKTY hFGR4 ...MRLL AL LGVLLSVPGP PVLSLEASEE VELEPC APS LEQQEQELTV mIL-lRD3 QIQVFEDEPA RIKCPLFEHF LKYNYSTAHS SGLTLLWYWT RQDRDLEEPI hI -lRD6 .EILSASQPF AFNCTFPPI TS GEVSVTWYKN SSKIPV rI -lRD6 .EMISEGQPF PFNCTYPPV TN GAVNLTWHRT .... PSKSPI mIL-lRD4 ..WGLENEAL IVRCPQRG R STYPVEWYYS ....DTNESI hI -lRD4 ..WGLENEAL IV3RCPRQG K PSYTVDWYYS .....QTNKSI hI -lRD2 REFRLEGEPV ALRCPQVPYW LWA SVS PRINLTWHKN DSARTV mI -lRD2 SELRLEGEPV VLRCPLAPHS DIS SS SHSFLTWSKL DSSQLI hlL-lRDlO hIL-lRD5 GEPFY LKHCSCSLAH El ETTTKSWYKS . SGSQEHV mI -lRD5 GEPFY LKPCGISAPV HRN ETATMRWFKG . SASHEYR mlL-lRDl LSV...NEID IRKCPLTPN KM HGDTIIWYKN .. DSKTPI hIL-lRDI VSS..ANEID VRPCPLNPN E HKGTITWYKD .. DSKTPV cIL-lRDl GEPT AISCPVITL PMLH SDYNLTWYRN .. GSNMPI hIL-lRD8 ..MALAGEPV RVKCALFYSY IRTNYSTAQS TGLRLMWYKN KGDLEEPI hFGR4 .... ALGQPV RLCCGRAERG G HWYKE .. GSRLAP mIL-lRD3 NFRLP ENRI SKEKDVLWFR PTLLNDTGNY TCMLRNTTYC SKVAFPLEW hIL-lRD6 SKII. QSRI HQDETWILFL PMEWGDSGVY QCVIKGRDSC HRIHVNLTVF rIL-lRD6 SINR. HVRI HQDQSWILFL PLALEDSGIY QCVIKDAHSC YRIAINLTVF mIL-lRD4 PTQK. RNRI FVSRDRLKFL PARVEDSGIY ACVIRSPNLN KTGYLNVTIH hIL-lRD4 PTQE. RNRV FASGQLLKFL PAEVADSGIY TCIVRSPTFN RTGYANVTIY hIL-lRD2 PGEE. ETRM WAQDGALWLL PALQEDSGTY VCTTRNASYC DKMSIELRVF mIL-lRD2 PRDEP ..RM WVKGNILWIL PAVQQDSGTY ICTFRNASHC EQMSVELKVF hIL-lRDIO hIL-lRD5 ELNPRSSSRI ALHDCVLEFW PVELNDTGSY FFQMKN.. YT QKWKLNVIRR mIL-lRD5 ELNNRSSPRV TFHDHTLEFW PVEMEDEGTY ISQVGN..DR RNWTLNVTKR mlL-lRDl SADR..DSRI HQQNEHLWFV PAKVEDSGYY YCIVRNSTYC LKTKVTVTVL hIL-lRDI STEQ..ASRI HQHKEKLWFV PAKVEDSGHY YCWRNSSYC LRIKISAKFV cIL-lRDl TTER..RARI HQRKGLLWFI PAALEDSGLY ECEVRSLNRS KQKIINLKVF hIL-lRD8 IFS...EVRM SKEEDSIWFH SAEAQDSGFY TCVLRNSTYC MKVSMSLTVA hFGR4 AG RV RGWRGRLEIA SFLPEDAGRY LCLARGSMIV LQNLTLITGD mIL-lRD3 QK DSC FNSAMRFPVH KMYIEHGIHK hIL-lRD6 EK HWCDTSIGG LP.NLSDEYK QILHLGKDDS rIL-lRD6 RK HWCDSSNEE SSINSSDEYQ QWLPIGKSGS mIL-lRD4 KK PPSCN . IPDY.LMYS TVRGSDKNFK hIL-lRD4 KK QSDCN -VPDY.LMYS TVSGSEKNSK hIL-lRD2 EN TDA FLPFI .. SYP QILTLSTSGV mIL-lRD2 KN TEA SLPHV.. SYL QISALSTTGL hIL-lRDIO hIL-lRD5 NK HSC FTERQ..VTS KIVEVKKFFQ mIL-lRD5 NK HSC FSDKL..VTS RDVEVNKSLH mlL-lRDl EN DPGIC .YSTQ.ATFP QRLHIAGDGS hIL-lRDI EN EPNLC .YNAQ.AIFK QKLPVAGDGG CIL-lRDl KN DNGLC -FNGE.MKYD QIVKSANAGK ML-1RD8 EN ESGLC .YNSR.IRYL EKSEVTKRKE hFGR4 SLTSSNDDED PKSHRDPSNR HSYPQQAPYW THPQRMEKKL HAVPAGNTVK mlL- 1RD3 ITCPNVDGYF P . SSVKPSVT WYKGCTEIVD FH3N...VLPE GMNLSFFIPL hlL- 1RD6 LTCHLHFPKS ... CVLGPIK WYKDCNEIKG E RFT VLETRLLVSN rlL- 1RD6 LTCHLYFPES ... CVLDSIK WYKGCEEIKV S KKFC PTGTKLLVNN mlL- 1RD4 ITCPTIDLY . ...NWTAPVQ WFKNCKALQE P RFR AHRSYLFIDN hlL- 1RD4 IYCPTIDLY. ...NWTAPLE WFKNCQALQG S RYR AHKSFLVIDN hlL- 1RD2 LVCPDLSEFT R.DKTDVKIQ WYKDSLLLDK DNEK..FLSV RGTTHLLVHD mlL- 1RD2 LVCPDLKEFI S . SNADGKIQ WYKGAILLDK GNKE .. FLSA GDPTRLLISN hlL- 1RD10 hlL- 1RD5 ITCENSYYQ . ... TLVNSTS LYKNCKKLLL ENN.. ..KNP TIKKNAEF . mlL- 1RD5 ITCKNPNYE . ... ELIQDTW LYKNCKEISK TPRI... LKD AEFGDAEF .. mlL- 1RD1 LVCPYVSYFK DENNELPEVQ WYKNCKPLLL DN....VSFF GVKDKLLVRN hlL- 1RD1 LVCPYMEFFK NENNELPKLQ WYKDCKPLLL DN.... IHFS GVKDRLIVMN cIL- 1RD1 IICPDLENFK DEDNINPEIH WYKECKSGFL EDKR..LVLA EGENAILILN hlL- 1RD8 ISCPDMDDFK KSD.QEPDW WYKECKPKMW R.....SIII QKGNALLIQE hFGR4 FRCPAAG . . . . NPTPTIR WLKDGQAFHG ENRIGGIRLR HQHWSLVMES mIL-lRD3 VSNN..GNYT CWTYPENGR LFHLTRTVTV KWGS . PKDA LPPQIYSPND hIL-lRD6 VSAEDRGNYA CQAILTHSGK QYEVLNGITV SITERAGYGG SVP.KIIYPK rIL-lRD6 IDVEDSGSYA CSARLTHLGR IFTVRNYIAV NTKE.VGSGG RIP.NITYPK mIL-lRD4 VTHDDEGDYT CQFTHAENGT NYIVTATRSF TVE . EKGFS . MFPVITNPPY hIL-lRD4 VMTEDAGDYT CKFIHNENGA NYSVTATRSF TVKDEQGFS . LF V3t,GAPAQ hIL-lRD2 VALEDAGYYR CVLTFAHEGQ QYNITRSIEL RIKKK..KEE TIPVIISP.. mIL-lRD2 TSMDDAGYYR CVMTFTYNGQ EYNITRNIEL RVKGT..TTE PIPVIISP.. hIL-lRDIO ... EFG.. S CEL .. KYGGF V..VRRTTEL TVTAPLTDKP PKLLYPMESK hIL-lRD5 ... EDQGYYS CVHFLHHNGK LFNITKTFNI TIVED .. RSN IVPVLLGP . K mIL-lRD5 ...GDEGYYS CVFSVHHNGT RYNITKTVNI TVIEG..RSK VTPAILGP . K mlL-lRDl VAEEHRGDYI CRMSYTFRGK QYPVIRVIQF ITIDE ..NKR DRPVILSP . R hIL-lRDI VAEKHRGNYT CHASYTYLGK QYPITRVIEF ITLEE ..NKP TRPVIVSP.A CIL-lRDl VTIQDKGNYT CRMVYTYMGK QYNVSRTMNL EVKES .. PLK MRPEFIYP. N hIL-lRD8 VQEEDGGNYT CEL .. KYEGK L..VRRTTEL KVTALLTDKP PKPLFPMENQ hFGR4 WPSDRGTYT CLVENAVGSI RYNYLLDVLE RSPH..RPIL QAGLPANT . mIL-lRD3 RWYEKEPGE ELVIPCKVYF SFIMD.SHNE VWWTIDGKKP . DDVTVDITI hIL-lRD6 NHSIEVQLGT TLIVDCNVTD TK..D.NTNL RCWRVNNTLV DDYYDESKRI rIL-lRD6 NNSIEVQLGS TLIVDCNITD TK..E.NTNL RCWRVNNTLV DDYYNDFKRI mIL-lRD4 NHTMEVEIGK PASIACSACF GKGSH.FLAD VLWQINKTW GNFGEARIQE hIL-lRD4 NEIKEVEIGK NANLTCSACF GKGTQ.FLAA VLWQLNGTKI TDFGEPRIQQ hIL-lRD2 LKTISASLGS RLTIPCKVFL GTGTP . LTTM LWWTANDTHI ESAYPGGRV mIL-lRD2 LETIPASLGS RLIVPCKVFL GTGTS . SNTI VWWLANSTFI SAAYPRGRV hIL-lRDIO LTIQETQLGD SANLTCRAFF GYSGD.VSPL lYWMKGEKFI EDLDENRVWE hIL-lRD5 LNHVAVELGK NVRLNCSALL N EEDV IYWMFGEENG .. SDPNIHE mIL-lRD5 CEKVGVELGK DVELNCSASL N KDDL FYWSIRKEDS .. SDPNVQE mlL-lRDl NE IEADPGS MIQLICNVTG Q FSDL VYWKWNGSEI EWNDPFLAE hIL-lRDI NETMEVDLGS QIQLICNVTG Q LSDI AYWKWNGSVI DEDDPVLGE CIL-lRDl NNTIEVELGS HWMECNVSS GV....YGLL PYWQVNDEDV DSFDSTYRE hIL-lRD8 PSVIDVQLGK PLNIPCKAFF GFSGE.SGPM lYWMKGEKFI EELAGHIRE hFGR4 AWGS DVELLCKVYS DA...QPHIQ ..WLKHIVIN GSSFGA..DG mIL-lRD3 NESVSYSSTE D..ETRTQIL SIKKVTPEDL RRNYVCHARN TKGEAEQAAK hIL-lRD6 REGVETHVSF REHNLYTVNI TFLEVKMEDY GLPFMCHAG . ...VSTAYII rIL-lRD6 QEGIETNLSL RNHILYTVNI TFLEVKMEDY GHPFTCHAA. ...VSAAYII mIL-lRD4 EEGRNESSSN D. DCLTSVL RITGVTEKDL SLEYDCLALN LHGMIRHTIR hIL-lRD4 EEGQNQSFSN G. LACLDMVL RIADVKEEDL LLQYDCLALN LHGLRRHTVR hIL-lRD2 TEGPRQEYSE NNENYIEVPL IFDPVTREDL HMDFKCWHN TLSFQTLRTT mIL-lRD2 TEGLHHQYSE NDENYVEVSL IFDPVTREDL HTDFKCVASN PRSSQSLHTT hIL-lRDIO SDIRILKEHL G.EQEVSISL IVDSVEEGDL .GNYSCYVEN GNGRRHASVL hIL-lRD5 EKEMRIMTPE G . KWHASKVL RIENIGESNL NVLYNCTVAS TGGTDTKSFI mIL-lRD5 DRKETTTWIS EGKLHASKIL RFQKITENYL NVLYNCTVAN EEAIDTKSFV mlL-lRDl DYQFVEHPST KRKYTLITTL NISEVKSQFY RYPFICWKN TNIFES HVQ hIL-lRDI DYYSVENPAN KRRSTLITVL NISEIESRFY KHPFTCFAKN THGIDAAYIQ CIL-lRDl QFYEEGMPHG .. IAVSGTKF NISEVKLKDY AYKFFCHFIY DSQEFTSYIK hIL-lRD8 GEIRLLKEHL G . EKEVELAL IFDSWEADL AN.YTCHVEN RNGRKHASVL hFGR4 FPYVQVLKTA DINSSEVEVL YLRNVSAED . AGEYTCLAGN SIGLSYQSAW mIL- -1RD3 VKQKV. PPRYTVELAC GFGATVFLW VLIWY hIL- ■1RD6 LQLP .. PDFRAYLIGG LIALVAVAVS YIYNIFKI DIVLWY rIL- •1RD6 LKRP.. PDFRAYLIGG LMAFLLLAVS ILYIYNTFKV DIVLWY mIL- -1RD4 LRRK.. PSKECPSHIA IYYIVAGCSL LLMFINVLVI VL hIL' -1RD4 LSRK.. PSKEC hIL- -1RD2 VKEASS . FSWGIVLA PLSLAFLVLG GIWM mIL- -1RD2 VKEVSS .TFSWSIALA PLSLIILWG AIW. hIL- -1RD10 LHKREL .MYTVELAGG LGAILLLLVC LVTIYKCY hIL- •1RD5 LVRKADMADI P ..GHVFTRG MIIAVLILVA WCLVTVCVI Y mIL- -1RD5 LVRKEIPDIP ... GHVFTGG VTVLVLASVA AVCIVILCVI Y mIL- -1RD1 LIYP V PDFKNYLIGG FIILTATIVC CVCIY hIL- -1RD1 LIYP V TNFQKHMIGI CVTLTVIIVC SVFIY cIL- -1RD1 LEHP V QNIRGYLIGG GISLIFLLFL ILIVY hIL- -1RD8 LRKKDL . IYKIELAGG LGAIFLLLVL LWIYKCY hFGR4 LTVLP . . . . E EDPTWTAAAP EARYTDIILY ASGSLALAVL LLLAGLY
Table 2
Alignment of the intracellular domains of various IL-lRs. hIL-lRD9 is SEQ ID NO: 8; mIL-lRD9 is SEQ ID NO: 14; hIL-lRDI is™ GenBank
X16896; hIL-lRD6 is GenBank U49065; mIL-lRD3 is GenBank X85999; huIL-lRD8 is SEQ ID NO: 3; and mIL-lRD4 is GenBank Y07519.
HuIL -1RD1 SDGKTYDAYI LYPKTVGEG. ..STSDCDIF VFKVLPEVLE KQCGYKLFIY HuIL -1RD6 VDGKLYDAYV LYPKPHKES . ..QRHAVDAL VLNILPEVLE RQCGYKLFIF MoIL--1RD3 LDGKEYDIYV SYAR NVEEEEF VLLTLRGVLE NEFGYKLCIF HuIL--1RD8 DDNKEYDAYL SYTKVDQDTL DCDNPEEEQF ALEVLPDVLE KHYGYKLFIP HuIL--1RD5 TDGKTYDAFV SYLKECRP .. .. ENGEEHTF AVEILPRVLE KHFGYKLCIF MoIL--1RD9 HuIL--1RD9 KYGYSLCLL MoIL--1RD4 NDGKLYDAYI IYPRVFRGS . AAGTHSVEYF VHHTLPDVLE NKCGYKLCIY
HuIL- -1RD1 GRDDYV.GED IVEVINENVK KSRRLIIILV RETSGFSWLG GSSEEQIAMY HuIL--1RD6 GRDEFP . GQA VANVIDENVK LCRRLIVIW PESLGFGLLK NLSEEQIAVY MoIL--1RD3 DRDSLPGGIV TDETLS . FIQ KSRRLLWLS PNYVLQG . TQ ALLELKAGLE HuIL--1RD8 ERDLIPSG.T YMEDLTRYVE QSRRLIIVLT PDYILRR . GW SIFELESRLH HuIL--1RD5 ERDWPGGAV VDEIHS.LIE KSRRLIIVLS KSYMSN ... E VRYELESGLH MoIL--1RD9 DRDVTP .GGV YADDIVSIIK KSRRGIFILS PSYLNG ... P RVFELQAAVN HuIL--1RD9 ERDVAP .GGV YAEDIVSIIK RSRRGIFILS PNYVNG ... P SIFELQAAVN MoIL--1RD4 GRDLLP . GQD AATWESSIQ NSRRQVFVLA PHMMHSK..E FAYEQEIALH
HuIL- -1RD1 NALVQDGIKV VLLELEKIQ DYEKM PESIKFIKQK HGAIRWSGDF HuIL--1RD6 SALIQDGMKV ILIELEKIE DYTVM PESIQYIKQK HGAIRWHGDF MoIL--1RD3 NMASRGNINV ILVQYKAVK. ...DMKVKEL KRAKTVLT .. ..VIKWKGEK HuIL--1RD8 NMLVSGEIKV ILIECTELKG KVNCQEVESL KRSIKLLS .. .. LIKWKGSK HuIL--1RD5 EALVERKIKI ILIEFTPVT DFTFL PQSLKLLKSH R.VLKWKADK MoIL--1RD9 LALVDQTLKL ILIKFCSFQ EPESL PYLVKKALRV LPTVTWKGLK HuIL--1RD9 LALDDQTLKL ILIKFCYFQ EPESL PHLVKKALRV LPTVTWRGLK MoIL-•1RD4 SALIQNNSKV ILIEMEPLG. EASRLQVGDL QDSLQHLVKI QGTIKWREDH
HuIL- -1RD1 TQGPQSAKTR FWKNVRYHMP VQRRSPSSKH HuIL-•1RD6 TEQSQCMKTK FWKTVRYHMP PRRCRPFLRS MoIL--1RD3 SKYPQ ...GR FWKQLQVAMP VKKSPRWSSN HuIL-•1RD8 SSKLN. SK FWKHLVYEMP IKKKEMLPRC HuIL--1RD5 SLSYN. SR FWKNLLYLMP AKTVKPGRDE MoIL--1RD9 SVHAS. SR FWTQIRYHMP VKNSNRFMFN HuIL--1RD9 SVPPN. SR FWAKMRYHMP VKNSQGFTWN MoIL--1RD4 VADKQSLSSK FWKHVRYQMP VPERASKTAS
Table 3 Alignment of primate IL-1RD8 and primate IL-1RD10.
RD8 MKPPFLLALV VCSWSTNLK MVSKRNSVDG CIDWSVDLKT YMALAGEPVR R1D0
RD8 VKCALFYSYI RTNYSTAQST GLRLMWYKNK GDLEEPIIFS EVRMSKEEDS RD10
RD8 IWFHSAEAQD SGFYTCVLRN STYCMKVSMS LTVAENESGL CYNSRIRYLE RD10
RD8 KSEVTKRKEI SCPDMDDFKK SDQEPDWWY KECKPKMWRS IIIQKGNALL RD10
RD8 IQEVQEEDGG NYTCELKYEG KLVRRTTELK VTALLTDKPP KPLFPMENQP
RD10 EFG. .TSCELKYGG FWRRTTELT VTAPLTDKPP KLLYPMESKL
RD8 SVIDVQLGKP LNIPCKAFFG FSGESGPMIY WMKGEKFIEE LAG.HIREGE
RD10 TIQETQLGDS ANLTCRAFFG YSGDVSPLIY WMKGEKFIED LDENRVWESt)
RD8 IRLLKEHLGE KEVELALIFD SWEADLANY TCHVENRNGR KHASVLLRKK RD10 IRILKEHLGE QEVSISLIVD SVEEGDLGNY SCYVENGNGR RHASVLLHKR
RD8 DLIYKIELAG GLGAIFLLLV LLWIYKCYN lELMLFYRQH FGADETNDDN
RD10 ELMYTVELAG GLGAILLLLV CLVTIYKCYK IEIMLFYRNH FGAEELDGDN RD8 KEYDAYLSYT KVDQDTLDCD NPEEEQFALE VLPDVLEKHY GYKLFIPERD
RD10 KDYDAYLSYT KVDPDQWNQE TGEEERFALE ILPDMLEKHY GYKLFIPDRD
RD8 LIPSGTYMED LTRYVEQSRR LIIVLTPDYI LRRGWSIFEL ESRLHNMLVS
RD10 LIPTGTYIED VARCVDQSKR LIIVMTPN3TV VRRGWSIFEL ETRLRNMLVT
RD8 GEIKVILIEC TELKGKVNCQ EVESLKRSIK LLSLIKWKGS KSSKLNSKFW RD10 GEIKVILIEC SELRGIMNYQ EVEALKHTIK LLTVIKWHGP KCNKLNSKFW
RD8 KHLVYEMPIK KKEMLPRCHV LDSAEQGLFG ELQPIPSIAM TS . SATLVS RD10 KRLQYEMPFK RIEPITHEQA LDVSEQGPFG ELQTVSAISM AAATSTALAT
RD8 SQADLP . EFH PS .. DSMQIR HCCRGYKHEI PA . LPVPS LGNHHTYCNL
RD10 AHPDLRSTFH NTYHSQMRQK HYYRSYEYDV PPTGTLPLTS IGNQHTYCNI RD8 PLTLLNGQLP LNNTLKD..T QEFHRNSSLL PLSSKELSFT SDIW
RD10 PMTLINGQRP QTKSSREQNP DEAHTNSAIL PLLPRETSIS SVIW
Table 4 Alignment and comparison of primate and rodent IL-1RD9. hIL-lRD9 MLCLGWIFLWLVAGERIKGFNISGCSTKKLLWTYSTRSEEEFVLFCDLPE mIL-1RD9 MLCLGWVFLWFVAGEKTTGFNHSACATKKLLWTYSARGAENFVLFCDLQE ****** *** **** *** * * ********* * * ******* ••* hIL-1RD9 PQKSHFCHRNRLSPKQVPEHLPFMGSN-DLSDVQWYQQPSNGDPLEDIRK mIL-lRD9 LQEQKFSHASQLSPTQSPAHKPCSGSQKDLSDVQWYMQPRSGSPLEEISR * * * *** * * * * ** ******** ** * *** * hIL-lRD9 SYPHIIQDKCTLHFLTPGVNNSGSYICRPKMIKSPYDVACCVKMILEVKP mIL-lRD9 NSPHMQSE-GMLHILAPQTNSIWSYICRPR-IRSPQDMACCIKTVLEVKP ** ** *.* * ****** * ** * *** * ****** hIL-1RD9 QTNASCEYSASHKQDLLLGSTGSISCPSLSCQSDAQSPAVTWYKNGKLLS mIL-lRD9 QRNVSCGNTAQDEQVLLLGSTGSIHCPSLSCQSDVQSPEMTWYKDGRLLP * * ** * * ********* ********* *** **** * ** hIL-1RD9 VERSNRIWDEVYDYHQGTYVCDYTQSDTVSSWTVRAWQVR IVGDTKL mIL-lRD9 EHKKNPIEMADIYVFNQGLYVCDYTQSDNVSSWTVRAWKVRTIGKDINV * * * ** ********* ********** **** * hIL-lRD9 KPDILDPVEDTLEVELGKPLTISCKARFGFERVFNPVIKWYIKDSDLEWE mIL-lRD9 KPEILDPITDTLDVELGKPLTLPCRVQFGFQRLSKPVIKWYVKESTQEWE ** **** *** ******** * *** * ****** * * *** hIL-lRD9 VSVPEAKSIKSTLKDEIIERNIILEKVTQRDLRRKFVCFVQNSIGNTTQS mIL-lRD9 MSVFEEKRIQSTFKNEVIERTIFLREVTQRDLSRKFVCFAQNSIGNTTRT ** * * * ** * * *** * * ****** ****** ******** hIL-1RD9 VQLKEKRGWLLYILLGTIGTLVAVLAASALLYRHWIEIVLLYRTYQSKD mIL-1RD9 IRLRKKEEWFVYILLGTALMLVGVLVAAAFLYWYWIEWLLCRTYKNKD * * ** ****** ** ** * * ** *** *** *** ** hIL-lRD9 QTLGDKKDFDAFVSYAKWSSFPSEATSSLSEEHLALSLFPDVLENKYGYS mIL-lRD9 ETLGDKKEFDAFVSYSNWSSPETDAVGSLSEEHLALNLFPEVLEDTYGYR ****** ******* *** * ********* *** *** *** hIL-lRD9 LCLLERDVAPGGVYAEDIVSIIKRSRRGIFILSPNYVNGPSIFELQAAVN mIL-lRD9 LCLLDRDVTPGGVYADDIVSIIKKSRRGIFILSPSYLNGPRVFELQAAVN **** *** ****** ******* ********** * *** ******** hIL-lRD9 LALDDQTLKLILIKFCYFQEPESLPHLVKKALRVLPTVTWRGLKSVPPNS mIL-lRD9 LALVDQTLKLILIKFCSFQEPESLPYLVKKALRVLPTVTWKGLKSVHASS *** ************ ******** ************** ***** * hIL-1RD9 RFWAKMRYHMPVKNSQGFTWNQLRITSRIFQ WKGLSRTETTGR mIL-lRD9 RFWTQIRYHMPVKNSNRFMFNGLRIFLKGFSPEKDLVTQKPLEGMPKSGN *** ********* * * *** * * * * hIL-lRD9 SSQPKEW mIL-lRD9 DHGAQNLLLYSDQKRC Structural analysis of the primate IL-lRDlO sequence (SEQ ID NO: 18 and 20) , in comparison with other IL-lRs, shows characteristic features exist, which are conserved with the IL-lRDIO embodiment described herein. For example, there are characteristic Ig domains, and" " subdomains therein. The corresponding regions of the IL- 1RD10 (SEQ ID NO: 18 and 20) are about: f2 to gly7; g2 from vallO to thr23; a3 from leu30 to met33; a3 ' from thr38 to gln40; b3 from ala48 to ala54; c3 from pro64 to lys70; c3 ' from glu72 to phe74; d3 from val83 to lys92; e3 from gln98 to vall06; and f3 from tyr117 to trpl26.
Structural analysis of the rodent IL-1RD9 sequence (SEQ ID NO: 12, 14, and 16), in comparison with other IL- lRs, shows characteristic features exist (see Tables) . For example, there are characteristic Ig domains, and subdomains therein. The corresponding regions of the IL- 1RD9 (SEQ ID NO: 12, 14, and 16) are about: Igl domain from glylδ to prol27, with cysl05 probably linked to cys52 (or possibly cys48) ; Ig2 domain from glyl28 to pro229, with cysl53 probably linked to cysl99; and the Ig3 domain from glu230 to lys333, with cys251 probably linked to cys315; transmembrane segment from val336 to tyr360; THD domain from gly381 to val539; conserved trp residues probably correspond to residues 64, 169, and 267. Alignment of the IL-1RD9 embodiments is shown in Table 4. There are characteristic beta strand sections, and alpha helical structures, as described above for IL- 1RD10. The corresponding segments of the human IL-1RD9 sequence (SEQ ID NO: 6, 8, and 10) are roughly: βB from gly3 to val13; α2 from prol5 to lys28; βc from ser30 to ser46; α3 from ile47 to gln61; βD from lys64 to glu75; α4 from glu77 to leu87; βE from val93 to leu98; and α5 from argl06 to valll7. The corresponding segments of the mouse IL-1RD9 sequence (SEQ ID NO: 12, 14, and 16) are roughly: α3 to glnlO; βD from lys13 to glu24; α4 from glu26 to leu36; βE from va42 to leu47; and α5 from arg55 to val66.
As used herein, the terms IL-1 like receptor D8 (IL- 1RD8), IL-1 like receptor D9 (IL-1RD9), or IL-1 like receptor D10 (IL-lRDIO) shall be used to describe a polypeptide comprising a segment having or sharing the amino acid sequence shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 and 20, or a substantial fragment thereof. The invention also includes a polypeptide variation" of the respective IL-1RD8, IL-1RD9, IL-1RD10 alleles whose sequences are provided, e.g., a mutein or soluble extracellular or intracellular construct. Typically, such agonists or antagonists will exhibit less than about 10% sequence differences, and thus will often have between 1- and 11-fold substitutions, e.g., 2-, 3-, 5-, 7-fold, and others. It also encompasses allelic and other variants, e.g., natural polymorphic, of the polypeptide described. Typically, it will bind to its corresponding biological ligand, perhaps in a dimerized state with an alpha receptor subunit, with high affinity, e.g., at least about 100 nM, usually better than about 30 nM, preferably better than about 10 nM, and more preferably at better than about 3 nM. The term shall also be used herein to refer to related naturally occurring forms, e.g., alleles, polymorphic variants, and metabolic variants of the mammalian protein.
This invention also encompasses polypeptides having substantial amino acid sequence identity with the amino acid sequences shown in SEQ ID NO: 2, 4, 6, 8, 10, 12,
14, 16, 18 and 20, preferably having segments of contiguous amino acid residues identical to segments of SEQ ID NO: 4, 10, or 20. It will include sequence variants with relatively few substitutions, e.g., typically less than about 25, ordinarily less than about
15, preferably less than about 3-5. Other embodiments include forms in association with an alpha subunit, e.g., an IL-1RD4, IL-1RD5, or IL-1RD6.
A substantial polypeptide "fragment", or "segment", is a stretch of amino acid residues of at least about 8 contiguous amino acids, generally at least 10 contiguous amino acids, more generally at least 12 contiguous amino acids, often at least 14 contiguous amino acids, more often at least 16 contiguous amino acids, typically at least 18 contiguous amino acids, more typically at least 20 contiguous amino acids, usually at least 22 contiguous amino acids, more usually at least 24 contiguous amino acids, preferably at least 26 contiguous amino acids, more preferably at least 28 contiguous amino acids," and, in particularly preferred embodiments, at least about 30 or more contiguous amino acids, usually 40, 50, 70, 90, 110, etc. Sequences of segments of different polypeptides can be compared to one another over appropriate length stretches. In many cases, the matching will involve a plurality of distinct, e.g., nonoverlapping, segments of the specified length. Typically, the plurality will be at least two, more usually at least three, and preferably 5, 7, or even more. While the length minima are provided, longer lengths, of various sizes, may be appropriate, e.g., one of length 7, and two of length 12. Similar features apply to segments of nucleic acid.
