Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NOVEL RECEPTOR-TYPE PHOSPHOTYROSINE PHOSPHATASE-KAPPA
Document Type and Number:
WIPO Patent Application WO/1994/024161
Kind Code:
A1
Abstract:
A novel receptor-type protein tyrosine phosphatase-kappa (RPTPkappa) protein or glycoprotein and the DNA coding therefor is expressed in a wide variety of mammalian tissues. The RPTPkappa protein or glycoprotein may be produced by recombinant means. Antibodies to the protein, methods for measuring the quantity of the protein, methods for screening compounds, such as drugs, which can bind to the protein and inhibit or stimulate their enzymatic activity, are provided. Further, methods for inhibiting homophilic binding of Type II RPTP, especially RPTPkappa molecules are provided.

Inventors:
SCHLESSINGER JOSEPH
SAP JAN M
ULLRICH AXEL
VOGEL WOLFGANG
FUCHS MIRIAM
Application Number:
PCT/US1994/004377
Publication Date:
October 27, 1994
Filing Date:
April 20, 1994
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV NEW YORK MED CT (US)
MAX PLANCK GESELLSCHAFT (DE)
International Classes:
A61K39/395; A61K45/00; C07H21/04; C07K14/705; C07K16/18; G01N33/50; C12N5/00; C12N5/10; C12N9/16; C12N15/00; C12N15/02; C12P21/08; C12Q1/68; C12R1/91; (IPC1-7): C07K13/00; C12N15/00; C12P21/02; A61K37/02
Other References:
MOLECULAR AND CELLULAR BIOLOGY, Volume 13, Number 5, issued May 1993, JIANG et al., "Cloning and Characterization of R-PTPk, a New Member of the Receptor Protein Tyrosine Phosphatase Family with a Proteolytically Cleaved Cellular Adhesion Molecule-Like Extracellular Region", pages 2942-2951.
THE EMBO JOURNAL, Volume 9, Number 10, issued 1990, KRUEGER et al., "Structural Diversity and Evolution of Human Receptor-Like Protein Tyrosine Phosphatase", pages 3241-3252.
FEDERATION OF EUROPEAN BIOLOGICAL SOCIETIES, Volume 290, Number 1,2, issued September 1991, GEBBINK et al., "Cloning, Expression and Chromosomal Localization of a New Putative Receptor-Like Protein Tyrosine Phosphatase", pages 123-130.
SCIENCE, Volume 253, issued 26 July 1991, FISCHER et al., "Protein Tyrosine Phosphatase: A Diverse Family of Intracellular and Transmembrane Enzymes", pages 401-406.
PROCEEDINGS OF NATIONAL ACADEMY OF SCIENCE USA, Volume 80, issued September 1983, HOFFMAN et al., "Kinetics of Homophilic Binding by Embryonic and Adult Forms of the Neural Cell Adhesion Molecule", pages 5762-5766.
Download PDF:
Claims:
WHAT IS CLAIMED IS;
1. An isolated mammalian receptortype protein tyrosine phosphataεe K protein or glycoprotein molecule.
2. The molecule of Claim 1 wherein the molecule compriεeε the amino acid εequence SEQ ID N0:1, εhown in FIG. 3.
3. The molecule of Claim 1 wherein the molecule iε a human receptortype protein tyroεine phosphatase K .
4. The molecule of Claim 3 wherein the molecule comprises the amino acid sequence SEQ ID NO:2, shown in FIG. 15AE.
5. An isolated nucleic acid molecule comprising a nucleotide sequence encoding the receptortype protein tyrosine phoεphataεe K protein of Claim 1.
6. The nucleic acid of Claim 5 wherein the nucleotide εequence encoding the receptortype protein tyroεine phoεphataεe K protein iε the nucleotide εequence SEQ ID No:3, εhown in FIG. 1AH.
7. An iεolated nucleic acid molecule comprising a nucleotide sequence encoding the human receptortype protein tyrosine phosphatase K protein of Claim 3.
8. The nucleic acid molecule of Claim 7 wherein the nucleotide sequence encoding the human receptor type protein tyroεine phoεphatase K is the nucleotide sequence SEQ ID NO:4, shown in FIG. 15AE.
9. The nucleic acid molecule of Claim 5 or 7 wherein the nucleic acid molecule is cDNA.
10. The nucleic acid molecule of Claim 5 or 7 wherein the nucleic acid iε genomic DNA.
11. The nucleic acid molecule of Claim 5 or 7 wherein the nucleic acid molecule iε an expression vehicle.
12. The nucleic acid molecule of Claim 11 wherein the expression vehicle is a plasmid.
13. A prokaryotic host cell having the plasmid of Claim 12.
14. A eukaryotic host cell having the plasmid of Claim 12.
15. A method for preparing the receptortype protein tyrosine phosphataεe K protein or glycoprotein of Claim 1, compriεing: (a) culturing a hoεt cell capable of expreεεing the protein or glycoprotein under culturing conditions that permit expression of the protein or glycoprotein; and (b) recovering the protein or glycoprotein from the culture.
16. The method of Claim 15 wherein the host cell is a prokaryotic cell.
17. The method of Claim 15 wherein the host cell is a eukaryotic cell.
18. An antibody specific for the protein or glycoprotein of Claim 1.
19. The antibody of Claim 18 wherein the antibody is a monoclonal antibody.
20. A method for detecting the presence of the nucleic acid molecule of Claim 5 in a cell, comprising: (a) contacting the cell with a nucleic acid probe capable of specifically hybridizing to least a portion of the nucleic acid molecule of Claim 5 under hybridizing conditions; and (b) measuring the hybridization of the probe to the nucleic acid of the cell, thereby detecting the presence of the nucleic acid sequence.
21. A method for detecting the presence of the nucleic acid molecule of Claim 5 in a cell, comprising: (a) contacting the cell with nucleic acid primers capable of specifically binding to at least a portion of the nucleic acid molecule of Claim 5 under hybridizing conditions; (b) selectively amplifying at least a portion of the nucleic acid molecule of Claim 5; and (c) detecting the amplified nucleic acid of the cell, thereby detecting the presence of the nucleic acid sequence.
22. A method for detecting in a cell the presence of or meaεuring the quantity of a receptor protein tyrosine phosphatase K protein or glycoprotein, comprising: (a) contacting the cell or an extract thereof with the antibody of Claim 18; and (b) detecting binding of the antibody to the cell or extract thereof, or measuring the quantity of antibody bound, thereby determining the presence of or measuring the quantity of the receptor protein tyrosine phosphataseic protein or glycoprotein.
23. A method for identifying in a chemical or biological preparation a compound capable of binding to the receptor protein tyrosine phosphatase K protein, glycoprotein or derivative of Claim 1, comprising: (a) attaching the receptor protein tyrosine phosphatase K protein, glycoprotein or derivative, or the ligandbinding portion thereof, to a solid phase matrix; (b) contacting the chemical or biological preparation with the solid phase matrix allowing the compound to bind; (c) washing away any unbound material; and (d) detecting the presence of the compound bound to the solid phase matrix.
24. A method for isolating from a complex mixture a compound capable of binding to the receptor protein tyrosine phosphataεe K protein or glycoprotein of Claim 1, comprising: (a) attaching the receptor protein tyroεine phoεphataεe , or the ligandbinding portion thereof, to a solid phase matrix; (b) contacting the complex mixture with the solid phase matrix allowing the compound to bind; (c) washing away any unbound material; and (d) eluting the bound compound, thereby isolating the compound.
25. A method for identifying an agent capable of modulating the enzymatic activity of receptor protein tyrosine phosphatase K, comprising: (a) contacting the agent with a receptor protein tyrosine phosphatase K in pure form, in a membrane preparation, or in a whole cell; (b) incubating the mixture of step (a) for a sufficient interval; (c) measuring the enzymatic activity of the receptor protein tyrosine phosphataεe*; and (d) comparing the enzymatic activity to that of receptor protein tyroεine phoεphataεeκ incubated without the agent, thereby determining whether the agent modulates enzymatic activity.
26. The method of Claim 25 wherein the agent is capable of decreasing the activity of the receptor protein.
27. The method of Claim 25 wherein the agent is capable of inhibiting the activity of the receptor protein.
28. A method for inhibiting homophilic binding of endogenous Type II receptor protein tyrosine phosphatase molecules in a mammal comprising administering to the mammal an effective amount of an inhibitory compound so that the homophilic binding is inhibited.
29. The method of Claim 28 wherein the homophilic binding is intercellular.
30. The method of Claim 28 wherein the inhibitory compound comprises an extracellular domain of a Type II receptor protein tyrosine phoεphataεe molecule.
31. The method of Claim 28 wherein inhibiting of homophilic binding modulates the endogenous enzymatic activity of the Type II receptor protein tyrosine phosphatase moleculeε.
32. The method of Claim 31 wherein the endogenous enzymatic activity of the Type II receptor protein tyrosine phosphataεe molecules is increased.
33. The method of Claim 31 wherein the endogenouε enzymatic activity of the Type II receptor protein tyrosine phosphatase molecules is decreased.
34. The method of Claim 28 or 31 wherein inhibiting homophilic binding of endogenous Type II receptor protein tyrosine phosphatase moleculeε regulates a cellular function comprising differentiation or cell cycle control.
35. The method of Claim 28 or 31 wherein inhibiting homophilic binding of endogenouε Type II receptor protein tyroεine phoεphataεe molecules regulates cellular behavior comprising motility, contact inhibition, cell adhesion, or signal transduction.
36. The method of Claim 28 or 31 wherein inhibiting homophilic binding of endogenous Type II receptor protein tyrosine phosphataεe moleculeε regulates an abnormal or deleteriouε process comprising cellular tranεformation to a cancerous state.
37. A method for inhibiting homophilic binding of endogenous receptortype protein tyrosine phosphatase K molecules compriεing adminiεtering to the mammal an effective amount of an inhibitory compound so that the homophilic binding is inhibited.
38. The method of Claim 37 wherein the homophilic binding iε intercellular.
39. The method of Claim 37 wherein the inhibitory compound comprises a soluble receptortype protein tyrosine phosphatase K protein extracellular domain.
40. The method of Claim 37 wherein inhibiting homophilic binding of endogenous receptortype protein tyrosine phoεphataεe K protein modulateε endogenous receptortype protein tyroεine phoεphataεe K enzymatic activity.
41. The method of Claim 40 wherein the endogenous enzymatic activity of the receptortype protein tyrosine phosphatase K protein is decreased.
42. The method of Claim 40 wherein the endogenous enzymatic activity of the receptortype protein tyrosine phosphatase K protein is increased.
43. The method of Claim 37 or 40 wherein inhibiting homophilic binding of endogenous receptor type protein tyrosine phosphatase K molecules regulates a cellular function comprising differentiation or cell cycle control.
44. The method of Claim 37 or 40 wherein inhibiting homophilic binding of endogenous receptor type protein tyrosine phosphatase K molecules regulates a cellular behavior comprising motility, contact inhibition, cell adhesion, or signal transduction.
45. The method of Claim 37 or 40 wherein inhibiting homophilic binding of endogenous receptor type protein tyroεine phosphatase K molecules regulates an abnormal or deleteriouε process comprising cellular transformation to a cancerous state.
46. A method for identifying a compound capable of inhibiting homophilic binding of Type II receptor type protein tyroεine phoεphataεe molecules comprising: (a) exposing an immobilized molecule comprising a Type II receptortype protein tyrosine phosphataεe protein extracellular domain to a Type II receptor type protein tyroεine phoεphataεe protein molecule and a compound; (b) removing all moleculeε that do not attach to the immobilized molecule; and (c) determining if the Type II receptortype protein tyrosine phosphataεe protein molecule bindε the immobilized molecule.
47. A method for identifying a compound capable of inhibiting homophilic binding of Type II receptor type protein tyrosine phosphatase molecules comprising exposing Type II receptortype protein tyrosine phosphataseexpressing cells to a compound and determining if the Type II receptortype protein tyrosine phosphataseexpressing cells aggregate.
48. The method of Claim 46 or 47 wherein the Type II receptortype protein tyrosine phosphatase molecule is receptortype protein tyrosine phosphatase K .
Description:
NOVEL RECEPTOR-TYPE PHOSPHOTYROSINE PHOSPHATASE-KAPPA

