AU778362B2 - Retinoid metabolizing protein - Google Patents

Retinoid metabolizing protein Download PDF

Info

Publication number
AU778362B2
AU778362B2 AU65483/01A AU6548301A AU778362B2 AU 778362 B2 AU778362 B2 AU 778362B2 AU 65483/01 A AU65483/01 A AU 65483/01A AU 6548301 A AU6548301 A AU 6548301A AU 778362 B2 AU778362 B2 AU 778362B2
Authority
AU
Australia
Prior art keywords
seq
protein
sequence
nucleic acid
retinoid
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.)
Ceased
Application number
AU65483/01A
Other versions
AU6548301A (en
Inventor
Barbara R. Beckett
Glenville Jones
P. Martin Petkovich
Jay A. White
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.)
Queens University at Kingston
Original Assignee
Queens University at Kingston
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
Priority claimed from AU31620/97A external-priority patent/AU3162097A/en
Application filed by Queens University at Kingston filed Critical Queens University at Kingston
Publication of AU6548301A publication Critical patent/AU6548301A/en
Application granted granted Critical
Publication of AU778362B2 publication Critical patent/AU778362B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Landscapes

  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)

Description

-1-
AUSTRALIA
PATENTS ACT 1990 COMPLETE SPECIFICATION FOR A STANDARD PATENT
ORIGINAL
a a a a a Name of Applicant: Queen's University At Kingston Actual Inventor: P. Martin Petkovich and Jay A. White and Barbara R. Beckett and Glenville Jones Address for Service: BALDWIN SHELSTON WATERS MARGARET STREET SYDNEY NSW 2000 CCN: 3710000352 Invention Title: 'RETINOID METABOLIZING PROTEIN' Details of Original Application No. 31620/97 dated 23 Jun 1997 The following statement is a full description of this invention, including the best method of performing it known to me/us:- File: 32944AUP00 la Retinoid Metabolizing Protein This application claims priority from U.S. patent application Serial No.
08/724,466, filed October 1, 1996, and from U.S. patent application Serial No. 08/667,546, filed June 21, 1996, the specifications of which applications are incorporated herein by reference.
Background of the Invention Vitamin A metabolism gives rise to several active forms of retinoic acid (RA) which are involved in regulating gene expression during development, regeneration, and in the growth and differentiation of epithelial tissues [Maden, 1992; Chambon, 1995; Mangelsdorf, 1995; Gudas, 1994; Lotan, 1995; Morriss-Kay, 1996]; has been linked apoptosis, or programmed cell death in a number of cell types; and to have anticarcinogenic and antitumoral properties [Lotan, 1996].
Early studies of retinol deficiency indicated a correlation between vitamin A depletion and a higher incidence of cancer and increased susceptability to chemical carcinogenesis [Chytil, 1984]. Several animal models have been used to demonstrate the effectiveness of retinoids in supressing carcinogenesis in a variety of tissues including skin, mammary epithelia, oral cavity, aerodigestive tract, liver, bladder and prostate [Moon, 1994].
These studies have led to the preventative use of retinoids to treat premalignant lesions Sincluding actinic keratosis and oral leukoplakia, as well as in the prevention of secondary tumours of the head and neck and the recurrence of non-small cell lung carcinomas, and 20 basal cell carcinomas [Hong, 1994; Lppman, 1995]. RA itself has been found to be useful therapeutically, notably in the treatment of cancers, including acute promyelocytic leukemia tumors of the head and neck, and skin cancer, as well as in the treatment of sKin disorders such as the premalignancy associated actinic keratoses, acne, psoriasis and ichthyosis. There is evidence that the effectiveness of RA as an anti-tumour agent is at least partially due to induction of cellular differentiation and/or inhibition of proliferation [Lotan, 1996].
Studies over the past several years indicate that a high proportion of patients with acute promyelocytic leukemia (APL) achieve complete remission after a short period of treatment S with all-trans RA. Unfortunately, this high rate of remission is in most cases brief. Following relapse, patients are clinically resistant to further treatment with RA [Warrell, 1994; Warrell, et 30 al., 1994; Chomienne, 1996; Muindi, 1992]. The nature of this resistance is unknown.
Interestingly, leukemic cells taken from patients exhibiting clinical resistance to RA have been shown to be sensitive to the differentiating action of RA when grown in vitro (Muindi, 1992; Muindi, 1994). This suggests that pharmacokinetic mechanisms may accoient for the acquired resistance to RA. This possibility is supported by studies showing that peak plasma concentrations of RA were much higher in patients after initial administration than in patients treated following relapse. This decrease in peak plasma RA concentration was accompanied by a 10-fold increase in urinary 4-oxo-retinoic acid concentration. In addition, ketoconazole, a broad spectrum inhibitor of cytochrome P450 function was shown to modulate RA pharmacokinetics in vivo [Muindi, 1992; Muindi, 1994]. It is therefore likely that RA increases the rate of its own metabolism which in turn results in the inability to sustain effective therapeutic doses of RA. Therapeutic administration of RA can result in a variety of undesirable side effects and it is therefore important to establish and maintain the minimal requisite doses of RA in treatment. For example, RA treatments during pregnancy can lead to severe teratogenic effects on the fetus. Adverse reactions to RA treatment also include headache, nausea, chelitis, facial dermatitis, conjunctivitis, and dryness of nasal mucosa. Prolonged exposure to RA can cause major elevations in serum triglycerides and can lead to severe abnormalities of liver function, including hepatomegaly, cirrhosis and portal hypertension.
RA metabolism may also account for the lack of response of certain tumors to RA treatment. For example, recent studies have shown that cytochrome P450 inhibitors that block RA metabolism, resulting in increased tissue levels of RA, may be useful therapeutic agents in the treatment of prostate cancer [Wouters, 1992; De Coster, 1996]. Thus RA metabolizing cytochrome P450s may be useful targets for the treatment of a number of different types of cancer.
The classical view of vitamin A metabolism holds that all trans-RA, the most active metabolite is derived from conversion of retinol to retinaldehyde to RA through two oxidation steps and that RA is further metabolized to the polar derivatives 4-OH RA and 4-oxo RA [Blaner, 1994; Napoli, 1995; Formelli, 1996; Napoli, 1996]. It is unknown whether the 4oxo- and 4-OH- metabolites are simply intermediates in the RA catabolic pathway or whether they can also have specific activities which differ from those of all-trans RA and 9-cis RA.
Pijnappel et al. [Pijnappel, 1993] have shown that, in Xenopus, 4-oxo-RA can efficiently modulate positional specification in early embryos and exhibits a more potent ability to regulate Hoxb-9 and Hoxb-4 gene expression than all-trans RA. 4-oxo-RA has been found to bind to retinoic acid receptor-p (RAR-p) with affinity comparable to all-trans RA [Pijnappel, 1993] but 25 poorly to RAR-y [Reddy, 1992], suggesting that this metabolite exhibits some receptor selectivity. 4-oxo-RA also binds to cellular retinoic acid binding protein (CRABP) but with an affinity slightly lower than that of all-trans RA [Fiorella, 1993]. Takatsuka et al. [Takatsuka, 1996] have shown that growth inhibitory effects of RA correlate with RA metabolic activity but it is unknown whether there is a causal relationship between production of RA metabolites and growth inhibition. The asymmetric distribution of these metabolites in developing embryos suggests that they may be preferentially sequestered or generated by tissue specific isomerases [Creech Kraft, 1994]. The normal balance of these metabolites is dependent upon rate of formation from metabolic precursors, retinol and retinaldehyde [Leo, 1989], and rate of catabolism. Lttle is presently known about the enzymes involved in this metabolic scheme, in particular the catabolism of RA.
The catabolism of RA is thought to be initiated by hydroxylation either at the C4-, or C18-position of the 1-ionone ring of RA [Napoli, 1996]. The C4- hydroxylation step is mediated by cytochrome P450 activity, as judged by the ability of broad spectrum P450 inhibitors such as ketocoazole and liarazole to block 4-hydroxylation [Williams, 1987, Van Wauwe, 1988; Van Wauwe, 1990, Van Wauwe, 1992, Wouters, 1992]. In certain tissues, including testis, skin and lung and in numerous cell lines, such as NIH3T3 fibroblasts, HL 60 myelomonocytic leukemic cells, F9 and P19 murine embryonal carcinoma cell lines and MCF7, RA metabolism can be induced by RA pretreatment [Frolik, 1979, Roberts, 1979a and b; Duell, 1992; Wouters, 1992].
Studies involving targetted disruption of RAR genes in F9 cells suggest that RAR-a and RAR-y isoforms may play a role in regulating the enzymes responsible for this increased metabolism [Boylan, 1995].
The glucuronidation of RA is a significant metabolic step in the inactivation of RA [Blaner, 1994; Formelli, 1996]. The elimination of RA may require oxidation to 4-oxo, followed by conjugation to form the 4-oxo all-trans RA glucuronide. This is supported by studies in both primates and humans showing that the 4-oxo RA glucuronide is the only retinoid conjugate found in urine [Muindi, 1992; Muindi, 1994]. The fact that following RA therapy, 4-oxo RA is not present or barely detectable in serum, suggests that oxidation may be the rate limiting step in this process.
It has recently been shown that 4-oxoretinol (4-oxo-ROL) can have greater biological activity than retinol. The 4-oxo-ROL is inducible by RA in F9 and P19 mouse teratocarcinoma cells [Blumberg et al., 1995; Achkar etal., 1996].
It is known that zebrafish fins regenerate through an RA sensitive process which utilizes many gene regulatory pathways involved in early vertebrate development [White, 1994; Akimenko, 1995a b].
20 As far as the inventors are aware, cytochrome P450s involved in the metabolism of RA in extrahepatic tissues remain uncharacterized at the molecular level.
SUMMARY OF THE INVENTION The present inventors are the first to identify, clone and sequence a gene (cDNA) encoding a retinoic acid-inducible, retinoic acid-metabolizing protein, including a cDNA which is RA-inducible in humans. The protein has been found to be expressed in epithelia.
A cDNA has been isolated from zebra fish and sequenced. A protein encoded by the cDNA has been expressed and shown to have the ability to hydroxylate retinoic acid at 4 position of the p-ionone ring of retinoic acid. The protein has been found to be inducible in epithelial cells exposed to retinoic acid.
A human cDNA encoding a protein with similar functionality has also been isolated and sequenced.
A cDNA has also been isolated from mouse and sequenced.
Homology between sequences from the three species, be they nucleic acids encoding the protein, or the amino acid sequences of the proteins, has been found to be relatively high and all three proteins contain a heme-binding motif characteristic of the group of proteins known as cytochrome P450s. The overall homology between the amino acid sequences of these newly obtained proteins and known cytochrome P450s is less than Notwithstanding this relatively low overall homology, a higher degree of homology has been observed in the heme binding region for certain other P450s. For example, homology between the approximately 20 amino acids defining respective heme binding regions of the new zebrafish protein and CYP4503A12 is about 50% and between the new zebrafish protein and hCYTFAOH is 65%. The homology between the heme binding region itself of a protein of the present invention and another P450 could well be 70%, 75%, 80%, 85%, 90%, 95% or even 100%.
A first aspect of the present invention is thus a purified protein having the ability to oxidize a retinoid, and having an amino acid sequence which is at least about 30% conserved in relation to the amino acid sequence identified as SEQ ID NO:2 or identified as SEQ ID NO:4 or identified as SEQ ID NO:32, or a functionally equivalent homolog thereof. The amino acid sequence identified as SEQ ID NO:2 is of the protein, termed here "zP450RAI", obtained from zebrafish. The amino acid sequence of the human protein is identified as SEQ ID NO:4 and the protein is referred to herein as "hP450RAI". The sequence of the mouse protein is identified as SEQ ID NO:32 and the protein is referred to herein as mP450RAI.
Such a protein which is at least about 35% conserved in relation to the amino acid sequence identified as SEQ ID NO:2 (zebrafish) or identified as SEQ ID NO:4 (human) or identified as SEQ ID NO:32 (mouse), or a functionally equivalent homolog thereof, also forms part of the invention disclosed herein. Likewise, the degree of sequence conservation of a protein could be 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or of course 100% of either SEQ ID NO:2 or SEQ ID NO:4 or SEQ ID NO:32 or a functionally equivalent homoiog thereof, variants being possible so lor as the ability of the native protein to oxidize a retinoid is retained. Also within the scope of the invention is any such protein which has the ability to hydroxylate retinoic acid at the 4 position of the p-ionone ring. Of course, conservatively substituted variants of proteins disclosed are within the scope of the present invention.
25 A retinoid oxidized by a protein of the present invention may be a retinoic acid or a retinol and the protein may have the ability to oxidize the carbon occupying the 4-position of the 1-ionone ring of the retinoid. In particular, all-trans retinoids may be metabolized by proteins of the present invention.
In the context of this specification, the term "conserved" describes similarity between sequences. The degree of conservation between two sequences can be determined oby optimally aligning the sequences for comparison, as is commonly known in the art, and S. comparing a position in the first sequence with a corresponding position in the second sequence. When the compared positions are occupied by the same nucleotide or amino acid, as the case may be, the two sequences are conserved at that position. The degree of conservation between two sequences is often expressed, as it is here, as a percentage representing the ratio of the number of matching positions in the two sequences to the total number of positions compared.
The generic term "retinoids" means a group of compounds which includes retinoic acid, vitamin A (retinol) and a series of natural and synthetic derivatives that can exert profound effects on development and differentiation in a wide variety of systems. For purposes of this disclosure "retinoid" is also intended to encompass an equivalent thereof having the same functional characteristics which may be produced, for example, by computational chemistry.
In another aspect, the present invention is an isolated nucleic aid molecule encoding a protein of the present invention.
The present invention thus includes an isolated nucleic acid molecule encoding a protein having an amino acid sequence which is at least about 30% conserved in relation to the amino acid sequence identified as SEQ ID NO:2 or identified as SEQ ID NO:4 or identified as SEQ ID NO:32, or a functionally equivalent homolog thereof, for example, or a nucleic acid strand capable of hybridizing with the nucleic acid molecule under stringent hybridization conditions. Of course, the degree of conservation of the protein which the nucleic acid encodes can be higher, that is, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 90%, 95% or more.
Particularly, the invention is an isolated nucleic acid molecule encoding a protein having the ability to oxidize a retinoid at the carbon occupying the 4-position of the Bionone ring of the retinoid ring, and more particularly, having all-trans retinoic acid 4hydroxylase activity. For the purposes of this invention, the term "isolated" refers to a nucleic acid that is substantially free of other cellular material or culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when produced by 20 chemical synthesis.
Cellular expression of preferred proteins of the present invention, preferred embodiments being described in more detail below, can for certain types of cells be induced by exposure of the cells to a retinoid, particularly, retinoic acid. A protein of the present invention, when described as being a "retinoic acid inducible protein", is a protein normally encoded by 25 DNA of a cell and whose expression by that cell can be induced by exposure of the cell to retinoic acid. It will be appreciated that not every cell, even if it contains DNA encoding such a protein, possesses all the attributes necessary to express the protein on exposure to RA.
Nucleotide sequences possessing promoter acivity associated with the .proteins have also been isolated and identified by the inventors. The human nucleotide sequence having promoter activity is identified as SEQ ID NO:33. The murine nucleotide 3 osequence having promoter activity is identified as SEQ ID NO:34. The zebrafish nucleotide sequence having promoter activity is identified as SEQ ID In fact, the invention includes genomic sequences for the human (SEQ ID Nos:36 and 37) and mouse (SEQ ID NO:38).
In another respect, the present invention is thus a microbial cell containing and expressing heterologous DNA encoding a retinoic acid inducible protein having all-trans retinoic acid 4-hydroxylase activity.
The sequence of a nucleic acid molecule of the present invention can correspond to a part of a human genome or of a fish genome or of a mouse genome, or vary therefrom due to the degeneracy of the genetic code. More particularly, a nucleic acid -6molecule of the present invention can be a DNA molecule having the sequence identified as SEQ ID NO:3 (zP450RAI) or SEQ ID NO:5 (hP450RAI), or SEQ ID NO:31 (mP450RAI), or the sequence can be one which varies from one of these sequences due to the degeneracy of the genetic code, or it can be a nucleic acid strand capable of hybridizing with at least one of these nucleic acid molecules under high or low stringency hybridization conditions.
"Stringent hybridization conditions" takes on its common meaning to a person skilled in the art here. Appropriate stringency conditions which promote nucleic acid hybridization, for example, 6x sodium chloride/sodium citrate (SSC) at about 45"C are known to those skilled in the art. The following examples are found in Current Protocols in Molecular Biology, John Wiley Sons, NY (1989), 6.3.1-6.3.6: For 50 ml of a first suitable hybridization solution, mix together 24 ml formamide, 12 ml 20x SSC, 0.5 ml 2 M Tris-HCI pH 7.6, 0.5 ml 100x Denhardt's solution, 2.5 ml deionized HO, 10 ml 50% dextran sulfate, and 0.5 ml SDS. A second suitable hybridization solution can be 1% crystalline BSA (fraction 1 mM EDTA, 0.5 M Na,HPO, pH 7.2, 7% SDS. The salt concentration in the wash step can be selected from a low stringency of about 2x SSC at 50C to a high stringency of about 0.2x SSC at 50"C. Both of these wash solutions may contain 0.1% SDS. In addition, the temperature in the wash step can be increased from low stringency conditions at room temperature, about 22°C, to high stringency conditions, at about 65C. The cited reference gives more detail, but appropriate wash stringency depends on degree of homology and length of probe. If homology 20 is 100%, a high temperature (65C to 75°C) may be used. If homology is low, lower wash temperatures must be used. However, if the probe is very short (<100bp), lower temperatures must be used even with 100% homology. In general, one starts washing at low temperatures (37*C to 40'C), and raises the temperature by 3-5C intervals until background is low enough not to be a major factor in autoradiography.
25 Another aspect of this invention is isolated mRNA transcribed from DNA having a sequence encoding a protein of the present invention.
In another aspect, the present invention is isolated DNA having a sequence according to a nucleotide sequence described above operatively linked in a recombinant cloning vector. In the context of this invention, the two-part term "operatively linked" means both that the regulatory sequence contains sufficient element(s) to allow expression of the nucleic acid in question and that the nucleic acid is linked to the regulatory sequence appropriately. For example, the nucleic acid of the invention is in the appropriate orientation and in phase with an initiation codon. The present invention thus includes a stably transfected cell line which expresses a protein having the ability to hydroxylate retinoic acid at the 4 position of the p-ionone ring of retinoic acid. The invention includes a culture of cells transformed with a recombinant DNA molecule having a nucleic acid sequence which encodes a protein having the ability to hydroxylate retinoic acid at the 4 position of the p-ionone ring of retinoic acid.
Another aspect of the present inventici is a host cell that has been engineered genetically to produce a protein of the invention described above, the cell having incorporated expressibly therein heterologous DNA encoding said protein. The cell may be selected such that production of the protein is inducible by exposing the cell to a retinoid, preferably, retinoic acid. The cell can be eukaryotic.
The present invention also includes a process for producing an abovedescribed protein of the invention. Such a process includes: preparing a DNA fragment containing a nucleotide sequence which encodes the protein; incorporating the DNA fragment into an expression vector to obtain a recombinant DNA molecule which contains the DNA fragment and is capable of undergoing replication; transforming a host cell with the recombinant DNA molecule to produce a transformant which can express the protein; culturing the transformant to produce the protein; and recovering the protein from resulting cultured mixture.
The present invention includes an antibody to a protein of the invention. Here, the term "antibody" is intended to include a Fab fragment and it can be a monoclonal antibody.
The antibody can be specifically raised to the amino acid sequence identified as SEQ ID NO:4, hP450RAI.
The present invention includes a purified protein for use in metabolizing retinoic acid in an organism or cell in need of such metabolizing. Likewise, the invention includes a method for metabolizing retinoic acid in an organism or cell in need of retinoic acid metabolizing wherein the method includes administering a protein of the invention as described 20 above.
The invention includes a method for inhibiting retinoic acid hydroxylation in an organism in need of such inhibition, comprising introducing into cells of the organism an effective amount of an antisense RNA or oligonucleotide substantially complementary to at least a portion of the sequence identified as SEQ ID NO:5. The organism can be human and/or the organism can be in need of treatment against a cancerous disease or a disease selected from the group consisting of cancer, actinic keratosis, oral leukoplakia, a secondary tumor of the head and/or neck, a non-small cell lung carcinoma, a basal cell carcinoma, acute promyelocytic leukemia, skin cancer, and a premalignancy associated actinic keratosis, acne, psoriasis and/or ichthyosis. Such a method can include use of at least one delivery vehicle or technique selected from the set of viral vectors, microinjection, electroporation, coprecipitation, liposomes, aerosol delivery and lavage. The portion of the sequence may be 5 bases in length, between 5 and 50 bases in length, 5 and 30 bases in length between 10 and 20 bases in length, or another suitable length may be found. The organism may be a human patient and the method can include treating the patient against a cancerous disease.
The invention also includes a method of inhibiting retinoic acid hydroxylation in an organism in need of such inhibition by administering to the organism an effective amount of an antibody, such antibodies being described above. A particularly useful antibody for the treatment of a human would be an antibody to the protein having the amino acid sequence identified as SEQ ID NO:4, or a portion thereof. It would be advantageous to adapt such an antibody for administration to a human by "humanizing" the antibody, as is understood by those skilled in the art [Hozumi, 1993].
The invention includes a method for producing a desired protein, comprising providing a cell which can produce an endogenous protein in response to exposure to a retinoid; incorporating into DNA of the cell a DNA sequence encoding the desired protein at or near a site which is normally occupied by a DNA sequence encoding the endogenous protein; and exposing the cell to the retinoid so as to induce production of the desired protein.
In another embodiment, the present invention is a kit for determining the presence of a protein having the ability to oxidize a retinoid, and having an amino acid sequence which is at least about 30% conserved in relation to the amino acid sequence identified as SEQ ID NO:2 or identified as SEQ ID NO:32, or more likely for determining the presence of a protein (or peptide fragment thereof) having an amino acid sequence identified as SEQ ID NO:4, the human protein. The kit includes an antibody to the protein linked to a reporter system wherein the reporter system produces a detectable response when a predetermined amount of the protein and the antibody are bound together.
In another aspect, the present invention is a kit for determining the presence of a nucleic acid encoding a protein of invention, or having the sequence identified as SEQ ID NO:3 or SEQ ID NO:5 or SEQ ID NO:31, or which varies from the sequence due to the degeneracy of the genetic code, or a nucleic acid strand capable of hybridizing with at least one 20 said nucleic acid under stringent hybridization conditions. The kit includes a nucleic acid molecule capable of hybridizing with at least a portion of a said nucleic acid or nucleic acid S: strand under stringent conditions in which the nucleic acid molecule is linked to a reporter *system wherein the reporter system produces a detectable response when a predetermined amount of the nucleic acid or nucleic acid strand and nucleic acid molecule are hybridized with 25 each other. The-molecule can be 5 bases in length or longer; between 5 and 50 bases in length, between 5 and 30 or 40 bases in length, or between 10 and 20 bases in length. Of course it might be possible to find a more suitable base length.
The present invention includes an isolated DNA molecule having a nucleotide sequence selected from the group consisting of: SEQ ID NO:33; SEQ ID NO:34; SEQ ID NO:35; and a fragment of or wherein the DNA molecule possesses promoter activity.
In a more particular aspect, the invetio-h includes a DNA molecule of one of the indicated sequences in which the DNA molecule includes the sequence TGAACT(N),TGAACT, wherein x has a value of up to 5. This sequence is conserved in all three of the nucleotide sequences identified as having promoter activity, particularly where x is 5. Even more particularly, the invention includes such a DNA molecule in which the sequence TCTGASSAAGKTAAC occurs downstream from the sequence TGAACT(N),TGAACT. Even more particularly, the sequence includes the sequence AATT between the sequence TGAACT(N),TGAACT and the sequence TCTGASSAAGKTAAC, the AATT having been found immediately downstream of the sequence TGAACT(N),TGAACT. It has been observed that there can be up to six nucleotides between the sequence TGAACT(N),TGAACT and the sequence TCTGASSAAGKTAAC. There can also be the sequence CAATTAAAGA upstream of the sequence TGAACT(N),TGAACT. In a particular aspect, the nucleotide sequence having promoter activity includes the sequence CAATTAAAGATGAACTTTGGGTGAACTAATT and the sequence TATAA. Particularly, the sequence TATAA is downstream of the sequence TGAACT(N),TGAACT, and more particularly, the sequence TATAA is downstream of the sequence TCTGASSAAGKTAAC and it can be spaced up to about 55 nucleotides downstream from the sequence TGAACT(N),TGAACT.
The invention includes a recombinant DNA having such a DNA sequence having promoter activity. The recombinant DNA can also include one or more structural genes.
The structural gene(s) can encode cytochrome P450 protein(s). The invention includes an expression plasmid including such recombinant DNA and an isolated cell containing the recombinant DNA. The cell is usually eukaryotic.
The invention includes a process for the production of recombinant protein(s), including steps of culturing a cell according to the invention containing recombinant DNA and Srecovering the protein(s) produced. The protein(s) can be a cytochrome P450, but is not 20 necessarily so. The protein(s) can be a fusion protein.
The invention further includes a method of screening drugs for their effect on :activity of a retinoic acid inducible protein, comprising exposing a purified said protein to a said drug and determining the effect on the activity. The activity can be hydroxylation of a retinoic acid, particularly all-trans retinoic acid. In particular, the activity can be hydroxylation of a 25 retinoic acid, particularly all trans-retinoic acid, at the 4 position of the p-ionone ring thereof.
The invention provides a method of screening drugs for their effect on activity of any protein having P450RAI activity of the present invention, particularly ascertaining the effect a potential drug has on the oxidation of retinoic acid, specifically all-trans retinoic acid.
S In yet another aspect, the invention includes a method of screening drugs for their effect on expression of a gene wherein the gene is inducible by a retinoid, including the step of exposing a recombinant DNA described above to a potential drug and determining the effect on gene expression. This can include exposing the recombinant DNA in the presence of a said retinoid. The gene can be a reporter gene which does not metabolize retinoic acid. The method can include ascertaining the effect on transcription of the gene.
The invention includes a method of screening a drug for its effect on the metabolism of a retinoid by a cytochrome P450 encoded by a nucleotide sequence of the invention which is incorporated into an expression system so as to be under control of a nucleotide sequence having native promoter activity for a said cytochrome P450, comprising: exposing the system to the drug in the presence of the retinoid so as to determine the effect of the drug on metabolism of the retinoid.