Amino acid sequence homology, or sequence identity, is determined by optimizing residue matches, if necessary, by introducing gaps as required. See, e.g., Needleham, et al . (1970) J. Mol. Biol. 48:443-453; Sankoff, et al . (1983) chapter one in Time Warps , String Edits, and Macromolecules : The Theory and Practice of Sequence Comparison, Addison-Wesley, Reading, MA; and software packages from IntelliGenetics, Mountain View, CA; and the University of Wisconsin Genetics Computer Group (GCG) , Madison, WI; each of which is incorporated herein by reference. This changes when considering conservative substitutions as matches. Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. Homologous amino acid sequences are intended to include natural allelic and interspecies variations in the cytokine sequence. Typical homologous polypeptides will have from 50-100% homology (if gaps can be introduced) , to 60-100% homology (if conservative substitutions are included) with an amino acid sequence segment shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 and 20. Homology measures will be at least about 70%, generally at least 76%, more generally at least 81%, often at least 85%, more often at least 88%, typically at least 90%, more typically at least 92%, usually at least 94%, more usually at least 95%, preferably at least 96%, and more preferably at least 97%, and in particularly preferred embodiments, at least 98% or more. The degree of homology will vary with the length of the compared segments. Homologous polypeptides, such as the allelic variants, will share most biological activities with the embodiments described in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 and 20. As used herein, the term "biological activity" is used to describe, without limitation, effects on inflammatory responses, innate immunity, and/or morphogenic development by respective ligands . For example, these receptors should, like IL-1 receptors, mediate phosphatase or phosphorylase activities, which activities are easily measured by standard procedures. See, e.g., Hardie, et al. (eds. 1995) The Protein Kinase FactBook vols . I and II, Academic Press, San Diego, CA; Hanks, et al . (1991) Meth. Enzvmol . 200:38-62; Hunter, et al. (1992) Cell 70:375-388; Lewin (1990) Cell 61:743-752; Pines, et al. (1991) Cold Soring Harbor Svmp. Quant. Biol. 56:449-463; and Parker, et al . (1993) Nature 363:736-738. Other activities include antigenic or immunogenic functions . The receptors exhibit biological activities much like regulatable enzymes, regulated by ligand binding. However, the enzyme turnover number is more close to an enzyme than a receptor complex. Moreover, the numbers of occupied receptors necessary to induce such enzymatic activity is less than most receptor systems, and may number closer to dozens per cell, in contrast to most receptors which will trigger at numbers in the thousands per cell. The receptors, or portions thereof, may be useful as phosphate labeling enzymes to label general or specific substrates. The terms ligand, agonist, antagonist, and analog of, e.g., an IL-1RD8, IL-1RD9, or IL-lRDIO, include molecules that modulate the characteristic cellular responses to IL-1 ligand proteins, as well as molecules possessing the more standard structural binding competition features of ligand-receptor interactions, e.g., where the receptor is a natural receptor or an antibody. The cellular responses likely are mediated through binding of various IL-1 ligands to cellular receptors related to, but possibly distinct from, the type I or type II IL-1 receptors. See, e.g., Belvin and Anderson (1996) Ann. Rev. Cell Dev. Biol. 12:393-416; Morisato and Anderson (1995) Ann. Rev. Genetics 29:371- 3991 and Hultmark (1994) Nature 367:116-117. Also, a ligand is a molecule which serves either as a natural ligand to which said receptor, or an analog thereof, binds, or a molecule which is a functional analog of the natural ligand. The functional analog may be a ligand with structural modifications, or may be a wholly unrelated molecule which has a molecular shape which interacts with the appropriate ligand binding determinants. The ligands may serve as agonists or antagonists, see, e.g., Goodman, et al . (eds. 1990) Goodman & Gilman's: The Pharmacological Bases of Therapeutics , Pergamon Press, New York.
Rational drug design may also be based upon structural studies of the molecular shapes of a receptor or antibody and other effectors or ligands. Effectors may be other proteins which mediate other functions in response to ligand binding, or other proteins which normally interact with the receptor. One means for determining which sites interact with specific other proteins is a physical structure determination, e.g., x- ray crystallography or 2 dimensional NMR techniques . These will provide guidance as to which amino acid residues form molecular contact regions. For a detailed description of protein structural determination, see, e.g., Blundell and Johnson (1976) Protein Crystallography. Academic Press, New York, which is hereby incorporated herein by reference.
II. Activities The IL-1 receptor-like polypeptides will have "a number of different biological activities, e.g., in phosphate metabolism, being added to or removed from specific substrates, typically proteins. Such will generally result in modulation of an inflammatory function, other innate immunity response, or a morphological effect. For example, a human IL-1RD9 gene coding sequence probably has about 60-80% identity with the nucleotide coding sequence of mouse IL-1RD9. At the amino acid level, there is also likely to be reasonable identity.
The receptors will also exhibit immunogenic activity, e.g., in being capable of eliciting a selective immune response. Antiserum or antibodies resulting therefrom will exhibit both selectivity and affinity of binding. The polypeptides will also be antigenic, in binding antibodies raised thereto, in the native state, or in denatured.
The biological activities of the IL-lRDs will generally be related to addition or removal of phosphate moieties to substrates, typically in a specific manner, but occasionally in a non specific manner. Substrates may be identified, or conditions for enzymatic activity may be assayed by standard methods, e.g., as described in Hardie, et al. (eds. 1995) The Protein Kinase FactBook vols. I and II, Academic Press, San Diego, CA; Hanks, et al. (1991) Meth. Enzvmol . 200:38-62; Hunter, et al. (1992) Cell 70:375-388; Lewin (1990) Cell 61:743-752; Pines, et al . (1991) Cold Spring Harbor Svmp. Quant. Biol. 56:449-463; and Parker, et al . (1993) Nature 363:736-738.
III. Nucleic Acids
This invention contemplates use of isolated nucleic acid or fragments, e.g., which encode these or closely related proteins, or fragments thereof, e.g., to encode a corresponding polypeptide, preferably one which is biologically active. In addition, this invention covers isolated or recombinant DNA which encodes such polypeptides or polypeptides having characteristic " sequences of the respective IL-lRDs, individually or as a group. Typically, the nucleic acid is capable of hybridizing, under appropriate conditions, with a nucleic acid coding sequence segment shown in SEQ ID NO: 1, 3, 5, 7, 9, 11, 13, 15, 17 or 19 but preferably not with a' corresponding segment of other receptors. Said biologically active polypeptide can be a full length polypeptide, or fragment, and will typically have a segment of amino acid sequence highly homologous to one shown in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 and 20. Further, this invention covers the use of isolated or recombinant nucleic acid, or fragments thereof, which encode polypeptides having fragments which are equivalent to the IL-1RD9 proteins. The isolated nucleic acids can have the respective regulatory sequences in the 5' and 3' flanks, e.g., promoters, enhancers, poly-A addition signals, and others from the natural gene.
An "isolated" nucleic acid is a nucleic acid, e.g., an RNA, DNA, or a mixed polymer, which is substantially pure, e.g., separated from other components which naturally accompany a native sequence, e.g., ribosomes, polymerases, and flanking genomic sequences from the originating species . The term embraces a nucleic acid sequence which has been removed from its naturally occurring environment, and includes recombinant or cloned DNA isolates, which are thereby distinguishable from naturally occurring compositions, and chemically synthesized analogs or analogs biologically synthesized by heterologous systems. A substantially pure molecule includes isolated forms of the molecule, either completely or substantially pure.
An isolated nucleic acid will generally be a homogeneous composition of molecules, but will, in some embodiments, contain heterogeneity, preferably minor. This heterogeneity is typically found at the polymer ends or portions not critical to a desired biological function or activity.
A "recombinant" nucleic acid is typically defined either by its method of production or its structure" In reference to its method of production, e.g., a product made by a process, the process is use of recombinant nucleic acid techniques, e.g., involving human intervention in the nucleotide sequence. Typically this intervention involves in vitro manipulation, although under certain circumstances it may involve more classical animal breeding techniques. Alternatively, it can be a nucleic acid made by generating a sequence comprising fusion of two fragments which are not naturally contiguous to each other, but is meant to exclude products of nature, e.g., naturally occurring mutants as found in their natural state. Thus, for example, products made by transforming cells with an unnaturally occurring vector is encompassed, as are nucleic acids comprising sequence derived using any synthetic oligonucleotide process . Such a process is often done to replace a codon with a redundant codon encoding the same or a conservative amino acid, while typically introducing or removing a restriction enzyme sequence recognition site. Alternatively, the process is performed to join together nucleic acid segments of desired functions to generate a single genetic entity comprising a desired combination of functions not found in the commonly available natural forms, e.g., encoding a fusion protein. Restriction enzyme recognition sites are often the target of such artificial manipulations, but other site specific targets, e.g., promoters, DNA replication sites, regulation sequences, control sequences, or other useful features may be incorporated by design. A similar concept is intended for a recombinant, e.g., fusion, polypeptide. This will include a dimeric repeat. Specifically included are synthetic nucleic acids which, by genetic code redundancy, encode equivalent polypeptides to fragments of IL-1RD9 and fusions of sequences from various different related molecules, e.g., other IL-1 receptor family members.
A "fragment" in a nucleic acid context is a contiguous segment of at least about 17 contiguous nucleotides, generally at least 21 contiguous " " nucleotides, more generally at least 25 contiguous nucleotides, ordinarily at least 30 contiguous nucleotides, more ordinarily at least 35 contiguous nucleotides, often at least 39 contiguous nucleotides, more often at least 45 contiguous nucleotides, typically at least 50 contiguous nucleotides, more typically at least 55 contiguous nucleotides, usually at least 60 contiguous nucleotides, more usually at least 66 contiguous nucleotides, preferably at least 72 contiguous nucleotides, more preferably at least 79 contiguous nucleotides, and in particularly preferred embodiments will be at least 85 or more contiguous nucleotides, e.g., 100, 120, 140, etc. Typically, fragments of different genetic sequences can be compared to one another over appropriate length stretches, particularly defined segments such as the domains described below.
A nucleic acid which codes for an IL-1RD8, IL-1RD9, or IL-1RD10 will be particularly useful to identify genes, mRNA, and cDNA species which code for itself or closely related proteins, as well as DNAs which code for polymorphic, allelic, or other genetic variants, e.g., from different individuals or related species. Preferred probes for such screens are those regions of the interleukin which are conserved between different polymorphic variants or which contain nucleotides which lack specificity, and will preferably be full length or nearly so. In other situations, polymorphic variant specific sequences will be more useful.
This invention further covers recombinant nucleic acid molecules and fragments having a nucleic acid sequence identical to or highly homologous to the isolated DNA set forth herein. In particular, the sequences will often be operably linked to DNA segments which control transcription, translation, and DNA replication. These additional segments typically assist in expression of the desired nucleic acid segment. Homologous, or highly identical, nucleic acid sequences, when compared to one another, e.g., IL-1RD9 sequences, exhibit significant similarity. The standards for homology in nucleic acids are either measures for homology generally used in the art by sequence comparison or based upon hybridization conditions . Comparative hybridization conditions are described in greater detail below.
Substantial identity in the nucleic acid sequence comparison context means either that the segments, or their complementary strands, when compared, are identical when optimally aligned, with appropriate nucleotide insertions or deletions, in at least about 60% of the nucleotides, generally at least 66%, ordinarily at least 71%, often at least 76%, more often at least 80%, usually at least 84%, more usually at least 88%, typically at least 91%, more typically at least about 93%, preferably at least about 95%, more preferably at least about 96 to 98% or more, and in particular embodiments, as high at about 99% or more of the nucleotides, including, e.g., segments encoding structural domains such as the segments described below. Alternatively, substantial identity will exist when the segments will hybridize under selective hybridization conditions, to a strand or its complement, typically using a sequence derived from SEQ ID NO: 1, 3, 5, 7, 9, 11, 13, 15, 17 or 19. Typically, selective hybridization will occur when there is at least about 55% homology over a stretch of at least about 14 nucleotides, more typically at least about 65%, preferably at least about 75%, and more preferably at least about 90%. See, Kanehisa (1984) Nuc . Acids Res . 12:203-213, which is incorporated herein by reference. The length of homology comparison, as described, may be over longer stretches, and in certain embodiments will be over a stretch of at least about 17 nucleotides, generally at least about 20 nucleotides, ordinarily at least about 24 nucleotides, usually at least about 28 nucleotides, typically at least about 32 nucleotides, more typically at least about 40 nucleotides, preferably at least about 50 nucleotides, and more preferably at least about 75 to 100 or more nucleotides. Stringent conditions, in referring to homology" in the hybridization context, will be stringent combined conditions of salt, temperature, organic solvents, and other parameters typically controlled in hybridization reactions . Stringent temperature conditions will usually include temperatures in excess of about 30° C, more usually in excess of about 37* C, typically in excess of about 45' C, more typically in excess of about 55" C, preferably in excess of about 65° C, and more preferably in excess of about 70° C. Stringent salt conditions will ordinarily be less than about 500 mM, usually less than about 400 mM, more usually less than about 300 mM, typically less than about 200 mM, preferably less than about 100 mM, and more preferably less than about 80 mM, even down to less than about 20 mM. However, the combination of parameters is much more important than the measure of any single parameter. See, e.g., Wet ur and Davidson (1968) J. Mol. Biol. 31:349-370, which is hereby incorporated herein by reference. • The signal should be at least 2X over background, generally at least 5-10X over background, and preferably even more.
For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are input into a computer, subsequent coordinates are designated, if necessary, and sequence algorithm program parameters are designated. The sequence comparison algorithm then calculates the percent sequence identity for the test sequence (s) relative to the reference sequence, based on the designated program parameters.
Optical alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1981) Adv. APPI . Math. 2:482, by the homology alignment algorithm of Needleman and Wunsch (1970) CT . Mol. Biol. 48:443, by the search for similarity method of Pearson and Lipman (1988) Proc . Nat ' 1 Acad. Sci . USA 85:2444, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Com uter Group, 575 Science Dr., Madison, WI) , or by visual inspection (see generally Ausubel et al., supra).
One example of a useful algorithm is PILEUP. PILEUP creates a multiple sequence alignment from a group of related sequences using progressive, pairwise alignments to show relationship and percent sequence identity. It also plots a tree or dendrogram showing the clustering relationships used to create the alignment. PILEUP uses a simplification of the progressive alignment method of Feng and Doolittle (1987) J. Mol. Evol. 35:351-360. The method used is similar to the method described by Higgins and Sharp (1989) CABIOS 5:151-153. The program can align up to 300 sequences, each of a maximum length of 5,000 nucleotides or amino acids. The multiple alignment procedure begins with the pairwise alignment of the two most similar sequences, producing a cluster of two aligned sequences . This cluster is then aligned to the next most related sequence or cluster of aligned sequences. Two clusters of sequences are aligned by a simple extension of the pairwise alignment of two individual sequences . The final alignment is achieved by a series of progressive, pairwise alignments. The program is run by designating specific sequences and their amino acid or nucleotide coordinates for regions of sequence comparison and by designating the program parameters. For example, a reference sequence can be compared to other test sequences to determine the percent sequence identity relationship using the following parameters: default gap weight (3.00), default gap length weight (0.10) , and weighted end gaps.
