AU773649B2 - Methods and compositions for modulating morphogenic protein expression - Google Patents

Methods and compositions for modulating morphogenic protein expression Download PDF

Info

Publication number
AU773649B2
AU773649B2 AU34387/02A AU3438702A AU773649B2 AU 773649 B2 AU773649 B2 AU 773649B2 AU 34387/02 A AU34387/02 A AU 34387/02A AU 3438702 A AU3438702 A AU 3438702A AU 773649 B2 AU773649 B2 AU 773649B2
Authority
AU
Australia
Prior art keywords
expression
reporter gene
nucleic acid
sequence
cell
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
AU34387/02A
Other versions
AU3438702A (en
Inventor
Hermann Oppermann
Engin Ozkaynak
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.)
Stryker Corp
Original Assignee
Curis Inc
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 AU36757/99A external-priority patent/AU743061B2/en
Application filed by Curis Inc filed Critical Curis Inc
Priority to AU34387/02A priority Critical patent/AU773649B2/en
Publication of AU3438702A publication Critical patent/AU3438702A/en
Application granted granted Critical
Publication of AU773649B2 publication Critical patent/AU773649B2/en
Assigned to STRYKER CORPORATION reassignment STRYKER CORPORATION Alteration of Name(s) in Register under S187 Assignors: CURIS, INC.
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Landscapes