The retinoid can be retinoic acid and it can be all trans-retinoic acid.
The invention includes any drug identified according to such a method as having the effect of modulating, and preferably reducing, the activity of a said protein or as having the effect of modulating, and preferably reducing, gene expression.
The invention includes a method for inhibiting retinoic acid metabolism in an organism in need of such inhibition, comprising administering to the organism an effective amount of a drug identified according to a method of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 is a schematic representation of the steps used to isolate retinoidregulated genes using differential display of mRNA. Schematic representation of the steps involved in the isolation of retinoid-regulated genes using the differential display technique. The cloned products isolated in step 6 can then be used for sequencing, Northern blotting or screening of cDNA libraries. P1, P2 and P3 correspond to fragments from RA induced mRNAs. P4 represents a PCR product from an mRNA which is down-regulated.
Figure 2(a) shows a polyacrylamide gel of PCR amplified mRNA in duplicate obtained using retinoic acid-treated fish (lanes 1 and 2) and dimethyl sulfoxide (DMSO-) treated control fish (lanes 3 and The arrow indicates a PCR amplified band present in the RA- S*treated samples and not observed in the controls.
Figure 2(b) shows the nucleotide sequence (SEQ ID NO:1) of the 337 base 20 pair PCR product isolated from the band (arrow) of Figure The arrows indicate the nucleotide sequences where the upstream and downstream priming sites for differential display PCR amplification were located in the untranslated portion of zP450RAI.
Figure 2(c) shows an amino acid sequence (SEQ ID NO:2) corresponding to cDNA (492 amino acid open reading frame). The boxed residues indicate the heme-binding 25 motif characteristic of cytochrome P450s.
Figure 2(d) shows amino acid sequence comparisons between zP450RAI and several other cytochrome P450s (SEQ ID Nos: 6,7,8,9,10) in the area of the conserved hemebinding motif found in the superfamily. The cysteine, designated 0 in the figure, which has been shown to be directly involved in heme-binding [Gotoh, 1989] is surrounded by several highly 30 conserved amino acids.
Figure 3(a) shows Northern blot analysis of mRNAs obtained from regenerate tissue of RA-treated fish in lane 5, and controls (DMSO-treated fish) in lane 4, using a zP450RAI cDNA probe. Comparison to an RNA ladder (lane 1) shows the major zP450RAI transcript to be in the 1.4-2.4 kb range.
Figure 3(b) shows localization of zP450RAI transcripts in regenerating caudal fin tissue 72 hours post-amputation by whole mount in situ hybrization. zP450RAI transcripts were found to be undetectable in DMSO-treated regenerates. The original plane of amputation is indicated by the white line with arrowhead: m (soft mesenchyme) and r (bony rays) are labelled. (ii) In a sample obtained from an RA-treated fish, zP450RAI transcripts, indicated by -11 the black arrowhead, were found to be localized to a band of cells extending across the distal tip of the regenerate. Lower levels of expression of zP450RAI were also evident in nonregenerate tissue at the proximal base of the isolated fin, as indicated by the black line with arrowhead. The plane of amputation is indicated by the white line with arrowhead as in Figure (iii) A histological section taken through the plane is indicated by the line. (iv) A histological section of RA-treated fins post-hybridizationJs shown. Localized expression of zP450RAI was detected in a subset of epithelial cells (black arrowhead) which lie at the distal tip of the regenerate. Basement membrane separating the dense blastemae and the wound epithelium is indicated by the grey arrowhead.
Figure 4 shows elution profiles of lipid soluble extracts obtained from treated media of pSG5-zP450RAI transfected COS-1 cells and pSG5 transfected control cells.
Figures 4(a) and 4(b) are plots of cpm vs fraction number for cells incubated with 575 pM [11,12-'HJRA for 4 hours and 24 hours, respectively, pSG5-zP450RAI COS-1 cells and control cells Metabolism of [11.12- 3 H]RA to total aqueous soluble metabolites of total cpm) was measured using aliquots of the aqueous soluble extract subjected to p-scintillation counting. See insets of Figures 4(a) and Figures 4(c) and 4(d) are plots of absorbance vs retention time for cells incubated with 1pM RA for 4 and 24 hours, respectively. Peaks observed in zP450RAI transfected cell are shaded black. The region of the chromatogram from 4 to 6 min has been expanded (see insets of Figures 4(c) and In cells transfected 20 with zP450RAI cDNA, the generation of peaks corresponding to 4-oxo RA and 4-OH RA was observed.
Figure 5 shows results obtained with human cell lines probed with a a-("P]-dATP labeled probe having the sequence identified as SEQ ID NO:11: HEK293; EL-E; MCF10A; LC-T; SK-LC6; and MCF7. indicates pretreatment with 10- 6 M RA and indicates no RA pretreatment. The blot was also probed with hGAPDH to control for RNA loading of the gel, shown in the bottom panel.
Figure 6 is similar to Figure 5 for the cell lines U937 and HepG2.
Figure 7 is similar to Figure 5 for the NT2 cell line.
Figure 8 is similar to Figure 5 for a normal NB4 cell line (first two lanes) and 30 three individually derived retinoic acid resistant NB4 derivative cell lines.
Figure 9 shows the murine cDNA amino acid sequence (top row, SEQ ID o NO:32) aligned with the human P450RAI amino acid sequence (middle row; SEQ ID NO:4) and *o the zebrafish cDNA amino acid sequence (bottom row: SEQ ID NO:2).
Figure 10(a) shows elution profiles of lipid soluble extracts obtained from treated media of pSG5-hP450RAI transfected COS-1 cells and pSG5 transfected control cells.
Plots of cpm vs fraction number for cellc incubated with [11,12- 3 H]RA for 24 hours of hP450RAI COS-1 cells and control cells are shown. Figure 10(b) shows measurement of aliquots of the aqueous soluble extract subjected to p-scintillation counting taken to determine metabolism of [11,12- 3 HIRA to total aqueous soluble metabolites. Figure 10(c) shows plots of absorbance vs retention time for hP450RAI transfected cell and -12control cells cells incubated with 1 pM RA for 24 hours. The inset is the region around minutes, expanded for clarity.
Figure 11 shows 4-oxo-RA production of pSG5-hP450RAI transfected COS- 1 cells and pSG5 transfected control cells.
Figure 11(b) shows 4-OH-RA production of pSG5-hP450RAI transfected COS- 1 cells and pSG5 transfected control cells.
Figure 11(c) shows formation of aqueous soluble metabolites of hP450RAI transfected COS-1 cells and pSG5 transfected control cells.
Figure 11(d) shows unmetabolized RA of pSG5-hP450RAI transfected COS-1 cells and pSG5 transfected control cells.
Figure 12(a) shows elution profiles of lipid soluble extracts obtained from media of MCF10A cells exposed to RA and unexposed MCF1OA control cells. Plots of cpm vs fraction number for cells incubated with [11,12- 3 H]RA for 24 hours of RA-induced cells and control are shown.
Figure 12(b) shows elution profiles of lipid soluble extracts obtained from treated media of MCF7 cells exposed to RA and unexposed MCF7 control cells. Plots of cpm vs fraction number for cells incubated with [11,12- 3 H]RA for 24 hours of RA-induced MCF7 cells and control are shown.
Figure 12(c) shows the total aqueous soluble metabolites measured using 20 aliquots of the aqueous soluble extracts of the cell lines described in Figures 12(a) and (b) subjected to p-scintillation counting. The first two bars are for unexposed MCF7 cells and MCF7 cells exposed to RA, respectively. The third and fourth bars are for unexposed MCF1OA cells and MCF10A cells exposed to RA, respectively.
Figure 13(a) shows elution profiles of lipid soluble extracts obtained from 25 microsomal preparations after incubation with radiolabelled RA for ninety minutes, as described in Example 7. Plots of dpm vs fraction number for HeLa P microsomes and HeLa RAI microsomes Figure 13(b) shows fractions 5 to 15 of Figure 13(a) on a larger scale.
Figure 14(a) shows SDS-PAGE of the GST-hP450RAI fusion protein. Lane 1 shows the whole cell lysate of E. coli expressing the plasmid containing the fusion protein, .o without induction. Lane 2 shows the whole cell lysate of E. coli expressing the plasmid containing the fusion protein after induction with 0.1 mM IPTG.
Figure 14(b) shows SDS-PAGE of the purified GST-hP450RAI fusion protein after induction with 0.1 mM IPTG, with molecular weight markers at the right hand side indicating the fusion protein to have a molecular weight.of about 75 kDa.
Figure 15 shows promoter sequences of human, mouse and zebrafish P450RAI. The boxed regions show highly conserved regions while the arrows indicate spaced apart consensus sequences of RAREs.
Figure 16 shows relative luciferase activity induced in cells containing a luciferase vector into which was cloned a portion of the putative promoter for mP450RAI.
-13- Luciferase activity was measured in cell extract supernatants from cells transfected with 3 concentrations of expression vectors comprising cDNAs encoding RXRa and RARy (100 ng, 500 ng, and 1 pg) along with a luciferase-based reporter gene including either the sense or antisence promoter sequence, or no promoter sequence, grown in presence and absence of
RA.
Figure 17 shows inhibition of P450RAI mRNA in MCF7 cells by 4-hydroxyphenylretinamide (4-HPR). Cells were treated for twelve hours with the indicated concentrations of all-trans retinoic acid (atRA) and 4-HPR. Total RNA was extracted using TRIzol, and, following electrophoresis. Northern blotting was performed as described. The nitrocellulose was probed with radiolabelled P450RAI then GAPDH.
Figure 18 shows expression of cytochrome P450RAI in MCF7 cells by northern blot analysis, over time, after administration of all-trans retinoic acid.
Figure 19 shows expression of cytochrome P450RAI in MCF7 cells by northern blot analysis. over time, after administration of all-trans retinoic acid alone, and in the presence of all-trans retinoic acid and ketoconazole.
Figure 20 shows expression of cytochrome P450RAI in MCF7 cells by northern blot analysis, over time, after administration of all-trans retinoic acid and after administration of Am580.
DESCRIPTION OF PREFERRED EMBODIMENTS 20 Figure 1 outlines the steps used to isolate retinoid-regulated genes using differential display of mRNA. The cloned products isolated in step 6 of Figure 1 were used for sequencing and screening of Danio rerio rerio) cDNA libraries. P1, P2 and P3 correspond to fragments from RA induced mRNAs. P4 is a PCR product from a down-regulated mRNA.
Details of procedures followed in determination of gene sequences described herein follow.
Danio rerio stocks D. rerio were kept at 28.5'C in 40 L tanks with ?5-30 fish per tank on a 14 hour light-10 hour dark cycle. Tap water was conditioned by the addition of 10 ml of Water Conditioner (Sera Aqutan) and 10 ml of 250 g/L Aquarium Salt (Nutra Fin) per 20 L. 2-3 L of water was changed daily. Amputation of fins was carried out following anaesthetization of the 30 fish in a solution of 0.2% ethyl-m-aminobenzoate methanesulfonic acid (ICN) in conditioned water. Retinoic acid treatment was performed by adding all-trans RA, to a final concentration of 10.6 M, directly into the tank water two days following amputation. Both control- and RAtreated fish were kept in the dark during the experiments.
Differential Display of mRNAs Differential mRNA display was performed essentially as described by Liang and Pardee (1992) with appropriate modifications as described herein. Regenerating tissues were collected 3 days post-amputation (24 hours post-RA addition) and quick frozen in liquid 13a nitrogen. Poly RNA was isolated using the Micro Fast-Track kit. Duplicate independent reverse transcription reactions were performed on the isolated poly(A)* RNA from both the treated and untreated samples for each specific 3' poly-T primer used The symbol represents A or C or G and not Tor U. Several combinations of the 3' poly-T primers given in the first column of Table 1 and the upstream primers given in the second column were utilized for PCR amplification. For each reaction 0.1 pg poly(A)' RNA was reverse o 9*4 a* r -14transcribed in a 20 pi reaction volume containing 300U Superscript Reverse Transcriptase (Gibco/BRL), 1X Buffer, 20 pM each dGTP, dATP, dCTP and dTTP, 10 pM dithiothreitol (DTT) and 5 pmol of primer. The reactions were mixed and incubated at 35 0 C for minutes, followed by 5 minutes at 95 0 C. PCR amplification was performed in a Perkin Elmer Cetus PCR machine as follows: 1 pl cDNA synthesis reaction, 5U Taq DNA polymerase (Gibco/BRL), 1X PCR Buffer, 2 pM each dGTP, dATP, dCTP and dTTP, 10 pCi a-['S]dATP (redivue, Amersham) 1.2 mM MgCI,, 0.5 pM upstream primer and 0.5 pM of the corresponding primer. PCR conditions were as follows: 1 cycle, 94°C for 5 minutes; 40 cycles, 94°C for 30 seconds, 42*C for 1 minute, 72 0 C for 30 seconds; followed by a final extension of 5 minutes at 72°C. 4 pl of the PCR reactions were loaded onto a 6% non-denaturing polyacrylamide gel and electrophoresed at 60 watts, 45"C. The gel was dried and exposed for 12 to 24 hours on Kodak XAR film at room temperature.
6* Se *fee 05 a 5 0 .5.5 so S& 0.
S
S
*SSS
55 0 *0@ 0@ 005 6 *0 0 0 20 25 Table 1. Sequences of the downstream Poly oligonucleotides for the differential display procedure.
Poly(T) primers: 5'-degenerate primers: TTT TTT TTT GG-3' 5'-AAG CGA CCG A-3' TTT TTT TTT GA-3' 5'-TGT TCG CCA G-3' 5'-TTT TTT TTT TTT GT-3' 5'-TGC CAG TGG A-3' 5'-TTT TTT TTT TTT GC-3' 5'-GGC TGC AAA C-3' 5'-CCT AGC GTT G-3' 5'-TTT TTT TTT TTT AG-3' 5'-TTT TTT TTT TTT AA-3' TTT TTT TTT AT-3' TTT TTT TTT AC-3' 5'-TTT TTT TTT CG-3' 5'-TTTTTT TTT TTT CA-3' TTT TTT TTT CT-3' 5'-TTT TTT TTT TTT CC-3' In Table 1, the sequences in the first column are identified as SEQ ID NOs: 12, 13, 14, 15, 16, 17, 18, 19, 20, 21,22 and 23, respectively. The sequences in the second 30 column are identified as SEQ ID NOs: 24, 25, 26, 27 and 28, respectively.
Gel Purification and Peamplification Bands demonstrating reproducible differential amplifications (see Figure 2a) were found for the upstream-downstream primer combination of 5'-TGCCAGTGGA-3'- poly-T primer, 5'-TTT TTT TTT TTT AG-3' (SEQ ID NOs: 26 and 16, respectively). These bands were excised from the gel by overlaying the X-ray film and cutting out the corresponding piece of dried gel and filter paper. The PCR product corresponding to a fragment of the protein described herein was isolated from the band in Figure Samples were placed in 100 pl of nuclease free water, incubated for 10 minutes at room temperature, then boiled for 15 minutes.
The supernatant was recovered following a 15 minute centrifugation at 12,000 x g.
In order to facilitate cloning of the PCR products, several changes were made to the reactions. Primers which included Eagl restriction endonuclease sites were used in the reamplification. Based on results obtained in the differential display analysis, the upstream TGCCAGTGGA-3' primer was replaced by 5'-GTAGCGGCCGCTGCCAGTGGA-3' (SEQ ID NO: 29) and the downstream poly-T primer, 5'-TTT TTT TTT TTT AG-3', was replaced by
GTAGCGGCCGCT,
1 (SEQ ID NO:30). In addition, the reaction volume was increased to pi, isotope was omitted and 20 as opposed to 40 cycles were performed. 5 pl aliquots of the PCR reactions were removed and the products were visualized by electrophoresis in a 1% agarose gel followed by ethidium bromide staining and UV illumination.
Cloning PCR Products The reamplified products were purified by phenol/chloroform extraction followed by ethanol precipitation. The resulting DNA pellet was resuspended in 17 pl of sterile water and digested at 37°C for 1 hour by the inclusion of 10U Eagl (New England Biolabs), and 1X NEB 3 buffer. Eagl restriction endonuclease was heat inactivated by incubation at 65°C for 20 minutes. pBluescript SK* vector was prepared by digestion with EagI, followed by dephosphorylation using calf intestinal alkaline phosphatase (CAP, Promega). Restriction digests were purified using the GeneClean II Kit (Bio 101) following electrophoresis in a 1% agarose gel. In a total ligation volume of 10 pl, 2 pl of digested PCR product, 1 pl digested SK*, 1U T4 DNA ligase (Gibco/BRL) and iX buffer were incubated at 16°C overnight. E. coli 20 bacterial strain JM109 was transformed with 1 pl of the ligation product using a BioRad Gene Pulser, then plated on LB+ampicillin plates and incubated overnight at 37 0
C.
Colony Selection Individual colonies were transferred in duplicate to fresh LB plates and.grown overnight at 37°C. Colonies were tranferred to nitrocellulose membrane and denatured in a 25 solution of 1.5M NaCI, 0.5M NaOH for 5 minutes, neutralized in 1.5M NaCI, 0.5M Tris-HCI, pH 8.0 for 5 minutes, followed by two 5 minute washes in 2X SSC. Membranes were then UV cross-linked (Stratalinker UV Crosslinker, Stratagene). Prehybridization and hybridization were performed using Quickhyb (Stratagene) following the manufacturer's directions. Each colony lift was probed with the corresponding PCR product isolated during the gel reamplification and purification step. a-[ 32 P]-dATP labelled probes were generated using the Prime-It Kit II (Stratagene). Subsequent to hybridization, filters were washed twice for 20 minutes in 2X SSC, 0.1% SDS solution at room temperature and exposed to Kodak X-omat autoradiography film overnight at -70*C. Positive colonies were selected from the duplicate plates, grown overnight in LB+ampicillin (100 pg/ml) and plasmid DNA isolated using the Qiaprep Spin Plasmid Kit (Qiagen).
Clones were sequenced using the T7 Sequencing Kit (Pharmacia Biotech).
Sequence comparisons were generated using the GeneWorks software package (Intelligenetics).
-16- Screening of a D. rerio cDNA Library A random primed D. rerio 6-18 hour embryo cDNA library constructed in Uni- ZAP II (Stratagene) was produced. 4.5 x 10 5 independent pfu were screened using the random primed, 3 P]-dATP labelled 337 bp PCR fragment isolated by mRNA differential display as a probe. Filters were prehybridized for 1-4 hours at 42 0 °Cin 50% formamide, 5X SSPE, 1X Denhardt's solution, 0.2 mg/ml denatured salmon sperm DNA. Hybridization was performed at 42*C by adding denatured probe to the prehybridization solution. Filters were washed two times for 20 minutes in 2X SSC, 0.05% SDS at room temperature and exposed to Kodak XAR film overnight at -70"C. Positive plaques were picked into 500 pl SM buffer and subjected to additional rounds of rescreening until purified. Positive plaques were exposed to the in vivo excision protocol following the manufacturer's directions (Stratagene). pBluescript containing colonies were plated onto LB+amp plates and grown overnight at 37°C. Sequence data were generated using the T7 Sequencing Kit (Pharmacia) and analysed using the GeneWorks software package (Intelligenetics).
Whole Mount in situ hybridization RA- and DMSO-treated regenerates were isolated 72 hours post-amputation (24 hours post RA/DMSO addition), washed in PBS and prepared for whole mount in situ hybridization. In situ hybridizations were undertaken as previously described [White, 1994].
Northern Blot Analysis Fish were allowed to regenerate their caudal fins for 72 hours. At 48 hours all-trans RA in DMSO vehicle or DMSO alone was added directly to the tank water.
S•mRNA was prepared using the Micro Fast-Track mRNA isolation kit (Invitrogen, CA) according to the manufacturer's directions. 3.0-5.0 pg poly A' RNA was electrophoresed, blotted and probed using a previously described method [White, 1994) with the full length zP450RAI cDNA 25 obtained as described below. Ethidium bromide stained agarose gel showed that equivalent amounts of mRNA were used in the blotting experiments. See lanes 2 and 3 of Figure 3(a).
HPLC Analysis Media from transfected cells incubated with 575 pM [11,12-'H]RA (Figures 4(a) and or 1 pM RA (Figures 4(c) and for either 4 hrs (Figures 4 and or 24 hrs (Figures 4(b) and were acidified with 0.1% acetic acid. Lipid soluble metabolites were separated from aqueous soluble metabolites using a total lipid extraction of the medium [Bligh, 1957]. Metabolism of [11,12-'H]RA to total aqueous soluble metabolites was measured using aliquots of the aqueous soluble extract subjected to l-scintillation counting (See the insets of Figures 4(a) and Upid soluble extracts were evaporated to dryness under a stream of nitrogen and resuspended in 93.5/5/1/0.5 hexane/isopropanol/methanollacetic acid (H/I/M/AA).
Metabolites were separated by HPLC using a Zorbax-SIL (3p, 8 x 0.62 cm) column eluted with a solvent system of 93.5/5/1/0.5 H/I/M/AA at a flow rate of 1 ml/min.
-17- EXAMPLE 1 Characterization of a novel cytochrome P450 Transcripts present in fin tissue regenerating in the presence or absence of RA were compared using the differential display PCR technique developed by Liang and Pardee [Liang, 1992] (Fig One of the differential display products which exhibited a dependence on the presence of RA for its expression, indicated by the arrow in Figure was isolated and sequenced. The sequence is identified as SEQ ID NO:1 and is also shown in Figure The amino acid sequence corresponding to the cDNA, termed here, "zP450RAI", is shown in Figure 2(c) and identified as SEQ ID NO:2. BLAST search analyses revealed sequence homology between zP450RAI and multiple members of the cytochrome P450 superfamily. Alignments between zP450RAI cDNA deduced amino acid sequence and those of other cytochrome P450s indicated that zP450RAI exhibited less than 30% overall amino acid identity with members of previously defined subfamilies [Nelson, 1993]. zP450RAI contains many of the structural motifs which are common to cytochrome P450 family members, including the hemebinding domain located in the C-terminal portion of the protein. See Figure The P450RAI family has been designated "CYP26".
EXAMPLE 2 Cell specific induction of zP450RAI by all-trans RA Northern blot analysis of mRNAs expressed in regenerate tissue isolated from 20 control (dimethyl sulfoxide-treated) and RA-treated fish was performed with a full-length zP450RAI cDNA probe. zP450RAI transcripts were not detectable in regenerate tissue from control fish (Figure lane 4) but were very noticeably present in tissues isolated from fish exposed to RA for 24 hours (Figure lane Whole mount in situ hybridization was used to determine the cellular localization of zP450RAI expression in regenerating fin tissue. Figure 3(b) shows regenerating fins from control and RA-treated fish. zP450RAI transcripts are not detectable in control fin tissue (Figure In regenerating tissue from RA-treated fish, zP450RAI transcripts were found to be abundant in a layer of epithelial cells extending across the distal edge of the wound epithelium as indicated by the black arrowhead in Figure Some low level staining was 30 also observed in inter-ray tissue as indicated by the black line with arrowhead in Figure 3(b)(ii).
A histological section of an RA-treated fin, taken along the line shown in Figure 3(b)(iii), is shown in Figure The section indicates that cells expressing zP450RAI are located deep within the epithelial layer at the distal tip of the blastemal mesenchyme. Whole mount in situ hybridization thus illustrates the usefulness of nucleic acid probes of the invention for the localization of cytochrome P450RAI mRNA in whole tissue.
-18- EXAMPLE 3 Metabolism of all-trans RA by zP450RAI transfected cells Retinoic acid as a substrate of zP450RAI was studied. The full-length zebrafish zP450RAI cDNA was cloned into the eukaryotic expression vector pSG5 [Green, 1988]. COS-1 cells were transiently transfected with either pSG5 or pSG5-zP450RAI and then incubated with either picomolar concentrations of [11,12 -H]all-trans-RA or micromolar concentrations of non-radioactive all-trans-RA. COS-1 cells are an African green monkey kidney "fibroblast-like" cell line. zP450RAI expression in COS-1 cells promoted the rapid conversion of RA into both lipid- and aqueous-soluble metabolites. See Figures 4(a) and 4(b).
Fractions of total lipid extracts of transfected cells were initially separated by normal-phase HPLC on Zorbax-SIL. Comparison between extracts from pSG5 and pSG5-zP450RAItransfected cells indicated that zP450RAI significantly increased RA metabolism. Incubation of zP450RAI-transfected cells with 575 pM [11 ,12- 3 Hall-trans-RA for either 4 or 24 hours resulted in accumulation of RA metabolites, one of which co-migrated on a column with synthetic standards 4-OH-RA and 18-OH-RA, and a second slightly less polar metabolite which comigrated with 4-oxo-RA standard (Figures 4(a) and Rechromatography of RA metabolites using other HPLC systems confirmed the identity of these two metabolites as 4- OH-RA and 4-oxo-RA (Table It is possible that the aqueous-soluble radioactivity represents glucuronides of RA metabolites or glucuronides of RA itself. Rapid glucuronidation of 4- and 18-hydroxy-RA in mammalian cell extracts has been reported by others (Wouters, 1992; Takatsuka, 1996].
Table 2 Chromatoaraphic properties of RA metabolites.
Metabolite Retention Time (min) Z-Sil Z-CNb Z-ODSc 25 RA (std) 2.57 4.47 19.92 4-oxo-RA (std) -4.79 11.33 11.73 4-OH-RA (std) 5.17 9.65 12.65 18-OH-RA (std) 5.06 9.53 14.03 Peak 1 (RA) 2.57 4.48 19.73 30 Peak 2 (4-oxo-RA) 4.87 11.38 11.57 Peak 3 (4-OH-RA) 5.16 9.68 12.68 HPLC conditions: Zorbax-SIL column eluted with 93.5/5//0.5 m/m b'HPLC conditions: Zorbax-SIL column eluted with 93.5/5/1/0.5 H/I/M/A.A (1 ml/min) "HPLC conditions: Zorbax-CN column eluted with 93.5/5/1/0.5 H/I/M/A.A (1 ml/min) HPLC conditions: Zorbax-ODS column eluted with a 20 min linear gradient with solvent containing 10 mM ammonium acetate which ranged from 55.45 to 5.95 H,O /MeOH (2 ml/min).
A similar pattern of zP450RAI-dependent metabolism was also observed using a much higher RA concentration (1 pM). zP450RAI-transfected COS-1 cells incubated for 4 or 24 hours with 1 pM RA generated two closely-running peaks which were discernible inca 350nm HPLC trace shown in Figures 4(c) and but which were essentially undetectable in control cells (See the insets of Figures 4(c) and These peaks co-migrated with those of 4-oxo-RA and 4-OH-RA standards, respectively. Diode array spectrophotometric -19detection of the zP450RAI-generated peaks showed that the spectral properties of the two metabolite peaks matched the standard retinoids [In hexane-based solvents: 4-OH-RA, m,.=350nm; 4-oxo-RA, l,.=355nm; in methanol-based solvents: 4-OH-RA, Am,=340nm; 4oxo-RA, A,=360nmj.
The invention thus includes a retinoic acid metabolizing protein belonging to the family of cytochrome P450s, designated CYP26, and generation of the protein in zebrafish caudal fin wound epithelium being induced in response to RA treatment. While RA metabolizing activity has previously been detected in epithelial tissues of several species [Frolik, 1979; Roberts, 1979; Wouters, 1992; Duell, 1992], an actual enzyme responsible for such activity has heretofore been unknown.
zP450RAI is up-regulated by RA treatment and apparently this up-regulation occurs in a specific set of cells in the wound epithelium of regenerating zebrafish caudal fins.
It might be of relevance to the regulation of the generation of this enzyme in vivo that experiments with F9 cells where RARs have been selectively ablated indicate that RAR-a and RAR-y might have a role in the regulation of RA metabolism [Boylan, 1995]. The expression of both RAR-a and RAR-y in the regenerating caudal fin is consistent with the suggestion that they may be involved in the regulation of P450RAI expression by RA [White, 1994].
EXAMPLE 4 20 Cloning of Human P450RAI The amino acid sequence corresponding to the DNA of zebrafish P450RAI (zP450RAI) (SEQ ID NO:2) was used to search an express sequence tag (EST) database. A oe commercially available EST clone (SEQ ID NO:11) having a high degree of homology with a Cterminal portion of the zP450RAI (from Glu 292 to Phe 410 of SEQ ID NO:2) was purchased (Research Genetics, Huntsville, AL). The clone is reportedly from a human infant brain cDNA library (Bento Soares and M. Fatima Bonaldo) and is apparently otherwise unpublished. The purchased clone was sequenced using the T7 sequencing kit (Pharmacia) and sequence data was generated using the Geneworks Software Package (Intelligenetics).
A cDNA library generated from an NT2 cell line treated with retinoic acid is 30 commercially available (Stratagene, cat#939231) and this product was used for further studies.
The cDNA library was probed with a nucleic acid having a sequence identified as SEQ ID NO:11. Eleven positively hybridizing clones were isolated and purified according to the manufacturer's directions. Sequence data for these clones were generated using the T7 Sequencing Kit (Pharmacia) and analyzed using the Geneworks package (Intelligenetics). The human DNA sequence is identified as SEQ ID NO:5 and the corresponding polypeptide as SEQ ID NO:4.
EXAMPLE Isolating Mouse P450RAI It has been found by the present inventors (results not shown) that RA can induce mRNA transcripts which cross hybridize with a hP450RAI cDNA probe in either of the F9 and P19 mouse cell lines having 4-hydroxylase activity,_as described by Blumberg etal.
[Blumberg et 1995; Achkar et al., 1996].
A retinoic acid-treated P19 teratocarcinoma cDNA library in lambda Unizap XR (Stratagene) was screened using 2 P-labelled full length human P450RAI as a probe.
Nitrocellulose filters were hybridized overnight at 37°C in a buffer containing 50% formamide, 1x Denhardt's, 5x SSPE, 0.1% SDS, and 100 pg/ml boiled salmon sperm DNA, and washed in 2x SSC, 0.1% SDS for 2x 15 minutes at room temperature followed by 20 minutes at 37 0
C.
From a total of 600,000 plaque forming units of phage, 72 positives were isolated. Of these, 18 were purified, as described above, and one was found to have a full-length cDNA insert 1.7 kb in length homologous to the full-length human clone previously isolated. The murine DNA sequence is identified as SEQ ID NO:31 and the corresponding polypeptide as SEQ ID NO:32.
Figure 9 shows aligned portions of the amino acid sequence of the mouse protein (SEQ ID NO:32) with those of the human protein (SEQ ID NO:4) and the zebrafish protein (SEQ ID NO:2).