Another example of algorithm that is suitable for determining percent sequence identity and sequence similarity is the BLAST algorithm, which is described Altschul, et al. (1990) J. Mol. Biol. 215:403-410. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http:www.ncbi.nlm.nih.gov/) . This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul, et al . , supra). These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLAST program uses as defaults a wordlength (W) of 11, the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1989) Proc. Nat'l Acad. Sci. USA 89:10915) alignments (B) of
50, expectation (E) of 10, M=5, N=4, and a comparison of both strands .
In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Nat'l Acad. Sci. USA 90:5873-5787) . One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.
A further indication that two nucleic acid sequences of polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is " " immunologically cross reactive with the polypeptide encoded by the second nucleic acid, as described below. Thus, a polypeptide is typically substantially identical to a second polypeptide, e.g., where the two peptides differ only by conservative substitutions. Another indication that two nucleic acid sequences are substantially identical is that the two molecules hybridize to each other under stringent conditions, as described below. The isolated DNA can be readily modified by nucleotide substitutions, nucleotide deletions, nucleotide insertions, and inversions of nucleotide stretches. These modifications result in novel DNA sequences which encode this polypeptide or its derivatives. These modified sequences can be used to produce mutant proteins (muteins) or to enhance the expression of variant species. Enhanced expression may involve gene amplification, increased transcription, increased translation, and other mechanisms. Such mutant IL-lRD9-like derivatives include predetermined or site-specific mutations of the polypeptide or its fragments, including silent mutations using genetic code degeneracy. "Mutant IL-1RD9" as used herein encompasses a polypeptide otherwise falling within the homology definition of the IL-1R9 as set forth above, but having an amino acid sequence which differs from that of other IL-lRD-like polypeptides as found in nature, whether by way of deletion, substitution, or insertion. In particular, "site specific mutant IL-1RD9" encompasses a polypeptide having substantial homology with a polypeptide of SEQ ID NO: 6, 8, 10, 12, 14 or 14, and typically shares most of the biological activities or effects of the forms disclosed herein. Although site specific mutation sites are predetermined, mutants need not be site specific. Mammalian IL-1RD9 mutagenesis can be achieved by making amino acid insertions or deletions in the gene, coupled with expression. Substitutions, deletions, insertions, or many combinations may be generated to arrive at a final construct. Insertions include amino- or carboxy- terminal fusions. Random mutagenesis can be conducted at a target codon and the expressed mammalian IL-1RD9 mutants can then be screened for the desired activity, providing some aspect of a structure-activity relationship. Methods for making substitution mutations at predetermined sites in DNA having a known sequence are well known in the art, e.g., by M13 primer mutagenesis. See also Sambrook, et al . (1989) and Ausubel, et al . (1987 and periodic Supplements) .
The mutations in the DNA normally should not place coding sequences out of reading frames and preferably will not create complementary regions that could hybridize to produce secondary mRNA structure such as loops or hairpins .
The phosphoramidite method described by Beaucage and Carruthers (1981) Tetra. Letts. 22:1859-1862, will produce suitable synthetic DNA fragments . A double stranded fragment will often be obtained either by synthesizing the complementary strand and annealing the strand together under appropriate conditions or by adding the complementary strand using DNA polymerase with an appropriate primer sequence. Polymerase chain reaction (PCR) techniques can often be applied in mutagenesis. Alternatively, mutagenesis primers are commonly used methods for generating defined mutations at predetermined sites. See, e.g., Innis, et al. (eds. 1990) PCR Protocols: A Guide to Methods and Applications Academic Press, San Diego, CA; and
Dieffenbach and Dveksler (1995; eds.) PCR Primer: A Laboratory Manual Cold Spring Harbor Press, CSH, NY. Appropriate primers of length, e.g., 15, 20, 25, or longer can be made using sequence provided. IV. Proteins, Peptides
As described above, the present invention encompasses primate IL-1RD8, primate or rodent IL-1RD9, and primate IL-lRD10,e.g. , whose sequences are disclosed e.g., in Tables 1-3, and described herein. Descriptions of features of IL-1RD9 are applicable in most cases, with appropriate modifications, also to IL-1RD8 and/or to IL- 1RD10. Allelic and other variants are also contemplated, including, e.g., fusion proteins combining portions of such sequences with others, including epitope tags and functional domains. Particularly interesting constructs will be intact extracellular or intracellular domains . The present invention also provides recombinant polypeptides, e.g., heterologous fusion proteins using segments from these rodent proteins. A heterologous fusion protein is a fusion of proteins or segments which are naturally not normally fused in the same manner. Thus, the fusion product of, e.g., an IL-1RD9 with another IL-1 receptor is a continuous protein molecule having sequences fused in a typical polypeptide linkage, typically made as a single translation product and exhibiting properties, e.g., sequence or antigenicity, derived from each source peptide. A similar concept applies to heterologous nucleic acid sequences.
In addition, new constructs may be made from combining similar functional or structural domains from other related proteins, e.g., IL-1 receptors or Toll-like receptors, including species variants. For example, ligand-binding or other segments may be "swapped" between different new fusion polypeptides or fragments. See, e.g., Cunningham, et al . (1989) Science 243:1330-1336; and O'Dowd, et al. (1988) J. Biol. Chem. 263:15985-15992, each of which is incorporated herein by reference. Thus, new chimeric polypeptides exhibiting new combinations of specificities will result from the functional linkage of receptor-binding specificities. For example, the ligand binding domains from other related receptor molecules may be added or substituted for other domains of this or related proteins. The resulting protein will often have hybrid function and properties. For example, a fusion protein may include a targeting domain which may serve to provide sequestering of the fusion protein to a particular subcellular organelle.
Candidate fusion partners and sequences can be selected from various sequence data bases, e.g., GenBank, c/o NCBI, and BCG, University of Wisconsin Biotechnology Computing Group, Madison, WI, which are each incorporated herein by reference.
The present invention particularly provides muteins which bind IL-1-like ligands, and/or which are affected in signal transduction. Structural alignment of human IL-1RD9 with other members of the IL-1R family show conserved features/residues. See Tables 1-4. Alignment of the human IL-1RD9 sequence with other members of the IL-1R family indicates various structural and functionally shared features. See also, Bazan, et al . (1996) Nature 379:591; Lodi, et al . (1994) Science 263:1762-1766; Sayle and Milner-White (1995) TIBS 20:374- 376; and Gronenberg, et al . (1991) Protein Engineering
4:263-269.
The IL-lα and IL-lβ ligands bind an IL-1 receptor type I (IL-1RD1) as the primary receptor and this complex then forms a high affinity receptor complex with the IL-1 receptor type III (IL-1RD3) . Such receptor subunits are probably shared with the receptors for the new IL-1 ligand family members. See, e.g., USSN 60/044,165 and USSN 60/055,111. It is likely that the IL-lγ ligand signals through a receptor comprising the association of IL-1RD9 (alpha component) with IL-1RD5 (beta component) . The IL-lδ and IL-lε ligands each probably signal through a receptor comprising the association of one of IL-1RD4, IL-1RD6, or IL-1RD9 (alpha components) with one of IL- 1RD3, IL-1RD5, IL-1RD7, IL-1RD8, or IL-1RD10 (beta components) .
Similar variations in other species counterparts of IL-1R sequences, e.g., receptors D1-D6, D8, D9, or D10, in the corresponding regions, should provide similar interactions with ligand or substrate. Substitutions with either rodent or primate, e.g., mouse sequences or human sequences, are particularly preferred. Conversely, conservative substitutions away from the ligand binding interaction regions will probably preserve most signaling activities; and conservative substitutions away from the intracellular domains will probably preserve most ligand binding properties .
"Derivatives" of the primate or mouse IL-1RD9 include amino acid sequence mutants, glycosylation variants, metabolic derivatives and covalent or aggregative conjugates with other chemical moieties. Covalent derivatives can be prepared by linkage of functionalities to groups which are found in the IL-1RD9 amino acid side chains or at the N- or C- termini, e.g., by means which are well known in the art. These derivatives can include, without limitation, aliphatic esters or amides of the carboxyl terminus, or of residues containing carboxyl side chains, O-acyl derivatives of hydroxyl group-containing residues, and N-acyl derivatives of the amino terminal amino acid or amino-group containing residues, e.g., lysine or arginine. Acyl groups are selected from the group of alkyl-moieties including C3 to C18 normal alkyl, thereby forming alkanoyl aroyl species.
In particular, glycosylation alterations are included, e.g., made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing, or in further processing steps. Particularly preferred means for accomplishing this are by exposing the polypeptide to glycosylating enzymes derived from cells which normally provide such processing, e.g., mammalian glycosylation enzymes. Deglycosylation enzymes are also contemplated. Also embraced are versions of the same primary amino acid sequence which have other minor modifications, including phosphorylated amino acid residues, e.g., phosphotyrosine, phosphoserine, or phosphothreonine . A major group of derivatives are covalent conjugates of the receptors or fragments thereof with other polypeptides. These derivatives can be synthesized in recombinant culture such as N- or C-terminal fusions or by the use of agents known in the art for their " " usefulness in cross-linking proteins through reactive side groups. Preferred derivatization sites with cross-linking agents are at free amino groups, carbohydrate moieties, and cysteine residues. Fusion polypeptides between the receptors and other homologous or heterologous proteins are also provided. Homologous polypeptides may be fusions between different receptors, resulting in, for instance, a hybrid protein exhibiting binding specificity for multiple different IL- 1 ligands, or a receptor which may have broadened or weakened specificity of substrate effect. Likewise, heterologous fusions may be constructed which would exhibit a combination of properties or activities of the derivative proteins. Typical examples are fusions of a reporter polypeptide, e.g., luciferase, with a segment or domain of a receptor, e.g., a ligand-binding segment, so that the presence or location of a desired ligand may be easily determined. See, e.g., Dull, et al . , U.S. Patent No. 4,859,609, which is hereby incorporated herein by reference. Other gene fusion partners include glutathione-S-transferase (GST) , bacterial β- galactosidase, trpE, Protein A, β-lactamase, alpha amylase, alcohol dehydrogenase, and yeast alpha mating factor. See, e.g., Godowski, et al . (1988) Science 241:812-816.
The phosphoramidite method described by Beaucage and Carruthers (1981) Tetra. Letts. 22:1859-1862, will produce suitable synthetic DNA fragments . A double stranded fragment will often be obtained either by synthesizing the complementary strand and annealing the strand together under appropriate conditions or by adding the complementary strand using DNA polymerase with an appropriate primer sequence. Such polypeptides may also have amino acid residues which have been chemically modified by phosphorylation, sulfonation, biotinylation, or the addition or removal of other moieties, particularly those which have molecular shapes similar to phosphate groups. In some emboα.iTnents , the modifications will be useful labeling reagents, or serve as purification targets, e.g., affinity ligands. Fusion proteins will typically be made by either recombinant nucleic acid methods or by synthetic polypeptide methods. Techniques for nucleic acid manipulation and expression are described generally, e.g., in Sambrook, et al . (1989) Molecular Cloning: A Laboratory Manual (2d ed. ) , Vols. 1-3, Cold Spring Harbor Laboratory, and Ausubel, et al . (eds. 1987 and periodic supplements) Current Protocols in Molecular Biology. Greene/Wiley, New York, which are each incorporated herein by reference. Techniques for synthesis of polypeptides are described, e.g., in Merrifield (1963) J. Amer. Chem. Soc . 85:2149-2156; Merrifield (1986) Science 232: 341-347; and Atherton, et al . (1989) Solid Phase Peptide Synthesis: A Practical Approach, IRL Press, Oxford; each of which is incorporated herein by reference. See also Dawson, et al. (1994) Science 266:776-779 for methods to make larger polypeptides. This invention also contemplates the use of derivatives of an IL-1RD8, IL-1RD9, or IL-lRDIO other than variations in amino acid sequence or glycosylation. Such derivatives may involve covalent or aggregative association with chemical moieties . These derivatives generally fall into three classes: (1) salts, (2) side chain and terminal residue covalent modifications, and (3) adsorption complexes, for example with cell membranes . Such covalent or aggregative derivatives are useful as immunogens, as reagents in immunoassays, or in purification methods such as for affinity purification of a receptor or other binding molecule, e.g., an antibody. For example, an IL-1 ligand can be immobilized by covalent bonding to a solid support such as cyanogen bromide-activated Sepharose, by methods which are well known in the art, or adsorbed onto polyolefin surfaces, with or without glutaraldehyde cross-linking, for use in the assay or purification of an IL-1 receptor, antibodies, or other similar molecules. The ligand can also be labeled with a detectable group, e.g., " " radioiodinated by the chloramine T procedure, covalently bound to rare earth chelates, or conjugated to another fluorescent moiety for use in diagnostic assays.
An IL-1RD8, IL-1RD9, or IL-1RD10 of this invention can be used as an immunogen for the production of antisera or antibodies specific, e.g., capable of distinguishing between other IL-1 receptor family members, for the IL-1RD8, IL-1RD9, or IL-lRDIO or various fragments thereof. The purified IL-1RD8, IL-1RD9, or IL- 1RD10 can be used to screen monoclonal antibodies or antigen-binding fragments prepared by immunization with various forms of impure preparations containing the protein. In particular, the term "antibodies" also encompasses antigen binding fragments of natural antibodies, e.g., Fab, Fab2, Fv, etc. The purified IL- 1RD9 can also be used as a reagent to detect antibodies generated in response to the presence of elevated levels of expression, or immunological disorders which lead to antibody production to the endogenous receptor. Additionally, IL-1RD8, IL-1RD9, or IL-lRDIO fragments may also serve as immunogens to produce the antibodies of the present invention, as described immediately below. For example, this invention contemplates antibodies having binding affinity to or being raised against the amino acid sequences shown, e.g., in SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18 and 20, fragments thereof, or various homologous peptides. In particular, this invention contemplates antibodies having binding affinity to, or having been raised against, specific fragments which are predicted to be, or actually are, exposed at the exterior polypeptide surface of the native IL-1RD8, IL-1RD9, or IL-1RD10. Various preparations of desired selectivity in binding can be prepared by appropriate cross absorptions, etc. The blocking of physiological response to the receptor ligands may result from the inhibition of binding of the ligand to the receptor, likely through competitive inhibition. Thus, in vitro assays of the present invention will often use antibodies or antigen binding segments of these antibodies, or fragments attached to solid phase substrates. These assays will also allow for the diagnostic determination of the effects of either ligand binding region mutations and modifications, or other mutations and modifications, e.g., which affect signaling or enzymatic function.