  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Description

Regulation 3.2
AUSTRALIA
Patents Act 1990 COMPLETE SPECIFICATION DIVISIONAL APPLICATION
(ORIGINAL)
Name of Applicant: Actual Inventor(s): Address for Service: Invention Title: Curis, Inc.
Engin Ozkaynak; Hermnann Oppermann.
DAVIES COLLISO8N GAVE, Patent Attelmey t Httlee i 1 Adelbounie Vituria 300 2 8adiJL.t~ 31veido6 062L#CC. 4oe 21, 60 HAV-,4~ zoo "Methods and compositions for modulating morphogenic protein expression" The following statement is a full description of this invention, including the best method of performing it known to us: Q:/OPER'JMS\2S23630-107. DOC 17/4/02 -1- Methods and Compositions for Modulating Morphogenic Protein Expression Field of the Invention The invention relates generally to the field of drug screening assays. More particularly, the invention relates to methods and compositions for identifying molecules that modulate production of true tissue morphogenic proteins.
Background of the Invention Any discussion of the prior art throughout the specification should in no way be considered as an admission that such prior art is widely known or forms part of common general knowledge in the field.
A class of proteins recently has been identified, the members of which are true tissue morphogenic proteins. The members of this class or proteins are characterized as component for inducing the developmental cascade of cellular and molecular events that culminate in the formation of new organ-specific tissue, including any vascular or connective tissue formation as required by the naturally occurring tissue. Specifically, the morphogens are competent for inducing all of the following biological functions in a *morphogenically permissive environment: stimulating proliferation of progenitor cells; stimulating differentiation of progenitor cells; stimulating the proliferation of differentiated cells and supporting the growth and maintenance of differentiated cells. For example, the morphogenic proteins can induce the full developmental cascade of bone tissue morphogenesis, including the migration and proliferation of mesenchymal cells, proliferation and differentiation of chondrocytes, cartilage matrix formation and calcification, vascular invasion, osteoblast proliferation, bone formation, bone remodelling, and heatopoietic bone marrow differentiation. These proteins also have remodelling, and hematopoietic bone marrow differentiation. These proteins also have 500322742 1.DOC/BSW -2been shown to induce true tissue morphogensis of non-chondrogenic tissue, including dentin, liver, and nerve tissue.
A particularly useful tissue morphogenic protein is human OP-1 (Osteogenic Protein-1), described in US 5,011,691; US Pat. No. 5,266,683 and Ozkaynak et al.
(1990) EMBO J. 9: 2085-2093. Species homologues identified to date include mouse OP-1 (see US Pat. 5,266,683) and the Drosophila homologue 60A, described in Wharton et al. (1991) PNAS 88: 9214-9218). Other closely related proteins include OP-2 (Ozkaynak (1992) J. Biol. Chem. 267: 25220-25227 and US Pat. No. 5,266,683); BMP6 (Celeste et al. (1991) PNAS 87: 9843-9847) and Vgr-1 (Lyons et al. (1989).
These disclosures are incorporated herein by reference.
It previously has been contemplated that these tissue morphogens can be administered to an animal to regenerate lost or damaged tissue. Alternatively, one can envision administering a molecule capable of modulating expression of the endogenous tissue morphogen as a means for providing morphogen to a site in vivo.
15 Advantageously, the present invention relates to compositions and methods of screening compounds which can modulate expression of an endogenous tissue morphogen, particularly OP-1 and closely related genes. The compounds thus identified have utility both in vitro and in vivo. Useful compounds contemplated include those capable of stimulating transcription and/or translation of the OP-1 gene, as well as 20 compounds capable of inhibiting transcription and/or translation of the OP-1 gene.
These and other advantages and features of the invention will be apparent from the description, drawings and claims which follow.
i Summary of the Invention The invention features compositions and methods for screening candidate compounds for the ability to modulate the effective local or systemic quantity of 500322742 I.DOC/BSW -3endogenous OP-I in an organism, and methods for producing the compounds identified.
In one aspect, the method is practiced by: incubating one or more candidate compounds with cells transfected with a DNA sequence encoding, in operative association with reporter gene, a portion of an OP-I non-coding DNA sequence that is competent to act on and affect expression of the associated receptor gene; measuring the level of reporter gene expression in the transfected cell, and comparing the level of reporter gene expressed in the presence of the candidate compound with the level of reporter gene expressed in the absence of the candidate compound. In a related aspect, the invention features the compound that is identified by use of the method of the invention.
The screening method of the invention provides a simple method of determining a change in the level of a reporter gene product expressed by a cell following exposure to one or more compound(s). The level of an expressedreporter gene product in a given cell culture, or a change in that level resulting from exposure to one or more compound(s) indicates that application of the compound can modulate the level of the morphogen expressed and normally associated with the non-coding sequence.
Specifically, an increase in the level of reporter gene expression is indicative of a candidate compound's ability to increase OP-I expression in vivo. Similarly, a decrease in the level of reporter gene expression is indicative of a candidate compound's ability to decrease or otherwise interfere with OP-1 expression in vivo.
The methods and compositions of the invention can be used to identify compounds showing promise as therapeutics for various in vivo and ex vivo mammalian S: applications, as well as to identify compounds having numerous utilities. For example, morphogen expression inducing compounds can be used in vivo to correct or alleviate a diseased condition, to regenerate lost or damaged tissue, to induce cell 500322742_I.DOC/BSW 3aproliferation and differentiation, and/or to maintain cell and tissue viability and/or a differentiated phenotype in vivo or ex vivo. The compounds also can be used to maintain the viability of, and the differentiated phenotype of, cells in culture. The various in vivo, ex vivo, and in vitro utilities and applications of the morphogenic proteins described herein are well documented in the art. See, for example, US 92/01968 (WO 94/03200), filed March 11, 1992; US 92/07358 (WO 93/04692), filed August 28; PCT US 92/0743 (W093/057541), filed August 28, 1992; US 93/07321 (WO 94/03200), filed July, 29, 1993; US 93/08808 (WO 94/06449), filed September 16, 1993; US 93/08885 (WO 94/06420), filed September 15, 1993, and US Pat. No. 5,266,683.
Morphogen expression inhibiting compounds identified by the methods, kits and compositions described herein can be used to modulate the degree and/or timing of morphogen expression in a cell. Such components can be used both in vitro and in vivo to more closely regulate the production and/or available concentration of morphogen.
Accordingly, a first aspect of the present invention provides use of a compound for 15 modulating OP-1 expression, said compound identified by a method comprising the steps of: incubating a candidate compound with a cell transfected with an isolated nucleic acid comprising a reporter gene in operative association with a single nucleic acid fragment of an OP-1 specific upstream non-coding 20 sequence, wherein said nucleic acid fragment: 6 consists ofnucleotides 3170 to 3317, 3020 to 3317, 2790 to 3317, 2548 to 3317, 2300 to 3317, 1300 to 3317, 2548 to 2790, 1549 to 2790, or 1 to 2790 of SEQ ID NO:1; (ii) comprises not more than one nucleic acid fragment of said OP-1 upstream sequence; and 500322742 I.DOC/BSW -3b (iii) is operative to regulate expression of said reporter gene; measuring the level of said reporter gene expressed in said cell; comparing said level with that of said reporter gene expressed in said cell in the absence of said candidate compound, wherein an increase in reporter gene expression level is indicative of said candidate's ability to increase OP-1 expression in vivo, and a decrease in reporter gene expression level is indicative of the candidate's ability to inhibit OP-1 expression in vivo.
According to a second aspect, the present invention provides use of a compound for modulating OP-1 expression, said compound identified by a method comprising the steps of: incubating a candidate compound with a cell transfected with an isolated nucleic acid, said nucleic acid comprising a reporter gene in operative association with: a first DNA sequence, wherein said first DNA sequence is: 15 a single nucleic acid fragment of an OP-1 specific upstream non-coding sequence, wherein said nucleic acid fragment consists ofnucleotides 2548 to 3317 or 2548 to *0 2790 of SEQ ID NO: 1; a nucleic acid fragment of an OP-1 specific upstream non- 20 coding sequence, wherein said nucleic acid fragment *0 consists ofnucleotides 1549 to 2297 or 1549 to 1788 of a *000 SEQ ID NO: 2; or a variant of a nucleic acid fragment of which a hybridises with a nucleic acid complementary to the nucleic acid fragment of (ii) under conditions of 500322742_ .DOC/BSW -3c hybridisation in 40% formamide, 5XSSPE, solution, and 0.1% SDS at 37°C, followed by washing in 0.1% SSPE, and 0.1% SDS at 50 0 C; and (ii) a second DNA sequence comprising a sequence which interacts with a DNA binding molecule and affects expression of said reporter gene, wherein; said second DNA sequence optionally comprises at least one Wt-1/Egr-1 consensus element SEQ ID NO: 4; or said second DNA sequence is selected from the group of sequences consisting of a TTC element, a Fushi-tarazu protein binding element and a steroid responsive element; wherein said isolated nucleic acid comprises not more than one nucleic acid fragment of measuring the level of reporter gene expressed in said cell; and 15 comparing said level with that of said reporter gene expressed in said cell in the absence of said candidate compound, wherein an increase in reporter gene expression level is indicative of said candidate's ability to increase OP-1 expression in vivo, and a decrease in reporter gene expression level is indicative of the candidate's ability to inhibit OP-1 20 expression in vivo.
According to a third aspect, the present invention provides use of a compound for modulating OP-1 expression, said compound identified by a method comprising the S. steps of: o• 500322742_1.DOC/BSW -3d incubating a candidate compound with a cell transfected with an isolated nucleic acid, said nucleic acid comprising a reporter gene in operative association with: a first DNA sequence, wherein said first DNA sequence is: a single nucleic acid fragment of an OP-1 specific upstream non-coding sequence, wherein said nucleic acid fragment consists ofnucleotides 2548 to 3317 or 2548 to 2790 of SEQ ID NO: 1; a nucleic acid fragment of an OP-1 specific upstream noncoding sequence, wherein said nucleic acid fragment consists of nucleotides 1549 to 2297 or 1549 to 1788 of SEQ ID NO: 2; or a variant of a nucleic acid fragment of which hybridises with a nucleic acid complementary to the 15 nucleic acid fragment of (ii) under conditions of o hybridisation in 40% formamide, 5XSSPE, solution, and 0.1% SDS at 37°C, followed by washing in 0.1% SSPE, and 0.1% SDS at 50 0
C;
(ii) a second DNA sequence comprising a sequence which interacts 20 with a DNA binding molecule and affects expression of said reporter gene, wherein; said second DNA sequence optionally comprises at least one Wt-1/Egr-1 consensus element SEQ ID NO: 4; or said second DNA sequence is selected from the group of sequences consisting of a TTC element, a Fushi-tarazu 500322742_I.DOC/BSW -3e protein binding element and a steroid responsive element; and (iii) a third DNA sequence independently selected from the group consisting of a TCC element, a Fushi-tarazu protein binding element, and a steroid responsive element; wherein said isolated nucleic acid comprises not more than one nucleic acid fragment of(l); measuring the level of reporter gene expressed in said cell; and comparing said level with that of said reporter gene expressed in said cell in the absence of said candidate compound, wherein an increase in reporter gene expression level is indicative of said candidate's ability to increase OP-1 expression in vivo, and a decrease in reporter gene expression level is indicative of the candidate's ability to inhibit OP-1 expression in vivo.
According to a fourth aspect, the present invention provides a method for screening 15 a candidate compound for the ability to modulate expression of OP-i, said method comprising the steps of: incubating a candidate compound with an epithelial cell transfected with an isolated nucleic acid comprising part or all of a nucleotide sequence encoding an OP-1 pro protein in operative association with a reporter 20 gene, said reporter gene further in operative association with a single nucleic acid fragment of an OP-1 specific upstream non-coding sequence, wherein said nucleic acid fragment: consists ofnucleotides 3170 to 3317, 3020 to 3317, 2790 to 3317, 2548 to 3317, 2300 to 3317, 1300 to 3317, 2548 to 2790, 1549 to 2790, or 1 to 2790 of SEQ ID NO: 1; 500322742 I.DOC/BSW -3f- (ii) comprises not more than one nucleic acid fragment of said OP-I upstream sequence; and (iii) is operative to regulate expression of said reporter gene; measuring the level of said reporter gene expressed in said cell; comparing said level with that of said reporter gene expressed in said cell in the absence of said candidate compound, wherein an increase in reporter gene expression level is indicative of said candidate's ability to increase OP-I expression in vivo, and a decrease in reporter gene expression level is indicative of the candidate's ability to inhibit OP-I expression in vivo.
According to a fifth aspect, the present invention provides a method for screening a candidate compound for the ability to modulate expression of OP-i, said method comprising the steps of: incubating a candidate compound with a cell transfected with an isolated i 15 nucleic acid comprising part or all of a nucleotide sequence encoding an OP-I pro protein in operative association with a reporter gene, said i reporter gene further in operative association with a single nucleic acid fragment of an OP-l specific upstream non-coding sequence, wherein said nucleic acid fragment: consists ofnucleotides 3170 to 3317, 3020 to 3317, 2790 to 3317, 2548 to 3317, 2300 to 3317, 1300 to 3317, 2548 to 2790, 1549 to 2790, or 1 to 2790 of SEQ ID NO:l; (ii) comprises not more than one nucleic acid fragment of said OP-1 upstream sequence; and (iii) is operative to regulate expression of said reporter gene; 500322742_ .Doc/BSW -3gsaid cell being of urogenital, liver, bone, cardia, lung, or nerve cell origin; measuring the level of said reporter gene expressed in said cell; comparing said level with that of said reporter gene expressed in said cell in the absence of said candidate compound, wherein an increase in reporter gene expression level is indicative of said candidate's ability to increase OP-1 expression in vivo, and a decrease in reporter gene expression level is indicative of the candidate's ability to inhibit OP-1 expression in vivo.
e go.
*••go gig o *°o **o 500322742_ .DOC/BSW P:\OPERUJMS\36757DIV.WPD- 17/4/02 -4- List of Useful Terms and Definitions As used herein, "gene expression" is understood to refer to the production of the protein product encoded by a DNA sequence of interest, including the transcription of the DNA sequence and translation of the mRNA transcript.
As used herein, "operative association" is a fusion of the described DNA sequences with a reporter gene in such a reading frame as to be co-transcribed, or at such a relative positioning as to be competent to modulate expression of the reporter gene.
As used herein, "vector" is understood to mean any nucleic acid comprising a nucleotide sequence of interest and competent to be incorporated into a host cell and recombining with and integrating into the host cell genome. Such vectors include linear nucleic acids, plasmid, phagemids, cosmids, YAC'S (yeast artificial chromosomes) and the like.
As used herein, "non-coding sequence" or "non-coding DNA" includes DNA sequences that are not transcribed into RNA sequence, and/or RNA sequences that are not translated into protein. This category of "non-coding sequence" has been defined for ease of reference in the application, and includes sequences occurring to the ATG site which indicates the start codon and sequences 3' to the stop codon, as well as intervening intron sequences that occur within the coding region of the gene.
As used herein, an "OP -specific" non-coding sequence is understood to define a noncoding sequence that lies contiguous to OP1 specific coding sequence at an OP-1 gene locus under naturally-occurring conditions. The sequences may include 3' and intron sequence.
As used herein, "allelic, species and other sequence variants thereof' includes point mutations, insertions and deletions such as would be naturally occurring or which can genetically engineered into an OP-1 non-coding DNA sequence and which do not affect substantially the regulation of a reporter gene by the OP-1 non-coding sequence. For example, one or ordinary skill in the art can use site directed P.\OPER\JMS\36757DIV WPD- 17/4/02 mutagenesis to modify, as by deletion, for example, one or more of the OP-1 noncoding sequences described herein without substantially affecting the regulation of OP- 1 or a reporter gene by the modification. Such modifications are considered to be within the scope of the disclosure provided herein.
As used herein, a "Wt-1/Egr-1 consensus binding sequence" or Wt-1/Egr-1 consensus binding element" is a nine base sequence which has been shown to be bound by the DNA binding proteins Wt-1 and Egr-1. The consensus sequence of the Wt-1/Egr-1 binding site has been determined by homology to be GNGNGGGNG, Seq.
ID No. 4 (Rauscher et al., Science 250: 1259-1262 (1990), incorporated herein by reference).
As used herein, a "TCC binding sequence" or "TCC binding element" is an approximately 15 to 20 base sequence of DNA which contains at least three contiguous or non-contiguous repeats of the DNA sequence TCC. The TCC binding sequence identified in human OP-1 genomic DNA is shown in Seq. ID No. 5, and the TCC binding sequence identified in murine OP-1 genomic DNA is shown in Seq. ID No. 6. The TCC binding sequence has also been shown to be bound by the DNA binding proteins Wt-1 and Egr-1 (Wang et al., Proc. Natl. Acad. Sci. 90: 8896-8900 (1993); Wang et al., Biochem Biophys Res. Comm.. 188: 433-439 (1992)).
As used herein, a "FTZ binding sequence" or "FTZ binding element" is a Fushitarazu DNA sequence (FTZ) that has been shown to be bound by the DNA binding protein Fushi-tarazu (FTZ-F1). The FTZ binding sequence identified in human OP-1 genomic DNA is shown in Seq. ID No. 7. The FTZ consensus sequence, a consensus sequence for the nuclear hormone receptor superfamily, is YCAAGGYCR.
As used herein, a "steroid binding sequence" or "steroid binding element" is a DNA sequence that has been shown to be bound by one or more elements, in response to activating signal molecules. Examples of such "activitating signal molecules" include retinoids, Vitamin D, and also include steroids such as estrogen and progesterone. Useful elements are anticipated to include the FTZ-F1 protein, WT- P:\OPERUMS\36757DIV.WPD- 17/4/02 -6- 1 and Egr-1. Activating signal molecules of the nuclear receptor family have recently been shown to bind to DNA as homodimers, heterodimers or as monomers (Parker, Curr. Op. Cell Biol., 1993, 5: 499-504). The formation of heterodimers among the nuclear receptor family molecules may significantly increase the diversity of binding elements which are recognized by these nuclear receptors, and provide for differential regulation of genes containing the specific binding sites. In addition, the nuclear receptors have been shown to interact with other accessory factors, such as transcription factors, to stimulate or repress transcription. These interactions, between the nuclear receptors and the nuclear receptors and accessory factors, indicate that there could be significant number of nuclear receptor/accessory factor interactions which have widely different transcriptional activities.
While the method of the invention is described with reference to a single cell, as will be appreciated by those having ordinary skill in the art, this is only for ease of description, and the method is most efficiently carried out using a plurality of cells.
With respect to transfection of DNA sequences in the cell and the method of the invention, all means for introducing nucleic acids into a cell are contemplated including, without limitation, CaPO 4 co-precipitation, electroporation, DEAE-dextran mediated uptake, protoplast fusion, microinjection and lipofusion. A key to the invention is the DNA sequences with which the cell is transfected, rather than the mechanical or chemical process by which the DNA incorporation is accomplished.
Useful reporter genes are characterized as being easy to transfect into a suitable host cell, easy to detect using an established assay protocol, and genes whose expression can be tightly regulated. Other reporter genes contemplated to have utility include, without limitation, the luciferase gene, the Green Fluorescent Protein (GFP) gene, the chloramphenicol Acetyl Transferase gene (CAT), human growth hormone, and beta-galactosidase. Additional useful reporter genes are any well characterized genes the expression of which is readily assayed, and examples of such reporter genes can be found in, for example, F.A. Ausubel et al., Eds., Current Protocols in Molecular Biology, John Wiley Sons, New York, (1989). As will be P:\OPER\JMS\36757DIV.WPD- 17/4/02 -7appreciated by those having ordinary skill in the art, the listed reporter genes are only a few of the possible reporter genes, and it is only for ease of description that all available reporter genes are not listed.
While the method, vectors, and cells described recite the use of a reporter gene in operative association with an OP-1 non-coding DNA sequence, it will be apparent to those of ordinary skill in the art that the DNA sequence OP-1, including human OP1, shown in Seq. ID No. 1 or murine OP-1, disclosed in U.S. Patent No. 5,266,683, is also within the scope of a suitable reporter gene. Other suitable reporter genes can be used for ease in assaying for the presence of the reporter mRNA or reporter gene product.
Where a cell line is to be established, particularly where the transfected DNA is to be incorporated into the cell's genome, lines that can be immortalized are especially desirable. As used herein, "immortalized" cell lines are viable for multiple passages greater than 50 generations) without significant reduction in growth rate or protein production.
While the selected non-coding DNA sequences disclosed herein are described using defined bases, as will be appreciated by those having ordinary skill in the art, to some degree the lengths of the selected DNA sequences recited are arbitrary and are defined for convenience. As will be understood by those of ordinary skill in the art, shorter sequences of OP-1 non-coding DNA sequence and other fusion DNA's can be used in a vector according to the invention, and can be transfected into a cell, or used in the method of the invention for screening a candidate compound for its ability to modulate OP-1 expression. Specifically, it is standard procedure for molecular biologists to first identify useful regulatory sequences, and then to determine the minimum sequence required, by systematic digestion and mutagenesis by exonuclease or endonuclease digestion, site directed mutagenesis and the like.
Accordingly, subsequent, standard routine experimentation is anticipated to identify minimum sequences and these, shorter sequences are contemplated by the invention disclosed herein.
P:\OPER\MS\36757DIV.WPD- 17/4/02 -8- Useful call types for the method and compositions according to the invention include any eukaryotic cell. Currently preferred are cell types known to express OP-1.
Such cells include epithelial cells and cells or uro-genital cell origin, including renal (kidney or bladder) cells, as well as liver, bone, nerve, ovary, cardiac muscle and the like. The cells may be derived from tissue or cultured from an established cell line.
See, for example Ozkaynak et al. (1991) Biochem. BioPhys. Res. Comm. 179: 116- 123 for a detailed description of tissues known to express OP-1. Other useful cells include those known to exhibit a steroid receptor, including cells having an estrogen receptor and cells responsive to the FTZ-F1 protein. Currently preferred cells also have simple media component requirements. Other useful representative cells include, but are not limited to, Chinese hamster ovary (CHO), canine kidney (MDCK); or rat bladder (NBT-2) and the like. Useful cell types can be obtained from the American Type Culture Collection (ATCC), Rockville, MD or from the European Collection of Animal Cell Cultures, Porton Down, Salisbury SP4 OJG, U.K. As used herein, "derived" means the cells are from the cultured tissue itself, or are a cell line whose parent cells are of the tissue itself.
Aspects and Embodiments of the Invention In one aspect, the invention features a vector having a reporter gene operatively associated with a portion of one or more OP-1 non-coding sequences. The OP-1 noncoding sequence chosen is independently selected from the 5' (or "upstream") noncoding human or murine OP-1 sequence shown in Seq. ID Nos. 1 and 2, respectively, the 3' (or "downstream") non-coding human or murine OP-1 sequence shown in Seq.