EXAMPLE 6 Transient Tranfection Analysis COS-1 cells were subcultured 20 hours prior to transfection which was carried out according to the standard DEAE-dextran method [Sambrook, 1989; Maniatis, 1982]. Cells were transfected with pE-AR (adrenodoxin expression vector, 1 pg/P100 plate) and pE-ADX (adrenodoxin reductase expression vector, 1 pg/P100 plate) together with 3 pg per plate of either pSG5 (control) or hP450RAI-pSG5 (experimental). [11,12- 3 H]all-trans retinoic acid (60,000 cpm per plate) was added 24 hours after transfection. Analyses were carried out as described in Example 3 and results obtained are shown in Figures 10 and 11(a) to 11(d). As indicated in the Figures, hP450RAI expression in COS-1 cells promoted conversion of RA into 4-OH-RA and 4-oxo-RA. Total amounts of 4-oxo-RA and 4-OH-RA produced in the transfected 30 cells in comparison to amounts produced in the control cells are shown in Figures 11(a) and respectively. Overall, greater amounts of aqueous soluble metabolites were produced in the transfected cells (Figure 11(c)) and greater amounts of unmetabolized RA were found in control cells (Figure 11(d)) The clone sequence (SEQ ID NO:11) was prepared as a "[P]-dATP labeled probe to study the inducibility of hP450RAI by RA in several cell lines: HEK293; EL-E; LC-T; SK-LC6; MCF7; U937; HepG2; NT2 (See Figures 5 to As can be seen, a variety of expression patterns were observed. The SK-LC6 human lung (epithelial) line appeared to constitutively express corresponding mRNA. There was apparently some increase in expression in the HEK293 (human embryonic kidney), LC-T (human lung epithelial), HepG2 -21- (human liver, epithelial in morphology), NT2 (pluripotent human embryonic carcinoma) and U937 (human monomyelocytes) cell lines in response to addition of RA. There was a large dependence on exposure to RA in the MCF7 (human breast carcinoma (epithelial)) cell line.
Some cell lines showed no expression in the absence or presence of RA: EL-E; HL-60 and The 3 2 [P]-dATP labeled probe was also used to study hP450RAI mRNA expression in a human acute promyelocytic leukemia cell line. Experiments were carried out using the NB4 cell line, isolated from a human acute promyelocytic leukemia patient, and three retinoic acid resistant cell lines were independently derived from NB4. Results are shown in Figure 8. As can be seen, the normal cells expressed hP450RAI mRNA after treatment with RA, while such expression appeared to be absent for the other cell lines both in the absence and presence of RA.
Analysis of metabolites of MCF10A and MCF7 cell lines exposed to RA was carried out, MCF10A cells having displayed no expression of mRNA and latter having displayed a large dependence of mRNA expression on exposure to RA. The results are shown in Figures 12(a) to 12(c). Consistent with the results shown in Figure 5, the results shown in Figure 12(a) indicate there was little difference in the lipid soluble activity profiles of the MCF10A cell line exposed to RA and the control. The last two bars of Figure 12 indicate that total aqueous soluble metabolites were about the same for both the induced and control MCF1 A cells. As 20 indicated in Figure 12(b), the MCF7 cell line exposed to RA had an elution profile which indicated significantly greater concentrations of 4-OH-RA and 4-oxo-RA than the same cell line not exposed to RA. Figure 12(c) indicates that the amount of total aqueous soluble metabolites of the MCF7 cells exposed to RA was much greater than that for the control cells. Again, these results are consistent with those obtained in the Northern blot analysis shown in Figure 5 for the MCF7 cell line.
EXAMPLE 7 Generation of a Stable Cell Line using Human P450RAI: HeLa Cells Expressing Sense Construct For expression in HeLa cells, the human cytochrome P450RAI cDNA (SEQ ID 30 NO:5) was inserted into the Xhol-Notl sites of the multiple cloning site of the Epstein-Barr virusbased vector pCEBV7 [Wilson, 1995]. Stable transfection was performed via the calcium phosphate method [Sambrook, 1989]. Prior to the day of transfection, HeLa cells were seeded at 3.0 X 106 cells per 100 mm plate. Approximately 12 pg of DNA were transfected per plate and triplicate plates were employed for the transfection. Selection using hygromycin B began three days after the transfection and continued for approximately three weeks until the development of foci on the plates. The concentration of hygromycin B (100 pg/ml) was chosen for selection of cells with high expression of the consiruct. A killing curve was determined prior to selection which showed that 50 pg/ml of hygromycin was sufficient to kill 50% of the cells in 4 -22days. Confirmation of the selected HeLa cells expressing the sense construct was determined by Northern blot analysis and probing with full length hP450RAI cDNA (data not shown).
Microsomes were prepared from HeLa cells transfected with the pCEBV7 alone (HeLa P) or from the HeLa cells expressing the P450RAI (HeLa RAI) and exposed to radiolabelled RA for ninety minutes. The results are shown in Figures 13(a) and 13(b), in which it can be seen that when microsomes prepared from these cells are incubated with RA, only those from HeLa RAI showed any conversion of retinoic acid to 4-hydroxy-retinoic acid or 4oxo-retinoic acid.
EXAMPLE 8 Expressing a Fusion Protein Containing Human P450RAI The cDNA of hP450RAI was inserted into the GST gene fusion vector, pGEX3X using the Smal site. The construct generated lacked 177 bp of the N-terminal portion of the hP450RAI cDNA, which encodes a hydrophobic region characteristic of a signal sequence. E. coli (JM,109) cells were used for expression of vectors and construct. After screening for positive colonies expressing the construct in the proper orientation, expression of the fusion protein was carried out according to the manual provided by Pharmacia.
GST-hP450RAI fusion protein was generated via electroporation of E.coli (JM109) cells with the construct, followed by induction of gene expression. Expression of the fusion protein is under the control of the tac promoter which is inducible by the lactose analog, isopropylbeta-thiogalactoside
(IPTG).
Cultures were grown overnight in LB-ampicillin broth. The following day, approximately 1:100 dilution factor of the overnight culture was transferred to a fresh tube "I containing LB-ampicillin broth. The culture was allowed to grow for three hours and induced with IPTG to a final concentration of 0.1 mM for an additional 2.5 hours. After induction, the culture was centrifuged at 7,700x g for 10 minutes at 4°C to sediment the cells. The pellet was resuspended in a ratio of 50pl of culture/ml of ice-cold 1X PBS. The cells were lysed with a sonicator at 30 second intervals while submerged in ice-water. Triton X-100 was added to the lysed cells to a final concentration of 1% and the mixture was incubated for 30 minutes at 4°C to allow solubilization. The suspension was subjected to centrifugation at 12,000x g for 30 minutes at 4"C.
Binding of the GST portion of the fusion protein to glutathione beads was used to purify the protein. 2 ml of a 50% slurry of reduced glutathione beads equilibrated with 1 X PBS were used per 100 ml of sonicate mixture from above. After the addition of the beads, the material was gently agitated for 30 minutes at room temperature to allow binding of the fusion protein to the beads. The suspension was centrifuged at 500 x g for 5 minutes to separate glutathione beads with bound fusion protein from other cellular components. The glutathione beads were washed three times with cold PBS to remove non-specifically bound material.
The fusion protein was eluted from the glutathione beads using an elution buffer containing reduced glutathione. Approximately 1.0 ml of glutathione elution buffer per -23 100 ml of sonicate suspension was used. The bound glutathione beads were allowed to mix at room temperature for 10 minutes to elute the fusion protein. Centrifugation of the eluted beads was performed at 500 x g for 5 minutes. Both the elution and centrifugation were performed three times. Figures 14(a) and show the results of SDS-PAGE of the GST-hP450RAI fusion protein.
It is generally possible to carry out some functional tests with the GST protein covalently attached to the protein of interest, but removal is preferred for the production of antibodies. The GST-hP450RAI fusion protein contains a site recognized by factor Xa.
Treatment of the eluted fusion protein with factor Xa yields both the GST protein and hP450RAI protein. Alternatively, it is possible to cleave the fusion protein prior to its elution from the beads, but inadequate cleavage many occur. Approximately 10 pg of factor Xa/mg of fusion protein is used. The material is mixed gently and incubated for approximately 2-16 hours at room temperature. Completion of digestion is checked by gel eletrophoresis at various time points. To purify the protein of interest from the GST protein, binding of cleaved fusion protein suspension with glutathione beads is carried out, followed by centrifugation. The protein of interest remains in the supernatant.
The GST fusion protein can be assayed immunologically or biochemically.
SBiochemically, treatment of the GST fusion protein with the GST substrate, 1-chloro-2,4dinitrobenzene (CDNB), indicates the amount of the fusion protein. The GST protein catalyzes 20 the conjugation of CDNB with glutathione, resulting in a CDNB-glutathione product with a strong molar absorptivity at 340 nm. Immunologically, a Western blot of the fusion protein can be probed with an anti-GST antibody which can be readily detected using a secondary antibody such as an anti-goat IgG alkaline phosphatase conjugate, and the standard colorimetric reaction.
EXAMPLE 9 SMapping of Human P450RAI A 1.3 kb cDNA of hP450RAI was mapped using a P-1 derived artificial.
chromosome (PAC) library. Mapping of the cDNA and genomic PAC clone was performed by fluorescence in situ hybridization [Lichter, 1990] to normal human lymphocyte chromosomes 30 counterstained with propidium iodide and DAPI. Biotinylated probe was detected with avidinfluorescein isothiocyanate (FITC). Images of metaphase preparations were captured by a thermoelectrically cooled charge coupled camera (Photometrics, Tucson, Arizona). Separate images of DAPI banded chromosomes [Heng, 1993] and FITC targeted chromosomes were obtained. Hybridization signals were acquired and merged using image analysis software and pseudo colored blue (DAPI) and yellow (FTIC) (Boyle, 1992] and overlaid electronically.
Positive hybridization signals were found to be localized to 10q23-24. The band assignment was determined by measuring the fractional chromosome length and by analyzing the banding pattern generated by the DAPI counterstained image.
-24- EXAMPLE Regulation of P450RAI transcription Cloning of P450RAI Promoters Cloning of Zebrafish P450RAI promoter. An adult zebrafish genomic library (1.0 x 106 pfu) was screened with the full length cDNA corresponding to zP450RAI and positive plaques purified through secondary and tertiary screening. Lambda DNA corresponding to positive plaques was prepared, and the inserts from these cloneswere excised by restriction enzyme digestion with Notl and subcloned into the plasmid SK+ (Stratagene). Genomic clones were analyzed by restriction enzyme digestion using enzymes from the polylinker of SK+, followed by Southern blotting using an oligonucleotide (5'-GTAGCACGGATGGTG-3') which hybridizes to the nucleotide sequence encoding the N-terminus of the zP450RAI cDNA to identify fragments of the genomic clones which encode the N-terminal region. A 772 base pair Pstl fragment which hybridized with the oligonucleotide probe was purified, ligated into the vector SK+ and sequenced using the Core Facility for Protein/DNA Chemistry at Queen's University, Kingston, Canada. Sequence analysis identified this clone as containing the putative initial methionine followed by 129 base pairs of coding sequence, plus 651 nucleotides upstream Within this 772 base pair fragment, a 402 base pair Hindlll fragment was found to contain the putative retinoic acid response element (RARE). This fragment was subcloned into the pGL3B luciferase vector, in both the forward and reverse orientations, (Promega) for transient transfection analyses.
20 Cloning Human P450RAI promoter. The full length hP450RAI cDNA was used as a probe to S. identify PAC (P1 artificial chromosome) clones which contain the hP450RAI gene. cDNA from hP450RAI was sent to Canadian Genome Analysis and Technology Program at the Hospital for Sick Children in Toronto, Ontario, Canada for screening of PAC libraries. 5 PAC clones were obtained from this screening, which were verified to contain the hP450RAI gene by restriction 25 enzyme digestion and Southern blotting using the full length hP450RAI cDNA as a probe. One of these clones, 245C7, was found to hybridize to an N-terminal probe from hP450RAI. The probe used was an approximately 130 bp NotI fragment generated from the hP450RAI cDNA.
Digestion and Southern blotting of clone 245C7 identified an approximately 3.5 Kb BamHI fragment which hybridized with the NotI fragment. This fragment was subcloned into the plasmid SK+ and sequence data generated at the Core Facility for Protein/DNA Chemistry at Queen's University, Kingston, Canada. Comparison of the sequence data generated with the hP450RAI cDNA identified this 3.5 Kb clone as containing the potential initial methionine and approximately 675 bp upstream Cloning of mouse P450RAI promoter. A clone of mouse P450RAI genomic DNA approximately 17 kb long was isolated from an SV129 ADASH library and subcloned into SK.
DNA prepared from this plasmid was digested with various restriction endonucleases, electrophoresed on an agarose gel, and Southern blotted onto nitrocellulose. The resulting blot was hybridized with a "P-labelled 230 base pair probe from the N-terminal region of a mouse P450RAI cDNA clone. A Sad fragment approximately 520 base pairs in length was found to hybridize strongly to the probe. This fragment was subcloned into SK cleaved with Sacl. Sequence analysis revealed the presence of a DRS-type RARE in the proximal promoter.
Flanking this RARE were two BssHII sites 193 base pairs apart. This 193 base pair BssHII fragment was subcloned into the Mlul site of pGL3B. Diagnostic restriction digests with Xhol were done to isolate clones with the BssHII fragments in both forward and reverse orientations.
Within the 193 base pair fragment is also found the TATA box, 45 base pairs downstream of the RARE. Also included in the 193 base pair BssHII fragment is 83 base pairs of DNA upstream of the DR5 RARE.
The existence of retinoic acid response elements (RAREs), which bind retinoic acid receptors (RARs) and retinoid-x-receptors RXRs is known. The three promoters of P450RAIs from different species described above were thus examined for the presence of RAREs, which might indicate that the regulation of expression of P450RAI is at the transcriptional level. RAREs bind RARs in the form of a heterodimer and typically include a direct repeat of the consensus motif The repeated sequences can be separated by 1, 2 or 5 nucleotides. Figure 15 shows the promoter sequences for the zebrafish, human and mouse P450RAI genes. There is a highly conserved motif, indicated by the arrows, which corresponds to a consensus retinoic acid response element known as a DR5, consisting of a direct repeat of a motif AGTTCA separated by 5 nucleotides. This motif in the P450RAI promoters extends over the sequence TGAACT (the complement of AGTTCA). There is also a highly conserved region extending on either side of the DR5, the entire region being boxed.
20 The degree of conservation of this region of the promoter between all three species suggests that this region is important to the control of RA response and that RAR/RXRs are acting in conjunction or competition with other factors binding to this region whose identities are currently unknown. A portion of the conserved region corresponds to the sequence 5'-CAATTAAA-3' which corresponds very closely to a homeodomain binding motif, suggesting that homeobox proteins may facilitate or compete with RAR/RXR heterodimers in the regulation of P450RAI expression.
EXAMPLE 11 Transfection with Mouse P450RAI Promoter A 195 basepair fragment of genomic DNA containing a portion of the putative S. 30 promoter for mP450RAI was cloned into the luciferase vector, pGL3B (Promega). For analyses of promoter activity, HepG2 cells were transfected in 6-well dishes with 2 pg BssHIpGL3B sense or antisense constructs, described in Example 10, using 5 pl lipofectAMINE reagent (Gibco, BRL).
On the first day, 48 hours prior to transfection, cells were replated into 6-well plates, with 2.5-3 mis Minimal Essential Medium (MEM) (Gibco, BRL)+ 10% Fetal Calf Serum (Gibco, BRL). On the third day, cells are generally about 50% confluent (HepG2). Before beginning transfection, the medium was replaced with fresh medium and the cells were allowed to grow while preparing DNA/lipofectamine mix. In individual wells of a 48 well plate were mixed 1-5 pg DNA in 100 pl optiMEM (Gibco, BRL) and 5 pl lipofectAMINE reagent in 100 -26pl optiMEM, by addition of the DNA/optiMEM to the lipofectAMINE/optiMEM with gentle mixing.
This was left to sit 15 min to 1 hour at room temperature. To each well were added 800 pi optiMEM to obtain a final volume of 1 ml. The cells were washed 2 times in 1X PBS and once in optiMEM. The 1 ml DNA/lipofectamine/optiMEM mixture was added to the cells and incubated for 20 hours at 37"C.
The effects of retinoic acid on promoter activity were analyzed by cotransfecting with varying amounts (100 ng to 1 pg) of expression vectors including cDNA sequences encoding zebrafish retinoic acid receptor gamma (RAR-y) and zebrafish retinoid x receptor alpha (RXR-a). Comparisons were made between cells transfected with the control pGL3B vector, those with the sense construct and those with the antisense construct by incubating one set of the transfected cells with DMSO and the other set with 10' M RA in
DMSO.
On the fourth day, the medium was removed and replaced with fresh medium 10% FBS). The RA or vehicle was added as required to the wells and mixed gently by swirling the plate. For a 6 well plate 3 pl of 10' M RA in DMSO were added. Control wells received 3 pl DMSO. The plates were covered in foil and incubated for 24 hours at 37'C.
On the fifth day, cells were harvested by removing the medium and washing twice in 1X PBS. Then, on ice, 100 pl lysis buffer Triton X-100, 25 mM glycylglycine, pH 7.8, 15 mM MgSO,, 4 mM EGTA, 1mM DTT (added fresh just before use)) were added, and the 20 cells were scraped off the bottom of the dish and transferred to a 1.5 ml microcentrifuge tube, spun for 5 minutes at 12000 x g, and the supernatant recovered..
To assay for luciferase activity, 20 pl supernatant from lysed, pelleted cells were transferred to a fresh tube. 80 pl luciferase assay buffer were added and a reading in millivolts in a luminometer was taken immediately. The luciferase assay buffer was 20 mM Tricine, 1.07 mM (MgCO 3 ),Mg(OH),*5HO, 2.67 mM MgSO,, 0.1 mM EDTA, 33.3 mM DTT, 0.27 mM Coenzyme A, 0.47 mM Luciferin, 0.53 mM ATP. The results are shown in Figure 16, in which it can be seen that enhanced luciferase activity was observed in the presence of RA, RXRa and RARy, for both orientations of the promoter sequence, although enhancement appears to be greater for the sense construct.
30 EXAMPLE 12 Inhibition of P450RAI Induction by 4-Hydroxyphenylretinamide It has recently been reported that 4-hydroxyphenylretinamide (4-HPR) inhibits RA-induced RA catabolism by NB4 cells [Taimi, 1997]. It was suggested that 4-HPR may inhibit the cytochrome P450 enzyme(s) responsible for RA oxidation to competitively inhibit RA catabolism. However, any such enzymes were not identified, no explanation of the nature of 4- HPR inhibition of RA catabolism was provided, and no evidence of 4-HPR metabolism was observed.
SExperiments to determine the effect of 4-HPR on the induction of P450RAI were thus carried out. Figure 17 illustrates the ability of the synthetic retinoid, 4-HPR to inhibit -27the induction of P450RAI by RA. MCF7 cells were grown in culture in minimal essential medium (MEM) (Gibco) supplemented with 10 fetal calf serum, insulin (.01 mg/mL), MEM non-essential amino acids (as directed by the manufacturer Gibco), sodium pyruvate (500 nM), L-glutamine (2 mM) gentamycin (10 pg/mL), penicillin (5 pg/mL), streptomycin (5 pg/mL), and fungizone (200 ng/mL). MCF7 cells cultured in parallel were treated for 12 hours with: pM 4HPR; 1 pM 4HPR; 1 pM RA; 1 pM RA and 10 pM 4-HPR; or 1 pM RA and 1 pM 4-HPR.
At the end of the 12 hour treatment, total RNA was extracted from the cells using TRIzol reagent (as outlined by the manufacturer Gibco). P450RAI message in the total RNA preparations was analyzed by northern blot hybridization. The blot was reprobed with a probe corresponding to the GAPDH cDNA to control for equivalent loading of RNA in each lane of the blot. The results indicate that 4-HPR treatment alone does not induce the P450RAI message.
As expected, RA treatment of MCF7 cells results in a marked induction of P450RAI message following 12 hours of incubation. However, when cells are treated with RA in the presence of pM 4-HPR, there is a noticeable suppression of P450RAI induction.
EXAMPLE 13 P450RAI expression in the presence of retinoic acid Figure 18 shows a time course of expression of cytochrome p450 RAI following treatment of MCF7 breast epithelium adenocarcinoma cells with 1 pM all-trans retinoic acid. In this northern blot analysis, total RNA was _xtracted at the indicated time points 20 and transferred to nitrocellulose following electrophoresis in a 1% agarose, 0.66 M formaldehyde gel. The nitrocellulose was then probed with radioactively labelled human cytochrome p450RAI cDNA or GAPDH cDNA to control for equivalence of mRNA loading.
This shows that after 3 hours of incubation with retinoic acid, the MCF7 cells express high levels of P450RAI message and following 12 hours of exposure, the message declines sharply, possibly indicating that the metabolic activity of induced P450RAI protein is reducing the concentration of active retinoic acid in the surrounding medium. This strongly suggests that the induction of P450RAI in MCF7 cells forms an autoregulatory negative feedback loop.
EXAMPLE 14 P450RAI expression in the presence of retinoic acid and ketoconazole Figure 19 shows a time course of P450RAI mRNA expression in MCF7 cells similar to that described in Figure 18, except that the effect of the broad spectrum cytochrome p450 inhibitor ketoconazole on P450RAI expression was examined. In the absence of ketoconazole, lanes 1 to 5 in Figure 19 a time course similar to that shown in Figure 18 is shown. In cells which were exposed to 1 pM ketoconazole (replenished every 12 hours following initial treatment), cytochrome P450RAI message was detectable at high levels at 24 and 48 hour time points indicating that the breakdown of retinoic acid can be inhibited by a cytochrome P450 inhibitor and that P450RAI metabolism may be responsible for the sharp -28drop in P450RAI message in the absence of ketoconazole. The example illustrates a method of identifying P450RAI inhibitors.
EXAMPLE P450RAI expression in the presence of Am580 Figure 20 shows a time course of P450RAI mRNA expression in MCF7 cells similar to that described in Figure 18, except that a comparison was made between the retinobenzoic acid derivative Am580 and all-trans retinoic acid. Retinoic acid shows a typical time course of induction of P450RAI message, lanes 1 to 4, Figure 20. Am580 induces P450RAI message to levels comparable to those observed following treatment with retinoic acid. Notably, whereas P450RAI message has declined sharply between 24 and 48 hours, for retinoic acid treated cells, the levels of P450RAI in Am580 treated cells remains high at this time point. This indicates that the synthetic retinoid Am580 is resilient to metabolism in these cells and illustrates the utility of identifying such compounds for therapeutic use. For example, the resistance to retinoic acid treatment observed in acute promyelocytic leukemia due to increased retinoic acid metabolism [Warrell, 1994] might be circumvented by treatment with a metabolism-resistant retinoid. P450RAI protein may be a useful agent for screening for these types of compounds.
EXAMPLE 16 Monoclonal antibodies to P450RAI 20 Monoclonal antibodies (Mab's) specific for P450RAI are useful, for example, for diagnostic purposes such as for determining P450RAI protein levels in the identification of normal and tumor tissues which metabolize RA. To produce these antibodies, purified P450RAI protein is prepared. The human P450RAI protein is produced in bacterial cells as a fusion protein with glutathione-S-transferase using the vector pGEX2 (Pharmacia). This 25 permits purification of the fusion protein by GSH affinity chromatography. In another approach, P450RAI is expressed as a fusion protein with the bacterial m;atose binding domain. The fusion protein is thus recovered from bacterial extracts by passing the extract over an amylose resin column followed by elution of the fusion protein with maltose. For this fusion construct, the vector pMalC2, commercially available from New England Biolabs, is used. This vector has been used in the past, for example, to overexpress nuclear receptor proteins which were recovered in high yields for functional studies and the production of receptor specific antisera [Ohno, 1993]. The preparation of a second fusion protein is also useful in the preliminary screening of MAb's.
The generation of hybridomas expressing monoclonal antibodies recognizing P450RAI protein is carried out as follows: BALB/c mice are injected intraperitoneally with protein/adjuvant three times at one-month intervals, followed by a final injection into the tail vein shortly prior to cell fusion. Spleen cells are harvested and fused with NS-1 myeloma cells (American Type Culture Collection, Rockville, MD) using polyethylene glycol 4000 according to -29standard protocols [Kennett, 1979; Mirski, 1989]. The cell fusion process is carried out as described in more detail below.
The fused cells are plated into 96-well plates with peritoneal exudate cells and irradiated spleen cells from BALB/Ccmice as feeder layers and selection with hypoxanthine, aminopterin, and thymidine (HAT medium) is performed.
An ELISA assay is used as an initial screening procedure. 1-10 pg of purified P450RAI (cleaved from the fusion protein) in PBS is used to coat individual wells, and 50-100 pl per well of hybridoma supernatants is incubated. Horseradish peroxidase-conjugated antimouse antibodies are used for the colorimetric assay.
As a secondary screening, breast epithelial carcinoma MCF-7 cells are used as a source of P450RAI determinant. As indicated above, untreated MCF-7 cells exhibit no detectable expression of P450RAI message and no detectable RA metabolizing activity, whereas there is a rapid and striking induction of both in cells treated with RA. Untreated MCF- 7 cells thus serve as a negative control for background. MCF-7 cells are aliquotted into 96 well plates one day prior to assay. Cells are fixed and permeabilized with methanol. Hybridoma supernatants are added and fluorescein isothiocyanate (FITC) -conjugated anti-mouse antibodies are used for screening using a standard fluorescence microscope.
S* Positive hybridomas are cloned by limiting-dilution and grown to large-scale for freezing and antibody production. Various positive hybridomas are selected for usefulness in 20 western blotting and immunohistochemistry, as well as for cross reactivity with P450RAI proteins from different species.
The selected MAb's are useful for monitoring the levels of expression of P450RAI protein following RA treatment in cell culture and in tissues. P450RAI protein expression may be a prognostic indicator for determining whether a particular tumor will 25 respond to RA treatment. There is also a wide intersubject variability in baseline RA metabolism and there is evidence suggesting that subjects with a high rate of RA metabolism have a higher incidence of squamous or large cell cancers of the lung [Rigas, 1996]. Once useful antibodies are characterized, these antibodies are used to survey tumor tissue samples for P450RAI expression.
Protocol For Production of Mouse Hybridomas Fusion. Feeder cells (spleen and peritoneal exudate cells) are plated. 24 to 48 hours before fusion, mouse myeloma cells are taken off drug (8-azaguanine 20 pg/ml) and counted to ensure that there are at least 50 X 106 cells. 2 g of PEG 4000 are autoclaved in a glass tube for 15 minutes and maintained at 60°C for use or alternatively stored at room temperture and remelted in a 60°C water bath when needed.
BALB/c mice are immunized as per desired schedule. The final injection is given intravenously in the tail vein. Fusion of immun;zed spleen cells is carried out 3 or 4 days after the intravenous injection. Spleen from each animal is collected separately; eye sera for ELISA if desired.
A single cell suspension is prepared using a Teflon pestle and decanting connective tissue. The suspension is washed lx in serum-free medium. Each spleen has about 10 x 106 cells. The myeloma cells are collected, counted and wased in in serum-free medium.
The cells are then fused. A small beaker of water, and serum-free medium (37"C) are prepared and the PEG melted at 50-60°C. The immunized spleen cells and myeloma cells are mixed in a 50 ml TC tube (recommended ratios vary from 1:1 to 2:1) and the cells are washed once with serum-free medium. The supernatant is carefully discarded.
2.4 ml pre-warmed serum-free medium is added immediately with pipette to the melted PEG and mixed, maintaining the temperature at 37*C in beaker of warm water. The PEG should be light pink. If it is yellow, another aliquot should be used. 0.5-1.0 ml of PEG is added dropwise to the cell pellet over 1 minute with gentle rotation of the tube or gentle stirring to ensure mixing.
The tip of the addition pipette is placed directly over the cell pellet. The tube is swirled gently in 37*C water bath for 90 seconds with the blunt end of a 3 ml pipette tip and 10 ml warm serumfree medium added slowly over 6-10 minutes while rotating tube gently to bring the volume up to 20-50 ml. The tube is maintained at 37"C for at least 20 minutes to obtain cell fusion and then the cells are washed 2x with serum-free medium. The cells are centrifuged and gently resuspended in 100 ml of pre-warmed medium 10-20% FBS. 100 pi/well aliquotted in 96well plates. Assuming one spleen fused with 100 x 106 cells myeloma fusion partner, about 20 plates are needed. On the following day, 100 pl medium is removed and 100 ul 2 x HAT i. added. Feed with with 1 x HAT medium for 1 to 3 weeks, then feed with HT medium remove 1/2 HAT medium and replace with equal volume HT medium).