This invention also contemplates the use of competitive drug screening assays, e.g., where neutralizing antibodies to the receptor or fragments compete with a test compound for binding to a ligand or other antibody. In this manner, the neutralizing antibodies or fragments can be used to detect the presence of a polypeptide which shares one or more binding sites to a receptor and can also be used to occupy binding sites on a receptor that might otherwise bind a ligand.
V. Making Nucleic Acids and Protein
DNA which encodes the polypeptides or fragments thereof can be obtained by chemical synthesis, screening cDNA libraries, or by screening genomic libraries prepared from a wide variety of cell lines or tissue samples. Natural sequences can be isolated using standard methods and the sequences provided herein, e.g., in Tables 1-3. Other species counterparts can be identified by hybridization techniques, or by various PCR techniques, combined with or by searching in sequence databases , e.g., GenBank.
This DNA can be expressed in a wide variety of host cells for the synthesis of a full-length receptor or fragments which can in turn, e.g., be used to generate polyclonal or monoclonal antibodies; for binding studies; for construction and expression of modified ligand binding or kinase/phosphatase domains; and for structure/function studies. Variants or fragments can be expressed in host cells that are transformed or transfected with appropriate expression vectors . These molecules can be substantially free of protein or cellular contaminants, other than those derived from the recombinant host, and therefore are particularly useful in pharmaceutical compositions when combined with a pharmaceutically acceptable carrier and/or diluent . The protein, or portions thereof, may be expressed as fusions with other proteins .
Expression vectors are typically self-replicating DNA or RNA constructs containing the desired receptor gene or its fragments, usually operably linked to suitable genetic control elements that are recognized in a suitable host cell. These control elements are capable of effecting expression within a suitable host. The specific type of control elements necessary to effect expression will depend upon the eventual host cell used. Generally, the genetic control elements can include a prokaryotic promoter system or a eukaryotic promoter expression control system, and typically include a transcriptional promoter, an optional operator to control the onset of transcription, transcription enhancers to elevate the level of mRNA expression, a sequence that encodes a suitable ribosome binding site, and sequences that terminate transcription and translation. Expression vectors also usually contain an origin of replication that allows the vector to replicate independently of the host cell. The vectors of this invention include those which contain DNA which encodes a protein, as described, or a fragment thereof encoding a biologically active equivalent polypeptide. The DNA can be under the control of a viral promoter and can encode a selection marker. This invention further contemplates use of such expression vectors which are capable of expressing eukaryotic cDNA coding for such a polypeptide in a prokaryotic or eukaryotic host, where the vector is compatible with the host and where the eukaryotic cDNA coding for the receptor is inserted into the vector such that growth of the host containing the vector expresses the cDNA in question. Usually, expression vectors are designed for stable replication in their host cells or for amplification to greatly increase the total number of copies of the desirable gene per cell . It is not always necessary to require that an expression vector replicate in a host cell, e.g., it is possible to effect transient expression of the polypeptide or its fragments in various hosts using vectors that do not contain a replication origin that is recognized by the host cell. It is also possible to use vectors that cause integration of the polypeptide encoding portion or its fragments into the host DNA by recombination. Vectors, as used herein, comprise plasmids, viruses, bacteriophage, integratable DNA fragments, and other vehicles which enable the integration of DNA fragments into the genome of the host. Expression vectors are specialized vectors which contain genetic control elements that effect expression of operably linked genes. Plasmids are the most commonly used form of vector but all other forms of vectors which serve an equivalent function and which are, or become, known in the art are suitable for use herein. See, e.g., Pouwels, et al . (1985 and Supplements) Cloning Vectors : A Laboratory Manual , Elsevier, N.Y. , and Rodriquez, et al . (eds.) Vectors : A Survey of Molecular Cloning Vectors and Their Uses, Buttersworth, Boston, 1988, which are incorporated herein by reference. Transformed cells are cells, preferably mammalian, that have been transformed or transfected with receptor vectors constructed using recombinant DNA techniques. Transformed host cells usually express the desired polypeptide or its fragments, but for purposes of cloning, amplifying, and manipulating its DNA, do not need to express the subject protein. This invention further contemplates culturing transformed cells in a nutrient medium, thus permitting the receptor to accumulate in the cell membrane. The polypeptide can be recovered, either from the culture or, in certain instances, from the culture medium.
For purposes of this invention, nucleic sequences are operably linked when they are functionally related to each other. For example, DNA for a presequence or secretory leader is operably linked to a polypeptide if it is expressed as a preprotein or participates in directing the polypeptide to the cell membrane or in secretion of the polypeptide. A promoter is operably linked to a coding sequence if it controls the transcription of the polypeptide; a ribosome binding site is operably linked to a coding sequence if it is positioned to permit translation. Usually, operably linked means contiguous and in reading frame, however, certain genetic elements such as repressor genes are not contiguously linked but still bind to operator sequences that in turn control expression.
Suitable host cells include prokaryotes, lower eukaryotes, and higher eukaryotes . Prokaryotes include both gram negative and gram positive organisms, e.g., E. coli and B. subtilis. Lower eukaryotes include yeasts, e.g., S. cerevisiae and Pichia, and species of the genus Dictvostelium. Higher eukaryotes include established tissue culture cell lines from animal cells, both of non-mammalian origin, e.g., insect cells, and birds, and of mammalian origin, e.g., human, primates, and rodents.
Prokaryotic host-vector systems include a wide variety of vectors for many different species. As used herein, E. coli and its vectors will be used generically to include equivalent vectors used in other prokaryotes. A representative vector for amplifying DNA is pBR322 or many of its derivatives . Vectors that can be used to express the receptor or its fragments include, but are not limited to, such vectors as those containing the lac promoter (pUC-series) ; trp promoter (pBR322-trp) ; Ipp promoter (the pIN-series) ; lambda-pP or pR promoters (pOTS) ; or hybrid promoters such as ptac (pDR540) . See Brosius, et al. (1988) "Expression Vectors Employing Lambda-, trp-, lac-, and Ipp-derived Promoters", in Vectors : A Survey of Molecular Cloning Vectors and Their Uses, (eds. Rodriguez and Denhardt) , Buttersworth, Boston, Chapter 10, pp. 205-236, which is incorporated herein by reference. Lower eukaryotes, e.g., yeasts and DictvosteJium. may be transformed with IL-1RD9 sequence containing vectors. For purposes of this invention, the most common lower eukaryotic host is the baker's yeast, Saccharomyces cerevisiae. It will be used to generically represent lower eukaryotes although a number of other strains and species are also available. Yeast vectors typically consist of a replication origin (unless of the integrating type) , a selection gene, a promoter, DNA encoding the receptor or its fragments, and sequences for translation termination, polyadenylation, and transcription termination. Suitable expression vectors for yeast include such constitutive promoters as 3-phosphoglycerate kinase and various other glycolytic enzyme gene promoters or such inducible promoters as the alcohol dehydrogenase 2 promoter or metallothionine promoter. Suitable vectors include derivatives of the following types: self-replicating low copy number (such as the YRp-series) , self-replicating high copy number (such as the YEp-series) ; integrating types (such as the Yip-series) , or mini-chromosomes (such as the YCp-series) .
Higher eukaryotic tissue culture cells are normally the preferred host cells for expression of the functionally active interleukin protein. In principle, many higher eukaryotic tissue culture cell lines are workable, e.g., insect baculovirus expression systems, whether from an invertebrate or vertebrate source. However, mammalian cells are preferred. Transformation or transfection and propagation of such cells has become a routine procedure. Examples of useful cell lines include HeLa cells, Chinese hamster ovary (CHO) cell lines, baby rat kidney (BRK) cell lines, insect cell lines, bird cell lines, and monkey (COS) cell lines. Expression vectors for such cell lines usually include an origin of replication, a promoter, a translation initiation site, RNA splice sites (if genomic DNA is used) , a polyadenylation site, and a transcription termination site. These vectors also usually contain a selection gene or amplification gene. Suitable expression vectors may be plasmids, viruses, or retroviruses carrying promoters derived, e.g., from such sources as from adenovirus, SV40, parvoviruses , vaccinia virus, or cytomegalovirus . Representative examples of suitable expression vectors include pCDNAl; pCD, see Okayama, et al. (1985) Mol. Cell Biol. 5:1136-1142; pMClneo PolyA, see Thomas, et al . (1987) Cell 51:503-512; and a baculovirus vector such as pAC 373 or pAC 610.
For secreted proteins, an open reading frame usually encodes a polypeptide that consists of a mature or secreted product covalently linked at its N-terminus to a signal peptide. The signal peptide is cleaved prior to secretion of the mature, or active, polypeptide. The cleavage site can be predicted with a high degree of accuracy from empirical rules, e.g., von-Heijne (1986) Nucleic Acids Research 14:4683-4690 and Nielsen, et al . (1997) Protein Enq. 10:1-12, and the precise amino acid composition of the signal peptide often does not appear to be critical to its function, e.g., Randall, et al. (1989) Science 243:1156-1159; Kaiser, et al. (1987) Science 235:312-317.
It will often be desired to express these polypeptides in a system which provides a specific or defined glycosylation pattern. In this case, the usual pattern will be that provided naturally by the expression system. However, the pattern will be modifiable by exposing the polypeptide, e.g., an unglycosylated form, to appropriate glycosylating proteins introduced into a heterologous expression system. For example, the receptor gene may be co-transformed with one or more genes encoding mammalian or other glycosylating enzymes . Using this approach, certain mammalian glycosylation patterns will be achievable in prokaryote or other cells. The source of IL-1RD8, IL-1RD9, or IL-lRDIO can be a eukaryotic or prokaryotic host expressing recombinant IL- 1RD8, IL-1RD9, or IL-lRDIO such as is described above. The source can also be a cell line such as mouse Swiss 3T3 fibroblasts, but other mammalian cell lines are also contemplated by this invention, with the preferred cell line being from the human species.
Now that the sequences are known, the primate IL- lRs, fragments, or derivatives thereof can be prepared by conventional processes for synthesizing peptides. These include processes such as are described in Stewart and Young (1984) Solid Phase Peptide Synthesis. Pierce Chemical Co., Rockford, IL; Bodanszky and Bodanszky (1984) The Practice of Peptide Synthesis, Springer-Verlag, New York; and Bodanszky (1984) The
Principles of Peptide Synthesis, Springer-Verlag, New York; all of each which are incorporated herein by reference. For example, an azide process, an acid chloride process, an acid anhydride process, a mixed anhydride process, an active ester process (e.g., p-nitrophenyl ester, N-hydroxysuccinimide ester, or cyanomethyl ester), a carbodiimidazole process, an oxidative-reductive process, or a dicyclohexylcarbodiimide (DCCD) /additive process can be used. Solid phase and solution phase syntheses are both applicable to the foregoing processes. Similar techniques can be used with partial IL-1RD9 sequences.
The IL-1RD8, IL-1RD9, or IL-1RD10 proteins, polypeptides, fragments, or derivatives are suitably prepared in accordance with the above processes as typically employed in peptide synthesis, generally either by a so-called stepwise process which comprises condensing an amino acid to the terminal amino acid, one by one in sequence, or by coupling peptide fragments to the terminal amino acid. Amino groups that are not being used in the coupling reaction typically must be protected to prevent coupling at an incorrect location.
If a solid phase synthesis is adopted, the C-terminal amino acid is bound to an insoluble carrier or support through its carboxyl group. The insoluble carrier is not particularly limited as long as it has a binding capability to a reactive carboxyl group. Examples of such insoluble carriers include halomethyl resins, such as chloromethyl resin or bromomethyl resin, hydroxymethyl resins, phenol resins, tert-alkyloxycarbonylhydrazidated resins, and the like.
An amino group-protected amino acid is bound in sequence through condensation of its activated carboxyl group and the reactive amino group of the previously formed peptide or chain, to synthesize the peptide step by step. After synthesizing the complete sequence, the peptide is split off from the insoluble carrier to produce the peptide. This solid-phase approach is generally described by Merrifield, et al . (1963) in J.
Am. Chem. Soc. 85:2149-2156, which is incorporated herein by reference.
The prepared protein and fragments thereof can be isolated and purified from the reaction mixture by means of peptide separation, e.g., by extraction, precipitation, electrophoresis, various forms of chromatography, and the like. The receptors of this invention can be obtained in varying degrees of purity depending upon desired uses. Purification can be accomplished by use of the protein purification techniques disclosed herein, see below, or by the use of the antibodies herein described in methods of immunoabsorbant affinity chromatography. This immunoabsorbant affinity chromatography is carried out by first linking the antibodies to a solid support and then contacting the linked antibodies with solubilized lysates of appropriate cells, lysates of other cells expressing the receptor, or lysates or supernatants of cells producing the polypeptide as a result of DNA techniques, see below.
Generally, the purified protein will be at least .. about 40% pure, ordinarily at least about 50% pure, usually at least about 60% pure, typically at least about 70% pure, more typically at least about 80% pure, preferable at least about 90% pure and more preferably at least about 95% pure, and in particular embodiments, 97%- 99% or more. Purity will usually be on a weight basis, but can also be on a molar basis. Different assays will be applied as appropriate. Similar concepts apply to polynucleotides and antibodies .
VI . Antibodies
Antibodies can be raised to the various mammalian IL-1RD8, IL-1RD9, or IL-lRDIO described herein, e.g.^ primate IL-1RD9 polypeptides and fragments thereof, both in naturally occurring native forms and in their recombinant forms, the difference being that antibodies to the active receptor are more likely to recognize epitopes which are only present in the native conformations . Denatured antigen detection can also be useful in, e.g., Western analysis. Anti-idiotypic antibodies are also contemplated, which would be useful as agonists or antagonists of a natural receptor or an antibody.
Antibodies, including binding fragments and single chain versions, against predetermined fragments of the polypeptide can be raised by immunization of animals with conjugates of the fragments with immunogenic proteins. Monoclonal antibodies are prepared from cells secreting the desired antibody. These antibodies can be screened for binding to normal or defective protein, or screened for agonistic or antagonistic activity. These monoclonal antibodies will usually bind with at least a K^, of about 1 mM, more usually at least about 300 μM, typically at least about 100μM, more typically at least about 30 μM, preferably at least about 10 μM, and more preferably at least about 3 μM or better.
The antibodies, including antigen binding fragments, of this invention can have significant diagnostic or therapeutic value. They can be potent antagonists that bind to the receptor and inhibit binding to ligand or inhibit the ability of the receptor to elicit a biological response, e.g., act on its substrate. They also can be useful as non-neutralizing antibodies and can be coupled to toxins or radionuclides to bind producing cells, or cells localized to the source of the interleukin. Further, these antibodies can be conjugated to drugs or other therapeutic agents, either directly or indirectly by means of a linker.
The antibodies of this invention can also be useful in diagnostic applications. As capture or non-neutralizing antibodies, they might bind to the receptor without inhibiting ligand or substrate binding. As neutralizing antibodies, they can be useful in competitive binding assays. They will also be useful in detecting or quantifying ligand. They may be used as reagents for Western blot analysis, or for immunoprecipitation or immunopurification of the respective protein.
