AU5668194A - Allergenic proteins and peptides from japanese cedar pollen - Google Patents

Allergenic proteins and peptides from japanese cedar pollen

Info

Publication number
AU5668194A
AU5668194A AU56681/94A AU5668194A AU5668194A AU 5668194 A AU5668194 A AU 5668194A AU 56681/94 A AU56681/94 A AU 56681/94A AU 5668194 A AU5668194 A AU 5668194A AU 5668194 A AU5668194 A AU 5668194A
Authority
AU
Australia
Prior art keywords
protein
seq
cryj
fragment
sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU56681/94A
Inventor
Andrew W Brauer
Mei-Chang Kuo
Joanne Pollock
Siu-Mei Helena Yeung
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Immulogic Pharmaceutical Corp
Original Assignee
Immulogic Pharmaceutical Corp
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 Immulogic Pharmaceutical Corp filed Critical Immulogic Pharmaceutical Corp
Publication of AU5668194A publication Critical patent/AU5668194A/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/415Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies

Description

ALLERGENIC PROTEINS AND PEPTIDES FROM JAPANESE CEDAR POLLEN
Background of the Invention
Genetically predisposed individuals, who make up about 10% of the population, become hypersensitized (allergic) to antigens from a variety of environmental sources to which they are exposed. Those antigens that can induce immediate and/or delayed types of hypersensitivity are known as allergens. (King,
T.P., Adv. Immunol. 23: 77-105, (1976)). Anaphylaxis or atopy, which includes the symptoms of hay fever, asthma, and hives, is one form of immediate allergy. It can be caused by a variety of atopic allergens, such as products of grasses, trees, weeds, animal dander, insects, food, drugs, and chemicals. The antibodies involved in atopic allergy belong primarily to the IgE class of immunoglobulins. IgE binds to mast cells and basophils. Upon combination of a specific allergen with IgE bound to mast cells or basophils, the IgE may be cross- linked on the cell surface, resulting in the physiological effects of IgE-antigen interaction. These physiological effects include the release of, among other substances, histamine, serotonin, heparin, a chemotactic factor for eosinophilic leukocytes and/or the leukotrienes, C4, D4, and E4, which cause prolonged constriction of bronchial smooth muscle cells (Hood, L.E. et al. Immunology (2nd ed.), The Benjamin/Cumming Publishing Co., Inc. (1984)). These released substances are the mediators which result in allergic symptoms caused by a combination of IgE with a specific allergen. Through them, the effects of an allergen are manifested. Such effects may be systemic or local in nature, depending on the route by which the antigen entered the body and the pattern of deposition of IgE on mast cells or basophils. Local manifestations generally occur on epithelial surfaces at the location at which the allergen entered the body. Systemic effects can include anaphylaxis (anaphy lactic shock), which is the result of an IgE-basophil response to circulating (intravascular) antigen.
Japanese cedar (Sugi; Cryptomeria japonica) pollinosis is one of the most important allergic diseases in Japan. The number of patients suffering from this disease is on the increase and in some areas, more than 10% of the population are affected. Treatment of Japanese cedar pollinosis by administration of Japanese cedar pollen extract to effect hyposensitization to the allergen has been attempted. Hyposensitization using Japanese cedar pollen extract, however, has drawbacks in that it can elicit anaphylaxis if high doses are used, whereas when low doses are used to avoid anaphylaxis, treatment must be continued for several years to build up a tolerance for the extract.
The major allergen from Japanese cedar pollen has been purified and designated as Sugi basic protein (SBP) or Cry j I. This protein is reported to be a basic protein with a molecular weight of 41-50 kDa and a pi of 8.8. There appear to be multiple isoforms of the allergen, apparently due in part to differential glycosylation (Yasueda et al. (1983) J. Allergy Clin. Immunol. 71: 77-86; and Taniai et al. (1988) FEBS Letters 239: 329-332. The sequence of the first twenty amino acids at the N-terminal end of Cryj I and a sixteen amino acid internal sequence have been determined (Taniai supra).
A second allergen has recently been isolated from the pollen of Cryptomeria japonica (Japanese cedar) (Sakaguchi et al. (1990) Allergy 45:309-312). This allergen, designated Cryj II, has been reported to have a molecular weight of approximately 37 kDa and 45 kDa when assayed on sodium dodecyl sulfate- polyacrylamide gel electrophoresis (SDS-PAGE) under non-reducing and reducing conditions, respectively (Sukaguchi et al., supra). Cryj II was found to have no immunological cross-reactivity with Cryj I (Sakaguchi (1990) supra: Kawashima et al. (1992) Int. Arch. Allergy Immunol. 98:110-117). Most patients with Japanese cedar pollinosis were found to have IgE antibodies to both Cryj I and Cryj II. however, 29% of allergic patients had IgE that only reacted with Cryj I and 14% of allergic patients had IgE that only reacted with Cry j II (Sakaguchi (1990) supra). Isoelectric focusing of Cryj II indicated that this protein has a pi above 9.5, as compared to pi 8.6-8.8 for Cryj I (Sakaguchi (1990) supra). Further, the reported NH2-terminal sequence for Cryj II, NH2-AlaIleAsnIlePheAsnNalGluLysTyr-
COOH, did not match that reported for Cryj I (Sakaguchi (1990) supra).
Despite the attention Japanese cedar pollinosis allergens have received, definition or characterization of the allergens responsible for its adverse effects on people is far from complete. Current desensitization therapy involves treatment with pollen extract with its attendant risks of anaphylaxis if high doses of pollen extract are administered, or long desensitization times when low doses of pollen extract are administered.
Summary of the Invention The present invention provides nucleic acid sequences coding for the
Cryptomeria japonica major pollen allergen Cry j II and fragments thereof. The present invention also provides purified Cryj II and at least one fragment thereof produced in a host cell transformed with a nucleic acid sequence coding for Cryj II or at least one fragment thereof and fragments of Cryj II prepared synthetically. As used herein, a fragment of the nucleic acid sequence coding for the entire amino acid sequence of Cryj II refers to a nucleotide sequence having fewer bases than the nucleotide sequence coding for the entire amino acid sequence of Cryj II and/or mature Cryj II. Cryj II and fragments thereof are useful for diagnosing, treating, and preventing Japanese cedar pollinosis. This invention is more particularly described in the appended claims and is described in its preferred embodiments in the following description.
Description of the Drawings
Fig. la shows an SDS-PAGE (12%) analysis of Cryj II under non-reducing conditions.
Fig. lb shows an SDS-PAGE (12%) analysis of Cryj II under reducing conditions.
Fig. 2 shows the results of mono S column chromatography of Cryj II eluted with a step gradient of NaCl in lOmM sodium acetate buffer, pH 5.0.
Fig. 3 shows an SDS-PAGE (12%) of purified subfractions of Cryj II analyzed under reducing conditions. Fig. 4 shows the nucleic acid sequence (SEQ ID NO: 1) and the deduced amino acid (SEQ ID NO: 2) coding for Cry j II.
Fig. 5 shows the deduced amino acid sequence of Cryj II (SEQ ID NO: 2). Fig. 6 shows the long form (SEQ ID NO: 4) and short form (SEQ ID NO: 5) NH2-terminii amino acid sequences of Cry j II determined by protein sequence analysis as discussed in Example 2 aligned with the ten amino acid sequence of Cry j II (SEQ ID NO: 3) defined by Sakaguchi et al., supra (SEQ ID NO: 6).
Fig. 7 is a graphic representation of the results of a direct ELISA assay showing the binding response of the monoclonal antibody 4B11 and seven patients' (Batch 1) plasma IgE to purified Cryj I as the coating antigen. Fig. 8 is a graphic representation of a direct ELISA assay showing the binding response of the monoclonal antibody 4B11, and seven patients' (Batch 1) plasma IgE to purified native Cryj II as the coating antigen.
Fig. 9 is a graphic representation of a direct ELISA assay showing the binding response of the monoclonal antibody, 4B11, and seven patients' (Batch 1) plasma IgE to recombinant Cryj II (rCryj II) as the coating antigen.
Fig. 10 is a graphic representation of a direct ELISA assay showing the binding response of eight patients' (Batch 2) plasma IgE to purified native Cryj I. Fig. 11 is a graphic representation of a direct ELISA assay showing the binding response of eight patients' (Batch 2) plasma IgE to purified native Cryj II.
Fig. 12 is a graphic representation of a direct ELISA assay showing the binding response of eight patients' (Batch 2) plasma IgE to recombinant Cryj II. Fig. 13 is a graphic representation of a direct ELISA assay showing the binding response of eight patients' (Batch 3) plasma IgE to purified native Cryj I.
Fig. 14 is a graphic representation of a direct ELISA assay showing the binding response of eight patients' (Batch 3) plasma IgE to purified native Cryj II. Fig. 15 is a graphic representation of a direct ELISA assay showing the binding response of eight patients' (Batch 3) plasma IgE to recombinant Cryj II.
Fig. 16 is a table which summarizes both the MAST scores performed on patient's plasma samples (Batch 1-3) and the direct ELISA results shown in Figs. 7- 15; a positive response is indicated by a (+) sign and the number of positive responses for each antigen is shown at the bottom of each column.
Detailed Description of the Invention
The present invention provides nucleic acid sequences coding for Cryj II, an allergen found in Japanese cedar pollen. The nucleic acid sequence coding for Cryj II shown in Fig. 4 (SEQ ID NO: 1) encodes a protein of 514 amino acids. The deduced Cryj II amino acid sequence is shown in Figs. 4 and 5 (SEQ ID NO: 2) .
Direct protein sequence analysis of native purified Cryj II resulted in two separate overlapping NH2-termini sequences, designated Long and Short, corresponding respectively to amino acids 46 through 89 (SEQ ID NO: 4) and 51 through 89 (SEQ ID NO: 5) of Figs. 4, 5 and 6. The ten amino acid sequence NH2-AlaIleAsnIlePhe- AsnValGluLysTry-COOH (SEQ ID NO: 6) previously defined by Sakaguchi et al, supra for Cryj II corresponds to amino acids 55 through 64 of Figs. 4 and 6. The full-length Cryj II sequence contains 20 cysteine residues and three potential N- linked glycosylation sites with the consensus sequence of Asn-Xxx-Ser/Thr. According to the program contained in PC Gene, Intelligenetics (Mountain View, CA) the proteins with the NH2-termini defined by the Long and Short forms of Cry j
II would contain 469 and 464 amino acids, respectively, and have predicted molecular weights of 51.5 kDa (long) and 50.9 kDa (short). The amino acid sequence representing the long form of Cryj II is encoded by the nucleotide sequence extending from bases 177-1586 (SEQ ID NO: 7) as shown in Fig. 4, and the amino acid sequence representing the short form of Cry j II is encoded by the nucleotide sequence extending from 192-1586 (SEQ ID NO: 8) as shown in Fig. 4. A host cell transformed with a vector containing the cDNA insert coding for full- length Cryj II has been deposited with the American Type Culture Collection, ATCC No. 69105.
Fragments of the nucleic acid sequence coding for fragments of Cryj II are also within the scope of the invention. Fragments within the scope of the invention include those coding for parts of Cryj II which induce an immune response in mammals, preferably humans, such as stimulation of minimal amounts of IgE; binding of IgE; eliciting the production of IgG and IgM antibodies; or the eliciting of a T cell response such as proliferation and/or lymphokine secretion and/or the induction of T cell anergy. The foregoing fragments of Cryj II are referred to herein as antigenic fragments. Fragments within the scope of the invention also include those capable of hybridizing with nucleic acid from other plant species for use in screening protocols to detect allergens that are cross-reactive with Cryj II. As used herein, a fragment of the nucleic acid sequence coding for Cryj II refers to a nucleotide sequence having fewer bases than the nucleotide sequence coding for the entire amino acid sequence of Cryj II and/or mature Cryj II. Generally, the nucleic acid sequence coding for the fragment or fragments of Cryj II will be selected from the bases coding for the mature protein, however, in some instances it may be desirable to select all or a part of a fragment or fragments from the leader sequence portion of the nucleic acid sequence of the invention. The nucleic acid sequence of the invention may also contain linker sequences, modified restriction endonuclease sites and other sequences useful for cloning, expression or purification of Cryj II or fragments thereof.
A nucleic acid sequence coding for Cryj II may be obtained from Cryptomeria japonica plants. Applicants have found that fresh pollen and staminate cones are a good source of Cry j II mRNA. It may also be possible to obtain the nucleic acid sequence coding for Cryj II from genomic DNA. Cryptomeria japonica is a well-known species of cedar, and plant material may be obtained from wild, cultivated, or ornamental plants. The nucleic acid sequence coding for Cryj II may be obtained using the method disclosed herein or any other suitable techniques for isolation and cloning of genes. The nucleic acid sequence of the invention may be DNA or RNA.
The present invention provides expression vectors and host cells transformed to express the nucleic acid sequences of the invention. Nucleic acid coding for Cryj II, or at least one fragment thereof may be expressed in bacterial cells such as E. coli, insect cells (baculo virus), yeast, or mammalian cells such as Chinese hamster ovary cells (CHO). Suitable expression vectors, promoters, enhancers, and other expression control elements may be found in Sambrook et al. Molecular Cloning: A Laboratory Manual, second edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1989). Other suitable expression vectors, promoters, enhancers, and other expression elements are known to those skilled in the art. Expression in mammalian, yeast or insect cells leads to partial or complete glycosylation of the recombinant material and formation of any inter- or intra-chain disulfide bonds. Suitable vectors for expression in yeast include YepSecl (Baldari et al. (1987) Embo J. 6: 229-234); pMFa (Kurjan and Herskowitz (1982) Cell 30: 933- 943); JRY88 (Schultz et al. (1987) Gene 54. 113-123) and pYES2 (Invitrogen Corporation, San Diego, CA). These vectors are freely available. Baculovirus and mammalian expression systems are also available. For example, a baculovirus system is commercially available (PharMingen, San Diego, CA) for expression in insect cells while the pMSG vector is commerically available (Pharmacia, Piscataway, NJ) for expression in mammalian cells.
For expression in E. coli, suitable expression vectors include, among others, pTRC (Amann et al. (1988) Gene 69: 301-315); pGEX (Amrad Corp. , Melbourne,
Australia); pMAL (N.E. Biolabs, Beverly, MA); pRIT5 (Pharmacia, Piscataway, NJ); pET-lld (Novagen, Madison, WI) Jameel et al., (1990) J. Virol. 64:3963- 3966; and pSEM (Knapp et al. (1990) BioTechniques 8: 280-281). The use of pTRC, and pET-lld, for example, will lead to the expression of unfused protein. The use of pMAL, pRIT5 pSEM and pGEX will lead to the expression of allergen fused to maltose E binding protein (pMAL), protein A (pRIT5), truncated β- galactosidase (PSEM), or glutathione S-transferase (pGEX). When Cryj II, fragment, or fragments thereof is expressed as a fusion protein, it is particularly advantageous to introduce an enzymatic cleavage site at the fusion junction between the carrier protein and Cryj II or fragment thereof. Cryj II or fragment thereof may then be recovered from the fusion protein through enzymatic cleavage at the enzymatic site and biochemical purification using conventional techniques for purification of proteins and peptides. Suitable enzymatic cleavage sites include those for blood clotting Factor Xa or thrombin for which the appropriate enzymes and protocols for cleavage are commercially available from for example Sigma Chemical
Company, St. Louis, MO and N.E. Biolabs, Beverly, MA. The different vectors also have different promoter regions allowing constitutive or inducible expression with, for example, IPTG induction (PRTC, Amann et al., (1988) supra: pET-lld, Novagen, Madison, WI) or temperature induction (pRIT5, Pharmacia, Piscataway, NJ) . It may also be appropriate to express recombinant Cryj II in different E. coli hosts that have an altered capacity to degrade recombinantly expressed proteins (e.g. U.S. patent 4,758,512). Alternatively, it may be advantageous to alter the nucleic acid sequence to use codons preferentially utilized by E. coli, where such nucleic acid alteration would not affect the amino acid sequence of the expressed protein. Host cells can be transformed to express the nucleic acid sequences of the invention using conventional techniques such as calcium phosphate or calcium chloride co-precipitation, DΕAΕ-dextran-mediated transfection, or electroporation.
Suitable methods for transforming the host cells may be found in Sambrook et al. supra, and other laboratory textbooks. The nucleic acid sequences of the invention may also be synthesized using standard techniques.
The present invention also provides a method of producing purified Japanese cedar pollen allergen Cryj II or at least one fragment thereof comprising the steps of culturing a host cell transformed with a DNA sequence encoding Japanese cedar pollen allergen Cryj II or at least one fragment thereof in an appropriate medium to produce a mixture of cells and medium containing said Japanese cedar pollen allergen Cryj II or at least one fragment thereof; and purifying the mixture to produce substantially pure Japanese cedar pollen allergen Cryj II or at least one fragment thereof. Host cells transformed with an expression vector containing DNA coding for Cryj II or at least one fragment thereof are cultured in a suitable medium for the host cell. Cryj II protein and peptides can be purified from cell culture medium, host cells, or both using techniques known in the art for purifying peptides and proteins including ion-exchange chromatography, gel filtration chromatography, ultrafiltration, electrophoresis and immunopurification with antibodies specific for Cryj II or fragments thereof. The terms isolated and purified are used interchangeably herein and refer to peptides, protein, protein fragments, and nucleic acid sequences substantially free of cellular material or culture medium when produced by recombinant DNA techniques, or chemical precursors.
Cryj II protein may also be isolated from Japanese cedar pollen as described in Example 1. Cry j II isolated directly from Japanese cedar pollen is referred to herein as "purified native" Cryj II. It is preferable that purified native Cry j II of the invention be at least 80% pure, and more preferably at least 90% pure and even more preferably be purified to homogeneity (at least 99% pure).
Another aspect of the invention provides preparations comprising Japanese cedar pollen allergen Cryj II or at least one fragment thereof synthesized in a host cell transformed with a DNA sequence encoding all or a portion of Japanese cedar pollen allergen Cryj II, or chemically synthesized, and purified Japanese cedar pollen allergen Cryj II protein, or at least one antigenic fragment thereof produced in a host cell transformed with a nucleic acid sequence of the invention, or chemically synthesized. In preferred embodiments of the invention the Cryj II protein is produced in a host cell transformed with the nucleic acid sequence coding for at least the mature Cryj II protein.
Fragments of an allergen from Cry j II, eliciting a desired antigenic response (referred to herein as antigenic fragments) are defined herein as any protein fragment or peptide which can be derived from the Cryj II proteins, but does not include the ten amino acid fragments which extends from amino acid residues 55-64, as shown in Figs. 4, 5 and 6, but may include any portion of that ten amino acid fragment in conjunction with another fragment derived from Cryj II. Antigenic fragments of Cryj II may be obtained, for example, by screening peptides recombinantly produced from the corresponding fragment of the nucleic acid sequence of the invention coding for such peptides, or by screening peptides which have been synthesized chemically using techniques known in the art, or by screening peptides produced by chemical cleavage of the allergen. The allergen may be arbitrarily divided into fragments of a desired length with no overlap of the peptides, or preferably divided into fragments of a desired length with no overlap of the peptides, or preferably divided into overlapping fragments of a desired length. The fragments are tested to determine their antigenicity (e.g. the ability of the fragment to induce an immune response such as T cell proliferation as discussed in Example 7).