ID Nos. 1 or 3, and the human intron non-coding OP-1 sequences shown in Seq. ID No. 1. Also anticipated to be useful are the non-coding sequences 3' and intron) of other species homologs of OP-1 and proteins closely related to OP-1. In addition, the portion of OP-1 sequence included in the vector can be a combination of two or more 5' non-coding, 3' non-coding and/or intron OP-1 sequences.
In one embodiment, the vector can include a non-coding OP1-specific sequence selected from at least one of the following sequence segments of Seq. ID No. 1 presented below, and which define human genomic OP-1 sequence comprising P:NOPER\JMS\36757DIV.WPD- 17/4/02 -9approximately 3.3 Kb of 5' non-coding sequence. In Seq. ID No. 1, the start codon begins at position 3318, and the upstream sequence (bases 1 to 3317) is composed of untranscribed (1 to 2790) and untranslated (2791 to 3317) OP1-specific DNA; approximately 1 Kb of which is presented in Fig. 1 (bottom strand).
Useful sequence segments include bases 2548-3317, representing 750 bases sharing significant (greater than 70% identity) between the mouse and human OP-1 homologs (See Fig. and bases 3170-3317; 3020-3317, 2790-3317, 2548-2790 of Seq. ID No. 1, all shorter fragments of this region of the DNA. As base 2790 is the mRNA start site, other useful sequences include 2790-3317, representing transcribed but not translated 5' coding sequence and shorter fragments of this DNA region as noted above; upstream fragments of OP1-specific DNA, bases 2548-2790; 1549-2790; 1-2790 of Seq. ID No. 1. Also useful sequence segments include the approximately 750 bases that have homology between the human and mouse OP-1 sequences with additional upstream sequences 2300 to 3317; 1300 to 3317; 1-3317; all fragments of the disclosed upstream OP1-specific DNA sequences of Seq. ID No. 1.
In another embodiment, the sequences are defined by the non-coding sequences of the mouse OP-1 homolog, including the following 5' non-coding sequences (Seq. ID No. 2150-2296, 2000-2296, 1788-2296, and 1549-2296 all of which define the 750 bases sharing high sequence identity with the human homolog (See, Fig. 800-2296; 1-2296; 1549-1788, 800-1788 and 1-1788.
Within this region also exist a number of Egr/Wt-1 sites (8 in hOP-1; 7 in mOPknown in the art to bind the regulatory elements Egr and Wt-1. Accordingly, in another aspect, the invention contemplates a screening material for identifying compounds which modulate OP-1 expression, the assay comprising the step of identifying compounds which bind Egr/Wt-1 site. At least one Wt/Egr-1 element, preferably between 1-6 elements, or at least 6 Wt/Egr-1 elements are included in a sequence. The relative locations of these elements are indicated in Fig. 1 and at positions 3192-3200; 3143-3151; 3027-3035; 2956-2964; 2732-2740; 2697-2704 of Seq. ID No. 1, and positions 2003-2011; 1913-1922; 1818-1826; 1765-1776; 1757- P:\OPER\JMS\36757D[V WPD. 17/4102 1765; 1731-1739; 1699-1707; 1417-1425 of Seq. ID No. 2 of Seq. ID Nos. 1, 2 substantially the same Seq. alignment. The lengths of bases within these 5' noncoding sequences is selected to include portions of the sequence of DNA which was determined to be homologous between murine and human genomic OP-1, separately and as a part of a larger sequence including non-homologous DNA. Additionally, the portion of OP-1 sequence selected can be a portion of the region of homology between murine and human OP-1 DNA sequences, bases 2548-2790 or 2548-3317 of Seq. ID No. 1, or bases 1549 to 1788 or 1549 to 2296 of Seq. ID No. 2, and/or at least one of an Wt-1/Egr-1 consensus binding sequence. In still another aspect the portion of OP-1 sequence selected can include a TCC binding sequence, a FTZ binding sequence, a steroid binding sequence, or part or all of an OP-1 intron sequence. The relative positions of the TCC and FTZ elements are indicated in Fig. 1 and at positions 2758- 2778 (TCC); 2432-2441 (FTZ) of Seq. ID No. 1 and 1755-1769 (TCC) of Seq. ID No.
2.
In another aspect, the invention features a cell that has been transfected with a reporter gene in operative association with a portion of OP-1 non-coding DNA sequence. The portion of OP-1 non-coding sequence is independently selected from the 5' (or upstream) non-coding human or murine OP-1 sequence shown in Seq. ID Nos. 1 and 2, the 3' (or downstream) non-coding murine OP-1 sequence shown in Seq. ID No. 3, and the human intron non-coding OP-1 sequence shown in Seq. ID No.
1. The six human intron non-coding OP-1 sequences are at bases 3736 to 10700; bases 10897 to 11063; bases 11217 to 11424; bases 11623 to 13358; bases 13440 to 10548; bases 15166 to 17250; all of Seq. ID No. 1. In addition to portion of OP-1 sequence selected can be a combination of 5' non-coding, 3' non-coding and/or intron OP-1 sequence. Thus, the cell can have been transfected with a reporter gene in operative association with a portion of 5' non-coding OP-1 genomic sequence that is independently selected from bases 3170 to 3317; 3020 to 3317; 2790 to 3317; 2548 to 3317; 2300 to 3317; 1300 to 3317; 1 to 3317; 2548 to 2790; 1549 to 2790; and 1 to 2790; all of Seq. ID No. 1 or bases 2150 to 2296; 2000 to 2296; 1788 to 2296; 1549 to 2296; 800 to 2296; 1 to 2296; 1549 to 1788; 800 to 1788; 1 to 1788; all of Seq. ID No. 2. The lengths of bases within these 5' non-coding sequences is selected to P:\OPERUMS\36757DIV WPD- 17/4/02 11 include portions of the sequence of DNA which was determined to be homologous between murine and human genomic OP-1, separately and as a part of a larger sequence including non-homologous DNA. Additionally, the portion of OP-1 sequence selected can be a portion of the region of homology between murine and human OP-1 DNA sequences, such as bases 2548-2790 to 2548-3317 of Seq. ID No. 1, or bases 1549 to 1788 or 1549 to 2296 of Seq. ID No. 2, and at least one of an Wt-1/Egr-1 consensus binding sequence, a TCC binding sequence, a FTZ binding sequence, a steroid binding sequence, and an intron. Thus the portion of OP-1 sequence selected can be a portion of the 5' non-coding human or murine OP-1 genomic DNA sequences, as stated above, and at least one Wt-1/Egr-1 consensus binding sequence alone or in combination with at least one of a TCC binding sequence, a FTZ binding sequence, a steroid binding sequence, and a human OP-1 intron DNA sequence. In another embodiment more than one Wt-1/Egr-1 element is used, for example, between 1-6, or at least six. These cells are suitable for use in the method of the invention.
In one embodiment, part of the OP-1 coding region is anticipated to have an expression regulatory function and also can be added to a vector for use in the screening assay described herein. OP-1 protein is translated as a precursor polypeptide having an N-terminal signal peptide sequence (the "pre pro" region) which is typically less than about 30 amino acid residues, followed by a "pro" region which is about 260 amino acid residues, followed by the additional amino acid residues which comprise the mature protein. The pre pro regions are cleaved from the primary translation sequence to yield the mature protein sequence. The mature sequence comprises both a conserved C-terminal seven cysteine domain and an N-terminal sequence which varies significantly in sequence between the various morphogens.
The mature polypeptide chains dimerize and these dimers typically are stabilized by at least one interchain disulfide bond linking the two polypeptide chain subunits. After the pro domain is cleaved from the OP-1 protein it associates noncovalently with the mature dimeric protein, presumably to enhance solubility and/or targeting properties of the mature species. See, for example, PCT/US93/07189, filed July 29, 1993. The pro region represents the nucleotide sequence occurring approximately 87 bases downstream of the ATG start codon, and continues for about 980 bases. The P:\OPERMS\36757DIVWPD -17/4/02 -12nucleotide sequence encoding the pro region is highly enriched in a "GC" sequence, which well may be competent to form a secondary structure as part of the mRNA transcript) which itself may modulate OP-1 expression. Accordingly, part or all of the nucleotide sequence encoding an OP-1 pro region, particularly that portion corresponding to a GC rich region, may be used, preferably in combination with one or more OP-1 non coding sequences, in the compositions and methods of the invention.
In another embodiment, the method can be practiced using a cell known to express the OP-1 gene. Suitable DNA sequences for transfection are described below, as well as suitable cells containing transfected DNA sequences.
In another aspect, the invention provides molecules, vectors, methods and kits useful in the design and/or identification of OP-1 expression modulating compounds.
As used herein a "kit" comprises a cell transfected with a DNA sequence comprising a reporter gene in operative association with a portion of OP-1 upstream DNA sequence and the reagents necessary for detecting expression of the reporter gene.
The portion of the OP-1 upstream DNA chosen can be any of the various portions which have been described herein.
Following this disclosure, medium flux screen assays, and kits therefore, for identifying OP-1 expression modulating compounds are available. These compounds can be naturally occurring molecules, or they can be designed and biosynthetically created using a rational drug design and an established structure/function analysis methodology. The compounds can be amino acid-based or can be composed in part or whole of non-proteinaceous synthetic organic molecules.
The OP-i expression modulating compounds thus identified then can be produced in reasonable quantities using standard recombinant expression or chemical synthesis technology well know and characterized in the art and/or as described herein. For example, automated means for the chemical synthesis of nucleic and amino acid sequences are commercially available. Alternatively, promising candidates P:\OPER\JMS\36757DIV.WPD- 17/4/02 13 can be modified using standard biological or chemical methodologies to, for example, enhance the binding affinity of the compound for a DNA element and the preferred candidate derivative then can be produced in quantity.
Once a candidate compound has been identified it can be tested for its effect on OP-1 expression. For example, a compound which upregulates (increases) the production of OP-i in a kidney cell line is a candidate for systemic administration. The candidate can be assayed in an animal model to determine the candidate molecule's efficacy in vivo. For example, the ability of a compound to upregulate levels of circulating OP-1 in vivo can be used to correct bone metabolism diseases such as osteoporosis (See, for example, PCT/US92/07932, supra). Useful in vivo animal models for systemic administration are disclosed in the art below.
As demonstrated herein below, OP-1 is differentially expressed in different cell types. Accordingly, it further is anticipated that a candidate compound will have utility as an inducer of OP-1 expression in one cell type but not in another. Thus, the invention further contemplates testing a candidate compound for its utility in modulating expression of OP-1 in different cells in vivo, including different cells known to express OP-1 under native physiological conditions.
Thus, in view of this disclosure, one of ordinary skill in recombinant DNA techniques can design and construct appropriate DNA vectors and transfect cells with appropriate DNA sequences for use in the method according to the invention to assay for compounds which modulate the expression of OP-i. These identified compounds can be used to modulate OP-1 production and its available concentrations in both in vivo and in vitro contexts.
Brief Description of the Drawings Fig. 1 shows the alignment of upstream sequences of the murine and human OP-1 gene. The murine sequence is present in the upper sequence lines and the human sequence is the lower sequence on all lines. The murine sequence is numbered backwards, counting back from the first ATG of the translated sequence P'\OPER\JMS\36757DIV WPD- 17/4/02 14which is shown highlighted. For purposes of alignment, dashes are introduced into the DNA sequence, and three portions of human DNA sequence have been cut from the sequence and placed underneath a gap, below a solid triangle; Fig. 2 shows a time course of murine uterus OP-1 mRNA regulation by estrogen; and Fig. 3a shows a schematic of the 2 kb and 4 kb OP-1 mRNAs, the hybridization locations of probes 1 through 7 (indicated by the bars under the schematic). The solid line indicates OP-1 mRNA, the indicate potential poly A signals, the boxes indicate the translated portion of OP-1 mRNA with the hatched box showing the TGF-13-like domain. The dashed lines indicate genomic DNA sequences. The arrows mark the locations of the cleavage site for OP-1 maturation.
Fig. 3b shows a Northern blot hybridization analysis of OP-1 specific 2 kb and 4 kb mRNAs in murine uterine tissue. Lanes 1 through 7 correspond to probes 1 through 7 respectively. The 2 kb and 4 kb mRNAs are indicated by the 4- and 2-on the left side of Fig. 3b, and a 0.24 to 9.49 kb RNA size ladder is indicated by dashes to the right of the figure.
Detailed Description As will be more fully described below, we have identified regions of the OP1 genetic sequence useful in identifying molecules capable of modulating OP-1 expression in vivo. Also as described herein, we have determined that OP-1 expression in vivo can be dependent both on cell type and on the status of the cell in a tissue. Specifically, as described herein below, OP-1 protein expression is differentially regulated in uterine tissue depending on the status of the uterine tissue.
For example, OP-1 expression is dramatically down-regulated in uterine mouse tissue during pregnancy, whereas it is normally expressed in this tissue in virgin mice.
Moreover, OP-1 expression in other tissues such as renal tissue apparently is unaffected during pregnancy. Administration of estrogen to a virgin mouse is capable of duplicating this down-regulation of OP-1 gene expression.
P:\OPER\JMSU6757DIV.WPD- 17/4/02 We investigated the DNA sequences responsible for the regulation of OP-1 gene expression by cloning non-coding sequences for the human and mouse OP-1 gene. The tissue specific modulation of OP-1 gene expression and the significant homology which was found between an approximately 750 base region of human and murine 5' non-coding OP-1 genomic sequence, implicate these sequences as having utility in a method for the screening of compounds for their ability to modulate OP-1 expression.
In view of this disclosure and the examples provided below, a method for identifying molecules which can affect OP-1 expression in a particular cell type in vivo now is provided.
Cloning of Human and Mouse OP-1 Gene Non-coding Sequences In the Northern blot analysis of murine organs multiple OP-1 transcripts, are detected namely, three species of 1.8, 2.2, 2.4 kb and a prominent 4.0 kb RNA species (Ozkaynak et al., 1992, J. Biol. Chem., 267: 25220-25227; Ozkaynak et al; Biochem.
Biophys. Res. Comm., 179: 116-123). The pattern is similar in rats with only the 1.8 kb species absent. The estrogen-mediated downregulation of OP-1 mRNA affects all of these species. In order to prove that the 4.0 kb mRNA is in fact a transcript from the same OP-1 locus, cDNA clones were isolated from a mouse teratocarcinoma cDNA library.
Four independent dines were obtained that added sequence information to the published mouse cDNA sequence. Two of these cDNA clones have longer untranslated sequences (0.4 and 0.3 kb) than previously reported (0.1 kb). Three of the murine clones contain additional 1.4 kb at the 3'-end. The combined sequences add up to a total OP-1 cDNA size of 3.5 kb, about 0.5 kb shorter than the 4.0 kb mRNA observed on Northern blots. cDNA clones that represent the 2 kb and 4 kb messages are shown schematically in Figure 3a. Since the polyA-tail is lacking in those cDNA clones that extend the 3'-information, it was anticipated that missing 0.5 kb sequence occurs at the 3'-end.
P:\OPER\JMS\36757DIV.WPD- 17/4/02 16- In order to obtain the sequence immediately adjacent to the 3'-end of the kb cDNA sequence, a mouse genomic library, ML1039J (Clontech), was screened with a 3'-end cDNA specific probe (0.45 kb, 3'-end Xmnl-EcoRI fragment of murine DP-1 cDNA) according to the parameters described below for the cloning of upstream noncoding sequences. This screen yielded four lambda clones which were analyzed by Southern blotting. All clones yielded a 1.5 kb Xmnl fragment which was subcloned from lambda 071 into a Bluescript vector and sequenced. Three polyadenylation signals (AATAAA) (Proudfoot et al, (1976) Nature, 263: 211-214) were found in this genomic fragment, at 3.52-, 3.58-, and 3.59 kb (shown schematically in Fig. 3a by the The 3'-end cDNA and the genomic DNA sequences in the 1.5 kb Xmnl fragment overlap by 0.4 kb in a region that immediately precedes the second polyadenylation signal located at 3.5 kb (Figure 3a, region indicated by probe 6) and are in complete agreement within this stretch.
Human upstream non-coding sequence and additional mouse upstream noncoding sequence were obtained by screening human and mouse genomic libraries, HL1067J and ML1030J respectively (Clontech). All libraries were screened by an initial plating of 750,000 plaques (approximately 50,000 plaques/plate). Hybridizations were done in 40% formamide, 5 x SSPE, 5 x Denhardt's solution, and 0.1% SDS at 37°C. Nonspecific counts were removed in 0.1 x SSPE, 0.1 SDS by shaking at Human and mouse upstream genomic DNA sequences were obtained from clones lambda 03 and lambda 033, respectively (Clontech, HL1067J and ML1030J).
These lambda clones were isolated using a 32P-labeled probe made from a human 0.47 kb EcoRI OP-1 cDNA fragment (obtained from p1 15) containing mainly 5' noncoding and exon 1 sequences.
A 7 kb EcoRI fragment from the human genomic clone, lambda 03, was isolated which contains 5 kb of upstream non-coding sequence. Additional upstream sequence information for murine was obtained by subcloning a 1.1 kb Pstl fragment from the genomic phage clone lambda 033. This fragment overlaps with the of the longest murine cDNA clone by 0.3 kb in the 5' non-coding region and provided 0.8 kb additional sequence information. A schematic diagram of the 2- and 4 kb OP-1 P:\OPERUMS\36757DIV.WPD- 17/4/02 -17messages is shown in Figure 3a with dashed lines indicating supplementing information derived from murine upstream and downstream genomic DNA.
All sequencing was done according to Sanger et al. (1977) Proc. Natl. Acad.
Sci. 74: 5463-5467, using exonuclease Ill-mediated unidirectional deletion (Ozkaynak et al., (1987) BioTechniques, 5: 770-773), subcloning of restriction fragments, and synthetic primers. Compressions were resolved by performing the reactions at with Taq polymerase and using 7-deaza-GTP (US Biochemical Corp;, Cleveland, OH).
Verification of OP-1 mRNA Sequences by Northern Blotting To verify the structures of the short and long mRNA species observed, Northern blot hybridizations were performed with probes made from seven non-overlapping DNA fragments (Fig. 3a; probes 1 through 7) specific to the 5' and 3' non-coding region, the protein coding sequence, and genomic regions upstream or downstream of the predicted mRNAs, respectively.
Hybridization of these probes to individual Northern blot strips containing mouse kidney mRNA is consistent with the predicted 4 kb mRNA structure. As shown in Fig.
3a, and Fig. 3b, the genomic DNA probes 1 and 2 did not hybridize to any message.
Probe 2 is specific to the upstream sequences immediately adjacent to the cDNA.
Probes 3, 4 and 5, specific to 5' non-coding, coding, and 3' non-coding regions, respectively, hybridized to both the 2 kb and 4 kb messages, hence these sequences are present in both messages. Probe 6, specific to sequences between the first and second polyadenylation signals, hybridized only to the 4 kb message. Finally, probe 7 which is specific to sequences further downstream of the fourth (last) polyadenylation signal, did not hybridize to any message. The results obtained with these probes confirm the two OP-1 mRNA structures and the approximate and 3'-end boundaries of OP-1 transcripts shown in Figure 3a. This demonstrates that the 2 kb and 4 kb mRNA's are from the same OP-1 genomic locus rather than from multiple genes.
The extensive 3' sequence included in the 4 kb mRNA transcript suggests that P:\OPERUJMS\36757DIV.WPD- 17/4/02 -18the 3' untranslated sequence may play a role in OP-1 gene expression particularly as it has been detected across species namely, in mouse, rat, dog, human and chicken.
Multiple stop codons in all three possible translation reading frames rule out the likelihood that this sequence encodes a peptide. The untranslated sequence itself may act therefore to influence mRNA stability. For example, the sequence may interact with another protein as has been described for transferrin receptor mRNA. Here, IREbinding protein (IRE; iron response element) stabilizes the transferrin receptor mRNA by binding to the 3'-end of the mRNA (Standard et al., 1990, Gene Dev., 4: 2157-2168, incorporated herein by reference). Alternatively, the 3'-end sequences may be interacting with the 5'-end sequences thereby affecting initiation of protein synthesis or, the 3'-end sequences may be serving as a binding site for other RNAs which can interfere with the binding of an expression in modulating molecule, including repressor molecule. (Klausner et al., 1989, Science, 246: 870-872; Kazak, 1992, Ann. Rev. Cell Biol., 8: 197-225, incorporated herein by reference).
Comparison of 5' Non-coding Sequences of Human and Mouse OP-1 DNA The cloning of the 5' non-coding genomic murine and human OP-1 DNA sequences demonstrated that a high degree of sequence homology exists between the human and murine 5' non-coding DNA sequences. The homology extends from the base immediately upstream of the translation start site for the OP-1 morphogen protein to approximately 750 bases upstream of the translation start site, as is shown in the shaded regions of Fig. 1, with the murine sequences being the upper lines and the human sequences being the lower lines. The 5' nucleotide of the region of homology for the human OP-1 5' non-coding sequence is base 2548 of Seq. ID No. 1 and for the murine OP-1 5' non-coding sequence is base 1549 of Seq. ID No. 2. The significant homology between the human and murine 5' non-coding sequences of OP-1 suggest that this region may be important in the regulation of OP-1 expression. As will be discussed in more detail below, this region contains several conserved DNA sequences which have been identified as the DNA binding sequences for two DNA binding proteins, Wt-1 and Egr-1, which both recognize these DNA sequences. The DNA binding sequences for Wt-1/Egr-1 present in human and murine marked in Fig.
P:\OPERJMS\3b757DIV.WPD- 17/4/02 -19- 1 with a single line. Also, the TCC binding sequence, a DNA binding sequence for Wt- 1 and Egr-1, is marked in Fig. 1 by the double line. WT-1 and Egr-1 proteins have also been implicated in the regulation of expression of several genes which are unrelated to OP-1.
Alignments of mouse and OP-1 human genomic sequence reveals a conserved stretch of 0.75 kb just upstream of the first ATG that contains several patterns with marked similarity to the zinc-finger protein binding sequence (5'-GCG GGG GCG-3') specific for Egr-1 and Wt-1 (Christy et al., 1989, PNAS, 86: 8737-8741; Rauscher et al., 1990, Science, 250: 1259-1262; Drummond et al., 1992, Science, 257: 664-678).
In mouse, a total of 8 and in human 7, patterns, conforming to the degenerate Egr- 1/Wt-1 binding sequence (5'-GNG NGG GNG-3') (Rupprecht et al., 1994, J. Biol.
Chem., 269: 6198-6202; Werner et al., 1994, J. Biol. Chem., 269: 12940-12946 are located before and after the presumed transcriptional initiation site (Fig. 1, shown by solid single lines). The presence of these has significance in light of the elevated levels of Wt-1 mRNA in the rat uterus decidua during pregnancy (Zhou et al., 1993, Differentiation, 54: 109-114).
The analysis also revealed, in the human upstream region, a pattern of seven TCC repeats, present at -561, immediately 3' of two Egr/Wt-1 sequences (at -624 and -587) (Figure 1, shown by double solid lines and at position 2758-2778 of Seq. ID No.
The mouse upstream region contains a similar, albeit less obvious sequence at 356 and at position 1755-1769 of Seq. ID No. 2. This TCC-repeat pattern is found in the promoters of PDGF-A and several other growth-related genes, and Wt-1 has been found to activate transcription when either of the sequences are present and to suppress it when both sequences are present. (Wang et al., 1992, Biochem. Biophys Res. Comm., 188: 433-439; Wang et al., 1993, PNAS, 90: 8896-8900 incorporated herein by reference). Accordingly, estrogen receptor may exert its effect on OP-1 expression in uterus by upregulating Wt-1, either directly or indirectly. Alternatively or, in addition other regulatory elements, located further upstream of the OP-1 gene may be involved in estrogen regulation.
P:\OPEJMS\36757D1V.WPD- 17/4/02 Also on Fig. 1, the human 5' non-coding DNA sequence is shown to contain a Fushi-tarazu (FTZ) binding sequence which is marked by carats below the human DNA sequence. A FTZ binding sequence is bound by the Fushi-tarazu protein (FTZ-F1), which is a member of the superfamily of nuclear receptors (Parker, (1993) Current opinion in Cell Biology, 5: 499-504, The superfamily of nuclear receptor proteins include steroid hormones, retinoids, thyroid hormone, nerve growth factor and Fushitarazu, and are structurally related. FTZ-F1 is likely to belong to a subfamily of nuclear receptors that bind DNA as monomers.