Protein fragments may be joined to other materials, particularly polypeptides, as fused or covalently joined polypeptides to be used as immunogens . Mammalian IL-IRs and fragments may be fused or covalently linked to a variety of immunogens, such as keyhole limpet hemocyanin, bovine serum albumin, tetanus toxoid, etc. See Microbiology, Hoeber Medical Division, Harper and Row, 1969; Landsteiner (1962) Specificity of Serological Reactions , Dover Publications, New York; and Williams, et al . (1967) Methods in Immunology and Immunochemistry, Vol. 1, Academic Press, New York; each of which are incorporated herein by reference, for descriptions of methods of preparing polyclonal antisera. A typical method involves hyperimmunization of an animal with an antigen. The blood of the animal is then collected shortly after the repeated immunizations and the gamma globulin is isolated.
In some instances, it is desirable to prepare monoclonal antibodies from various mammalian hosts, such as mice, rodents, primates, humans, etc. Description of techniques for preparing such monoclonal antibodies may be found in, e.g., Stites, et al . (eds.) Basic and Clinical Immunology (4th ed. ) , Lange Medical Publications, Los Altos, CA, and references cited therein; Harlow and Lane (1988) Antibodies : A Laboratory Manual , CSH Press; Goding (1986) Monoclonal Antibodies: Principles and Practice (2d ed. ) Academic Press, New York; and particularly in Kohler and Milstein (1975*) in Nature 256:495-497, which discusses one method of generating monoclonal antibodies . Each of these references is incorporated herein by reference. Summarized briefly, this method involves injecting an animal with an immunogen. The animal is then sacrificed and cells taken from its spleen, which are then fused with myeloma cells. The result is a hybrid cell or "hybridoma" that is capable of reproducing in vitro. The population of hybridomas is then screened to isolate individual clones, each of which secrete a single antibody species to the immunogen. In this manner, the individual antibody species obtained are the products of immortalized and cloned single B cells from the immune animal generated in response to a specific site recognized on the immunogenic substance.
Other suitable techniques involve in vitro exposure of lymphocytes to the antigenic polypeptides or alternatively to selection of libraries of antibodies in phage or similar vectors. See, Huse, et al . (1989) "Generation of a Large Combinatorial Library of the Immunoglobulin Repertoire in Phage Lambda, " Science 246:1275-1281; and Ward, et al . (1989) Nature 341:544- 546, each of which is hereby incorporated herein by reference. The polypeptides and antibodies of the present invention may be used with or without modification, including chimeric or humanized antibodies. Frequently, the polypeptides and antibodies will be labeled by joining, either covalently or non-covalently, a substance which provides for a detectable signal . A wide variety of labels and conjugation techniques are known and are reported extensively in both the scientific and patent literature. Suitable labels include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent moieties, chemiluminescent moieties, magnetic particles, and the like. Patents, teaching the use of such labels include U.S. Patent Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241. Also, recombinant or chimeric immunoglobulins may be produced, see Cabilly, U.S. Patent No. 4,816,567; or made in transgenic mice, see Mendez, et al. (1997) Nature Genetics 15:146-156. These references are incorporated herein by reference .
The antibodies of this invention can also be used for affinity chromatography in isolating the IL-lRs.
Columns can be prepared where the antibodies are linked to a solid support, e.g., particles, such as agarose, Sephadex, or the like, where a cell lysate may be passed through the column, the column washed, followed by increasing concentrations of a mild denaturant, whereby the purified protein will be released. The protein may be used to purify antibody.
The antibodies may also be used to screen expression libraries for particular expression products . Usually the antibodies used in such a procedure will be labeled with a moiety allowing easy detection of presence of antigen by antibody binding.
Antibodies raised against an IL-1R will also be used to raise anti-idiotypic antibodies. These will be useful in detecting or diagnosing various immunological conditions related to expression of the protein or cells which express the protein. They also will be useful as agonists or antagonists of the ligand, which may be competitive inhibitors or substitutes for naturally occurring ligands.
An IL-1R polypeptide that specifically binds to or that is specifically immunoreactive with an antibody generated against a defined immunogen, such as an immunogen consisting of the amino acid sequence of, e.g., SEQ ID NO: 4, 10, or 20, is typically determined in an immunoassay. The immunoassay typically uses a polyclonal antiserum which was raised, e.g., to a polypeptide of SEQ ID NO: 4, 10, or 20. This antiserum is selected to have low crossreactivity against other IL-1R family members, e.g., IL-lRs Dl through D8, preferably from the same species, and any such crossreactivity is removed by immunoabsorption prior to use in the immunoassay.
To produce antisera for use in an immunoassay, the polypeptide of, e.g., SEQ ID NO: 4, 10, or 20, is" * isolated as described herein. For example, recombinant polypeptide may be produced in a mammalian cell line. An appropriate host, e.g., an inbred strain of mice such as Balb/c, is immunized with the selected protein, typically using a standard adjuvant, such as Freund's adjuvant, and a standard mouse immunization protocol (see Harlow and Lane, supra) . Alternatively, a synthetic peptide derived from the sequences disclosed herein and conjugated to a carrier polypeptide can be used an immunogen. Polyclonal sera are collected and titered against the immunogen polypeptide in an immunoassay, e.g., a solid phase immunoassay with the immunogen immobilized on a solid support. Polyclonal antisera with a titer of 10^ or greater are selected and tested for their cross reactivity against other IL-1R family members, e.g., IL- 1RD1 through IL-1RD6, using a competitive binding immunoassay such as the one described in Harlow and Lane, supra, at pages 570-573. Preferably at least two IL-1R family members are used in this determination. These IL- 1R family members can be produced as recombinant polypeptides and isolated using standard molecular biology and protein chemistry techniques as described herein.
Immunoassays in the competitive binding format can be used for the crossreactivity determinations. For example, the polypeptide of SEQ ID NO: 4, 10, or 20 can be immobilized to a solid support. Polypeptides added to the assay compete with the binding of the antisera to the immobilized antigen. The ability of the above polypeptides to compete with the binding of the antisera to the immobilized polypeptide is compared to the polypeptides of IL-1RD1 through IL-1RD6. The percent crossreactivity for the above polypeptides is calculated, using standard calculations. Those antisera with less than 10% crossreactivity with each of the polypeptides listed above are selected and pooled. The cross-reacting antibodies are then removed from the pooled antisera by immunoabsorption with the above-listed proteins . The immunoabsorbed and pooled antisera are then used in a competitive binding immunoassay as described above to compare a second polypeptide to the immunogen polypeptide (e.g., the IL-1RD8, IL-1RD9, or IL-1RD10 like polypeptide of SEQ ID NO: 4, 10, or 20) . To make this comparison, the two polypeptides are each assayed at a wide range of concentrations and the amount of each polypeptide required to inhibit 50% of the binding of the antisera to the immobilized polypeptide is determined. If the amount of the second polypeptide required is less than twice the amount of the polypeptide of the selected polypeptide or polypeptides that is required, then the second polypeptide is said to specifically bind to an antibody generated to the immunogen.
It is understood that these IL-1R polypeptides are members of a family of homologous polypeptides that comprise at least 7 genes previously identified. For a particular gene product, such as, e.g., IL-1RD9, the term refers not only to the amino acid sequences disclosed herein, but also to other polypeptides that are allelic, non-allelic, or species variants. It is also understood that the terms include nonnatural mutations introduced by deliberate mutation using conventional recombinant technology such as single site mutation, or by excising short sections of DNA encoding the respective proteins, or by substituting new amino acids, or adding new amino acids . Such minor alterations typically will substantially maintain the immunoidentity of the original molecule and/or its biological activity. Thus, these alterations include polypeptides that are specifically immunoreactive with a designated naturally occurring IL- 1RD8, IL-1RD9, or IL-1RD10 protein. The biological properties of the altered polypeptides can be determined by expressing the polypeptide in an appropriate cell line and measuring the appropriate effect, e.g., upon transfected lymphocytes . Particular polypeptide modifications considered minor would include conservative substitution of amino acids with similar chemical properties, as described above for the IL-1R family as a whole. By aligning a polypeptide optimally with the polypeptide of the IL-IRs and by using the conventional immunoassays described herein to determine immunoidentity, one can determine the polypeptide compositions of the invention.
VII . Kits and quantitation
Both naturally occurring and recombinant forms of the IL-1R like molecules of this invention are particularly useful in kits and assay methods. For example, these methods would also be applied to screening for binding activity, e.g., ligands for these proteins. Several methods of automating assays have been developed in recent years so as to permit screening of tens of thousands of compounds per year. See, e.g., a BIOMEK automated workstation, Beckman Instruments, Palo Alto,
California, and Fodor, et al . (1991) Science 251:767-773, which is incorporated herein by reference. The latter describes means for testing binding by a plurality of defined polymers synthesized on a solid substrate. The development of suitable assays to screen for a ligand or agonist/antagonist homologous polypeptides can be greatly facilitated by the availability of large amounts of purified, soluble IL-IRs in an active state such as is provided by this invention. Purified IL-1RD8, IL-1RD9, or IL-lRDIO can be coated directly onto plates for use in the aforementioned ligand screening techniques. However, non-neutralizing antibodies to these polypeptides can be used as capture antibodies to immobilize the respective receptor on the solid phase, useful, e.g., in diagnostic uses.
This invention also contemplates use of IL-1RD8, IL- 1RD9, or IL-lRDlO fragments thereof, peptides, and their fusion products in a variety of diagnostic kits and methods for detecting the presence of the protein or its ligand. Alternatively, or additionally, antibodies against the molecules may be incorporated into the kits and methods. Typically the kit will have a compartment containing, e.g., either an IL-1RD9 peptide or gene segment or a reagent which recognizes one or the other. Typically, recognition reagents, in the case of peptide, would be a ligand or antibody, or in the case of a gene segment, would usually be a hybridization probe.
A preferred kit for determining the concentration of IL-1RD8, IL-1RD9, or IL-lRDlO in a sample would typically comprise a labeled compound, e.g., ligand or antibody, having known binding affinity for IL-1RD9, a source of IL-1RD9 (naturally occurring or recombinant) as a positive control, and a means for separating the bound from free labeled compound, for example a solid phase for immobilizing the IL-1RD9 in the test sample. Compartments containing reagents, and instructions, will normally be provided.
Antibodies, including antigen binding fragments, specific for mammalian IL-1RD8 or a peptide fragment, or receptor fragments are useful in diagnostic applications to detect the presence of elevated levels of ligand and/or its fragments. Diagnostic assays may be homogeneous (without a separation step between free reagent and antibody-antigen complex) or heterogeneous (with a separation step) . Various commercial assays exist, such as radioimmunoassay (RIA) , enzyme-linked immunosorbent assay (ELISA) , enzyme immunoassay (EIA) , enzyme-multiplied immunoassay technique (EMIT) , substrate-labeled fluorescent immunoassay (SLFIA) and the like. For example, unlabeled antibodies can be employed by using a second antibody which is labeled and which recognizes the antibody to an IL-1R or to a particular fragment thereof . These assays have also been extensively discussed in the literature. See, e.g.,
Harlow and Lane (1988) Antibodies: A Laboratory Manual, CSH., and Coligan (ed. 1991) and periodic supplements, Current Protocols In Immunology Greene/Wiley, New York. Anti-idiotypic antibodies may have similar use to serve as agonists or antagonists of IL-IRs . These should be useful as therapeutic reagents under appropriate circumstances . Frequently, the reagents for diagnostic assays*" are supplied in kits, so as to optimize the sensitivity of the assay. For the subject invention, depending upon the nature of the assay, the protocol, and the label, either labeled or unlabeled antibody, or labeled ligand is provided. This is usually in conjunction with other additives, such as buffers, stabilizers, materials necessary for signal production such as substrates for enzymes, and the like. Preferably, the kit will also contain instructions for proper use and disposal of the contents after use. Typically the kit has compartments for each useful reagent, and will contain instructions for proper use and disposal of reagents. Desirably, the reagents are provided as a dry lyophilized powder, where the reagents may be reconstituted in an aqueous medium having appropriate concentrations for performing the assay.
The aforementioned constituents of the diagnostic assays may be used without modification or may be modified in a variety of ways. For example, labeling may be achieved by covalently or non-covalently joining a moiety which directly or indirectly provides a detectable signal. In many of these assays, a test compound, IL-1R, or antibodies thereto can be labeled either directly or indirectly. Possibilities for direct labeling include label groups: radiolabels such as ^-^1 , enzymes (U.S. Pat. No. 3,645,090) such as peroxidase and alkaline phosphatase, and fluorescent labels (U.S. Pat. No. 3,940,475) capable of monitoring the change in fluorescence intensity, wavelength shift, or fluorescence polarization. Both of the patents are incorporated herein by reference. Possibilities for indirect labeling include biotinylation of one constituent followed by binding to avidin coupled to one of the above label groups . There are also numerous methods of separating the bound from the free ligand, or alternatively the bound from the free test compound. The IL-1R can be immobilized on various matrixes followed by washing. Suitable matrices include plastic such as an ELISA plate, filters, and beads. Methods of immobilizing the receptor to a matrix include, without limitation, direct adhesion to plastic, use of a capture antibody, chemical coupling, and biotin-avidin. The last step in this approach involves the precipitation of antibody/antigen complex by any of several methods including those utilizing, e.g., an organic solvent such as polyethylene glycol or a salt such as ammonium sulfate. Other suitable separation techniques include, without limitation, the fluorescein antibody magnetizable particle method described in
Rattle, et al . (1984) Clin. Chem. 30 (9) : 1457-1461, and the double antibody magnetic particle separation as described in U.S. Pat. No. 4,659,678, each of which is incorporated herein by reference. The methods for linking protein or fragments to various labels have been extensively reported in the literature and do not require detailed discussion here. Many of the techniques involve the use of activated carboxyl groups either through the use of carbodiimide or active esters to form peptide bonds, the formation of thioethers by reaction of a mercapto group with an activated halogen such as chloroacetyl, or an activated olefin such as maleimide, for linkage, or the like. Fusion polypeptides will also find use in these applications.