Antigenic fragments may also be predicted using an algorithm such as that discussed in a paper by Hill et al, Journal of Immunology, 147:184-197 (1991).
Algorithms for predicting peptides which elicit T cell activity such as the algorithm discussed by Hill et al. are based on the protein's sequence wherein certain patterns within the sequence are likely to bind MHC and therefore may contain T cell epitopes. The peptides predicted by the algorithm such as Cry j II A and Cry j IIB discussed in Example 7 may be produced recombinantly or synthetically and tested for T cell activity as discussed in Example 7.
If fragments of Japanese cedar pollen allergen, e.g. Cryj II are to be used for therapeutic purposes, then the fragments of Japanese cedar pollen allergen which are capable of eliciting a T cell response such as stimulation (i.e., proliferation or lymphokine secretion) and/or are capable of inducing T cell anergy are particularly desirable and fragments of Japanese cedar pollen which have minimal IgE stimulating activity are also desirable. Additionally, for therapeutic purposes, purified Japanese cedar pollen allergens, e.g. Cryj II, and fragments thereof preferably do not bind IgE specific for Japanese cedar pollen or bind such IgE to a substantially lesser extent than the purified native Japanese cedar pollen allergen binds such IgE. If the purified Japanese cedar pollen allergen or fragment or fragments thereof bind IgE, it is preferable that such binding does not result in the release of mediators (e.g. histamines) from mast cells or basophils. Minimal IgE stimulating activity refers to IgE stimulating activity that is less than the amount of IgE production stimulated by the native Cryj II protein.
Isolated antigenic fragments or peptides of the present invention which have T cell stimulating activity, and thus comprise at least one T cell epitope are particularly desirable. T cell epitopes are believed to be involved in initiation and perpetuation of the immune response to a protein allergen which is responsible for the clinical symptoms of allergy. These T cell epitopes are thought to trigger early events at the level of the T helper cell by binding to an appropriate HLA molecule on the surface of an antigen presenting cell and stimulating the relevant T cell subpopulation. These events lead to T cell proliferation, lymphokine secretion, local inflammatory reactions, recruitment of additional immune cells to the site, and activation of the B cell cascade leading to production of antibodies. One isotype of these antibodies, IgE, is fundamentally important to the development of allergic symptoms and its production is influenced early in the cascade of events, at the level of the T helper cell, by the nature of the lymphokines secreted. An epitope is the basic element or smallest unit of recognition by a receptor, particularly immunoglobulins, histocompatibility antigens and T cell receptors, where the epitope comprises amino acids essential to receptor recognition. Amino acid sequences which mimic those of the epitopes particularly T cell epitopes and which modify the allergic response to protein allergens including those capable of down regulating allergic response to Cry j II, are within the scope of this invention.
As discussed in Example 7, human T cell stimulating activity can be tested by culturing T cells obtained from an individual sensitive to Japanese cedar pollen allergen, (i.e., an individual who has an IgE mediated immune response to Japanese cedar pollen allergen) with a peptide derived from the allergen and determining whether proliferation of T cells occurs in response to the peptide as measured, e.g., by cellular uptake of tritiated thymidine. Stimulation indices for responses by T cells to peptides can be calculated as the maximum CPM in response to a peptide divided by the control CPM. A stimulation index (S.I.) equal to or greater than two times the background level is considered "positive". Positive results are used to calculate the mean stimulation index for each peptide tested. Preferred peptides of this invention comprise at least one T cell epitope and have a mean T cell stimulation index of greater than or equal to 2.0. A peptide having a mean T cell stimulation index of greater than or equal to 2.0 is considered useful as a therapeutic agent. As shown in Fig. 17 Cryj II peptides Cry j II A and Cry j IIB have mean stimulation indexes of at least two and therefore comprise at least one T cell epitope as predicted.
Purified protein allergens from Japanese cedar pollen or preferred antigenic fragments thereof, when administered to a Japanese cedar pollen-sensitive individual, or an individual allergic to an allergen cross-reactive with Japanese cedar pollen allergen, are capable of modifying the allergic response of the individual to Japanese cedar pollen or such cross-reactive allergen of the individual, and preferably are capable of modifying the B-cell response, T-cell response or both the B-cell and the T-cell response of the individual to the allergen. As used herein, modification of the allergic response of an individual sensitive to a Japanese cedar pollen allergen can be defined as non-responsiveness or diminution in symptoms to the allergen, as determined by standard clinical procedures (See e.g. Narney et al, British Medical Journal, 302:265-269 (1990)) including diminution in Japanese cedar pollen induced asthmatic symptoms. As referred to herein, a diminution in symptoms includes any reduction in allergic response of an individual to the allergen after the individual has completed a treatment regimen with a peptide or protein of the invention. This diminution may be subjective (i.e. the patient feels more comfortable in the presence of the allergen). Diminution in symptoms can be determined clinically as well, using standard skin tests as is known in the art.
The purified Cryj II protein or fragments thereof are preferably tested in mammalian models of Japanese cedar pollinosis such as the mouse model disclosed in Tamura et al. (1986) Microbiol. Immunol. 30: 883-896, or U.S. patent 4,939,239; or the primate model disclosed in Chiba et al. (1990) Int. Arch. Allergy Immunol. 93: 83-88. Initial screening for IgE binding to the protein or fragments thereof may be performed by scratch tests or intradermal skin tests on laboratory animals or human volunteers, or in in vitro systems such as RAST
(radioallergosorbent test), RAST inhibition, ELISA assay, radioimmunoassay (RIA), or histamine release.
Exposure of allergic individuals to purified protein allergens of the present invention or to the antigenic fragments of the present invention which comprise at least one T cell epitope and are derived from protein allergens may tolerize or anergize appropriate T cell subpopulations such that they become unresponsive to the protein allergen and do not participate in stimulating an immune response upon such exposure. In addition, administration of the protein allergen of the invention or an antigenic fragment of the present invention which comprises at least one T cell epitope may modify the lymphokine secretion profile as compared with exposure to the naturally-occurring protein allergen or portion thereof (e.g. result in a decrease of IL-4 and/or an increase in IL-2). Furthermore, exposure to such antigenic fragment or protein allergen may influence T cell subpopulations which normally participate in the response to the allergen such that these T cells are drawn away from the site(s) of normal exposure to the allergen (e.g., nasal mucosa, skin, and lung) towards the site(s) of therapeutic administration of the fragment or protein allergen. This redistribution of T cell subpopulations may ameliorate or reduce the ability of an individual's immune system to stimulate the usual immune response at the site of normal exposure to the allergen, resulting in a dimunution in allergic symptoms.
The isolated Cryj II protein, and fragments or portions derived therefrom can be used in methods of diagnosing, treating and preventing allergic reactions to
Japanese cedar pollen allergen or a cross reactive protein allergen. Thus the present invention provides therapeutic compositions comprising purified Japanese cedar pollen allergen Cryj II or at least one fragment thereof produced in a host cell transformed to express Cryj II or at least one fragment thereof, and a pharmaceutically acceptable carrier or diluent. The therapeutic compositions of the invention may also comprise synthetically prepared Cry j II or at least one fragment thereof and a pharmaceutically acceptable carrier or diluent. Administration of the therapeutic compositions of the present invention to an individual to be desensitized can be carried out using known techniques. Cryj II protein or at least one fragment thereof may be administered to an individual in combination with, for example, an appropriate diluent, a carrier and/or an adjuvant. Pharmaceutically acceptable diluents include saline and aqueous buffer solutions. Pharmaceutically acceptable carriers include polyethylene glycol (Wie et al. (1981) Int. Arch. Allergy Appl. Immunol. 64:84-99) and liposomes (Strejan et al. (1984) J. Neuroimmunol 7: 27). For purposes of inducing T cell anergy, the therapeutic composition is preferably administered in nonimmunogenic form, e.g. it does not contain adjuvant. Such compositions will generally be administered by injection (subcutaneous, intravenous, etc.), oral administration, inhalation, transdermal application or rectal administration. The therapeutic compositions of the invention are administered to Japanese cedar pollen-sensitive individuals at dosages and for lengths of time effective to reduce sensitivity (i.e, reduce the allergic response) of the individual to Japanese cedar pollen. Effective amounts of the therapeutic compositions will vary according to factors such as the degree of sensitivity of the individual to Japanese cedar pollen, the age, sex, and weight of the individual, and the ability of the Cryj II protein or fragment thereof to elicit an antigenic response in the individual.
The Cryj II cDNA (or the mRNA from which it was transcribed) or a portion thereof can be used to identify similar sequences in any variety or type of plant and thus, to identify or "pull out" sequences which have sufficient homology to hybridize to the Cryj II cDNA or mRNA or portion thereof, for example, DNA from allergens of Cupressus sempervirens, Juniperus sabinoides etc. , under conditions of low stringency. Those sequences which have sufficient homology (generally greater than 40%) can be selected for further assessment using the method described herein. Alternatively, high stringency conditions can be used. In this manner, DNA of the present invention can be used to identify, in other types of plants, preferably related families, genera, or species such as Juniperus, or Cupressus, sequences encoding polypeptides having amino acid sequences similar to that of Japanese cedar pollen allergen Cryj II, and thus to identify allergens in other species. Thus, the present invention includes not only Cryj II, but also other allergens encoded by DNA which hybridizes to DNA of the present invention. The invention further includes previously unidentified isolated allergenic proteins or fragments thereof that are immunologically related to Cryj II or fragments thereof, such as by antibody cross-reactivity wherein the isolated allergenic proteins or fragments thereof are capable of binding to antibodies specific for the protein and peptides of the invention, or by T cell cross-reactivity wherein the isolated allergenic proteins or fragments thereof are capable of stimulating T cells specific for the protein and peptides of this invention. Proteins or peptides encoded by the cDNA of the present invention can be used, for example as "purified" allergens. Such purified allergens are useful in the standardization of allergen extracts which are key reagents for the diagnosis and treatment of Japanese cedar pollinosis. Furthermore, by using peptides based on the nucleic acid sequences of Cryj II, anti-peptide antisera or monoclonal antibodies can be made using standard methods. These sera or monoclonal antibodies can be used to standardize allergen extracts.
Through use of the peptides and protein of the present invention, preparations of consistent, well-defined composition and biological activity can be made and administered for therapeutic purposes (e.g. to modify the allergic response of a Japanese cedar sensitive individual to pollen of such trees). Administration of such peptides or protein may, for example, modify B-cell response to Cry j II allergen, modify T-cell response to Cryj II allergen or modify both B-cell and T-cell responses. Purified peptides can also be used to study the mechanism of immunotherapy of Cryptomeria japonica allergy and to design modified derivatives or analogues useful in immunotherapy.
Work by others has shown that high doses of allergens generally produce the best results (i.e., best symptom relief). However, many people are unable to tolerate large doses of allergens because of allergic reactions to the allergens. Modification of naturally-occurring allergens can be designed in such a manner that modified peptides or modified allergens which have the same or enhanced therapeutic properties as the corresponding naturally-occurring allergen but have reduced side effects (especially anaphy lactic reactions) can be produced. These can be, for example, a protein or peptide of the present invention (e.g., one having all or a portion of the amino acid sequence of Cry j II), or a modified protein or peptide, or protein or peptide analogue.
It is possible to modify the structure of a protein or peptide of the invention for such purposes as increasing solubility, enhancing therapeutic or preventive efficacy, or stability (e.g., shelf life ex vivo, and resistance to proteolytic degradation in vivo). A modified protein or peptide can be produced in which the amino acid sequence has been altered, such as by amino acid substitution, deletion, or addition, to modify immunogenicity and/or reduce allergenicity, or to which a component has been added for the same purpose. For example, the amino acid residues essential to T cell epitope function can be determined using known techniques (e.g., substitution of each residue and determination of the presence or absence of T cell reactivity).
For example, a peptide can be modified so that it maintains the ability to induce T cell anergy and bind MHC proteins without the ability to induce a strong proliferative response or possibly any proliferative response when administered in immunogenic form. In this instance, critical binding residues for the T cell receptor can be determined using known techniques (e.g., substitution of each residue and determination of the presence or absence of T cell reactivity). Those residues shown to be essential to interact with the T cell receptor can be modified by replacing the essential amino acid with another, preferably similar amino acid residue (a conservative substitution) whose presence is shown to enhance, diminish but not eliminate binding to relevant MHC.
Additionally, peptides of the invention can be modified by replacing an amino acid shown to be essential to interact with the MHC protein complex with another, preferably similar amino acid residue (conservative substitution) whose presence is shown to enhance, diminish but not eliminate or not effect T cell activity. In addition, amino acid residues which are not essential for interaction with the MHC protein complex but which still bind the MHC protein complex can be modified by being replaced by another amino acid whose incorporation may enhance, not effect, or diminish but not eliminate T cell reactivity. Preferred amino acid substitutions for non-essential amino acids include, but are not limited to substitutions with alanine, glutamic acid, or a methyl amino acid.
Another example of a modification of protein or peptides is substitution of cysteine residues preferably with alanine, serine, threonine, leucine or glutamic acid to minimize dimerization via disulfide linkages. Another example of modification of the peptides of the invention is by chemical modification of amino acid side chains or cyclization of the peptide.
In order to enhance stability and/or reactivity, the protein or peptides of the invention can also be modified to incorporate one or more polymorphisms in the amino acid sequence of the protein allergen resulting from natural allelic variation. Additionally, D-amino acids, non-natural amino acids or non-amino acid analogues can be substituted or added to produce a modified protein or peptide within the scope of this invention. Furthermore, proteins or peptides of the present invention can be modified using the polyethylene gly col (PEG) method of A. Sehon and co-workers (Wie et al. supra) to produce a protein or peptide conjugated with PEG. In addition, PEG can be added during chemical synthesis of a protein or peptide of the invention.
Modifications of proteins or peptides or portions thereof can also include reduction/ alyklation (Tarr in: Methods of Protein Microcharacterization, J.E. Silver ed. Humana Press, Clifton, NJ, pp 155-194 (1986)); acylation (Tarr, supra): chemical coupling to an appropriate carrier (Mishell and Shiigi, eds, Selected Methods in Cellular Immunology, WH Freeman, San Francisco, CA (1980); U.S. Patent
4,939,239; or mild formalin treatment (Marsh International Archives of Allergy and Applied Immunology, 41.199-215 (1971)).
To facilitate purification and potentially increase solubility of proteins or peptides of the invention, it is possible to add reporter group(s) to the peptide backbone. For example, poly-histidine can be added to a peptide to purify the peptide on immobilized metal ion affinity chromatography (Hochuli, E. et al., Bio/Technology, 6:1321-1325 (1988)). In addition, specific endoprotease cleavage sites can be introduced, if desired, between a reporter group and amino acid sequences of a peptide to facilitate isolation of peptides free of irrelevant sequences. In order to successfully desensitize an individual to a protein antigen, it may be necessary to increase the solubility of a protein or peptide by adding functional groups to the peptide or by not including hydrophobic T cell epitopes or regions containing hydrophobic epitopes in the peptides or hydrophobic regions of the protein or peptide. To potentially aid proper antigen processing of T cell epitopes within a peptide, canonical protease sensitive sites can be recombinantly or synthetically engineered between regions, each comprising at least one T cell epitope. For example, charged amino acid pairs, such as KK or RR, can be introduced between regions within a peptide during recombinant construction of the peptide. The resulting peptide can be rendered sensitive to cathepsin and/or other trypsin-like enzymes cleavage to generate portions of the peptide containing one or more T cell epitopes. In addition, such charged amino acid residues can result in an increase in solubility of a peptide.
Site-directed mutagenesis of DNA encoding a peptide or protein of the invention (e.g. Cryy II or a fragment thereof) can be used to modify the structure of the peptide or protein by methods known in the art. Such methods may, among others, include PCR with degenerate oligonucleotides (Ho et al., Gene, 77:51-59
(1989)) or total synthesis of mutated genes (Hostomsky, Z. et al., Biochem. Biophys, Res. Comm., 161: 1056-1063 (1989)). To enhance bacterial expression, the aforementioned methods can be used in conjunction with other procedures to change the eucaryotic codons in DNA constructs encoding protein or peptides of the invention to ones preferentially used in E. coli, yeast, mammalian cells, or other eukaryotic cells.
Using the structural information now available, it is possible to design Cryj II peptides which, when administered to a Japanese cedar pollen sensitive individual in sufficient quantities, will modify the individual's allergic response to Japanese cedar pollen. This can be done, for example, by examining the structure of Cryj II, producing peptides (via an expression system, synthetically or otherwise) to be examined for their ability to influence B-cell and/or T-cell responses in Japanese cedar pollen sensitive individuals and selecting appropriate peptides which contain epitopes recognized by the cells. It is now also possible to design an agent or a drug capable of blocking or inhibiting the ability of Japanese cedar pollen allergen to induce an allergic reaction in Japanese cedar pollen sensitive individuals. Such agents could be designed, for example, in such a manner that they would bind to relevant anti- Cry j II IgΕs, thus preventing IgΕ-allergen binding and subsequent mast cell degranulation. Alternatively, such agents could bind to cellular components of the immune system, resulting in suppression or desensitization of the allergic response to Cryptomeria japonica pollen allergens. A non-restrictive example of this is the use of appropriate B- and T-cell epitope peptides, or modifications thereof, based on the cDNA/protein structures of the present invention to suppress the allergic response to Japanese cedar pollen. This can be carried out by defining the structures of B- and T-cell epitope peptides which affect B- and T-cell function in in vitro studies with blood components from Japanese cedar pollen sensitive individuals. Protein, peptides or antibodies of the present invention can also be used for detecting and diagnosing Japanese cedar pollinosis. For example, this could be done by combining blood or blood products obtained from an individual to be assessed for sensitivity to Japanese cedar pollen with an isolated antigenic peptide or peptides of Cryj II, or isolated Cryj II protein, under conditions appropriate for binding of components in the blood (e.g., antibodies, T-cells, B-cells) with the peptide(s) or protein and detennining the extent to which such binding occurs. Other diagnostic methods for allergic diseases which the protein, peptides or antibodies of the present invention can be used include radio-allergosorbent test (RAST), paper radioimmunosorbent test (PRIST), enzyme linked immunosorbent assay (ELISA), radioimmunoassays (RIA), immuno-radiometric assays (IRMA), luminescence immunoassays (LIA), histamine release assays and IgE immunoblots.