1. INTRODUCTION

The invention in the field of biochemistry and cell and molecular biology relates to novel receptor- type protein tyrosine phosphataεe protein or glycoprotein, termed RPTPK (also known as RPTPase-ic) , DNA coding therefor, methods for production and identification of the protein, methods for screening compounds capable of binding to and inhibiting or stimulating PTPase enzymatic activity, methods for inhibiting homophilic binding of RPTPK, and methods for identifying compounds which are capable of inhibiting homophilic RPTPK binding.

2. BACKGROUND OF THE INVENTION

Tyrosine phosphorylation of proteins is involved in an increasing number of cellular signalling events. It was originally implicated in signalling by paracrine- or autocrine-acting growth factors, and endocrine hormones such as insulin (see Yarden, Y. et al . , Annu. Rev. Biochem. 57:443-478 (1988) for review) . It is now clear that this posttranslational modification is also involved in diverse processes such as the activation of cells of the immune system by antigens (Klausner, R.D. et al . , Cell 64:875-878), signalling by ly phokines (Hatakeyama, M. et al. , 1991 Science 252:1523-1528 (1991); Mills, G.B. et al. , J. Biol. Chem. 265:3561-3567 (1990)), and cellular differentiation and survival (Fu, X.-Y. 1992 Cell 70:323-335; Schlessinger, J. et al. 1992 Neuron 9:1- 20; Velazquez, L. et al. , 1992 Cell 70:313-322). In view of the diversity of processes in which tyrosine

phosphorylation is involved, it is not surprising that links are also emerging with the process of cell adhesion and cell-cell contact. The identification of several growth factor receptors and retroviral oncogenes as tyrosine- specific protein kinases indicated that protein phosphorylation on tyrosine residues plays a key role in cellular growth control. This notion has recently received support by the observation that the level of tyrosine phosphorylation of enzymes thought to play an important role in signal transduction (such as phospholipase C) correlates with their increased activity upon growth factor stimulation, thus establishing a functional role for tyrosine phosphorylation (Ullrich, A., et al., Cell 51:203-212 (1990)) .

Most of the processes in which tyrosine phosphorylation is implicated involve the transduction of a signal through the cell membrane. In its best understood fashion, this can occur through dimerization-mediated activation of members of the receptor tyrosine kinase family by soluble ligands (reviewed in Ullrich, A. et al . 1990 Cell 61:203- 212) . However, modulation of receptor tyrosine kinase activity can also occur by membrane-bound ligands on neighboring cells, as in the case of the interaction between the εevenless kinase and the Jbride of sevenless protein (Rubin, G.M. 1991, Trends in Genetics 7:372-376). Recently, receptor-like tyrosine kinases were described with an extracellular domain similar to that of cell adhesion molecules of the CAM- family (e.g. Axl and Ark (O'Bryan, J.P. et al . , 1991 Mol. Cell. Biol. 11:5016-5031; Rescigno, J. et al. , 1991 Oncogene 6:1909-1913)). Such observations may implicate tyrosine phosphorylation as a more broadly

used direct downstream effector mechanism for precise cell-cell recognition and signalling events. Members

« of the non-receptor family of tyrosine kinases have i also in several instances been shown to be associated with other proteins with a trans-membrane topology, examples being the association of the Lck and Fyn kinases with the CD4 protein and T-cell receptor complex components respectively (Haughn, L. et al.,

10 1992 Nature 358:328-331; Samelson, L.E. et al., 1992 Proc. Natl . Acad. Sex . USA 87:4358-4362; Veillette, A. et al., 1988 Cell 55:301-308). However, the mechanism by which kinase activity is modulated in these instances is not understood.

15 The degree and pattern of phosphorylation of tyrosine residues on cellular proteins are regulated by the opposing activities of protein-tyrosine kinases (PTKases; ATP:protein-tyrosine O-phosphotransferase, EC 2.7.1.112) and protein-tyrosine-phosphatases

2o (PTPases; protein-tyrosine-phosphate phosphohydrolase, EC 3.1.3.48). The structural characteristics and evolution of PTKases as well as their role in the regulation of cell growth have been reviewed (Hunter, T. , et al., Annu . Rev. Bioche . 54:897-930 (1985);

25 Ullrich, A., et al . , supra) .

2.1. PTKases

Tyrosine kinases comprise a discrete family of enzymes having common ancestry with, but major

30 differences from, serine/threonine-specific protein kinases (Hanks, S.K. et al . , (1988) Science 241:42- 52) . The mechanisms leading to changes in activity of tyrosine kinases are best understood for receptor-type tyrosine kinases which have a transmembrane topology

35 (Ullrich, A. et al . , supra ) . With such kinases, the binding of specific ligands to the extracellular

domain of these enzymes is thought to induce their oligomerization leading to an increase in tyrosine kinase activity and activation of the signal transduction pathways (Ullrich, A. et al., supra) . The importance of this activity is supported by the knowledge that dysregulation of kinase activity through mutation or over-expression is a mechanism for oncogenic transformation (Hunter, T. et al . , supra ; Ullrich, A. et a . , 1990, supra) .

2.2. PTPases

The protein phosphatases are composed of at least two separate and distinct families (Hunter, T. Cell,. 58:1013-1016 (1989)), the protein serine/threonine phosphatases and the protein tyrosine phosphatases. This is in contrast to protein kinases, which show clear sequence similarity between serine/threonine- specific and tyrosine-specific enzymes.

There appear to be two basic types of PTPase molecules. The first group is comprised of small, soluble enzymes that contain a single conserved phosphatase catalytic domain, and include (1) placental PTPase IB (Charbonneau, H. et al. , Proc .