Preparation of Peritoneal Exudates and Spleen Feeder Cells. A sacrificed mouse is sprayed with 70% alcohol, skin is nicked and torn apart, with care being taken not to cut the peritoneum. The peritoneum is lifted with forceps and a needle is introduced; 5 ml of serumfree medium is slowly injected. The abdomen is massaged and the fluid is slowly sucked up, collected in a sterile tube and kept on ice. The volume is brought up to 5 ml. The spleen is obtained and placed in a sterile tube containing serum-free medium. The spleen is gently mashed with a sterile Teflon pestle. Clumps are allowed to settle and the cells are decanted into a clean tube, care being taken to avoid including connective tissue, in order to minimize fibroblast growth. The sample is irradiated at 4500 R. Cells are washed once with serum-free medium, placed into 96-well plates [one spleen/10 plates (approximately 2-5 x 105 cells/well) and peritoneal exudate cell suspension (PECS) 3 x 103 cells/well) in a total volume of 100 pl/well] and incubated at 37°C until ready to be used. They can also be stored in sterile tube overnight at 4°C.
Discussion It is possible to compare the zP450RAI, mP450RAI and hP450RAI sequences described above. Of the 492 amino acids of zP450RAI (SEQ ID NO:2), it is possible to align 334 amino acids with the 497 amino acids of hP450RAI (SEQ ID NO:4). See Figure 9. On this -31 basis, there is about 68% homology between the human and fish proteins. The degree of homology between the two amino acid sequences is slightly greater towards the C-terminus than in the N-terminal region. It also appears as though nucleic acid sequences encoding the conserved sequence Met-Lys-Arg-Gln-Lys (amino acid numbers 70 to 74 of zP450RAI) can be used as a probe to obtain corresponding proteins from cDNA libraries of other species. The amino acid identity between the human and mouse P450RAI sequences is greater than 93%.
As shown above, RA-induced expression of a protein by the cells described herein involves a regulatory sequence which is located upstream of the coding sequence of DNA that it controls. As it occurs in nature, the protein is P450RAI, whether in cells of the zebrafish, human, mouse or other organism. Such a cell can be modified by incorporating DNA encoding a different protein into the region of the gene which encodes P450RAI, as exemplified above with DNA encoding luciferase. An aspect of the present invention is thus a regulatory DNA sequence, responsive to the presence of RA, operably linked to a proteinencoding sequence and incorporated into a conventional genetically engineered proteinproducing cell. Production of the desired protein can be induced by exposure of the cell to RA.
Antisense nucleic acids or oligonucleotides (RNA or preferably DNA) that inhibit cellular RA-induced P450RAI production can be used to inhibit metabolism of RA by P450RAI [Monia, 1996]. Antisense oligonucleotides, typically 15 to 20 bases long, bind to the sense mRNA or pre mRNA region coding for the protein of interest, which can inhibit translation 20 of the bound mRNA to protein. The cDNA sequence encoding hP450RAI can thus be used to design a series of oligonucleotides which together span a large portion, or even the entire cDNA sequence. These oligonucleotides can be tested to determine which provides the greatest inhibitory effect on the expression of the protein (Stewart, 1996]. This can be done by exposing cells to the various oligonucleotides and measuring subsequent changes in hP450RAI activity or by using antibodies to screen for inhibition of P450RAI synthesis. The most suitable mRNA target sites include and 3'-untranslated regions as well as the initiation codon. Other regions might be found to be more or less effective. Alternatively, an antisense nucleic acid or oligonucleotide may bind to P450RAI DNA coding or regulatory sequences.
Rather than reducing RA metabolism by inhibiting P450RAI gene expression at the nucleic acid level, activity of the P450RAI protein may be directly inhibited by binding to an agent, such as, for example, a suitable small molecule or a monoclonal antibody.
The present invention thus includes a method of screening drugs for their effect on activity of a retinoic acid inducible protein. The method includes exposing the protein to a prospective inhibitor drug and determining the effect on protein activity. The measured activity might be hydroxylation of a retinoid, particularly all-trans retinoic acid, or hydroxylation of a retinoic acid, particularly all-trans retinoic acid, at the 4 position of the 3-ionone ring thereof.
For screening drugs for use in humans, hP450RAI itself is particularly useful for testing the effectiveness of such drugs. Prospective drugs could also be tested for inhibition of the activity of other P450 cytochromes, which are desired not to be inhibited. In this way, drugs which selectively inhibit hP450RAI over other P450s could be identified.
-32- Another system for screening for potential inhibitors of a P450RAI protein includes a stably transfected cell line having incorporated therein DNA of a reporter gene P-galactosidase, firefly luciferase, or the like) and of the P450RAI, in which expression of both genes is inducible by exposure of the cells to RA. Expression of the reporter gene provides a measure of the induction of the expression system and therefore provides an indication of the amount of RA present. Exposure of the cells to RA leads to RA metabolism and, with time, such metabolism leads to a decrease in the degree of induction which is indicated by the reporter protein. Exposure of the cells to RA in the presence of an agent that inhibits P450RAI metabolism of RA results in decreased RA metabolism, whereas exposure of the cells to RA in the presence of an agent that does not inhibit P450RAI metabolism of RA has no effect on RA metabolism. A comparison of expression of the reporter gene in the presence of RA alone and in the presence of both RA and a potential inhibitory drug thus gives a measure of the effectiveness of the drug in inhibiting metabolism of RA by the P450RAI protein.
One system for screening for potential inhibitiors of a P450RAI protein includes a cell line in which the endogenous P450RAI gene is not present or not functional or not expressed. In this cell line, a cytochrome P450RAI expression vector and an RA-inducible reporter gene are incorporated such that exposure of the cell line to RA results in metabolism of RA by the expressed P450RAI protein and a degree of induction of the reporter gene based ~on remaining active RA. The addition of an inhibitor of P450RAI will decrease the rate of 20 metabolism/degradation of RA and therefore increase the activation/induction of the RA sensitive reporter gene.
Given the high degree of conservation of the promoter regions of the mouse, human and zebrafish P450RAI promoter regions, it is likely that RA regulates P450RAI expression through a transcriptional mechanism involving the RARE conserved in the S 25 promoters of all three species. This is supported by studies which show the rapid and RAdependent expression of P450RAI in a number of cell lines. Since P450RAI message is induced so strongly a reporter gene may be a useful indicator of RA activity in vivo as well as in **vitro. Thus, the P450RAI promoter linked to a reporter gene provides a tool for screening retinoids or other compounds which have the ability to block or inhibit P450RAI induction. For 30 example, the P450RAI reporter gene could be stably or transiently introduced into a cell line such that when the cells are exposed to a certain level of retinoid or other agent, the concentration will be reflected in reporter gene activity. Such transfection assays can be carried out in a manner similar to those described by Petkovich et al., for example [Petkovich, 1987; Ohno, 1994].
The invention thus provides a system for screening potential inhibitors of RA catabolism by a P450RAI protein. The system includes a transfected cell line having incorporated therein DNA of a reporter gene, for example the luciferase gene exemplified above, in which expression of the reporter gene is inducible by exposure of the cells to RA. In this system, the P450RAI gene is omitted, that is the reporter gene is under the control of the native promoter for the P450RAI gene. Expression of the reporter gene provides a measure of -33the induction of the expression system and therefore provides an indication of the amount of mRNA produced in response to exposure of the cells to RA. Exposure of the cells to RA in the presence of an agent that inhibits induction of the expression system indicates that the agent is a potential inhibitor of RA catabolism, provides a measure of the effectiveness of the agent as a drug in inhibiting the expression of P450RAI gene and thus metabolism of RA.
There is the possibility that cellular refinoic acid-binding protein (CRABP) [Adamson, 1993] is involved in binding of a retinoid substrate to a P450RAI protein of the present invention. The effect of the presence of CRABP, derivatives, synthetic fragments or analogs thereof could thus be determined according to screening methods of the present invention; effectiveness of such agents in enhancing RA metabolism can also be determined.
It will of course be understood, without the intention of being limited thereby, that a variety of substitutions of amino acids is possible while preserving the structure responsible for retinoid metabolizing acitivity of the proteins disclosed herein. Conservative substitutions are described in the patent literature, as for example, in United States Patent No.
5,264,558. It is thus expected, for example, that interchange among non-polar aliphatic neutral amino acids, glycine, alanine, proline, valine and isoleucine, would be possible. Likewise, substitutions among the polar aliphatic neutral amino acids, serine, threonine, methionine, asparagine and glutamine could possibly be made. Substitutions among the charged acidic amino acids, aspartic acid and glutamic acid, could probably be made, as could substitutions 20 among the charged basic amino acids, lysine and arginine. Substitutions among the aromatic amino acids, including phenyialanine, histidine, tryptophan and tyrosine would also likely be possible. These sorts of substitutions and interchanges are well known to those skilled in the art. Other substitutions might well be possible. Of course, it would also be expected that the greater the percentage of homology, sequence similarity, of a variant protein with a 25 naturally occuring protein, the greater the retention of metabolic activity. Of course, as protein variants having the activity of P450RAI as described herein are intended to be within the scope of this invention, so are nucleic acids encoding such variants.
A further advantage may be obtained through chimeric forms of the protein, as known in the art. A DNA sequence encoding the entire protein, or a portion of the protein, could thus be linked, for example, with a sequence coding for the C-terminal portion of E. coli B-galactosidase to produce a fusion protein. GST-CYPRAI fusion proteins are described in the
OO
above examples. An expression system for human respiratory syncytial virus glycoproteins F and G is described in United States Patent No. 5,288,630 issued February 22, 1994 and references cited therein, for example.
A recombinant expression vector of the invention can be a plasmid, as described above. The recombinant expression vector of the invention further can be a virus, or portion thereof, which allows for expression of a nucleic acid introduced into the viral nucleic acid. For example, replication defective retroviruses, adenoviruses and adeno-associated viruses can be used.
-34- The invention provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operatively linked to a regulatory sequence in a manner which allows for expression, by transcription of the DNA molecule, of an RNA molecule which is antisense to the nucleotide sequence of SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:31. Regulatory sequences operatively linked to the antisense nucleic acid can be chosen which direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance a viral promoter and/or enhancer, or other regulatory sequences can be chosen which direct tissue or cell type specific expression of antisense RNA.
The recombinant expression vectors of the invention can be used to make a transformant host cell including the recombinant expression vector. The term "transformant host cell" is intended to include prokaryotic and eukaryotic cells which have been transformed or transfected with a recombinant expression vector of the invention. The terms "transformed with", "transfected with", "transformation" and "transfection" are intended to encompass introduction of nucleic acid a vector) into a cell by one of many possible techniques known in the art. Prokaryotic cells can be transformed with nucleic acid by, for example, electroporation or calcium-chloride mediated transformation. Nucleic acid can be introduced into mammalian cells via conventional techniques such as calcium phosphate or calcium S. chloride coprecipitation, DEAE-dextran-mediated transfection, lipofection, electroporation or 20 microinjection. Suitable methods for transforming and transfecting host cells are known [Sambrook, 1989].
The number of host cells transformed with a recombinant expression vector of the invention by techniques such as those described above will depend upon the type of •recombinant expression vector used and the type of transformation technique used. Plasmid vectors introduced into mammalian cells are integrated into host cell DNA at only a low S* frequency. In order to identify these integrants, a gene that contains a selectable marker (e.g.
resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Preferred selectable markers include those which confer resistance to certain drugs, such as G418 and hygromycin. Selectable markers can be introduced on a separate plasmid from the nucleic acid of interest or, preferably, are introduced on the same plasmid. Host cells transformed with one or more recombinant expression vectors containing a nucleic acid of the invention and a gene for a selectable marker can be identified by selecting for cells using the selectable marker. For example, if the selectable marker encodes a gene conferring neomycin resistance (such as pRc/CMV), transformant cells can be selected with G418. Cells that have incorporated the selectable marker gene will survive, while the other cells die.
Certain nucleic acids of the invention encode proteins which "have biological activity of a cytochrome P450RAI". The biological activity of a cytochrome P450RAI is the ability to oxidize a retinoid. Such activity can be tested for as described above.
The invention provides purified proteins having biological activity of P450RAI.
The terms "isolated" and "purified" each refer to a protein substantially free of cellular material or culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized. In certain preferred embodiments, the protein having biological activity of P450RAI comprises an amino acid sequence identified as SEQ ID NO:2 or SEQ ID NO:4 or SEQ ID NO:32. Alternatively, preferred proteins encoded by a nucleic acid comprising the nucleotide sequence identified as SEQ ID NO:3 or SEQ ID NO:5 or SEQ ID NO:31, as defined above, are encompassed by the invention. Furthermore, proteins having biological activity of P450RAI that are encoded by nucleic acids which hybridize under stringent conditions, as discussed above, to a nucleic acid comprising a nucleotide sequence identified as SEQ ID NO:3 or SEQ ID NO:5 or SEQ ID NO:31 are encompassed by the invention.
P450RAIs of the invention can be obtained by expression in a suitable host cell using techniques known in the art. Suitable host cells include prokaryotic or eukaryotic organisms or cell lines, for example, yeast, E. coli, insect cells and COS 1 cells. The recombinant expression vectors of the invention, described above, can be used to express a protein having P450RAI activity in a host cell in order to isolate the protein. The invention provides a method of preparing an purified protein of the invention comprising introducing into a host cell a recombinant nucleic acid encoding the protein, allowing the protein to be expressed in the host cell and isolating and purifying the protein. Preferably, the recombinant nucleic acid is a recombinant expression vector. Proteins can be isolated from a host cell expressing the protein and purified according to standard procedures of the art, including ammonium sulfate .20 precipitation, column chromatography ion exchang:, gel filtration, affinity chromatography, etc.), electrophoresis, and ultimately, crystallization (see generally, "Enzyme Purification and *Related Techniques", Methods in Enzymology. 22,233-577 (1971)].
Alternatively, the protein or parts thereof can be prepared by chemical synthesis using techniques well known in the chemistry of proteins such as solid phase 25 synthesis [Merrifield, 1964], or synthesis in homogeneous solution [Houbenwcyl, 1987].
The protein of the invention, or portions thereof, can be used to prepare antibodies specific for the proteins. Antibodies can be prepared which bind to a distinct epitope in an unconserved region of a particular protein. An unconserved region of the protein is one which does not have substantial sequence homology to other proteins, for example other 30 members of the P450 family of cytochromes. Conventional methods can be used to prepare the antibodies. For example, by using a peptide of a P450RAI protein, polyclonal antisera or monoclonal antibodies can be made using standard methods. As demonstrated in Example 16, a mammal, a mouse, hamster, or rabbit) can be immunized with an immunogenic form of the peptide which elicits an antibody response in the mammal. Techniques for conferring immunogenicity on a peptide include conjugation to carriers or other techniques well known in the art. For example, the peptide can be administered in the presence of adjuvant.
The progress of immunization can be monitored by detection of antibody titers in plasma or serum. Standard ELISA or other immunoassay can be used to assess the levels of antibodies.
Following immunization, antisera can be obtained and, if desired, polyclonal antibodies isolated from the sera.
-36- To produce monoclonal antibodies, antibody producing cells (lymphocytes) can be harvested from an immunized animal and fused with myeloma cells by standard somatic cell fusion procedures, thus immortalizing these cells and yielding hybridoma cells.
Such techniques are well known in the art. For example, the hybridoma techrique originally developed by Kohler and Milstein [Kohler, 1975] as well as other techniques such as the human B-cell hybridoma technique [Kozbor, 1983], the EBV-hybridoma technique to produce human monoclonal antibodies [Cole, 1985], and screening of combinatorial antibody libraries [Huse, 1989]. Hybridoma cells can be screened immunochemically for production of antibodies specifically reactive with the peptide, and monoclonal antibodies isolated.
The term antibody as used herein is intended to include fragments thereof which are also specifically reactive with a protein having the biological activity of P450RAI, or a peptide fragment thereof; Antibodies can be fragmented using conventional techniques and the fragments screened for utility in the same manner as described above for whole antibodies.
For example, F(ab') 2 fragments can be generated by treating antibody with pepsin. The resulting F(ab') 2 fragment can be treated to reduce disulfide bridges to produce Fab' fragments.
It is also known in the art to make chimeric antibody molecules with human constant regions. See, for example, Morrison et Takeda et al., Cabilly et al., Boss et al., Tanaguchi etal., Teng etal. [Morrison, 1985; Takeda, 1985; Cabilly; Boss; Tanaguchi; Teng, 1982], European Patent Publication 0173494, United Kingdom Patent GB 2177096B, PCT 20 Publication W092/06193 and EP 0239400. It is expected that such chimeric antibodies would be less immunogenic in a human subject than the corresponding non-chimeric antibody.
Another method of generating specific antibodies, or antibody fragments, reactive against protein having the biological activity of a P450RAI, or a peptide fragment thereof, is to screen expression libraries encoding immunoglobulin genes, or portions thereof, expressed in bacteria, with peptides produced from the nucleic acid molecules of the present invention. For example, complete Fab fragments, VH regions and FV regions can be expressed in bacteria using phage expression libraries. See for example Ward et al., Huse et al., and McCafferty et al. [Ward, 1989; Huse, 1989; McCaffei.y, 1990]. Screening such libraries with, for example, a P450RAI peptide can identify immunoglobulin fragments reactive with P450RAI. Alternatively, the SCID-hu mouse developed by Genpharm can be used to produce antibodies, or fragments thereof.
The polyclonal, monoclonal or chimeric monoclonal antibodies can be used to S* detect the proteins of the invention, portions thereof or closely related isoforms in various biological materials, for example they can be used in an ELISA, radioimmunoassay or histochemical tests. Thus, the antibodies can be used to quantify the amount of a P450RAI protein of the invention, portions thereof or closely related isoforms in a sample in order to determine the role of P450RAI proteins in particular cellular events or pathological states.
Using methods described hereinbefore, polyclonal, monoclonal antibodies, or chimeric monoclonal antibodies can be raised to nonconserved regions of P450RAI and used to distinguish a particular P450RAI from other proteins.
-37- The polyclonal or monoclonal antibodies can be coupled to a detectable substance or reporter system. The term "coupled" is used to mean that the detectable substance is physically linked to the antibody. Suitable detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, p-galactosidase, and acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride and phycoerythrin; an example of a luminescent material includes luminol; and examples of suitable radioactive material include 1251; 11I, "S and 3 H. In a preferred embodiment, the reporter system allows quantitation of the amount of protein (antigen) present.
Such an antibody-linked reporter system could be used in a method for determining whether a fluid or tissue sample of a subject contains a deficient amount or an excessive amount of the protein. Given a normal threshold concentration of such a protein for a given type of subject, test kits could thus be developed.
The present invention allows the skilled artisan to prepare bispecific antibodies and tetrameric antibody complexes. Bispecific antibodies can be prepared by forming hybrid hybridomas [Staerz, 1986a 20 The present invention includes three types of compounds related to retinoids: those that inhibit enzymatic activity of P450RAI, thereby inhibiting metabolism of RA; those retinoids that evade metabolism by P450RAI, for example Am580, shown above; and those compounds that repress induction of P450RAI gene expression, for example 4-HPR, as shown above.
Compositions of the invention are administered to subjects in a biologically compatible form suitable for pharmaceutical administration in vivo. By "biologically compatible from suitable for administration in vivo" is meant a form of the composition to be administered in which any toxic effects are outweighed by the therapeutic effects of the composition. The term "subject" is intended to include living organisms in which a desired therapeutic response can be elicited, e.g. mammals. Examples of subjects include human, dogs, cats, mice, rats and transgenic species thereof. Administration of a therapeutically active amount of the therapeutic compositions of the present invention is defined as an amount effective, at dosages and for periods of time necessary to achieve the desired result. For example, a therapeutically active amount of a compound that inhibits catabolism of RA by a P450RAI protein may vary according to factors such as the disease state, age, sex, and weight of the individual, as well as target tissue and mode of delivery. Dosage regimes may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation.
Compounds of the present invention, such as those that are found to inhibit metabolism of RA by P450RAI enzymes and that are useful as anticancer agents and in the -38treatment, amelioration, or prevention of skin disorders for which retinoic acid is useful, for example, may be used topically. In this regard they may be included in compositions for therapy in animals, including humans, for premalignant epithelial cell lesions, as a prophylaxis against tumor promotion in epithelial cells and treatment for dermatoses such as ichthyoses, follicular disorders, benign epithelial disorders, and other proliferative skin diseases, such as acne, psoriasis, eczema, atopic dermatitis, nonspecific dermatitis and the like.
Topical compositions are usually formulated with a pharmaceutically acceptable carrier in liquid, semi-solid or solid form. A pharmaceutically acceptable carrier is a material that is nontoxic and generally inert and does not affect the functionality of the active ingredients adversely. Such materials are well known and include those materials sometimes referred to as diluents or vehicles (excipients) in the pharmaceutical formulation art. The carrier may be organic or inorganic in nature. Examples of pharmaceutically acceptable carriers are water, gelatin, lactose, starch, mineral oil, cocoa butter, dextrose, sucrose, sorbitol, mannitol, gum, acacia, alginates, cellulose, talc, magnesium stearate, polyoxyethylene sorbitan monolaurate, and other commonly used pharmaceutical carriers. In addition to an active ingredient and carrier, the formulation may contain minor amounts of additives such as flavoring agents, coloring agents, thickening or gelling agents, emulsifiers, wetting agents, buffers, stabilizers, and preservatives such as antioxidants.
Certain compositions may be administered enterally. For oral administration, suitable forms are, for example, tablets, pills, syrups, suspensions, emulsions, solutions, powders and granules.
As anti-tumor agents or as part of an anti-tumor formulation, for example, compounds of the present invention can be used in a similar manner to retinoids used for treating various tumours, such as all-trans retinoic acid. The dose to be administered, whether a single dose, multiple does or daily dose, will of course vary with the particular compound employed because of the varying potency of the active ingredient, the chosen route of administration, the size of the recipient, the type of tumor, and the nature of the patient's condition. The dosage to be administered is not subject to definite bounds, but it will usually be an effective amount, or the equivalent on a molar basis of the pharmacologically active free form produced from a dosage formulation upon the metabolic release of the active drug to achieve its desired pharmacological and physiological effects. An oncologist skilled in the art of •cancer treatment will be able to ascertain without undue experimentation, appropriate protocols for the effective administration of the compounds of this present invention.
Nucleic acids which encode proteins having biological activity of a P450RAI protein can be used to generate either transgenic animals or "knock out" animals which, in turn, are useful in the development and screening of therapeutically useful reagents. A transgenic animal a mouse) is an animal having cells that contain a transgene, which transgene was introduced into the animal or an ancestor of the animal at a prenatal, an embryonic stage.
A transgene is. a DNA which is integrated into the genome of a cell from which a transgenic animal develops. In one embodiment, a human P450RAI cDNA, comprising the nucleotide -39sequence shown in SEQ ID NO:5, or an appropriate variant or subsequence thereof, can be used to generate transgenic animals that contain cells which express human P450RAI protein.
Methods for generating transgenic animals, particularly animals such as mice, have become conventional in the art are described, for example, in U.S. Patent Nos. 4,736,866 and 4,870,009. In a preferred embodiment, plasmids containing recombinant molecules of the invention are microinjected into mouse embryos. In particular, the plasmids are microinjected into the male pronuclei of fertilized one-cell mouse eggs; the injected eggs are transferred to pseudo-pregnant foster females; and, the eggs in the foster females are allowed to develop to term. [Hogan, 1986]. Alternatively, an embryonal stem cell line can be transfected with an expression vector comprising nucleic acid encoding a protein having P450RAI activity, and cells containing the nucleic acid can be used to form aggregation chimeras with embryos from a suitable recipient mouse strain. The chimeric embryos can then be implanted into a suitable pseudopregnant female mouse of the appropriate strain and the embryo brought to term.
Progeny harboring the transfected DNA in their germ cells can be used to breed uniformly transgenic mice.
Typically, particular cells would be targeted for P450RAI transgene incorporation by use of tissue specific enhancers operatively linked to the P450RAI encoding gene. For example, promoters and/or enhancers which direct expression of a gene to which they are operatively linked preferentially in cardiac muscle cells can be used to create a 20 transgenic animal which expresses a P450RAI protein preferentially in cardiac muscle tissue.
Examples of suitable promoters and enhancers include those which regulate the expression of the genes for cardiac myosin and cardiac actin. Transgenic animals that include a copy of an P450RAI transgene introduced into the germ line of the animal at an embryonic stage can also be used to examine the effect of increased P450RAI expression in various tissues.