In another diagnostic test, the presence in individuals of IgE specific for Cryj II at least one protein allergen and the ability of T cells of the individuals to respond to T cell epitope(s) of Cryj II protein allergen can be determined by administering to the individuals an Immediate Type Hypersensitivity test and a Delayed Type Hypersensitivity test. The individuals are administered an Immediate Type Hypersensitivity test (see e.g. Immunology (1985) Roitt, I.M., Brostoff, J., Male, D.K. (eds), CN. Mosby Co., Gower Medical Publishing, London, ΝY, pp. 19.2-19.18; pp. 22.1-22.10) utilizing the Cry i II protein allergen or a portion thereof, or a modified form of the Cryy // protein allergen or a portion thereof, each of which binds IgE specific for the allergen. The same individuals are administered a Delayed Type Hypersensitivity test prior to, simultaneously with, or subsequent to administraiton of the Immediate Type Hypersensitivity test. Of course, if the Immediate Type Hypersensitivity test is administered prior to the
Delayed Type Hypersensitivity test, the Delayed Type Hypersensitivity test would be given to those individuals exhibiting a specific Immediate Type Hypersensitivity reaction. The Delayed Type Hypersensitivity test utilizes a modified form of the protein allergen or a portion thereof, the protein allergen produced recombinantly, or a recombitope peptide derived from the protein allergen, each of which has human T cell stimulating activity and each of which does not bind IgE specific for the allergen in a substantial percentage of the population of individuals sensitive to the allergen (e.g., at least about 75%). Based on the results of the above diagnostic tests, those individuals found to have both a specific Immediate Type Hypersensitivity reaction and a specific Delayed Type Hypersensitivity reaction are suitable candidates for administration of a therapeutically effective amount of a therapeutic composition. The therapeutic composition comprises the modified form of the protein or portion thereof, the recombinantly produced protein allergen, or the recombitope peptide, each as used in the Delayed Type Hypersensitivity test, and a pharmaceutically acceptable carrier or diluent.
The present invention also provides a method of producing Cryj II or fragment thereof comprising culturing a host cell containing an expression vector which contains DNA encoding all or at least one fragment of Cryj II under conditions appropriate for expression of Cryj II or at least one fragment. The expressed product is then recovered, using known techniques. Alternatively, Cryj II or fragment thereof can be synthesized using known mechanical or chemical techniques.
The DNA used in any embodiment of this invention can be cDNA obtained as described herein, or alternatively, can be any oligodeoxynucleotide sequence having all or a portion of a sequence represented herein, or their functional equivalents. Such oligodeoxynucleotide sequences can be produced chemically or enzymatically, using known techniques. A functional equivalent of an oligonucleotide sequence is one which is 1) a sequence capable of hybridizing to a complementary oligonucleotide to which the sequence (or corresponding sequence portions) of Cryj II or fragments thereof hybridizes, or 2) the sequence (or corresponding sequence portion) complementary to Cryj II, and/or 3) a sequence which encodes a product (e.g., a polypeptide or peptide) having the same functional characteristics of the product encoded by the sequence (or corresponding sequence portion) of Cryj II. Whether a functional equivalent must meet one or both criteria will depend on its use (e.g., if it is to be used only as an oligoprobe, it need meet only the first or second criteria and if it is to be used to produce a Cryj II allergen, it need only meet the third criterion).
The invention is further illustrated by the following non-limiting examples.
Example 1
Purification of Native Japanese Cedar Pollen Allergen (Cry II)
The following purification of native Cry j II from Japanese cedar pollen was modified from previously published reports (Yasueda et al, J. Allergy Clin. Immunol. 71:77 (1983); Sukaguchi et al., Allergy, 45:309 (1990)). lOOg of Japanese cedar pollen obtained from Japan (Hollister-Stier, Spokane, WA) was defatted in 1L diethyl ether three times, the pollen was collected after filtration and the ether was dried off in a vacuum.
The defatted pollen was extracted at 4°C overnight in 2L extraction buffer lθ
containing 50 mM tris-HCl, pH 7.8, 0.2 M NaCl and protease inhibitors in final concentrations: soybean trypsin inhibitor (2 μg/mL), leupeptin (1 μg/mL), pepstatin A (1 μg/mL) and phenyl methyl sulfonyl fluoride (0.17 mg/mL). The insoluble material was re-extrated with 1.2L extraction buffer at 4°C overnight and both extracts were combined together and depigmented by batch absorption with
Whatman DE-52 (200g dry weight) equilibrated with the extraction buffer.
The depigmented material was then fractionated by ammonium sulfate precipitation at 80% saturation (4°C), which removed much of the lower molecular weight material. The resulting pellet was resuspended in 0.4 L of 50 mM Na- acetate, pH 5.0 containing protease inhibitors and was dialyzed extensively against the same buffer.
The sample was further subjected to purification by either one of the two methods described below.
Method A
The sample was applied to a 100 mL DEAE cellulose column (Whatman DE- 52) equilibrated at 4°C with 50 mM Na-acetate, pH 5.0 with protease inhibitors. The unbound material (basic proteins) from the DEAE cellulose column was then applied to a 50 ml cation exchange column (Whatman CM-52) which was equilibrated with 10 mM Na-acetate, pH 5.0 at 4°C with protease inhibitors. A linear gradient of 0-0.3 M NaCl was used to elute the proteins. The early fractions were enriched in Cryj I whereas the later fractions were enriched in Cryj II. Fractions containing Cryj II were pooled and next applied to an 1 mL Mono S HR 5/5 column (Pharmacia, Piscataway, NJ) in 10 mM Na-acetate, pH 5.0, and proteins were eluted with a linear gradient of NaCl at room temperature. Residual Cryj I was eluted at "0.2 M NaCl and Cryj II was eluted between 0.3 to 0.4 M NaCl. The Cryj II peak was pooled and concentrated to twofold by lyophilization and subjected to gel filtration chromatography.
The sample was applied to FPLC Superdex 75 16/60 column (Pharmacia, Piscataway, NJ) in 10 mM acetate buffer, pH 5.0 and 0.15 M NaCl at a flow rate of
30 ml/min. at room temperature. Purified Cryj II was recovered in the 35-30 kD region. Cryj II migrated as two broad bands lower than Cry j I under non-reducing conditions (Fig. la) but both bands shifted upward and migrated as Cryj I under reducing condition (Fig. lb) when analyzed by silver-stained SDS-PAGE. This highly purified Cryj II still contained a small amount (~5%) of Cryj I as detected by
Western blot using MAb CBF2, which has been shown to bind to Cry j I and by N- terminal protein sequencing. This Cryj II preparation was used to generate primary protein sequence of Cry j II as described below.
Method B
The dialyzed sample from the ammonium sulfate precipitation was applied at 1 ml/min to an 5.0 ml Q-Sepharose Econapac anion exchange cartridge (BioRad,
Richmond, CA) equilibrated with 50 mM Na-acetate, pH 5.0 with protease inhibitors at 4°C. Elution was performed with the above buffer containing 0.5 M NaCl. The basic unbound material was then applied to a 5.0 ml CM-Sepharose Econopac cation exchange cartridge (BioRad, Richmond, CA) equilibrated in 50 mM sodium acetate pH 5.0 with protease inhibitors. Basic proteins were eluted with a linear gradient up to 0.1 M sodium phosphate pH 7.0, 0.3 M NaCl at 1 ml/min at 4°C. A Cry j II -enriched peak was collected late in the gradient and further purified by gel filtration chromatography.
FPLC gel filtration was performed using a 320 mL Superdex 75 26/60 (Pharmacia, Piscataway, NJ) column at 0.5 ml/min in 20 mM sodium acetate, pH
5.0, in the presence of 0.15 M NaCl. The major peak containing mostly Cry j II eluted between 160 and 190 ml. Contaminating Cry j I was next removed by FPLC using a 1.0 ml Mono S 5/5 (Pharmacia, Piscataway, NJ) cation exchange column equilibrated with 10 mM sodium acetate pH 5.0. A stepwise gradient of 0-1 M NaCl was utilized by holding isocratically at 0.2 M, 0.3 M, 0.4 M and 1 M salt concentration.
Multiple peaks (up to nine peaks) were obtained (Fig. 2) and analyzed by silver stained SDS-PAGE under reducing conditions (Fig. 3). Cryj I with a reported pi of 8.6-8.9 (Yasueda et al, J. Allergy Clin. Immunol, vol. 17 (1983)), eluted in the earlier peaks and displayed a molecular weight of about 40 kD. Cry j II was purified to homogeneity as two bands (Fig. 3) and eluted in the later multiple peaks, suggesting the existence of isoforms. ELISA analysis using the mouse monoclonal 8B11 IgG antibody which was raised against biochemically purified Cry j I confirmed the absence of Cryj I in these purified Cryj II preparation. This purified Cryj II was used in the human IgE reactivity studies (Example 6).
Physical properties of Cry II
The physiochemical properties of Cryj II were studied and summarized as below. Under non-reducing SDS-PAGE conditions Cryj II consists of two bands with molecular weights ranged 34000-32000. The molecular weights of both bands are shifted higher to about 38-36 kD under reducing conditions (Fig. lb). This shift in SDS-polyacrylamide gel has also been observed by others (Sakaguchi et al, -4//ergy45:309-312 (1990)). These results suggest that intra-disulfide bonds are probably present in the protein, and it is supported by the present findings that cloned Cryj II contains 20 cysteines deduced from the nucleotide sequence (Example 3). The pi of Cryj II estimated from IEF gel is about 10. The purified Cryj II binds human IgE of some allergic patients.
The two molecular weight bands of Cryj II were separated on a 12% SDS- polyacrylamide gel and was then electroblotted onto PNDF membrane (Applied Biosystems, Foster City, CA). The blot was stained with coomassie brilliant blue and was cut and subjected to Ν-terminal amino acid sequencing. (Example 2). The results showed that the upper and lower molecular weight bands had identical Ν- terminal sequences except the lower molecular weight band missed the first five amino acids. The estimated molecular weight of the upper band based on the cDΝA sequence is about 52,000, which is significantly higher than the molecular weight estimated from SDS-polyacrylamide gel either in the presence or absence of reducing reagent. It is also higher than that obtained from gel filtration and preliminary mass spectroscopy analysis. These are several possibilities to account for this difference. One possibility is that Cry j II protein is processed. It is probable that the Ν- terminal and C-terminal of the protein are cleaved. It is not clear at the present time whether this processing occurs in the cell or due to proteolysis during purification even though four different protease inhibitors were added in most of the purification steps. Nevertheless, the two N-terminal sequences obtained from the purified Cryj II (Example 2) also contained the N-terminal sequence (10 amino acid) published by Sakaguchi et al {Allergy, 45:309-312(1990)) suggesting that the N-terminal of Cryj II is probably hydrolyzed. Since Sakaguchi et al. (supra), did not use any protease inhibitors in their purification, a higher degree of hydrolysis might have occurred.
This could explain why the N-terminal amino acid sequence that Sakaguchi et al. obtained was downstream of the N-terminal sequences as discussed in Example 2.
Another approach which may be used to purify native Cryj II or recombinant Cryj II is immunoaffinity chromatography. This technique provides a very selective protein purification due to the specificity of the interaction between monoclonal antibodies and antigen. Murine polyclonal and monoclonal antibodies are generated against purified Cryj II. These antibodies are used for purification, characterization, analysis and diagnosis of the allergen Cry j II.
Example 2
Protein Sequencing of Purified Cryj II
Cryj II protein was isolated as in Example 1. The doublet band shown on SDS-PAGE (Fig. la) was electroblotted onto ProBlott (Applied Biosystems, Foster City, CA). Sequencing was performed with the Beckman/Porton Microsequencer (model LF3000, Beckman Instruments, Carlsbad, CA), a Programmable Solvent Module (Beckman System Gold Model 126, Beckman Instuments, Carlsbad, CA) and a Diode Array Detector Module for PTH-amino acid detection (Beckman System
Gold Model 168, Beckman Instruments, Carlsbad, CA) following manufacturers specifications.
A single N-terminal sequence analysis of the upper doublet band and multiple N-terminal sequence analyses of the lower doublet band showed that both bands contained two N-termini, designated "long" and "short". The lower doublet band contained approximately 3.3 picomoles of the long form and 8.3 picomoles of the short form. This difference in yields was sufficient to make sequence assignments according to the quantitation at each sequencer cycle. The upper doublet band contained approximately 8.3 picomoles of both sequences. The revealed long sequence was NH2-RKVEHSRHD- NIFNNEKYGANGDGKH-DCTEAFSTAW(Q)
( ) ( ) ( ) KΝP ( ) -COOH, (SEQ ID NO: 4) where (Q) indicates a tentative identification of glutamine at position 38 and () indicated unknown residues at positions 39-41 and 45. The revealed "short" sequence was NH2- SRHDAINIFNVEKYGAVGDGKHDCTEAFSTAWS-COOH (SEQ ID NO: 5). Thus the long Cryj II sequence had five additional amino terminal residues than the short form and the sequence of the short form exactly matched that of the long form. In addition, both the long and short forms of Cry j II contained the ten amino acids, NH2-AINIFNVEKY-COOH (SEQ ID NO: 6), previously described for Cryj II (Sakaguchi et al. 1990, supra). The previously published ten amino acids (Sakaguchi et al. 1990, supra) correspond to amino acids ten through 19 of the long form described above.
Example 3
Extraction of RNA From Japanese Cedar Pollen and Staminate Cones and Cloning of Crv i II
Fresh pollen and staminate cone samples, collected from a single Cryptomeria japonica (Japanese Cedar) tree at the Arnold Arboretum (Boston, MA), were frozen immediately on dry ice. RNA was prepared from 500 mg of each sample, essentially as described by Frankis and Mascarhenas (1980) Ann. Bot. 45: 595-599. The samples were ground by mortar and pestle on dry ice and suspended in 5 ml of 50 mM Tris pH 9.0 with 0.2 M NaCl, 1 mM EDTA, 0.1 % SDS that had been treated overnight with 0.1 % diethyl pyrocarbonate (DEPC). After five extractions with phenol/chloroform/isoamyl alcohol (mixed 25:24: 1), the RNA was precipitated from the aqueous phase with 0.1 volume 3M sodium acetate and 2 volumes ethanol. The pellets were recovered by centrifugation, resuspended in 2 ml dH2θ and heated to 65 °C for 5 minutes. Two ml 4M lithium chloride was added to the preparation and the RNA was precipitated overnight at 0°C. The RNA pellets were recovered by centrifugation, resuspended in 1 ml dH2θ, and again precipitated with 3M sodium acetate and ethanol on dry ice for one hour. The final pellet was washed with 70% ethanol, air dried and resuspended in 100 μl DEPC-treated dH2θ and stored at -80°C. Double stranded cDNA was synthesized from 4 μg pollen RNA or 8 μg flowerhead RNA using a commercially available kit (cDNA Synthesis System kit, BRL, Gaithersburg, MD). The double-stranded cDNA was phenol extracted, ethanol precipitated, blunted with T4 DNA polymerase (Promega, Madison, WI), and then ligated to ethanol precipitated, self annealed, AT and AL oligonucleotides for use in a modified Anchored PCR reaction, according to the method of Rafnar et al. (1990) J. Biol. Chem. 266: 1229-1236 ; Frohman et al. (1990) Proc. Natl. Acad. Sci. USA 85- 8998-9002; and Roux et al. (1990) BioTech. 8: 48-57. Oligonucleotide AT has the sequence (SEQ ID NO: 10) 5 ' -GGGTCTAGAGGTACCG-TCCGTCCGATCGATC ATT-3 ' (Rafnar et al. supra). Oligonucleotide AL has the sequence (SEQ ID NO: 11)
5'-AATGATCGATGCT (Rafnar et al. supra).
The first attempts at amplifying the amino terminus of Cryj II from the linkered cDNA (2 μl of a 20 μl reaction) was made using the degenerate oligonucleotide CP-11 and oligonucleotide AP. CP-11 has the sequence (SEQ ID NO: 12) 5'-ATACTTCTCIACGTTGAA-3' , wherein A at positon 1 can be G, C at position 4 can be T, C at position 7 can be T, I at position 10 is inosine to reduce degeneracy (Knoth et al. (1988) Nucleic Acids Res. 16: 10932), G at position 13 can be A, and G at position 16 can be A). AP, which has the sequence (SEQ ID NO: 13) 5'-GGGTCTAGAGGTA-CCGTCCG-3' , corresponds to nucleotides 1 through 20 of the oligonucleotide AT. CP-11 is the degenerate oligonucleotide sequence that is complementary to the coding strand sequence substantially encoding amino acids PheAsnValGluLysTyr (SEQ ID NO: 14) (amino acids 59 to 64 of Fig. 4), which correspond to the carboxy terminus of the previously published Cry j II sequence (Sakaguchi et al. , supra) shown in Fig. 4. All oligonucleotides were synthesized by Research Genetics Inc., Huntsville, AL.
Polymerase chain reactions (PCR) were carried out using a commercially available kit (GeneAmp DNA Amplification kit, Perkin Elmer Cetus, Norwalk, CT) whereby 10 μl lOx buffer containing dNTPs was mixed with 100 pmoles of each oligonucleotide, cDNA (3-5 μl of a 20 μl first strand cDNA reaction mix), 0.5 μl Amplitaq DNA polymerase, and distilled water to 100 μl.
The samples were amplified with a programmable thermal controller (MJ Research, Inc., Cambridge, MA). The first 5 rounds of amplification consisted of denaturation at 94 °C for 1 min, annealing of primers to the template at 45 °C for 1 min, and chain elongation at 72 °C for 1 min. The final 20 rounds of amplification consisted of denaturation as above, annealing at 55 °C for 1 min, and elongation as above. The primary PCR reaction was carried out with 100 pmol each of the oligonucleotides AP and CP-11. Five percent (5 μl) of this initial amplification was then used in a secondary amplification with 100 pmoles each of AP and CP-12. CP- 12 has the sequence (SEQ ID NO: 15) 5'-CCTGCAGTACTTCT- CIACGTTGAAIAT-3' , wherein C at position 10 can be T, C at position 13 can be T, I at positions 16 and 25 are inosines to reduce degeneracy as above, G at position 19 can be A, and G at position 22 can be A. The sequence (SEQ ID NO: 16) 5'-
CCTGCAG-3' (bases 1 through 7 of CP-12) represents a Pst I site added for cloning purposes; the remaining degenerate oligonucleotide sequence is complementary to the coding strand sequence that substantially encodes the amino acids IlePheAsnValGluLysTyr (SEQ ID NO: 17) (amino acids 58-64 of Fig. 4). Amplified DNA was recovered by sequential chloroform, phenol, and chloroform extractions, followed by precipitation on dry ice with 0.5 volumes of 7.5M ammonium acetate and 1.5 volumes of isopropanol. After precipitation and washing with 70% ethanol, the DNA was simultaneously digested with Xba I and Pst I in a 50 μl reaction, precipitated to reduce the volume to 10 μl, and electrophoresed through a preparative 2% GTG NuSeive low melt gel (FMC, Rockport, ME). The appropriate sized DNA area was visualized by ethidium bromide (EtBr) staining, excised, and ligated into appropriately digested pUC19 for sequencing by the dideoxy chain termination method of Sanger et al. (1977) Proc. Natl. Acad. Sci. USA 74: 5463-5476) using a commercially available sequencing kit (Sequenase kit, U.S. Biochemicals, Cleveland, OH). All resultant clones were sequenced, and none were found to contain Cryj II sequence. An alternate 2° PCR reaction was performed with AP and the nested oligonucleotide CP-21. CP-21 has the sequence (SEQ ID NO: 18) 5'-CCTGCAGTACTTCTCIACGTTGAAGAT-3* wherein C at position 10 can be T, C at position 13 can be T, I at position 16 is inosine to reduce degeneracy as above, G at position 19 can be A, G at position 22 can be A, and G at position 25 can be A or T. The sequence (SEQ ID NO: 16) 5'-CCTGCAG-3* (bases 1 through 7 of CP-21) represent a Pst I site added for cloning purposes; the remaining degenerate oligonucleotide sequence is the non-coding strand sequence corresponding to coding strand sequence substantially encoding amino acids IlePheAsnNalGluLysTyr (SEQ ID NO: 17) (amino acids 58 to 64 of Fig. 4).