Natl . Acad. Sex . 86:5252-5256 (1989); Chernoff, J. et al . , Proc. Natl . Acad. Sex . USA 87:2735-2789 (1990)), (2) T-cell PTPase (Cool, D.E. et al. , Proc. Natl . Acad. Sex . USA 86:5257- 5261 (1989)), and (3) rat brain PTPase (Guan, K. , et al . , Proc. Natl . Acad. Sex . USA, 87:1501- 1505 (1990).

The identification of a tyrosine phosphatase homology domain has raised new interest in the potential of PTPases to act as modulators of tyrosine phosphorylation (Kaplan, R. et al . 1990 Proc . Natl . Acad . Sex . USA 87:7000-7004; Krueger, N.X. et al . ,

1990 EMBO J. 9:3241-3252; see, for review, Fischer, E.H. et al . , 1991 Science 253:401-406).

The second group of PTPases is made up of the more complex, receptor-linked PTPases, termed R-

PTPases or RPTPs, which are of high molecular weight and contain two tandemly repeated conserved domains separated by 56-57 amino acids. RPTPs may be further subdivided into four types based on structural motifs within their extracellular segments.

One example of RPTPs are the leukocyte common antigens (LCA) (Ralph, S.J., EMBO J . , 6:1251-1257 (1987); Charbonneau, H. , et al . , Proc . Natl . Acad . Sex . USA, 85:7182-7186 (1988)). LCA, also known as CD45, T200 and Ly-5 (reviewed in Thomas, M.L. , Ann. Rev. _r.nunu.nol. 7:339-369 (1989)) comprises a group of membrane glycoproteins expressed exclusively in he opoietic (except late erythroid) cells, derived from a common gene by alternative splicing events involving the amino terminus of the proteins.

Other examples of RPTPs are the LCA-related protein, LAR (Streuli, M. et al . , J. Exp. Med . , 168:1523-1530 (1988)), and the LAR-related Drosophila proteins DLAR and DPTP (Streuli, M. , et al . , Proc . Natl. Acad. Sex . USA, 86:8698-8702 (1989)). Jirik et al . screened a cDNA library derived from the human hepatoblastoma cell line, HepG2, with a probe encoding the two PTPase domains of LCA (FASEB J. 4:A2082 (1990) , abstr. 2253) and discovered a cDNA clone encoding a new RPTP, named He-PTP. The HePTP gene appeared to be expressed in a variety of human and murine cell lines and tissues.

A large number of members of the RPTP family, called type II RPTPs, display an extracellular domain containing a combination of Ig-domainε and fibronectin type III repeats (Fn-III) , features typically

encountered in cell adhesion molecules (CAMs) (Gebbink, M.F.B.G. et al., 1991 FEBS Lett. 290:123- 130; Streuli, M. et al . , 1988 J. Exp. Med. 168: 1523-1530) . An analysis of the expression patten of several R-PTPases in the developing Drosophxla CNS suggests some function of these molecules in aspects of axon guidance and outgrowth (Tian, S.S. et al., 1991 Cell 67:675-685; Yang, X. et a . , 1991. Cell 67:661-673), an observation winch might be related to the ability of R-PTPases to control the activity of src-family tyrosine kinases (Mustelin, T. et a . , 1989 Proc.Natl.Acad.Sci.USA 86:6302-6306; Ostergaard, H.L. et al., 1989 Proc. Natl. Acad. Sci. USA 86:8959- 8963; Zheng, X.M. et al . , 1992 Nature 359:336-339).

Other studies have raised the possibility that certain R-PTPases may function as tumor suppressor genes, e.g. by controlling contact inhibition (LaForgia, S. et al., 1991 Proc . Natl . Acad . Sex . USA 88:5036-5040). Elevation of cellular phosphotyrosine may occur through mechanisms other than the activation of a tyrosine kinase itself. For instance, expression of the v-crk oncogene, though not a tyrosine kinase, induces the phosphorylation of tyrosine residues through a poorly understood mechanism (Mayer, B.J. et al . (1988) Nature 332 , 272-275). Potentially, such an outcome could result from either mutation of the substrate or through a general decrease in cellular phosphatase activity, especially in view of the normally high turnover rate of cellular tyrosine- phosphate (Sefton, B.M. et al . (1980) Cell 20:807- 816) . The latter possibility is suggested by the demonstration that tyrosine phosphatase inhibitors can ••reversibly transform" cells (Klarlund, J.K. Cell 41 : 707-717 (1985)). PTPases could therefor act as recessive oncogenes.

While we are beginning to understand more about the structure and diversity of the PTPases, much remains to be learned about their cellular functions. Thus, a better understanding of, and an ability to control, phosphotyrosine metabolism, requires knowledge not only the role of PTKase activity, but the action of PTPase enzymes as well. It is clear in the art that further delineation of structure-function relationships among these PTPases and RPTP membrane receptors are needed to gain important understanding of the mechanisms of cell growth, differentiation, and oncogenesis.

SUMMARY OF THE INVENTION

The present inventors have conceived of a role for RPTPs in cellular control mechanisms, both as potential anti-oncogenes, and as effectors in a newly discovered mechanism of transmembrane signalling. They therefore undertook a search for individual RPTP genes and proteins in mammals, including humans, which are potentially involved in such processes, and describe herein the identification of a novel, widely expressed member of the RPTP family, RPTPK, in both mice and in humans which has a transmembrane topology. The novel human RPTP disclosed herein consists of two associated subunits whose expression is modulated by cell-to-cell contact, and, in a manner analogous to receptor tyrosine kinases, is subject to direct regulation by extracellular ligands which bind to the extracellular portion. Further, as is demonstrated in the Working Example presented in Section 15, infra . RPTPK is shown to homophilically bind other RPTP* molecules.

The present invention thus provides a mammalian, preferably a human, receptor-type protein tyrosine phosphatase- K (RPTP/) protein or glycoprotein molecule, a functional derivative of the RPTP/c, or a homolog of the RPTP/c in another mammalian species. When the RPTP/c molecule is of natural origin, it is substantially free of other proteins or glycoproteins with which it is natively associated. RPTP/c is naturally expressed in mammalian brain and is developmentally and anatomically regulated. It is also expressed in other mammalian tissues. The RPTPc molecule of the present invention may also be prepared by chemical synthesis or by or recombinant means. Thus, the substantially pure RPTP/c protein or glycoprotein of the present invention may be produced by biochemical purification of the protein or glycoprotein of natural origin or by production using chemical synthesis or by recombinant expression in prokaryotic or eukaryotic hosts.

In particular, the invention is directed to a mammalian RPTP/c protein or glycoprotein having the amino acid sequence of RPTP/c shown in FIG. 3 (SEQ ID NO:l). In another embodiment is provided a functional derivative thereof. Preferably, the RPTP/c is of human origin, and has the amino acid sequence SEQ ID NO:2, as shown in FIG. 15A-15E.

The invention is further directed to a nucleic acid molecule, preferably DNA, which may consist essentially of a nucleotide sequence encoding a mammalian RPTP/c having the nucleotide sequence SEQ ID NO:3 (FIG. 1A-1H) . Preferably, the nucleic acid molecule consists essentially of a nucleotide sequence encoding human RPTP/c and having the nucleotide sequence SEQ ID NO:4 or encodes a funcitonal derivative thereof. The DNA molecule is

preferably cDNA or genomic DNA. The invention is further directed to the DNA molecule in the form of an expression vehicle, as well as prokaryotic and eukaryotic hosts transformed or transfected with the DNA molecule.

Also included in the present invention is a process for preparing a RPTPK protein or glycoprotein, or a functional derivative thereof, comprising: (a) culturing a host capable of expressing the protein, glycoprotein or functional derivative under culturing conditions, (b) expressing the protein, glycoprotein or functional derivative; and (c) recovering the protein, glycoprotein or functional derivative from the culture.

This invention is also directed to an antibody, either polyclonal, monoclonal, or chimeric, which is specific for the RPTPK protein or glycoprotein. This invention is also directed to a method for detecting the presence of nucleic acid encoding a normal or mutant RPTPK in a cell or in a subject, com¬ prising:

(a) contacting a cell or an extract thereof from the subject with an oligonucleotide probe encoding at least a portion of a normal or mutant RPTPK under hybridizing conditions; and

(b) measuring the hybridization of the probe to the nucleic acid of the cell, thereby detecting the presence of the nucleic acid, preferably DNA. The DNA can be selectively amplified, using the polymeraεe chain reaction, prior to assay.

The invention is further directed to a method for detecting the presence, or measuring the quantity of RPTPK in a cell or cells, comprising:

(a) contacting said cell or an extract thereof with an antibody specific for an epitope of RPTPK; and

(b) detecting the binding of the antibody to the cell or extract thereof, or measuring the quantity of antibody bound, thereby detecting the presence or measuring the quantity of the RPTPK.

The present invention is also directed to methods for identifying and isolating a compound capable of binding to RPTPK from a chemical or biological preparation comprising: (a) attaching RPTPK, or the ligand-binding portion thereof, to a solid phase matrix; (b) contacting the chemical or biological preparation with the solid phase matrix allowing the compound to bind, and washing away any unbound material;

(c) detecting the presence of the compound bound to the solid phase matrix; and, for purposes of isolation,

(d) eluting the bound compound, thereby isolating the compound.