The pattern and extent of expression of a recombinant molecule of the invention in a transgenic mouse is facilitated by fusing a reporter gene to the recombinant molecule such that both genes are co-transcribed to form a polycistronic mRNA. The reporter gene can be introduced into the recombinant molecule using conventional methods such as those described in Sambrook et al., [Sambrook,1989]. Efficient expression of both cistrons of the polycistronic mRNA encoding the protein of the invention and the reporter protein can be achieved by inclusion of a known internal translational initiation sequence such as that present in poliovirus mRNA. The reporter gene should be under the control of the regulatory sequence of the recombinant molecule of the invention and the pattern and extent of expression of the gene encoding a protein of the invention can accordingly be determined by assaying for the phenotype of the reporter gene. Preferably the reporter gene codes for a phenotype not displayed by the host cell and the phenotype can be assayed quantitatively. Examples of suitable reporter genes include lacZ (p-galactosidase), neo (neomyciin phosphotransferase), CAT (chloramphenicol acetyltransferase) dhfr (dihydrofolate reductase), aphlV (hygromycin phosphotransferase), lux (luciferase), uidA (p-glucuronidase). Preferably, the reporter gene is lacZ which codes for p-galactosidase. p-galactosidase can be assayed using the lactose analogue X-gal (5-bromo-4-chloro-3-indolyl-b-D-galactopyranoside) which is broken down by P-galactosidase to a product that is blue in color [Old].
Although experimental animals used in the preferred embodiment disclosed are mice, the invention should not be limited thereto. It can be desirable to use other species such as, for example, rats, hamsters, rabbits and sheep.
The transgenic animals of the invention can be used to investigate the molecular basis of RA metabolism. The transgenic animals of the invention can also be used to test substances for the ability to prevent, slow or enhance RA metabolism. A transgenic animal can be treated with the substance in parallel with an untreated control transgenic animal.
Cells from the transgenic animals of the invention can be cultured using standard tissue culture techniques. In particular, cells carrying the recombinant molecule of the invention can be cultured and used to test substances for the ability'to prevent, slew or enhance RA metabolism.
Additionally, the non-human homologues of genes encoding proteins having P450RAI activity can be used to construct a "knock out" animal which has a defective or altered P450RAI gene. For example, with established techniques, a portion of murine genomic P450RAI DNA an exon), can be deleted or replaced with another gene, such as a gene encoding a selectable marker which can be used to monitor integration. The altered P450RAI DNA can then be transfected into an embryonal stem cell line. The altered P450RAI DNA will homologously recombine with the endogenous P450RAI gene in certain cells and clones containing the altered gene can be selected. Cells containing the altered gene are injected into S* a blastocyst of an animal, such as a mouse, to form aggregation chimeras as described for transgenic animals. Chimeric embryos are implanted as described above. Transmission of the altered gene into the germline of a resultant animal can be confirmed using standard techniques and the animal can be used to breed animals having an altered P450RAI gene in every cell [Lemoine, 1996]. Accordingly, a knockout animal can be made which cannot express a functional P450RAI protein. Such a knockout animal can be used, for example, to test the effectiveness of an agent in the absence of a P450RAI protein.
The antisense nucleic acids and oligonucleotides of the invention are useful for inhibiting expression of nucleic acids mRNAs) encoding proteins having P450RAI activity.
Since proteins having P450RAI activity are associated with metabolism of agents which can act on the cell, RA, decreasing expression of such proteins can increase sensitivity of the cell to such agents. Antisense nucleic acids can be introduced into a drug resistant cell in culture to inhibit P450RAI expression. One or more antisense nucleic acids, such as oligonucleotides, can be added to cells in culture media, typically, for example, at 200 pg/ml.
The antisense nucleic acids of the invention, or oligonucleotides thereof, can thus be used in gene therapy to correct or prevent retinoic acid or other retinoid resistance in a subject. For example, antisense sequences can be used to render retinoic acid or other retinoid resistant malignant cells sensitive to chemotherapeutic agents. Administration of -41antisense nucleic acids to a subject may be most effective when the antisense nucleic acid is contained in a recombinant expression vector which allows for continuous production of antisense RNA. Recombinant molecules comprising an antisense nucleic acid or oligonucleotide thereof, can be directly introduced into tissues, including lung tissue in vivo, using delivery vehicles such as liposomes, retroviral vectors, adenoviral vectors and DNA virus vectors. A delivery vehicle can be chosen which can be targeted to a cell of interest in the subject a retinoid resistant tumor cell). Antisense nucleic acids can also be introduced into isolated cells, such as those of the haematopoietic system, ex vivo using viral vectors or physical techniques such as microinjection and electroporation or chemical methods such as coprecipitation and incorporation of DNA into liposomes, and such cells can be returned to the donor. Recombinant molecules can be delivered in the form of an aerosol or by lavage.
Accordingly, the invention provides a method for inhibiting retinoic acid or other retinoid resistance of a resistant cell by introducing into the resistant cell a nucleic acid which is antisense to a nucleic acid which encodes the protein identified as SEQ ID NO:2, SEQ ID NO:32, or particularly, the in case of human cells SEQ ID NO:4.
The nucleic acids of the invention can further be used to design ribozymes which are capable of cleaving a single-stranded nucleic acid encoding a protein having P450RAI activity, such as an mRNA. A catalytic RNA (ribozyme) having ribonuclease activity can be designed which has specificity for a P450RAI-encoding mRNA based upon the 20 sequence of a nucleic acid of the invention. For example, a derivative of a Tetrahymena L- 19IVS RNA can be constructed in which the base sequence of the active site is complementary to the base sequence to be cleaved in a P450RAI-encoding mRNA. [Cech a and b).
Alternatively, a nucleic acid of the invention could be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules [Bartel, 1993].
The isolated nucleic acids and antisense nucleic acids of the invention can be used to construct recombinant expression vectors as described previously. These recombinant expression vectors are then useful for making transformant host cells containing the recombinant expression vectors, for expressing protein encoded by the nucleic acids of the S°invention, and for isolating proteins of the invention as described previously. The isolated nucleic acids and antisense nucleic acids of the invention can also be used to construct transgenic and knockout animals as described previously.
The isolated proteins of the invention are useful for making antibodies reactive against proteins having P450RAI activity, as described previously. Alternatively, the antibodies of the invention can be used to isolate a protein of the invention by standard immunoaffinity techniques. Furthermore, the antibodies of the invention, including bispecific antibodies are useful for diagnostic purposes.
Molecules which bind to a protein comprising an amino acid sequence shown in SEQ ID NO:4 can also be used in a method for killing a cell which expresses the protein, wherein the cell.takes up the molecule. Preferably, the cell is a tumor cell. Destruction of such cells can be accomplished by labeling the molecule with a substance having toxic or -42therapeutic activity. The term "substance having toxic or therapeutic activity" as used herein is intended to include molecules whose action can destroy a cell, such as a radioactive isotope, a toxin diphtheria toxin or ricin), or a chemotherapeutic drug, as well as cells whose action can destroy a cell, such as a cytotoxic cell. The molecule binding to the P450RAI can be directly coupled to a substance having a toxic or therapeutic activity or may be indirectly linked to the substance. In one example, the toxicity of the molecule taken up by the cell is activated by P450RAI protein.
The invention also provides a diagnostic kit for identifying tumor cells comprising a molecule which binds to a protein comprising an amino acid sequence shown in SEQ ID NO:4, for example, for incubation with a sample of tumor cells; means for detecting the molecule bound to the protein, unreacted protein or unbound molecule; means for determining the amount of protein in the sample; and means for comparing the amount of protein in the sample with a standard. Preferably, the molecule is a monoclonal antibody. In some embodiments of the invention, the detectability of the molecule which binds to P450RAI is activated by said binding change in fluorescence spectrum, loss of radioisotopic label).
The diagnostic kit can also contain an instruction manual for use of the kit.
The invention further provides a diagnostic kit for identifying tumor cells comprising a nucleotide probe complementary to the sequence, or an oligonucleotide fragment thereof, shown in SEQ ID NO:3, for example, for hybridization with mRNA from a sample of S 20 tumor cells; means for detecting the nucleotide probe bound to mRNA in the sample with a standard. The diagnostic kit can also contain an instruction manual for use of the kit.
Those skilled in the art will know, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention °described herein. Such equivalents are intended to be encompassed by the following claims.
-43- References Particulars of references cited above are given below. All of the listed references are incorporated herein by reference.
Adamson, P. Boylan, J. Balis, F. Murphy, R. Godwin, K. Gudas, L. J. and Poplack, D. G. (1993). Time course of induction of metabolism of all-trans retinoic acid and the up-regulation of cellular retinoic acid-binding protein. Cancer Research 53, 472-476.
Achkar, Derguini, Blumberg, Langston, Arthur, A. Speck, Evans, R. M., Bolado, Jr., J. Nakanishi, K. and Buck, J. (1996) 4-Oxoreinol, a new natural ligand and transactivator of the retinoic acid receptors. Proc. Natl. Acad. Sci. USA 93, 4879-84.
Akimenko, M. A. and Ekker, M. (1995a). Anterior duplication of the Sonic hedgehog expression pattern in the pectoral fin buds of zebrafish treated with retinoic acid. Developmental Biology 170, 243-7.
Akimenko, M. Johnson, S. Westerfield, M. and Ekker, M. (1995b). Differential induction of four msx homeobox genes during fin development and regeneration in zebrafish.
Development 121, 347-57.
Akiyoshi-Shibata, Sakaki, Ohyama, Noshiro, Okuda, K. and Yabusaki, Y. (1994).
Further oxidation of hydroxycalcidiol by calcidiol 24-hydroxylase. A study with the mature enzyme expressed in Escherichia coli. European Journal of Biochemistry 224, 335-43.
Bartel, D. and Szostak, J.W. (1993). Science 261, 1411-1418.
Blaner, W. (1994). Retinol and retinoic acid metabolism. In: The Retinoids. Sporn, Roberts, A. and Goodman, Editors) Raven Press, Inc.: New York.
Bligh, E. G. and Dyer, W. J. (1957). A rapid method of total lipid extraction and purification.
Canadian Journal of Biochemistry 37, 911-917.
Blumberg, Bolado, Jr., Derguini, Craig, A. Moreno, T. Chakravarti, Heyman, R. Buck, J. and Evans, R. M. (1996) Novel retinoic acid receptor ligands in Xenopus embryos. Proc. Natl. Acad. Sci. USA 93, 4873-78.
Boss et al., United States Patent No. 4,816,397.
Boylan, J. Lufkin, Achkar, C. Taneha, Chambon, P. and Gudas, L. J. (1995).
Targeted Disruption of Retinoic Acid Receptor a (RARa) and RARg Results in Receptor- Specific Alterations in Retinoic Acid-Mediated Differentiation and Retinoic Acid Metabolism. Mol.
Cell Biol. 15, 843-851.
Boyle, A.L. etal. (1992). Genomics 12,106-15.
Cabilly et al. United States Patent No. 4,816,567.
Cech et al., United States Patent No. 4,987,071.
Cech et al., United States Patent No. 5,116,742.
Chambon, P. (1995). The molecular and genetic dissection of the retinoid signaling pathway.
[Review]. Recent Progress in Hormone Research 50, 317-32.
Chen, K. S. and DeLuca, H. F. (1995). Cloning of the human 1 alpha,25-dihydroxyvitamin D-3 24-hydroxylase gene promoter and identification of two vitamin D-responsive elements.
Biochimica et Biophysica Acta 1263,1-9.
-44- Chomienne, Fenaux and Degos, L. (1996). Retinoid differentiation therapy in promyelocytic leukemia. FASEB J. 1025-1030.
Chytil, F. (1984). Retinoic acid: Biochemistry, toxicology, pharmacology, and therapeutic use.
Pharmacol. Rev. 36, 93-99.
Cole et al. (1985). Monoclonal Antibodies in Cancer Therapy. Allen R. Bliss, Inc.
Costaridis, Horton, Zeitlinger, Holder, N. and Maden, M. (1996). Endogenous Retinoids in the Zebrafish Embryo and Adult. Developmental Dynamics 205, 41-51.
Creech Kraft, Schuh, Juchau, M. R. and Kimelman, D. (1994). Temporal distribution, localization and metabolism of all-trans retinol, didehydroretinol and all-trans retinal during Xenopus development. Biochem. J. 301, 111-119.
De Coster, Wouters, W. and Bruynseels, J. (1996). P450-dependent enzymes as targets for prostate cancer therapy. J. Ster. Biochem. Mol. Biol. 56, 133-43.
Duell, E. Astrom, Griffiths, C. Chambon, P. and Voorhees, J. J. (1992). Human skin levels of retinoic acid and cytochrome p-450-derived 4-hydroxyretinoic acid after topical application of retinoic acid in vivo compared to concentrations required to stimulate retinoic acid receptor-mediated transcription in vitro. Journal of Clinical Investigation 90, 1269-74.
Duell, Astrom, Kang, Griffiths, C. E. M. and Voorhees, J. (1994). All-trans, 9-cis and 13-cis retinoic acid each induce a cytochrome P450 4-retinoic acid hydroxylase which causes all-trans but not 9-cis or 13-cis retinoic acid to self-metabolize. Society for Investigative Dermatology Abstracts 102, 641.
Fiorella, P. Giguere, V. and Napoli, J. L. (1993). Expression of Cellular Retinoic Acid-binding Protein (Type II) in Escherichia coli. The Journal of Biological Chemistry 268, 21545-21552.
Formelli, Barua, A. and Olson,J. (1996). Bioactivities of N-(4-hydroxyphenyl) retinimide and retinoyl B-glucuronide. FASEB J. 10, 1014-1024.
25 Frolik, C. Roberts, A. Tavela, T. Roller, P. Newton, D. L. and Sporn, M. B. (1979).
Isolation and identification of 4-hydroxy- and 4-oxoretinoic acid. In vitro metabolites of all-trans retinoic acid in hamster trachea and liver. Biochemistry 18, 2092-7.
Gotoh, 0. and Fujii-Kuriyama, Y. (1989). Evolution, structure, and gene regulation of cytochrome P-450.
Green, Issemann, I. and Sheer, E. (1988). A versatile in vivo and in vitro eukaryotic expression vector for protein engineering. Nucleic Acids Research 16, 369-370.
Gudas, Sporn, M. and Roberts, A. (1994). Cellular biology and biochemistry of the retinoids.
In: The Retinoids. Sporn, Roberts, A. and Goodman, Editors) Raven Press, Inc.: New York.
35 Heng, H. and Tsui, L-C. (1993). Chromosome 102, 325-32.
Hogan, B. et al., (1986). A Laboratory Manual, Cold Spring Harbor, Cold Spring Harbor Laboratory.
Hong, W. (1994). Retinoids and human cancer. In: The Retinoids. Sporn, Roberts, A. and Goodman, Editors) Raven Press, Inc.: New York.
Houbenwcyl, (1987). Methods of Organic Chemistry, ed. E. Wansch. Vol. 15 I and II. Thieme, Stuttgart.
Hozumi, N and Sandhu, J.S. (1993). Recombinant antibody technology, its advent and advances. Cancer Invest. 11, 714-723.
Huse etal., (1989). Science 246, 1275 -1281.
Jones, B. Ohno, C. Allenby, Boffa, Levin, A. Grippo, J. F. and Petkovich, M.
(1995). New Retinoid X Receptor Subtypes in Zebra Fish (Danio rerio) Differentially Modulate Transcription and Do Not Bind 9-cis Retinoic Acid. Mol-Cell Biol. 15, 5226-5234.
Kennett, R. (1979). Cell fusion. Methods Enzymol. 58, 345-359.
Kohler and Milstein. (1975). Nature 256, 495-497.
Kozbor et al. (1983). Immunol. Today 4, 72.
Lammer, E. Chen, D. Hoar, R. Agnish, N. Benke, P. Braun, J. Curry, C. J., Fernhoff, P. Grix, A. Lott, I. T. et al. (1985). Retinoic acid embryopathy. New England Journal of Medicine 313, 837-41.
Lansdorp, United States Patent 4,868,109.
Lemoine, N.R. and Cooper, D.N. (1996). Gene Therapy, Human Molecular Genetics Series, BIOS Scientific Publishers, Oxford, U.K.
Leo et al. (1989). Metabolism of retinol and retinoic acid by human liver cytochrome P45011C8.
Arch. Biochem. Biophys. 269, 305-312.
Liang, P. and Pardee, A. B. (1992). Differential display of eukaryotic messenger RNA by means of the polymerase chain reaction. Science 257, 967-71.
Lichter, P. et al. (1990). High Resolution Mapping of Chromosome 11 by in situ hybridization by cosmid clones. Science 247, 64-9.
Lippman. S. Heyman, R. Kurie, J. Benner, S. E. and Hong, W. K. (1995). Retinoids and chemoprevention: clinical and basic studies. J. Cellular Biochem. Supplement 22, 1-10.
Lotan, R. M. (1995). Squamous differentiation and retinoids. Cancer Treat. Res. 74, 43-72.
Lotan, R. (1996). Retinoids in Cancer Chemoprevention. Faseb J. 10, 1031-1039.
Maden, M. and Holder, N. (1992). Retinoic acid and development of the central nervous system. [Review]. Bioessays 14, 431-8.
Makin, Lohnes, Byford, Ray, R. and Jones, G. (1989). Target cell metabolism of 1,25-dihydroxyvitamin D3 to calcitroic acid. Evidence for a pathway in kidney and bone involving 24-oxidation. Biochemical Journal 262, 173-80.
Maniatis et al. (1982) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York.
Mangelsdorf, D. J. and Evans, R. M. (1995). The RXR Heterodimers and Orphan Receptors.
Cell 83, 841-850.
Merrifield, (1964]. J. Am. Chem. Assoc. 85, 2149-2154.
McCafferty et al., (1990). Nature 348, 552-554.
Mirski, S. and Cole, S. P. C. (1989). Antigens associated with multidrug resistance in H69AR, a small cell lung cancer cell line. Cancer Res. 49, 5719-5724.
-46- Monia, B. Johnston, J. Geiger, Muller, M. and Fabbro, D. (1996). Antitumor activity of a phosphorothioate antisense oligodeoxynucleotide targeted against C-rafkinase. Nature Medicine 2, 668-75.
Moon, R. Mehta, R.G. and Rao, K.V.N. (1994). Retinoids and cancer in experimental animals. In: The Retinoids. Sporn, Roberts, A. and Goodman. Editors) Raven Press, Inc.: New York.
Morriss-Kay, G. (1993). Retinoic acid and craniofacial development: molecules and morphogenesis. [Review]. Bioessays 15, 9-15.
Morriss-Kay, G. M. (1996). Embryonic development and pattern formation. FASEB J. 10, 961- 968.
Morrison etal., (1985). Proc. Natl. Acad. Sci. USA 81, 6851.
Muindi, J. R. Frankel, S. Huselton, DeGrazia, Garland, Young, C. W. and Warrell, R. Jr. (1992). Clinical pharmacology of oral all-trans retinoic acid in patients with acute promyelocytic leukemia. Cancer Research 52, 2138-2142.
Muindi, J. Young, C. W. and Warrell, R. J. (1994a). Clinical pharmacology of all-trans retinoic acid. Leukemia 8, 1807-1812.
Muindi, J. Young, C. W. and Warrell, R. J. (1994b). Clinical pharmacology of all-trans retinoic acid. Leukemia 8, s16-s21.
Napoli, J. Boerman, M. Chai, Zhai, Y. and Fiorella, P. D. (1995). Enzymes and binding proteins affecting retinoic acid concentrations. J. Ster. Biochem. Mol. Biol. 53, 497-502.
Napoli, J. (1996). Retinoic acid biosynthesis and metabolism. FASEB J. 10, 993-1001.
Nelson, D. Kamataki, Waxman, D. Guengerich, F. Estabrook, R. Feyereisen, Gonzalez, F. Coon, M. Gunsalus, I. Gotoh, Okuda, K. and Nebert, D. W.
(1993). The P450 superfamily: update on new sequences, gene mapping, accession numbers, *25 early trivial names of enzymes, and nomenclature. DNA Cell Biology 12, 1-51.
Old, R.W. and Primrose, In: Principles of Gene Manipulation. An Introduction to Genetic Engineering, 4th ed. Oxford University Press. 63-66.
Ohno, C. K. and Petkovich, M. (1993). FTZ-F1 beta, a novel member of the Drosophila nuclear receptor family. Mechanisms of Development 40, 13-24.
30 Ohno, C. Ueda, H. and Petkovich, M. (1994). The Drosophila nuclear receptors FTZ-F1 alpha and FTZ-F1 beta compete as monomers for binding to a site in the fushi tarazu gene.
Mol. Cell Biol. 14, 3166-75.
Ohyama, Ozono, Uchida, Shinki, Kato, Suda, Yamamoto, Noshiro, M.
and Kato, Y. (1994). Identification of a vitamin D-responsive element in the 5'-flanking region of 35 the rat 25-hydroxyvitamin D3 24-hydroxylase gene. Journal of Biological Chemistry 269, 10545- Petkovich, Brand, N. Krust, A. and Chambon, P. (1987). A human retinoic acid receptor which belongs to the family of nuclear receptors. Nature 330, 444-50.
Pijnappel, W. Hendriks, H. Folkers, G. van den Brink, Dekker, E. J., Edelenbosch, van der Saag, P. and Durston, A. J. (1993). The retinoid ligand 4-oxo-retinoic acid is a highly active modulator of positional specification. Nature 366, 340-4.
-47- Reddy, A. Chen, Zacharewski, Gronemeyer, Voorhees, J. J. and Fisher, G. J.
(1992). Characterization and purification of human retinoic acid receptor-g1 overexpressed in the baculovirus-insect cell system. Biochem. J. 287, 833-840.
Rigas, Miller, Zhang, Z. Klimstra, Tong, Kris, M. and Warrell, R. (1996).
Metabolic phenotypes of retinoic acid and the risk of lung cancer. Cancer Res. 56, 2692-2696.
Roberts, A. Nichols, M. Newton, D. L. and Sporn; M. B. (1979a). In vitro metabolism of retinoic acid in hamster intestine and liver. Journal of Biological Chemistry 254, 6296-302.
Roberts, A. Frolik, C. Nichols, M. D. and Sporn, M. B. (1979b). Retinoid-dependent induction of the in vivo and in vitro metabolism of retinoic acid in tissues of the vitamin Adeficient hamster. Journal of Biological Chemistry 254, 6303-9.
Sambrook, Fritsch E.F. and Maniatis, T. (1989). Molecular Cloning: A Laboratory Manual.
Cold Spring Harbor Lab Press, Cold Spring Harbor, New York.
Staerz Bevan (1986a). Proc. Natl. Acad. Sci. (USA) 83, 1453.
Staerz Bevan (1986b). Immunology Today 7, 241.
Stewart, Canitrot, Baracchini, Dean, Deeley, and Cole, S.P.C. (1996).
Reduction of Expression of the multidrug resistance protein (MRP) in human tumor cells by antisense phophorothioate oligonucleotides. Biochem. Pharamcol. 51, 461-469.
Taimi, M. and Breitman, T.R. (1997). N-4-hydroxyphenylretinamide enhances retinoic acidinduced differentiation and retinoylation of proteins in the human acute promyelocytic leukemia cell line, NB4, by a mechanism that may involve inhibition of retinoic acid catabolism.
Biochemical and Biophysical Research Communications 232, 432-435.
Takatsuka, Takahashi, N. and De Luca, L. M. (1996). Retinoic Acid Metabolism and Inhibition of Cell Proliferation: An Unexpected Liaison. Cancer Research 56, 675-678.
Takeda et al., (1985). Nature 314, 452.
Tanaguchi et al., European Patent Publication EP171496.
Teng, et al. (1982) Meth. Enzymol. 92. 3-16.
Thaller, C. and Eichele, G. (1990). Isolation of 3,4-didehydroretinoic acid, a novel morphogenetic signal in the chick wing bud. Nature 345, 815-9.
Van Wauwe, J. Coene, Goossens, Van Nijen, Cools, W. and Lauwers, W.
(1988). Ketoconazole inhibits the in vitro and in vivo metabolism of all-trans retinoic acid. The Journal of Pharmacology and Experimental Therapeutics 245, 718-722.
oVan Wauwe, J. Coene, Goossens, Cools, W. and Monbaliu, J. (1990). Eiects of cytochrome P450 inhibitors on the in vivo metabolism of all-trans-retinoic acid in rats. The Journal of Pharmacology and Experimental Therapeutics 252, 365-369.
Van Wauwe, Van Nyen, Coene, Stoppie, Cools, Goossens, Borghgraef, P.
and Janssen, P. A. J. (1992). Liarazole, an Inhibitor of Retinoic Acid Metabolism, Exerts Retinoid-Mimetic Effects in Vivo. The Journal of Pharmacology and Experimental Therapeutics 261,773-779.
Ward et al., (1989). Nature 341. 544-546.
Warrell, R. J. (1994). Applications for retinoids in cancer therapy. Seminars in Hematol. 31, 1- -48- Warrell, R. Maslak, Eardley, Heller, Miller, W. J. and Frankel, S. R. (1994).
Treatment of acute promyelocytic leukemia with all-trans retinoic acid: an update of the New York experience. Leukemia 8, 929-933.
White, J. Boffa, M. Jones, B. and Petkovich, M. (1994). A zebrafish retinoic acid receptor expressed in the regenerating caudal fin. Development 120, 1861-72.
Williams, J. B. and Napoli, J. L. (1987). Inhibition of retinoic acid metabolism by imidazole antimycotics in F9 embryonal carcinoma cells. Biochemical Pharmacology 36, 1386-1388.
Wilson, G. M. and Deeley, R. G. (1995). An episomal expression vector system for monitoring sequence-specific effects on mRNA stability in human cell lines. Plasmid 33,198-207.
Windhorst, D. B. (1982). The use of isotretinoin in disorders of keratinization. Journal of the American Academy of Dermatology 6, 708-9.
Wouters, van, D. Dillen, Coene, M. Cools, W. and De, C. R. (1992). Effects of liarazole, a new antitumoral compound, on retinoic acid-induced inhibition of cell growth and on retinoic acid metabolism in MCF-7 human breast cancer cells. Cancer Research 52. 2841-6.
Zierold, Darwish, H. M. and DeLuca, H. F. (1995). Two vitamin D response elements function in the rat 1,25-dihydroxyvitamin D 24-hydroxylase promoter. Journal of Biological Chemistry 270, 1675-8.
.e, a -49- SEQUENCE LISTING GENERAL INFORMATION:
APPLICANTS:
NAME: QUEEN'S UNIVERSITY AT KINGSTON
STREET:
CITY: Kingston PROVINCE: Ontario COUNTRY: CA POSTAL CODE (ZIP) K7L 1J7 NAME: PETKOVICH, P. Martin STREET: 875 Everitt Avenue CITY: Kingston PROVINCE: Ontario COUNTRY: CA POSTAL CODE (ZIP) K7M 4R1 NAME: WHITE, Jay A.
STREET: 913 Purcell Crescent CITY: Kingston PROVINCE: Ontario COUNTRY: CA POSTAL CODE (ZIP) K7P 1C1 NAME: BECKETT, Barbara R.
STREET: 112 Westmorland Road CITY: Kingston PROVINCE: Ontario COUNTRY: CA POSTAL CODE (ZIP) K7M 1J7 NAME: JONES, Glenville STREET: 66 Inverness Crescent CITY: Kingston i. PROVINCE: Ontario COUNTRY: CA POSTAL CODE (ZIP) K7M 6N7 (ii) TITLE OF INVENTION: RETINOID METABOLIZING PROTEIN (iii) NUMBER OF SEQUENCES: 38
*S
(iv) COMPUTER READABLE FORM: MEDIUM TYPE: Diskette, 3 1/2 inch, 1.4 Mb storage COMPUTER: COMPAQ, IBM PC compatible OPERATING SYSTEM: MS-DOS 5.1 SOFTWARE: WORD PERFECT CURRENT APPLICATION DATA: APPLICATION NUMBER: (vi) PRIOR APPLICATION DATA: APPLICATION NUMBER: US 08/667,546; US 08/724,466 FILING DATE: 21-JUN-1996; 01-OCT-1996 INFORMATION FOR SEQ ID NO:1 SEQUENCE CHARACTERISTICS: LENGTH: 337 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1 TGCCAGTGGA CAATCTCCCT ACCAAATTCA CTAGTTATGT CCAGAAATTA GCCTAAACCG GAGCCTTTGT ACATATGTTT TTATTTTAGA TGAACTGTGA TGTATTGGAT ATTTTCTAAT TTGTTTATAT AAAGCAGATG TGTATATAAG TCTATGCGAA GAAGCGAAAA CGAGGGCACT ACTTTCTCAT GGATCACTGT AATGCTACAG AGTGTCTGTG ATGTATATTT ATAATGTAGT TGTGTCATAT AGCTTTTGTA CTGTATGCAA CTTATTTAAC TCGCTCTTTA TCTCATGGGT TTTATTTAAT AAAACATGTT CTTACAAAAA AAAAAAA 120 180 240 300 337 INFORMATION FOR SEQ ID NO:2 SEQUENCE CHARACTERISTICS: LENGTH: 492 amino acids TYPE: amino acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: Gly Leu Tyr Thr Leu Met Val 0O OS *0 S
S
0 .0.:0 0O *5~ .5*
S
*.eg o o oeoo Met 1 Pro SEQ ID NO:2 Thr Phe Leu 10 Val Lys Leu 5 Phe Cys Thr lle Val Leu Val Leu Leu 20 Leu Ala Ala Trp Glu 25 Arg Ile Arg Arg Val Asp Pro Asn Ser Pro Leu Met Leu Met Pro Gly Thr Leu Gin Arg Met Gly Arg Lys Leu Pro Phe Ile Glu Thr Leu Gin Leu Gly Phe Leu Arg Met Arg Gin Lys Cys Ile Tyr Thr His Leu Phe 50 S S 50 0550 o
SSOS
S.
0050 05*
S.
Gly 85 Leu Asn Pro Thr Val Val Met Gly Ala Asp Asn Val Arg Gin Pro Ala Ser 115 His Gly Val Leu Gly Glu His 105 Gly Lys Leu Val Ser Val Gin Trp 110 Ser Asn Val Arg Thr lie Ser Asp Thr Gin His Lys Lys Lys Ala 130 Ser Arg Ile 140 Ile Arg Ala Phe Gin Gin Glu Asp Ala Leu 145 Lys Ser Ala le Glu 150 Glu Lys Trp Leu Gin Leu Met Phe 185 Tyr Ile Pro Lys 170 Ser Cys Val Tyr Pro Glu Met 180 Arg Ile Ala Met Arg Ile Leu 190 -51- Thr Asp Glu Gin Glu Leu Val 205 Leu Gly Phe Glu Pro Glu Gin Ile Lys 195 200 o Glu Val 225 His Asn Asn Ala Ala 305 Lys Pro Gly Gly Pro 385 Ala Ser Gly Lys Asn 465 Asn Ala 210 Pro Ser Glu Ser Ala 290 Thr Val Gly Cys Phe 370 Lys Asp Lys Gly Ile 150 Gly .eu SPhe Phe Lys Asn Arg 275 Thr Ser Arg Lys Val 355 Arg Gly Val Gly Ser 435 Phe Pro Pro Gl Sei Ile Glu 260 Arg Glu Leu Glu Gly 340 Ile Val Trp Phe Leu 420 Arg Leu Pro Thr i Glu r Gly SGlu 245 Gin Ser Leu Val Glu 325 Leu Lys Ala Asn Pro 405 Glu Met Val Thr Lys I Met Leu 230 Glu Lys Asp Leu Met 310 Val Ser Glu Leu Val 390 Asn Asp Cys Glu 4et 470 Phe SIle 215 Tyi Asn Tyr Glu Phe 295 Phe Gin Met Thr Lys 375 Ile Lys Gly Val Leu 455 Lys Thr e Lys SArg Ile SLys Pro 280 .Gly Leu Glu Glu Leu 360 Thr Tyr Glu Ser Gly 440 Thr Thr Ser Asn Leu Phe Gly Leu Arg 235 Arg Lys Lys 250 Asp Ala Leu 265 Phe Ser Leu Gly His Glu Gly Leu Asn 315 Lys Val Glu 330 Leu Leu Asp 345 Arg Ile Asn Phe Glu Leu Ser Ile Cys 395 Glu Phe Gin 410 Arg Phe Asn 425 Lys Glu Phe Gin His Cys Gly Pro Thr 475 Tyr Val Arg A Ser 220 Ala Ilie Gin Gin Thr 300 Thr Met Gin Pro Asn 380 Asp Pro Tyr Ala \sn 160 :le SLeu Arg Gin Leu Ala 285 Thr Glu Gly Leu Pro 365 Gly Thr Glu Ile Lys 445 Trp Tyr E Pro Asn Asp Leu 270 Met Ala Val Met Lys 350 Val Tyr His Arg Pro 130 Val Ile Pro SIl Ph( Asp 255 Ile Lys Ser Val Tyr 335 Tyr Pro Gin Asp Phe 415 Phe Leu Leu Val e Asp e Ile 240 Asp Glu Glu Thr Gin 320 Thr Thr Gly Ile Val 400 Met Gly Leu Ser Asp 480 Lsn 490 INFORMATION FOR SEQ ID NO:3 SEQUENCE CHARACTERISTICS: LENGTH: 1850 base pairs TYPE: nucleic acid TOPOLOGY: linear 52 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:3 TGTCGCCGTT*GCTGTCGGTT GCTGTCGGAC GCTGTCTCCT CTCCAGAAGC TTGTTTTTCG TTTTGGCGAT CAGTTGCGCG CTTCAAC ATG GGG CTG TAC ACC CTT ATG GTC ACC Met Giy Leu Tyr Thr Leu Met Val Thr TTT CTC Phe Leu AAG TTG Lys Leu AGT CCT Ser Pro CTC CAG Leu Gin AAA TAC Lys Tyr 75 AGG GTG Arg Val AAG CTG Lys Leu TCT GAC Ser Asp AAG GCC Lys Ala CCC GTG Pro Val 155 AAA GAC Lys Asp 170 CGG ATA Arg Ile ACG GAC Thr Asp
TGC
Cys
TGG
Trp
CTA
Leu
CTG
Leu
GGG
Gi y
ATG
Met
GTG
Vali
ACC
Th r
ATT
Ile 140
ATC
Ile
TCC
Ser
GCT
Aa
GAG
31 u
ACC
Thr
GAG
Gi u
CCG
Pro AT C Ile
TGC
Cys
GGA
Gi y
TCT
Ser
CTC
Leu 125
ATG
Met
CAG
Gin
TGC
Cys
ATG
Met
CAA
Gin 205
ATC
Ile
ATG
Met C CA Pro
CTC
Le u AT C Ile
GCT
Al a
GTT
Vai 110
TCC
Ser
AGG
Arg
CAG
Gin
GTG
Val
AGA
Org 190
GAA
31 u Val 15
TTA
Leu
GGT
Gi y
CAG
Gin
TAC
Tyr
GAT
Asp 95
CAG
Gin
AAT
As n
GCG
Al a
GAG
Gi u
CTG
Leu 175
ATC
Ile
CTG
Leu Leu
ATC
Met
ACC
Thr
AGA
Arq
AAG
Lys 80
AAT
As n T GG T rp
GTC
Val
TTC
Ph e
GTG
Val 160
GTT
Vai
CTG
Leu
GTG
Val1 Pro
ATC
Ile
ATG
Met
AGG
Arg 65
ACG
Thr
GTG
Val
CCA
Pro
CAT
His
TCT
Ser 145
AAG
Lys
TAT
Tyr
CTT
Leu
GAA
Gi u Vai
CGA
Arg
GGC
Gi y 50
AAG
Lys
CAC
His
AGG
Arg
GCA
Al a
GGA
Gi y 130
CGA
Arg
AGC
Ser
CCA
Pro
GGT
Gly
GCT
Ala 210 Le u
CGA
Arg 35
TTG
Phe Leu GAT CCG Asp Pro Ala Ala Vai AAC TGC AGA Asn Cys Arg GGA GAA ACG CCG TTC ATT Leu Pro Phe Ile Gly Giu Thr
TTT
Ph e
CTC
Leu
CAG
Gin
TCA
Ser 115
GTT
Vai
GAT
A~sp
GCC
Al a
GAA
31 u
~TTT
E'he 195 rTT ?he
CTG
Leu
TTC
Phe
ATT
Ile 100
GTG
Val1
CA
Gin
GCT
Al a
ATA
Ile
ATG
Met 180
GAA
Gi u
GAG
Glu
COC
Arq
GGG
Gi y
CTG
Le u
AGA
Arg
CAC
His
CTG
Leu
CAG
Gin 165
AAG
Lys
CCA
Pro
.GAA
31 U
ATG
Met
AAC
As n CT 3 Leu
ACC
Thr
AAA
Lys
GAG
31 u 150
GAA
Gi u
AA
Lys
GAG
Gi u
ATG
Met Lys
CCG
Pro
GGC
Gly
ATC
Ile
AAC
As n 135
CAC
His T 33 T rp
CTC
Leu
CAA
Gin
ATC
Ile 215
CGG
Arg
ACT
Th r
GAA
Gi u
CTG
Leu 120
AAG
Lys
TAC
Tyi:
CTG
Leu