A primary PCR was also performed on double-stranded, linkered cDNA using CP-23D and AP, as above, to attempt to amplify the 3' end of the Cryj II cDNA. A secondary PCR was performed using 5 % of the primary reaction, using CP-24D and AP. CP-23D (sequence (SEQ ID NO: 19) 5'- GCIATTAATATTTTTAA-3' , wherein the T at position 6 can be C or A, T at position 9 can be C, T at position 12 can be C or A, and T at position 15 can be C ) is the coding strand sequence substantially encoding amino acids AlalleAsnllePheAsn
(SEQ ID NO: 20) (amino acids 55 to 60 of Fig. 4); CP-24D (SEQ ID NO: 21) (sequence 5'-GGAATTCCGCIATTAATATTTTTAATGT-3', wherein the T at position 14 can be C or A, T at position 17 can be C, T at position 20 can be C or A, T at position 23 can be C, and T at position 26 can be C ) contains the sequence 5'-GGAATTCC-3' (SEQ ID NO: 22) (bases 1 through 8 of CP-24), which represents an Eco RI site added for cloning purposes. The remaining degenerate oligonucleotide sequence of CP-24D substantially encodes amino acids AlalleAsnllePheAsnVal (SEQ ID NO: 23) (amino acids 55 to 61 of Fig. 4). Again, multiple clones were sequenced, none of which could be identified as Cryj II, and this approach was not pursued further.
Upon the characterization of novel Cryj II protein sequence data described in Example 2, new degenerate oligonucleotides for cloning Cryj II were designed and synthesized. All oligonucleotides mentioned hereafter were synthesized on an ABI 394 DNA/RNA Synthesizer (Applied Biosystems, Foster City, CA), and purified on NAP-10 columns (Pharmacia, Uppsala, Sweden) as per the manufacturers' instructions. Degenerate oligonucleotide CP-35 was used with AP on the double- stranded linkered cDNA in a primary PCR reaction carried out as described herein. CP-35 has the sequence (SEQ ID NO: 24) 5'-GCTTCGGTACAATCATGTTT-3' , wherein T at position 3 can also be C; G at position 6 can also be A, T or C; A at position 9 can also be G; A at position 12 can also be G; A at position 15 can be G; and T at position 18 can also be C; this degenerate oligonucleotide sequence is the non-coding strand sequence corresponding to coding strand sequence substantially encoding amino acids LysHisAspCysThrGluAla of Cry j II (SEQ ID NO: 25) (amino acids 71 to 77 of Fig. 4). Five percent (5 μl) of this initial amplification, designated JC136, was then used in a secondary amplification with 100 pmoles each of AP and degenerate Cryj II primer CP-36, an internally nested Cryj II oligonucleotide primer with the sequence (SEQ ID NO: 26) 5'- GGCTGCAGGTACAATCATGTTTGCCATC-3' wherein A at position 11 can also be G; A at position 14 can also be G; A at position 17 can also be G; T at position 20 can also be C; G at position 23 can also be A, T, or C; and A at position 26 can also be G. The nucleotides 5'-GGCTGCAG-3' (SEQ ID NO: 27) (bases 1 through 8 of CP-36) represent a Pst I restriction site added for cloning purposes. The remaining degenerate oligonucleotide sequence of CP-36 is the non-coding strand sequence corresponding to coding strand sequence substantially encoding amino acids AspGlyLysHisAspCysThr of Cryj II (SEQ ID NO: 28) (amino acids 69 to 75 of Fig. 4). The dominant amplified product, designated JC137, was a DNA band of approximately 265 base pairs, as visualized on an EtBr-stained 2% GTG agarose gel.
Amplified DNA was recovered by sequential chloroform, phenol, and chloroform extractions, followed by precipitation at -20°C with 0.5 volumes of 7.5 ammonium acetate and 1.5 volumes of isopropanol. After precipitation and washing with 70% ethanol, the DNA was simultaneously digested with Xba I and Pst I in a 15 μl reaction and electrophoresed through a preparative 2% GTG SeaPlaque low melt gel (FMC, Rockport, ME). The appropriate sized DNA band was visualized by EtBr staining, excised, and ligated into appropriately digested pUC19 for sequencing by the dideoxy chain termination method (Sanger et al. (1977) Proc. Natl Acad Sci. USA 74: 5463-5476) using a commercially available sequencing kit (Sequenase kit, U.S. Biochemicals, Cleveland, OH).
The clones designated pUC19JC137a, pUC19JC137b, and pUC19JC137e were found to contain sequences encoding the amino terminus of Cryj II. All three clones had identical sequence in their regions of overlap, although all three clones had different lengths in the 5' untranslated region. Clone pUC19JC137b was the longest clone. The translated sequence of these clones had complete identity to the disclosed 10 amino acid sequence of Cryj II (Sakaguchi et al., supra.), as well as to the Cryj II amino acid sequence described in Example 2. Amino acid numbering is based on the sequence of the full length protein; amino acid 1 corresponds to the initiating memionine (Met) of Cryj II. The position of the initiating Met was supported by the presence of an upstream in-frame-stop codon and by 78% homology of the surrounding nucleotide sequence with the plant consensus sequence that encompasses the initiating Met, as reported by Lutcke et al. (1987) EMBO J. 6:43-48.
The cDNA encoding the remainder of Cryj II gene was cloned from the linkered cDNA by using oligonucleotides CP-37 (SEQ ID NO: 29) (which has the sequence 5'-ATGTTGGACAGTGTTGTCGAA-3') and AP in a primary PCR, designated JC138U. Oligonucleotide CP-37 corresponds to nucleotides 129 to 149 of *
Fig. 4, and is based on the nucleotide sequence determined for the partial Cryj II clone pUC19JC137b.
A secondary PCR reaction was performed on 5% of the initial amplification mixture, with 100 pmoles each of AP and CP-38 (SEQ ID NO: 30) (which has the sequence 5'-GGGAATTCAGAAAAGTTGAGCATTCTCGT-3'), the nested primer.
The nucleotide sequence (SEQ ID NO: 31) 5'-GGGAATTC-3' (bases 1 through 8 of CP-38) represents an Eco RI restriction site added for cloning purposes. The remaining oligonucleotide sequence corresponds to nucleotides 177 to 197 of Fig. 4, and is based on the nucleotide sequence determined for the partial Cryj II clone pUC19JC137b. The amplified DNA product, designated JC140iii, was purified and precipitated as above, followed by digestion with Eco RI and Asp 718 and electrophoresis through a preparative 1 % low melt gel. The dominant DNA band, which was approximately 1.55 kb in length, was excised and ligated into pUC19 for sequencing. DNA was sequenced by the dideoxy chain termination method (Sanger et al. supra) using a commercially available kit (sequenase kit (U.S. Biochemicals,
Cleveland, OH). Both strands were completely sequenced using M13 forward and reverse primers (N.E. Biolabs, Beverly, MA) and internal sequencing primers CP- 35, CP-38, CP-40, CP-41, CP-42, CP-43, CP-44, CP-45, CP-46, CP-47, CP-48, CP-49,CP-50, and CP-51. CP-40 (SEQ ID NO: 32) has the sequence 5'- GTTCTTCAATGGGCCATGT-3' and corresponds to nucleotides 359 to 377 of Fig.
4. CP-41 (SEQ ID NO: 33) has the sequence 5'- GTGTTAGGACT- GTCTCTCGG-3', which is the non-coding strand sequence that corresponds to nucleotides 720 to 739 of Fig. 4. CP-42 (SEQ ID NO: 35) has the sequence 5'-TGTCCAGGCCAT-GGAATAAG-3', which corresponds to nucleotides 864 to 883 of Fig. 4 except that the first nucleotide was synthesized as a T rather than the correct G. CP-43 has the sequence (SEQ ID NO: 35) 5'-
GCCTTACATGGACTGCAACC-3', which is the non-coding strand sequence that corresponds to nucleotides 1476 to 1495 of Fig. 4. CP-44 has the sequence (SEQ ID NO: 36) 5'-TCCACGGGTCTGATAATCCA-3\ which corresponds to nucleotides 612 to 631 of Fig. 4. CP-45 has the sequence (SEQ ID NO: 37)
5'-AGGCAGGAAGCAATTTT-CCC-3', which is the non-coding strand sequence that corresponds to nucleotides 1254 to 1273 of Fig. 4. CP-46 has the sequence (SEQ ID NO: 38) 5'-TACTGCACTTCAGCT-TCTGC-3', which corresponds to nucleotides 1077 to 1096 of Fig. 4. CP-47 has the sequence (SEQ ID NO: 39) 5 ' -GGGGGTCTCCG AATTTATC A-3 ' , which is the non-coding strand sequence that substantially corresponds to nucleotides 1039 to 1058 of Fig. 4, except that the fifth nucleotide of CP-47 was synthesized as a G rather than the correct nucleotide, T. CP-48 (SEQ ID NO: 40), which has the sequence 5'-
GGATATTTCAGTGGACACGT-3', corresponds to nucleotides 1290 to 1309 of Fig. 4. CP-49 (SEQ ID NO: 41) has the sequence 5'-TATTAGAAGACC- CTGTGCCT-3', which is the non-coding strand sequence that corresponds to nucleotides 821 to 840 of Fig. 4. CP-50 (SEQ ID NO: 42) has the sequence
5'-CCATGTAAGGCCAAGTTAGT-3', which corresponds to nucleotides 1485 to 1504 of Fig. 4. CP-51 (SEQ ID NO: 43) has the sequence
5'-ACACCTTTACCCATTAGAGT-3' , which is the non-coding strand sequence that corresponds to nucleotides 486 to 505 of Fig. 4. Three clones, designated ρUC19JC140iiia, pUC19JC140iiid and pUC19JC140iiie, were subsequently found to contain partial Cryj II sequence. The sequence of clone pUC19JC140iiid was chosen as the consensus sequence since it had the longest 3' untranslated region. The sequences of pUC19JC140iiid and pUC19JC137b were used to construct the composite Cryj II sequence shown in Fig. 4. In this composite, nucleotide 230 is reported as the A found in pUC19JC137b
(also, pUC19JC137a, ρUC19JC140iiia and ρUC19JC140ϋie) not as the G found in pUC19JC140iiid; however both A and G at nucleotide 230 encode Lys at amino acid 63. The sequence of clone pUC19JC140iiia was identical to that of pUC19JC140iiid except for the following: pUC19JC140iiia has a T at nucleotide 357 in place of a C (no predicted change in amino acid 106), has C at nucleotide 754 instead of T
(changes amino acid 238 from He to Thr), C at nucleotide 1246 instead of T (changes amino acid 402 from Leu to Pro), and T at nucleotide 1672 instead of C (untranslated region). The sequence of clone pUC19JC140iiie was identical to that of pUC19JC140iiid except for G at nucleotide 794 instead of A (changes amino acid 251 from He to Met), and T at nucleotide 357 in place of C (no predicted change in amino acid 106).
An earlier attempt at cloning the JC140iii PCR product using an Eco RllXba I digest (oligonucleotide AP has both Xba I and Asp 718 restriction enzyme sites) yielded cDNA that was cut in half due to an internal Xba I restriction site in the Cry j II cDNA, giving rise to 800 and 750 bp bands; the 750 bp band was succesfully cloned into Eco RllXba I digested pUC19 and sequenced. Two 750 bp clones were sequenced and found to be the 5' half of the Cryj II molecule: clones pUC19JC140- 2a and pUC19JC140-2b. Clone pUC19JC140-2a has C for nucloeotide 297 instead of T (changes amino acid 86 from Cys to Arg) and clone pUC19JC140-2b has G for nucleotide 753 instead of A (changes amino acid 238 from He to Val). Both clone pUC19JC140-2a and clone pUC19JC140-2b have a T at nucleotide 357 in place of C (no predicted change in amino acid 106). Two different PCR amplifications were also sequenced directly to verify the clonal Cryj II sequence using the Amplitaq Cycle Sequencing kit (Perkin Elmer Cetus, Norwalk, CT). This procedure involves the [32p] -end-labelling of oligonucleotide sequencing primers which are then annealled (1.6 pmoles in 1 μl) to template DNA and elongated with dideoxy NTPs (methodology of Sanger et al.
(1977) Proc. Natl. Acad. Sci. USA 74:5463-5476) in a PCR reaction also containing 4 μl 10X Cycling Mix (contains 0.5 U/μl Amplitaq DNA Polymerase), 5 μl template DNA (10-100 fmoles) and dH2θ to 20 μl . The dGTP in the termination mixes in this kit have been replaced by 7-deaza-dGTP, which provides increased resolution of sequences containing high G+C regions of DNA. The template DNA was a PCR product that was recovered by sequential chloroform, phenol, and chloroform extractions, precipitated at -20°C with 0.5 volumes of 7.5 ammonium acetate and 1.5 volumes of isopropanol, then electrophoresed through a preparative 1 or 2% SeaPlaque low melt gel (FMC). Appropriate sized DNA bands were visualized by EtBr staining, excised, and treated with Gelase (Epicentre Technologies, Madison,
WI) to remove the agarose. The DNA was again precipitated, and resuspended in 50 μl TE (10 mM Tris, pH 7.4, 1 mM EDTA, pH 8.0) containing 20 μg/ml RNAse (Boehringer Mannheim, Indianapolis, IN). Two secondary amplifications which had been used to clone Cryj II were repeated, and used as template DNA for PCR cycle sequencing: JC137U, the 5' end PCR, (amplified from the 1° PCR JC136 above) was reamplified with oligonucleotides AP and CP-36; and JC140U, the 3' end PCR, (amplified from the 1 ° PCR JC138U above) was reamplified with oligonucleotides AP and CP-38. Both of the 1 ° amplifications used were precipitated, electrophoresed through a preparative 1 or 2% SeaPlaque low melt gel (FMC), and the appropriate sized bands were visualized by EtBr staining and excised. Two μl of each 1 ° amplification was then used in the corresponding 2° PCR reaction. The 2° PCR product was then prepared as DNA template for PCR cycle sequencing as described above. The oligonucleotides used as primers in PCR cycle sequencing, many of which were used to sequence the clones, are as follows: for JC137U, CP-36 and CP-39 (SEQ ID NO: 44), which has the sequence 5'-
CTGTCCAACATAATTTGGGC-3' and is the non-coding strand sequence corresponding to nucleotides 120 to 139 of Fig. 4. The oligonucleotide primers used for sequencing JC140U were CP-38, CP-40, CP-41, CP-42, CP-43, CP-44, CP-45, CP-46, CP-47, CP-49, CP-50, CP-54 (SEQ ID NO: 45), which has the sequence 5'- CATGGCAGGGTGGTTCAGGC-3 ' , corresponds to nucleotides 985 to 1004 of Fig.
4, CP-55 (SEQ ID NO: 46), which has the sequence 5'-TAGCCCCATTTACGTGCACG-3' and is the non-coding strand sequence that corresponds to nucleotides 929 to 948 of Fig. 4, and CP-56 (SEQ ID NO: 47), which has the sequence 5'-TTGGGGTCGAGGCCTCCGAA-3' and corresponds to nucleotides 1437 to 1456 of Fig. 4. The sequence of this full-length PCR cycle sequencing had only 2 nucleotide changes from the composite pUC19JC137b/ρUC19JC140iiid Cryj II sequence shown in Figure 4, neither of which lead to an amino acid change. There was a T instead of C at nucleotide 357 (no predicted change in amino acid 106), and a C instead of A at nucleotide 635 (no amino acid change).
The nucleotide and predicted amino acid sequences of Cryj II are shown in Figs. 4 and 5. This is a composite nucleotide sequence from the two overlapping clones pUC19JC137b and pUC19JC140iiid. Sequencing of multiple independent clones and cycle sequencing of PCR product confirmed the nucleotide sequence of Figure 4. There were several nucleotide changes resulting in predicted amino acid changes, as cited above. However, all nucleotide polymorphisms, with the exception of the T for C substitition at nucleotide 357, were only observed in single clones or sequencing reactions. Although T was seen at nucleotide 357 in all clones except pUC19JC140iiid, both C and T encode Leu at amino acid 106.
The complete cDNA sequence for Cryj H is composed of 1726 nucleotides, including 41 nucleotides of 5' untranslated sequence, an open reading frame of 1542 nucleotides starting with the codon for an initiating Met (nucleotides 42-44 of Fig.
4), and a 143 bp 3' untranslated region. There is a consensus polyadenylation signal sequence in the 3' untranslated region 64 nucleotides 5' to the poly A tail (nucleotides 1654-1659 of Fig. 4). The position of the initiating Met is confirmed by the presence of an in-frame upstream stop codon and by 78% homology with the plant consensus sequence that encompasses the initiating Met (TAAAAUGGC (bases
38 through 46 of Fig. 4 (SEQ ID NO: 48)) found in Cryj II compared with the AACAAUGGC (SEQ ID NO: 49) consensus sequence for plants, Lutcke et al. (1987) EMBO J. 6: 43-48). The open reading frame encodes a deduced protein of 514 amino acids that has complete sequence identity with the published partial protein sequence for Cryj II (Sakaguchi et al. supra), which corresponds to amino acids 55 through 64 of Fig. 4. The predicted Cry j II protein has 20 Cys, contains four potential N-linked glycosylation sites corresponding to the consensus sequence N-X-S/T, has a predicted molecular weight of 56.6 kDa and a predicted pi of 9.08. Detection of three separate NH2 termini sequences for Cryj II (the long form and the short form as determined in Example 2 and the NH2 terminus determined by
Sakaguchi et al., supra, as shown in Fig. 6) may suggest that the amino terminus of the mature Cry j II protein is blocked and that the sequences obtained by sequence analysis of purified protein represent proteolytic cleavage products. As shown in Fig. 6, the amino acid sequence of the long form of Cryj II begins at amino acid 46 and the amino acid sequence of the short form of Cryj II begins at amino acid 51 ; and the NH2-terminal sequence determed by Sakaguchi et al. begins at amino acid 54. It is also possible that amino acids 1 to 45 represent the leader/pre-pro position of Cryj II that is enzymatically cleaved to give a functionally active protein beginning at amino acid 46 of Fig. 4. The sequences beginning at amino acids 51 and 54 represent breakdown products of the protein beginning at amino acid 46. There is a predicted cleavage site between amino acids 22 and 23 of Fig. 4 using the method of von Heijne (Nucleic Acids Res. (1986) 14:4683-4690). If the mature Cry j II protein started at amino acid 23 in Fig. 4, the protein would be 492 amino acids long with a predicted molecular weight of 54.2 kDa and a predicted pi of 9.0.
Searching the Swiss-Prot data base with the Cryj II sequence demonstrated that Cryj II is 43.3% homologous (33.3% identical to polygalacturonase of tomato (Lycopersicon esculentum) and 48.4% homologous (32.6% identical) to polygalacturonase of corn, Zea mays. All nucleotide and amino acid sequence analyses were performed using PCGENE (Intelligenetics, Mountain View, CA.).
Example 4
Extraction of RNA from Japanese Cedar Pollen Collected in Japan and
Expression of Recombinant Cryj II
Fresh pollen collected from a pool of Cryptomeria japonica (Japanese cedar) trees in Japan was frozen immediately on dry ice. RNA was prepared from 500 mg of the pollen, essentially as described by Frankis and Mascarenhas Ann. Bot. 45:595- 599. The samples were ground by mortar and pestle on dry ice and suspended in 5 ml of 50 mM Tris pH 9.0 with 0.2 M NaCl, 1 mM EDTA, 1 % SDS that had been treated overnight with 0.1 % DEPC. After five extractions with phenol/chloroform
/isoamyl alcohol (mixed at 25:24:1), the RNA was precipitated from the aqueous phase with 0.1 volume 3 M sodium acetate and 2 volumes ethanol. The pellets were recovered by centrifugation, resuspended in 2 ml dH2θ and heated to 65°C for 5 minutes. Two ml of 4 M lithium chloride were added to the RNA preparations and they were incubated overnight at 0°C. The RNA pellets were recovered by centrifugation, resuspended in 1 ml dH2θ, and again precipitated with 3 M sodium acetate and ethanol overnight. The final pellets were resuspended in 100 μl dH2θ and stored at -80°C.
Double stranded cDNA was synthesized from 8 μg pollen RNA using the cDNA Synthesis Systems kit (BRL) with oligo dT priming according to the method of Gubler and Hoffman (1983) Gene 25:263-269. PCRs were carried out using the Gene Amp DNA Amplification kit (Perkin Elmer Cetus) whereby 10 μl lOx buffer containing dNTPs was mixed with 100 pmol each of a sense oligonucleotide and an anti-sense oligonucleotide, cDNA (10 μl of a 400 μl double stranded cDNA reaction mix), 0.5 μl Amplitaq DNA polymerase, and distilled water to 100 μl.
The samples were amplified with a programmable thermal controller from MJ Research, Inc. (Cambridge, MA). The first 5 rounds of amplification consisted of denaturation at 94°C for 1 min, annealing of primers to the template at 45°C for 1 min, and chain elongation at 72°C for 1 min. The final 20 rounds of amplification consisted of denaturation as above, annealing at 55°C for 1 min, and elongation as above.
A new set of primer pairs was synthesized for amplification of a Cryj II cDNA from the initiating Met to the stop codon. CP-52 (SEQ ID NO: 50) has the sequence 5'- GCCGAATTCATGGCCATGAAATTAATT-3 ' where the nucleotide sequence 5'-GCCGAATTC-3' (SEQ ID NO: 51) (bases 1 through 9 of CP-52 represents an Eco RI restriction site added for cloning purposes, and the remaining sequence corresponds to nucleotides 42 to 59 of Fig. 4. CP-53 (SEQ ID NO: 52) has the sequence 5'-CGGGGATCCTCATTATGGATG-GTAGAT-3' where the nucleotide sequence 5'-CGGGGATCC-3' (SEQ ID NO: 53) (bases 1 through 9 of CP-53 represents a Bam HI restriction site added for cloning purposes, and the remaining oligonucleotide sequence of CP-53 is complementary to coding strand sequence corresponding to nucleotides 1572 to 1589 of Fig. 4. The PCR reaction with CP-52 and CP-53 on the double stranded Japanese Cedar pollen cDNA yielded a band of approximately 1.55 kb on an EtBr-stained agarose minigel, and was called JC145. Amplified DNA was recovered by sequential chloroform, phenol, and chloroform extractions, followed by precipitation at -20°C with 0.5 volumes of 7.5 ammonium acetate and 1.5 volumes of isopropanol. After precipitation and washing with 70% ethanol, the DNA was simultaneously digested with Eco RI and Bam HI in a 15 μl reaction, and electrophoresed through a preparative 1 % SeaPlaque low melt gel (FMC). Appropriate sized DNA bands were visualized by EtBr staining, excised, and ligated into appropriately digested pUC19 for sequencing by the dideoxy chain termination method (Sanger et al. (1977) Proc. Natl. Acad. Sci. USA