Further, the present invention includes a method for identifying an agent capable of stimulating or inhibiting the phosphatase enzymatic activity of RPTPK, comprising:

(a) contacting the agent with the RPTPK in pure form, in a membrane preparation, or in a whole live or fixed cell; (b) incubating the mixture in step (a) for a sufficient interval;

(c) measuring the enzymatic activity of the RPTPK;

(d) comparing the enzymatic activity to that of the RPTPK incubated without the agent, thereby determining whether the agent stimulates or inhibits the enzymatic activity.

Still further, the invention provides methods for inhibiting the homophilic binding of Type II RPTP, preferably the homophilic binding of RPTPK, provides methods for identifying agents capable of inhibiting such Type II RPTP homophilic binding, and methods for inhibiting endogenous Type II RPTP homophilic binding in mammalian subjects.

4. DESCRIPTION OF THE FIGURES

FIG. IA - 1H shows the complete nucleotide sequence and amino acid sequence of murine RPTPK. The signal peptide, A5 homology region, transmembrane domain, and PTPase domains are designated by brackets.

FIG. 2 is a schematic representation of the various RPTPK cDNA clones isolated, and the proposed domain structure of the RPTPK protein. Translational start and stop codons as well as restriction sites mentioned in the text are indicated. The vertical arrow indicates the position of the furin cleavage site. TM: transmembrane segment.

FIG. 3 shows the predicted amino acid sequence of the RPTPK precursor protein. The putative signal peptide and transmembrane (TM) segment are underlined. The two tandem phosphatase domains are boxed (PTP- l,PTP-2) . The proteolytic cleavage site (RTKR 640- 643) is printed in bold, and the Ig-like domain (Ig, 214-270) shown in bold italic characters. A5: homology to A5 surface protein (Takagi, S. et al . , 1991 Neuron 7:295-307); FN-III: fibronectin type III repeats. The Genbank accession number for the cDNA sequence is L10106. FIG. 4 shows a proposed alignment of the four FN- III repeats of RPTPK and domain 7 of human fibronectin

(Kornblihtt, A.R. et a . , 1985 EMBO J. 4:1755-1759). Residues most typically conserved in FN-III repeats are highlighted in bold. Residues identical in three or more out of the five aligned sequences are indicated with an asterisk. This region of the protein also contains clearly detectable homology to LAR, Drosophxla PTPase 10D, and Drosophxla neuroglian, all of which have been reported to contain FN-III repeats.

FIG. 5 shows an alignment of the N-terminal domains of RPTPK and mRPTPμ with the cell surface protein A5 (Takagi et al . , supra) . Numbers indicate the first residue of the respective proteins shown in the alignment. Residues marked as consensus are identical between A5 and RPTPK, or between A5 and mRPTPμ. Conservative substitutions are present but not shown. Residues in bold (C,W) define a possible Ig-like domain structure. FIG. 6 shows the expression of RPTPK mRNA in adult tissues using Northern blot analysis of poly(A)+RNA from various mouse tissues. The entire cDNA fragment from clone λ-604 was used as a probe. A similar pattern of hybridization was seen using as a probe the λ-50 cDNA clone and the N-terminal half of the λ-35 cDNA clone. Positions of RNA molecular weight markers, in kb, are indicated on the left side.

FIG. 7 is a gel pattern showing the immunoprecipitation of the RPTPK protein. HeLa cells transiently transfected by the calcium phosphate technique with an RPTP-ic expression vector (+) or an empty expression vector (-) were analyzed by radio- i munoprecipitation using antiserum 116 directed against a synthetic peptide corresponding to residues 60 to 76 in the extracellular domain. The

immunoprecipitation was performed in the absence (-) or presence (+) of 20 μg of the immunogenic peptide (α-κ: anti RPTPK antiserum 116; pre: corresponding preimmune serum) . Positions of protein molecular weight standards (expressed in kDa) are indicated on the left side of the autoradiogram.

FIG. 8 shows the protein tyrosine phosphatase activity of anti-RPTPκ immunoprecipitates. The RPTPK protein was immunoprecipitated from transiently transfected COS cells using anti-N-terminal antibody 116 or corresponding preimmune serum. The PTPase activity in the immune complexes was analyzed in the absence (-) or presence (+) of vanadate. The amount of radioactivity released as inorganic phosphate is expressed as the percentage of the total input radioactivity. A representative of several experiments is shown.

FIG. 9 shows RPTPK immunoreactive species in COS cells, and effect of Endo F treatment on SDS-PAGE mobility. Total lysates from mock or RPTPK transfected COS cells were treated or not with Endo F. The lysates were resolved by SDS-PAGE and immunoblotted with anti-N-terminal antibody 116 (left panel) or anti-cytoplasmic antibody 122 (right panel) . The 95 kDa band in panel B also seen in mock- transfected cells is presumably due to fortuitous reactivity of antiserum 122 and not relevant to the analysis. No such protein species was detectable using an antiserum raised against the same antigen in a different rabbit.

FIG. 10. Pulse-chase analysis of RPTPK processing. Mock-transfected cells (lanes 1-2) and cells transfected with a wild type RPTPK expression vector (In. 3-6) were metabolically labeled with [ 35 S]- methionine (200 [μCi/ml) for 15 minutes ("pulse") and

chased for the time-periods indicated. Immunoprecipitation was performed using antiserum 116. Arrows indicate the positions of the 210 kDa RPTPK precursor and the 110 kDa N-terminal cleavage product. FIG. 11 shows the effect of mutagenesis of the furin cleavage motif RTKR on RPTPK processing. Total lysates from mock-transfected COS cells, cells expressing wt RPTPK, or RPTP(κ) carrying a mutation in the furin cleavage motif RTKR (CM K) were resolved by SDS-PAGE. Immunoblotting was performed using anti-N- terminal antiserum 116 (left panel) , or anti- cytoplasmic antiserum 122 (right panel) .

FIG. 12 shows the co-immunoprecipitation of the RPTPK processing products. Total lysate from mock or wild type RPTPK transfected COS cells was subjected to immunoprecipitation using anti-N-terminal antiserum 116, and the precipitate inununoblotted with anti- cytoplasmic antiserum 122. As a control, total lysate from RPTPK transfected cells was loaded in the right lane on the immunoblot.

FIG. 13A-B are a series of micrographs showing the in situ hybridization analysis of RPTPK expression during development and in the adult CNS. 13A panel shows localization of RPTPK mRNA in the rat at embryonic day 18. CTX, cerebral cortex; SC, spinal cord; L, liver; K, kidney; I, intestine. 13B panel shows localization of RPTPK mRNA in a sagittal section of rat brain at postnatal day 6. CTX, cerebral cortex; CB, cerebellum; DG, dentate gyrus. In the cerebral cortex, particularly in the occipital region, the labeling is not uniform in all the cortical cell layers. In the hippocampal formation labeling is more intense in the dentate gyrus and in CA3. In the cerebellum, the most intense labeling is seen in the external granular cell layer.

FIG. 14 is a molecular model describing the processing of the R-PTP-K precursor protein. A furin- like endoprotease cleaves the 210 kDa precursor protein, after which both cleavage products (110 and 100 kDa) remain associated. No suggestions as to the mechanism of association are intended. The numerals 116 and 122 designate the sites of epitopes recognized by antisera described in the text. FIG. 15A-15E shows the nucleotide sequence of the human RPTPK (SEQ ID NO:4), designated MCP7, and its derived amino acid sequence (SEQ ID NO:2).

FIG. 16A-B. A comparison of the amino acid sequence or RPTPKK (MCP7 sequence) to the amino acid sequence of hRPTPμ. Lack of designation of an amino acid in hRPTPμ indicates identity to the MCP7 sequence. The putative signal peptide and transmembrane region are overlined/asterisked, the cleavage site is underlined, the FN-III repeats are overbracketed, the Ig domain is overhatched, and the A5 and two PTPase domains are boxed.

FIG. 17 shows a Northern blot analysis of MCP7 mRNA from human tissues. Poly(A)+RNA (4μg per lane) prepared from the indicated tissues was probed with a 32 p-labeled fragment corresponding to the extracellular domain of MCP7. The blots were applied for a 5 day exposure using an intensifying screen.

FIG. 18 shows a Northern blot analysis of MCP7 mRNA from several different human breast cancer cell lines. Poly(A)+RNA (4μg per lane) prepared from the indicated cell line was probed as in FIG. 15 and the blots similarly exposed.

FIG. 19A-B show gel patterns indicating the transient expression of MCP7 mRNA in transfected cells. Cells of the 293 line were transfected with a MCP7 expression vector (or an empty vector as a

control) , metabolically labeled for 24 hours with [ 35 S] methionine and incubated with an anti-N-terminal antiserum 116. Cells were washed, lysed and protein- antibody complexes were removed by protein-A sepharose. 19A panel shows a SDS-PAGE gel of immunoprecipitates. 19B panel shows Western blots of SDS-PAGE gels of lysates of cells transfected by MCP7- CMV (lane 1) or "empty" CMV (lane 2) and immunoblotted with the anti-N-terminal antiserum 116.