The FTZ-F1 protein is a positive regulator at the fushi-tarazu gene in blastoderm stage embryos of Drosophila. FTZ-F1 is closely related in the silkworm (Bombyx) BmFTZ-F1 protein and the mouse embryonal long terminal repeat binding protein (ELP) and all of them are members of the nuclear hormone receptor superfamily, which recognizes the same 9 base pair sequence, 5'-PyCAAGGPyCPu-3'. The FTZ binding sequence does not apparently have a direct or inverted repeat. In contrast, other members of the nuclear hormone receptor superfamily usually bind to repeated sequences. Nevertheless, the FTZ-F1, BmFTZ-F1 and ELP proteins have high affinities for the FTZ binding site DNA, indicating that the mechanism that the binding is somewhat different from that of other members of the nuclear hormone receptor superfamily. (Hitachi et al., 1992, Mol. and Cell Biology December, pp. 5667-5672.).
The mRNA transcription initiation site for human OP-1 is marked on Fig. 1 by the upward arrow, and the OP-1 protein translation initiation site is marked on Fig. 1 by the solid triangles just prior to the highlighted ATG. The transcription initiation site for the human OP-1 gene is at base 2790 of Seq. ID No. 1 and the analogues site for murine is at base 1788 of Seq. ID No. 2. The translation initiation site for the human OP-1 gene is at base 3318 of Seq. ID No. 1 and for murine it is at base 2296 of Seq.
ID No. 2. The high degree of identity that the murine and human DNA sequences share in the region between the transcription initiation site and the translation initiation site, suggests that this region likely plays a role in the modulation of the expression of the OP-1 gene product.
P:\OPER\JMS\36757DIV.WPD- 17/4/02 -21 Analysis of OP-1 Gene Expression in Mouse Tissues A detailed analysis of the uro-genital tract of rats has revealed OP-1 mRNA expression in the renal (kidney), and bladder tissues, as well as at other sites of the urogenital organ system. The most abundant levels are present in renal and uterine tissue (8 week old mice), while much lower levels were found in ovaries. The mRNA level of G3DPH, a "housekeeping function" molecule, was used as an internal control for recovery and quality of mRNA preparations and equal amounts of poly(A)+ RNA were loaded into each lane.
Preparation of RNA and Northern blot hybridization analysis was conducted as follows. 8-week-old female mice, strain CD-1, were obtained from Charles River Laboratories, Wilmington, MA. Total RNA, from the various organs of mice was prepared using the acid-guanidine thiocyanate-phenol-chloroform method (Chomczynski et al., (1987) Anal. Biochem. 162: 156-159). The RNA was dissolved in TES buffer (10 mM Tris-HCI, 1 mM Na 2 -EDTA, 0.1% SDS, pH7.5) containing Proteinase K (Stratagene, La Jolla, CA; approx. 1 mg proteinase/ml TES) and incubated at 37 0 C for 1 hr. Poly RNA was selected in a batch procedure on oligo(dT)-cellulose (Stratagene, La Jolla, CA) in 0.5 M NaCI, 10 mM Tris-HCI, 1 mM Na 2 -EDTA, pH 7.4 (1 x binding buffer). For the selection of poly(A)+ RNA, total RNA obtained from 1 g of tissue was mixed with approximately 0.1g of oligo(dT)-cellulose (in 11 ml TES containing 0.5 M NaCI). The tubes containing the RNA and oligo(dT)cellulose were gently shaken for approx. 2 hrs. Thereafter, the oligo(dT)-cellulose was washed twice in 1x binding buffer and once in 0.5x binding buffer (0.25 M NaCI, 10 mM Tris-HCI, 1 mM Na 2 -EDTA, pH 7.4) and poly RNA was eluted with water and precipitated with ethanol.
Poly(A)+ RNA (5 mg per lane) was electrophoresed on 1.2% agaroseformaldehyde gels with 1 mg of 400 pg/ml ethidium bromide added to each sample prior to heat denaturation (Rosen et al., (1990) Focus, 12: 23-24). Electrophoresis was performed at 100 Volts with continuous circulation of the 1 x MOPS buffer (Ausubel et al., eds., (1990) Current Protocols in Molecular Biology, John Wiley Sons, New York). Following electrophoresis, the gels were photographed, rinsed briefly in water, P:\OPER\JMS\36757DIV.WPD- 17/4/02 -22and blotted overnight onto Nytran (Schleicher Schuell Inc., Keene, NH) or Duralon- UV (Stratagene) membranes in 10 x SSC. The membranes were dried at 80°C for min. and irradiated with UV light (1 mW 2 /cm 2 for 25 sec.).
The 32 P-labeled probe was made from a murine OP-1 cDNA fragment (0.68 kb BstXI-Bgll frg.) by random hexanucleotide priming (Feinberg et al., (1984) Anal.
Biochem., 137: 266-267). The hybridizations were done in 40% formamide, 5x SSPE, x Denhardt's, 0.1% SDS, pH 7.5 at 37 0 C overnight. The non-specific counts were washed off by shaking in 0.1x SSPE, 0.1% SDS at 500C. For re-use, filters were stripped in 1 mM Tris-HCI, 1 mM Na 2 -EDTA, 0.1% SDS, pH 7.5 at 80°C for 10 min.
Analysis of OP-1 Expression During Pregnancy in Mice An examination of the effect of pregnancy upon OP-1 expression was undertaken by measuring OP-1 mRNA level in kidney, ovary and uterus, before, during and after pregnancy (virgins, 2-day post-coital 4-day pc, 6-day pc, 8-day pc, 13day pc, 17-day pc, 3-day lactating, and retired breeders) by Northern blot hybridization of poly(A)+ RNA. These measurements demonstrated that, while kidneys show no pregnancy-related changes in OP-1 mRNA levels, the uterine level became nearly undetectable by 6-day pc. However, no changes were observed in the ovaries. A dramatic and rapid decline in OP-1 message in uterine tissue between day 3 and 4 of pregnancy is apparent in the comparison with virgin animals.
The levels of OP-1 mRNA in the embryo and maternal levels in uterus of 8 week old mice at day 13 and 16 of the pregnancy were also compared. While the OP-1 expression in the pregnant uterus is dramatically reduced, high levels of OP-1 message were found in the mouse embryo at 13- and 16-days. Thus, at a stage of pregnancy when OP-1 mRNA expression in the maternal uterus is almost undetectable, embryonal OP-1 expression is high. The high embryonal OP-1 expression also is detected consistent with the relatively high level of OP-1 mRNA, found in human placenta. The level of OP-1 mRNA measured in the embryo is in the same range as that measured in adult kidney or virgin uterus tissue. Hence, it is likely that OP-1 plays a critical role int he development of the embryo which may require P:\OPER\JMS\36757DV.WPD- 17/4/02 -23appropriate amounts of OP-1 at very specific stages of tissue and organ morphogensis. While not being limited to any given theory, it is possible that OP-1 expression in uterine tissue during pregnancy potentially could interfere with the level of OP-1 produced by the developing embryo, and thereby, interfere with proper development of the embryo. Therefore, a shut-down or inhibition of uterine OP-1 expression during pregnancy might be for the benefit of the fetus.
Effect of Estrogen and Progesterone on OP-1 Expression During pregnancy the estrogen and progesterone levels increase many fold and high level are sustained until birth. To determine whether these hormonal changes are responsible for the altered OP-1 transcription in pregnant uterine tissue, non-pregnant female mice were subcutaneously administered 17-estradiol or progesterone or a combination of both.
In the first experiment the rapid increase in estrogen and progesterone levels during pregnancy was simulated. Non-pregnant mice were injected subcutaneously on four consecutive days with increasing doses, starting with 20 mg 171-estradiol, or 100 mg progesterone or the combination of both and doubling the dose on each following day. On the fourth day the animals were sacrificed and mRNA was isolated from uteri and kidneys. A striking negative effect of 17p-estradiol on the uterine OP-1 mRNA expression was observed, but no effect by progesterone was seen. In the kidneys, however, mRNA levels did not change after 17p-estradiol or progsterone treatment.
Another experiment addressed the time course: 17p-estradiol was administered to virgin female mice at a constant dose of 200 mg (50 ml of 4 mg/ml 17p-estradiol per day, subcutaneously in DMSO [dimethyl sulfoxide] 150 ml 150 mM CaCI) (Figure 2).
Following this, their uteri were extracted, poly(A)+ RNA was prepared, equal amounts of poly(A)+ RNA (5 mg) was loaded into each lane of a 1.2% agarose-formaldehyde gel and analyzed by Northern blot hybridization. The effect was rapid, with considerable decrease of OP-1 mRNA 12 hours after administration of 17p-estradiol and almost undetectable levels by 48 hours, as shown in Fig. 2. In the figure, the P:\OPER\JMS\36757DIV WPD- 17/4/02 -24lanes correspond as follows: from left to right, 0-day (negative control), 0-day (negative control), and 8-days. The arrowheads mark the two major OP-1 mRNA species. A modest amount of message reappears a few days later (Figure 2).
The uterus has been identified as a major site of OP-1 expression. The level of OP-1 expression in uterine tissue is comparable to that observed in renal tissue.
However, during pregnancy, by day four, the uterine OP-1 mRNA levels are reduced to the limit of detection. The loss of OP-1 expression corresponds with also is rising levels of estrogen during this same time frame. The same dramatic loss of uterine OP- 1 message also is observed in estrogen-treated animals, suggesting that estrogen is involved in negative regulation of OP-1 expression in uterine tissue. The effect of estrogen is rapid, with most of the message disappearing after 12 hours of 173estradiol administration. The reappearance of some OP-1 message at later days may be due to a counter-regulatory mechanism. In contrast to the modulated OP-1 mRNA levels in the uterus, no substantial changes occur in renal tissue during pregnancy or in response to estrogen treatment. Therefore, OP-1 mRNA expression in these different organs is regulated independently. The differential expression may be due, for example, to a lack of estrogen receptors in renal tissue. Alternatively, co-regulation by means of none or more accessory molecules that interact with estrogen or a related nuclear receptor molecule(s) may allow for the independent regulation. For example, each of Wt-1 protein (which binds to the Wt-l/Egr-1 element) and OP-1 protein are required for normal kidney development, an each are expressed at high levels during kidney tissue development. As described above the OP-1 promoter region contains Wt-1 consensus binding elements. Wt-1 protein also has been shown to negatively regulate the transcription of the insulin growth factor II gene and the platelet-derived growth factor A chain gene. Kreidberg et al., Cell, 1993, 74: 679-691. Without being limited to a given theory, it may be that Wt-i protein, either alone or in combination with one or more molecules is involved in the expression of OP-i. For example, Wt-1 protein may act in concert with a nuclear hormone receptor element, including, for example, the estrogen receptor element.
P:\OPER\JMS\36757DIV.WPD- 17/4/02 Implication of Tissue Specific Differential Regulation of OP-1 Expression Estrogen also has been shown to inhibit the uterine expression of calbindin-D 2 8 k, a vitamin D dependent calcium binding protein, the a-subunit expression of the glycoprotein hormones, and other proteins involved in bone formation. Estrogen also has been shown to cause dramatic decreases in the steady state mRNA levels of the bone matrix proteins osteocalcin, prepro a2 chain type I collagen, osteonectin, osteopontin, and alkaline phosphatase in an ovariectomized rat, which is a rat model for osteoporosis.
Estrogen appears to mediate its beneficial effect on bone metabolism in the osteoporotic model through inhibition of osteoclasts. Estrogen does not reverse osteoporosis. By contrast, OP-1, which is expressed in uterine, renal and bone tissues, is able to induce an increase in bone mass in the osteoporotic model. Thus, the negative effect of estrogen on OP-1 expression in uterine tissue may seem unexpected in view of estrogen's effect on bone metabolism.
In addition to the 5' non-coding DNA sequences of OP-1, the other non-coding sequences such as introns and 3' non-coding sequences may be involved in the modulation of OP-1 protein expression. This invention presents a method in which these non-coding sequences are assayed while in operative association with a reporter gene for their influence on the expression of OP-1. Non-coding sequences which are involved in the modulation of OP-1 expression will be identified by culturing cells transfected with the non-coding sequences, in operative association with a reporter gene, with one or more compound(s), measuring the level of reporter gene expression, and comparing this level of expression to the level of reporter gene expression in the absence of the compound(s).
EXAMPLARY CELLS, VECTORS, REPORTER GENES AND ASSAYS FOR USE IN SCREENING COMPOUNDS WHICH MODULATE OP-1 REGULATORY
SEQUENCES
I. Useful Cells P:\OPER\JMS\36757DIV.WPD- 17/4/02 -26- Any eukaryotic cell, including an immortalized cell line suitable for long term culturing conditions is contemplated to be useful for the method and cell of the invention. Useful cells should be easy to transfect, are capable of stably maintaining foreign DNA with an unrearranged sequence, and have the necessary cellular components for efficient transcription and translation of the protein, including any elements required for post-translational modification and secretion, if necessary.
Where the cell is to be transfected with a non-dominating selection gene, the cell genotype preferably is deficient for the endogenous selection gene. Preferably, the cell line also has simple media composition requirements, and rapid generation times.
Particularly useful cell lines are mammalian cell lines, including myeloma, HeLa, fibroblast, embryonic and various tissue cell lines, kidney, liver, lung and the like.
A large number of cell lines now are available through the American Type Culture Collection (Rockville, MD) or through the European Collection of Animal Cell Cultures (Porton Down, Salisbury, SP4 OJG, U.K.) Where, as here, the expression of a reporter gene that is controlled by noncoding sequences of the morphogen OP-1 is to be analyzed, particularly useful cells and cell lines are envisioned to include eukaryotic, preferably mammalian cells of a tissue and cell type known to express OP-1 and/or closely related proteins. Such cells, include, without limitation, cells of uro-genital cell origin, including kidney, bladder and ovary cells, lung, liver, mammary gland and cardiac cells, cells of gonadal origin, cells of gastrointestinal origin, glial cells and other cell lines known to express endogenous genes encoding morphogenic proteins. Preferred cell lines are of epithelial origin.
11. Exemplary VectorsNector Construction Considerations Useful vectors for use in the invention include, but are not limited to cosmids, phagemids, yeast artificial chromosomes or other large vectors. Vectors that can be maintained within the nucleus or integrated into the genome by homologous recombination are also useful. For example a vector such as PSV2CAT would be useful.
Selected portions of non-coding OP-1 sequence can be cloned into a useful P:\OPER\JMS\36757DIV WPD- 17/4/02 -27 vector using standard molecular cloning techniques as will be apparent to one of ordinary skill in the art. Restriction endonuclease sites will be utilized when possible, and can be engineered into the sequence when needed. If restriction endonuclease sites are needed to be engineered into the sequence, eight base recognition sites are preferable because they generally occur infrequently in DNA and will enhance a practitioners ability to obtain the sequence of interest. Restriction endonuclease sites can be engineered into the non-coding sequence using the common techniques such as site directed mutagenesis and PCR with primers including the desired restriction endonuclease site.
As discussed above, murine and human OP-1 sequences share a region of high homology covering approximately 750 bases upstream of the translation initiation site as shown by the shading in Fig. 1. This region is positions 2548-3317 of Seq. ID No.
1 and positions 1549-2296 of Seq. ID No. 2. The mRNA transcription initiation site lie within this region at position 2790 of Seq. ID No. 1 and by analogy at position 1788 of Seq. ID No. 2, shown in Fig. 1 by the upward arrow. This suggests that positions 2548-2790 of Seq. ID No. 1 and 1549-1788 of Seq. ID No. 2 contain conserved promoter elements for the expression of OP-1 mRNA, and approximately 500 bases at positions 2791-3317 of Seq. ID No. 1 and positions 1790-2296 of Seq. ID No. 2 contain conserved elements of the transcribed, but not translated, sequences all or part of which may be involved in the regulation of OP-1 expression. Additionally sequences upstream of the homology region may also be involved in the regulation of OP-1 expression. Thus a range of upstream sequences, including sequences upstream of the transcription initiation site and not including the approximately 500 bases of transcribed sequence, can be fused in operative association with a reporter gene to modulate expression of the gene.
3' non-coding sequences and intron sequences also can be fused in operative association with a reporter gene, either separately or in combination with each other or with 5' non-coding sequences. For example, one can place the 5' sequences defined by positions 2790-3317; 2548-2790 or 2548-3317 of Seq. ID No. 1. and either/both of 3' sequences or intron sequences in operative association with a P:\OPERVMS\36757DIV.WPD- 17/4/02 -28reporter gene. The positions of the six introns are shown in Seq. ID No. 1 as bases 3736 to 10700; bases 10897 to 11063; bases 11217 to 11424; bases 11623 to 13358; bases 13440 to 10548; bases 15166 to 17250; Also envisioned is a nucleic acid construct comprising a small fragment of non-coding OP-1 sequence in combination with additional conserved elements such as one or more Wt-1/Egr-1 binding sequences; a TCC binding sequence and/or a FTZ binding sequence in operative association with a reporter gene. Such a nucleic acid construct also could include intron sequences and/or 3' non-coding sequences.
A range of useful 5' non-coding fragments has been provided, and as will be apparent to those of ordinary skill in the art, smaller fragments of OP-1 sequence also are useful. Such smaller fragments can be identified to deleting bases from one or both ends of the provided 5' non-coding fragments, using techniques that are well known in the art and testing the truncated constructs for their ability to modulate reporter gene expression. In this way, the shortest modulating sequences can be identified.
III. Transfection Considerations Any method for incorporating nucleic acids into cells of interest is contemplated in the method of the invention. Calcium phosphate (CaPO 4 followed by glycerol shock is a standard means used in the art for introducing vectors, particularly plasmid DNA into mammalian cells. A representative method is disclosed in Cockett et al..
(1990) Biotechnology 8: 662-667, incorporated herein by reference. Other methods that may be used include electroporation, protoplast fusion, particularly useful in myeloma transfections, microinjections, lipofections and DEAE-dextran mediated uptake. Methods for these procedures are described in F.M. Ausubel, ed., Current Protocols in Molecular Biology, John Wiley Sons, New York (1989).
As will be appreciated by those having skill in the art, optimal DNA concentrations per transfection will vary according to the transfection protocol. For calcium phosphate transfection, for example, preferably 5-10 pg plasmid DNA per P:\OPERJMS\36757DIV.WPD- 17/4/02 -29plasmid type is transfected. In addition, the DNA to be tranfected preferably is essentially free of contaminants that may interfere with DNA incorporation. A standard means used in the art for purifying DNA is by ethidium bromide banding.
IV. Exemplary Reporter Genes There are numerous reporter systems commercially available, which include, without limitation, the chloramphenicol acetyltransferase luciferase, GAL4, and the human growth hormone (hGH) assay systems.
CAT is a well characterized and frequently used reporter system and a major advantage of this system is that it is an extensively validated and widely accepted measure of promoter activity. See, for example, Gorman, Moffat, and Howard, B.H. (1982) Mol. Cell. Biol., 2: 1044-1051 for a description of the reporter gene and general methodology. In this system cells are harvested 2-3 days after transfection with CAT expression vectors and extracts prepared. The extracts are incubated with acetyl CoA and radioactive chloramphenicol. Following the incubation acetylated chloramphenicol is separated from nonacetylated form by thin layer chromatography. In this assay the degree of acetylation reflects the CAT gene activity with the particular promoter.
Another well-recognized reporter system is the firefly luciferase reporter system.
See, for example Gould, and Subramani, S. (1988) Anal. Biochem., 7: 404-408 for the description of the reporter gene and general methodology. The luciferase assay is fast and has increased sensitivity. The system also is particularly useful in bulk transfections or if the promoter of interest is weak. In this assay transfected cells are grown under standard conditions, and when cultured under assay conditions both ATP and the substrate luciferin is added to the cell lysate. The enzyme luciferase catalyzes a rapid, ATP dependent oxidation of the substrate which then emits light.
The total light output is measured using a luminometer according the manufacturer's instructions Cromega) and is proportional to the amount of luciferase present over a wide range of enzyme concentrations.
P:\OPER\JMS\36757DIV.WPD- 17/4/02 A third reporter system is based on immunologic detection of hGH, it is quick and easy to use. (Selden, Burke-Howie, K. Rowe, Goodman, and Moore, D.D. (1986), Mol. Cell. Biol., 6: 3173-3179 incorporated herein by reference).
hGH is assayed in the media, rather than in cell extracts. This allows direct monitoring over by a single population of transfected cells over time.
As indicated above and as will be appreciated by those having ordinary skill in the art, particular details of the conventional means for transfection, expression, and assay of recombinant genes are well documented in the art and are understood by those having ordinary skill in the art. The instant invention enables and discloses vectors, cells and a method for screening compounds to determine the capability of compounds to modulate the expression of OP-1 via the non-coding sequences of the OP-1 genomic DNA.
Further details on the various technical aspects of each of the steps used in recombinant production of foreign genes in mammalian expression systems can be found in a number of texts and laboratory manuals in the art, such as, for example, F.M. Ausubel et al., Ed., Current Protocols in Molecular Biology, John Wiley Sons, New York, (1989).
VIII. Exemplary Homologous/Non-Homologous Recombination One approach to screen for inducers of (organ-specific) OP-1 expression in a particular cell line derived from a particular tissue such as renal or uterine tissue, is through gene targeting by homologous recombination (Sedivy et al., W.H. Freeman Co., New York (1992); A.S. Waldman, Crit. Rev. Oncol. Hematol. 12, 49 (1992)).
In one strategy of endogenous (genomic) OP-1 gene is replaced by another reporter gene which is optimally suited for screening assays, such as the firefly luciferase gene.
To target the OP-1 gene in an appropriate cell line, e.g. a kidney cell line or NGT-2, the following arrangement of genetic elements can be assembled.
Genomic OP-1 upstream and promoter sequences preferably 3000 to 5000 nucleotides in length, and which mediate the homologous recombination, are attached to the luciferase gene. The OP-1 upstream sequences down to the first coding ATG P:\OPER\MS\36757DIV.WPD- 17/4/02 -31 can be attached at the start codon ATG of the luciferase coding sequence, using a restriction site such as Ncol, which can be introduced by site directed mutagenesis into both the promoter and the luciferase sequences.
Also included is a selective marker, preferably the neo gene, without its own promoter. Preferably, selectable marker (neo) is placed downstream of the reporter gene (luciferase), after an intercistronic sequence derived from the poliovirus genome and which allows translation of the sequence marker on the same transcript as the reporter gene transcripts. Details of this approach, including specific intercistronic sequences and the detailed steps of homologous recombination, are described in the art, including (Jasin et al., PNAS USA 85: 8583 (1988); Sedivy et al., PNAS USA 86, 227 (1989); Dorin et al., Science 243: 1357 (1989) the disclosures of which are incorporated herein by reference. As described therein, the endogenes OP-1 gene is replaced by the luciferase and neo coding sequences and the expression of these sequences then assayed in a standard A screening protocol.
A genetic arrangement of OP-1 promoter (as much genomic OP-1 upstream sequence as possible, up to 10,000 bp) and reporter gene (without its original promoter but joined directly to the OP-1 ATG or in its vicinity) can also be introduced into cells on standard eukaryotic expression vectors. These vectors carry selectable markers (neo, dhfr, etc.) and will typically be integrated into the host genome with variable copy number ranging from one to several copies without efforts at amplification. Also, if desired, the vector or gene copy number can be enhanced using a well characterized amplifiable gene, such as dhfr in conjunction with methotrexate. Commercial vectors designed for autonomous replication without integration are readily available. One source vector is the Episomal Expression Epstein Barr Virus Vector (pREP, Invitrogen Corp., San Diego CA).
Introns also can be tested for regulatory sequences as described hereinabove using the methods described herein. One or more intron sequences derived from a genomic OP-1 locus preferably is introduced into proper mammalian cells using, for example, a yeast artificial chromosome (pYACneo, Clontech, Inc., Palo Alto, CA) (Ref.
P:\OPERUMS\36757DIV.WPD- 17/4/02 32 Albertson, H.M. et al., PNAS USA, 87: 4256, 1990), or other vectors adapted to allow transfer of large sequences, up to 1 megabases. As for the OP-1 5' or 3' noncoding sequences described above, the intron sequence or a portion thereof is incorporated in operative association with a reporter gene and the ability of the sequence to modulate reporter gene expressions then associated.