74:5463-5476) using a commercially available sequencing kit (Sequenase kit, U.S. Biochemicals, Cleveland, OH).
Clones pUC19JC145a and pUC19JC145b were completely sequenced using M13 forward and reverse primers (N.E. Biolabs, Beverly, MA) and internal sequencing primers CP-41, CP-42, CP-44, CP-46, and CP-51. The nucleotide and deduced amino acid sequences of clones pUC19JC145a and pUC19JC145b were identical to the Cryj II sequence of Fig. 4, with the following exceptions. Clone pUC19JC145a was found to contain a single nucleotide difference from the previously known Cryj II sequence: it has a C at nucleotide position 1234 of Fig. 4 rather than the previously described T. This nucleotide change results in a predicted amino acid change from He to Thr at amino acid 398 of the Cryj II protein. Clone pUC19JC145b has a G at nucleotide position 1088 of Fig. 4 rather than the previously described A, and an A for a G at nucleotide 1339. The nucleotide change at 1088 is silent and does not result in a predicted amino acid change. The nucleotide change at position 1339 results in a predicted amino acid change from Ser to Asn at amino acid 433 of the Cryj II protein. None of these polymorphisms have yet been confirmed by independently-derived PCR clones or by direct amino acid sequencing and may be due to the inherent error rate of Taq polymerase (approximately 2 x 10"4, Saiki et al. (1988) Science 239:487-491). However, such polymorphisms in primary nucleotide and amino acid sequences are expected.
Expression of Cryj II was performed as follows. Ten μg of pUC19JC145b was digested simultaneously with Eco RI and Bam HI. The nucleotide insert encoding Cryj II (extending from nucleotide 42 through 1589 of Fig. 4) was isolated by electrophoresis of this digest through a 1 % SeaPlaque low melt agarose gel. The insert was then ligated into the appropriately digested expression vector pET-lld (Novagen, Madison, WI; Jameel et al. (1990) J. Virol. 64:3963-3966) modified to contain a sequence encoding 6 histidines (His 6) immediately 3' of the
ATG initiation codon followed by a unique Eco RI endonuclease restriction site. A second Eco RI endonuclease restriction site in the vector, along with neighboring Cla I and Hind III endonuclease restriction sites, had previously been removed by digestion with Eco RI and Hind III, blunting and religation. The histidine (Hisό) sequence was added for affinity purification of the recombinant protein (Cryj I) on a
Ni2 + chelating column (Hochuli et al. (1987) J. Chromatog. 411:177-184; Hochuli et al. (1988) Bio/Tech. 6:1321-1325.). A recombinant clone was used to transform Escherichia coli strain BL21-DE3, which harbors a plasmid that has an isopropyl-β- D-thiogalactopyranoside (IPTG)-inducible promoter preceding the gene encoding T7 polymerase. Induction with IPTG leads to high levels of T7 polymerase expression, which is necessary for expression of the recombinant protein in pET-1 Id. Clone pET-lldΔHRhis6JC145b.a was confirmed to be a Cry/ H clone in the correct reading frame for expression by dideoxy sequencing (Sanger et al. supra) with CP- 39. Expression of the recombinant protein was examined in an initial small culture. An overnight culture of clone pET-lldΔHRhis6JC145b.a was used to innoculate 50 ml of media (Brain Heart Infusion Media, Difco) containing ampicillin (200 μg/ml), grown to an A600 = 1.0 and then induced with IPTG (1 mM, final concentration) for 2 hrs. One ml aliquots of the bacteria were collected before and after induction, pelleted by centrifugation, and crude cell lysates prepared by boiling the pellets for 5 minutes in 50 mM Tris HC1, pH 6.8, 2 mM EDTA, 1 % SDS, 1 % β-mercaptoethanol, 10% glycerol, 0.25% bromophenol blue (Studier et al., (1990) Methods in Enzymology 185-60-89). Recombinant protein expression was examined on a 12% Coomassie blue-stained SDS-PAGE gel, according to the method in Sambrook et al. , supra, on which 25 μl of the crude lysates were loaded. A negative control consisted of crude lysate from uninduced bacteria containing the plasmid with Cryj II. There was no notable increase in production of any recombinant E. coli protein in the range of 58 Kd, the size predicted for the recombinant Cryj II with the His6 leader. The pET-1 ldΔHRhis6JC145b.a clone was then grown on a larger scale to examine if there was any recombinant protein being expressed. A 2 ml culture of bacteria containing the recombinant plasmid was grown for 8 hr, then 3 μl was spread onto each of 6 (100 x 15 mm) petri plates with 1.5% agarose in LB medium (Gibco-BRL, Gaithersburg, MD) containing 200 μg/ml ampicillin, grown to confluence overnight, then scraped into 6 L of liquid media (Brain Heart Infusion media, Difco) containing ampicillin (200 μg/ml). The culture was grown until the absorbance at A600 was 1.0, IPTG added (1 mM final concentration), and the culture grown for an additional 2 hours.
Bacteria were recovered by centrifugation (7,930 xg, 10 min) and lysed in 50 ml of 6M Guanidine-HCl, 0.1M Na2HPO4, pH 8.0, for 1 hour with vigorous shaking. Insoluble material was removed by centrifugation (11,000 xg, 10 min, 4° C). The pH of the lysate was adjusted to pH 8.0, and the lysate applied to a 50 ml
Nickel NTA agarose column (Qiagen) that had been equilibrated with 6 M Guanidine HC1, 100 mM Na2HPO4, pH 8.0. The column was sequentially washed with 6 M Guanidine HC1, 100 mM Na2HPO4, 10 mM Tris-HCl, pH 8.0, then 8 M urea, 100 mM Na2HPO4, pH 8.0, and finally 8 M urea, 100 mM sodium acetate, 10 mM Tris-HCl, pH 6.3. The column was washed with each buffer until the flow through had an A280_<- 0-05.
The recombinant Cryj II protein was eluted with 8 M urea, 100 mM sodium acetate, 10 mM Tris-HCl, pH 4.5, and collected in 10 ml aliquots. The protein concentration of each fraction was determined by A280 and the peak fractions pooled. An aliquot of the collected recombinant protein was analyzed on SDS-
PAGE according to the method in Sambrook et al. supra.
This 6L prep, JCIIpET-1, yielded 1.5 mg of recombinant Cr y II, which was resolved into 2 major bands on SDS-PAGE at 58 kDa and 24 kDa. The 58 kDa band, which represents recombinant Cryj II, was approximately 9-10% of the total protein as determined by densitometry measurement (Shimadzu Flying Spot Scanner,
Shimadzu Scientific Instruments, Inc., Braintree, MA). The 24 kDa band accounts for about 90% of the total protein and may represent a degradation product of the recombinant Cryj II or an E. coli contaminant.
Another Cryy II expression construct was made by the ligation of the pUC19JC140iiid Cryj II insert into appropriately digested pETl ldΔHR (with the 6 histidine leader). The vector was derived from another pETlldΔHR construct whose insert supplied an EcoR I site (at the 5' pETlldΔHR-insert junction) and an Asp 718 site (at the 3' end of the insert); the construct was digested with these two enzymes, run on a low melt minigel as above, and the vector recovered as a band in low melt agarose. The pUC19JC140iiid construct was digested with Eco R I and
Asp 718 to release the Cryy II insert, which was isolated on a low melt minigel and ligated into the Eco R I/Asp 718 digested pETlldΔHR vector prepared above. Five clones were found to contain the correct nucleotide sequence at the insert/vector 5' junction, when sequenced by dideoxy sequencing (as above) with CP-39. This new construct, when expressed, would begin at amino acid 46 of Cryy II as shown in Figs. 4 and 5. This recombinant protein is designated rCryy II Δ46. A 50 ml small scale expression test (as performed above) showed that the expression level of rCryy
II Δ46 from this construct, designated pETlldΔHRJC140iiid2, would be much greater than the initial expression level from pETlldΔHRJC145b2. A 9L prep, JCIIpET-3, was processed as above, and yielded 200 mg of rCryy II Δ46 at 80% purity as determined by densitometry of a Coomasie blue stained 12% SDS-PAGE gel.
Example 5
Northern blot on RNA from Japanese Cedar Pollen Sources
A northern blot analysis was performed on the RNA isolated from Japanese Cedar pollen from both the Arnold Arboretum tree and the pooled trees from Japan.
Using essentially the method of Sambrook, supra, ten μg of RNA isolated from Japanese cedar pollen collected from the Arnold Arboretum (Boston, MA) and 15 μg pooled RNA from Japanese cedar pollen collected from trees in Japan were run on a 1.2% agarose gel containing 38% formaldehyde and IX MOPS (20X = 0.4M MOPS, 0.02M EDTA, O.lM NaOAc, pH 7.0) solution. The RNA samples (first precipitated with 1/10 volume sodium acetate, 2 volumes ethanol to reduce volume and resuspended in 5.5 μl dH2O) were run with 10 μl formaldehyde/formamide buffer containing loading dyes with 15.5% formaldehyde, 42% formamide, and 1.3X MOPS solution, final concentration. The samples were transferred to Genescreen Plus (NEN Research Products, Boston, MA) by capillary transfer in 10X
SSC (20X = 3M NaCl, 0.3M Sodium Citrate), after which the membrane was baked 2 hrs at 80°C and UN irradiated for 3 minutes. Prehybridization of the membrane was at 60°C for 1 hour in 4 ml 0.5M ΝaPo4 (pH 7.2), lmM EDTA, 1 % BSA, and 7% SDS. The antisense probe was synthesized by asymmetric PCR on the JC145 amplification in low melt agarose (above), where 2 μl DNA is amplified with
2 μl dNTP mix (0.167mM dATP, 0.167mM dTTP, 0.167mM dGTP, and 0.033mM dCTP), 2 μl 10X PCR buffer, 10 μl 32P-dCTP (100 μCi; Amersham, Arlington Heights, II), 1 μl (100 pmoles) antisense primer CP-53, 0.5 μl Taq polymerase, and dH2O to 20 μl; the 10X PCR buffer, dNTPs and Taq polymerase were from Perkin Elmer Cetus (Norwalk, CT). Amplification consisted of 30 rounds of denaturation at 94°C for 45 sec, annealing of primer to the template at 60°C for 45 sec, and chain elongation at 72°C for 1 min. The reaction was stopped by addition of 100 μl TE, and the probe recovered over a 3cc G-50 spin column (2 ml G-50 Sephadex [Pharmacia, Uppsala, Sweden] in a 3cc syringe plugged with glass wool, equilibrated with TE) and counted on a 1500 TriCarb Liquid Scintillation Counter (Packard, Downers Grove, IL). The probe was added to the prehybridizing buffer at 106 cpm/ml and hybridization was carried out at 60°C for 16 hrs. The blot was washed in high stringency conditions: 3x15 min at 65°C with 0.2%SSC/1 % SDS, followed by wrapping in plastic wrap and exposure to film at -80°C. A seven hour exposure of this Northern blot analysis revealed a single thick band at approximately 1.7 kb for both RNA collected from the Arboretum tree and the RNA collected from the pooled trees from Japan. This message is the expected size for Cryj II as predicted by PCR analysis of the cDNA.
Example 6
Direct binding assay of IgE to Cry /' I. Cry y II and recombinant Cry i II. Corning assay plates (#25882-96) were coated with Cryy I or Cryy II at 2 μg/mL or recombinant Cr y II preparation at 10 μg/mL (approximately 20% pure) in a volume of 50 μL overnight at 4°C. The coating antigens were removed and the wells were blocked with 0.5 % gelatin, PVP (polyvinyl pyrolidine) 1 mg/ mL in PBS, 200 μL/well for 2 hours at room temperature. The anti- r y I monoclonal antibody, 4B11, was serially diluted in PBS-Tween 20 starting at a 1:1000 dilution.
The human plasma were serially diluted in PBS-Tween at a starting dilution of 1:2. For this set 23 plasma samples from patients symptomatic for Japanese cedar pollen allergy chosen for IgE binding analysis. The first antibody incubation proceeded overnight at 4°C. Following three washes with PBS-Tween the second antibodies were added (goat anti-mouse Ig or goat anti-human IgE both at 1:2000) and incubated for two hours at room temperature at 100 μL/well. This solution was removed and streptavidin-HRPO diluted to 1:10,000, was added at lOOμL/well. The color was allowed to develop for 2-5 minutes. The reaction was stopped by the addition of lOOμL/well of 1M phosphoric acid. Plates were read on a Microplate IL310 Autoreader (Biotek Instruments, Winooski, NT) with a 450nm filter. The absorbance levels of duplicate wells were averaged. The graphed results (log of the dilution vs. absorbance) of the ELISA assays are shown in Figs. 7 to 15. The summary of the results are given in Fig. 16. A positive binding result, indicated by a plus sign is determined to be a reading of two-fold or greater above background (no first antibody) at the second dilution of plasma (1:6).
In Fig. 7 the binding response of the monoclonal antibody, 4B11, and seven patients' (Batch 1) plasma IgE is shown to purified Cr y I as the coating antigen. The monoclonal antibody, raised against purified Cr y I shows a saturating level of binding for the whole dilution series. The individual patient samples show a variable response of IgE binding to the Cr y I preparation. One patient, #1034, has no detectable binding to this protein preparation. All the patient samples were obtained from individuals claiming to be symptomatic for Japanese cedar pollen allergy and the results of their MAST scores are shown in Fig. 16. Fig. 8 is a graph representing the binding of the same antibody set as in Fig. 7 to purified native Cry j II. The anti- Cryj I monoclonal antibody, 4B11, is negative on this preparation demonstrating lack of cross-reactivity between the two allergen antigens. In general, there is a lower overall response to this allergenic component of cedar pollen with more patient samples showing decreased binding. However, patient #1034, that was negative on Cryy I shows very strong reactivity to Cryj II. In the last antigen set, Fig. 9, using recombinant Cryy II (τCryj II), monoclonal antibody 4B11 reactivity is negative and there is further reduction in binding of the human IgE samples compared to biochemically purified Cryy II. Two of the patients, #1143 and #1146, are clearly positive for IgE binding to the recombinant form of Cryj II although the patient that reacted the strongest to biochemically purified form is negative here, 1034. Figs. 10-15 represent the application of the same antigen sets for the direct binding analysis of the next sixteen patients designated patient Batch 2 and patient Batch 3 in Figs. 10-15.
The table shown in Fig. 16 summarizes both the MAST scores, performed in Japan on the plasma samples before shipment using a commercially available kit, and the direct ELISA results outlined above. Two patients were negative by the MAST assay, however, one of these patients, #1143, was positive on all the ELISA antigens. The number of positive responses for each antigen is shown and this represents a measure relative allergenicity of the different allergen preparations. These results demonstrate that Cryy II is an allergen as defined by human allergic patient IgE reactivity and that there are some patients who are not reactive to Cryj I but are reactive to Cryy II. The frequency of response in this population of patients is less to Cryy II than to Cryy I.
Example 7
Japanese Cedar Pollen Allergic Patient T Cell Studies with Crv f II and Cry II Peptides.
Synthesis of Cryj II Peptides Japanese cedar pollen Cryy II peptides designated Cry j IIA Cry j IIB were synthesized using standard Fmoc/tBoc synthetic chemistry and purified by Reverse Phase HPLC. The amino acid sequence of peptide Cry J IIA is FTFKNDGIIAAYQ (SEQ ID NO: 54) which corresponds to amino acids 116-128 as shown if Figs 4 and 5. The amino acid sequence of peptide Cry j IIB is NGYFSGHNIPACKN (SEQ ID
NO: 55) which corresponds to amino acids 416-429 as shown in Figs 4 and 5. The peptide names are consistent throughout.
T Cell Responses to Japanese Cedar Pollen Antigen Peptides Peripheral blood mononuclear cells (PBMC) were purified by lymphocyte separation medium (LSM) centrifugation of 60 ml of heparinized blood from one Japanese cedar pollen-allergic patient who exhibited clinical symptoms of seasonal rhinitis and was MAST and/or skin test positive for Japanese cedar pollen. Long term T cell lines were established by stimulation of 2 X 10^ PBL/ml in bulk cultures of complete medium (RPMI-1640, 2 mM L-glutamine, 100 U/ml penicillin/streptomycin, 5xlO"5M 2-mercaptoethanol, and 10 mM HEPES supplemented with 5% heat inactivated human AB serum) with 10 μg/ml of partially purified native Cr y II for 7 days at 37°C in a humidified 5% CO2 incubator to select for Cry j II reactive T cells. This amount of priming antigen was determined to be optimal for the activation of T cells from most Japanese cedar pollen allergic patients. Viable cells were purified by LSM centrifugation and cultured in complete medium supplemented with 5 units recombinant human IL-2/ml and 5 units recombinant human IL-4/ml for up to three weeks until the cells no longer responded to lymphokines and were considered "rested". The ability of the T cells to proliferate to peptides Cry j IIA and Cry j IIB, recombinant Cryy II (τCryj II), purified native Cr y* II, or purified native Cryj I was then assessed. For assay, 2 X 104 rested cells were restimulated in the presence of 2 X 104 autologous Epstein- Barr virus (EBN)-transformed B cells (prepared as described below) (gamma- irradiated with 25,000 RADS) with 2-50 μg/ml of rCryy II, purified native Cryy II, peptides Cry j IIA and Cry j HB, of purified native Cryj I, in a volume of 200 μl complete medium in duplicate or triplicate wells in 96-well round bottom plates for 2-4 days. The optimal incubation was found to be 3 days. Each well then received 1 μCi tritiated thymidine for 16-20 hours. The counts incorporated were collected onto glass fiber filter mats and processed for liquid scintillation counting. The maximum response in a titration of each peptide is expressed as the stimulation index
(S.I.). The S.I. is the counts per minute (CPM) incorporated by cells in response to peptide, divided by the CPM incorporated by cells in medium only. An S.I. value equal to or greater than 2 times the background level is considered "positive" and indicates that the peptide contains a T cell epitope. The results of this assay indicated that peptides Crj II, and Cr y HB did noit appear to contain a T cell epitope for this particular allergenic patient. However, additional Japanese cedar pollen allergic patients will be tested in this assay system and one or both of these peptides may contain T cell epitopes for other allergic individuals.
Preparation of (EBV)-transformed B Cells for Use as Antigen Presenting Cells
Autologous EBN-transformed cell lines were γ-irradiated with 25,000 Rad and used as antigen presenting cells in secondary proliferation assays and secondary bulk stimulations. These cell lines were also used as a control in the immuno- fluorescence flow cytometry analysis. These EBN-transformed cell lines were made by incubating 5 X 106 PBL with 1 ml of B-59/8 Marmoset cell line (ATCC CRL1612, American Type Culture Collection, Rockville, MD) conditioned medium in the presence of 1 μg/ml phorbol 12-myristate 13-acetate (PMA) at 37°C for 60 minutes in 12 X 75 mm polypropylene round-bottom Falcon snap cap tubes (Becton Dickinson Labware, Lincoln Park, ΝJ). These cells were then diluted to 1.25 X 10^ cells/ml in RPMI- 1640 as described above except supplemented with 10% heat- inactivated fetal bovine serum and cultured in 200 μl aliquots in flat bottom culture plates until visible colonies were detected. They were then transferred to larger wells until the cell lines were established. Although the invention has been described with reference to its preferred embodiments, other embodiments, can achieve the same results. Variations and modifications to the present invention will be obvious to those skilled in the art and it is intended to cover in the appended claims all such modification and equivalents and follow in the true spirit and scope of this invention.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT:
(A) NAME: IMMULOGIC PHARMACEUTICAL CORPORATION
(B) STREET: 610 Lincoln Street
(C) CITY: Waltham (D) STATE: MA
(E) COUNTRY: USA
(F) POSTAL CODE (ZIP) : 02154
(G) TELEPHONE: (617) 466-6000 (H) TELEFAX: (617)466-6040
(ii) TITLE OF INVENTION: Allergenic Proteins and Peptides From
Japanese Cedar Pollen
(iii) NUMBER OF SEQUENCES: 55
(iv) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS (D) SOFTWARE: ASCII (TEXT)
(v) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE:
(vi) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE: (viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Vanstone, Darlene
(B) REGISTRATION NUMBER: 35,729
(C) REFERENCE/DOCKET NUMBER: IPC-033PC (ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (617) 466-6000
(B) TELEFAX: (617) 466-6040
(2) INFORMATION FOR SEQ ID NO:l;
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1726 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS : single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 42..1586 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:l: TGAGTTCGAG ACAAGTATAG AAAGAATTTT CTTTTATTAA A ATG GCC ATG AAA 53