FIG. 20A-B shows Western blot patterns indicating co-expression of MCP7 with different RTKs. Semiconfluent 293 cells were transfected with expression plasmids encoding the indicated RTK together with either an equal amount of MCP7 expression vector or a control plasmid. After stimulation with the appropriate ligand: stem cell factor (SCF) for the pl45 c"ki* RTK; epidermal growth factor for all other RTKs; insulin for I-R, cells were lysed, aliquots run on SDS-PAGE and transferred to nitrocellulose. Proteins were immunoblotted with anti-phosphotyrosine antibody 5E.2. Molecular mass markers are indicated.

FIG. 21A-B shows Northern blots indicating the relationship between MCP7 mRNA levels and the state of cell confluence in SK-BR-3 cells (21A) and HT-29 cells (21B) in culture. Poly(A)+ RNA (4 μg per lane) was prepared from cells obtained at different levels of confluence (lanes 1 and 4:40%; lanes 2 and 5: 70%, lane 3 and 6: 100%) and was probed with a 32 P-labeled DNA probe corresponding to the extracellular domain of MCP7 (upper blots) and with a fragment coding for GAPDH (lower blots) .

FIG. 22A. Expression of the R-PTPκ protein in transfected S2 cells. Detergent lysates were prepared from transfected cells, resolved by SDS-PAGE, and

immunoblotted with an antiserum directed against the extracellular domain of the R-PTPκ protein (Y.-P. Jiang et al . Mol. Cell. Biol. 13, 2942 (1993)). Lanes: 1, R-PTPκ anti-sense transfected cells, not heat-shocked; 2, anti-sense transfected after heat- shock; 3, sense transfected cells, not heat-shocked; 4, sense-transfected cells after heat-shock; 5, lysate from COS cells transiently transfected with an R-PTPκ expression vector (Y.-P. Jiang et al . Mol. Cell. Biol. 13, 2942 (1993)). Molecular weight standards are indicated in kilodaltons.

The entire RPTPK CDNA was introduced in both orientations as a Hpal/EcoRV fragment into the Hpal site of a derivative of the pCasper expression vector containing the hsp70 promoter, and the resulting construct co-transfected with the pPC4 plasmid (conferring α-amanitin resistance) into S2 cells using calcium phosphate precipitation. Pools of stably transfected cells were selected in the presence of

5μg/ml α-amanitin for three weeks. Transfected cells were heat-shocked at 37°C for 30 minutes and allowed to recover for 2 hours. Adherent cells were collected, and washed twice in BSS (Kramer, H. et al., 1991, Nature 352:207; Snow, P. et al., 1989, Cell 59:313) . FIG. 22B-C. Photographs of transfected cell populations after heat-shock induction and aggregation for 2 hours. 22B panel, control (anti-sense transfected) cells; 22C panel, cells transfected with an expression vector carrying the R-PTPκ cDNA in the sense orientation; insert: higher magnification of a typical aggregate.

FIG. 22D. Time-course and quantitation of aggregation by Coulter-counting of above-threshold particles. Open squares: anti-sense vector transfected cells, uninduced; full squares, idem,

induced; open circles, cells transfected with an expression vector containing the R-PTPK cDNA in the sense orientation, uninduced; full circles, sense, induced. Standard errors are indicated by error bars. Adherent, transfected cells were collected, washed twice with BSS, resuspended in BSS at a concentration of 4xl0 6 cells/ml, and incubated in Coulter-Counter vials on a rotary shaker for 2 hours at 100 rpm at room temperature. For each time-point, 1 ml was counted using the Coulter-counter with the following settings: 1/amplification=4; threshold=10; 1/aperture current=32.

FIG. 22E. Effect of deletion of the intracellular domain of the R-PTPκ protein, and mutation of the furin cleavage site. Parental S2 cells were transiently transfected with expression vectors encoding an R-PTPκ cDNA in which the furin cleavage site had been mutated (CM) (Y.-P. Jiang et a-i« Mol. Cell. Biol. 13, 2942 (1993)), a cDNA encoding a catalytically inactive deletion mutant of R-PTPκ lacking most of the intracellular (PTPase) domain (Δ- PTP) , or a wt R-PTPκ cDNA (wt) . For the deletion mutant, a cDNA encoding a truncated, catalytically inactive form (ΔK) of RPTPK was constructed by restriction digestion with BspEI and Klenow till-in of the wild type cDNA. This leads to the introduction of a stop codon after amino acid residue 1083, and the generation of a protein lacking the cysteine residues essential for catalysis in the two intracellular catalytic homology domains of RPTPK. Cells were heat- induced 72 hours after transfection, subjected to aggregating conditions for 2 hours, and above- threshold aggregates counted with a Coulter-counter. Error bars indicate standard errors. Transfected, but non heat-shock induced cells behaved as untransfected

parental cells. The apparent differences in aggregation intensity between the different forms of R-PTPK may reflect protein expression levels. The numbers provided by Coulter-counter counting actually provide an underestimation of the amount of aggregation as determined by visual inspection and counting of aggregates, since only large particles above a certain threshold size are scored by the Coulter-Counter.

FIG. 23A-C. Aggregates consist solely of cells expressing the R-PTPκ protein. Two different cell populations, one of which had been labeled with the fluorescent dye dil (J. Schlessinger et al . Science 195. 307 (1977) , were allowed to co-aggregate and the resulting aggregates inspected by visible and fluorescence microscopy, dil-fluorescence is white in the photographs. 23A: a pool of R-PTPκ expressing cells was allowed to aggregate in the presence of an equal number of dil- stained R-PTPκ-negative cells.

23B: R-PTPK expressing cells were stained with dil and allowed to aggregate in the presence of unstained RPTP -negative cells. 23C: mixture of stained and unstained R-PTPκ-positive cells.

In each case, ten aggregates were randomly localized under visible light only. Subsequent inspection under U.V. light consistently showed the staining pattern exemplified in the photographs, dil dye (Molecular Probes, Inc.) was added to the growth medium at a concentration of 3.2μM during heat shock, and washed away prior to recovery and assay. 2xl0 6 cells of each population were mixed and allowed to co-aggregate in a total volume of 1 ml.

FIG. 24A-D. Adhesion of R-PTPK transfected cells to a surface coated with recombinant purified R-PTPK extracellular domain protein. R-PTPκ-negative, 24A, or positive, 24B, S2 cells, or R-PTPκ-negative, 24C, and positive, 24D, L6 cells were incubated with a surface partially coated with the K2AP protein (circle) , and the adherent cells fixed and stained. Amino acids 1-639 of the RPTPK proprotein were fused in-frame with human placental alkaline phosphatase in the vector pBacblue III (Invitrogen) by a series of appropriate cloning steps. Recombinant virus was generated and used to infect High-Five cells for production of the K2AP fusion protein using standard procedures. A secreted alkaline phosphatase (AP) control protein was generated in L6 myoblast cells by stable transfection with a modified version of the AP- TAG vector encoding a fusion protein of AP with a signal peptide. Both proteins were affinity purified by elution from an anti-alkaline phosphatase monoclonal antibody (Medix Biotech) column using 100 mM diethanolamine pH 11.5, or 50% ethylene glycol, dialyzed against PBS, and stored at 4°C. The K2AP and AP proteins were approximately 90% and 50% pure, resp. as determined by silver staining. To generate a mammalian cell line expressing the RPTPK protein, an MJ 30-based RPTPK expression vector was co-transfected with pSVneo into L6 cells, and individual clones surviving G418 selection screened for expression using immunoblotting. This procedure did not detect endogenous RPTPK protein in the parental L6 cells. The expressed protein underwent appropriate furin cleavage as described (Jiang, Y.-P. et al., 1993, Mol. Cell. Biol. 13_:2942) . For adhesion assays, 4μl aliquots of protein samples (20μg/ml) were spotted on 35 mm

bacteriological Petri dishes and incubated at room temperature for 30 minutes. The solutions were removed by aspiration, and the surface of the entire plate blocked with 1% heat-inactivated BSA for 60-90 minutes. The plates were incubated with a suspension of S2 cells (4xl0 6 /ml) in BSS with shaking (50 rpm) for one hour at room temperature, or with L6 cells in S- MEM (2xl0 6 /ml) without shaking, at 37°C, washed three times with PBS, fixed and stained.

5. DETAILED DESCRIPTION OF THE INVENTION Through the use of recombinant DNA methods, the present inventors have identified novel mammalian receptor-type (transmembrane) protein tyrosine phosphatases (PTPase; EC 3.1.3.48). In view of its receptor-like structure, and the likelihood that it is part of a family, the inventors have termed this protein, RPTPK (receptor protein tyrosine phosphatase- K.) . The family is designated herein as the "RPTPs." Human RPTPK has 1444 amino acids (SEQ. ID N0:2).