X. Exemplary Screening Assay for Compounds which Alter OP-1 Gene or Reporter Gene Levels Candidate compound(s) which may be administered to affect the level of a given endogenous morphogen, such as OP-1, or a reporter gene that is fused to OP-1 non-coding sequence may be found using the following screening assay, in which the level of reporter gene production by a cell type which produces measurable levels of the reporter gene expression product by incubating the cell in culture with and without the candidate compound, in order to assess the effects of the compound on the cell.
This can be accomplished by detection of the reporter expression product either at a protein or RNA level. The protocol is based on a procedure for identifying compounds which alter endogenous levels of morphogen expression, a detailed description also may be found in PCT US 92/07359.
Cultured cells are transfected with portions of OP-1 non-coding sequences in operative association with a reporter gene, and such transfected cells are maintained with the vector remaining as a plasmid in the cell nucleus or the vector can be integrated into the host cell genome, preferably at the OP-1 genomic locus.
Cell samples for testing the level of reporter gene expression are collected periodically and evaluated for reporter gene expression using the appropriate assay for the given reporter gene as indicated in the section describing reporter gene assays, or, alternatively, a portion of the cell culture itself can be collected periodically and used to prepare polyA(+) RNA for mRNA analysis.
Once candidate compounds are identified, they can be produced in reasonable, useful quantities using standard methodologies known in the art. Amino acid-based P:\OPER\JMS\36757DIV.WPD- 17/4/02 -33molecules can be encoded by synthetic nucleic acid molecules, and expressed in a recombinant expression system as described herein above or in the art. Alternatively, such molecules can be chemically synthesized, by means of an automated peptide synthesizer, for example. Non-amino acid-based molecules can be produced by standard organic chemical synthesis procedures.
Provided below is an exemplary protocol for carrying out the method of the invention, using the CAT gene as the reporter gene and one or more mammalian cell lines known to express OP-1. The example is non limiting, and other cells, reporter genes and OP-1 non-coding sequences are envisioned.
Exemplary Construction of Representative Vectors for Transfection A DNA fragment containing the OP-1 promoter can be joined to a reporter gene for transfection into a cell line that expresses endogenous OP-1. Suitable cell lines are selected by Northern blot hybridization to an OP-1 specific probe (by analyzing the cell extracts for OP-1 mRNA). Using this technology we have found several cell lines which make high levels of OP-1 mRNA, and some of these lines are the kidney line IMCD, the bladder line NGT II.
An approximately 5 Kb EcoRI, BamHI genomic fragment containing approximately 4 Kb of upstream OP-1 sequences as well as part of the first intron is blunt-ended with T4 DNA polymerase and cloned into a polylinker of a pUC vector (p0146-1). An approximately 3.5 kb DNA fragment containing human OP-1 upstream sequences is obtained by deleting a portion of coding sequences and the first intron from p0146-1 with the restriction enzyme Ehel. The -3.5kb fragment has blunt ends and contains mostly 5' non-coding sequences and also includes a short stretch of bases into the OP-1 gene. This upstream fragment is of -3.5kb ligated to a 1.6 kb Hindlll-BamHI fragment from the CAT gene obtained from the vector SV2CAT by Hindlll end blunted ligation. The 1.6kb CAT gene fragment contains about 70 bases of upstream sequences. These ligated fragments are cloned into Bluescript KS(-) vector (Stratgene, La Jolla, CA). This construct in turn is subjected to site specific mutagenesis to delete the extra sequences (approximately 30 bases) from the 3' end P:\OPER\MS\36757DIV.WPD- 17/4/02 -34of the OP-1 upstream sequences and the adjacent 5' non-coding sequences (approximately 70 bases) from the CAT gene. This mutagenesis results in the elimination of any OP-1 coding sequences from the promoter fragment as well as any non-coding sequences upstream of the CAT gene. Thus the resulting construct is a fusion of OP-1 upstream sequences with the CAT gene sequences which encode the CAT protein. This approximately 5 kb fragment is then excised from Bluescript using Hindlll and BamHI and ligated into a Hindlll-BamHI cut and gel purified back-bone of the pSV2CAT vector, for transfection into suitable cell lines.
Suitable cell lines include cell line that have been shown to contain high level of OP-1 mRNA, indicating that the OP-1 promoter is active in the cells. Two of these cell lines are mouse inner medullary collecting duct (IMCD) cells, and the rat bladder carcinoma line (NBT II). However other cell lines of the uro-genital system that produce high levels of the OP-1 message can be used in addition to the many previously mentioned cell types and cell lines.
The transfection of this vector into an OP-1 producing cell line is accomplished following standard techniques, transfection using calcium phosphate, liposome mediated transfection, electroporation, or DEAE-dextran transfection.
The transfected cells are harvested 48-72 hours after transfection with the CAT expression vector and extracts are made by successive freeze-thawing. 2 pl of 200 pCi/ml 14C-choramphenicol (35 to 55 mCi/mmol), 20 pl of 4 mM acetyl CoA, 32.5 pl of 1 M Tris-Hcl, pH 7.5, and 75.5 pl of water is added to 20 ml of cell extract, and incubated for 1 hour at 37 degrees Celsius. Upon completion of incubation, 1 ml ethyl acetate is added to the reaction, microcentrifuged for 1 minute and the top layer is removed. This top layer is dried down in a SpeedVac for 45 minutes, and each sample is resuspended in 30 ml of ethyl acetate. The samples are spotted onto a plasticbacked TLC sheet for chromatograph. The thin layer is then developed in a tank containing 200 ml of 19:1 chloroform/methanol. The chromatography is run for 2 hours and placed under film for autoradiography. The activity of the C 1 4 in the monoacetylated chloramphenicol series is calculated as described in Current Protocols P:\OPER\JMS\36757DIV.WPD- 1714102 in Molecular Biology, 1993 (Ausubel et al., eds. John Wiley Sons, New York).
Upon determination of CAT activity, the main construct can be deleted in sections to determine the regions that are responsible for the observed CAT activity.
Alternatively, he upstream sequences can be deleted unidirectionally, using an exonuclease such as Ba131, and the deletion product can be analyzed in the CAT activity assay. This system can also be used in the method of the invention to screen compounds for their ability to modulate OP-1 expression by dividing the cells into several groups, and culturing one group in the absence of any added compounds, and culturing the other groups with one or more candidate compound, and comparing the resulting levels of CAT activity.
While a readily assayable, well characterized, non OP-1 reporter gene is preferred in the method disclosed herein, as will be appreciated by those having ordinary skill in the art, OP-1 coding sequence also may be used in the screening method of the invention. The OP-1 expression preferably is determined by an immunoassay or by Northern or dot blot or other means for measuring mRNA transcript. See, for example, WO 95/11983, published May 4, 1995 for a detailed description on assaying changes in OP-1 levels in a cell or fluid.
XI. Exemplary Screening Assay for Compounds which Alter OP-1 Gene Expression in Endogenous Cell Type Models OP-1 is expressed in a variety of different cell types, including renal, bone, lung, heart, uterine, cardia and neural tissue. Candidate compounds can be identified which have a modulating effect on cells of one tissue type but not another, and/or wherein the effect is modulated in the different cells. The assay described below can be used to evaluate the effect of a candidate compound(s) in a particular cell type known to express OP-1 under physiological conditions.
Cell cultures of kidney, adrenals, urinary bladder, brain or other organs, may be prepared as described widely in the literature. For example, kidneys may be explanted from neonatal or new born or young or adult rodents (mouse or rat) and used in organ P:\OPER\JMS\36757DIV.WPD -17/4/02 -36culture as whole or slice (1-4mm) tissues. Primary tissue cultures and established cell lines, also derived from kidney, adrenals, urinary, bladder, brain, mammary, or other tissues may be established in multiwell plates (6 well or 24 well) according to conventional cell culture techniques, and are cultures in the absence or presence of serum for a period of time (1-7 days). Cells may be cultured, for example, in Dulbecco's Modified Eagle medium (Gibco, Long Island, NY) containing serum fetal calf serum at Gibco) or in serum-deprived medium, as desired, or in defined medium containing insulin, transferrin, glucose, albumin, or other growth factors).
Samples for testing the level of OP-1 production includes culture supernatants or cell lysates, collected periodically and evaluated for OP-1 production by immunoblot analysis (Sambrook et al., ed., 1989, Molecular Cloning, Cold Spring Harbor Press, Cold Spring Harbor, NY), or a portion of the cell culture itself, collected periodically and used to prepare polyA+ RNA for RNA analysis. To monitor de nova OP-1 synthesis, some cultures are labeled according to conventional procedures with an methionine/ 35 S-cysteine mixture for 6-24 hours and then evaluated to OP-1 synthesis by conventional immunoprecipitation methods.
XII. Exemplary In vivo Animal Model for Testing Efficacy of Compounds to Modulate OP-1 Expression It previously has been to demonstrated that OP1 can effect osteoporosis on the standard ovariectomized rat model, as indicated by the dose-response increase in alkaline phosphate and osteocalcin levels following injection with OP-1. The osteoporotic rat model provides an in vivo model for evaluating the efficacy of a candidate modulating compound. In order to determine the effect of a candidate morphogen stimulating agent on OP-1 production and, thereby, on bone production in vivo, alkaline phosphate and osteocalcin levels are measured under conditions which promote osteoporosis, wherein osteoporosis is induced by ovary removal in rats and in the presence and absence of a candidate modulating compound. A compound competent to enhance or induce endogenous OP-1 expression should result in increase osteocalcin and alkaline phosphate levels.
P:\OPER\JMS\36757DIV.WPD- 17/4/02 -37- Forty Long-Evans rats (Charles River Laboratories, Wilmington) weighting about 200g each are ovariectomized (OVX) using standard surgical procedures, and ten rats are sham operated. The ovariectomization of the rats produces an osteoporotic condition within the rats as a result of decreased estrogen production. Food and water are provided ad libitum. Eight days after ovariectomy, the rats, prepared as described above, are divided into three groups: sham-operated rates; ovariectomized rats receiving 1 ml of phosphate-buffer saline (PBS) i.v. in the tail vein; and (C) ovariectomized rats receiving various dose ranges of the candidate stimulating agent either by intravenous injection through the tail vein or direct administration to kidney tissue.
The effect of the candidate compound on in vivo bone formation can be determined by preparing sections of bone tissue from the ovariectomized rats. Each rat is injected with 5 mg of tetracycline, which will stain the new bone (visualized as a yellow color by fluorescence), on the 15th and 21st day of the study, and on day 22 the rats are sacrificed. The body weights, uterine weights, serum alkaline phosphate levels, serum calcium levels and serum osteocalcin levels then were determined for each rat. Bone sections are prepared and the distance separating each tetracycline straining is measured to determine the amount of new bone growth. The levels of OP- 1 in serum following injection of the candidate agent also can be monitored on a periodic basis using, for example, the immunoassay described in sections V and VII above.
V. Exemplary Determination of OP-1 Protein Production Where OP-1 acts as the reporter gene, detection of the gene product readily can be assayed using antibodies specific to the protein and standard immunoassay testings. For example, OP-1 may be detected using a polyclonal antibody specific for OP-1 in an ELISA, as follows.
1 ug/100 /l of affinity-purified polyclonal rabbit IgG specific for OP-1 is added to each well of a 96-well plate and incubated at 37 0 C for an hour. The wells are P:\OPERU\MSU6757DIV.WPD- 17/4/02 -38washed four time with 0.167M sodium borate buffer with 0.15 M NaCI (BSB), pH 8.2 containing 0.1% Tween 20. To minimize non-specific binding, the wells are blocked by filling completely with 1% bovine serum albumin (BS) in BSB and incubating for 1 hour at 370C. The wells are then washed four times with BSB containing 0.1% Tween 20. A 100 ul aliquot of an appropriate dilution of each of the test samples of cell culture supernatant is added to each well in triplicate and incubated at 370C for 30 min.
After incubation, 100 1 l biotinylated rabbit anti-OP-1 serum (stock solution is about 1 mg/ml and diluted 1:400 in BSB containing 1% BSA before use) is added to each well and incubated at 370C for 30 min. The wells are then washed four times with BSB containing 0.1% Tween 20. 100 streptavidin-alkaline (Southern Biotechnology Associates, Inc. Birmingham, Alabama, diluted 1:2000 in BSB containing 01% Tween before use) is added to each well and incubated at 370C for 30 min. The plates are washed four times with 0.5M Tris buffered Saline (TBS), pH 7.2. 50 substrate (ELISA Amplification System Kit, Life Technologies, Inc., Bethesda, MD) is added to each well incubated at room temperature for 15 min. Then, 50 Ml amplifier (from the same amplification system kit) is added and incubated for another 15 min at room temperature. The reaction is stopped by the addition of 50 /l 0.3M sulphuric acid. The OD at 490 nm of the solution in each well is recorded. To quantitate OP-1 in culture media, a OP-1 standard curve is performed in parallel with the test samples.
VI. Exemplary Production of OP-1 Polyclonal and Monoclonal Antibody Polyclonal antibody for OP-1 protein may be prepared as follows. Each rabbit is given a primary immunization of 100 kg/500 E. coli produced OP-1 monomer (amino acids 328-431 in SEQ ID NO:5) in 0.1%SDS mixed with 500 /l Complete Freund's Adjuvant. The antigen is injected subcutaneously at multiple sites on the back and flanks of the animal. The rabbit is boosted after a month in the same manner using incomplete Freund's Adjuvant. Test bleeds are taken from the ear vein seven days later. Two additional boosts and test bleeds are performed at monthly intervals until antibody against OP-1 is detected in the serum using an ELISA assay. Then, the rabbit is boosted monthly with 100 Mg of antigen and bled (15 ml per bleed) at days seven and ten after boosting.
P:\OPERUMS\36757DIV.WPD- 17/4/02 -39- Monoclonal antibody specific for OP-1 protein may be prepared as follows. A mouse is given two injections of E. coli produced OP-1 monomer. The first injection contains 100 /g of OP-1 in complete Freund's adjuvant and is given subcutaneously.
The second injection contains 50 ug of OP-1 in incomplete adjuvant and is given intraperitoneally. The mouse then receives a total of 230 Mg of OP-1 (amino acids 307-431 in SEQ ID NO:5) in four intraperitoneal injections at various times over an eight month period. One week prior to fusion, both mice are boosted intraperitoneally with 100 mg of OP-1 (307-431) and 30 /g of the N-terminal peptide (Ser 293 -Asn 309 -Cys) conjugated through the added cysteine to bovine serum albumin with SMCC crosslinking agent. This boost was repeated five days four days three days (IP) and one day (IV) prior to fusion. The mouse spleen cells are then fused to myeloma 653) cells at a ratio of 1:1 using PEG 1500 (Boeringer Mannheim), and the cell fusion is plated and screened for OP-1-specific antibodies using OP-1 (307- 431) as antigen. The cell fusion and monoclonal screening then are according to standard procedures well described in standard texts widely available in the art.
VII. Exemplary Process for Detecting OP-1 in Serum Presented below is a sample protocol for identifying OP-1 in serum. Following this general methodology OP-1 may be detected in body fluids, including serum, and can be used in a protocol for evaluating the efficacy of an OP-1 modulating compound in vivo.
A monoclonal antibody raised against mammalian, recombinantly produced OP- 1 using standard immunology techniques well described in the art and described generally in example VI., above, was immobilized by passing the antibody over an agarose-activated gel Affi-GelTM, from Bio-Rad Laboratories, Richmond, CA, prepared following manufacturer's instructions) and used to purify OP-1 from serum.
Human serum then was passed over the column and eluted with 3M K-thiocyanate.
K-thiocyanate fractions then were dialyzed in 6M urea, 20mM PO 4 pH 7.0, applied to a C8 HPLC column, and eluted with a 20 minute, 25-50% acetonitrile/0,1% TFA gradient. Mature, recombinantly produced OP-1 homodimers elute between 20-22 minutes, and are used as a positive control. Fractions then were collected and tested P:\OPERVMS\36757DIV.WPD- 17/4/02 for the presence of OP-1 by standard immunoblot using an OP-1 specific antibody.
Using this method OP-1 readily was detected in human serum. See also, PCT/US92/07432 for a detailed description of the assay.
IX. Considerations for Formulations and Methods for Administering Therapeutic Agents Where the OP-I-modulating agent identified herein comprises part of a tissue or organ preservation solution, any commercially available preservation solution may be used to advantage. For example, useful solutions known in the art include Collins solution, Wisconsin solution, Belzer solution, Eurocollins solution and lactated Ringer's solution. Generally, an organ preservation solution usually possesses one or more of the following properties: an osmotic pressure substantially equal to that of the inside of a mammalian cell, (solutions typically are hyperosmolar and have K+ and/or Mg++ ions present in an amount sufficient to produce an osmotic pressure slightly higher than the inside of a mammalian cell); the solution typically is capable of maintaining substantially normal ATP levels in the cells; and the solution usually allows optimum maintenance of glucose metabolism in the cells. Organ preservation solutions also may contain anticoagulants, energy sources such as glucose, fructose and other sugars, metabolites, heavy metal chelators, glycerol and other materials of high viscosity to enhance survival at low temperatures, free oxygen radical inhibiting agents and a pH indicator. A detailed description of preservation solutions and useful components may be found, for example, in US patent No. 5,002,965.
Where the OP-i-modulating agent is to be provided to an individual, the donor prior to harvest, or the recipient prior to or concomitant with transplantation, the therapeutic agent may be provided by any suitable means, preferably directly locally, as by injection to the tissue or organ locus) or systemically parenterally or orally).
Useful solutions for parenteral administration may be prepared by any of the methods well known in the pharmaceutical art, described, for example, in Remington's Pharmaceutical Sciences (Gennaro, Mack Pub., 1990. Formulations may P:\OPERVMS\36757D1V.WPD 17/4/02 -41 include, for example, polyalkylene glycols such as polyethylene glycol, oils of vegetable original, hydrogenated naphthalenes, and the like. Formulations for direct administration, in particular, may include glycerol and other compositions of high viscosity to help maintain the agent at the desired locus. Biocompatible, preferably bioresorbable, polymers, including, for example, hyaluronic acid, collagen, tricalcium phosphate, polybutyrate, lactide and glycolide polymers and lactide/glycolide copolymers, may be useful excipients to control the release of the agent in vivo.
As will be appreciated by those skilled in the art, the concentration of the compounds described in a therapeutic composition will vary depending upon a number of factors, including the dosage of the drug to be administered, the chemical characteristics hydrophobicity) of the compounds employed, and the route of administration. Where the morphogen-stimulating agent is part of a preservation solution, the dosage likely will depend for example, on the size of the tissue or organ to be transplanted, the overall health status of the organ or tissue itself, the length of time between harvest and transplantation the duration in storage), the frequency with which the preservation solution is changed, and the type of storage anticipated, low temperature. In general terms, preferred ranges include a concentration range between about 0.1 ng to 100 ag/kg per tissue or organ weight per day.
Where the therapeutic agent is to be administered to a donor or recipient, the preferred dosage of drug to be administered also is likely to depend on such variables as the type and extent of progression of the disease, the overall health status of the particular patient, the relative biological efficacy of the compound selected, the formulation of the compound excipients, and its route of administration. In general terms, a suitable compound of this invention may be provided in an aqueous physiological buffer solution containing about 0.001% to 10% w/v compound for parenteral administration. Typical dose ranges are from about 10 ng/kg to about 1 g/kg of body weight per day; and preferred dose range is from about 0.1 /g/kg to 100 mg/kg of body weight per day.
The invention may be embodied in other specific forms without departing from P:\OPERJMS\36757DIV.WPD- 17/4/02 -42the spirit or essential characteristics thereof. The present embodiments are therefore to be considered in all respects as illustrative and not restrictive, the scope of the invention being indicated by the appended claims rather than by the foregoing description, and all changes which come within the meaning and range of equivalency of the claims are therefore intended to be embraced therein.
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.
P:\OPER\JMS36757DIV.WPD- 17/4/02 -43- SEQUENCE LISTING GENERAL INFORMATION:
APPLICANT:
NAME: CREATIVE BIOMOLECULES, INC.
STREET: 45 SOUTH STREET CITY: HOPKINTON STATE: MA COUNTRY: USA POSTAL CODE (ZIP): 01748 TELEPHONE: 508-435-9001 TELEFAX: 508-435-0454
TELEX:
(ii) TITLE OF INVENTION: METHODS AND COMPOSITIONS FOR MODULATING MORPHOGEN EXPRESSION (iii) NUMBER OF SEQUENCES: 7 (iv) CORRESPONDENCE ADDRESS: ADDRESSEE: PATENT ADMINISTRATOR, CREATIVE BIOMOLECULES
INC.
STREET: 45 SOUTH STREET CITY: HOPKINTON STATE: MA COUNTRY: USA ZIP: 07148 COMPUTER READABLE FORM: MEDIUM TYPE: Floppy disk COMPUTER: IBM PC compatible OPERATING SYSTEM: PC-DOS/MS-DOS SOFTWARE: PatentIn Release Version #1.25 (vi) CURRENT APPLICATION DATA: APPLICATION NUMBER: AU 36757/99 FILING DATE: 24 JUN-1999
CLASSIFICATION:
INFORMATION FOR SEQ ID NO:1: SEQUENCE CHARACTERISTICS: LENGTH: 17415 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1: TCAACCGGTC TCTTTAGGTT TTGGCTGTGC TTATTACTAT GAGCACCTGC TGTGTGCCAG GCTCAGAATA GGCTCAGGTG ACTAGAATCC TTGCTTAGAC ACTGACGGAT CAGTTGTTTC AGACCTGCTG CCCCTGCCCC CAGCCTCACC TGCTTGTGAA GAGTAGATAA AAAGCAGAGA CTACTACTGA AGAACAGGGC
TCATTCAACA
AGATGCACAA
ATATGTAAAT
GATCCCTCCA
GGTACTAATT
AGAAGGGTAA
TGTAGCACCA
AAAGATTTGA
TGCTTTGGCT CCTTATTATT 300 P:\OPER\JMS\36757D1VWPD- 17/4/02 44
TCAGACTTTG
TAGCAAGCCA
TGCTCTGCAG
GAAGAAAATG
GGCCTGGAGG
CTCAAGCACT
ACCTCCTTTT
GCGTGTGCAG
TGCTGTTCCT
GGACTCAGGT TGTCTCTTTC TTGTCTTTCA CCACCCTCCA TCACCTCACC CTGTTTATCT TGCAGGCTGA AGTGACCCAG GCTCTCCAGT GTGGCACTCC TGCAATTTCC
CACCTTTCCT
AGAGTGATAG
TTGCTTCCCT
CCATGTCTTC
AGGTGCAAAA
TGGTGACCTC
ACTGTCCAGG
GTTGAAGGAA
TGGTGGGTGG
CTGGGGCTGT
CTCTGGCTCC
CAAGGGAGAC
ATAATTCTCT
GAGCCCCTCC
AAGGCCCTCT
TTTTACCCCG
GGCAGAGGGA
CGGTATTGAT
CTCAGGCACA
ACTGCCACGA
GCTGAGGGGT
CAGCTCACAC
ACCAGGCGCT
GGCCCCAGTG
TCTCCCTCCT
ACAAGATCTG
GGCCTGGAGG
CTGCCCAGCA
TTAGTACGGG
AGAGTAGGAA
TTGCAGGCCC
GGAGTGAATG
GTAGAGGCCC
GTGTAAATGG
CCTTTCTCCA
CCAGAGGCCA
TCCCCAGCTC
CAGACAAGCG
GGGGACCCAC
CTGCCCTCAC
CCGGTGGGTT
TGAGGGATGA
GACCCTTGGA
GGAGGGCTGA
GGCTGGAAAG
GCAGTTCACT
GGGTTCAAGG
CTCTGGGTGT
CCAGTCTCCC
GCATGTCCTC
CCTCTCCTTC
CGGCTCCTTG
TGGTGCTTGG
GCAGGGGGTA
ACCAAGTGTG
CACAGCTCGC
GATGAAGAGC
CTCCCTGGCA
CCAGGACCTG
CCCATGTGGC
CTTGGCAAAC
TCCTGCTCCA
CCCCCGCTTC
TCTCTCACCC
TGCCACTCTG
CTGAGTGGTC
ATGGATGAGG
GGGGAAGAGG
TGGGAAGCCC
ATACAGGGCC
TATTCACTCA
CACTGCGGGT
CTAGCGGGGG
TCTCTACTGG
GGCTGGGGAC
CCACCTGGGA
CAGTGGCATC
GCGCCCCCGC
TGTACACTCT
TGGGGTAGGA
CTCTGCCTGG
AGTCCCTGAA
AtGGAGTCTG
CCCTGGGACT
CTTTGTTCAT
GGGCCCTTAA
CTCCTGATAT
AGGTGGTGGG
TGGGGCTCAA
CAGGCTGGCA
CCCTCAGGCT
TACTTCTGCT
GGAAGGACAG
CCCAACCTCA
AGCCCCAGGA
GGTGCAGTAA
TGGGGACTCC
AGAGACAGGA
ATGTGAAGAC
CACTGAGTGC
CGAGCCTGGT
TGCCTTTCCC
TCCTCCAAAA
CCCCACTGCC
CAGATGGACC
TTCCTGCTTG
CTTCTGAATT
TCTTATTTAA
CCATGTGGGT
GGCCACATAA
AGGTGATTCA
GAGCTGGGGG
CCTGTGCTCC
GGGTGGTTTG
GACCTGTTCC
TTTTTCTGCT
GCCATCTAGT
CCAGAAAACT
CCCCAAAGTG
GCCCTTCCCA
GGGAAGGAGA
TTCCCTTGAG
CTGTGACAGC
GAGGAGGCCG
CAGCGGCTGG
ATAGCTGACC
TTCTGTTCCC
TGGAAAAGCC
ACTCCCCTTC
TGTCACTTAT
CTGGACGACT
CCAGGATGCC
GTTTCCAGAG
GGCTCTTGCT
TCGCTCATGG
TATCTGGGCC
GAACCACATA
GCCCTGTACG
ATGCTTGTCT
AGGGCCTTGC
ATCCCACAGC
CCTTGCCTGG
CCAAAAGTCC
GTAGAAGACC
AGGCTTAGCA
GTTCATCCCA
CAGGATGAAC
ATCCCACACC
GTGCATGGCT
ATGGGGAGGT
CTCCCCAGGC
720 780 840 900 960 1020 1080 1140 1200 1260 1320 1380 1440 1500 1560 1620 1680 1740 1800 1860 1920 1980 2040 2100 2160 2220 2280 2340 TAGTGGTGGG GCTGGGGTGT CTGGTCTCAG
CCGAAGAGGA
ACAAATCGTG
ACCAGAGGTG
GGAGGTGGGA
ACTGCGCACG
CGGAGAAGAT
AGAACCCCCC
TACAGTGGCT
CGCTGATCCG
CACGGAGTAA
CGTGCGAGGG
GGAGAA.AGCA