Met Ala Met Lys
1 TTA ATT GCT CCA ATG GCC TTT CTG GCC ATG CAA TTG ATT ATA ATG GCG 101
Leu lie Ala Pro Met Ala Phe Leu Ala Met Gin Leu He He Met Ala 5 10 15 20 GCA GCA GAA GAT CAA TCT GCC CAA ATT ATG TTG GAC AGT GTT GTC GAA 149
Ala Ala Glu Asp Gin Ser Ala Gin He Met Leu Asp Ser Val Val Glu 25 30 35 AAA TAT CTT AGA TCG AAT CGG AGT TTA AGA AAA GTT GAG CAT TCT CGT 197
Lys Tyr Leu Arg Ser Asn Arg Ser Leu Arg Lys Val Glu His Ser Arg 40 45 50 CAT GAT GCT ATC AAC ATC TTC AAT GTG GAA AAG TAT GGC GCA GTA GGC 245
His Asp Ala He Asn He Phe Asn Val Glu Lys Tyr Gly Ala Val Gly 55 60 65 GAT GGA AAG CAT GAT TGC ACT GAG GCA TTT TCA ACA GCA TGG CAA GCT 293
Asp Gly Lys His Asp Cys Thr Glu Ala Phe Ser Thr Ala Trp Gin Ala 70 75 80 GCA TGC AAA AAC CCA TCA GCA ATG TTG CTT GTG CCA GGC AGC AAG AAA 341
Ala Cys Lys Asn Pro Ser Ala Met Leu Leu Val Pro Gly Ser Lys Lys 85 90 95 100 TTT GTT GTA AAC AAT CTG TTC TTC AAT GGG CCA TGT CAA CCT CAC TTT 389
Phe Val Val Asn Asn Leu Phe Phe Asn Gly Pro Cys Gin Pro His Phe 105 110 115 ACT TTT AAG GTA GAT GGG ATA ATA GCT GCG TAC CAA AAT CCA GCG AGC 437
Thr Phe Lys Val Asp Gly He He Ala Ala Tyr Gin Asn Pro Ala Ser 120 125 130 TGG AAG AAT AAT AGA ATA TGG TTG CAG TTT GCT AAA CTT ACA GGT TTT 485
Trp Lys Asn Asn Arg He Trp Leu Gin Phe Ala Lys Leu Thr Gly Phe
135 140 145 ACT CTA ATG GGT AAA GGT GTA ATT GAT GGG CAA GGA AAA CAA TGG TGG 533
Thr Leu Met Gly Lys Gly Val He Asp Gly Gin Gly Lys Gin Trp Trp 150 155 160
GCT GGC CAA TGT AAA TGG GTC AAT GGA CGA GAA ATT TGC AAC GAT CGT
581
Ala Gly Gin Cys Lys Trp Val Asn Gly Arg Glu He Cys Asn Asp Arg
165 170 175 180
GAT AGA CCA ACA GCC ATT AAA TTC GAT TTT TCC ACG GGT CTG ATA ATC 629
Asp Arg Pro Thr Ala He Lys Phe Asp Phe Ser Thr Gly Leu He He 185 190 195 CAA GGA CTG AAA CTA ATG AAC AGT CCC GAA TTT CAT TTA GTT TTT GGG 677
Gin Gly Leu Lys Leu Met Asn Ser Pro Glu Phe His Leu Val Phe Gly
200 205 210 AAT TGT GAG GGA GTA AAA ATC ATC GGC ATT AGT ATT ACG GCA CCG AGA 725
Asn Cys Glu Gly Val Lys He He Gly He Ser He Thr Ala Pro Arg
215 220 225 GAC AGT CCT AAC ACT GAT GGA ATT GAT ATC TTT GCA TCT AAA AAC TTT 773
Asp Ser Pro Asn Thr Asp Gly He Asp He Phe Ala Ser Lys Asn Phe 230 235 240 CAC TTA CAA AAG AAC ACG ATA GGA ACA GGG GAT GAC TGC GTC GCT ATA 821
His Leu Gin Lys Asn Thr He Gly Thr Gly Asp Asp Cys Val Ala He 245 250 255 260 GGC ACA GGG TCT TCT AAT ATT GTG ATT GAG GAT CTG ATT TGC GGT CCA 869
Gly Thr Gly Ser Ser Asn He Val He Glu Asp Leu He Cys Gly Pro 265 270 275 GGC CAT GGA ATA AGT ATA GGA AGT CTT GGG AGG GAA AAC TCT AGA GCA 917
Gly His Gly He Ser He Gly Ser Leu Gly Arg Glu Asn Ser Arg Ala
280 285 290 GAG GTT TCA TAC GTG CAC GTA AAT GGG GCT AAA TTC ATA GAC ACA CAA 965
Glu Val Ser Tyr Val His Val Asn Gly Ala Lys Phe He Asp Thr Gin 295 300 305 AAT GGA TTA AGA ATC AAA ACA TGG CAG GGT GGT TCA GGC ATG GCA AGC 1013
Asn Gly Leu Arg He Lys Thr Trp Gin Gly Gly Ser Gly Met Ala Ser 310 315 320 CAT ATA ATT TAT GAG AAT GTT GAA ATG ATA AAT TCG GAG AAC CCC ATA 1061
His He He Tyr Glu Asn Val Glu Met He Asn Ser Glu Asn Pro He 325 330 335 340 TTA ATA AAT CAA TTC TAC TGC ACT TCA GCT TCT GCT TGC CAA AAC CAG 1109
Leu He Asn Gin Phe Tyr Cys Thr Ser Ala Ser Ala Cys Gin Asn Gin 345 350 355 AGG TCT GCG GTT CAA ATC CAA GAT GTG ACA TAC AAG AAC ATA CGT GGG 1157
Arg Ser Ala Val Gin He Gin Asp Val Thr Tyr Lys Asn He Arg Gly 360 365 370 ACA TCA GCA ACA GCA GCA GCA ATT CAA CTT AAG TGC AGT GAC AGT ATG 1205
Thr Ser Ala Thr Ala Ala Ala He Gin Leu Lys Cys Ser Asp Ser Met 375 380 385 CCC TGC AAA GAT ATA AAG CTA AGT GAT ATA TCT TTG AAG CTT ACC TCA 1253 Pro Cys Lys Asp He Lys Leu Ser Asp He Ser Leu Lys Leu Thr Ser 390 395 400
GGG AAA ATT GCT TCC TGC CTT AAT GAT AAT GCA AAT GGA TAT TTC AGT 1301
Gly Lys He Ala Ser Cys Leu Asn Asp Asn Ala Asn Gly Tyr Phe Ser 405 410 415 420
GGA CAC GTC ATC CCT GCA TGC AAG AAT TTA AGT CCA AGT GCT AAG CGA 1349
Gly His Val He Pro Ala Cys Lys Asn Leu Ser Pro Ser Ala Lys Arg
425 430 435
AAA GAA TCT AAA TCC CAT AAA CAC CCA AAA ACT GTA ATG GTT GAA AAT 1397
Lys Glu Ser Lys Ser His Lys His Pro Lys Thr Val Met Val Glu Asn
440 445 450
ATG CGA GCA TAT GAC AAG GGT AAC AGA ACA CGC ATA TTG TTG GGG TCG 1445
Met Arg Ala Tyr Asp Lys Gly Asn Arg Thr Arg He Leu Leu Gly Ser 455 460 465
AGG CCT CCG AAT TGT ACA AAC AAA TGT CAT GGT TGC AGT CCA TGT AAG 1493
Arg Pro Pro Asn Cys Thr Asn Lys Cys His Gly Cys Ser Pro Cys Lys 470 475 480
GCC AAG TTA GTT ATT GTT CAT CGT ATT ATG CCG CAG GAG TAT TAT CCT 1541
Ala Lys Leu Val He Val His Arg He Met Pro Gin Glu Tyr Tyr Pro 485 490 495 500
CAG AGG TGG ATA TGC AGC TGT CAT GGC AAA ATC TAC CAT CCA TAATGAGATA 1593
Gin Arg Trp He Cys Ser Cys His Gly Lys He Tyr His Pro 505 510
CATTGAAACT GTATGTGCTA GTGAATATTC TTGTGGTACA ATATTAGAAC TGATATTGAA 1653
AATAAATCAT CAATGTTTCT AAGGCATTTA TAATAGATTA TATTAATGGT TCAGCCTGGT 1713 GCAAAAAAAA AAA 1726
(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 514 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
Met Ala Met Lys Leu He Ala Pro Met Ala Phe Leu Ala Met Gin Leu 1 5 10 15
He He Met Ala Ala Ala Glu Asp Gin Ser Ala Gin He Met Leu Asp 20 25 30 Ser Val Val Glu Lys Tyr Leu Arg Ser Asn Arg Ser Leu Arg Lys Val * 35 40 45
Glu His Ser Arg His Asp Ala He Asn He Phe Asn Val Glu Lys Tyr 50 55 60
Gly Ala Val Gly Asp Gly Lys His Asp Cys Thr Glu Ala Phe Ser Thr 65 70 75 80 Ala Trp Gin Ala Ala Cys Lys Asn Pro Ser Ala Met Leu Leu Val Pro
85 90 95
Gly Ser Lys Lys Phe Val Val Asn Asn Leu Phe Phe Asn Gly Pro Cys 100 105 110
Gin Pro His Phe Thr Phe Lys Val Asp Gly He He Ala Ala Tyr Gin 115 120 125
Asn Pro Ala Ser" Trp Lys Asn Asn Arg He Trp Leu Gin Phe Ala Lys 130 135 140
Leu Thr Gly Phe Thr Leu Met Gly Lys Gly Val He Asp Gly Gin Gly
145 150 155 160 Lys Gin Trp Trp Ala Gly Gin Cys Lys Trp Val Asn Gly Arg Glu He
165 170 175
Cys Asn Asp Arg Asp Arg Pro Thr Ala He Lys Phe Asp Phe Ser Thr 180 185 190
Gly Leu He He Gin Gly Leu Lys Leu Met Asn Ser Pro Glu Phe His 195 200 205
Leu Val Phe Gly Asn Cys Glu Gly Val Lys He He Gly He Ser He 210 215 220
Thr Ala Pro Arg Asp Ser Pro Asn Thr Asp Gly He Asp He Phe Ala 225 230 235 240
Ser Lys Asn Phe His Leu Gin Lys Asn Thr He Gly Thr Gly Asp Asp 245 250 255
Cys Val Ala He Gly Thr Gly Ser Ser Asn He Val He Glu Asp Leu 260 265 270
He Cys Gly Pro Gly His Gly He Ser He Gly Ser Leu Gly Arg Glu 275 280 285 Asn Ser Arg Ala Glu Val Ser Tyr Val His Val Asn Gly Ala Lys Phe 290 295 300
He Asp Thr Gin Asn Gly Leu Arg He Lys Thr Trp Gin Gly Gly Ser 305 310 315 320
Gly Met Ala Ser His He He Tyr Glu Asn Val Glu Met He Asn Ser 325 330 335
Glu Asn Pro He Leu He Asn Gin Phe Tyr Cys Thr Ser Ala Ser Ala 340 345 350
Cys Gin Asn Gin Arg Ser Ala Val Gin He Gin Asp Val Thr Tyr Lys 355 360 365 Asn He Arg Gly Thr Ser Ala Thr Ala Ala Ala He Gin Leu Lys Cys 370 375 380 Ser Asp Ser Met Pro Cys Lys Asp He Lys Leu Ser Asp He Ser Leu 385 390 395 400
Lys Leu Thr Ser Gly Lys He Ala Ser Cys Leu Asn Asp Asn Ala Asn 405 410 415
Gly Tyr Phe Ser Gly His Val He Pro Ala Cys Lys Asn Leu Ser Pro 420 425 430
Ser Ala Lys Arg Lys Glu Ser Lys Ser His Lys His Pro Lys Thr Val 435 440 445
Met Val Glu Asn Met Arg Ala Tyr Asp Lys Gly Asn Arg Thr Arg He 450 455 460
Leu Leu Gly Ser Arg Pro Pro Asn Cys Thr Asn Lys Cys His Gly Cys 465 470 475 480
Ser Pro Cys Lys Ala Lys Leu Val He Val His Arg He Met Pro Gin 485 490 495
Glu Tyr Tyr Pro Gin Arg Trp He Cys Ser Cys His Gly Lys He Tyr 500 505 510
His Pro
(2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 45 amino acids (B) TYPE: amino acid
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
Arg Lys Val Glu His Ser Arg His Asp Ala He Asn He Phe Asn Val 1 5 10 15
Glu Lys Tyr Gly Ala Val Gly Asp Gly Lys His Asp Cys Thr Glu Ala 20 25 30
Phe Ser Thr Ala Trp Gin Ala Ala Cys Lys Asn Pro Ser 35 40 45 (2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 41 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(v) FRAGMENT TYPE: internal (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:
Arg Lys Val Glu His Ser Arg His Asp Ala He Asn He Phe Asn Val 1 5 10 15
Glu Lys Tyr Gly Ala Val Gly Asp Gly Lys His Asp Cys Thr Glu Ala 20 25 30 Phe Ser Thr Ala Trp Gin Lys Asn Pro
35 40
(2) INFORMATION FOR SEQ ID NO:5: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 36 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide
(v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:5:
Ser Arg His Asp Ala He Asn He Phe Asn Val Glu Lys Tyr Gly Ala 1 5 10 15
Val Gly Asp Gly Lys His Asp Cys Thr Glu Ala Phe Ser Thr Ala Trp 20 25 30
Gin Lys Asn Pro 35
(2) INFORMATION FOR SEQ ID NO:6:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 10 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:6:
Ala He Asn He Phe Asn Val Glu Lys Tyr 1 5 10
(2) INFORMATION FOR SEQ ID NO:7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1410 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7:
AGAAAAGTTG AGCATTCTCG TCATGATGCT ATCAACATCT TCAATGTGGA AAAGTATGGC 60
GCAGTAGGCG ATGGAAAGCA TGATTGCACT GAGGCATTTT CAACAGCATG GCAAGCTGCA 120
TGCAAAAACC CATCAGCAAT GTTGCTTGTG CCAGGCAGCA AGAAATTTGT TGTAAACAAT 180
CTGTTCTTCA ATGGGCCATG TCAACCTCAC TTTACTTTTA AGGTAGATGG GATAATAGCT 240 GCGTACCAAA ATCCAGCGAG CTGGAAGAAT AATAGAATAT GGTTGCAGTT TGCTAAACTT 300
ACAGGTTTTA CTCTAATGGG TAAAGGTGTA ATTGATGGGC AAGGAAAACA ATGGTGGGCT 360
GGCCAATGTA AATGGGTCAA TGGACGAGAA ATTTGCAACG ATCGTGATAG ACCAACAGCC 420
ATTAAATTCG ATTTTTCCAC GGGTCTGATA ATCCAAGGAC TGAAACTAAT GAACAGTCCC 480
GAATTTCATT TAGTTTTTGG GAATTGTGAG GGAGTAAAAA TCATCGGCAT TAGTATTACG 540 GCACCGAGAG ACAGTCCTAA CACTGATGGA ATTGATATCT TTGCATCTAA AAACTTTCAC 600
TTACAAAAGA ACACGATAGG AACAGGGGAT GACTGCGTCG CTATAGGCAC AGGGTCTTCT 660
AATATTGTGA TTGAGGATCT GATTTGCGGT CCAGGCCATG GAATAAGTAT AGGAAGTCTT 720
GGGAGGGAAA ACTCTAGAGC AGAGGTTTCA TACGTGCACG TAAATGGGGC TAAATTCATA 780
GACACACAAA ATGGATTAAG AATCAAAACA TGGCAGGGTG GTTCAGGCAT GGCAAGCCAT 840 ATAATTTATG AGAATGTTGA AATGATAAAT TCGGAGAACC CCATATTAAT AAATCAATTC 900
TACTGCACTT CAGCTTCTGC TTGCCAAAAC CAGAGGTCTG CGGTTCAAAT CCAAGATGTG 960
ACATACAAGA ACATACGTGG GACATCAGCA ACAGCAGCAG CAATTCAACT TAAGTGCAGT 1020
GACAGTATGC CCTGCAAAGA TATAAAGCTA AGTGATATAT CTTTGAAGCT TACCTCAGGG 1080
AAAATTGCTT CCTGCCTTAA TGATAATGCA AATGGATATT TCAGTGGACA CGTCATCCCT 1140 GCATGCAAGA ATTTAAGTCC AAGTGCTAAG CGAAAAGAAT CTAAATCCCA TAAACACCCA 1200
AAAACTGTAA TGGTTGAAAA TATGCGAGCA TATGACAAGG GTAACAGAAC ACGCATATTG 1260
TTGGGGTCGA GGCCTCCGAA TTGTACAAAC AAATGTCATG GTTGCAGTCC ATGTAAGGCC 1320
AAGTTAGTTA TTGTTCATCG TATTATGCCG CAGGAGTATT ATCCTCAGAG GTGGATATGC 1380
AGCTGTCATG GCAAAATCTA CCATCCATAA 1410
(2) INFORMATION FOR SEQ ID NO:8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1395 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:8: TCTCGTCATG ATGCTATCAA CATCTTCAAT GTGGAAAAGT ATGGCGCAGT AGGCGATGGA. 60
AAGCATGATT GCACTGAGGC ATTTTCAACA GCATGGCAAG CTGCATGCAA AAACCCATCA 120
GCAATGTTGC TTGTGCCAGG CAGCAAGAAA TTTGTTGTAA ACAATCTGTT CTTCAATGGG 180
CCATGTCAAC CTCACTTTAC TTTTAAGGTA GATGGGATAA TAGCTGCGTA CCAAAATCCA 240
GCGAGCTGGA AGAATAATAG AATATGGTTG CAGTTTGCTA AACTTACAGG TTTTACTCTA 300 ATGGGTAAAG GTGTAATTGA TGGGCAAGGA AAACAATGGT GGGCTGGCCA ATGTAAATGG 360
GTCAATGGAC GAGAAATTTG CAACGATCGT GATAGACCAA CAGCCATTAA ATTCGATTTT 420
TCCACGGGTC TGATAATCCA AGGACTGAAA CTAATGAACA GTCCCGAATT TCATTTAGTT 480
TTTGGGAATT GTGAGGGAGT AAAAATCATC GGCATTAGTA TTACGGCACC GAGAGACAGT 540
CCTAACACTG ATGGAATTGA TATCTTTGCA TCTAAAAACT TTCACTTACA AAAGAACACG 600 ATAGGAACAG GGGATGACTG CGTCGCTATA GGCACAGGGT CTTCTAATAT TGTGATTGAG 660
GATCTGATTT GCGGTCCAGG CCATGGAATA AGTATAGGAA GTCTTGGGAG GGAAAACTCT 720
AGAGCAGAGG TTTCATACGT GCACGTAAAT GGGGCTAAAT TCATAGACAC ACAAAATGGA 780
TTAAGAATCA AAACATGGCA GGGTGGTTCA GGCATGGCAA GCCATATAAT TTATGAGAAT 840 GTTGAAATGA TAAATTCGGA GAACCCCATA TTAATAAATC AATTCTACTG CACTTCAGCT * 900
TCTGCTTGCC AAAACCAGAG GTCTGCGGTT CAAATCCAAG ATGTGACATA CAAGAACATA 960
CGTGGGACAT CAGCAACAGC AGCAGCAATT CAACTTAAGT GCAGTGACAG TATGCCCTGC 1020 AAAGATATAA AGCTAAGTGA TATATCTTTG AAGCTTACCT CAGGGAAAAT TGCTTCCTGC 1080
CTTAATGATA ATGCAAATGG ATATTTCAGT GGACACGTCA TCCCTGCATG CAAGAATTTA 1140
AGTCCAAGTG CTAAGCGAAA AGAATCTAAA TCCCATAAAC ACCCAAAAAC TGTAATGGTT 1200
GAAAATATGC GAGCATATGA CAAGGGTAAC AGAACACGCA TATTGTTGGG GTCGAGGCCT 1260
CCGAATTGTA CAAACAAATG TCATGGTTGC AGTCCATGTA AGGCCAAGTT AGTTATTGTT 1320 CATCGTATTA TGCCGCAGGA GTATTATCCT CAGAGGTGGA TATGCAGCTG TCATGGCAAA 1380
ATCTACCATC CATAA 1395
(2) INFORMATION FOR SEQ ID NO:9:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 1479 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:9:
GAAGATCAAT CTGCCCAAAT TATGTTGGAC AGTGTTGTCG AAAAATATCT TAGATCGAAT 60
CGGAGTTTAA GAAAAGTTGA GCATTCTCGT CATGATGCTA TCAACATCTT CAATGTGGAA 120
AAGTATGGCG CAGTAGGCGA TGGAAAGCAT GATTGCACTG AGGCATTTTC AACAGCATGG 180 CAAGCTGCAT GCAAAAACCC ATCAGCAATG TTGCTTGTGC CAGGCAGCAA GAAATTTGTT 240
GTAAACAATC TGTTCTTCAA TGGGCCATGT CAACCTCACT TTACTTTTAA GGTAGATGGG 300
ATAATAGCTG CGTACCAAAA TCCAGCGAGC TGGAAGAATA ATAGAATATG GTTGCAGTTT 360
GCTAAACTTA CAGGTTTTAC TCTAATGGGT AAAGGTGTAA TTGATGGGCA AGGAAAACAA 420 TGGTGGGCTG GCCAATGTAA ATGGGTCAAT GGACGAGAAA TTTGCAACGA TCGTGATAGA* 480
CCAACAGCCA TTAAATTCGA TTTTTCCACG GGTCTGATAA TCCAAGGACT GAAACTAATG 540
AACAGTCCCG AATTTCATTT AGTTTTTGGG AATTGTGAGG GAGTAAAAAT CATCGGCATT 600 AGTATTACGG CACCGAGAGA CAGTCCTAAC ACTGATGGAA TTGATATCTT TGCATCTAAA 660
AACTTTCACT TACAAAAGAA CACGATAGGA ACAGGGGATG ACTGCGTCGC TATAGGCACA 720
GGGTCTTCTA ATATTGTGAT TGAGGATCTG ATTTGCGGTC CAGGCCATGG AATAAGTATA 780
GGAAGTCTTG GGAGGGAAAA CTCTAGAGCA GAGGTTTCAT ACGTGCACGT AAATGGGGCT 840
AAATTCATAG ACACACAAAA TGGATTAAGA ATCAAAACAT GGCAGGGTGG TTCAGGCATG 900 GCAAGCCATA TAATTTATGA GAATGTTGAA ATGATAAATT CGGAGAACCC CATATTAATA 960
AATCAATTCT ACTGCACTTC AGCTTCTGCT TGCCAAAACC AGAGGTCTGC GGTTCAAATC 1020
CAAGATGTGA CATACAAGAA CATACGTGGG ACATCAGCAA CAGCAGCAGC AATTCAACTT 1080
AAGTGCAGTG ACAGTATGCC CTGCAAAGAT ATAAAGCTAA GTGATATATC TTTGAAGCTT 1140
ACCTCAGGGA AAATTGCTTC CTGCCTTAAT GATAATGCAA ATGGATATTT CAGTGGACAC 1200 GTCATCCCTG CATGCAAGAA TTTAAGTCCA AGTGCTAAGC GAAAAGAATC TAAATCCCAT 1260
AAACACCCAA AAACTGTAAT GGTTGAAAAT ATGCGAGCAT ATGACAAGGG TAACAGAACA 1320
CGCATATTGT TGGGGTCGAG GCCTCCGAAT TGTACAAACA AATGTCATGG TTGCAGTCCA 1380
TGTAAGGCCA AGTTAGTTAT TGTTCATCGT ATTATGCCGC AGGAGTATTA TCCTCAGAGG 1440
TGGATATGCA GCTGTCATGG CAAAATCTAC CATCCATAA 1479
(2) INFORMATION FOR SEQ ID NO:10:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 35 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:10:
GGGTCTAGAG GTACCGTCCG TCCGATCGAT CCATT 35
(2) INFORMATION FOR SEQ ID NO:11:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 13 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:11: AATGATCGAT GCT 13
(2) INFORMATION FOR SEQ ID NO:12: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 18 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:12:
RTAYTTYTCN ACRTTRAA 18 (2) INFORMATION FOR SEQ ID NO:13:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 13 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:13:
GGGTCTAGAG GTA 13
(2) INFORMATION FOR SEQ ID NO:14:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 6 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:14:
Phe Asn Val Glu Lys Tyr 1 5
(2) INFORMATION FOR SEQ ID NO:15:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 27 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:15; CCTGCAGTAY TTYTCNACRT TRAANAT 27
(2) INFORMATION FOR SEQ ID NO:16: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 7 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:16:
CCTGCAG
7
(2) INFORMATION FOR SEQ ID NO:17:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 7 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:17:
He Phe Asn Val Glu Lys Tyr
1 5 (2) INFORMATION FOR SEQ ID NO:18: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 27 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:18:
CCTGCAGTAY TTYTCNACRT TRAADAT 27
(2) INFORMATION FOR SEQ ID NO:19:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 17 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:19: GCNATHAAYA THTTYAA 17
(2) INFORMATION FOR SEQ ID NO:20:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 6 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:20: Ala He Asn He Phe Asn
1 5
(2) INFORMATION FOR SEQ ID NO:21: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 28 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:21: GGAATTCCGC NATHAAYATH TTYAAYGT 28
(2) INFORMATION FOR SEQ ID NO:22:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 8 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:22:
GGAATTCC 8
(2) INFORMATION FOR SEQ ID NO:23: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 7 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide
(v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:23:
Ala He Asn He Phe Asn Val
1 5
(2) INFORMATION FOR SEQ ID NO:24:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 20 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:24: GCYTCNGTRC ARTCRTGYTT 20
(2) INFORMATION FOR SEQ ID NO:25: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 7 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:25:
Lys His Asp Cys Thr Glu Ala 1 5
(2) INFORMATION FOR SEQ ID NO:26:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 28 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:26:
GGCTGCAGGT RCARTCRTGY TTNCCRTC 28
(2) INFORMATION FOR SEQ ID NO:27:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 8 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:27:
GGCTGCAG 8
(2) INFORMATION FOR SEQ ID NO:28:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 7 amino acids (B) TYPE: amino acid
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:28:
Asp Gly Lys His Asp Cys Thr 1 5
(2) INFORMATION FOR SEQ ID NO:29: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:29:
ATGTTGGACA GTGTTGTCGA A 21
(2) INFORMATION FOR SEQ ID NO:30:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 29 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:30:
GGGAATTCAG AAAAGTTGAG CATTCTCGT 29
(2) INFORMATION FOR SEQ ID NO:31:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 8 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:31:
GGGAATTC 8
(2) INFORMATION FOR SEQ ID NO:32:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:32: GTTCTTCAAT GGGCCATGT 19 (2) INFORMATION FOR SEQ ID NO:33:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:33:
GTGTTAGGAC TGTCTCTCGG 20
(2) INFORMATION FOR SEQ ID NO:34:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 20 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:34:
TGTCCAGGCC ATGGAATAAG 20
(2) INFORMATION FOR SEQ ID NO:35:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 20 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:35:
GCCTTACATG GACTGCAACC 20
(2) INFORMATION FOR SEQ ID NO:36:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:36:
TCCACGGGTC TGATAATCCA 20 (2) INFORMATION FOR SEQ ID NO:37:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:37:
AGGCAGGAAG CAATTTTCCC 20
(2) INFORMATION FOR SEQ ID NO:38:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:38:
TACTGCACTT CAGCTTCTGC 20
(2) INFORMATION FOR SEQ ID NO:39:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 20 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:39: GGGGGTCTCC GAATTTATCA 20
(2) INFORMATION FOR SEQ ID NO:40: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:40:
GGATATTTCA GTGGACACGT 20
(2) INFORMATION FOR SEQ ID NO:41:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 20 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:41: TATTAGAAGA CCCTGCGCCT 20
(2) INFORMATION FOR SEQ ID NO:42: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:42
CCATGTAAGG CCAAGTTAGT 20 (2) INFORMATION FOR SEQ ID NO:43:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:43:
ACACCTTTAC CCATTAGAGT 20 (2) INFORMATION FOR SEQ ID NO:44:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:44:
CTGTCCAACA TAATTTGGGC 20
(2) INFORMATION FOR SEQ ID NO:45:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:45
CATGGCAGGG TGGTTCAGGC 20
(2) INFORMATION FOR SEQ ID NO:46:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:46;
TAGCCCCATT TACGTGCACG 20
(2) INFORMATION FOR SEQ ID NO:47:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:47: TTGGGGTCGA GGCCTCCGAA 20
(2) INFORMATION FOR SEQ ID NO:48: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 9 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:48:
TAAAAUGGC 9 (2) INFORMATION FOR SEQ ID NO:49:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 9 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:49:
AACAAUGGC 9
(2) INFORMATION FOR SEQ ID NO:50:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 27 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:50:
GCCGAATTCA TGGCCATGAA ATTAATT 27
(2) INFORMATION FOR SEQ ID NO:51:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 9 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:51:
GCCGAATTC 9
(2) INFORMATION FOR SEQ ID NO:52: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 27 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO.-52:
CGGGGATCCT CATTATGGAT GGTAGAT 27
(2) INFORMATION FOR SEQ ID NO:53:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 9 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:53:
CGGGGATCC 9
(2) INFORMATION FOR SEQ ID NO:54:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 13 amino acids (B) TYPE: amino acid
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:54 : Phe Thr Phe Lys Val Asp Gly He He Ala Ala Tyr Gin 1 5 10
(2) INFORMATION FOR SEQ ID NO:55:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 14 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:55: Asn Gly Tyr Phe Ser Gly His Val He Pro Ala Cys Lys Asn
1 5 10