Human RPTPK (also designated MCP7) has an extracellular domain composed of one "MAM" domain, which is a sequence motif spanning about 170 amino acid residues, which was recently established by comparison of several functionally diverse receptors (including RPTPμ and the A5 protein) and is thought to play a role in cell adhesion (Beck ann & Bork, 1993, TIBS 18.:40-41). The extracellular domain further includes one Ig-like, and four FN-type Ill-like segments. It therefore shares structural features with some cell adhesion molecules, permitting the classification of RPTPK into the type II PTPase class. The cDNA cloning of human RPTPK and the complete DNA and amino acid sequences of human RPTPK and its murine homologue are described herein. Northern

analysis has been used to identify the natural expres¬ sion of the protein in various cells and tissues. A partial cDNA clone of the catalytic domain of RPTPK/HPTPK has been previously described (commonly assigned U.S. Patent Application Serial No. 07/654,188, from which the present application claims priority; Kaplan et al., Proc. Natl . Acad. Sci . 87:7000-7004 (1990); Krueger et al., EMBO J. 9:3241- 3252 (1990)).

RPTPK has been shown to be expressed in anatomically distinct regions of rat brain and its expression has been found to be developmentally regulated. Remarkably, in addition to being composed of intracellular domains having enzymatic activity, the receptor family to which RPTPs belong includes transmembrane proteins having and N-terminal extracellular domains, analogous to the tyrosine kinase enzyme family (Tonks, N.K. et al . (1988) Biochemistry 27:8695-8701; Charbonneau, H. et al . (1988) Proc . Natl . Acad . Sci . USA 85:7182-7186; Streuli, M. et al . (1988) J. Exp. Med . 168:1523- 2530; Streuli, M. et al . (1989) Proc . Natl . Acad . Sci . USA 86:8698-8702). The present inventors have therefore concluded that ligands in the extracellular environment can control the activity of this membrane- associated subclass of PTPases.

Further, results presented in the current invention demonstrate that Type II RPTPs undergo homophilic binding, i.e.. Type II RPTP receptor molecules have the ability to bind to each other. Homophilic binding, as defined here, may include intercellular binding and/or binding of at least two Type II RPTP receptor proteins present on the surface of the same cell. In addition, homophilic binding may

include not only binding of identical Type II RPTP molecules to each other, for example binding of at least two RPTPK molecules to each other, but may also include the binding of any two Type II RPTP molecules to each other, such as, for example, the binding of RPTPK to another Type II RPTP molecule. As demonstrated in the Working Example presented in Section 15, below, RPTPK undergoes intercellular homophilic binding to other RPTPK molecules. This result represents the first example of such a homophilic binding mechanism observed within the RPTP family of molecules, and provides a link between cell- cell contact and cellular signaling events involving tyrosine phosphorylation.

RPTPK is useful in methods for screening drugs and other agents which are capable of activating or inhibiting the PTPase enzymatic activity, and thereby affecting major pathways of cellular metabolism. By attaching an intact RPTPK, or the ligand-binding portion thereof, to a solid phase matrix, an affinity probe is created which can be used to screen biological products or chemical agents for their capacity to interact with the receptor on the basis of their binding activity. Bound material can then be eluted from the affinity probe in purified form.

RPTPK is also useful in methods for screening drugs and other agents which are capable in inhibiting Type II RPTP homophilic binding, and thus affecting major processes involving, but not limited to, cell- cell and/or cell-ECM (extracellular matrix) interactions. By attaching an intact Type II RPTP such as RPTPK, or an extracellular domain thereof, to a solid matrix, drugs or other agents may be screened for their ability to bind the RPTP. Those agents

which bind the RPTP with specificity may be eluted off the solid phase matrix in purified form and further tested for their ability to inhibit RPTP homophilic binding. Note that it is intended to be within the scope of this invention that the inhibition of RPTP homophilic binding described herein refers to not only the binding of at least two identical Type II RPTP molecules, such as at least two RPTPK molecules to each other, but also to binding of any Type II RPTP class of molecules to each other, such as, for example, the binding of RPTPK to another Type II RPTP molecule. Potential agents which may inhibit such Type II RPTP binding may include, but are not limited to, soluble portions of Type II RPTP extracellular domains, antibodies directed against Type II RPTP extracellular domain epitopes, or small synthetic molecules. RPTP extracellular domains may include all or any inhibitory portion of the MAM, Ig, and/or fibronectin Type III (FN-III) domains, as well as peptides which include the HAV, and/or the RXR/LR consensus sequences, as described below. Any of the inhibitory compounds which inhibit homophilic RPTP binding may but are not required to modulate the phosphatase activity of the RPTP molecules whose binding capability is affected.

Further, the ability of a compound to inhibit Type II RPTPK homophilic binding may be tested in a variety of ways. RPTPK will be used as an example, but it should be kept clear that such techniques may be used for any Type II RPTP molecule. RPTPK, or an extracellular domain thereof, may first be immobilized by attachment to a solid matrix, using techniques well known to those of ordinary skill in the art. Such a solid matrix may include but is not limited to a petri dish, microtiter well, or a glass, plastic or agarose

bead. Second, RPTPK, either in a purified protein form or, alternatively, present in a cell membrane preparation or present on the surface of an intact cell, may be incubated in the presence of the solid matrix together with a compound of interest. The ability of the compound to inhibit RPTRK homophilic binding to the solid matrix may then be assayed by determining if RPTPK molecules bind the immobilized molecules. Such a determination may be accomplished using a variety of techniques well known to those of ordinary skill in the art and include, but are not limited to the labelling of the RPTPK present in purified form, in a cell membrane preparation, or in an intact cell. Alternatively, a compound of interest may be tested by incubating RPTP -expressing cells in the presence of the compound of interest and subsequently assaying the ability of the cells to undergo aggregation. Aggregation assays may include, b ut are not limited to directly counting aggregates using the aid of a microscope, and/or determining super-threshold particles with a coulter-counter.

Methods for coupling proteins and peptides to a solid phase matrix or carrier, the solid phase matrix materials useful in these methods, and means for elution, are well known to those of skill in the art. The RPTPK protein, or derivatives thereof having enzymatic activity, can be used for testing agents or compounds capable of enhancing or inhibiting the phosphatase activity. The ability of a compound under test to modify phosphatase activity can be tested in an in vitro system wherein the test compound is added to purified RPTPK protein, or an enzymatically active derivative thereof, and the effects on enzyme activity measured using standard

enzymological procedures well known to those of skill in the art.

Alternatively, the action of a compound on RPTPK enzymatic activity can be measured in a whole cell preparation using live or fixed cells, or a membrane fraction derived from live or fixed cells. This method is useful or screening compounds acting via the extracellular receptor portion of the protein, as well as compounds acting directly on the enzymatic portion of the protein. A test compound is incubated with cells, or with a membrane preparation derived therefrom, which express high amounts of RPTPK, such as transfected COS or NIH-3T3 cells. The amount of cellular phosphotyrosine is then measured, using methods well-known in the art (Honegger, A.M. et al.. Cell 51:199-209 (1987); Margolis, B. et al.. Cell 57:1101-1107 (1989)). The results are compared to results obtained in the absence of the test compound, or in the absence or presence of a known activator of RPTPK enzymatic activity. In such studies, the action of the test compound in the presence of an activator of tyrosine kinase can also be measured. A compound which stimulates RPTPK enzymatic activity will result in a net decrease in the amount of phosphotyrosine, whereas a compound which inhibits RPTPK enzymatic activity will result in a net increase in the amount of phosphotyrosine. Compounds which inhibit homophilic Type II RPTP binding may also modulate the enzymatic activity of the RPTP molecules they affect, either by increasing or decreasing the RPTPs' phosphatase activity.

In the case of growth factor receptors which are tyrosine kinases, such as the receptors for epidermal growth factor (EGF) and for platelet-derived growth factor (PDGF) , tyrosine phosphorylation is linked to

cell growth and to oncogenic transformation. Activation of a PTPase, leading to dephosphorylation, would serve as a counterregulatory mechanism to prevent or inhibit growth, and might serve as an endogenous regulatory mechanism against cancer. Thus, mutation or dysregulation of this receptor/enzyme system may promote susceptibility to cancer.

Inhibitory compounds which are found that are capable of inhibiting Type II RPTP homophilic binding may be used to modulate a variety of cellular processes including, but not limited to those involving cell-cell and/or cell-ECM interactions. Such processes include, but are not limited to normal cellular functions such as differentiation and cell cycle control; normal cellular behaviors including, but not limited to motility, contact inhibition, cell adhesion, and signal transduction; and abnormal or potentially deleterious processes such as cellular transformation to a cancerous state.

Inhibitory compounds which inhibit Type II RPTP homophilic binding may be used to modulate such processes in mammals by administration of an effective concentration of the inhibitory compound to a mammal, using techniques well known to those of ordinary skill in the art. Inhibitory compounds may include, but are not limited to, compounds comprising soluble RPTP Type II extracellular domains, for example, soluble RPTPK extracellular domains. Depending on the conditions being treated, agents may be formulated and administered systemically or locally. Techniques for formulation and administration may be found in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, latest edition. Suitable routes may include oral, rectal, transmucosal, or intestinal administration;

parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventicular, intravenous, intraperitoneal, intranasal, or intraocular injections, just to name a few. For injection, the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution. Ringer's solution, or physiological saline bffer. For such transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.