Another diagnostic aspect of this invention involves use of oligonucleotide or polynucleotide sequences taken from the sequence of an IL-1R. These sequences can be used as probes for detecting levels of the respective IL- 1R in patients suspected of having an immunological disorder. The preparation of both RNA and DNA nucleotide sequences, the labeling of the sequences, and the preferred size of the sequences has received ample description and discussion in the literature. Normally an oligonucleotide probe should have at least about 14 nucleotides, usually at least about 18 nucleotides, and the polynucleotide probes may be up to several kilobases . Various labels may be employed, most commonly radionuclides, particularly 32p. However, other techniques may also be employed, such as using biotin modified nucleotides for introduction into a polynucleotide. The biotin then serves as the site for binding to avidin or antibodies, which may be labeled with a wide variety of labels, such as radionuclides, fluorescers, enzymes, or the like. Alternatively, antibodies may be employed which can recognize specific duplexes, including DNA duplexes, RNA duplexes, DNA-RNA hybrid duplexes, or DNA-protein duplexes. The antibodies in turn may be labeled and the assay carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected. The use of probes to the novel anti-sense RNA may be carried out in conventional techniques such as nucleic acid hybridization, plus and minus screening, recombinational probing, hybrid released translation (HRT) , and hybrid arrested translation (HART) . This also includes amplification techniques such as polymerase chain reaction (PCR) .
Diagnostic kits which also test for the qualitative or quantitative presence of other markers are also contemplated. Diagnosis or prognosis may depend on the combination of multiple indications used as markers . Thus, kits may test for combinations of markers. See, e.g., Viallet, et al. (1989) Progress in Growth Factor Res. 1:89-97.
VIII. Therapeutic Utility This invention provides reagents with significant therapeutic value. The IL-IRs (naturally occurring or recombinant) , fragments thereof, mutein receptors, and antibodies, along with compounds identified as having binding affinity to the receptors or antibodies, should be useful in the treatment of conditions exhibiting abnormal expression of the receptors of their ligands. Such abnormality will typically be manifested by immunological disorders. Additionally, this invention should provide therapeutic value in various disease's or disorders associated with abnormal expression or abnormal triggering of response to the ligand. The IL-1 ligands have been suggested to be involved in morphologic development, e.g., dorso-ventral polarity determination, and immune responses, particularly the primitive innate responses. See, e.g., Sun, et al . (1991) Eur . J . Biochem. 196:247-254; Hultmark (1994) Nature 367:116-117.
Recombinant IL-IRs, muteins, agonist or antagonist antibodies thereto, or antibodies can be purified and then administered to a patient. These reagents can be combined for therapeutic use with additional active ingredients, e.g. , in conventional pharmaceutically acceptable carriers or diluents, along with physiologically innocuous stabilizers and excipients. These combinations can be sterile, e.g., filtered, and placed into dosage forms as by lyophilization in dosage vials or storage in stabilized aqueous preparations. This invention also contemplates use of antibodies or binding fragments thereof which are not complement binding.
Ligand screening using IL-1R or fragments thereof can be performed to identify molecules having binding affinity to the receptors. Subsequent biological assays can then be utilized to determine if a putative ligand can provide competitive binding, which can block intrinsic stimulating activity. Receptor fragments can be used as a blocker or antagonist in that it blocks the activity of ligand. Likewise, a compound having intrinsic stimulating activity can activate the receptor and is thus an agonist in that it simulates the activity of ligand, e.g., inducing signaling. This invention further contemplates the therapeutic use of antibodies to IL-IRs as antagonists. The quantities of reagents necessary for effective therapy will depend upon many different factors, including means of administration, target site, reagent physiological life, pharmacological life, physiological state of the patient, and other medicants administered. Thus, treatment dosages should be titrated to optimize safety and efficacy. Typically, dosages used in vitro may provide useful guidance in the amounts useful for in situ administration of these reagents. Animal testing of effective doses for treatment of particular disorders will provide further predictive indication of human dosage. Various considerations are described, e.g., in Gilman, et al . (eds., 1990) Goodman and Gilman's: The Pharmacological Bases of Therapeutics, 8th Ed. , Pergamon Press; and Remington ' s Pharmaceutical Sciences , 17th ed. (1990), Mack Publishing Co., Easton, Perm. ; each of which is hereby incorporated herein by reference. Methods for administration are discussed therein and below, e.g., for oral, intravenous, intraperitoneal, or intramuscular administration, transdermal diffusion, and others.
Pharmaceutically acceptable carriers will include water, saline, buffers, and other compounds described, e.g., in the Merck Index, Merck & Co., Rahway, New Jersey. Because of the likely high affinity binding, or turnover numbers, between a putative ligand and its receptors, low dosages of these reagents would be initially expected to be effective. And the signaling pathway suggests extremely low amounts of ligand may have effect. Thus, dosage ranges would ordinarily be expected to be in amounts lower than 1 mM concentrations, typically less than about 10 μM concentrations, usually less than about 100 nM, preferably less than about 10 pM (picomolar) , and most preferably less than about 1 fM (femtomolar) , with an appropriate carrier. Slow release formulations, or slow release apparatus will often be utilized for continuous administration.
IL-IRs, fragments thereof, and antibodies or its fragments, antagonists, and agonists, may be administered directly to the host to be treated or, depending on the size of the compounds, it may be desirable to conjugate them to carrier proteins such as ovalbumin or serum albumin prior to their administration. Therapeutic formulations may be administered in many conventional dosage formulations . While it is possible for the active ingredient to be administered alone, it is preferable to present it as a pharmaceutical formulation. Formulations comprise at least one active ingredient, as defined above, together with one or more acceptable carriers thereof. Each carrier must be both pharmaceutically and physiologically acceptable in the sense of being compatible with the other ingredients and not injurious to the patient. Formulations include those suitable for oral, rectal, nasal, or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration. The formulations may conveniently be presented in unit dosage form and may be prepared by methods well known in the art of pharmacy. See, e.g., Gilman, et al . (eds. 1990) Goodman and Gilman's: The Pharmacological Bases of Therapeutics , 8th Ed. , Pergamon Press; and Remington's Pharmaceutical Sciences, 17th ed. (1990), Mack Publishing Co., Easton, Perm.; Avis, et al . (eds. 1993) Pharmaceutical Dosage Forms : Parenteral Medications Dekker, NY; Lieberman, et al . (eds. 1990) Pharmaceutical Dosage Forms : Tablets Dekker, NY; and Lieberman, et al. (eds. 1990) Pharmaceutical Dosage Forms : Disperse Systems Dekker, NY. The therapy of this invention may be combined with or used in association with other therapeutic agents, particularly agonists or antagonists of other IL-1 family members.
IX. Ligands
The description of the IL-1 receptors herein provide means to identify ligands, as described above. Such ligand should bind specifically to the respective receptor with reasonably high affinity. Typical ligand receptor binding constants will be at least about 30 mM, e.g., generally at least about 3 mM, more generally at least about 300 μM, typically at least about 30 μM, 3 μM, 300 nM, 30 nM, etc. Various constructs are made available which allow either labeling of the receptor to detect its ligand. For example, directly labeling IL-1R, fusing onto it markers for secondary labeling, e.g., FLAG or other epitope tags, etc., will allow detection of receptor. This can be histological, as an affinity" method for biochemical purification, or labeling or selection in an expression cloning approach. A two- hybrid selection system may also be applied making appropriate constructs with the available IL-1R sequences. See, e.g., Fields and Song (1989) Nature 340:245-246.
Generally, descriptions of IL-IRs will be analogously applicable to individual specific embodiments directed to IL-1RD8, IL-1RD9, OR IL-lRDIO reagents and compositions .
The broad scope of this invention is best understood with reference to the following examples, which are not intended to limit the inventions to the specific embodiments .
EXAMPLES
I. General Methods
Some of the standard methods are described or referenced, e.g., in Maniatis, et al . (1982) Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor Press; Sambrook, et al . (1989) Molecular Cloning: A Laboratory Manual, (2d ed. ) , vols. 1-3, CSH Press, NY; Ausubel, et al . Biology Greene Publishing Associates, Brooklyn, NY; or Ausubel, et al .
(1987 and Supplements) Current Protocols in Molecular
Biology, Greene/Wiley, New York. Methods for protein purification include such methods as ammonium sulfate precipitation, column chromatography, electrophoresis, centrifugation, crystallization, and others. See, e.g., Ausubel, et al. (1987 and periodic supplements); Coligan, et al. (ed. 1996 and periodic supplements) Current Protocols In Protein Science Greene/Wiley, New York; Deutscher (1990) "Guide to Protein Purification" in Methods in Enzvmology, vol. 182, and other volumes in this series; and manufacturer's literature on use of protein purification products, e.g., Pharmacia, Piscataway, N.J., or Bio-Rad, Richmond, CA. Combination with recombinant techniques allow fusion to appropriate segments, e.g., to a FLAG sequence or an equivalent" which can be fused via a protease-removable sequence. See, e.g., Hochuli (1989) Chemische Industrie 12:69-70; Hochuli (1990) "Purification of Recombinant Proteins with Metal Chelate Absorbent" in Setlow (ed.) Genetic Engineering, Principle and Methods 12:87-98, Plenum
Press, N.Y.; and Crowe, et al . (1992) OIAexpress : The High Level Expression & Protein Purification System QUIAGEN, Inc., Chatsworth, CA.
Computer sequence analysis is performed, e.g., using available software programs, including those from the GCG (U. Wisconsin) and GenBank sources. Public sequence databases were also used, e.g., from GenBank, NCBI, SWISSPROT, and others.
Many techniques applicable to IL-10 receptors may be applied to IL-IRs, as described, e.g., in USSN 08/110,683 (IL-10 receptor) , which is incorporated herein by reference for all purposes. Also, while many of the techniques described are directed to the IL-1RD9 reagents, corresponding methods will typically be applicable with the IL-1RD8, and IL-1RD10 reagents. See also, USSN 60/065,776, filed November 17, 1997, and USSN 60/078,008, filed March 12, 1998, both of which are incorporated herein by reference.
II. Computational Analysis.
Human sequences related to IL-IRs were identified from various EST databases using, e.g., the BLAST server (Altschul, et al . (1994) Nature Genet. 6:119-129). More sensitive pattern- and profile-based methods (Bork and Gibson (1996) Meth. Enzvmol. 266:162-184) were used to identify a fragment of a gene which exhibited certain homology to the IL-IRs .
III. Cloning of full-length human IL-1R cDNAs . PCR primers derived from the IL-1RD8, IL-1RD9, or IL-lRDIO sequences are used (Nomura, et al . (1994) DNA Res . 1:27-35) to probe an appropriate human cDNA library to yield a full length IL-1RD9 or IL-1RD10 cDNA sequence or to probe a human erythroleukemic, TF-1 cell line- derived cDNA library (Kitamura, et al . (1989) Blood 73:375-380) to yield the IL-1R8 cDNA sequence. Full length cDNAs for human IL-1RD9 are cloned, e.g., by DNA hybridization screening of λgtlO phage. PCR reactions were conducted using T. aquaticus Taqplus DNA polymerase (Stratagene) under appropriate conditions.
IV. Localization of IL-1RD8, IL-1RD9, and IL-1RD10 mRNA Human multiple tissue (Cat# 1, 2) and cancer cell line blots (Cat# 7757-1) , containing approximately 2 μg of poly (A) + RNA per lane, are purchased from Clontech (Palo Alto, CA) . Probes are radiolabeled with [α-32p] dATP, e.g., using the Amersham Rediprime random primer labeling kit (RPN1633) . Prehybridization and hybridizations are performed at 65° C in 0.5 M Na2HPθ4,
7% SDS, 0.5 M EDTA (pH 8.0) . High stringency washes are conducted, e.g., at 65° C with two initial washes in 2 x SSC, 0.1% SDS for 40 min followed by a subsequent wash in 0.1 x SSC, 0.1% SDS for 20 min. Membranes are then exposed at -70° C to X-Ray film (Kodak) in the presence of intensifying screens. More detailed studies by cDNA library Southerns are performed with selected human IL- 1RD9 clones to examine their expression in hemopoietic or other cell subsets . Two prediction algorithms that take advantage of the patterns of conservation and variation in multiply aligned sequences, PHD (Rost and Sander (1994) Proteins 19:55-72) and DSC (King and Sternberg (1996) Protein Sci. 5:2298-2310), are used. Alternatively, two appropriate primers are selected from Tables 1, 2, or 3. RT-PCR is used on an appropriate mRNA sample selected for the presence of message to produce a cDNA, e.g., a sample which expresses the gene. Full length clones may be isolated by hybridization of cDNA libraries from appropriate tissues pre-selected by PCR signal. Northern blots can be performed.
Message for genes encoding, e.g., IL-1RD9 will be assayed by appropriate technology, e.g., PCR, immunoassay, hybridization, or otherwise. Tissue and organ cDNA preparations are available, e.g., from Clontech, Mountain View, CA. Identification of sources of natural expression are useful, as described. And the identification of functional receptor subunit pairings will allow for prediction of what cells express the combination of receptor subunits which will result in a physiological responsiveness to each of the IL-1 ligands. The message for IL-1RD9 is quite rare, as it is not found with a degree of frequency in the available sequence databases. This suggests, e.g., a very rare message, or a highly restricted distribution. IL-1R9 is expressed predominantly on T cells, NK cells, monocytes and dendritic cells . Southern Analysis on cDNA libraries can be performed:
DNA (5 μg) from a primary amplified cDNA library is digested with appropriate restriction enzymes to release the inserts, run on a 1% agarose gel and transferred to a nylon membrane' (Schleicher and Schuell, Keene, NH) . Samples for human mRNA isolation may include, e.g. : peripheral blood mononuclear cells (monocytes, T cells, NK cells, granulocytes, B cells), resting (T100); peripheral blood mononuclear cells, activated with anti- CD3 for 2, 6, 12 h pooled (T101) ; T cell, THO clone Mot 72, resting (T102) ; T cell, THO clone Mot 72, activated with anti-CD28 and anti-CD3 for 3, 6, 12 h pooled (T103); T cell, THO clone Mot 72, anergic treated with specific peptide for 2, 7, 12 h pooled (T104) ; T cell, THl clone HY06, resting (T107) ; T cell, THl clone HY06, activated with anti-CD28 and anti-CD3 for 3, 6, 12 h pooled (T108) ; T cell, THl clone HY06, anergic treated with specific peptide for 2, 6, 12 h pooled (T109) ; T cell, TH2 clone HY935, resting (THO) ; T cell, TH2 clone HY935, activated with anti-CD28 and anti-CD3 for 2, 7, 12 h pooled (Till); T cells CD4+CD45RO- T cells polarized 27 days in anti- CD28, IL-4, and anti IFN-γ, TH2 polarized, activated with anti-CD3 and anti-CD28 4 h (T116) ; T cell tumor lines Jurkat and Hut78, resting (T117) ; T cell clones, pooled AD130.2, Tc783.12, Tc783.13, Tc783.58, Tc782.69, resting (T118); T cell random γδ T cell clones, resting (T119) ;
Splenocytes, resting (B100); Splenocytes, activated with anti-CD40 and IL-4 (B101) ; B cell EBV lines pooled WT49, RSB, JY, CVIR, 721.221, RM3 , HSY, resting (B102); B cell line JY, activated with PMA and ionomycin for 1, 6 h pooled (B103); NK 20 clones pooled, resting (K100) ; NK 20 clones pooled, activated with PMA and ionomycin for 6 h (K101) ; NKL clone, derived from peripheral blood of LGL leukemia patient, IL-2 treated (K106) ; NK cytotoxic clone 640-A30-1, resting (K107) ; hematopoietic precursor line TF1, activated with PMA and ionomycin for 1, 6 h pooled (C100); U937 premonocytic line, resting (M100) ; U937 premonocytic line, activated with PMA and ionomycin for 1, 6 h pooled (M101) ; elutriated monocytes, activated with LPS, IFNγ, anti-IL-10 for 1, 2, 6, 12, 24 h pooled (M102); elutriated monocytes, activated with LPS, IFNγ,
IL-10 for 1, 2, 6, 12, 24 h pooled (M103); elutriated monocytes, activated with LPS, IFNγ, anti-IL-10 for 4, 16 h pooled (M106) ; elutriated monocytes, activated with LPS, IFNγ, IL-10 for 4, 16 h pooled (M107); elutriated monocytes, activated LPS for 1 h (M108) ; elutriated monocytes, activated LPS for 6 h (M109) ; DC 70% CDla+, from CD34+ GM-CSF, TNFα 12 days, resting (D101) ; DC 70% CDla+, from CD34+ GM-CSF, TNFα 12 days, activated with PMA and ionomycin for 1 hr (D102); DC 70% CDla+, from CD34+ GM-CSF, TNFα 12 days, activated with PMA and ionomycin for 6 hr (D103); DC 95% CDla+, from CD34+ GM- CSF, TNFα 12 days FACS sorted, activated with PMA and ionomycin for 1, 6 h pooled (D104) ; DC 95% CD14+, ex CD34+ GM-CSF, TNFα 12 days FACS sorted, activated with