Claims (1)

  1. Claims:
    1. A nucleic acid having a nucleotide sequence coding for a Japanese Cedar pollen allergen Cryj II, or at least one antigenic fragment thereof, or the functional equivalent of said nucleotide sequence.
    2. A nucleic acid of claim 1 wherein said nucleotide sequence consists essentially of at least one fragment of the coding portion of the nucleotide sequence of Fig. 4 (SEQ ID NO: 1).
    3. A nucleic acid of claim 2 wherein said fragment comprises bases 108 through
    1586 (SEQ ID NO: 9) of the nucleotide sequence of Fig. 4 (SEQ ID NO: 1).
    4. A nucleic acid of claim 1 wherein said nucleotide sequence consists essentially of the nucleotide sequence of Fig. 4 (SEQ ID NO: 1).
    5. A nucleic acid of claim 1 wherein said fragment comprises bases selected from the group consisting of bases 177 through 1586 (SEQ ID NO: 7) of the nucleotide sequence of Fig. 4, and bases 192 through 1586 (SEQ ID NO: 8) of the nucleotide sequence of Fig. 4 (SEQ ID NO: 1).
    6. An expression vector comprising a nucleotide sequence coding for a Japanese cedar pollen allergen Cr y π, or at least one antigenic fragment thereof, or the functional equivalent of said nucleotide sequence.
    7. An expression vector of claim 6 wherein said nucleotide sequence consists essentially of at least one fragment of the coding portion of the nucleotide sequence of Fig. 4 (SEQ ID NO: 1).
    8. An expression vector of claim 6 wherein said nucleotide sequence comprises bases 108 through 1586 (SEQ ID NO: 9) of the nucleotide sequence of Fig.
    4.
    9. A host cell transformed to express a protein or peptide encoded by the nucleic acid of claim 1.
    10. Isolated Cryy II protein, or at least one antigenic fragment thereof, produced in a host cell transformed with the nucleic acid of claim 1. 11. An antigenic fragment of claim 10 which does not bind immunoglobulin E specific for a Japanese cedar pollen allergen, or if binding of said antigenic fragment to said immunoglobulin E occurs, such binding does not result in histamine release from mast cells or basophils.
    12. An antigenic fragment of claim 10 which binds immunoglobulin E to a substantially lesser extent than purified, native Cryj II protein binds said immunoglobulin E.
    13. Isolated Cryy II protein of claim 10 wherein the host cell is E.coli.
    14. A method of producing Cryj II protein, or at least one fragment thereof, comprising the steps of: a. culturing a host cell transformed with a DNA sequence encoding Cryj
    II protein or fragment thereof, in an appropriate medium to produce a mixture of cells and medium containing Cryy* II protein or at least one fragment thereof; and b. purifying said mixture to produce substantially pure Cr y II protein, or at least one fragment thereof.
    15. A protein preparation comprising Cryy II protein, or at least one fragment thereof, synthesized in a host cell transformed with a nucleic acid comprising a nucleotide sequence encoding all or a portion of Cryj II.
    16. A protein preparation of claim 15 wherein said at least one fragment of Cryy II is an antigenic fragment.
    17. A protein preparation comprising chemically synthesized Cryy II protein, or at least one fragment thereof.
    18. A protein preparation of claim 15 wherein said Cryy II protein comprises an amino acid sequence shown in Fig. 4 (SEQ ID NO: 2).
    19. A protein preparation of claim 17 wherein said Cryj II protein comprises an amino acid sequence shown in Fig. 4 (SEQ ID NO: 2). 20. An isolated peptide comprising at least one T cell epitope of Cryj II.
    21. An isolated peptide of claim 20 which as minimal immunoglobulin E stimulating activity.
    22. An isolated peptide of claim 20 which does not bind immunoglobulin E specific for a Japanese cedar pollen allergen, or if binding of the peptide to said immunoglobulin E occurs, such binding does not result in histamine release from mast cells or basophils.
    23. An isolated peptide of claim 20 which binds immunoglobulin E to a substantially lesser extent than purified native Cryy II protein binds said immunoglobulin E.
    24. Isolated Cr y II protein, or an antigenic fragment thereof, which modifies, in an individual sensitive to Japanese cedar pollen to whom it is administered, the allergic response of the individual to a Japanese cedar pollen allergen.
    25. Isolated Cry j II protein or antigenic fragment of claim 24 which modifies B- cell response of the individual to a Japanese cedar pollen allergen, T-cell response of the individual to a Japanese cedar pollen allergen, or both the B- cell response and the T-cell response of the individual to a Japanese cedar pollen allergen.
    26. Modified Cryy II protein or at least one modified fragment thereof, which when administered to an individual sensitive to Japanese cedar pollen, reduces the allergic response of the individual to Cr y II.
    27. A therapeutic composition comprising isolated Cryy II protein, or at least one fragment thereof, and a pharmaceutically acceptable carrier or diluent.
    28. A therapeutic composition of claim 27 wherein said Cry j II protein comprises an amino acid sequence shown in Fig. 4 (SEQ ID NO: 1).
    29. A method of treating sensitivity to a Japanese cedar pollen allergen, or an allergen immunologically cross-reactive with a Japanese cedar pollen allergen, in an individual sensitive to said allergen, comprising administering to the individual a therapeutically effective amount of the composition of claim 27.
    30. A method of detecting sensitivity in an individual to a Japanese cedar pollen allergen, comprising combining a blood sample obtained from the individual with isolated Cryy II protein, or antigenic fragment thereof, produced in a host cell transformed with the nucleic acid of claim 1 or chemically synthesized, under conditions appropriate for binding of blood components with the protein or fragment thereof, and determining the extent to which such binding occurs.
    31. A method of claim 30 wherein the extent to which binding occurs is determined by assessing T cell function, T cell proliferation, B cell function, binding of the protein or fragment thereof to antibodies present in the blood or a combination thereof.
    32. A monoclonal antibody, polyclonal antibody or immunoreactive fragment thereof, specifically reactive with Cryj II protein, or at least one antigenic fragment thereof.
    33. Cryy II protein isolated from Japanese cedar pollen, said protein having a molecular weight of about 40 kD as determined by sodium dodecyl sulfate- polyacrylamide gel electrophoresis. 34. A host cell transformed with a vector containing the cDNA insert of Cryj II, said host cell having ATCC deposit number 69105.
    35. A recombinant DNA molecule comprising a DNA coding for a polypeptide having at least one epitope of the protein allergen, Cryy II.
AU56681/94A 1992-11-12 1993-11-12 Allergenic proteins and peptides from japanese cedar pollen Abandoned AU5668194A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US97517992A 1992-11-12 1992-11-12
US975179 1992-11-12
PCT/US1993/011000 WO1994011512A2 (en) 1992-11-12 1993-11-12 Allergenic proteins and peptides from japanese cedar pollen