The insulin receptor is also a tyrosine kinase, and phosphorylation of tyrosine in cells bearing insulin receptors would be associated with normal physiological function. In contrast to the case of cell growth and cancer, activation of an RPTP would counteract insulin effects. Subnormal RPTP levels or enzymatic activity would act to remove a normal counterregulatory mechanisms. Perhaps more important, though, over-activity, or inappropriate activation, of an RPTP, such as RPTPK, would be expected to partially or totally inhibit the action of insulin on cells, leading to diabetes (of an insulin-resistant variety) . Thus, susceptibility to diabetes may be associated with RPTPK dysregulation.

Therefore, the methods of the present invention for identifying normal or mutant genes encoding RPTPK, or for measuring the amount or activity of RPTPK associated with a cell or tissue, can serve as methods for identifying susceptibility to cancer, diabetes, or other diseases associated with alterations in cellular phosphotyrosine metabolism. The present invention provides methods for evaluating the presence of, and the level of, normal

or mutant RPTPK in a cell or in a subject. Absence, or more typically, low expression of the RPTPK, or presence of a mutant RPTPK, in an individual may serve as an important predictor of susceptibility to oncogenic transformation and the development of cancer. Alternatively, over-expression of RPTPK, possibly due to a mutant receptor/enzyme system insensitive to negative regulation, or due to overabundance of a stimulatory ligand in the body, may serve as an important predictor of susceptibility to diabetes.

An oligonucleotide probe corresponding to a DNA sequences encoding a part of RPTPK (see below) is used to test cells from a subject for the presence of DNA or RNA sequences encoding the RPTPK A preferred probe would be one directed to the nucleic acid sequence encoding at least 4 amino acid residues, and preferably at least 5 amino acid residues, of the RPTPK. Qualitative or quantitative assays can be performed using such probes. For example. Northern analysis (see Section 7, below) is used to measure expression of an RPTPK mRNA in a cell or tissue preparation. Such methods can be used even with very small amounts of DNA obtained from an individual, following use of selective amplification techniques. Recombinant DNA methodologies capable of amplifying purified nucleic acid fragments have long been recognized. Typically, such methodologies involve the introduction of the nucleic acid fragment into a DNA or RNA vector, the clonal amplification of the vector, and the recovery of the amplified nucleic acid fragment. Examples of such methodologies are provided by Cohen et al. (U.S. Patent 4,237,224), Sambrook et al . , MOLECULAR CLONING : A LABORATORY MANUAL , Second

Edition, Cold Spring Harbor Press, Cold Spring Harbor, NY (1989) , which references are herein incorporated by reference) . An in vitro enzymatic method which is capable of increasing the concentration of such desired nucleic acid molecules is called the "polymerase chain reaction or "PCR" (Mullis, K. et al., Cold Spring Harbor Symp. Quant . Biol . 51:263-273 (1986); Erlich, H. et al., EP 50424, EP 84796, EP 258017, EP 237362; Mullis, K., EP 201184; Mullis, K. et al . , US 4,683,202; Erlich, H., US 4,582,788; and Saiki, R. et al . , US 4,683,194) .

The PCR provides a method for selectively increasing the concentration of a particular nucleic acid sequence even when that sequence has not been previously purified and is present only in a single copy in a particular sample. The method can be used to amplify either single- or double-stranded DNA. The method uses two oligonucleotide probes to serve as primers for the template-dependent, polymerase mediated replication of a desired nucleic acid molecule.

The precise nature of the two oligonucleotide probes of the PCR method is critical to the success of the method. Polymerase dependent amplification of a nucleic acid molecule proceeds by the addition of a 5' nucleotide triphosphate to the 3• hydroxyl end of a nucleic acid molecule. Thus, the action of a polymerase extends the 3' end of a nucleic acid molecule. These inherent properties are exploited in the selection of the oligonucleotide probes of the PCR. The oligonucleotide sequences of the probes are selected such that they contain sequences identical to, or complementary to, sequences which flank the particular nucleic acid sequence whose amplification

is desired. More specifically, the oligonucleotide sequence of the "first" probe is selected such that it is capable of hybridizing to an oligonucleotide sequence located 3' to the desired sequence, whereas the oligonucleotide sequence of the "second" probe is selected such that it contains an oligonucleotide sequence identical to one present 5' to the desired region. Both probes possess 3' hydroxy groups, and therefore can serve as primers for nucleic acid synthesis.

PCR reaction conditions are cycled between (a) those conducive to hybridization and nucleic acid polymerization, and (b) those which result in the denaturation of duplex molecules. In the first step of the reaction, the nucleic acids of the sample are transiently heated, and then cooled, in order to denature any double-stranded molecules. The "first" and "second" probes are then added to the sample at a concentration which greatly exceeds that of the desired nucleic acid molecule. Upon incubation under conditions conducive to hybridization and polymerization, the "first" probe will hybridize to the sample nucleic acid molecule at a position 3• to the sequence to be amplified. If the nucleic acid molecule of the sample was initially double-stranded, the "second" probe will hybridize to the complementary strand of the nucleic acid molecule at a position 3' to the sequence which is the complement of the sequence whose amplification is desired. Upon addition of a polymerase, the 3' ends of the "first" and (if the nucleic acid molecule was double-stranded) "second" probes will be extended. The extension of the "first" probe will result in the synthesis of an oligonucleotide having the exact sequence of the desired nucleic acid. Extension of the "second" probe

will result in the synthesis of an oligonucleotide having the exact sequence of the complement of the desired nucleic acid. The PCR reaction is capable of exponential amplification of specific nucleic acid sequences because the extension product of the "first" probe, of necessity, contains a sequence which is complementary to a sequence of the "second" probe, and thus can serve as a template for the production of an extension product of the "second" probe. Similarly, the extension product of the "second" probe, of necessity, contains a sequence which is complementary to a sequence of the "first" probe, and thus can serve as a template for the production of an extension product of the "first" probe. Thus, by permitting cycles of polymerization, and denaturation, a geometric increase in the concentration of the desired nucleic acid molecule can be achieved. For reviews of the PCR, see: Mullis, K.B., Cold Spring Harbor Symp. Quant . Biol . 51:263-273 (1986); Saiki, R.K. et al . BioTechnology 3:1008-1012 (1985); Mullis, K.B. et al . Meth . Enzymol . 155:335-350 (1987).

In one embodiment, the present invention is directed to a naturally occurring mammalian RPTPK. In another embodiment, the present invention is directed to a recombinant mammalian RPTPK. The preferred mammalian RPTPK of the present invention is of human origin. The invention provides the naturally occurring molecule substantially free of other proteins with which it is natively associated. "Substantially free of other proteins or glycoproteins" indicates that the protein has been purified away from at least 90 per cent (on a weight basis) , and from even at least 99 per cent if desired, of other proteins and glycoproteins with which it is

natively associated, and is therefore substantially free of them. That can be achieved by subjecting the cells, tissue or fluid containing the RPTPK to standard protein purification techniques such as an immunoabsorbent column bearing an antibody specific for the protein. Other forms of affinity purification utilize solid-phase substrates which bind the RPTP'ε enzymatic domain, or a ligand that will bind to the receptor domain. Alternatively, the purification can be achieved by a combination of standard methods, such as ammonium sulfate precipitation, molecular sieve chromatography, and ion exchange chromatography. It will be understood that the RPTPK of the present invention can be biochemically purified from a variety of cell or tissue sources. For preparation of naturally occurring RPTPK, tissues such as mammalian brain, especially of human origin, are preferred.

Alternatively, because the gene for the RPTPK can be isolated or synthesized, the polypeptide can be synthesized substantially free of other mammalian proteins or glycoproteins in a prokaryotic organism or in a non-mammalian eukaryotic organism, if desired. As intended by the present invention, a recombinant RPTPK molecule produced in mammalian cells, such as transfected COS, NIH-3T3, or CHO cells, for example, is a protein with the naturally occurring amino acid sequence or is a functional derivative thereof. Where a naturally occurring protein or glycoprotein is produced by recombinant means, it is provided substantially free of the other proteins and glycoproteins with which it is natively associated. Alternatively, methods are well known for the synthesis of polypeptides of desired sequence on solid phase supports and their subsequent separation from the support.

The present invention provides any of a number of "functional derivatives" of the RPTPK. By "functional derivative" is meant a "fragment," "variant," "analog," or "chemical derivative" of the RPTPK, which terms are defined below. A functional derivative retains at least a portion of the function of the RPTPK, such as (a) binding to a specific antibody, (b) phosphatase enzymatic activity, or (c) binding of the extracellular "receptor" domain to a ligand, which permits its utility in accordance with the present invention.