CTCAAACAGC AATTCCAGAC AGCAAGAGAA GCGAGGCGAG GAAACAACCT CAGCTTGGCA TAAGAAGGCA ATAAAGAAGG ACTACAGGAA AGAAAACAAA GGTCTTGGAG GTCTCTGGGA GCGTCTGATG GGGGCGGGAG GTGGTGAGTG GGGAGAGGTC CAGGCGGAGG GAATGGCGAG P:\OPERJMS\36757DIV.WPD 1 7/4/02 45 CGAGAGACAG GCTGGCAACG AAGGATACAT GGGACTAGGG
GCTTCAGGGA
GCAAGACCGG
TTCCGCGCCA CCTCCGCGCT CCCCCAACTT TGCTCGGGCC GCCTTCCTGG TCCGGACCGC TCCTCCAGCC GCATCCCCGC GACGTCCCGC AAACATTTGT CGAGCGCTCT AGAGGGAATG GGCGGGGCCG AGGGCAGGTG GGAGGCCGCC
TCCTCCTCCT
CCTCTTGTGC
CGCCGGAGGT
GGAAGAAGGA
TGGACTCCTA
CGTTCGCCGG
CCCGGGGCCA
GGCCATGTCT
GGCCGCCTGC
CGCGTAGAGC
TGGCGCTCTG
ACGAGGTGCA
AGCGCGAGAT
AGCACAACTC
GCGGCGGGCC
GCCCCCCTCT
GCTTCGTCAA
TTTGAGACTG
GGCTGAAATC
GTGTGCGCCC
GGATCCTCCG
GGGCGTTCAA
CCTGGATGTA
AGCCCTGCCT
CCTCCTCCGC
GATCCAGGGC
TGGAAGAGGG
GCGCTCGCCC
GGCTTGCTGG
GGCTGCTTTC
GCACCGAGCA
GGCGCGGGCG
CCCCTCTGCC
CGGCGCGATG
GGCACCCCTG
CTCGAGCTTC
CCTCTCCATT
GGCACCCATG
CGGCGGCCAG
GGCCAGCCTG
CCTCGGTGAG
GGAGGGAGGG
GCAGTGCCTG
CCAGGTCGGG
AAGTCCCCTC
AGCGCGGGGC
AAGGGCCCTT
GGCTCTGGCA
CCAGGCCCCA
GCACAAGGCT
TGGGTTGCCG
GCCCGCCTGC
CTGCTCCTCC
CAAGCCCTGC
GGGGGCGGGG
CAGCGGGGCC
ACCTGGGGCG
CACGTGCGCT
TTCCTGCTGC
ATCCACCGGC
TTGGGCTTGC
TTCATGCTGG
GGCTTCTCCT
CAAGATAGCC
TAAGGGCAGG
AGCCGCTTCT
CCCGAGGGTC
CCGCTGGGTC
CATGTTACGC
TCGGTCATGT
CCCGGCGAGG
TCGCGGCCGT
GGCGCGGAGG
CTCAAGGTCA
TTCCCACCGC
GAGTGCCGAG
CAGGCTCTAG
AATGAACCCA
GGCGCGGGAG
GCGCGTACCA
GGGAGAGCGC
CCGCCCGAGG
CTCCTCGCTG
CACCCGCGCC
GGTGCGCCCG
GTCCGGGCAG
CGTCTGCAGC
GTGCGGGCCC
CACTGCGAGC
GCTCCGCCCT
GCCTCCGCAG
CCCACCGCCC
ACCTGTACAA
ACCCCTACAA
ATTTCCTCAC
CGAGGGTACG
TCTATGCAGC
TCCCACCCAC
GGTGAGCCTG
CGCCGGCCGC
GAGCTGTCCC
CTGCCTTGCC
CGCACCCCCT
GCTACCGGCC
GGTCAGCGTG
CCGCTTCCAG
GGTCCGCAGC
AGGGCAGGGC
GCACCCCGTG
CTGGGCACAG
GGGCCCCTCG
CTCTGGCGCT
CCCGGGGCCC
GCGAGAGCGC
CCTCCCCGGC
CGCCTCCTCA
GGCGAGTGCG
AGCGCGGCCG
AAGTGACCGA
GGAGCCCGGA
TGCGGCGCCG
GGCCGACTTC
CCAGGAGCGG
GCGCCCGCAC
CGCCATGGCG
GGCCGTCTTC
CGACGCCGAC
CCGTCTCCTT
CCGCCCAGCT
AGCCCTATGA
TAGGGGTTAC
ATCTCTGGGG
GGGCCGGCGC
GCCCTTCCTG
TACCCTCCCT
GCTCCGAGCC
GGGGAAGAAA
GCTGGCTTAA
GACCTTCTAT
CCACCCGTCC
CGGCTCCGAT
GGCACTCAGT
CTGGGGGGAG
AAGCCCGTCC
CCCGAGGCGG
CTGCTATCCG
CAGAGGAGCG
GTTGGCTCTC
CTCGCCTTTT
GGGCGAGGGG
GCCGGGGAGG
GCGGCGCGAC
GCCCGGGTAG
CACAGCTTCG
AGCCTGGACA
CGGGAGATGC
CTCCAGGGCA
GTGGAGGAGG
AGTACCCAGG
ATGGTCATGA
TCGGGGGCAC
TTCCGCTCCT
CTCCCAAGCT
TGGGAAGGAG
CTGGAGGCAA
CGGTCCGTGA
GGCCCCTCTC
GTCAAGCCCT
TTGGGGCCCC
CGGTGCCATT
2400 2460 2520 2580 2640 2700 2760 2820 2880 2940 3000 3060 3120 3180 3240 3300 3360 3420 3480 3540 3600 3660 3720 3780 3840 3900 3960 4020 4080 4140 4200 4260 4320 4380 ACCTGTCCCC TCGTGGTGCG CCCGCCTTAG TCTCCGGGCG CCGATGCCCC ATTCTCTCTT GGCTGGAGCT GCTAATTTTC TTTGTTTTCT TTCTTTGTTT ATTTTTTTCT TTTTTCTTTT TTTTTCTTTT P:\OPER\JMS\36757D1 V.WPD. 17/4/02 46
CTTTTCTTTT
AGTGCAATGG
GTGCCTCAGC
GTATTTTAGT
CTTTTTTTTT
CGCGATCTCT
CTCCCGAGTA
AGAGACAGGG
CAGGTGATCC TCCCGCCTCA GCCCTGCCGC TAGTCTTCTA
ATTTTCAGCA
GGATCTCAGA
CGACCCCGGC
GTGCGCGCAC
ACTGATGATC
CGCTTTCATT
TCACGCCCAC
CTGGGGCGTG
CTGATAAGGT
ACTAGTGAAC
CAAAAAAGTT
ATTTAGAACC
GCATGTGTGC
ATTTTACTGG
TCTGATTTTT
TTAACTATAC
GAGCTCTGTC
TATAGCATCT
GCTGCCTTCA
GAAGGCCCTG
TAGGCCCGGG
AACTTTGGTG
GGAAGGCCAC
CTTTCATCTT
TTGGGCTTTG
GATGGCAGAG
AAACTCGAGG
TTTACCACGT
GCTGACGACC
GACGGCAGCC
ATTCTCCAGA
AAATATTTGG
CTACTTGTGT
GGGCCTGTGT
GACTGGGGGC
CCCTGGAGTT
CAAGCTTCAG
TAAATTATAC
ATCTCCAAGA
TTGTATATCA
AGGGGTATAT
CGTCGTCTGC
CAAAAAATAA
GCCCTCCTGA
TCCCTTCTGT
GAGCCTGGCT
CAAACCAAGA
GCTCCCTGTC
CGGGCTATTT
AGCTGGGTGG
GAAAGGACAA
GGGACCCTGA
TGGCTTTTAA
TCATTAGTTA
TTTTTTGAGA CGGAGTTTCA GCTCACCGCA ACCTCTGCCT GCTGGGATTA CAGGCATGCG TTTCTCCATG TTAGGCAGGC GCCTCCCAAA GTGGTGCTGG TTTTAAGTAT TTAGTGGTAG GTGGCGCGCA AACCACAGGT GCGGGGGCCT GGGGGTCCCG TGGGGCTGCA GCCGAGGGCC CTTGCTCAAA CTAACCCCCC TTTCCGAGAT AACACACCCC AACTTGCTGC GAAAACCCGA CAACATGGAA ATAATGATAC GCGGGGGAAG CGCAGATCCG CCCGGGAGCC ATTGTCTGTA CGAGGCAAGG GGAGGGAGGT TAAGCAGCCA GTTAAGAAGG TGTATGAGGT GGAAAGAAGC TCGTGTCCTC CTGGAGGAAG GGCGGGGGCA TAGCTGGGGC ACTTCTGTAT TTGTGATTTT AGGAAAATTC CAAATACATA AGCCTGGGGT TAGCCAGGGC TGGGTACCCC GGACTCCCAC GGAATCATTA AAAAGGTATT GCAAAAAAGC CCCCAGTGCT CCCAAGAGAA AGACCAGGTT GCTCCCCCCA TGGCGGCAGG TTTTCCTCCT CTGGCTGTAC AGACCGGCTT GTCTGAGCCT CTTTCTCAGG TCTAGCTTTC CAGAGCGCAC TGACCTTGTT GGTGAAGACC TGGGCTGCAG
CTCTTGCTCG
CCCGGGTTCA
CACCATGCCT
TGGTCTCGAA
GATTACAGGC
GTCCCGGGCC
TTTGGCGATT
GTTTCCGACT
GGGGAGCTCC
GGAGCAGCGC
GATAGCGCTG
ACCAAGTCAA
TGAAGCCCCA
CCTTCATGCT
CTTAATAATA
TTAGATGCCA
AAGCAGCAAT
AAGGTGCAGA
ACACCAGGAA
TTACGGAGTG
TTTAAAACAA
CATATAA.ATA
CCTTTCTCTG
TGAATGTGCA
TGTAATCTCT
TATGGGCCGG
GCTCGGAGGG
AGCAAGCTGG
ATACACCTTT
CTTAATCAGT
TGGGACATCA
TTCTTTCTCT
TTTGGCGAGA
CCCAGACTGG
AGCGATTCTC
GGCTAATTTT
CTCCCGATCT
GTGAAGCTGT
GGCAGAATCT
GGGTTGCGCG
GGAGCCGCGA
CCCTCCATAT
ACGGGCTGGG
TTTCCTGAGC
GACAGCAAAC
CGCTGGGCAC
TCCCCTCCTC
ACTAAATCCA
AA.ATTACCTT
ATATGACCTG
TGAGTGGGCT
CTGGAGAGAG
GGAGGTGGGG
TGTGTATTTA
ATGAACCGCA
GTGGGGGATT
GGTCCCAGTG
GGCTTCTGCA
CAGTGTGGGC
TGCCTCTGGG
GACTTGTTTG
CAATCCATTT
CAGGCTGGCT
CTCCAAATTA
CTCTGTCCCT
CACTTCCTGT
4440 4500 4560 4620 4680 4740 4800 4860 4920 4980 5040 5100 5160 5220 5280 5340 5400 5460 5520 5580 5640 5700 5760 5820 5880 5940 6000 6060 6120 6180 6240 6300 6360 6420 AGATGCTTCT AATGTTGGCC TTTAATTTCT GCTAAGCAGC AGCACACAAA TAAATGGCCT P:\OPER\JMS\36757DIV.WPD 17/4/02 47
GTCCCTTCTA
AGGGTTGGAG
AAAGTACTGC
TGAAGTGGAT
TGGAAGAGGG
GGAAATATCA
GTCTCCAGCT
TCCAGGAGCG
TTCACTCAAG
TCAAAACCTG
TGGAGTGCAA
TCTCCTGCCT
TTTTTTTGTA
CCTGACCTCG
CCGTGCCCGG
GTCACTCAGT
TTGTCGAGTT
TCCAGGGAGG
TACTCATCAT
CAAGCTCACA
GGTGCAGTGG
TGACCTCAGT
TTGTATTTTT
AACTCCTGGA
TGAGCCATCA
CGTGAGTCCA
TGAATGGCAA
ACATGTAAAA
TGACATTACC
ATGGTGAGCA
TCCTGTTGTA
AGGGTTGGGG
TATAGTGTTT
TGAGAGGATG
AAAGGTACAA
GAGGTGAGTG
TTTGTCCCAC
CTGAGGTTCT
AGCGTATTGG
CTCTAATTTT
TGGCGCGATC
CAGCCTCCCG
TTTTTAGTAG
TGATCCACCC
AATCTGCTCT
AAGATATTAT
AGGCATCTGG
ATGTTTCACC
GTAACTTCAG
CTGACTCCAG
CCAAATCTCG
CTCCCAAGTA
GTAATTTTTT
CTCAAGCAAT
TGCTAGTTGC
CTCAGTAAGA
TAAACAACTT
GAAACAGAGT
TTTTCTTGGA
CTTGGTCTTT
GCTTGGAATT
GGAGGTGTAG
TTCCTTGGAT
GAACAATAGA
AGAGGTGTTG
ACAAAGAGAA
CCTAAGGGAT
GGGAATTCCC
GATATGCGTG
TTTTTTTTTT
TCAGCTCACT
AGTAGCTGGG
AGACGGGGTT
GCCTCGGCCT
AATTTTTTAA
TTACAACCCC
CTTGCAGCAA
CTTCATATTG
TGGGATGGTC
CAAGATCTTA
GCTCACAGCA
GCTGGGACCA
GTAGAGACAG
CTTCCCACCT
GCACAGTTGG
GACTCCCTAC
TCTCCATAGG
GAGACTTGTC
TGCAAATCAT
AGGAGGATAT
GCACTGAATG
CTCTAGTCGA
GGAGCATGAA
AGTGCTGGCT
CATGAAAGCA
GGAGATGGAG
GCAAGCTCAG
AATACAGGCG
TCACTGTGTT
CCCAAAGTTC
AGATATCATT
ACCATAGATT
CAGCTGGCTT
AGGAAATGGG
AGATCTATCT
AACTAGAAGG
CCTTCTGCCT
TAGGCATGCA
AGTTTCACCA
TTGCCTACCA
AGGGACTTGG
TGGCCACTGA
GTTGATCTGA
GGCTCAGGAC
ACCCTGAACA
AGGTCTGGAA
CTTCATGCAT
ACCATGCCAT
ATGTAATTAT
TCTCGCTCCA
ACCTCCAGGG
CCCGCACCAT
AGCCAGGATG
TGGGATTACA
TGCAAACTTT
CAAACCTCTG
TCCTGTCTAT
CACAGAGAAC
TTAACCTGGC
GGGTGGCAGT
GTTTGCTGAG
ACTGCTGATT
AGTCAAGTAC
GGGCTGCCCT
GTCAATTATT
GTAACATCCC
TCTTTTCTCA
GGGCACAACC
TCACCCAGGC
TTCACACCAT
GCGCGGCTAA
GTCTCGATCT
GGCGTGACAG
GGGCACTTGA
TCCTAGAATG
GCTGTCTCCT
CCATTTCTCT
CACTCTTCCA
6480 6540 6600 6660 6720 6780 6840 6900 6960 7020 7080 7140 7200 7260 7320 7380 7440 7500 7560 7620 7680 7740 7800 7860 7920 7980 8040 8100 8160 8220 8280 8340 8400 8460 CAGGAGTTCA AATCCTAGCT CCTGGGCTCA AGCGATCCTC
CCACTATGCC
TGTTGCCCAG
TGGCTAATTT
CCCAGTCTTG
GAGTGCCGGG ATTACAGGTG GCGAAACTGA CAGATGAGAA AGCAGAACCT TTTCTTTCTG AGTCTTTGTT TCTCATCAAT GGTGGCCCAA GAGTTGATGA CAACAGTCCT ATAAGATTAT
ATTCTACAAA
ACTTGATGAA
ATTTATGGCT
GAAGTCTGAA
CCAAGCCCCT
CTTACTTATT
TATCAGTTAT TGATAGTTCA ATAGGCAACC
CAACTCAGAA
GTAAGAGAAG
ATCAACAGTT
ACTCATTAAT ATCAAACCCA AAATTGAATT AACTCTATGT TTCCTCGCTC ATACGACACA AAATGCCAAC TAAGAACATC CCCAAACTCA AGCAGTGGTT AAGCTTAGAA GTGAATTCTA TTGGAAAATA CCATGGGCTA ACGACTTTAA ACTTAAAAGT GGTTCTCAAA CTTCAAGGTG AATCAAAATC ATCTGTAGAG CTTGTTAAAA CACAGGTTGC TGGTCCACCC CAAGAGTGTC P:\OPER\JMS\3(67S7D[V WPD 17/4/02 48 TTGAGTCAGT AGGTCTCAAG TAGGGCTCAA GAATATGCAT TTCTAATGAG CTCCAGGTGA GTCTAAGTGT TAGTCGTCGG TCTTGGGACC ACAACTTTGG GAACAATTGA TTTAGAAGAA
CTCAAAGATC
AAAGGTACAA
ATCAAAGGTG
CTGTGGAGCA
TCCCTACAAA
ACTTTTAAGA
GGAGTGAGTG
AGAAAGGGGT
TTTTAACCAC
AGTTCAGATA
ACCCAATGCT
AACTCATGTT
GGCATTTGGG
AGTTCCCATT
GGAATATTTT TAAAATTGTG TTAGAGAGAG GTGGGATCTA AGCATTATTA AATGGTATCT GTAAAATACG CATAAACAGA AGAACAGAAA TTGTTATGCC ATGGATAAAC TTCAGGGGCC TATGGTTTGG ATGTGGTTTG ACATTATTGA GAGGTTATGG
GGGATGGGGT
GAAATTTAAT
TCATGAGGGA
CCAGGTGTGC
TGCCAGTGTA
TCCACCTTCA GGGATTAGTG CAGTCTCCAG CTAGTGGGAC TGGATTAGTT ACCATACAGT GGTTGTTATA AAGTGAGGCT GCTTCTGGTG TTTTATCTGT TTGCAGGCAC TTCCTTCCCC TTCCACTTCT CTGCCAGGTT AGGATGCAGC ATGAGGCCCT CACCAGAAGC TGACCAGATG ATCTTGAACT TCCCAGTCCC CAGAACCATG ACGCAGTCTA GAGTATTCTA GAACACTACT GACTTCTCCC TGAATATCAG GGCCCTCAGG CCAGTGACTG CCAGCTAATG GAAGCAGCCT CCTGATCCAT
GACACTAGGA
GTCAGGTACC
GTGGAGAGGG
TGAGAGACCT
CTCACAGCTG
CCTCTTTCTT
ACTAACCCAG
GGAACCCCGG
CCTCTTCAGC
TCATGGTGGA
CTCAATGGTC
TGATAACAGG
GACCTCTAAG
ACAATTTTCA
TCACAGGCTT
ACTGCTCACA
AGATGGCCCT
CACCAGGGCC
CTGGCTGGAG
TGGAGGGAGA
AGGCCTAACT
GGGTCAATGG
TCTGGGGTGG
ACTTTCAAGA
AATGGCCCAC
GGGGCAGTCT
TGAAAAAGAG
GGCAGAGTTT
TATCTCAGTT
ATAACATAGT
ACAGGGCACC
TCCAGAGCCT
TTATAGCAAC
ATACTCTGGC
CACATTCCTC
CTATCATAGA
GGCCAACAAT
TGAGCTGTGA
ATGTGCAAAC
AGAGACGGGC
GGTAATAAAA
ATCTTTATGT
GGGATGGTTG
GCTTTTAGAC
CTGCTACCTA
TCACCTTCAA
GGGAGGTTTT
AAGGACCAGC
CAAGATGAGA
AATTAAAGGA
CTACCCTCCC
TCTTAGAGCC
GTGCTACTTA
AGCTAAATAA
ACAAGACAGA
CTATGGACAA
TCTGCCCCTT
CCCCACCTTT
CAGATTCACT
GTCCTACACT
TGAGATAATG
AGAGGAAAGC
GGAGGCAAAG
CCATAAGAGG
CAAGGGACCA
TAGTCTTCCT
GCACACCAGT
AAAGGCTGAA
AAGGTTGAAG
AAGTGAACTG
AGCTGTGGCA
GTCAAACTCT
CTCCTTCCCC
AGGCACGGTG
AGAAGCTTCC
ACCTTTTTTC
CTAAGACACA
GAGTGACAGA
CCTCCCTTCT
CCCCTGACTT
TTCAAGAATT
TGAAAGTTCT
GGGACATGGA
CCTGCCAAGA
ATGATTTTCC
CATCCTTGTG
TCAGTAGGAA
CCCATGCTCC
GCACCAGATG
GAGCAGACTG
ATGAAAACTT
AAGCCTCCTG
CTTACTCTGG
AGACTCGAAG
CACCTTCACC
TTGGGATCAG
AGGACCTCTT
TGGCTGCCTG
TCTATAAATT
GTGGTAGAAA
CAGACAAGAG
TGCAGAGTCT
GATTGGACCA
TGAACTAAGA
TAGCATCTTG
ACAAGGGTAA
GGAGCAGAGA
ATGCTTACAA
TCGAACCTCT
GGCATAGTAC
TCCTCCCATT
TCACTCAAAA
GCTGGGTTCT
TCACTTTTGG
GAAAGCATCT
CTTTGGAAAT
GAGAAGATCT
TCTTCTTTCA
GAACAAGGCC
GGATGGCTGT
8520 8580 8640 8700 8760 8820 8880 8940 9000 9060 9120 9180 9240 9300 9360 9420 9480 9540 9600 9660 9720 9780 9840 9900 9960 10020 10080 10140 10200 10260 10320 10380 10440 10500 GGTTAGTGCC CTACTTTTCC CAGCAGGTTG GATGCAGAAT CATGCTCTTG TCGTTCAGGA P:\OPER\JMS\36757D1V.WPD- 17/4102 49
TGACCATGGG
AAGGAGCAGT
ATGCTTAGAC
TGTGTTCTCT
ATCGAGAGTT
AATTCCGGAT
GCGTTTATCA
AGGTGCTGAG
ACCCCATGAG
GCCCGTGTCT
CTCTGGGCCT
GTGGTCAATC
TCCCCCGCCA
AGTGCTCATG
GTTCAGCCTT
GTGCACTTGG
GGGCGGCACG
GTCCACTTCC
ACGCCCAAGA
GATCACAGAC
TCATTCAGTG
AAGATCTAAG
CTTGACTCAC
GGGCGACACT
GCCAGTTGGG
CAACCAGTAG
GATCCAGGAG
TCAGTGCTGC
TGCTCCTGGG
TGGGAACTTT
TGAACCACTT
ATAGTGACTT
CAACAAAGAT
GACCATGGGT
TGTCACTGGG
TCAGAGCCAG
CTGAGCCTGT
GTCACTGGCA
AGAGTGAAAC
GACCCTCCAG
ATGGGCATGC
CCAGACACTA
TCTACAGTGT GTTGGTGAGG CTCCATCTAG CTTAGGCAAG ATGAGCTGTC GGTGTTGGTG TCCTCTTCCA GTGGAACATG ACAAGGAATT CTTCCACCCA CGCTACCACC CCGGTTTGAT CTTTCCAAGA TCCCAGAAGG GGAAGCTGTC ACGGCAGCCG CTACAAGGAC TACATCCGGG AACGCTTCGA CAATGAGACG TTCCGGATCA
GGTGCTCCAG
TTTCCTCTGG
CTCTGCTTCC
CTCCTCCTCT
CGGAGGAGGG
CGCGGCACAA
CTGCCAGTCC
CTTGCTTCAC
TTGATCCTGC
CTGCTTCCTG
GGCCCCAGAA
GCAGCATCCG
ACCAGGAAGC
CCACCACAGG
CTCACCAAGA
GAGAGCTGGG
GAGTTAGACT
GGGCTCAAGC
ATGGGTTGAG
AAAATTCACC
CTCAACAGTG
TTCCTCTCAA
CAGTAGGAGG
TTCCAGAAGT
CCTGACTCCT
TACAAAGCCT
TGCAGGGAGG
GAGCACTTGG
GGGCAGAGGA
CATCTGTTGG
CTCGCAGGGA
CTGGCTGGTG
CCTGGGCCTG
TAATGCAGCC
TACAAACAGG
AGCCCTGTCC
TCCAGGGCAG
CAAGCAGCCC
GTCCACGGGG
CCTCGGATGG
AACCCAGCAG
TGCTCTGAGC
GGTTTGACTC
CCACTCAGAG
AGACTGCCGT
GTTTGGCTGT
AGCCCGCAGA
TCCTCTGAGC
GGCCACAGCT
AAGAGAATGA
CTCTATGTCT
GGACAGGATG
CTTTGAGAAG
GGCATTGCAG
GCAGGTGGGT
AGAAGGTGGT
GGTAGTGGAG
ATCGGATCTC
TTTGACATCA
CAGCTCTCGG
TGTGCTCCTG
CTTCCCCGCC
CGCTCGTGGC
ACGATCAACC
TTCATGGTGG
AGCAAACAGC
CCAACGTGGC
GCCCCGGCGA
TCTCTTCGAT
CGAGAGCTCG
GCTGACTGTC
TTTCTATATG
AGACATCAGA
GCTAAGGTTG
CCCAGCTCCT
GTAGTTGGCC
ATGTCTCTCC
TTTAGTTTGT
TGCACTGATG
GTGACATTGG
GTGGAGGAAA
GCTATACGGG TATCTGGGAG GAGGGTTTCC CTCCCCTCCC CTGTGACCTG CTAACGCGAA TTCCTGCTCG ACAGCCGTAC CAGCCACCAG CAACCACTGG TGGAGACGCT GGATGGTGAG GACTTCAGGA GGGTCTCAGC CCTCCCAACC AGTACTCCAT CCTCCTGTAA CTGCTCTTCT CCAAGTTGGC GGGCCTGATT CTTTCTTCAA GGCCACGGAG GCAGCCAGAA CCGCTCCAAG 10560 10620 10680 10740 10800 10860 10920 10980 11040 11100 11160 11220 11280 11340 11400 11460 11520 11580 11640 11700 11760 11820 11880 11940 12000 12060 12120 12180 12240 12300 12360 12420 12480 12540
AGGGTATCTT
CCGCAGGAGA
TTTAGCAA.AC
AGCAGTCCCC
TCCAGGGTCT
GGATGAGCCT
AACCCAAGTC
GGTGGACATT
TCTGCCCCAC
AGGGGGGCTT
ATGGGTCTTT
GTTGGGTCAC
AGCTTAGCTT
AACCAAGGCT
CGGCACATGC
AGGTGGGAGG
CTGACTAAAA
CAGGAGTCCG
AAGACCTGGT
ACACCTCTAA
TCACAGGGCA
AAATGCGCTT
AGGGTTGGTT
CCCACCATCT
CATTATTTTT
CTTAGGAATG
TTGCCCTTCC
TGGGGATCTA
TGAGCAGACA
AAGAGCCCTG
CGTGGGAGTG AGCTTGGTGT TTGGTCAATC AGTTGTCAGA GCACACCGGG CCCTGTCAGC P:\OPER\JMS\36757DIV.WPD. 17/4102 50
AGGCACAGCC
GAAAGACAGG
TGGGCCTGCT CTGAGTATGA TCATGACTAG GAAAAAAGCA
CAGAGAGCCC
ATCCCTCTGT
CTGGGAAGTT
TGTGGGGTGG
CTTCTCAATG
GTAGGTGGAG
AAGGAAGGTT
TTTACAAGTG GCAGTGTGTG TGAGAGAGAG ACAAGACAGA CAGACAGACA CTCAGGCCCT GACCCGAATG CTTCCAAATT TACGTAGTTC TGGAAAACCC CCTGTATCAT TTTCACTACT CAAAGAAACC TCGGGAGTGT TTTCTTCTGA AAGGTCATCA GGTTTTGACT CTCTGCTGTC TCATTTCTTC TTGCTGGTGG TGGTGATGGT TGCTTGTCCC AGGCCCTGTC CCGCATCCTC TTGCCCCTGC AGAGGGATGA TCATAAGCAG ATCTCTTTGA TTCGATTCCC TTATGGAATC AAACCAGTGT CCGCAGAAGG ATGGGGCATT GGCACCTCCC CAGGTTAGAC CCTCGAACTG TTCATTGCTG TTCACATCAC CTCCTCACTG GGCTCTGCTT AGAGGCAGGC CTTGTAAGAA GTAAGGGGCT GGCTGGGTCT GCGGGTGCTG TGCTCAGTCT GCCTCGCTAG GGAAGGGGAC CAGAAATGTG GTTGAGACTT CCATTCTCTG CATGCACCTC GGCCGTGTGT TTGAATGTAG TTTACCCCAT TCCCATTCCC TTCCTGAACC AGTCAACTGT TCTGGAGCCA TTTGCATGGA GGGTCATGTG ATCCACCTGC rATCTCTArr nTTrTnnnrr
GCAGGGCGCC
CAGGCAGATG
TTCCAGAAAG
TTAGTAGGGC
GCTTTTGAAT
TGCCTAAGAT
CTTCACTTCC
GCACGAGCTG
GTCTTGGGTG
TGTTTCTCAT
TTGGGCTAAG
GAACTCGAAC
ATTCCCATCC
ATACCTGGGG
ACGGGAGGGA
TTCAGGGGTG
GTGTGTTGGG
TGCAGTGGCC
TAGCATTTAA
TATTATGGGA
CTTTGCTGGG
CGGGAAATTT
GGAGGAACTT
TTGTCAATGC
TATGTCAGCT
TGGGCCCTCT
CTCTGCCAGT
GATACAGGGA
CAGGAACCCA
CTTGGCTGGC
AGCCATCTCC
CGGATCTCCC
GGGTCATGTT
GCCTCACGAG
TTGTGTGAGG
ACAACACACG
TAAGACTACA
GGTAGAAATG
ACCCCCCAGC
TGATGTGTGT
AGAGAACAGC
TCCGAGACCT
GGCGTGGGCT
TAAGACTCCA
GGCCTCATGA
AACACTTTGG
TGCTTCTCAA
CCCTCTGCCC
CAGCTTGGTT
ACTGGCACAT
TGAATCTAGG
TCTCATTCAG
GAGATGCAGC
GGGTATTGTC
TTCAGTGGGA
GTGCACACAG
TACCTGCAGC
TAAGCATCAC
GAGGCAAGGA
TGCCATTATG
TAAGGTGAAC
TTGAGGTTGT
CTGGAGGGGT
TGTATAAAAG
TGAGAGAGGA
AGTTTTAAGG
CGTTCTGTGC
GTGTTTCTTT
AGCAGGCACC
GGGCTGGCAG
CCCACAGGCA
GTATCAAGTG
AATCCGAGAG
ACTCTGAACC
GATGATGCCG
TCTGACTTCA
CAGGCGCTTG
GGCTGCCCCC
AGAGGAGTGA
CAGTCACGAG
GATGAACACT
ATGAGGTCAT
GAGTCCAGAA
CCAGCTCTGA
TGGGACAGAA
TGAGGAGTAT
TGGCACATGG
AGTTATTAGC
TGCCAGTGTG
12600 12660 12720 12780 12840 12900 12960 13020 13080 13140 13200 13260 13320 13380 13440 13500 13560 13620 13680 13740 13800 13860 13920 13980 14040 14100 14160 14220 14280 14340 14400 14460 14520 14580 GTGAGGCAAA AGGCAGGGGA CGTGAGCTCT GGATCGTGAT CTnTTCTAnn TACTGGATTn
GCAATGTGTC
CAGGGCAACT
TCTTCCCTGG
GCCCTCCTGT
CCCAAACTCC
ATTGATTATT
ATCCCTTACT
CTCTCTGGCA
TGCCCTGTGG
GGTGGTATTC
GGATTATGCC
GAGCCTGCCC
AGGGGCCTCT
GGGCAACATT
GACCAGTGCA
GGTGGGCAAA
GGCTCAAATA
TACCCTCAGG
CAGACACACT
TCAGGACTGA
GCTGGAAGAT
TCTGTGCCAC
CCCGGAGCCA
CCGAGGCATG
TCCCTGGAGA
GAGCTTGTAG
CAGGGCGTAC
TGACCACATC
TCCATGTGCT
CCAGACCCTA
GCATGGGTGA
GAGGTTTGTT
P:\OPER'JMS\36757D]V. WPD 17/4/02 -51 TGACCACCTA GTGGGGGTAG AGCTGATGAT TGCCTCACAC CGGAGGCTCC TTCCTGTGCC GCGTTCTGTC CAGAAGACAC AGCCATGGAT GTCCATTTTA GGATCAGCCA AGCCCGTGGG GCTTTCCTTC ATTTTTATTT TATGTTTTTT TAGAAATGGG GTCTTGCTCT GTCACCCAGG CTGGGGTGCA GTGGTGTGAT CATACGTCAC CGCAGCTTTG AGCCGTCTTC CCACTCAGTC TACTAAGCTT GGACTATAGG CCAAGACTAT AGAGTGGTCC TTCTTTCCAT TCTTTTGGGA CCATGAGAGG CCACCCATGT TTCCTGCCCC TGCTGGGCCC TGCTGCTCAG AAGGCATGGT CTGAGGCTTT CACCTTGGTC GTGAGCCTTC GTGGTGGTTT CTTTCAGCAT GGGGTTGGGA TGCTGTGCTC AGGCTTCTGC ATGGTTTCCC ACACTCTCTT CTCCTCCTCA GGACTGGATC
ATCGCGCCTG
TCCTACATGA
TGGGGTGTGG
CTGGGGCCTG
CGTTAAPAAA
CTTCGACCCT
CTCCTTTGAG
CCCTGCTTCA
ACAGTGATCA
GCCATTTCAA
ACGTGCCCAG
AGTTGATAAG
TGGTTATGTA
CCCTGCCTTG
TTTAGTAATC
ATCTCATCTA
TTTGTTTTTT
AAGCCCTCAT
TCCCTGGCCT
CCCGGCTGCC
ACAGGAGTGC
GGATCAAAGT
GGGCAACTGT
GGAATTTGCT
GTTTGAAATT
GCCTGGAGGT
AAGGCTACGC
ACGCCACCAA
TCACCTGGGC
AGTAGATGTC
AAATCAAAAA
GGAGTTCATT
GATGCTTGTA
TTTTCCAGTA
GAAGCGGGCC
GTTAAATTTC
TCACGGCGAC
AGCTGTTAGG
GCTCCCTGTC
AATGCTTCTA
TGCCTCCTTC
TCCCTTTCAT
TAAGTGTTGG
TGGGATTGAA
GGATGTGCTG
AGGGGACACA
CATCGCCCTG
GAATTCTCCA
CTTCTAGAGA
TTAAAATCAT
GTCCATCATA
GGTAACAGAG
GCGCTACTAC
CCACGCCATC
CGGGCAGGCT
AGCCCATTGC
TTGTACTTTA
TATTTCTCCT
GTATTGTGGG
GAGGAAACAG
AGCAGGATAA
AGTCGGGTCA
ACTCAGTAGG
GATTTTGTCC
ATGAACATCA
GTGACAGAGA
TTTTCTTGTC
TTGATTCTGC
TTTTCCAAAG
GGTCCTTAGG
TGCTGTGCCA
TCTCTATCCT
TGTGCACCTA
GGGATTGTGA
GTCCTGATGA
TCAAGGTGGA
AAATGTGTAA
TGCCAGTCAC
TGTGAGGGGG
GTGCAGACGC
GCGGGGCCAC
CATGTCATGA
TGACTGGTTT
AATTTTTAAA
TGCTGGTTAC
GTAAACAGAT
TGGGATGGAG
CCAGGAAGAT
TGTTATTCCT
TTTTGCTTAG
TCTGAGCCTT
GCTCACTACC
CCTGTCCTGT
TCTTTGAGGG
CCCTTGCTCC
CTGGAAACAG
GTATCCCCTG
TCTCCAACCC
CTTCCATCAG
AATGATGCGA
AATGCTTCCA
GCAGGTGGGG
AAAGCATGCT
TGATGCTCAA
AGTGTGCCTT CCCTCTGAAC
TGGTGGGTGT
CAGATCCTGC
CTTTTGGGGG
GGTATAAAGA
GTAACTAAAA
GGTGCCTAAG
GAGAAATTTC
AGATGAGTGG
TCCATGTGAT
GCTCTGCCAA
AATCCAAGGT
TCCTGCCTAC
AGGACTACTC
GTGTTAAGTC
CAGGGGTTTT
CCTCCTCAAT
AAGAGTCCTC
GAAGGTGCCA
CTGCCTTCAT
TATTTCACCA
TTGTGGTCAT
GAGGAAATGA
A.AGGGTATGG
GGCCTATGTC
GCCTGGCACC
CACGCCATCT
TGCCTCCAAG
CCCCTTGCGC
GGAGTATAAT
GTTGTATGGG
AGCACTGGGC
AGTGAGGGGC
GGACCCATGG
AATGAGATTA
CAGCAACCAT
TCAAGGACCT
TGATCATCCA
CCTCCTTTCA
CTGGAGAAAA
TGTTTCTTTG
TGAAACTTCA
CCCAACCTGT
GGCATGTCTC
GGCCCATGGA
GAGATCTGCA
GTTTAAAAGG
GCTGATGGCT
ATGTGTAAGA
AGCAGTCACA
TACAGTTGCT
14640 14700 14760 14820 14880 14940 15000 15060 15120 15180 15240 15300 15360 15420 15480 15540 15600 15660 1S720 15780 15840 15900 15960 16020 16080 16140 16200 16260 16320 16380 16440 16500 16560 16620 PAOPERUJMS\36757D1V.WPD- 17/4/02 52
GGAAACCCAG
AACACGTGGC
GCGGTCTTCC
TTGCTTATCA
CACATCCTCT
AAGTCAGGGA
CCTGGTCCTG
ATGCTTTGTT
AGTGCTTAGA
CAGACAGGAT
CCACTGCCCA
AAGCCCTGCT
TCCAACGTCA
AAGTTTCACG
AGATCTGCTA
CTTAGTTTGC
GTGGACCTCA
GTAAGGTGCA
CACTATTCAG
AATGTCGGAT
CCCCTTGTCT
ACAGGGAGTG
GTTTCTTTAT
TGTTTTGGTG
GTGCGCCCAC
TCCTGAAGAA
TTGAAAACAA
ACACTGATCT
TTAGTCCCCG
CGAGGCACTC
GAGAAGTCCA
CAATCTACAG
CTGGCCCCTT
GCAAAACAGG
CTTGGCTTGG
AGAAACGTGG
TGTGTTTCAG
GCAGCTCAAT
CAGGACAGTG
TGGTTGGCTG
CTATTCCCTA
ATAGGCATTT
TGAGCAAGAT
TGCACAGGGC
CCTTCCCCAC
GATAATCCCA
GGAGTGTCAC
AGGCCAGTTA
GTCCACTTCA
GAATCTCTGG
CCGTCAGCTT
TTGTCTTACC
GAGTCTATGT
GGAGCACTTC
AGTTCCCCAA
TGTATAATGT
GAACTGAGTT
CTGCAGTCAT
GAACGACTCA
TCAACCCGGA
CCCTGTCTTG
AGGTTGAGTG
TCGGTCTATT
GTCCCTGTCC
TAGTGGGTCC
CAGAGAATTA
GAAAACCTCT
GTCCATGTAA
TCATTATGCC
CCGCTTCTCA
AACGGTGCCC
16680 16740 16800 16860 16920 16980 17040 17100 17160 17220 17280 17340 17400 17415 GCCATCTCCG TCCTCTACTT CGATGACAGC ATACAGAAAC ATGGTGGTCC GGGCCTGTGG CTGCCACTAG CTCCTCCGAG AATTC INFORMATION FOR SEQ ID NO:2: SEQUENCE CHARACTERISTICS: LENGTH: 2299 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: NAME/KEY: misc feature LOCATION: 1. .2299 OTHER INFORMATION: /note= 1"MOPi UPSTREAM SEQUENCE" (xi) SEQUENCE DESCRIPTION: SEQ ID NO:2: TGCATAGGTC ACACATCCCT TTCGGTGCCT CCTCCGACTG TTGCTACCCT TCTGTTCCGG CCAGGTAGCC GTGGAGGTTG GCCTGTGTGT GGCACCCATG
CCTCTACCCA
GGCTCTGACG
ACCTTACTGA
CCATGCTTGC
CAGGCCACAG
GGGCCCCTTT GTTTATTCAA TATCAAGAAC
AGGCTAGCCA
TTCTCAGAGA
AGGGCCTTAG
CTGCCCACTC
AAGGCCACAC
TGTAACGTGG
CACCACAGCA
GGTGGGCATC
CATCACCTAA
GAAGCCTGAA
AAGGCCCTTC
GGTGCCCTAT
GAACGAAAGG
TGTTTCCAGG
ACCCAACGTT
ACAGCCTTCA
TCACCGGAGG
GGTCCAGGGA
CAAGGCCTGG
AATTCCTACC
AGACCAGGCT
CCCAGCCCCT
CTCTCCCTTC
GAAAGACTGC
GGCCCAAGAA
CTCCTGCCTG
GGATGAGGCA
TCATGTCTCC
GGGGCCTCTC
GCTAAGACCT
AAGGAGCCCT
TCCCATCTCA
ACTTGCCATT
AGCTCGCAGC
AGGTCTGCAC
GCAAGTTTCT
GAGAGACCTA
CCCTAGGACT
CTGCTTGGCC
TGGGCCAGGG
TTTAGCCCCT
GGTAGTTATC
TCAAGAGGGA
TCAGATCACC
ACTCAGTACT
CAGACAGTCA
TCTGTTCCAG
CCCCCTACTC
P:\OPERJMS\36757D1 V. WPD 17/4/02 53 TTACCACCCC TGAAACGCTT GCTTGTCGCC CACCTTCAGC AAAGCAGGAA GCCTGGCTCA
CCATCCCCAC
TAGGAAACGG
GTTTCCTCCT
TCTTCTGAAC
GGGGTTATTA
GGAGGGGGTG
CATCTGGGGT
CTGGGCCCGG
GTGTGTGTGT
ATTATCTCTG
GGGAGAACGT
CGAGGGGGTT
GCCAGGTCCA
ATGCTTTCCT
AGAGAGCTGG
GGACACCCGG
CATTGGGCCA
GGGGCGGGGA
CCGGGGCTCC
GCGGGAGATC
GAGGAGGGAG
TGTAGGTCTG
TCTCTGGAGT
CGGGCCAGAA
GGCCATGTGT
CCGCTGCCCT
TCACTCACTG
AGGCTCCAAA
GGGAGGAGAG
TTGGGTACCA
GACAGTGAGG
GGAACATTCT
TTGAAGGCTT
ATCCCTTGGG
ATGTATGTGT
CCCGTCCTGG
TCCCCGAGGA
TTGCATGACT
AGAGTGGCCA
AGCACAGCCC
GCAGATTGGG
TGGGCACTCG
GGCTTGGGGA
AGTCAGTCCT
CGAGGCGGCG
GGAAAGGGGT
CTAGGGTTCG
CAAGCTGCTG
TGCTGTGCTA
CTGAGTAAAG
GGCGAGGCCG
GCCCCCTCCG
CCATTCTGGG
GAGCAGCAAT
GGAGGCTAGT
ACTGCCTACT
TACCAAAGGA
CTAGTCCCAG
TGGGTTAAAA
TGCTGGACTT
GTGTTGGGGG
AAGGTTCCCA
ACAAAAGCCA
GGAGCAAATG
CCTCCGAGGA
TTAGTTGCTA
ATTCTTCTCT
GTAAATATTT
GGGCGGGGAC
CCCGCTCCTC
GGCGGGCGAT
TTGTTGCTGG
CTCAGCGCCC
CTCCTCCCAC
GCCTTGCCGT
GACAGGGGCG
CCTTGAAGCT
CTGCCACCTG
TGAAGGCTGC
CCACTGAGAG
CCTTTGAGAC
CCTCAGGCCC
CTCATGTCAG
ATTTCACTTA
GATACTTGGG
GAGGGGAGGT
AGTGAAGTGT
GAGGAAGTGG
GGATAGCAGA
GAGTGTTGGG
GCCTTCTCGG
CACTTTGGCC
CTGGGTCCCT
GTAGAGCGCC
AGGCGCAGGT
CCCCGCTCCC
CCGGGCGCGC
TGCCCGCGGG
AGCTGCCTCT
CCCGGCCCGC
GCGTCCTGGC
TCCCGGGCAA
CGCCTGCAGC
GGGCGGCGCG
TTTGCTCCCA
ACCCAGTATC
AGGAAAATCG
CTGACCTGGG
GACCCCGCCC
TGTACTCTGT
AAGGAAAAGC
GTGTGTGTGT
AGAAAGAACT
CACCCGAGGG
GGGGCAAGCG
GGGGGCGGTT
A.TTCCTGCGC
ACTTCCAAGT
GCGGCGTCTG
CTGGGAGGAA
GGGAGGCAGC
CGGCCCCAGC
AGGGCCCTTG
CCTGAGCGCG
CCGGCACTCG
CTCCTCGCTC
G.AGTGCGGGC
AGCGCAGCCG
AAGTGACCTC
GGCCCGGTGC
TTTTTCAGAT
TGTCTGGGAC
AGTCGGGAGC
GCTAGGGGTA
CCCCAAGAGA
AGAGCTGCAG
CGAGAAGTAC
GTGTGTGTGA
TTATCTCCAC
GGGAGGGGCA
GTGGGGGTAC
CGAAAGATGA
TCCCTCCTGG
GCGAGTCCCG
TCCCAGTGCC
TGAATGAAGC
GGGAGCGGGA
GCGCCCAACT
TATTGGGCAC
ATCAGAGCGG
CTCTCCGGAC
TCTTGCTCGC
CGAGGGGCCC
GCCGGGGAGT
GGGTCGTGGA
CCCGGATCGC
780 840 900 960 1020 1080 1140 1200 1260 1320 1380 1440 1500 1560 1620 1680 1740 1800 1860 1920 1980 2040 2100 2160 2220 2280 2299 GCGTAGAGCC GGCGCGATG INFORMATION FOR SEQ ID NO:3: SEQUENCE CHARACTERISTICS: LENGTH: 2997 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: PA0PER\JMS\36757D1V.WPD- I 7/4t02 54 NAME/KEY: misc feature LOCATION: 1. .2997 OTHER INFORMATION: /note= "MOPi TERMINAL SEQUENCE" (xi) SEQUENCE DESCRIPTION: SEQ ID NO:3: TAGCTCTTCC TGAGACCCTG ACCTTTGCGG GGCCACACCT TTCCAAATCT ACCATCTAAG TCTCTCACTG CCTTCCCCTC ACCTCCCCAA CAGCTGATGA GCGCCCTTTC GCAAACGCCT AAGAGCAGGA CTGGCGCTCT GAGTCTTTGA GGCTTAGCCA GGGTGGGCGC AATGATATGT CACAATAAAA AAACAATTTA TCCCCGTTTC TGGAGGATGG AGAGGCCCCT CCCAAATCCA GCCTGTAGGG ATGTGGCCTC AAAGGGTGTT CACACACACA CACACACACA TGCACACACA CACACACACA CATGCATGCA CACACACACG CTGCCAGTCC CTCAGCCCCA CTAACAAGCA CAGCGGACCC TGTCCTCCAA ACAAGGCCCC CCTTCACAGC TGCCTCTCTT TCTGGATCTG AGGGAGATGG TCCTGCTTCA GAGGTGGGAT CAGAGCCCCA GCTGCCCCAA AGTGTTTCCT GCATCCACAC CATCTGGTCA GGTGGGGAAC TTCTGTTCTG TTTTAGTTTT TTCGATGTGG GGGAAGGCCA GCTTTTTGTG TGACTCCCCT TCGGTGTCTC TTATACGTTT TTTCCATCTG AAGCTCTTTC AGTCTGTTTG TTTTCTTATT
CCCACCTTGG
CCGGAAGCAT
CTTCTGGCAC
AAAATGTCTG
GGAGTAATCG
TGGCGTCTGT
CCCATGAATG
TTGGTTTATT
CCTTTCCGTC
AGGAGGAGGA
GGGATGAAGA
CATGCATGAT
CACACACACA
CACACACACA
ATTCCTGCCC
TACCTGAGGA
TCTGGCACCT
CACTCTCAAC
CAGAACAGGG
GTTGGTTATG
TGGTTCCTAG
AAAGAATGAG
CAATTCTTCA
TCTTCTTCAG
TTTCTTTTTG
GTGGTGCACA
AAATGTGTAA.
CAAAGGCTCC
TAAGACTATT
CGAGGAGCCA ACAGACCAAC GTAAGGGTTC CAGAA.ACCTG GTGACGGACA AGATCCTACC CCAGGAAAGT GTCCATTGGC CAAGCCTCGT TCAGCTGCAG GTTGAAGGAA AACCAAGCAG AAAATGGTTA GGATACAGAT CTGACTTTGT AAACAGAAAA
TCGTCTCGTT
GGATGTCTGC
TGGAGGGAGG
ACACACACAC
CACACACGCA
TCTGAAGCGC
CATGGTAGGA
AGCACAGGGG
CTGTGGCCGA
CCTAACAGAA
TTGTACTGGC
GCTCAAACAA
CTTCAAATGC
GTTAGCCAGG
TGTCTGTGTC
TAGCTTGGGC
TAATTTGTTC
TTTTACTTTA
ATAGTTGTGA
TCACAGAGAA
TATTAACAGT
GTGTGTGTTT
TCAGAAGAGG
TATGCATGCA
ACACACACAC
CGCACGCACG
ATGTAGACTT
AATCCATGAG
ATGCAGGCTC
GCTCCGGAGC
GGTTCTGCGA
TTAGAAGGTT
GGCCTCTCTG
AGAGGGTTAA
CAGGACCTAT
CCTGGAAACA
TGCAGCTTCT
TGTGTGTTTG
GAGCCCTAGT
CAAGACAAAG
CAATGAGGCC
TGGACCGATG
TCGATGTCTC
CTCTCCTGAG
AGCGTGCAGG
AGCTACCACA
CACATGGCCC
CAGAAGGAAG
AAGCCACTGT
ATATTTTCCT
GCCGGGGCTG
ACCAGACCTG
CCAGTGAGGG
CACACACACA
ACACACACGA
CACACGCACG
TGGAATGGCT
AAAAGCAAAG
TTCAGGACAC
CAGGTCCTGG
CAGATTGGTT
CAACCATGCT
CCTGAGTTTG
ACTGGCTGCC
GGCCATGTCG
CTGGGCTCTC
ACTCTGCCCA
CAGATCTGGG
CTGCCTGCAG
AAATTATTTA
GACTTCCTTC
TACCCATAGC
120 180 240 300 360 420 480 540 600 660 720 780 840 900 960 1020 1080 1140 1200 1260 1320 1380 1440 1500 1560 1620 1680 1740 1800 1860 TGTCGAATAA AGTGGTCCTT AGTGAAAATT CTGTATAAAT AGAGTAAGAA GGGGTTTGAC P:\OPER\JMS\36757D1V.WPD- 17/4/02 55
TTTGCAATAA
TCCCCCTGAA
ACTGTTAACG
GTGCAGCTGT
GGTAAGGCTG
CTTCAA-AGAG
TCATCCCAAT
AAGCCATGCT
GCTGCCCTCG
GCCCCCCACC
TGCGCACGAA.
GCTCTCTCTT
CCTGCGATTT
CAGGCTGAAG
TTAGCCTCTC
ACTAATCTGT
ACCTGGTTCC
CATACACACA
AAGGAGACAT
CTCCTGGACA
GGATGTTTAT
GTGGGGCTCC
GATGGCAAAG
GCAGGGGGTA
AAAATAGAGG
GGCCTCACAG
CAGGCCAAGC
TCCCCGGCAA
GGTCACCTGA
CCCCTGTTGA
CTAAACCTTT
CCCAGAGCCA
AACCCCACCC
TGCCAATATG
TCCCTGTGTG
CCCCCGCCAT
TTGGTTCTGG
CTGGAGTCTC
CATCCAGGCC
CTGGGCGGGA
AGTGCCTTTG
AATTAGCTGT
CACAAGAGAG
CTGGCGTCAT
TGTGGAGGTG
GCTCAGGGCA
CGGATGTTTC
CTCAGACCCA
CCTGTCACTG
CAAGCCGGAG
CCACCCCCAA
TTTTTGCAAA
CTGCTCCTAA.
GGCCTCATCC
TTGTCCGACC CATGTGTGTA ATCGGCTGAG AACCCTCGAC CAGGGGAAGT CGGGCGCTCC GAGACGGAAC CAAACAACAA ATTGAACTAC AGCCCAGCTG GTTTACTGCT AACATAGTCG AAGAGGGGGG GGTGTATACC TCAGTGCCCG TCACACCCGG GGCAGCCCAC CGCAGGCTGG GTGCTCATCT GGCTACATCG TAGAATCCCA GGCGATGCTT GCAACCCAGC CGTCCTAACA TCCCGACAAC TCAGCTTTTG GGTCCAGATG TGGCCTCTCA CCCCAGTGAT GTTTACACAT TAAGGAGTTT GGGCTTCTCT CTGAACAGAG GTGCCAGGGC ACAAACGGTC GAGGTCAGCT CGACCCAAAA GGAATAGGAA
TTTGTGTCTT
CTTGATCTCG
TCAATATTTG
ATGTGAGTTT
TCAGCAGCTG
AAAGATTTAG
CCAAACTTGA
GCAGTTGGGG
AGAAGGGAGT
GTCTTTGAAG
GGGACAGGCT
CATTCCAGCC
TTCTGTTTTC
GATGTGTGCC
CTTAAAAAAC
TGAGCGGGCC
CGTTGTCACA
GACATCTTCA
AATGGAA
1920 1980 2040 2100 2160 2220 2280 2340 2400 2460 2520 2S80 2640 2700 2760 2820 2880 2940 2997 AAATGGCTGA CGGATGTCTA CTTGTGCCCA INFORMATION FOR SEQ ID NO:4: Wi SEQUENCE CHARACTERISTICS: LENGTH: 9 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: NAME/KEY: misc feature LOCATION: l. .9 OTHER INFORMATION: /note= IIWTl/EGR CONSENSUS SEQUENCE,, (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:
GNGNGGANG
INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 21 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA P:\OPER\JMS\36757DIV.WPD- 17/4/02 -56- (ix) FEATURE: NAME/KEY: miscfeature LOCATION: 1..21 OTHER INFORMATION: /note= "WT1 HUMAN TCC BINDING SITE" (xi) SEQUENCE DESCRIPTION: SEQ ID TCCTCCTCCT CCTCCTCCTC C 21 INFORMATION FOR SEQ ID NO:6: SEQUENCE CHARACTERISTICS: LENGTH: 15 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: NAME/KEY: miscfeature LOCATION: 1..15 OTHER INFORMATION: /note= "WT1 MOUSE TCC BINDING SITE" (xi) SEQUENCE DESCRIPTION: SEQ ID NO:6: TCCTCCCGCT CCTCC INFORMATION FOR SEQ ID NO:7: SEQUENCE CHARACTERISTICS: LENGTH: 9 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: NAME/KEY: miscfeature LOCATION: 1..9 OTHER INFORMATION: /note= "HUMAN TFZ BINDING SITE" (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7:
TCAAGGTCA