PMA and ionomycin 1, 6 hr pooled (D105) ; DC CDla+ CD86+, from CD34+ GM-CSF, TNFα 12 days FACS sorted, activated with PMA and ionomycin for 1, 6 h pooled (D106) ; DC from monocytes GM-CSF, IL-4 5 days, resting (D107); DC from monocytes GM-CSF, IL-4 5 days, resting (D108) ; DC from monocytes GM-CSF, IL-4 5 days, activated LPS 4, 16 h pooled (D109); DC from monocytes GM-CSF, IL-4 5 days, activated TNFα, monocyte supe for 4, 16 h pooled (DUO) ; leiomyoma Lll benign tumor (X101) ; normal myometrium M5 (0115); malignant leiomyosarcoma GS1 (X103); lung fibroblast sarcoma line MRC5, activated with PMA and ionomycin for 1, 6 h pooled (C101) ; kidney epithelial carcinoma cell line CHA, activated with PMA and ionomycin for 1, 6 h pooled (C102); kidney fetal 28 wk male (O100) ; lung fetal 28 wk male (O101) ; liver fetal 28 wk male (O102); heart fetal 28 wk male (0103); brain fetal 28 wk male (0104) ; gallbladder fetal 28 wk male (0106) ; small intestine fetal 28 wk male (O107) ; adipose tissue fetal 28 wk male (0108) ; ovary fetal 25 wk female (0109) ; uterus fetal 25 wk female (OllO); testes fetal 28 wk male (0111); spleen fetal 28 wk male (0112); adult placenta 28 wk (0113); and tonsil inflamed, from 12 year old (X100) ; psoriasis human skin sample; normal human skin sample; pool of rheumatioid arthritis human; Hashimoto's thryroiditis thryroid; normal human throid; ulceratived colitis human colon; normal human colon; normal weight monkey colon; pheumocysitc carnii pneumonia lung; allergic lung; pool of three heavy smoker human lung; pool of two normal human lung; Ascaris-challenged monkey lung, 24hr; Ascaris-challenged monkey lung, 4hr; normal weight monkey lung..
IL-1RD8 message is described below in Table 5. There appears to be a correlation between developmental stage of tissues and the levels of messages: fetal and transformed tissues express high levels, whereas normal, adult tissues express low levels (with the exception of skeletal muscle) . Further insights into this phenomenon will need further experiments. Message for genes encoding IL-1RD8 will be assayed by appropriate technology, e.g., PCR, immunoassay, hybridization, or otherwise. Tissue and organ cDNA preparations are available, e.g., from Clontech, Mountain View, CA. Identification of sources of natural expression are useful, as described. And the identification of functional receptor subunit pairings will allow for prediction of what cells express the combination of receptor subunits which will result in a physiological responsiveness to each of the IL-1 ligands.
Table 5
Multiple Tissue Northern Blots were screened with a radiolabeled probe, encompassing the cytoplasmic region of Interleu in-1 receptor R8 (IL-1RD8) . The results are summarized below:
In all cases listed there is a smaller band at 3.4 Kb and in a few cases a larger band at 4.0 Kb as well . Tissue 3.4 kb 4.0 kb
Spleen weak
Thymus weak
Prostate weak Testis weak
Ovary weak
Small Intestine weak
Colon (mucosal lining) weak
Peripheral Blood Leukocyte weak Heart moderate
Brain weak
Placenta moderate
Lung weak
Liver weak Skeletal Muscle strong
Kidney weak
Pancreas weak
Fetal brain strong weak
Fetal lung strong weak Fetal Liver strong weak
Fetal Kidney strong weak proleukocytic leukemia HL-60 strong
HeLa Cell S3 very strong weak Chronic myelogenous leukemia, K-562 very strong weak
Lymphoblastic leukemia, MOLT-4 weak
Burkitt's lymphoma Rajii moderate
Colorectal adenocarcinoma S 40 very strong strong
Lung carcinoma A549 strong strong Melanoma very strong weak
V. Cloning of species counterparts of IL-lRDs Various strategies are used to obtain species counterparts of IL-1RD8, IL-1RD9, and IL-1RD10 preferably from other primates . One method is by cross hybridization using closely related species DNA probes. It may be useful to go into evolutionarily similar species as intermediate steps . Another method is by using specific PCR primers based on the identification of blocks of similarity or difference between genes, e.g., areas of highly conserved or nonconserved polypeptide or nucleotide sequence. In addition, gene sequence databases may be screened for related sequences from other species .
VI. Production of mammalian IL-1RD8, IL-1RD9, and IL- 1RD10 protein
An appropriate, e.g., GST, fusion construct is engineered for expression, e.g., in E. coli. For example, a mouse IGIF pGex plasmid is constructed and transformed into E. coli. Freshly transformed cells are grown, e.g.., in LB medium containing 50 μg/ml ampicillin and induced with IPTG (Sigma, St. Louis, MO). After overnight induction, the bacteria are harvested and the pellets containing, e.g., the IL-1R8 polypeptide are isolated. The pellets are homogenized, e.g., in TE buffer (50 mM Tris-base pH 8.0, 10 mM EDTA and 2 mM pefabloc) in 2 liters. This material is passed through a microfluidizer (Microfluidics, Newton, MA) three times.
The fluidized supernatant is spun down on a Sorvall GS-3 rotor for 1 h at 13,000 rpm. The resulting supernatant containing the IL-1R polypeptide is filtered and passed over a glutathione-SEPHAROSE column equilibrated in 50 mM Tris-base pH 8.0. The fractions containing the IL-1RD9- GST fusion protein are pooled and cleaved, e.g., with thrombin (Enzyme Research Laboratories, Inc., South Bend, IN) . The cleaved pool is then passed over a Q-SEPHAROSE column equilibrated in 50 mM Tris-base. Fractions containing IL-1RD9 are pooled and diluted in cold distilled H2O, to lower the conductivity, and passed back over a fresh Q-Sepharose column, alone or in succession with an immunoaffinity antibody column.. Fractions containing the IL-1RD9 polypeptide are pooled, aliquoted, and stored in the -70° C freezer.
Comparison of the CD spectrum with IL-1R polypeptide may suggest that the protein is correctly folded. See Hazuda, et al. (1969) J. Biol. Chem. 264:1689-1693. VII. Determining physiological forms of receptors
The IL-lα and IL-lβ ligands bind an IL-1RD1 as the primary receptor and this complex then forms a high affinity receptor complex with the IL-1RD3. Such receptor subunits are probably shared with the receptors for the new IL-1 ligand family members. See, e.g., USSN 60/044,165 and USSN 60/055,111. Combination of the IL- 1RD9 (α subunit type, based upon sequence analysis) will combine with the IL-1RD5 (β subunit type, based upon sequence analysis) to form a heterodimer receptor. The IL-lδ and IL-lε ligands each probably signal through a receptor comprising the association of IL-1RD4, IL-1RD6, or IL-1RD9 (alpha components) with IL-1RD3 , IL-1RD8, or IL-1RD10 (beta components) . These defined subunit combinations can be tested now with the provided reagents. In particular, appropriate constructs can be made for transformation or transfeetion of subunits into cells. Constructs for the alpha chains, e.g., IL-1RD1, IL-1RD4, IL-1RD6, and IL-1RD9 forms can be made. Likewise for the beta subunits IL-1RD3, IL-1RD5,
IL-1RD7, and IL-1RD8. Structurally, the IL-1RD10 is most similar to the IL-1RD8 , suggesting that it may also be a beta receptor subunit. Combinatorial transfections of transformations can make cells expressing defined subunits, which can be tested for response to each of the IL-1 ligands. Appropriate cell types can be used, e.g., 293 T cells, Jurkat cells, with, e.g., a nuclear kappa B (NFKb) -controlled luciferase reporter construct such as described e.g., in Otieno et al.,(1997) Am J Physiol 273-xxx.
Such combinations of various IL-1 ligands and receptors were tested to determine if a functional signaling complex had been formed using an NFKb- controlled luciferase reporter construct to indicate formation of a functional signaling complex (+) or failure to form a functional signaling complex (-) . The results, presented below,
IL-lα + IL-lβ + IL-1RD1 + IL-1RD3 = +; IL-lα + IL-lβ + IL-1RD1 + IL-1RD5 = +; IL-lα + IL-lβ + IL-1RD1 + IL-1RD8 = +; IL-lα + IL-lβ + IL-1RD1 + IL-lRDIO may = +/?;
suggest that IL-1RD3, IL-1RD5, IL-1RD8, and IL-lRDIO may functionally substitute for each other when in combination with IL-lα + IL-lβ + IL-1RD1.
Other combinations (below) demonstrate a failure of functional substitution; suggesting the importance of contextual dependence on substitution e.g., IL-1RD3, and IL-1RD8 cannot functionally replace IL-1RD5 in the following combination: IL-lγ+ IL-1RD9 + IL-1RD5.
IL-lγ+ IL-1RD9 + IL-1RD5 = + IL-lγ+ IL-1RD9 + IL-1RD3 = -
IL-lγ+ IL-1RD9 + IL-1RD8 = -
A further series of experiments tested the ability of mouse (m) and human (h) homologues to functionally substitute for each other. The results, shown below,
mIL-lγ+mIL-lRD5 + mIL-lRD9 = + mIL-lγ+mIL-lRD5 + ML-1RD9 = - mIL-lγ+hIL-lRD5 + ML-1RD9 = - mIL-lγ+hIL-lRD5 + mIL-lRD9 = -
hIL-lγ+mIL-lRD5 + mIL-lRD9 = - hIL-lγ+mIL-lRD5 + ML-1RD9 = - hIL-lγ+hIL-lRD5 + mIL-lRD9 = - hIL-lγ+hIL-lRD5 + hIL-lRD9 = +
suggest that species homogeneity is required to form a functioning complex in this particular constellation of ligand and receptor units . Biological assays will generally be directed to the ligand binding feature of the protein or to the kinase/phosphatase activity of the receptor. The activity will typically be reversible, as are many other enzyme actions mediate phosphatase or phosphorylase activities, which activities are easily measured by standard procedures. See, e.g., Hardie, et al . (eds. 1995) The Protein Kinase FactBook vols. I and II, Academic Press, San Diego, CA; Hanks, et al . (1991) Meth. Enzvmol . 200:38-62; Hunter, et al. (1992) Cell 7θ":3,75- 388; Lewin (1990) Cell 61:743-752; Pines, et al . (1991) Cold Spring Harbor Svmp. Quant. Biol. 56:449-463; and Parker, et al . (1993) Nature 363:736-738.
The family of interleukins 1 contains molecules, each of which is an important mediator of inflammatory disease. For a comprehensive review, see Dinarello (1996) "Biologic basis for interleukin-1 in disease" Blood 87:2095-2147. There are suggestions that the various IL-1 ligands may play important roles in the initiation of disease, particularly inflammatory responses . The finding of novel polypeptides related to the IL-1 family furthers the identification of molecules that provide the molecular basis for initiation of disease and allow for the development of therapeutic strategies of increased range and efficacy.
VIII. Preparation of antibodies specific for IL-IRs
Inbred Balb/c mice are immunized intraperitoneally with recombinant forms of the polypeptide, e.g., purified IL-1RD8, IL-1RD9, and IL-1RD10, or stable transfected
NIH-3T3 cells. Animals are boosted at appropriate time points with protein, with or without additional adjuvant, to further stimulate antibody production. Serum is collected, or hybridomas produced with harvested spleens. Alternatively, Balb/c mice are immunized with cells transformed with the gene or fragments thereof, either endogenous or exogenous cells, or with isolated membranes enriched for expression of the antigen. Serum is collected at the appropriate time, typically after numerous further administrations. Various gene therapy techniques may be useful, e.g., in producing protein in situ, for generating an immune response.
Monoclonal antibodies may be made. For example, splenocytes are fused with an appropriate fusion partner and hybridomas are selected in growth medium by standard procedures . Hybridoma supernatants are screened for the presence of antibodies which bind to the desired IL-IR, e.g., by ELISA or other assay. Antibodies which selectively recognize specific IL-IR embodiments may also be selected or prepared.
In another method, synthetic peptides or purified protein are presented to an immune system to generate monoclonal or polyclonal antibodies. See, e.g., Coligan (1991) Current Protocols in Immunology Wiley/Greene; and Harlow and Lane (1989) Antibodies : A Laboratory Manual Cold Spring Harbor Press. In appropriate situations, the binding reagent is either labeled as described above, e.g., fluorescence or otherwise, or immobilized to a substrate for panning methods. Nucleic acids may also be introduced into cells in an animal to produce the antigen, which serves to elicit an immune response. See, e.g., Wang, et al . (1993) Proc. Nat'l. Acad. Sci. 90:4156-4160; Barry, et al . (1994) BioTechni ues 16:616- 619; and Xiang, et al . (1995) Immunity 2:129-135. Moreover, antibodies which may be useful to determine the combination of the IL-1RD8, IL-1RD9, or IL- 1RD10 with a functional beta subunit may be generated. Thus, e.g., epitopes characteristic of a particular functional alpha/beta combination may be identified with appropriate antibodies .
IX. Production of fusion proteins with IL-IRs
Various fusion constructs are made with IL-IRs. A portion of the appropriate gene is fused to an epitope tag, e.g., a FLAG tag, or to a two hybrid system construct. See, e.g., Fields and Song (1989) Nature 340:245-246.
The epitope tag may be used in an expression cloning procedure with detection with anti-FLAG antibodies to detect a binding partner, e.g., ligand for the respective IL-IR. The two hybrid system may also be used to isolate proteins which specifically bind, e.g., to IL-1RD9. X. Structure activity relationship
Information on the criticality of particular residues is determined using standard procedures and analysis. Standard mutagenesis analysis is performed, e.g., by generating many different variants at determined positions, e.g., at the positions identified above, and evaluating biological activities of the variants. This may be performed to the extent of determining positions which modify activity, or to focus on specific positions to determine the residues which can be substituted to either retain, block, or modulate biological activity.
Alternatively, analysis of natural variants can indicate what positions tolerate natural mutations . This may result from population analysis of variation among individuals, or across strains or species. Samples from selected individuals are analyzed, e.g., by PCR analysis and sequencing. This allows evaluation of population polymorphisms .
XI. Isolation of a ligand for IL-IRs
An IL-IR can be used as a specific binding reagent to identify its binding partner, by taking advantage of its specificity of binding, much like an antibody would be used. Typically, the binding receptor is a heterodimer of receptor subunits. A binding reagent is either labeled as described above, e.g., fluorescence or otherwise, or immobilized to a substrate for panning methods .
The binding composition is used to screen an expression library made from a cell line which expresses a binding partner, i.e., ligand, preferably membrane associated. Standard staining techniques are used to detect or sort surface expressed ligand, or surface expressing transformed cells are screened by panning. Screening of intracellular expression is performed by various staining or immunofluorescence procedures. See also McMahan, et al . (1991) EMBO J. 10:2821-2832.
For example, on day 0, precoat 2-chamber permanox slides with 1 ml per chamber of fibronectin, 10 ng/ml in PBS, for 30 min at room temperature. Rinse once with PBS. Then plate COS cells at 2-3 x 105 cells per chamber in 1.5 ml of growth media. Incubate overnight at 37° C. On day 1 for each sample, prepare 0.5 ml of a solution of 66 μg/ml DEAE-dextran, 66 μM chloroqulrife, and 4 μg DNA in serum free DME. For each set, a positive control is prepared, e.g., of IL-1R-FLAG cDNA at 1 and 1/200 dilution, and a negative mock. Rinse cells with serum free DME. Add the DNA solution and incubate 5 hr at 37° C. Remove the medium and add 0.5 ml 10% DMSO in
DME for 2.5 min. Remove and wash once with DME. Add 1.5 ml growth medium and incubate overnight .
On day 2, change the medium. On days 3 or 4, the cells are fixed and stained. Rinse the cells twice with Hank's Buffered Saline Solution (HBSS) and fix in 4% paraformaldehyde (PFA) /glucose for 5 min. Wash 3X with
HBSS. The slides may be stored at -80° C after all liquid is repoved. For each chamber, 0.5 ml incubations are performed as follows. Add HBSS/saponin (0.1%) with 32 μl/ml of 1 M NaN3 for 20 min. Cells are then washed with HBSS/saponin IX. Add appropriate IL-IR or IL- lR/antibody complex to cells and incubate for 30 min. Wash cells twice with HBSS/saponin. If appropriate, add first antibody for 30 min. Add second antibody, e.g., Vector anti-mouse antibody, at 1/200 dilution, and incubate for 30 min. Prepare ELISA solution, e.g., Vector Elite ABC horseradish peroxidase solution, and preincubate for 30 min. Use, e.g., 1 drop of solution A (avidin) and 1 drop solution B (biotin) per 2.5 ml HBSS/saponin. Wash cells twice with HBSS/saponin. Add
ABC HRP solution and incubate for 30 min. Wash cells twice with HBSS, second wash for 2 min, which closes cells. Then add Vector diaminobenzoic acid (DAB) for 5 to 10 min. Use 2 drops of buffer plus 4 drops DAB plus 2 drops of H2O2 per 5 ml of glass distilled water.
Carefully remove chamber and rinse slide in water. Air- dry for a few minutes, then add 1 drop of Crystal Mount and a cover slip. Bake for 5 min at 85-90° C. Evaluate positive staining of , ools and progressively subclone to isolation of single genes responsible for the binding.
Alternatively, IL-IR reagents are used to affinity purify or sort out cells expressing a putative ligand. See, e.g., Sambrook, et al . or Ausubel, et al .
Another strategy is to screen for a membrane bound receptor by panning. The receptor cDNA is constructed as described above. The ligand can be immobilized and used to immobilize expressing cells. Immobilization may be achieved by use of appropriate antibodies which recognize, e.g., a FLAG sequence of an IL-IR fusion construct, or by use of antibodies raised against the first antibodies. Recursive cycles of selection and amplification lead to enrichment of appropriate clones and eventual isolation of receptor expressing clones.
Phage expression libraries can be screened by mammalian IL-IRs. Appropriate label techniques, e.g., anti-FLAG antibodies, will allow specific labeling of appropriate clones.
Many modifications and variations of this invention can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. The specific embodiments described herein are offered by way of example only, and the invention is to be limited by the terms of the appended claims, along with the full scope of equivalents to which such claims are entitled; and the invention is not to be limited by the specific embodiments that have been presented herein by way of example .