ATG
Met
ATA
Ile 200
AAA~
Lys
CAG
Gin
GTC
Vai
CAC
His 105
GGC
Gly
AAA
Lys
ATT
Ile
CAA
Gin
TTT
Phe 185
AAG
Lys
AAC
DRsn 210 258 306 354 402 GTG CTA CCC GTT TTA CTC TTT- CTC GCC GCG GTG 546 TTG TTC TCC TTG CCA ATC GAC GTT CCT TTC AGT GGT CTG TAC AGG GGT Leu Phe Ser Leu Pro Ile Asp Vai Pro Phe Ser Giy Leu Tyr Arg Gly 220 225 230 53 TTG AGG GCA CGC AAT TTC ATT CAC TCC AAA ATT GAG GAA AAC ATC AGG Leu Arg Ala Arg Asn Phe Ile His Ser Lys Ile Giu Giu Asn Ile Arg AAG AAA ATT CAA GAT GAC GAC AAT Lys Lys Ile Gin Asp Asp Asp Asn 250 255 GAA AAC GAA Giu Asn Giu 260 CAG AAA TAC AAA gin Lys Tyr Lys
GAC
Asp 265 GCC CTT CAG CTG Ala Leu Gin Leu
TTG
Leu 270 ATC GAG AAC AGC Ile Glu Asn Ser
AGA
Arg 275 AGA AG T GAC GAA Arg S' r Asp Giu CCT TTT Pro Phe 280 AGT TTG CAG Ser Leu Gin CAT GAA ACC His Giu Thr 300
GCG
Aila 285 ATG AAA GAA GCA Met Lys Giu Ala ACA GAG CTT CTA Thr Glu Leu Leu TTT GGA GGT Phe Gly Giy 295 TTT CTG GGT Phe Leu Giy ACC GCC AGC ACT Thr Ala Ser Thr
GCA
Aila 305 ACC TCA CTT GTC Thr Ser Leu Val
ATG
Met 310 CTG AAC Leu Asn 315 ACA GAA GTG GTG Thr Giu Val Val
CAG
Gin 320 AAG GTC AGA GAG Lys Val Arg Giu
GAG
Gi u 325 GTT CAG GAG AAG Val Gin Giu Lys GAA ATG GGC ATG Giu Met Giy Met ACA CCT GGA AAG Thr Pro Giy Lys
GGC
Gi y 340 TTG AGT ATG GAG Leu Ser Met Giu
CTG
Leu 345 1026 1074 1122 1170 1218 TTG GAC CAG CTG Leu Asp Gin Leu
AAG
Lys 350 TAC ACT GGA TGT Tyr Thr Gly Cys
GTG
Vai 355 ATT AAA GAG ACT Ile Lys Glu Thr CTT AGA Leu Arg 360 ATC AAC CCT Ile Asn Pro GAA TTG AAT Giu Leu Asn 380
CCT
Pro 365 GTT CCC GGA GGA Vai Pro Gly Giy AGA GTC GCA CTC Arg Val Aia Leu AAA ACC TTT Lys Thr Phe 375 ATT TAC AGC Ile Tyr Ser GGT TAC CAA ATT Giy Tyr Gin Ile
CCT
Pro 385 AAA GGA TGG AAC Lys Gly Trp Asn
GTC
Val 390 1266 ATC TGT Ile Cys 395 GAC ACG CAC GAT Aso Thr His Asp GCC GAC GTC TTT Aia Asp Val Phe
CCA
Pro 405 AAC AAA. GAG GAG Asn Lys Giu Giu
TTC
Phe 410 CAG CCG GAG AGA Gin Pro Glu Arg
TTC
Phe 415 ATG AGC AAA GGT Met Ser Lys Giy CTG GAG GAC GGG TCC AGG Leu Giu Asp Giy Ser Arg 420 425 TTT AAC TAC ATC Phe Asn Tyr Ile
CCC:
Pro 430 TTC GGA GGA GGA TCC AGG ATG TGT GTG Phe Gly Giy Giy Ser Arg*Met Cys Val 435 GGC AAA Gly Lys 440 1314 1362 1410 1458 1506 1554 GAG TTC GCC AAA GTG TTA CTC AAG Giu Phe Ala Lys Vai Leu Leu Lys 445
ATC
Ile 450 TTT TTA GTT GAG Phe Leu Val Glu TTA ACG CAG Leu Thr Gin 455 CAT TGC AAT TGG ATT CTC TCA AAC GGA CCC CCG ACA ATG AAA ACA GGC His Cys Asn Trp Ile Leu Ser Asn Giy Pro Pro Thr Met Lys Thr Gly 460 465 470 CCG ACT ATT Pro Thr Ile 475 TAC CCA GTG GAC AAT CTC CCT ACC Tyr Pro Vai Asp Asn Leu Pro ThzL AAA TTC ACT AGT TAT Lys Phe Thr Ser Tyr 485 -54- GTC AGA AAT TAGCCTAACC GGAGCTTTGT ACATATGTTT TTATTTTAGA Val Arg Asn 490 TGAACTGTGA TGTATTGGAT ATTTTCTATT TTGTTTATAT AAAGCAGATG TGTATATAAG TCTATGCGAG GAAGCGAJAAA CGAGGGCACT ACTTTCTCAT GGATCACTGT AATGCTACAG AGTGTCTGTG ATGTATATTT ATAATGTAGT TGTGTTATAT AGCTTTTGTA CTGTATGCAA CTTATTTAAC TCGCTCTTTA TCTCATGGGT TTTATTTAAT AAAACATGTT CTTACAAAAA INFORMATION FOR SEQ ID NO:4 SEQUENCE CHARACTERISTICS: LENGTH: 497 amino acids TYPE: amino acid TOPOLOGY: linear 1603 1663 1723 1783 1843 1850 (xi) SEQUENCE DESCRIPTION: Met Gly Leu Pro Ala Leu Leu Ala P rc Val Met Arg Thr Val Pro His Se r 145 Gi y Val Leu Gin Leu Ser Gly Lys His Arg Al a Asp 130 Arg Ser Tyr Leu Leu 210 Leu Gi y Phe Phe Leu Arg Ser 115 Ser Gl u Ser Pro Gl y 195 Val Leu 20 Arg Pro Leu Phe Ile 100 Val Ser Al a Leu Gi u 180 Cys Gi u Ph e Asp Phe Gin GI y Leu Arg His Leu Gi u 165 Val1 Giu A-l a Leu Arg Phe Met 70 Arg Le u Th r Lys Gi u Gin Lys Pro Phe Al a Ser Gly 55 Lys Pro Gi y Ile Gin 135 Cys Trp Arg Gin Giu 215 Aila Cys 40 Gi u Arq Thr AsD Le u 120 Arg Tyr Leu Le u Le u 200 31 U SEQ ID NO:4 Ser Ala Leu Ile Lys Leu 25 Ala Leu Pro Thr Leu Gin Arg Lys Tyr 75 Val. Arg Val 90 Asp Arg Leu 105 Gly Ser Gly Lys Lys Val Val Pro Val 155 Ser Cys Gly 170 Met Phe Arg 185 Ala Gly Asp Met Thr Arg T rp Leu Met Gi y Met Val Cys Ile 140 Ile Gi u Ile 'sly ksn 220 Asp Pro Val Phe Gly Ser Leu 125 Met Thr Arg Ala Asp 205 Leu Leu Pro Leu Ile Al a Val 110 Ser Arg Gi u GI y Met 190 Ser Phe Cys Thr Phe Val Leu Tyr GI y Gin Tyr Asp His As n Al a Gi u Leu 175 Arg Giu Ser Cys Thr Arg Lys As n T rp Leu Phe Val 160 Leu Ile Gin Le u Pro Ile Asp Vai Pro Phe Ser Gly Leu 225 230 Asn Gi y Leu Al a Thr 305 His Cys Leu Pro Gi y 385 Thr Asp Ile Lys T rp 465 Tyr Le u Leu Leu Le u 290 Al a Val Lys Lys Val 370 Tyr His Arg Pro Ile 450 Gin Pro Ile Arg le 275 Lys Ser Leu Ser Tyr 355 Pro Gin Asp Phe Phe 435 Leu Leu Val His Al.a 260 GI u Gin Al a Gin As n 340 Ile GI y Ile Val1 Ser 420 GI y Le u Leu A~sp Al a 245 Ser His Ser Al a Lys 325 Glin Gi y Gi y Pro Al a 405 Aila Gi y Lys Asn Asn 485 Arg GI u Ser Ser Th r 310 Val Asp C ys Ph e Lys 390 Gi u Pro G I y Ile Gly 470 Ile Al a Trp Th r 295 Ser Arq As n Vai Ara 375 Gi y Ile His Le u Phe 455 Pro Gi u Gi y GI u 280 Gi u Leu Giu Lys Ile 360 Val T rp Phe Pro Arg 440 Thr Pro Gin Gin 265 Arg Leu Ile Gi u Leu 345 Lys Aila Asn Thr Giu 425 Ser Val Thr Tyr As n 250 Gi y Gi y Leu Thr Le u 330 Asp Glu Leu Val As n 410 Aksp :ys 31 u ,let Arg 235 Ile Cys Gi u Phe Tyr 315 Lys Met Th r Lys Ile 395 Lys Al a Vali Leu Lys 475 Arg Lys Arg Gi y 300 Leu Ser Gi u Leu Thr 380 Tyr Gi u Ser GI y Al a 460 Thr Gly Met Lys Ala Al a Asp Leu 285 GI y Gi y Lys Ile Arg 365 Phe Ser Gi u Arg Lys 445 Arg Ser Lys Al a 270 Asp His Leu Gly Leu 350 Leu Gi u Ile Phe Phe 430 Glu His Pro Ile 255 Leu Met Giu Tyr Leu 335 Giu As n Leu Cys As n 415 Ser Phe Dys ['hr Arg 240 Cys Gin Gin Thr Pro 320 Le u Gin Pro Asn Asp 400 Pro Phe Al a Asp Val 480 Leu Pro Ala Arg Phe Thr His Phe His Gly Glu 495 INFORMA~TION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 1494 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID ATG GGG CTC CCG GCG CTG CTG GCC AGT GCG CTC TGC ACC TTC GTG CTG Met Gly Leu Pro Ala Leu Leu A-la. Ser Ala Leu Cys Thr Phe Val Leu 1 5 10 -56- CCG CTG CTG Pro Leu Leu GTG AGC GGC Val Ser Gly
CTC
Leu TTC CTG GCT GCG Phe Leu Ala Ala ATC AAG Ile Lys 25 CTC TGG GAC CTG TAC TGC Leu Trp Asp Leu Tyr Cys CGC GAC CGC AGT TGT GCC CTC CCA TTG CCC CCC GGG ACT Arg Asp Arg Ser Cys 40 Ala Leu Pro Leu Pro Pro Gly Thr ATG GGC Met Gly TTC CCC TTC Phe Pro Phe TTT GGG Phe Gly 55 ATG AAG Met Lys 70 GAA ACC TTG Glu Thr Leu CGC AGG AAA Arg Arg Lys CAG ATG Gin Met TAC GGC Tyr Gly 75 GTA CTG CAG CGG Val Leu Gin Arg TTC ATC TAC AAG Phe Ile Tyr Lys
AGG
Arg AAG TTC CTG CAG Lys Phe Leu Gin ACG CAT CTG TTC Thr His Leu Phe
GGG
Gly CGG CCC ACC GTA Arg Pro Thr Val
CGG
Arg 90 GTG ATG GGC GCG Val Met Gly Ala GAC AAT Asp Asn GTG CGG Val Arg CCA GCG Pro Ala CAC GAC His Asp 130
CGC
Arg
ATC
Ile 100 TTG CTC GGA GAC Leu Leu Gly Asp CGG CTG GTG TCG Arg Leu Val Ser GTC CAC TGG Val His Trp 110 TCT AAC CTG Ser Asn Leu 1 r r rr r r r TCG GTG CGC ACC ATT Ser Val Arg Thr Ile 115 TCC TCG CAC AAG CAG Ser Ser His Lys Gin 135
CTG
Leu 120 GGA TCT GGC TGC Gly Ser Gly Cys CGC AAG AAG GTG ATT ATG CGG GCC TTC Arg Lys Lys Val Ile Met Arg Ala Phe 140 CGC GAG GCA CTC Arg Giu Ala Leu
GAA
Glu 150 TGC TAC GTG CCG Cys Tyr Val Pro ATC ACC GAG GAA Ile Thr Giu Glu GGC AGC AGC CTG Gly Ser Ser Leu CAG TGG CTG AGC Gin Trp Leu Ser GGC GAG CGC GGC Gly Glu Arq Gly CTC CTG Leu Leu 175 GTC TAC CCC Val Tyr Pro CTA CTG GGC Leu Leu Gly 195
GAG
Glu 180 GTG AAG CGC CTC ATG TTC CGA ATC GCC Val Lys Arg Leu Met Phe Arg Ile Ala ATG CGC ATC Met Arg Ile 190 TCC GAG CAG Ser Giu Gin TGC GAA CCC CAA Cys Glu Pro Gin
CTG
Leu 200 GCG GGC GAC GGG Ala Gly Asp Gly
GAC
Asp 205 CAG CTT Gin Leu 210 GTG GAG GCC TTC Val Giu Ala Phe
GAG
Glu 215 GAA ATG ACC CGC Glu Met Thr Arg
AAT
Asn 220 CTC TTC TCG CTG Leu Phe Ser Leu CCC ATC GAC GTG CCC TTC Pro Ile Asp Vai Pro Phe 225 230 AGC GGG CTG TAC Ser Gly Leu Tyr CGG GGC ATG AAG GCG CGG Ara Gly Met Lys Ala Arg 235 240 AAC CTC ATT Asn Leu Ile GGG CTG CGG Gly Leu Arg CAC GCG His Ala 245 CGC ATC GAG Arg Ile Glu CAG AAC Gin Asn 250 CAG GGC Gin Gly 265 ATT CGC GCC Ile Arg Ala TGC AAA GAC Cys Lys Asp AAG ATC TGC Lys Ile Cys 255 GCG CTG CAG Ala Leu Gin 270 768 816
GCA
Ala 260 TCC GAG GCG GGC Ser Giu Ala Gly -57- CTG TTG ATC Leu Leu Ile 275 GAG CAC TCG Glu His Ser TGG GAG AGG GGA Trp Giu Arg Gly 280 GAG CGG CTG GAC ATG CAG Glu Arg Leu Asp Met Gin 285 GCA CTA Ala Leu 290 AAG CAA TCT TCA ACC GAA CTC CTC TTT Lys Gin Ser Ser Thr Glu Leu Leu Phe 295 GGA GGA CAC GAA ACC Gly Giy His Giu Thr 300
ACG
Thr 305 GCC AGT GCA GCC Ala Ser Ala Ala
ACA
Thr 310 TCT CTG ATC ACT Ser Leu Ile Thr TAC CTG GGG CTC TAC CCA Tyr Leu Giy Leu Tyr Pro 315 320 CAT GTT CTC CAG His Val Leu Gin GTG CGA GAA GAG Vai Arg Giu Glu
CTG
Leu 330 AAG AGT AAG GGT Lys Ser Lys Gly TTA CTT Leu Leu 335 TGC AAG AGC Cys Lys Ser CTT AAA TAC Leu Lys Tyr 355
AAT
Asn 340 CAA GAC AAC AAG Gin Asp Asn Lys TTG GAC ATG GAA ATT TTG GAA CAA Leu Asp Met Glu Ile Leu Giu Gin 345 350 ATC GGG TGT GTT lie Giy Cys Vai
ATT
Ile 360 AAG GAG ACC CTT Lys Giu Thr Leu CTG AAT CCC Leu Asn Pro CCA GTT Pro Vai 370 CCA GGA GGG TTT Pro Giy Giy Phe GTT GCT CTG AAG Val Ala Leu Lys
ACT
Thr 380 TTT GAA TTA AAT Phe Glu Leu Asn r r r
GGA
Gly 385 TAC CAG ATT CCC Tyr Gin Ile Pro
AAG
Lys 390 GGC TGG AAT GTT Gly Trp Asn Vai
ATC
Ile 395 TAC AGT ATC TGT Tyr Ser Ile Cys
GAT
Asp 400 ACT CAT GAT GTG GCA GAG ATC TTC ACC AAC AAG GAA GAA TTT Thr His Asp Vai Aia Giu Ile Phe Thr Asn Lys Giu Glu Phe 405 410 AAT CCT Asn Pro 415 1008 1056 1104 1152 1200 1248 1296 1344 1392 1440 1488 GAC CGA TTC Asp Arg Phe ATT CCA TTT Lie Pro Phe 435 GCT CCT CAC CCA Ala Pro His Pro
GAG
Glu 425 GAT GCA TCC AGG Asp Ala Ser Arg TTC AGC TTC Phe Ser Phe 430 GAA TTT GCA Glu Phe Ala GGA GGA GGC CTT Gly Giy Gly Leu
AGG
Arg 440 AGC TGT GTA GGC Ser Cys Val Gly
AAA
Lys 445 AAA ATT CTT CTC AAA ATA Lys Ile Leu Leu Lys Ile ACA GTG GAG CTG Thr Val Giu Leu AGG CAT TGT GAC Arg His Cys Asp
TGG
Trp 465 CAG CTT CTA Gin Leu Leu AAT GGA Asn Gly 470 AAT CTC Asn Leu 485 CCT CCT ACA ATG Pro Pro Thr Met
AAA
Lys 475 ACC AGT CCC ACC Thr Ser Pro Thr TAT CCT GTG GAC Tyr Pro Val Asp CCT GCA AGA TTC ACC CAT TTC CAT Pro Ala Arg Phe Thr His Phe His GGG GAA Gly Glu 495 ATC TGA Ile 1494 -58- INFORMATION FOR SEQ ID NO:6 SEQUENCE CHARACTERISTICS: LENGTH: 20 amino acids TYPE: amino acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:6 Pro Phe Gly Gly Gly Pro Arg Leu Cys Pro Gly Tyr Glu Leu Ala Arg 1 5 10 Val Ala Leu Ser INFORMATION FOR SEQ ID NO:7 SEQUENCE CHARACTERISTICS: LENGTH: 20 amino acids TYPE: amino acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7 Pro Phe Ser Gly Gly Ala Arg Asn Cys Ile Gly Lys Gln Phe Ala Met 1 5 10 Ser Glu Met Lys INFORMATION FOR SEQ ID NO:8 SEQUENCE CHARACTERISTICS: LENGTH: 20 amino acids TYPE: amino acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:8 Pro Phe Ser Gly Gly Ala Arg Asn Cys Ile Gly Lys Gln Phe Ala Met 1 5 10 Asn Glu Leu Lys INFORMATION FOR SEQ ID NO:9 SEQUENCE CHARACTERISTICS: LENGTH: 20 amino acids TYPE: amino acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:9 Pro Phe Gly Thr Gly Pro Arg Asn Cys Ile Gly Met Arg Phe Ala Ile 1 5 10 Met Asn Met Lys -59- INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 20 amino acids TYPE: amino acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID Pro Phe Ser Gly Gly Ser Arg Asn Cys Ile Gly Lys Gin Phe Ala Met 1 5 10 Asn Glu Leu Lys INFORMATION FOR SEQ ID NO:11 SEQUENCE CHARACTERISTICS: LENGTH: 351 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:ll GAACTCCTCT TTGGAGGACA CGAAACCACG GCCAGTGCAG CCACATCTCT GATCACTTAC CTGGGGCTCT ACCCACATGT TCTCCAGAAA GTGCGAGAAG AGCTGAAGAG TAAGGGTTTA 120 CTTTGCAAGA GCAATCAAGA CAACAAGTTG GACATGGAAA TTTTGGAACA ACTTAAATAC 180 ATCGGGTGTG TTATTAAGGA GACCCTTCGA CTGAATCCCC CAGTTCCAGG AGGGTTTCGG 240 SGTTGCTCTGA AGACTTTTGA ATTAAATGGA TACCAGATTC CCAAGGGCTG GAATGTTATC 300 TACAGTATCT GTGATACTCA TGATGTGGCA GAGATCTTCA CCAACAAGGA A 351 INFORMATION FOR SEQ ID NO:12 SEQUENCE CHARACTERISTICS: LENGTH: 14 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:12 TTTTTTTTTT TTGG 14 INFORMATION FOR SEQ ID NO:13 SEQUENCE CHARACTERISTICS: LENGTH: 14 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:13 TTTTTTTTTT TTGA 14 INFORMATION FOR SEQ ID NO:14 SEQUENCE CHARACTERISTICS: LENGTH: 14 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:14 TTTTTTTTTT TTGT iA INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 14 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ I'D TTTTTTTTTT TTGC 14 INFORMATION FOR SEQ ID NO:16 SEQUENCE CHARACTERISTICS: LENGTH: 14 base pairs TYPE: nucleic acid TOPOLOGY: linear (xI) SEQUENCE DESCRIPTION: SEQ ID NO:16 TTTTTTTTTT TTAG 14 INFORMATION FOR SEQ ID NO:17 SEQUENCE CHARACTERISTICS: LENGTH: 14 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:17 TTTTTTTTTT TTAA 14 INFORMATION FOR SEQ ID NO:18 SEQUENCE CHARACTERISTICS: LENGTH: 14 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:18 TTTTTTTTTT TTAT 14 e INFORMATION FOR SEQ ID NO:19 SEQUENCE CHARACTERISTICS: LENGTH: 14 base pairs TYPE: nucleic acid S. TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:19 TTTTTTTTTT TTAC 14 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 14 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID TTTTTTTTTT TTCG -61- INFORMATION FOR SEQ ID NO:21 SEQUENCE CHARACTERISTICS: LENGTH: 14 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:21 TTTTTTTTTT TTCA 14 INFORMATION FOR SEQ ID NO:22 SEQUENCE CHARACTERISTICS: LENGTH: 14 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:22 TTTTTTTTTT TTCT 14 INFORMATION FOR SEQ ID NO:23 SEQUENCE CHARACTERISTICS: LENGTH: 14 base pairs TYPE: nucleic acid TOPOLOGY: linear SEQUENCE DESCRIPTION: SEQ ID NO:23 TTTTTTTTTT TTCC 14 INFORMATION FOR SEQ ID NO:24 SEQUENCE CHARACTERISTICS: LENGTH: 10 base pairs S(B) TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:24 AAGCGACCGA INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 10 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID TGTTCGCCAG INFORMATION FOR SEQ ID NO:26 SEQUENCE CHARACTERISTICS: LENGTH: 10 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:26
TGCCAGTGGA
-62- INFORMATION FOR SEQ ID NO:27 SEQUENCE CHARACTERISTICS: LENGTH: 10 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:27 GGCTGCAAAC INFORMATION FOR SEQ ID NO:28 SEQUENCE CHARACTERISTICS: LENGTH: 10 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:28 CCTAGCGTTG INFORMATION FOR SEQ ID NO:29 SEQUENCE CHARACTERISTICS: LENGTH: 21 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:29 GTAGCGGCCG CTGCCAGTGG A 21 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 12 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID GTAGCGGCCG CT 12 INFORMATION FOR SEQ ID NO:31 SEQUENCE CHARACTERISTICS: LENGTH: 1725 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:31 GCACGAGGGA GGCTGAAGCG TGCC ATG GGG CTC CCG GCG CTG CTG GCC AGT 51 Met Gly Leu Pro Ala Leu Leu Ala Ser 1 GCG CTC TGC ACC TTC GTG CTG CCG CTG CTG CTC TTC CTG GCG GCG CTC 99 Ala Leu Cys Thr Phe Val Leu Pro Leu Leu Leu Phe Leu Ala Ala Leu 15 20 AAG CTC TGG GAC CTG TAC TGT GTG AGC AGC CGC GAT CGC AGC TGC GCC 147 Lys Leu Trp Asp Leu Tyr Cys Val Ser Ser Arg Asp Arg Ser Cys Ala 3D 35 -63- CTC CCC TTG Leu Pro Leu TTG CAG ATG Leu Gin Met
CCC
Pro CCC GGT ACC ATG Pro Gly Thr Met.
GGC
Gi y 50 TTC CCA TTC TTT Phe Pro Phe Phe GTG CTT CAG CGG Val Leu Gin Arg
AGG
Arg 65 AAG TTT CTG CAG ATG Lys Phe Leu Gin Met GGG GAA ACA Gly Giu Thr AAG CGC AGG Lys Arg Arg CCC ACG GTG Pro Thr Val AAA TAC GGC TTC ATC TAC AAG ACG CAT CTG Lys Tyr Giy Phe Ile Tyr Lys Thr His Leu .80 TTT GGG CGG Phe Giy Arg
CGG
Arg GTG ATG GGC GCG Vai Met Gly Ala
GAT
Asp 95 AAT GTG CGG CGC Asn Val Arg Arg ATC TTG CTG GGA GAG CAC Ile Leu 100 Leu Giy Giu His 105 CGG TTG GTG TCG GTG CAC TGG CCC GCG TCG GTG CGC ACC ATC CTG GGC Arg Leu. Val Ser Vai His Trp Pro Ala Ser Vai Arg Thr Ile Leu Gly 110 115 120 GCT GGC TGC Aia Gly Cys AAG GTG ATT Lys Val Ile 140
CTC
Leu 125 TCC AAC CTG CAC Ser Asn Leu His
GAT
As p 130 TCC TCG CAC AAG Ser Ser His Lys CAG CGA AAG Gin Arg Lys 135 TGC TAC GTG Cys Tyr Vai ATG CAG GCC TTC Met Gin Ala Phe
AGC
Ser 145 CGC GAG GCA CTC Arg Giu Ala Leu
CAG
Gin 150 CTC GTG Leu Val 155 ATC GCT GAG GAA Ile Al1a Glu Giu AGC AGT TGT CTG Ser Ser Cys Leu CAG TGG CTA AGC Gin Trp Leu Ser
TGC
Cys 170 GGC GAG CGC GGC Giy Glu Arg Gly CTG GTC TAC CCC Leu Vai Tyr Pro
GAG
Gi u 180 GTG AAG CGC CTC Val Lys Arg Leu TTC CGC ATC GCC Phe Arg Ile Ala
ATG
Met 190 CGC ATC CTG CTG Ar Ile Leu Leu TGC GAG CCG GGT Cys Giu Pro Gly CCA GCG Pro Ala 200 GGC GGC GGG Gly Giy Giy ACC CGC AAT Thr Arg Asn 220
GAG
Gi u 205 GAC GAG CAA CAG Asp Giu Gin Gin
CTC
Leu 210 GTG GAG GCT TTC Val Giu Ala Phe GAG GAG ATG Giu Giu Met 215 AGC GGC CTG Ser Gly Leu CTC TTC TCT CTT Leu Phe Ser Leu
CCC
Pro 225 ATT GAC GTG CCC Ile Asp Val Pro
TTT
Phe 230 TAC CGG Tyr Arg 235 GGC GTG AAG GCG Gly Val Lys Ala
CGG
Arg 240 AAC CTT ATA CAC Asn Leu Ile His CGC ATC GAG GAG Arg Ile Giu Giu
AAC
Asn 250 ATT CGC GCC AAG Ile Arg Ala Lys ATC CGC CGG CTT CAG GCT ACA GAG CCG GAT GGG Ile Arg Arg Leu Gin Ala Thr Giu Pro Asp Gly 255 260 265 GGT TGC AAG GAC Gly Cys Lys Asp
GCG
Al a 270 CTG CAG CTC CTG Leu Gin Leu Leu
ATT
Ile 275 GAG CAC TCG TGG Giu His Ser Trp GAG AGG Glu Arg 280 GGA GAG AGG Gly Giu Arg
CTG
Leu 285 GAT ATG CAG GCA Asp Met Gin Ada CTA AAA CAA TCG Leu Lys Gin Ser 290 TCA ACA GAG CTC Ser Thr Giu Leu 295 CTC TTT GGT Leu Phe Gly 300 GGT CAT GAA ACT ACA GCC Gly His Glu Thr Thr Ala 305 -64 AGT GCT GCG ACA TCA CTG ATC Set Ala Ala Thr Ser Leu Ile 31.0 ACT TAC CTA GGA CTC TAC CCA CAT GTC CTC CAG AAA GTT CGA GAA GAG Thr Tyr 315 Leu Gly Leu Tyr
C
C
C
a.
C
ATA
Ile 330
GAC
Asp
GAG
Gi u
CTG
Leu
GTT
Val
AAC
As n 410
GAT
Asp
TGT
Cys
GAG
Gi u
ATG
Met
TTC
AAG
Lys
ATG
Met
ACC
Thr
AAG
Lys
ATT
Ile 395
AAG
Lys
GCT
Ala
GTA
Val
CTG
Leu
AAG
Lys 475
ACC
AGC
Ser
GAA
GI u
CTG
Leu
ACT
Thr 380
TAC
Tyr
GAG
GI u
TCC
Ser
GGC
Gi y
GCT
Al a 460
ACA
Thr
TAC
AAG
Lys
ACT
Thr
CGA
Arg 365
TTT
Ph e
AGT
Ser
GGC
Gi y
TTG
Leu 350
TTG
Leu
GAG
G I u
ATC
Ile GAA TTT Glu Phe CGG TTC Arg Phe 430 AAA GAG Lys Giu 445 AGG CAC Arg His PLGC CCC Ser Pro TTC CAG T T Leu 335
GAA
Gi u
AAT
As n
CTG
Leu
TGT
Cys
AAT
Asn 415
AGC
Ser
TTT
Phe T GT Cys
ACT
Thr
GGA
Pro 320
CTT
Leu
CAG
Gin
CCT
Pro
AAT
As n
GAC
Asp 400
CCC
Pro
TTC
Phe
GCA
Ala
GAT
Asp
GTG
Val 480
GAT
His
TGC
Cys
CTT
Leu
CCG
Pro
GGA
Gi y 385
ACC
Thr
GAC
Asp
ATT
Ile Lys
TGG
T'rp 465
PAC
L'yr Val Leu Gin AAG AGC AAT CP- GAC AAC AAG TTA Lys
AAA
Lys
GTT
Val 370
TAC
Tyr
CAC
H is
CGC
Arg
CCA
Pro
ATT
Ile 450
CAG
Gin
CCT
Ser
TAC
Tyr 355 C CA Pro
CAG
Gin
GAT
Asp
TTT
Phe
TTT
Phe 435
CTT
Leu
CTT
Leu
GTG
Asr 340
ATT
Ile
GGA
GI y
ATC
Ile
GTG
Val
ATA
Ile 420
GGA
Gi y
CTT
Leu
CTA
Leu
GAC
Gin
GGG
Gi y
GGG
Gi y
CCC
Pro
GCA
Al a 405
GTG
Val
GGA
Gi y
PAG
Ly s
PAT
As n
PAT
Asp
TGT
Cys
TTT
Phe
AAG
Lys 390
GAT
Asp
CCT
Pro
GGC
Gi y
ATA
Ile
GGA
Gly 470
CTC
A~ r
GTC
Val
CGG
Arg 375
GGC
GI y
ATC
Ile
CAT
His
CTT
Leu
TTT
Phe 455 Pro
CCT
Lys
ATT
Ile 360
GTT
Val T GG Trp
TTC
Phe
CCA
Pro
CGG
Arg 440
ACA
Thr
CCT.
Pro
GCA
Leu 345
AAG
Lys
GCT
Al a
AAT
As n
ACT
Thr
GAG
Gi u 425
AGC
Ser
GTG
Val
ACA
Thr
AGA
Lys 325 Val Arg Glu Glu 1011 1059 1107 1155 1203 1251 1299 1347 1395 1443 1491 1535 1595 1655 1715 1725 Pro Val Asp Asn Leu Pro A-la Arg 485 kTC TGATAGCTAT TTCAATTCTT Phe Thr Tyr Phe Gin Gly Asp Ile 490 495 GGACTTATTT GAPLGTGTATA TTGGTTTTTT TTAAP.AATAG TGTCATGTTG ACTTTATTTA ATTTCTAAAT GTATAGTATG ATATTTATGT GTCTCTACTA CAGTCCCGTG G.CTTTAAAT ATTAA).ATAA TGAATTTGTA TGATTTCCCA ATAAAGTPAA ATTPAAP.AGT GAAAA..PAAA INFORMATION FOR SEQ ID NO:32 SEQUENCE CHARACTERISTICS: LENGTH: 497 amino acids TYPE: amino acid TOPOLOGY: linear TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:32 Met Gly Leu Pro Ala Leu Leu Ala Ser Ala Leu Cys Thr Phe Val Leu 0* 9 *9 9 9 9* 9 9* S 9 Pro ValI Met Arg Thr Val1 Pro His Ser 145 Ser Val Leu Gin Pro 225 Asn Arg Leu Let.
Ser Gi y Lys His Arg Al a Asp 130 Arg Ser Tyr Leu Leu 210 Ile Leu Leu Leu Let Ser Phe Phe Le u Arg Ser 115 Ser Gl u Cys Pro Gly 195 Val1 Asp Ile Gin Ile 1Leu Arg Pro Le u Phe Ile 100 Vai Ser Al a Leu Gi u 180 Cys Glu Val His Al a 260 Glu Phe Asp Phe Gin Gi y 85 Leu Arq His Leu Gi u 165 Val1 GI u Al a Pro Al a 245 I'hr iis Let Arc Phe Met 70 Arg Leu Thr Lys Gln 150 Gln Lys Pro Phe Phe 230 Arg Giu Ser 1 Ala SSer Gly 55 Lys Pro GI y Ile Gln 135 Cys T rp Arg Gi y Glu 215 Ser Ile Pro TrpC Al 2 Cys 40 GI u Arg Thr Glu Leu 120 Arg Tyr Leu Leu Pro 200 Gl u Gly Gl u ksp ;i u so0 Leu 25 Al a Thr Arg Val His 105 Gi y Lys ValI Ser Met 185 Al a Met Leu Giu Gly 265 Arg Lys Leu Leu Lys Arg 90 Arg Al a Lys Le u Cys 170 Phe GI y Thr Tyr Asn 250 Giy 31y Let P rc Gi r Tyr 75 Val Le u Gl y Val Val 155 Gi y Arg Gl y Arg Arg 235 Ile Cys Glu Trp Leu Met Gly Met Val1 Cys Ile 140 Ile GI u Ile Gi y As n 220 Gly Arg Lys Arg Asp Pro Val Ph e G.1 y Ser Leu 125 Met Al a Ar g Al a Gi u 205 Leu Val Al a tksp Leu Le.
P rc Leu Ile Al a Val 110 Ser Gin Gi u Gly Met 190 Asp Phe Lys Lys Al a 270 Asp Tyr Gly Gin Tyr Asp His As n Ala Glu Leu 175 Arg Glu Ser Al a2 Ile 255 Leu C Met C Cys Thr Arg Lys As n T rp Leu Phe Val 160 Le u Ile Gln Leu krg 240 krg ln ;in 275 Ala Leu Lys Gin Ser Ser Thr Glu Leu Leu Phe Gi y 300 Gly His Giu Thr -66 Thr Ala Ser Ala Ala Thr Ser Leu Ile Thr Tyr Leu Gly Leu Tyr Pro 315 320 His Val Leu Cys Lys Ser Leu Lys Tyr 355 Pro Val Pro Gin Lys 325 Asn Gin 340 Ile Gly Vai Arg.Giu GiU le Lys Sex 330 Asp Asn Lys Leu Asp 345 Lys Giu Met Gitu Thr Leu Lys Giy Leu Leu 335 Thr Leu Giu Gin 350 Arg Leu Asn Pro 365 Phe Glu Leu Asn Cys Val Ile 360 Val1 Gly Gly Phe Aia Leu Lys 370 Gly Tyr Gin Ile Pro 385 Th r Lys 390 Asp Trp Asn Vai Ile 395 Lys Ser Ile Cys His Asp Vai Al a 405 Val Ile Phe Thr As n 410 Asp Giu Giu Phe As n 415 Asp Arg Phe Ile Pro Phe 435 Lys Ile Leu Pro His Pro Aia Ser Arg Giy Gly Leu Arg 440 Th r Cys Val Gly Lys 445 Arg Phe Ser Phe 430 Giu Phe Aia His Cys Asp
S.
*0 S
S*
S@
S
S S
S
S V Leu Lys Ile Val Giu Leu 450 Trp Gin Ala 460 Th r Leu Leu Asn 465 Tyr Gi y 470 Pro Thr Met Lys 475 Thr Ser Pro Thr Pro Val Asp As n 485 Leu Pro Ala Arg Phe Tyr Phe Gin Gi y 495
S.
5 INFORMATION FOR SEQ ID NO:33 SEQUENCE CHARACTERISTICS: (A)LENGTH: base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:33 CGCACCCCAG GAGGCGCGCT CGGAGGGAAG CCGCCACCGC CGCCGCCTCT GCCTCGGCGC GGAACAAACG GTTAAAGATT TTGGGCCASC GCCTCCGCGG GGGGAGGAGC CAGGGGCCCC AATCCCGCAA TTAAAGATGA ACTTTGGGTG AACTAATTGT CTGACCAAGG TAACGTGGGC AGCAACCTGG GCCGCCTATA. AAGCGGCAGC GCCGTGGGGT TTGAAGCGCT GGCGGCGGCG GCAGGTGGCG CGGGAGGTCG CGGCGCGCCA TGG 120 180 240 273 -67 INFORMATION FOR SEQ ID NO:34 SEQUENCE CHARACTERISTICS: LENGTH: base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:34 CGCACCCCCA GGAGGCGCGC TCAGAGGGAA GCCGCCAGTG C6CCGCCTCT GCCTCGGCGC GGAPLCAAACG GTTAAAGATT TTTTTGGGCA GCGCCTCGAG GGGGGAGGAG CCAGGGGCCC GATCCGCAA~mT TAAAGATGAA CTTTGGGTGA ACTAATTTGT CTGACCAAGG TAACGTGGGC AGTAACCTGG GCGGCCTTAT AAAGAGGGCG CGCGGCGGGG TTCGGAGCTA GGGAGGCGGC GGCAGGTGGC GCGGGAGGCT GAAGCGTGCC ATGG 120 180 240 274 64.06 0 0 9*0O 00 *se 0 0 00 0 0..:S INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 319 base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID TCGGGGGAAT TAACACCTTT TCAAAGTGAA ATCTCAGGAT TGGTATTAAA ATGCGCCTAT AACAAATGGT TGAGAGTTTG GCGGGGCGAG ATGACACCAC AATTAAAGAT GAACTTTGGG PLGTTAACAGG AGGAGACCTG CGCGCAATGG ATATATAAGG rCAGTTTGTG CGTAAAGACG CGTCTCCTCT CCAGAAGCTT GTTGCGCGCT TCAACATGG TGTCTGCCTT CTACAGGAGG GAGCCGCTTC TGCCCTGTGG rrQIACTAATT TATCTGAGGA 3CGCGCAGGC GAGGACGCCC 3TTTTTCGTT TTGGCGATCA 120 180 240 300 319 INFORMATION FOR SEQ ID NO:36 SEQUENCE CHARACTERISTICS: LENGTH: base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:36
GATCCCAGATCTGCCTATTGCGCCCGATGCCCCGAGGCTCTCTCTTGGAC
TCTGGCCCTGAGTTCTTCTGCGCGATCCTTCGGAGACGTCTGGAGGCCTG
CTTTATGCATCTCTCTTGGACCTCAGTTTCCCCACACGTGGGAGGAGGCA
GCTGGACGATTCCTGAAAGGACTTTCCCTTGCTtCCTCATCACGTGGAAG
AGAGCCCACCCGGCACCTGGAATGGAAAGCCAGTGAAGGCTGCTTTGGG
CCGGGGCAKCGGGTGGGACCGGGCGGGAGGGATTCCAAAGAGACCGCCGG
GAAGGCTAGAGCTTGGAATTCCGGCTCCTCGGAGTCCTGGCCCTCCC-CCA
CCGCCGCCTCGGAGCTCAGCACCTTGGATGGGGGAGCGGCGCTCC
TAGCCCCGCACCCCAGGAGGCGCGCTCGGAGGGAGCCGcr-cCGCCGCC
GCCTCTGCCTCGGCGCGGAACAAACGGTTAAAGATTTTGGGCCASCGCCT
CCCG7GAGGC6GCCATCGATAAAGAT
TGGGTGAACTAATTGTCTGACCAAGGTAACGTGGGCAGCAACCTGGGCCG
CCTATAAAGCGGCAGCGCCGTGGGGTTTGAGCGCTGGCGGCGGCGGCAG
GTGGCGCGGGAGGTCGCGGCGCGCCATGGGGCTCCCGGCGCTGCTGGCA
GTGCGCTCTGCACCTTCGTGCTGCCGCTGCTGCTCTTCCTGGCTGCGATC
AAGCTCTGGGACCTGTACTGCGTGAGCGGCCGCGACCGCAGTTGTGCCCT
CCCATTGCCCCCCGGGACTATSGGSTTCCCCTTCTTTGGGGAAACCTTGC
-68-
AGATGNTACTNCAGGTAAGGGAGGGTGGGGCGGGACAGGCTGCTTCCCCG
GAGCCCGGCGCGGCTCTGGGCTTCTGCTGAAGTCGGGGTAGGCGCCCCCG
GGAGGCATGCTATTGCGGCTAGGAGCAGGGCTGGCGGGAGCGCGGCGCTC
CCCGGMKYMCS CTCAWGCS CRCWWKTMWCCTCCGCCTYMCTCCCAMAGCG
GARSAARWKCYKGMRGATGAAGCGCAGGAAATACGGCTTCATCTACAAGA
CGCATCTGTTCGGGCGGCCCACCGTACGGGTGATGGGCGCGGACAATGTG
CGGCGCATCTTGCTCGGAGAGCACCGGCTGGTGTCGGTCCACTGGCCAGC
GTCGGTGCGCACCATTCTGGGATCTGGCTGCCTCTCTAACCTGCACGACT
CCTCGCACAAGCAGCGCAAGAAGGTGGGGGCAGGAGGCGACGGCTGGACA
GGGAGGGGGACCCCATTTATGAGCGGAATTCCGGCTGATGGATGCTAGGC
GCGGGCTAGCAGCTTGAGGTGGGCTAGGACCCTCTGCCAGCTCCAGGTTA
GCTTTCCCAGCTCGGAGAGTGCCATGTGTCTGGCAGGACTGGGGGTGTCT
GGAAGGGGACGGCGGTAGACGAGAGGGGCGGATGGAGGCTTTTAACGCTG
TCCCCTCCTCGGGACT CAGGTGATTATGCGGGCCTTCAGCCGCGAGGCAC
TCGAATGCTACGTGCCGGTGATCACCGAGGAAGTGGGCAGCAGCCTGGAG
CAGTGGCTGAGC.TGCGGCGAGCGCGGCCTCCTGGTCTACCCCGAGGTGAA
GCGCCTCATGTTCCGAATCGCCATGCGCATCCTACTGGGCTGCGAACCCC
AACTGGCGGGCGACGGGGACTCCGAGCAGCAGCTTGTGGAGGCCTTCGAG
GAAATGACCCGCAATCTCTTCTCGCTGCCCATCGACGTGCCCTTCAGCGG
GCTGTACCGGGTAAGGGCGGCAAACGGGCTGCGGACTAGGGGCGCGGGAC
CTGGGCGTCTGCTCACCGCCGCGCGCTCTCTGCGCTCAGGGCATGAAGGC
GCGGAACCTCATTCACGCGCGCATCGAGCAGAACATTCGCGCCAAGATCT
GCGGGCTGCGGGCATCCGAGGCGGGCCAGGGCTGCAAAGACGCGCTGCAG
CTGTTGATCGAGCACTCGTGGGAGAGGGGAGAGCGGCTGGACATGCAGGT
GAGTAGCAGCTTCAGACCAGGCACTGCGGAGTTTGGTCCC CTGGCTTTCC AAGGCGCTGTTCCT GGGGCCCCCAAAGCGCGCGCCTGGGGCCCAGCTTTC
TGGAGTGGGCGGCCGGCTCAGACTACAGCTATGGAATCCCGAAGGAAGGC
TGAGACACCCGGTCAGGAGAGCTGCGGALGGGGCTGCGGMGGAAACTGGG
*~.AGCATCCCCTAGCCTTTAMCAGGTTTCAAAGGGAAAGTTGGAATTTGCAA
9 AAATGTTAATAAAGAACCTTGCGATTTTAATAAAACTAAGACTTTAACTC 9. .AGGAGTTTCCGGTAGRGCGGGGTCGTACTCGCCTTACTGCTCCAGCTGAA
CTAAAGGGACGTTGCATTTTGTTTAAAGATATTGCTTTCCTTGACTTTCT
GTCAGCAAAACATTTAGCCCTTCTAGTCTTCCCTCCAGAACTCTCAGTTC
*GATTCTGAGTAATCCTTCTGTCAAACCGCAGGCAGACTTGTGAGAATGTG
GGTCTCACTCTATTCTTAGGCACTAAAGCAATCTTCAACCGAACTCCTCT
*.TTGGAGGACACG-AAACCACGGCCAGTGCAGCCACATCTCTGATCACTTAC
CTGGGGCTCTACCCACAT GTTCTCCAG INFORMATION FOR SEQ ID NO:37 SEQUENCE CHARACTERISTICS: LENGTH: base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:37
GATCCAGGTTGCTGAAACATATCTCCATATAGGGCAGAACAATTATCAAA
AGCATAAGAATTGCAGCCACAGCATAGGGAAGAAAGAGGAGTTTTTAAAC
CACAACAAAAGGGAGAAAGAAGAGAATTTTAACTTACATTTAATTCAAAA
GTCTTCAGAGCAACCCGAAACCCTCCTGGAACTGGGGGATTCAGTCGAAG
GGTCTCCTTAATAACACACCCGATGTATYTAAGTTGTTCCAAAATTTCCA
TGTCCAACTTGTTGTCTTGATTGCTCTTGAGTAACCCTAYCAAY
AGTCATACAGAGGTGACGTYATTTGTGCTCCAATTAAAATCAGCCCA
GCAGACGTAAACAGGGCTTAAGTGGAGACTAAACCCAAAGGGCCCCATGA
TGGGAGAGACTGGGAGGGGGTAAACAGCAGCTAATGGCCATTTGCCTGCCC
AAATCCACTATCTATTTACAATCCCAGGAGAATGCTGCTCACCAGTTAGA
AGGACCAAGTTTCTCCCCACGCCCCCCCALCCCCACACTCACCACC-ACCAC
CC-ACACTAATCAGCTATTCACACTATGTATGCCCTTGGACACACCAATTC
AAGAAAAGTGGAACCTATCTGAGAATCTCCACGGTTCACAAAAAGGTGGA.
GGAGGGGTAGGAATACAAGGTCAAACCCTGCCC
INFORMATION FOR SEQ ID NO:38 SEQUENCE CHARACTERISTICS: -69- (A)LENGTH: base pairs TYPE: nucleic acid TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:38
TCGCGAGGAGCGACCCGGCTTGAGAGGGGTAGACGAGACCAGATGCTC
CCCGGCGCCCCCTCATGCGGGTTGCGGTCTCTCTCCTCCACCTCCCTCTC
AGCGGAGGAAGTTTCTGCAGATGAAGCGCAGGAAATACGGCTTCA7TCTAC AAGACGCATCTGTTTGGGCGGCCCACGGTGCGGGTGATGGGCGC
SGATAA
TGTGCGGCGCATCTTGCTGGGAGAGCACCGGTTGGTGTCGGTGCACTGGC
CCGCGTCGGTGCGCACCATCCTGGGCGCTGGCTGCCTCTCCAACCTGCAC
GATTCCTCGCACAAGCAGCGAGAAGGTGAGGGTGAGCTGCACTCCT
TGGCTGGCAGGGAGACCTCATCCTATGGCTTGGTTCAGGCAAATAGAT
GCGGGGCGAGGGCTAGTCCTAT
GTGGTGGGGACCAGGACCCTCTCTATCT
GAGATCCACTTTAGCTTTTCTGCTAGCACGTGGGTTAGTCCTGGGGGGGA
CTAATTGAGGATGAAGCAGGGGGCGG
GAAAGTAGAGGATTGTAACACT
CTCTGCTCCTGGGGGGTGCTCAGGTGAT
TATGCAGGCCTTCAGCCGCGAGGCACTCCAGTGCTACGTGCCCGTGATCG
CTGAGGAGTCAGCGTTGTCTGGAGCAGTGGCTAAGCTGCGGCGGCGC
GGCCTCCTGGTCTACCCCGAGGTGAAGCGCCTCATGTTCCGCATCGCCAT
GCGCATCCT GCTGGGCTGCGAGCCGGGTCCAGCGGGCGGCGGGAGGCG
AGCAGCAGCTCGTGGAGGCTTTCGAGGAGATGACCCGCATCTCTTCTCT
CTTCCCATTGACGTGCCCTTTAGCGGCCTGTACCGGGTAAGGGCGGTTTG
CGGAGTCGGAGTAGGGGACGCAAGCTCGGGCATCCGCTCACCGCCACGC
TCTCTCCGCGCTCAGGGCGTGAGGCGCGGACCTTATACACGCGCGCT
CGAGGAGAACATTCGCGCCAAGATCCGCCGGCTTCAGGCTACAGAGCCGG
ATGGGGGTTGCAAGGACGCGCTGCAGCTCCTGATTGAGCACTCGTGGGAG
*.AGGGGAGAGAGGCTGGATATGCAGGTGAGAAGCATTTCA.PAGGTGCCA
AGGGCCGGGGAGTGCCTCTGACTTTCCAGACACACTTTCTGGGGTCTCCA
AAGCCCTGTCAAGGCCCCAGCTACTTCCAAGTGGGCGGCGATGCTAGGTC
TAGAGCTTTTCAACCTGTGGGTCGTGACCCCTTCACGGAGCCAAACAACC
CTTTCAGAGGGTCGCCTAGAGCATCTGATATCCGATATTTAATA
::GAAACATAACAGTAGCAAATTACCGTTATGAAGTAGCAACAAAGATAT
*TTTATCGTTGGGGGTCACCACAACGAGGACCGTATTAAGGGTGGCA
TTGGTCTAGAGAGCTGTGGAGGGGGTGGCTGAGCATGGGAGA~TCCC.
AAGTTCAAAGGGCAAGGCTCATCTACAAAGGTTAAGCGGAGAGCAGG
ATAGGGTTCTTTTTGGTTTATTTTAC
AAACGGATTTTACCCTTGAAGTCTTCCGTGCAATATTCTCAGGTCAGGTC
~TAT-CCTTAGGCACTAACATCGTCACAGGCTCCTCTTTGGTGGTCA
TGAACTACAGCCAGTGCTGCGCGTCACTGATCACTTACCTAGGACTCT
AC CCACAT GT CCTC CAGAAAGTT CGAGAAGAGATAAAGAGCAAGGTAGGA TGATTCTAGAGGTTCCCCATTTGCCTAGGACATTCCT
CTATTAACCACCA
**CCACCACCCCCACTGTATATAAGTTTGCTCGATACACCCAGTACTATGAC
AGTGAAGATCTGAGAGCTAGGTGGGACTGTGGGGGA~CTCCACCTCG
TGAATTTAAAAAGGCAGTTGTTTGTACTGGGCTCTCTCTTGGG
CAGAATT
TGACCCTCTCCTCCTCCTCCTCCTCCTCCTCCTCTTCCTCCTCCACCACC
ACCACCATCACCACCTTTTATAGAGCAAJGGTTCTCCTTTCCCTGACCAAG
AAAGAATTATGGCAACGTAGT~-GGT
GTGAGGGCTCAGGGTATGACCCTTTATATACCTGATAGCAACATTGTCT
:GGATAATGGGTTTAGGCTGAGGGTGTG
.PGGAGGCCATCAGGCCA
TCGCTCTTTCCCTTTTATCCTCTCCCATCCAGACGCCTTCGGTTTAGT
TACAGGTGAGTCCTGCTGGGCTGACTTTTTTTTTGGGTGCCCAGGGAT
CCTA~-CTTTTTTTCAAGrTATTCAAC ATAcAAcA-TAAAGAACTGCrACTATCC GGGTGTGTCATTAAGGA~cCCTGCGATTGTCCTCCGGTTCCAAG
GTTTCGGGTTGCTCTGAGACTTTTGAGCTGAATGTGGTGCACCTCCTG.
TCCCCCACCCCCAGCCCTCGTCCACGTCCACTCTGCTATGCTGTTGAGCA
TCAGCTGCCCAGAGCAGTGGCTCACTGCCCTTGACAGTGTCCTGCCTCCT
ATGGTACTGGGAACCAJTTTGCTCTCCTCTCTTAATGCCATCCATGCTAG
TATGACTTTTTGTTGTTGCAAGCTCAGGGCCGGGATTGTCAATTCTTAG
GATTTTTTTTTTTTTTTCAGGATACCGATCC
CCAAGGGCTGG.AT
GTTATTTACAGTATCTGTGACACCCACATGTGGCAGATATCTTCACTA
CAAGGAGGAATTTAATCCCGACCGCTTTATAGTGCCTCATCCAGAGGATG
70
CTTCCCGGTTCAGCTTCATTCCATTTGGAGGAGGCCTTCGGAGCTGTGTA
GGCAAAGAGTTTGCAAAAATTCTTCTTAAGATATTTACAGTGGAGCTGGC
TAGGCACTGTGATTGGCAGCTTCTAAATGGACCTCCTACAATGAAGACAA
GCCCCACTGTGTACCCTGTGGACAATCTCCCTGCAAGATTTACCCACTTC
CAGGGAGATATCTGATAGCTATTTCAATTCTTGGACTTATTTGAAGTGTA
TATTGTTTTTTTTAAAATAGTGTCATGTTGACTTTATTTAATTTCTAAAT
GTATAGTATGATATTTATGTGTCTC TACTACAGTCCCGTGGTCTTAAATA
TTAAATAATGAATTTGTATGATTTCCCAATAAAGI'AAAATTAAAAAGTG
CTTCTCTTGCTTTTTAAGATTCTTGTTGGCAAGCTGCCCATGGTGGTACA
TTGCTGTAATACTAGGACTTGGAAGGTGGAGGCAAGAAGAGCAGGCATTC
P.AGGCTAGCCTGGGCTACAGAAATCCTGTCTTAAACAAACACTACAACAA
AAAGTCCTGTTAGGGAATCTGACTGGCTCAGTGTTTGTACTTTGTGTATT
TAAAATGATTTAGAGTGAAACCATAGGTCTCTCCCCCATGTCAGAAAATA
TATATTATTATGTGTATGCTGATCCAAAGTATCTTTGTAACTTTTTCTAA
GGTCATTGAGACTTCATATTTTGAAATTGTATGGAGGCTAGTTATATTAC
ATTATTTATTTATTTATTTATTTACATTTTTATGGTGCTGGGGATTGGAT
CGAAGGCTTCACACCTCTAGGGCAAGCCCTTTGTCATTAAGGCGCTGCCT
CTCCCTTTCAGCCCAACGTTAATTCTAGATTCTTTTTCTTTGGTGCTTTT
GGGAGGTAAACCTGGGATGCTGCAGTTATTTGGTGGTGGTCGTTGGTTTT
ACTCTAGAGAGAAGGCAACTTTGGGAAGGCAACACTGCTGCTGGTGAGTC
GGGAAGCATCATCCCAGAGCAACGGGGTCAGCATAGCTAACATTTTAAAT.
CAGCATAATGAATCCCT GTCATATGGAGGAGGCAGAACTCCT CTTTGAAG
TTGATATTTTAGATAAGACAGAGCCAGCCCCTCTGGTTATGGACAGTTCT
TACCCAAAATGAAACAGAGAAGAAAACCACTGGTGTGTCACCTTTCCTTA
GAAGTGCTTCAGGA