Claims (4)

1. Use of a compound for modulating OP-1 expression, said compound identified by a method comprising the steps of: incubating a candidate compound with a cell transfected with an isolated nucleic acid comprising a reporter gene in operative association with a single nucleic acid fragment of an OP-1 specific upstream non-coding sequence, wherein said nucleic acid fragment: consists ofnucleotides 3170 to 3317, 3020 to 3317, 2790 to 3317, 2548 to 3317, 2300 to 3317, 1300 to 3317, 2548 to 2790, 1549 to 2790, or 1 to 2790 of SEQ ID NO: 1; (ii) comprises not more than one nucleic acid fragment of said OP-i upstream sequence; and (iii) is operative to regulate expression of said reporter gene; measuring the level of said reporter gene expressed in said cell; comparing said level with that of said reporter gene expressed in said cell in the absence of said candidate compound, wherein an increase in reporter gene expression level is indicative of said candidate's ability to increase OP-1 expression in vivo, and a decrease in reporter gene expression level is indicative of the candidate's ability to inhibit OP-1 expression in vivo. S.. 2. Use of a compound for modulating OP-1 expression, said compound identified by a method comprising the steps of: o* o•* 500322742 I.DOC/B SW -58- incubating a candidate compound with a cell transfected with an isolated nucleic acid, said nucleic acid comprising a reporter gene in operative association with: a first DNA sequence, wherein said first DNA sequence is: a single nucleic acid fragment of an OP-1 specific upstream non-coding sequence, wherein said nucleic acid fragment consists ofnucleotides 2548 to 3317 or 2548 to 2790 of SEQ ID NO: 1; a nucleic acid fragment of an OP-1 specific upstream non- coding sequence, wherein said nucleic acid fragment consists of nucleotides 1549 to 2297 or 1549 to 1788 of SEQ ID NO: 2; or a variant of a nucleic acid fragment of which hybridises with a nucleic acid complementary to the 15 nucleic acid fragment of (ii) under conditions of hybridisation in 40% formamide, 5XSSPE, solution, and 0.1% SDS at 37 0 C, followed by washing in 0.1% SSPE, and 0.1% SDS at 50 0 C; and (ii) a second DNA sequence comprising a sequence which interacts 20 with a DNA binding molecule and affects expression of said reporter gene, wherein; said second DNA sequence optionally comprises at least one Wt-1/Egr-1 consensus element SEQ ID NO: 4; or 500322742 I.DOC/BSW -59- said second DNA sequence is selected from the group of sequences consisting of a TTC element, a Fushi-tarazu protein binding element and a steroid responsive element; wherein said isolated nucleic acid comprises not more than one nucleic acid fragment of(1); measuring the level of reporter gene expressed in said cell; and comparing said level with that of said reporter gene expressed in said cell in the absence of said candidate compound, wherein an increase in reporter gene expression level is indicative of said candidate's ability to increase OP-1 expression in vivo, and a decrease in reporter gene expression level is indicative of the candidate's ability to inhibit OP-1 expression in vivo.
3. Use of a compound for modulating OP- 1 expression, said compound identified by a method comprising the steps of: V 15 incubating a candidate compound with a cell transfected with an isolated nucleic acid, said nucleic acid comprising a reporter gene in operative 0.o°0. association with: a first DNA sequence, wherein said first DNA sequence is: a single nucleic acid fragment of an OP-I specific upstream non-coding sequence, wherein said nucleic acid fragment consists of nucleotides 2548 to 3317 or 2548 to 2790 of SEQ ID NO: 1; a nucleic acid fragment of an OP- I specific upstream non- coding sequence, wherein said nucleic acid fragment 500322742_l.DOC/BSW consists ofnucleotides 1549 to 2297 or 1549 to 1788 of SEQ ID NO: 2; or a variant of a nucleic acid fragment of which hybridises with a nucleic acid complementary to the nucleic acid fragment of (ii) under conditions of hybridisation in 40% formamide, 5XSSPE, solution, and 0.1% SDS at 37 0 C, followed by washing in 0.1% SSPE, and 0.1% SDS at 50 0 C; (ii) a second DNA sequence comprising a sequence which interacts with a DNA binding molecule and affects expression of said reporter gene, wherein; said second DNA sequence optionally comprises at least one Wt-1/Egr-1 consensus element SEQ ID NO: 4; or said second DNA sequence is selected from the group of 15 sequences consisting of a TTC element, a Fushi-tarazu protein binding element and a steroid responsive element; and (iii) a third DNA sequence independently selected from the group consisting of a TCC element, a Fushi-tarazu protein binding 20 element, and a steroid responsive element; wherein said isolated nucleic acid comprises not more than one nucleic acid fragment of(1); measuring the level of reporter gene expressed in said cell; and comparing said level with that of said reporter gene expressed in said cell in the absence of said candidate compound, wherein an increase in reporter gene 500322742 I.DOC/BSW -61- expression level is indicative of said candidate's ability to increase OP-I expression in vivo, and a decrease in reporter gene expression level is indicative of the candidate's ability to inhibit OP-I expression in vivo.
4. A method for screening a candidate compound for the ability to modulate expression of OP-i, said method comprising the steps of: incubating a candidate compound with an epithelial cell transfected with an isolated nucleic acid comprising part or all of a nucleotide sequence encoding an OP-I pro protein in operative association with a reporter gene, said reporter gene further in operative association with a single nucleic acid fragment of an OP-l specific upstream non-coding sequence, wherein said nucleic acid fragment: consists of nucleotides 3170 to 3317, 3020 to 3317, 2790 to 3317, 2548 to 3317, 2300 to 3317, 1300 to 3317, 2548 to 2790, 1549 to 2790, or 1 to 2790 of SEQ ID NO:1; 15 (ii) comprises not more than one nucleic acid fragment of said OP-I upstream sequence; and S-(iii) is operative to regulate expression of said reporter gene; measuring the level of said reporter gene expressed in said cell;* S(b) comeasuring satheid level with that of said reporter gene expressed in said cell comparing said level with that of said reporter gene expressed in said cell 20 in the absence of said candidate compound, wherein an increase in 000. •reporter gene expression level is indicative of said candidate's ability to •increase OP-I expression in vivo, and a decrease in reporter gene S: •expression level is indicative of the candidate's ability to inhibit OP-I expression in vivo. 500322742 I.DO/BSW -62- A method for screening a candidate compound for the ability to modulate expression of OP-1, said method comprising the steps of: incubating a candidate compound with a cell transfected with an isolated nucleic acid comprising part or all of a nucleotide sequence encoding an OP-1 pro protein in operative association with a reporter gene, said reporter gene further in operative association with a single nucleic acid fragment of an OP-1 specific upstream non-coding sequence, wherein said nucleic acid fragment: consists ofnucleotides 3170 to 3317, 3020 to 3317, 2790 to 3317, 2548 to 3317, 2300 to 3317, 1300 to 3317, 2548 to 2790, 1549 to 2790, or 1 to 2790 of SEQ ID NO:1; (ii) comprises not more than one nucleic acid fragment of said OP-1 upstream sequence; and (iii) is operative to regulate expression of said reporter gene; i. 15 said cell being of urogenital, liver, bone, cardia, lung, or nerve cell origin; measuring the level of said reporter gene expressed in said cell; o comparing said level with that of said reporter gene expressed in said cell in the absence of said candidate compound, wherein an increase in reporter gene expression level is indicative of said candidate's ability to 20 increase OP-1 expression in vivo, and a decrease in reporter gene o expression level is indicative of the candidate's ability to inhibit OP-1 expression in vivo. S°6. Use of a compound for modulating OP-1 expression, substantially as herein described with reference to any one of the examples but excluding comparative examples. 500322742 I.DOC/BSW -63-
7. A method for screening a candidate compound for the ability to modulate expression of OP-1, substantially as herein described with reference to any one of the examples but excluding comparative examples. DATED this 29 th Day of March, 2004. BALDWIN SHELSTON WATERS Attorneys for: CURIS, INC. 500322742 l.DOC/BSW
AU34387/02A 1994-06-07 2002-04-17 Methods and compositions for modulating morphogenic protein expression Ceased AU773649B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU34387/02A AU773649B2 (en) 1994-06-07 2002-04-17 Methods and compositions for modulating morphogenic protein expression