Claims

WHAT IS CLAIMED IS:
1. An isolated or recombinant IL-1RD9 polypeptide: a) consisting of SEQ ID NO: 6, 8, 10, 12, 14, or 16; b) encoded by a polynucleotide comprising the open reading frame of SEQ ID NO: 5, 7, 9, 11, 13, or 15; or c) encoded by a naturally occurring allelic variant of a polynucleotide comprising the open reading frame of SEQ ID NO: 5, 7, 9, 11, 13, or 15.
2. The polypeptide of claim 1, encoded by a naturally occurring allelic variant of a polynucleotide comprising the open reading frame of SEQ ID NO: 5, 7, 9, 11, 13, or 15.
3. An isolated or recombinant I1-1RD9 polypeptide which: a) has an apparent molecular weight 68.3 kD as determined by SDS/polyacrylamide gel electrophoresis; b) has as estimated pi of 9.04; and c) is found on T cells; and wherein said polypeptide has at least one of the following properties : i) is a heterodimer; iii) is an IL-1 α subunit type, or iii) when brought into contact with IL-1RD5 and IL- lα, for a sufficient time, forms a functional high affinity receptor complex that activates an NFKB transcription factor reporter construct.
4. An isolated or recombinant polypeptide comprising a segment of contiguous amino acid residues selected from the following group: a) 15 contiguous amino acid residues of said polypeptide of claim 2 ; b) 20 contiguous amino acid residues of said polypeptide of claim 2; c) 25 contiguous amino acid residues of said polypeptide of claim 2 ; d) 30 contiguous amino acid residues of said polypeptide of claim 2; e) 35 contiguous amino acid residues of said polypeptide of claim 2; or f) 40 contiguous amino acid residues of said polypeptide of claim 2.
5. The polypeptide of claim 1 which is immunogenic .
6. An isolated or recombinant polypeptide comprising an immunogenic peptide of said polypeptide of claim 3.
7. An isolated or recombinant polypeptide comprising an immunogenic polypeptide of claim 4.
8. A fusion protein comprising said polypeptide of claim 4 and: a) a detection or purification tag selected from the group consisting of a FLAG, His6, and immunoglobulin peptide; b) a carrier protein selected from the group consisting of keyhole limpet hemocyanin, bovine serum albumin, and tetanus toxoid; or c) another peptide selected from the group consisting of luciferase, bacterial β- galactosidase, trpE, protein A, β-lactamase, alpha amylase, alcohol dehydrogenase, and yeast alpha mating factor.
9. A fusion protein comprising said polypeptide of claim 5 and: a) a detection or purification tag selected from the group consisting of a FLAG, His6, and immunoglobulin peptide; b) a carrier protein selected from the group consisting of keyhole limpet hemocyanin, bovine serum albumin, the tetanus toxoid; or c) another peptide selected from the group consisting of luciferase, bacterial β- - - galactosidase, trpE, protein A, β-lactamase, alpha amylase, alcohol dehydrogenase, and yeast alpha mating factor.
10. A composition comprising said polypeptide of claim
I , that is : a) in a pharmaceutically acceptable carrier; b) in a sterile composition; c) in a buffered solution; or d) in an aqueous suspension.
II . A composition comprising said polypeptide of claim 4, that is: a) in a pharmaceutically acceptable carrier; b) in a sterile composition; c) in a buffered solution; or d) in an aqueous suspension.
12. A polypeptide of claim 4, that is: a) denatured; b) immunopurified; c) attached to a solid substrate; d) detectably labeled; or e) chemically synthesized.
13. A polypeptide of claim 5, that is: a) denatured; b) immunopurified; c) attached to a solid substrate; d) detectably labeled; or e) chemically synthesized.
14. A kit comprising said polypeptide of claim 1, and: a) a compartment comprising said protein; or b) instructions for use or disposal of reagents in said kit.
15. A kit comprising said polypeptide of claim 4, and: a) a compartment comprising said protein; or " b) instructions for use or disposal of reagents in said kit.
16. A method of raising an antibody, comprising immunizing an animal with a polypeptide of claim 5.
17. A method of producing an antibody: antigen complex, comprising contacting a polypeptide of claim 5 with an antibody which specifically binds said polypeptide, thereby forming said complex.
18. A composition of matter selected from the group consisting of: a) a substantially pure or recombinant IL-1RD8 polypeptide exhibiting identity over a length of at least about 12 amino acids to SEQ ID NO: 4; b) a natural sequence IL-1RD8 comprising SEQ ID NO:
4; c) a fusion polypeptide comprising IL-1RD8 sequence; d) a substantially pure or recombinant IL-1RD10 polypeptide exhibiting identity over a length of at least about 12 amino acids to SEQ ID NO: 20; e) a natural sequence IL-1RD10 comprising SEQ ID
NO: 20; and f) a fusion protein comprising IL-1RD10 sequence.
19. A substantially pure or isolated polypeptide comprising a segment exhibiting sequence identity to a corresponding portion of an: a) IL-1RD8 of claim 18, wherein: i) said polypeptide further exhibits identity to a distinct segment of 9 amino acids; ii) said length of identity is at least 17 amino acids; iii) said length of identity is at least" about
25 amino acids; or b) IL-lRDlO of claim 18, wherein: i) said polypeptide further exhibits identity to a distinct segment of 9 amino acids; ii) said length of identity is at least 17 amino acids; iii) said length of identity is at least about 25 amino acids.
20. The composition of matter of claim 18, wherein said: a) IL-1RD8 comprises a mature sequence of SEQ ID NO
2 or 4; b) IL-1RD10 comprises a mature sequence of Seq ID
NO: 18 or 20; or c) polypeptide: i) is from a warm blooded animal selected from a primate, such as a human; ii) comprises at least one polypeptide segment of SEQ ID NO: 4 or 20; iii) exhibits a plurality of portions exhibiting said identity; iv) is a natural allelic variant of a primate or rodent IL-1RD8 or primate IL-lRDIO; v) has a length at least about 30 amino acids; vi) exhibits at least two non-overlapping epitopes which are specific for a primate or rodent IL-1RD8 or primate IL-lRDIO; vii) exhibits a sequence identity at least about 90% over a length of at least about 20 amino acids to a primate IL-1RD8 or IL-
1RD10; viii) has a molecular weight of at least 100 kD with natural glycosylation; ix) is a synthetic polypeptide; x) is attached to a solid substrate; xi) is conjugated to another chemical moiety; xii) is a 5-fold or less substitution from natural sequence; or xiii) is a deletion or insertion variant" from a natural sequence.
21. A composition comprising: a) a sterile IL-1RD8 polypeptide of claim 18; b) said IL-1RD8 protein or peptide of claim 18 and a carrier, wherein said carrier is: i) an aqueous compound, including water, saline, and/or buffer; and/or ii) formulated for oral, rectal, nasal, topical, or parenteral administration; c) a sterile IL-lRDIO polypeptide of claim 18; or d) said IL-lRDlO polypeptide of claim 18 and a carrier, wherein said carrier is: i) an aqueous compound, including water, saline, and/or buffer; and/or ii) formulated for oral, rectal, nasal, topical, or parenteral administration.
21. A fusion protein of claim 18, comprising: a) mature protein sequence of SEQ ID NO: 2, 4, 18 or 20; b) a detection or purification tag, including a
FLAG, His6, or Ig sequence; or c) sequence of another receptor protein.
22. A kit comprising a polypeptide of claim 18, and: a) a compartment comprising said polypeptide; and/or b) instructions for use or disposal of reagents in said kit.
23. A binding compound comprising an antigen binding site from an antibody, which specifically binds to a natural : A) IL-1RD8 protein of claim 18, wherein: a) said protein is a primate or rodent protein; b) said binding compound is an Fv, Fab, or Fab2 fragment; ' ~ c) said binding compound is conjugated to another chemical moiety; or d) said antibody: i) is raised against a peptide sequence of a mature polypeptide of Seq ID NO
2 or 4; ii) is raised against a mature primate or rodent IL-1RD8; iii) is raised to a purified human IL- 1RD8; iv) is raised to a purified mouse IL-
1RD8; v) is immunoselected; vi) is a polyclonal antibody; vii) binds to a denatured IL-1RD8 ; viii) exhibits a Kd to antigen of at least 30 μM; ix) is attached to a solid substrate, including a bead or plastic membrane; x) is in a sterile composition; or xi) is detectably labeled, including a radioactive or fluorescent label; or
B) IL-lRDlO polypeptide of claim 18, wherein: a) said polypeptide is a primate polypeptide; b) said binding compound is an Fv, Fab, or
Fab2 fragment; c) said binding compound is conjugated to another chemical moiety; or d) said antibody: i) is raised against a peptide sequence of a mature polypeptide of SEQ ID NO: 18 or 20; ii) is raised against a mature primate IL-1RD10; iii) is raised to a purified human IL-
1RD10; iv) is immunoselected; v) is a polyclonal antibody; vi) binds to a denatured IL-1RD10; " vii) exhibits a Kd to antigen of at least
30 μM; viii) is attached to a solid substrate, including a bead or plastic membrane; ix) is in a sterile composition; or x) is detectably labeled, including a radioactive or fluorescent label
24. A kit comprising said binding compound of claim 25, and: a) a compartment comprising said binding compound; and/or b) instructions for use or disposal of reagents in said kit.
26. A method of:
A) making an antibody of claim 23, comprising immunizing an immune system with an immunogenic amount of: a) a primate IL-1RD8 polypeptide; b) a primate IL-1RD10 polypeptide; or thereby causing said antibody to be produced; or
B) producing an antigen: antibody complex, comprising contacting: a) a primate IL-1RD8 polypeptide with an antibody of claim 23A; or b) a primate IL-1RD10 polypeptide with an antibody of claim 23B; thereby allowing said complex to form.
27. A composition comprising: a) a sterile binding compound of claim 23, or b) said binding compound of claim 23 and a carrier, wherein said carrier is : i) an aqueous compound, including water, saline, and/or buffer; and/or ii) formulated for oral, rectal, nasal, topical, or parenteral administration.
28. An isolated or recombinant nucleic acid encoding a protein or peptide or fusion protein of claim 18, wherein: a) said IL-1RD8 or IL-1RD10 is from a mammal; or b) said nucleic acid: i) encodes an antigenic polypeptide sequence of SEQ ID NO: 2, 4, 18 or 20; ii) encodes a plurality of antigenic polypeptide sequences of SEQ ID NO: 2, 4,
18 or 20; iii) exhibits identity to a natural cDNA encoding said segment; iv) is an expression vector; v) further comprises an origin of replication; vi) is from a natural source; vii) comprises a detectable label; viii) comprises synthetic nucleotide sequence; ix) is less than 6 kb, preferably less than 3 kb; x) is from a mammal, including a primate, such as a human; xi) comprises a natural full length coding sequence; xii) is a hybridization probe for a gene encoding said IL-1RD8 or IL-1RD10; xiii) comprises a plurality of nonoverlapping segments of at least 15 nucleotides from SEQ ID NO: 1, 3, 17 or 19; or xiv) is a PCR primer, PCR product, or mutagenesis primer.
29. A cell transfected or transformed with a recombinant nucleic acid of claim 28.
30. The cell of claim 29, wherein said cell is: a) a prokaryotic cell; b) a eukaryotic cell; c) a bacterial cell; d) a yeast cell; e) an insect cell; f) a mammalian cell; g) a mouse cell; h) a primate cell; or i) a human cell.
31. A kit comprising said nucleic acid of claim 28, and: a) a compartment comprising said nucleic acid; b) a compartment further comprising a primate or rodent IL-1RD8 or primate IL-lRDIO polypeptide; and/or b) instructions for use or disposal of reagents in said kit.
32. A method of:
A) making a polypeptide, comprising expressing said nucleic acid of claim 28, thereby producing said polypeptide; or
B) making a duplex nucleic acid, comprising contacting said nucleic acid of claim 28 with a hybridizing nucleic acid, thereby allowing said duplex to form.
33. A nucleic acid which: a) hybridizes under wash conditions of 40° C and less than 2M salt to SEQ ID NO: 3 or 19; or b) exhibits identity over a stretch of at least about 30 nucleotides to a primate IL-1RD8 or IL-1RD10.
34. The nucleic acid of claim 33, wherein: a) said wash conditions are at 55° C and/or 500 mM salt; or b) said stretch is at least 55 nucleotides.
35. The nucleic acid of claim 34, wherein: a) said wash conditions are at 65° C and/or 150 mM salt; or b) said stretch is at least 75 nucleotides.
36. A method of modulating physiology or development of a cell or tissue culture cells comprising contacting said cell with an agonist or antagonist of a primate IL-1RD8 or IL-1RD10.
37. The method of claim 36, wherein said cell is transformed with a nucleic acid encoding either an IL- 1RD8 or IL-1RD10, and another IL-IR.
PCT/US1998/020939 1997-10-15 1998-10-14 Human receptor proteins; related reagents and methods WO1999019480A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP98952080A EP1029043A1 (en) 1997-10-15 1998-10-14 Human receptor proteins; related reagents and methods
JP2000516032A JP2004512801A (en) 1997-10-15 1998-10-14 Human receptor proteins; related reagents and methods
AU97864/98A AU9786498A (en) 1997-10-15 1998-10-14 Human receptor proteins; related reagents and methods
CA002306455A CA2306455A1 (en) 1997-10-15 1998-10-14 Human receptor proteins; related reagents and methods

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
US95182997A 1997-10-15 1997-10-15
US08/951,829 1997-10-15
US97163597A 1997-11-17 1997-11-17
US08/971,635 1997-11-17
US7800898P 1998-03-12 1998-03-12
US60/078,008 1998-03-12
US4071498A 1998-03-18 1998-03-18
US09/040,714 1998-03-18
US8188398P 1998-04-15 1998-04-15
US60/081,883 1998-04-15
US9598798P 1998-08-10 1998-08-10
US60/095,987 1998-08-10

Publications (2)

Publication Number Publication Date
WO1999019480A2 true WO1999019480A2 (en) 1999-04-22
WO1999019480A3 WO1999019480A3 (en) 1999-06-24

Family

ID=27556387

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/020939 WO1999019480A2 (en) 1997-10-15 1998-10-14 Human receptor proteins; related reagents and methods

Country Status (6)

Country Link
EP (1) EP1029043A1 (en)
JP (1) JP2004512801A (en)
AR (1) AR017518A1 (en)
AU (1) AU9786498A (en)
CA (1) CA2306455A1 (en)
WO (1) WO1999019480A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999032629A1 (en) * 1997-12-23 1999-07-01 Immunex Corporation Tigirr dna and polypeptides
WO1999037773A1 (en) * 1998-01-23 1999-07-29 Immunex Corporation Acpl dna and polypeptides
US7202029B2 (en) 1998-09-30 2007-04-10 Millennium Pharmaceuticals, Inc. Nucleic acids encoding human TANGO 195
US11267893B2 (en) 2018-07-16 2022-03-08 Regeneron Pharmaceuticals, Inc. Anti-IL36R antibodies

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996007739A2 (en) * 1994-09-09 1996-03-14 Neurocrine Biosciences, Incorporated Interleukin-1 type 3 receptors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996007739A2 (en) * 1994-09-09 1996-03-14 Neurocrine Biosciences, Incorporated Interleukin-1 type 3 receptors

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
A.O. CHUA AND U. GUBLER: "Sequence of the cDNA for the human fibroblast type interleukin-1 receptor" NUCLEIC ACIDS RESEARCH, vol. 17, no. 23, 11 December 1989, page 10114 XP002091875 IRL PRESS LIMITED,OXFORD,ENGLAND *
C.J. MCMAHAN ET AL.: "A novel Il-1 receptor, cloned from B cells by mammalian expression, is expressed in many cell types" EMBO J., vol. 10, no. 10, October 1991, pages 2821-2832, XP002091855 OXFORD UNIVERSITY PRESS,GB; *
D. BUCK: "Human DNA sequence from cosmid cU72E5, between markers DXS366 and DS87" EMBL SEQUENCE DATABASE,24 December 1995, XP002099439 Heidelberg, FRG *
D. MUZNY ET AL.: "Xp22-164-166; HTG phase 1, 73 unordered pieces" EMBL SEQUENCE DATABASE,5 October 1998, XP002099440 Heidelberg, FRG *
J.E. SIMS ET AL.: "cDNA expression cloning of the IL-1 receptor, a member of the immunglobulin superfamily" SCIENCE, vol. 241, 29 July 1988, pages 585-589, XP002091876 AAAS,WASHINGTON,DC,US *
P. PARNET ET AL.: "IL-1Rrp is similar to the type I interleukin-1 reecptor and its homologues T1/ST2 and IL-1R AcP" J. BIOL. CHEM., vol. 271, no. 8, 23 February 1996, pages 3967-3970, XP002091852 AM. SOC. BIOCHEM. MOL.BIOL.,INC.,BALTIMORE,US *
S. TOMINAGA : "A putative protein of a growth-specific cDNA from BALB/c-3T3cells is highly similar to the extracellular portion of mouse interleukin I receptor" FEBS LETTERS, vol. 258, 1989, pages 301-304, XP002091854 ELSEVIER, AMSTERDAM, NL *
S.A. GREENFEDER ET AL.: "Molecular cloning and characterization of a second subunit of the interleukin 1 receptor complex" J. BIOL. CHEM., vol. 270, no. 23, 9 June 1995, pages 13757-13756, XP002091874 AM. SOC. BIOCHEM. MOL.BIOL.,INC.,BALTIMORE,US *
T. TETSUKA ET AL.: "Nucleotide sequence of a complementary DNA for human ST2" EMBL SEQUENCE DATABASE,3 September 1992, XP002091853 Heidelberg, FRG *
T.L. BORN ET AL.: "Cloning of a novel receptor subunit, AcPL, required for interleukin-18 signalling" J. BIOL. CHEM., vol. 273, no. 45, 6 November 1998, pages 29445-29450, XP002091850 AM. SOC. BIOCHEM. MOL.BIOL.,INC.,BALTIMORE,US *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999032629A1 (en) * 1997-12-23 1999-07-01 Immunex Corporation Tigirr dna and polypeptides
WO1999037773A1 (en) * 1998-01-23 1999-07-29 Immunex Corporation Acpl dna and polypeptides
AU750767B2 (en) * 1998-01-23 2002-07-25 Immunex Corporation ACPL DNA and polypeptides
US7270817B2 (en) 1998-01-23 2007-09-18 Immunex Corporation ACPL antibodies and methods of use thereof
US7838248B2 (en) 1998-01-23 2010-11-23 Immunex Corporation ACPL-related assays
US7993845B2 (en) 1998-01-23 2011-08-09 Immunex Corporation ACPL-related assays
US7202029B2 (en) 1998-09-30 2007-04-10 Millennium Pharmaceuticals, Inc. Nucleic acids encoding human TANGO 195
US7408038B2 (en) 1998-09-30 2008-08-05 Millennium Pharmaceuticals, Inc. Secreted immunomodulatory proteins
US11267893B2 (en) 2018-07-16 2022-03-08 Regeneron Pharmaceuticals, Inc. Anti-IL36R antibodies
US12060428B2 (en) 2018-07-16 2024-08-13 Regeneron Pharmaceuticals, Inc. Anti-IL36R antibodies

Also Published As

Publication number Publication date
WO1999019480A3 (en) 1999-06-24
CA2306455A1 (en) 1999-04-22
JP2004512801A (en) 2004-04-30
AU9786498A (en) 1999-05-03
EP1029043A1 (en) 2000-08-23
AR017518A1 (en) 2001-09-12

Similar Documents

Publication Publication Date Title
US7393530B2 (en) IL-1-like cytokine antibodies
EP0977779A1 (en) Mammalian cytokines; related reagents and methods
US20050009145A1 (en) Mammalian receptor proteins; related reagents and methods
JP2015007129A (en) Mammalian receptor proteins; related reagents and methods
WO1999040195A1 (en) Mammalian receptor proteins; related reagents and methods
CA2392109A1 (en) Mammalian receptor proteins; related reagents and methods
US6326472B1 (en) Human receptor proteins; related reagents and methods
EP1062332A2 (en) Human receptor proteins; related reagents and methods
US7812126B2 (en) DIRS1 polypeptides
US20050106673A1 (en) Mammalian receptor proteins; related reagents and methods
WO1999019480A2 (en) Human receptor proteins; related reagents and methods
WO2000017363A2 (en) Interleukin-1 like molecule: interleukin-1-zeta; nucleic acids, polypeptides, antibodies, their uses
US7205381B2 (en) Mammalian cytokines; related reagents and methods
MXPA00003774A (en) Human receptor proteins;related reagents and methods
MXPA00008848A (en) Human receptor proteins;related reagents and methods

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AL AM AU AZ BA BB BG BR BY CA CN CZ EE GD GE HR HU ID IL IS JP KG KR KZ LC LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK SL TJ TM TR TT UA US UZ VN

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
AK Designated states

Kind code of ref document: A3

Designated state(s): AL AM AU AZ BA BB BG BR BY CA CN CZ EE GD GE HR HU ID IL IS JP KG KR KZ LC LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK SL TJ TM TR TT UA US UZ VN

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)

Free format text: (EXCEPT GD)

ENP Entry into the national phase in:

Ref document number: 2306455

Country of ref document: CA

Ref country code: CA

Ref document number: 2306455

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase in:

Ref country code: JP

Ref document number: 2000 516032

Kind code of ref document: A

Format of ref document f/p: F

NENP Non-entry into the national phase in:

Ref country code: KR

WWE Wipo information: entry into national phase

Ref document number: PA/a/2000/003774

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1998952080

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1998952080

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1998952080

Country of ref document: EP