Claims (102)

19.0ct. 2004 16:00 Shelston IP No. 1810 P. -71- THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:- 1. An isolated nucleic acid molecule comprising a nucleotide sequence that hybridizes under high stringency conditions, wherein high stringency conditions include a wash step of about 0.2xSSC at 50 0 C, to the nucleotide sequence shown as SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:31, and encodes a protein that oxidizes a retinoid. 2. An isolated nucleic acid molecule comprising a nucleotide sequence that hybridizes under high stringency conditions, wherein high stringency conditions include a wash step of about 0.2xSSC at 50C, to the nucleotide sequence shown as SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:31, and encodes a protein that hydroxylates a 10 retinoic acid at the 4-position of the P-ionone ring. 3. The isolated nucleic acid molecule of claim 1 or 2, wherein the protein comprises an amino acid sequence that is at least 60 percent conserved with respect to SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32. 4. The isolated nucleic acid molecule of claim 1 or 2, wherein the protein comprises an amino acid sequence that is at least 70 percent conserved with respect to SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32. 5. The isolated nucleic acid molecule of claim 1 or 2, wherein the protein comprises an amino acid sequence that is at least 80 percent conserved with respect to SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32. 9. 20 6. The isolated nucleic acid molecule of claim 1 or 2, wherein the protein comprises 9 an amino acid sequence that is at least 90 percent conserved with respect to SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32. 7. The isolated nucleic acid molecule of claim 1 or 2, wherein the protein comprises an amino acid sequence that is at least 95 percent conserved with respect to SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32. 8. An isolated nucleic acid molecule comprising a nucleotide sequence identified as SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:31, or which varies from a said sequence in a coding region due to the degeneracy of the genetic code. 9. Isolated mRNA transcribed from DNA comprising a nucleic acid molecule of claim 3, wherein the mRNA is the full length of said nucleotide sequence. COMS ID No: SBMI-00961543 Received by IP Australia: Time 16:07 Date 2004-10-19 19. Oct. 2004 16:01 Shelston IP No. 1810 P. 6 -72- Isolated DNA comprising a nucleic acid molecule of claim 3 in a recombinant cloning vector. 11. The nucleic acid molecule of any one of claims 1 to 8 wherein the sequence of the nucleic acid molecule corresponds to a part of a fish genome, or of a human genome, or of a mouse genome, or varies there from due to the degeneracy of the genetic code. 12. An isolated nucleic acid molecule encoding a protein comprising an amino acid sequence selected from the group consisting of SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32, or a conservatively substituted variant thereof. 13. The nucleic acid molecule of claim 1, wherein the retinoid is a rctinol or a 1jo retinoic acid. 14. The nucleic acid molecule of claim 1, wherein the protein oxidizes the carbon at the 4-position of the p-ionone ring of the retinoid. 15. The nucleic acid molecule of claim 1, wherein the retinoid is an all-trans retinoid. 16. The nucleic acid molecule of claim 2, wherein the retinoic acid is all-trans. :15 17. An isolated nucleic acid molecule which is antisense to the full length of the nucleic acid molecule of claim 1 or 2. 18. An isolated nucleic acid molecule which is antisense to a nucleic acid molecule •comprising a nucleotide sequence shown as SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:31. 20 19. An isolated nucleic acid molecule that hybridizes under high stringency conditions, wherein high stringency conditions include a wash step of about 0.2xSSC at 0 C, to 5' untranslated region of the nucleotide sequence identified as SEQ ID NO:3. A recombinant expression vector suitable for transformation of a host cell comprising the nucleic acid molecule of any one of claims 1 to 8 or 11 to 16 and at least one regulatory sequence operatively linked to the nucleic acid molecule.
21. The recombinant expression vector of claim 20, wherein the nucleic acid molecule is operatively linked to said at least one regulatory sequence to allow expression of said encoded protein. COMS ID No: SBMI-00961543 Received by IP Australia: Time 16:07 Date 2004-10-19 19. Oct. 2004 16:01 Shelston IP No. 1810 P. 7 -73-
22. The recombinant expression vector of claim 20, wherein the nucleic acid molecule is operatively linked to said at least one regulatory sequence to allow expression of an RNA molecule which is antisense to the nucleic acid molecule.
23. A transformant host cell including the recombinant expression vector of claim 21.
24. A transformant host cell including the recombinant expression vector of claim 22. The transformant host cell of claim 23 or 24 which is a mammalian cell.
26. A microbial cell containing and expressing heterologous DNA encoding a 10 protein that oxidizes a retinoid, the DNA comprising the nucleic acid molecule of any one of claims 1 to 8 or 11 to 16.
27. The microbial cell of claim 26, wherein the protein has all-trans-retinoic acid 4- hydroxylase activity.
28. A microbial cell containing and expressing heterologous DNA which is complementary to the full coding region of the nucleic acid molecule of any one of claims 1 to 8 or 11 to 16. S29. A stably transfected cell line which expresses a protein that oxidizes a retinoid, the cell line having incorporated transfected DNA encoding said protein and comprising the nucleic acid molecule of any one of claims 1, 3 to 8 or 11 to 20 30. A stably transfected cell line which expresses a protein that hydroxylates retinoic acid at the 4-position of the p-ionone ring, the cell line having incorporated transfected DNA encoding said protein, and comprising the nucleic acid molecule of any one of claims 2 to 8, 11, 12 or 16.
31. The cell line of claim 29 or 30 which is a mammalian cell line.
32. A process for producing a retinoid metabolizing protein, comprising the steps of: preparing a DNA fragment comprising the nucleic acid molecule of any one of claims 1 to 8 or 11 to 16; incorporating the DNA fragment into an expression vector to obtain a recombinant DNA molecule which comprises the DNA fragment and which undergoes replication in a cell; COMS ID No: SBMI-00961543 Received by IP Australia: Time 16:07 Date 2004-10-19 19.0ct. 2004 16:01 Shelston IP No. 1810 P. 8 -74- transforming a host cell with the recombinant DNA molecule to produce a transformant cell which expresses said protein under pre-selected conditions; culturing the transformant cell to produce said protein; and recovering said protein from resulting culture mixture.
33. A method for preparing a protein comprising culturing a transformant host cell including the recombinant expression vector of any one of claims 10, 20 or 21 in a suitable medium until the protein is formed and isolating the protein.
34. A microsomal preparation comprising a recombinant protein expressed by a cell that has been transfected with a nucleic acid molecule encoding the protein, or by a t descendent cell thercof, wherein said protein oxidizes a retinoid, said nucleic acid S: molecule comprising a nucleotide sequence that hybridizes under high stringency conditions, wherein high stringency conditions include a wash step of about 0.2 x SSC at 50°C, to a polynucleotide having the nucleotide sequence shown as SEQ ID NO:3, SEQ D NO:5 or SEQ ID NO:31.
35. A microsomal preparation comprising a recombinant protein expressed by a cell that has been transfected with a nucleic acid molecule encoding the protein, or by a descendent cell thereof, wherein said protein bydroxylates a retinoid at the C4-position of the -ionone ring, said nucleic acid molecule comprising a nucleotide sequence that hybridizes under high stringency conditions, wherein high stringency conditions include 20 a wash step of about 0.2 x SSC at 50°C, to a nucleic acid molecule having the nucleotide sequence shown as SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:31.
36. The preparation of claim 34 wherein said oxidation is at the C4-position of the 3- ionone ring of the retinoid.
37. The preparation of any one of claims 34 to 36, wherein the retinoid is all-trans retinoic acid.
38. The preparation of any one of claims 34 to 37, wherein the amino acid sequence identity between the protein and SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32 is at least about 93 percent. 39, The preparation of any one of claims 34 to 38, wherein the protein comprises the amino acid sequence identified as SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32. COMS ID No: SBMI-00961543 Received by IP Australia: Time 16:07 Date 2004-10-19 19. ct. 2004 16:02 Shelston IP No. 1810 P. 9 The preparation of any one of claims 34 to 39, wherein the protein oxidizes the C18-position of all-trans retinoic acid.
41. A microsomal preparation comprising a recombinant protein expressed by a cell that has been transfected with a nucleic acid molecule encoding the protein, or by a descendent cell thereof, wherein said protein oxidizes all-trans retinoic acid at the C4- position of the p-ionone ring, said nucleic acid molecule encoding an amino acid sequence that is at least 60 percent conserved with respect to SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32.
42. The preparation of claim 41, wherein the nucleic acid molecule encodes an amino acid sequence that is at least 95 percent conserved with respect to SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32. 0
43. The preparation of any one of claims 34 to 42, wherein the microsomal S. preparation is substantially free of other proteins that are cytochromes expressed by epidermal cells.
44. The preparation of claim 34, wherein the preparation is enriched in said oxidase activity at least 6.3 fold with respect to a microsomal preparation obtained from a non- transfected said cell under the same conditions.
45. The preparation of claim 35, wherein the preparation is enriched at least 7.8 fold in said hydroxylase activity with respect to a microsomal preparation obtained from a 20 non-transfected said cell under the same conditions.
46. A probe comprising an isolated nucleic acid molecule comprising: a sequence of at least 20 consecutive bases selected from bases 1 to 884 and 1257 to 1494 of SEQ ID NO:5, or the complement thereof; (ii) a sequence of at least 20 consecutive bases selected from SEQ ID NO:3, or the complement thereof; or (iii) a sequence of at least consecutive bases selected from SEQ ID NO:31, or the complement thereof.
47. The probe of claim 46, the sequence comprising at least 30 consecutive bases selected from: bases 1 to 884 and 1257 to 1494 of SEQ ID NO:5, or the complement thereof; SEQ ID NO:3, or the complement thereof; or SEQ ID NO:31, or the complement thereof.
48. The probe of claim 46, the sequence comprising at least 40 consecutive bases selected from: bases 1 to 884 and 1257 to 1494 of SEQ ID NO:5, or the complement COMS ID No: SBMI-00961543 Received by IP Australia: Time 16:07 Date 2004-10-19 19.0ct. 2004 16:02 Shelston IP No. i810 P. -76- thereof; SEQ ID NO:3, or the complement thereof; or SEQ ID NO:31, or the complement thereof.
49. The probe of claim 46, the sequence comprising at least 50 consecutive bases selected from: bases 1 to 884 and 1257 to 1494 of SEQ ID NO:5, or the complement thereof; SEQ ID NO:3, or the complement thereof; or SEQ ID NO:31, or the complement thereof. A kit for determining the presence of a nucleic acid strand, the kit comprising the probe of any one of claims 46 to 49.
51. The kit of claim 50, further comprising a reporter system wherein the reporter 10 system produces a detectable response when a predetermined amount of the nucleic acid strand and the nucleic acid molecule are hybridized with each other. S52. A method of screening drugs for their effect on activity of a retinoid metabolizing protein comprising exposing the microsomal preparation of any one of claims 33 to 45 to a said drug and determining the effect on the activity of the protein.
53. A method of claim 52, wherein the activity is oxidation of a retinoid. 1 54. A method of claim 52, wherein the activity is hydroxylation of a retinoid.
55. A method of claim 53 or 54, wherein the retinoid is a retinoic acid.
56. A method of claim 55, wherein the retinoic acid is all-trans-retinoic acid.
57. A method of screening drugs for their effect on activity of a retinoid 20 metabolizing protein, comprising exposing a cell that has been transfected with a nucleic acid molecule encoding the protein, or a descendent cell thereof, to a said drug, wherein said transfected cell or descendent cell thereof expresses the protein, and determining the effect on the activity of the protein, wherein said protein oxidizes a retinoid, said nucleic acid molecule comprising a nucleotide sequence that hybridizes under high stringency conditions, wherein high stringency conditions include a wash step of about 0.2 x SSC at 0 C, to the nucleotide sequence shown as SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:31.
58. The method of claim 57, wherein the nucleic acid molecule encodes an amino acid sequence that is at least 60 percent conserved with respect to SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32. COMS ID No: SBMI-00961543 Received by IP Australia: Time 16:07 Date 2004-10-19 19.0ct. 2004 16:02 Shelston IP No. 1810 P. 11 -77-
59. The method of claim 58, wherein the nucleic acid molecule encodes an amino acid sequence that is at least 95 percent conserved with respect to SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32. The method of any one of claims 57 to 59, wherein the method includes determining the effect a said drug has on the oxidation of a retinoid which is a retinoic acid.
61. The method of claim 60, wherein the retinoic acid is all-trans-retinoic acid.
62. A method of screening drugs for their effect on activity of a retinoid metabolizing protein, comprising exposing a cell that has been transfected with a nucleic 10 acid molecule encoding the protein, or a descendent cell thereof, to a said drug, wherein said transfected cell or descendent cell thereof expresses the protein, and determining the effect on the activity of the protein, wherein said protein hydroxylates a retinoid at the 4-position of the j-ionone ring, said nucleic acid molecule comprising a nucleotide :sequence that hybridizes under high stringency conditions, wherein high stringency conditions include a wash step of about 0.2 x SSC at 50*C, to the nucleotide sequence shown as SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:31.
63. The method of claim 62, wherein the nucleic acid molecule encodes an amino acid sequence that is at least 60 percent conserved with respect to SEQ ID NO:2, SEQ :ID NO:4, or SEQ ID NO:32. 20 64. The method of claim 63, wherein the nucleic acid molecule encodes an amino *i *acid sequence that is at least 95 percent conserved with respect to SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32. The method of any one of claims 62 to 64, wherein the method includes determining the effect a said drug has on the hydroxylation of a retinoid which is a retinoic acid.
66. The method of claim 65, wherein the retinoic acid is all-trans-retinoic acid.
67. A method of screening drugs for their effect on activity of a retinoid metabolizing protein comprising exposing the stably transfected cell line of any one of claims 29 to 31 to a said drug and determining the effect on the activity of the protein.
68. The method of claim 67, wherein the activity is oxidation of a retinoid. COMS ID No: SBMI-00961543 Received by IP Australia: Time 16:07 Date 2004-10-19 19.0ct. 2004 16:03 Shelston IP No. 1810 P. 12 -78-
69. The method of claim 67, wherein the activity is hydroxylation of a retinoid. The method of claim 68 or 69, wherein the retinoid is a retinoic acid.
71. The method of claim 70, wherein the retinoic acid is all-trans-retinoic acid.
72. A method of screening a drug for its effect on the metabolism of a retinoid by a cytochrome P450 encoded by the nucleic acid molecule of any one of claims 1 to 8 or 11 to 16 which is incorporated into an expression system so as to be under control of a nucleotide sequence having native promoter activity for the cytocbrome P450, comprising: exposing the system to the drug in the presence of the retinoid so as to 10 determine the effect of the drug on metabolism of the retinoid.
73. The method of claim 72, wherein the retinoid is an all-trans-retinoid. 0*00
74. The method of claim 73, wherein the retinoic acid is all-trans-retinoic acid.
75. An antibody which specifically binds to an unconserved region of a protein that oxidizes a retinoid and is encoded by a nucleic acid molecule comprising a nucleotide i 15 sequence that hybridizes under high stringency conditions, wherein high stringency conditions include a wash step of about 0.2xSSC at 50 0 C, to the nucleotide sequence shown as SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:31.
76. An antibody which specifically binds to an unconserved region of a protein that hydroxylates a retinoic acid at the 4-position of the 1-ionone ring and is encoded by a 20 nucleic molecule comprising a nucleotide sequence that hybridizes under high stringency conditions, wherein high stringency conditions include a wash step of about 0.2xSSC at 50 0 C, to the nucleotide sequence shown as SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:31.
77. An antibody which specifically binds to an unconserved region of a protein encoded by a nucleic molecule comprising a nucleotide sequence identified as SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:31, or which varies from a said sequence in a coding region due to the degeneracy of the genetic code.
78. The antibody of claim 75, wherein the retinoid is a retinol or a retinoic acid.
79. The antibody of claim 78, wherein the protein oxidizes the carbon at the 4- position of the p-ionone ring of the retinoic acid. COMS ID No: SBMI-00961543 Received by IP Australia: Time 16:07 Date 2004-10-19 19. Oct. 2004 16:03 Shelston IP No. 1810 P. 13 -79- The antibody of claim 75, wherein the retinoid is an all-trans retinoid.
81. The antibody of claim 76 or 79, wherein the retinoic acid is all-trans.
82. An antibody which specifically binds to a protein having the sequence identified as SEQ ID NO:4.
83. An antibody which specifically binds to a protein having the sequence identified as SEQ ID NO:2.
84. An antibody which specifically binds to a protein having the sequence identified as SEQ ID NO:32. An antibody which specifically binds to an epitope located within an unconserved region of a protein, the unconserved region consisting of amino acids 1 to 432 of SEQ ID NO:2.
86. The antibody of claim 85, wherein the epitope comprises amino acids 70 to 74 of SEQ ID NO: 2.
87. An antibody which specifically binds to an epitope located within an 15 unconserved region of a protein, the unconserved region consisting of amino acids 1 to 436 ofSEQ ID NO:4.
88. The antibody of claim 87, wherein the epitope comprises amino acids 70 to 74 of SEQ ID NO:4.
89. An antibody which specifically binds to an epitope located within an 20 unconserved region of a protein, the unconserved region consisting of amino acids 1 to 436 of SEQ ID NO:32. The antibody of claim 89, wherein the epitope comprises amino acids 70 to 74 of SEQ ID NO:32.
91. The antibody of any one of claims 75 to 81, wherein the amino acid sequence identity between the protein and SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:32 is at least about 93 percent.
92. The antibody of any one of claims 75, 76 or 78 to 81, wherein the nucleic acid molecule encodes an amino acid sequence that is at least 60 percent conserved with respect to SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32. COMS ID No: SBMI-00961543 Received by IP Australia: Time 16:07 Oate 2004-10-19 19. Oct. 2004 16:03 Shelston IP No. 1810 P. 14 80
93. The antibody of any one of claims 75 to 81, wherein the nucleic acid molecule encodes an amino acid sequence that is at least 95 percent conserved with respect to SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:32.
94. The antibody of any one of claims 75 to 85, 87, 89 or 91 to 93, wherein the antibody does not bind only to a heme-binding motif of the protein corresponding to amino acid residues from about 431 to about 449 of SEQ ID NO: 2, or only to a heme- binding motif of the protein corresponding to amino acid residues from about 435 to about 453 of SEQ ID NO: 4, or only to a heme-binding motif of the protein corresponding to amino acid residues from about 435 to about 453 of SEQ ID NO: 32.
95. The antibody of any one of claims 75 to 94, wherein the protein is a recombinant protein. The antibody of any one of claims 75 to 95, wherein the antibody is a :monoclonal antibody. co 0
97. A kit for determining the presence of the protein, comprising the antibody of any one of claims 75 to 96 linked to a reporter system, wherein the reporter system produces a detectable response when a predetermined amount of the protein and the antibody are bound together.
98. The protein of any one of claims 34 to 45 for use in metabolizing retinoic acid in an organism or cell in need of such metabolizing.
99. Use of the protein of any one of claims 34 to 45 for preparing a composition for metabolizing retinoic acid in an organism or cell in need of retinoic acid metabolizing.
100. Use of an antisense nucleic acid or oligonucleotide substantially complementary to at least a portion of the sequence identified as SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:31, wherein the portion is at least 5 bases in length, for preparing a medicament for inhibiting retinoic acid hydroxylation in an organism in need of such inhibition.
101. The use of claim 99 or 100 wherein the organism is human.
102. The use of claim 101 wherein the organism is being treated for a disease selected from the group consisting of cancer, actinic keratosis, oral leukoplakia, a secondary tumor of the head and/or neck, a non-small cell lung carcinoma, a basal cell carcinoma, COMS ID No: SBMI-00961543 Received by IP Australia: Time 16:07 Date 2004-10-19 19. 0ct. 2004 16:04 Shelston IP No. 1810 P. -81. acute promyelocytic leukemia, skin cancer, and a premalignancy associated actinic keratosis, acne, psoriasis and/or ichthyosis.
103. The use of claim 102 wherein the disease is acute promyelocytic leukemia.
104. An isolated DNA molecule comprising a nucleotide sequence selected from the group consisting of: SEQ ID NO:33; SEQ ID NO:34; SEQ ID NO:35; and a fragment of or 10 wherein said DNA molecule possesses promoter activity.
105. The DNA molecule of claim 104 wherein said DNA molecule includes the sequence TGAACT(N)xTGAACT, wherein x has a value of up to *106. The DNA molecule of claim 105 wherein x is
107. The DNA molecule of claim 105 further including the sequence 15 TCTGASSAAGKTAAC downstream from the sequence TGAACT(N)xTGAACT.
108. The DNA molecule of claim 107 further including the sequence AATT between the sequence TGAACT(N)XTGAACT and the sequence TCTGASSAAGKTAAC.
109. The DNA molecule of claim 107 wherein there are up to six nucleotides between the sequence TGAACT(N)xTGAACT and the sequence TCTGASSAAGKTAAC.
110. The DNA molecule of claim 105 further including the sequence CAATTAAAGA upstream of the sequence TGAACT(N)xTGAACT.
111. The DNA molecule of claim 104 wherein said DNA molecule includes the sequence CAATTAAAGATGAACTTTGGGTGAACTAATT,
112. The DNA molecule of any of claims 104 to 111 wherein said DNA molecule includes the sequence TATAA.
113. The DNA molecule of any of claims 105, 106 or 110, wherein said DNA molecule includes the sequence TATAA downstream of the sequence TGAACT(N)xTGAACT. COMS ID No: SBMI-00961543 Received by IP Australia: Time 16:07 Date 2004-10-19 19. Oct. 2004 16:04 Shelston IP No. 1810 P. 16 82
114. The DNA molecule of claim 107 or 108, wherein said DNA molecule includes the sequence TATAA downstream of the sequence TCTGASSAAGKTAAC.
115. The DNA molecule of claim 105 wherein said DNA molecule includes the sequence TATAA located downstream of the sequence TGAACT(N)TGAACT and spaced up to about 55 nucleotides there from.
116. A recombinant DNA comprising the isolated DNA molecule of any one of claims 104 to 115.
117. The recombinant DNA of claim 116, further comprising one or more structural genes. 10 118. The recombinant DNA of claim 117, wherein the one or more structural genes oencodes a cytochrome P450 protein(s).
119. An expression plasmid comprising the recombinant DNA of claim 116.
120. An isolated cell containing the recombinant DNA of claim 117.
121. The isolated cell of claim 120 wherein said cell is eukaryotic. S"15 122. A process for the production of protein(s), comprising culturing the cell of claim .00. 120 and recovering the protein(s) produced. 66-6 123. The process of claim 122 wherein the protein(s) comprise a cytochrome P450.
124. The process of claim 122 wherein the protein(s) comprise a fusion protein.
125. A method of screening drugs for their effect on expression of a gene wherein the gene is inducible by a retinoid, comprising exposing the recombinant DNA of claim 117 to a said drug and determining the effect on gene expression.
126. The method of claim 125 including exposing the recombinant DNA in the presence of a said retinoid.
127. The method of claim 125 wherein said structural gene is a reporter gene which does not metabolize retinoic acid.
128. A method of screening drugs for their effect on expression of a gene wherein the gene is inducible by a retinoid, comprising exposing the recombinant DNA of claim 116 to a said drug and determining the effect on gene expression, wherein determining the effect on gene expression includes ascertaining the effect on transcription of the gene. COMS ID No: SBMI-00961543 Received by IP Australia: Time 16:07 Date 2004-10-19 19.00. 21004 16:04 Shelston 1P INo.1810 P.I 83
129. An isolated nucleic acid molecule according to any one of claims I to 8 or 11 to 19 and substantially as herein described wit reference to any one of the examples but excluding comparative examples.
130. Isolated mENA according to claim 9 and substantially as herein described with reference to any one of the examples but excluding comparative examples.
131. Isolated DNA according to claim 10 and substantially as herein described with reference to any one of the examples but excluding comparative examples.
132. A recombinant expression vector according to any one of claims 20 to 22 and substantially as herein described with reference to any one of the examples but 1 excluding comparative examples.
133. A transforrnant host cell according to any one of claims 23 to 25 and substantially as herein described with reference to any one of the examples but excluding comparative examples.
134. A microbial cell according to any one of claims 26 to 28 and substantially as herein described wit reference to any one of the examples but excluding comparative examples. *135. A stably transfected cell line according to any one of claims 29 to 31 and substantially as herein described with reference to any one of the examples but excluding comparative examples.
136. A process for producing a retinoid metabolizing protein, substantially as herein described wvith reference to any one of the examples but excluding comparative examples.
137. A microsomal preparation, substantially as herein described with reference to any one of the examples but excluding comparative examples.
138. A probe according to any one of claims 46 to 49 and substantially as herein described with reference to any one of the examples but excluding comparative examples.
139. A kit according to claim 50 or 51 and substantially as herein described with reference to any one of the examples but excluding comparative examples. COMS ID No: 5BMI-00961543 Received by IP Australia: Time 16:07 Date 2004-10-19 19. Oct. 2 00 4 16:0 5 S he Is t on I P No. 1810 P. 18 84
140. A method of screening drugs for their effect on activity of a retinoid metabolizing protein, substantially as herein described with reference to any one of the examples but excluding comparative examples.
141. An antibody which specifically binds to an unconserved region of a protein that oxidizes a retinoid, substantially as herein described with reference to any one of the examples but excluding comparative examples.
142. An isolated DNA molecule according to claim 104 and substantially as herein described With reference to any one of the examples but excluding comparative examples. io1 DATED this 19"' day of October 2004 Sheiston TP Attorneys for: Queen's University At Kingston so. COMS ID No: SBMI-00961543 Received by IP Australia: Time 16:07 Date 2004-10-19
AU65483/01A 1996-06-21 2001-08-27 Retinoid metabolizing protein Ceased AU778362B2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US08667546 1996-06-21
US08724466 1996-10-01
AU31620/97A AU3162097A (en) 1996-06-21 1997-06-23 Retinoid metabolizing protein
PCT/CA1997/000440 WO1997049815A1 (en) 1996-06-21 1997-06-23 Retinoid metabolizing protein