Publications (1)

Publication Number Publication Date
AU5668194A true AU5668194A (en) 1994-06-08

Family

ID=25522765

Family Applications (1)

Application Number Title Priority Date Filing Date
AU56681/94A Abandoned AU5668194A (en) 1992-11-12 1993-11-12 Allergenic proteins and peptides from japanese cedar pollen

Country Status (6)

Country Link
EP (1) EP0669983A1 (en)
JP (1) JPH08505284A (en)
AU (1) AU5668194A (en)
CA (1) CA2148713A1 (en)
NZ (1) NZ258701A (en)
WO (1) WO1994011512A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU748104B2 (en) * 1994-09-02 2002-05-30 Merck Patent Gmbh Peptide compositions capable of down regulating an antigen specific immune response

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4176820B2 (en) * 1993-11-05 2008-11-05 明治乳業株式会社 Cedar pollen allergen CryjII epitope
EP0655500B1 (en) * 1993-11-05 1999-05-12 Kabushiki Kaisha Hayashibara Seibutsu Kagaku Kenkyujo Allergenic polypeptide from Japanese cedar pollen and dna encoding it
JP3649460B2 (en) * 1993-11-05 2005-05-18 明治乳業株式会社 Japanese cedar pollen allergen Cry j II epitope
EP0783322A1 (en) * 1994-09-02 1997-07-16 Immunologic Pharmaceutical Corporation Peptide compositions capable of down regulating an antigen specific immune response
US6759234B1 (en) 1994-09-02 2004-07-06 Immulogic Pharmaceutical Corporation Compositions and methods for administering to humans, peptides capable of down regulating an antigen specific immune response
EP0700929A3 (en) * 1994-09-10 1999-09-01 Kabushiki Kaisha Hayashibara Seibutsu Kagaku Kenkyujo Peptide derived from cedar pollen allergens and uses thereof
US6719976B1 (en) * 1996-03-10 2004-04-13 Meiji Milk Products Co., Ltd. Peptide-based immunotherapeutic agent for treating allergic diseases
AU730198C (en) 1996-03-21 2002-03-14 Circassia Limited Cryptic peptides and method for their identification
JP4176750B2 (en) * 1996-06-14 2008-11-05 明治乳業株式会社 T cell epitope peptide
JP3734040B2 (en) 1996-06-14 2006-01-11 明治乳業株式会社 T cell epitope peptide
DE19957904A1 (en) * 1999-12-01 2001-06-07 Merck Patent Gmbh Insect venom allergens with reduced IgE reactivity and process for their preparation
WO2011052697A1 (en) 2009-10-30 2011-05-05 日本製紙株式会社 Protein having cedar pollen immunogenicity, polynucleotide for coding for said protein, and uses of said protein and polynucleotide
BR112014031327B1 (en) 2012-06-15 2022-02-01 Immunomic Therapeutics, Inc Nucleic acid molecules for the treatment of allergies

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2838800B2 (en) * 1989-09-02 1998-12-16 株式会社林原生物化学研究所 Desensitizer
CA2067182A1 (en) * 1990-08-13 1992-02-14 Rudolf Valenta Birch pollen allergen p14 for diagnosis and therapy of allergic diseases

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU748104B2 (en) * 1994-09-02 2002-05-30 Merck Patent Gmbh Peptide compositions capable of down regulating an antigen specific immune response

Also Published As

Publication number Publication date
WO1994011512A3 (en) 1994-07-07
WO1994011512A2 (en) 1994-05-26
CA2148713A1 (en) 1994-05-26
NZ258701A (en) 1996-11-26
JPH08505284A (en) 1996-06-11
EP0669983A1 (en) 1995-09-06

Similar Documents

Publication Publication Date Title
AU678412B2 (en) Allergenic proteins and peptides from Johnson grass pollen
WO1994001560A1 (en) Allergenic proteins and peptides from japanese cedar pollen
JP3575802B2 (en) Allergenic proteins and peptides from Japanese cedar pollen
AU5668194A (en) Allergenic proteins and peptides from japanese cedar pollen
WO1994011512A9 (en) Allergenic proteins and peptides from japanese cedar pollen
NZ263913A (en) Lol pi protein allergen from ryegrass and related peptides coding sequences, vectors, protein production and use
US5710126A (en) T cell epitopes of ryegrass pollen allergen
US8540999B2 (en) Allergenic proteins and peptides from Japanese cedar pollen
JP4102432B2 (en) Allergenic proteins and peptides derived from cedar pollen
WO1995006728A2 (en) T cell epitopes of ryegrass pollen allergen
AU4046293A (en) T cell epitopes of the major allergens from (ambrosia artemisiifolia)
US6090386A (en) T cell peptides of the CRX JII allergen
NO317850B1 (en) Isolated nucleic acid encoding the Der p III protein allergen, isolated Der p III protein allergen, pharmaceutical composition comprising the allergen, and its use for the preparation of pharmaceutical composition and methods for preparing such.
US6214358B1 (en) Protein allergens of the species Cynodon dactylon
WO1992016554A1 (en) Protein allergens of the species cynodon dactylon
US6441157B1 (en) Nucleic acid sequences encoding protein allergens of the species Cynodon dactylon
JP3618342B2 (en) Allergen of the protein of the species of Cynodon dactylon
AU3438493A (en) Allergenic proteins and peptides from japanese cedar pollen