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US255250 1994-06-07
AU36757/99A AU743061B2 (en) 1994-06-07 1999-06-24 Methods and compositions for modulating morphogenic protein expression
AU34387/02A AU773649B2 (en) 1994-06-07 2002-04-17 Methods and compositions for modulating morphogenic protein expression

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU36757/99A Division AU743061B2 (en) 1994-06-07 1999-06-24 Methods and compositions for modulating morphogenic protein expression

Publications (2)

Publication Number Publication Date
AU3438702A AU3438702A (en) 2002-06-27
AU773649B2 true AU773649B2 (en) 2004-05-27

Family

ID=32398673

Family Applications (1)

Application Number Title Priority Date Filing Date
AU34387/02A Ceased AU773649B2 (en) 1994-06-07 2002-04-17 Methods and compositions for modulating morphogenic protein expression

Country Status (1)

Country Link
AU (1) AU773649B2 (en)

Also Published As

Publication number Publication date
AU3438702A (en) 2002-06-27

Similar Documents

Publication Publication Date Title
US6083690A (en) Methods and compositions for identifying osteogenic agents
AU703445B2 (en) Methods and compositions for modulating morphogenic protein expression
US5916763A (en) Promoter for VEGF receptor
Ko et al. Murine and human T-lymphocyte GATA-3 factors mediate transcription through a cis-regulatory element within the human T-cell receptor δ gene enhancer
Luo et al. The matrix Gla protein gene is a marker of the chondrogenesis cell lineage during mouse development
Mittanck et al. Essential promoter elements are located within the 5′ untranslated region of human insulin-like growth factor-I exon I
WO1996038590A9 (en) Methods and compositions for identifying osteogenic agents
ZA200502131B (en) BMP-2 estrogen responsive element and methods of using the same.
US6071695A (en) Methods and products for identification of modulators of osteogenic protein-1 gene expression
EP1181056B1 (en) Lim mineralization protein splice variants
EP0842268B1 (en) Methods and compositions for identifying morphogen analogs
US5834188A (en) Methods and compositions for identifying morphogen analogs
Bunyaratavej et al. Bone morphogenetic proteins secreted by breast cancer cells upregulate bone sialoprotein expression in preosteoblast cells
US20080003675A1 (en) Methods and compositions for identifying morphogen analogs
AU773649B2 (en) Methods and compositions for modulating morphogenic protein expression
AU743061B2 (en) Methods and compositions for modulating morphogenic protein expression
Slater et al. Glucocorticoid receptor binding site in the mouse alpha-amylase 2 gene mediates response to the hormone.
AU745514B2 (en) Modulators of morphogen expression and methods of identifying the same
US6090544A (en) Methods and compositions for identifying morphogen analogs
US6103491A (en) Methods and compositions for identifying morphogen analogs
AU9719101A (en) Modulators of morphogen expression and methods of identifying the same
US6818446B2 (en) Compositions and methods for the analysis of mucin gene expression and identification of drugs having the ability to inhibit mucin gene expression
Matsui et al. The mechanism of biogenesis and potential function of the two alternatively spliced mRNAs encoded by the murine Msx3 gene
Agarwal Regulation of alpha-1 (I) collagen gene transcription by TGF-beta
US20090169526A1 (en) Bmp-6 estrogen responsive element and methods of use thereof

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)