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU31620/97A Division AU3162097A (en) 1996-06-21 1997-06-23 Retinoid metabolizing protein

Publications (2)

Publication Number Publication Date
AU6548301A AU6548301A (en) 2001-10-25
AU778362B2 true AU778362B2 (en) 2004-12-02

Family

ID=33545807

Family Applications (1)

Application Number Title Priority Date Filing Date
AU65483/01A Ceased AU778362B2 (en) 1996-06-21 2001-08-27 Retinoid metabolizing protein

Country Status (1)

Country Link
AU (1) AU778362B2 (en)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
RANER ET AL, LEO ET AL (SEE 31620/97) *

Also Published As

Publication number Publication date
AU6548301A (en) 2001-10-25

Similar Documents

Publication Publication Date Title
US5639616A (en) Isolated nucleic acid encoding a ubiquitous nuclear receptor
EP0910644B1 (en) Retinoid metabolizing protein
EP1007074A1 (en) Hypothalamus-specific polypeptides
US7579329B2 (en) P450RAI-2 (P450 cytochrome 26B), encoding nucleic acid molecules and methods and uses thereof
US7537919B2 (en) Cytochrome P450RAI-2 and related proteins
AU688282B2 (en) Stably-transformed mammalian cells expressing a regulated, inflammatory cyclooxygenase
JP2000506396A (en) Nucleotide sequence encoding phosphatidylinositol-3&#39;-kinase-related protein and use thereof
US7335484B2 (en) Retinoid metabolizing protein
AU778362B2 (en) Retinoid metabolizing protein
JP2001512986A (en) Tazarotene-induced gene 3 (TIG3)
WO1999016783A1 (en) Potent inhibitors of human 9-cis retinol dehydrogenase
US6063606A (en) Retinoid metabolizing protein
MXPA98010433A (en) Protein that metabolizes retinol
WO2002048334A2 (en) P450rai-2 (p450 cytochrome 26b), encoding nucleic acid molecules and methods and uses thereof
US7335756B2 (en) Retinoic acid metabolizing cytochrome P450
WO1996040720A1 (en) Mammalian prostaglandin h synthase-2
CA2431640A1 (en) P450rai-2 (p450 cytochrome 26b), encoding nucleic acid molecules and methods and uses thereof
CA2351627A1 (en) Methods and compositions for diagnosis and treatment of cancer based on the transcription factor ets2
MXPA98010434A (en) Method to identify citocrome p
WO2004046349A2 (en) Human methionine aminopeptidase type 3
AU2002302253A1 (en) A retinoic acid metabolizing cytochrome P450