A "fragment" of the RPTPK refers to any subset of the molecule, that is, a shorter peptide. A "variant" of the RPTPK refers to a molecule substantially similar to either the entire peptide or a fragment thereof. Variant peptides may be conveniently prepared by direct chemical synthesis of the variant peptide, using methods well-known in the ar -

Alternatively, amino acid sequence variants of the peptide can be prepared by mutations in the DNA which encodes the synthesized peptide. Such variants include, for example, deletions from, or insertions or substitutions of, residues within the amino acid sequence. Any combination of deletion, insertion, and substitution may also be made to arrive at the final construct, provided that the final construct possesses the desired activity. Obviously, the mutations that will be made in the DNA encoding the variant peptide must not alter the reading frame and preferably will not create complementary regions that could produce secondary mRNA structure (see European Patent Publication EP 75444) . At the genetic level, these variants ordinarily are prepared by site-directed mutagenesis (as

exemplified by Adelman et al . , DNA 2:183 (1983)) of nucleotides in the DNA encoding the protein or peptide molecule, thereby producing DNA encoding the variant, 5 and thereafter expressing the DNA in recombinant cell culture. The variants typically exhibit the same qualitative biological activity as the nonvariant protein or peptide.

An "analog" of the RPTPK refers to a non-natural Q molecule substantially similar to either the entire molecule or a fragment thereof.

A "chemical derivative" of the RPTPK contains additional chemical moieties not normally a part of the peptide. Covalent modifications of the RPTPK 5 protein or of a peptide derived therefrom, are included within the scope of this invention. Such modifications may be introduced into the molecule by reacting targeted amino acid residues of the peptide with an organic derivatizing agent that is capable of o reacting with selected side chains or terminal residues.

Cysteinyl residues most commonly are reacted with alpha-haloacetates (and corresponding amines) , such as chloroacetic acid or chloroacetamide, to give 5 carboxymethyl or carboxyamidomethyl derivatives.

Cysteinyl residues also are derivatized by reaction with bromotrifluoroacetone, α-bromo-/3-(5- imidozoyl)propionic acid, chloroacetyl phosphate, N- alkylmaleimides, 3-nitro-2-pyridyl disulfide, methyl 0 2-pyridyl disulfide, p-chloromercuribenzoate, 2- chloromercuri-4- nitrophenol, or chloro-7-nitrobenzo- 2-oxa-l,3-diazole.

Histidyl residues are derivatized by reaction with diethylprocarbonate, pH 5.5-7.0, because this 5 agent is relatively specific for the histidyl side chain. Para-bromophenacyl bromide also is useful; the

reaction is preferably performed in 0.1 M sodium cacodylate at pH 6.0.

Lysinyl and amino terminal residues are reacted with succinic or other carboxylic acid anhydrides. Derivatization with these agents has the effect of reversing the charge of the lysinyl residues. Other suitable reagents for derivatizing α-amino-containing residues include imidoesters such as methyl picolinimidate; pyridoxal phosphate; pyridoxal; chloroborohydride; trinitrobenzenesulfonic acid; 0- methylisourea; 2,4 pentanedione; and transaminaεe- catalyzed reaction with glyoxylate.

Arginyl residues are modified by reaction with one or several conventional reagents, among them phenylglyoxal, 2,3- butanedione, 1,2-cyclohexanedione, and ninhydrin. Derivatization of arginine residues requires that the reaction be performed in alkaline conditions because of the high pK. of the guanidine functional group. Furthermore, these reagents may react with the groups of lysine as well as the arginine e-amino group.

The specific modification of tyrosyl residues per se has been studied extensively, with particular interest in introducing spectral labels into tyrosyl residues by reaction with aromatic diazonium compounds or tetranitromethane. Most commonly, N-acetylimidizol and tetranitromethane are used to form O-acetyl tyrosyl species and 3-nitro derivatives, respectively. Carboxyl side groups (aspartyl or glutamyl) are selectively modified by reaction with carbodiimides (R'-N-C-N-R*) such as l-cyclohexyl-3-(2-morpholinyl- (4-ethyl) carbodiimide or l-ethyl-3-(4-azonia-4,4- dimethylpenty1) carbodiimide. Furthermore, aspartyl and glutamyl residues are converted to asparaginyl and glutaminyl residues by reaction with ammonium ions.

Glutaminyl and asparaginyl residues may be deamidated to the corresponding glutamyl and aspartyl residues, under mildly acidic conditions. Either form of these residues falls within the scope of this invention.

Derivatization with bifunctional agents is useful for cross-linking the protein or peptide to a water- insoluble support matrix or to other macromolecular carriers. Commonly used cross-linking agents include, e.g. , 1,l-bis(diazoacetyl)-2-phenylethane, glutaraldehyde, N-hydroxysuccinimide esters, for example, esters with 4-azidosalicylic acid, homobifunctional imidoesterε, including disuccinimidyl esters such as 3,3'- dithiobis(succinimidyl- propionate) , and bifunctional maleimides such as bis- N-maleimido-l,8-octane. Derivatizing agents such as methyl-3-[ (p-azidophenyl)dithio]propioimidate yield photoactivatable intermediates that are capable of forming crosslinks in the presence of light.

Alternatively, reactive water-insoluble matrices such as cyanogen bromide-activated carbohydrates and the reactive substrates described in U.S. Patent Nos. 3,969,287; 3,691,016; 4,195,128; 4,247,642; 4,229,537; and 4,330,440 are employed for protein immobilization. Other modifications include hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the X- a ino groups of lysine, arginine, and histidine side chains (T.E. Creighton, PROTEINS: STRUCTURE AND MOLECULE PROPERTIES , W.H. Freeman & Co. , San Francisco, pp. 79-86 (1983)), acetylation of the N- terminal amine, and, in some instances, amidation of the C-terminal carboxyl groups. Such derivatized moieties may improve the solubility, absorption, biological half life, and the

like. The moieties may alternatively eliminate or attenuate any undesirable side effect of the protein and the like. Moieties capable of mediating such effects are disclosed, for example, in REMINGTON' S PHARMACEUTICAL SCIENCES , 16th ed. , Mack Publishing Co., Easton, PA (1980)

This invention is also directed to an antibody specific for an epitope of RPTPK, preferably, of human RPTPK, and the use of such an antibody to detect the presence of, or measure the quantity or concentration of, the RPTPK in a cell, a cell or tissue extract, or a biological fluid.

The term "antibody" is meant to include polyclonal antibodies, monoclonal antibodies (mAbs) , chimeric antibodies, and anti-idiotypic (anti-Id) antibodies.

Polyclonal antibodies are heterogeneous populations of antibody molecules derived from the sera of animals immunized with an antigen, preferably the RPTPK protein or glycoprotein, a peptide derived therefrom or an epitope thereof.

Monoclonal antibodies are a substantially homogeneous population of antibodies to specific antigens. MAbs may be obtained by methods known to those skilled in the art. See, for example Kohler and Milstein, Nature 256:495-497 (1975) and U.S. Patent No. 4,376,110. Such antibodies may be of any immunoglobulin class including IgG, IgM, IgE, IgA, and any subclass thereof. The hybridoma producing the mAbs of this invention may be cultivated in vitro or in vivo . Production of high titers of mAbs in vivo production makes this the presently preferred method of production. Briefly, cells from the individual hybridomas are injected intraperitoneally into pristane-primed BALB/c mice to produce ascites fluid

containing high concentrations of the desired mAbs. MAbs of isotype IgM or IgG may be purified from such ascites fluids, or from culture supernatants, using column chromatography methods well known to those of skill in the art.

Chimeric antibodies are molecules different portions of which are derived from different animal species, such as those having variable region derived from a murine mAb and a human immunoglobulin constant region. Chimeric antibodies and methods for their production are well-known in the art (Cabilly et al, Proc . Natl . Acad. Sci . USA 81:3273-3277 (1984); Morrison et al . , Proc . Natl . Acad . Sci . USA 81:6851- 6855 (1984); Boulianne et al.. Nature 312:643-646

(1984); Neuberger et al. , Nature 314 : 268-210 (1985); Taniguchi et al., European Patent Publication EP171496 (February 19, 1985); Morrison et al . , European Patent Publication EP 173494 (March 5, 1986); Neuberger et al., PCT Publication WO 86/01533 (March 13, 1986); Kudo et al . , European Patent Publication EP 184187 (June 11, 1986); Sahagan et a . , J. Immunol. 137:1066- 1074 (1986); Robinson et a . , International Patent Publication #PCT/US86/02269 (7 May 1987); Liu et al . , Proc . Natl . Acad . Sci . USA 84:3439-3443 (1987); Sun et al., Proc. Natl. Acad. Sci . USA 84:214-218 (1987); Better et al . , Science 240:1041-1043 (1988)). These references are hereby incorporated by reference. An anti-idiotypic (anti-Id) antibody is an antibody which recognizes unique determinants generally associated with the antigen-binding site of an antibody. An anti-Id antibody can be prepared by immunizing an animal of the same species and genetic type (e.g. mouse strain) as the source of the mAb with the mAb to which an anti-Id is being prepared. The immunized animal will recognize and respond to the

idiotypic determinants of the immunizing antibody by producing an antibody to these idiotypic determinants (the anti-Id antibody) . The anti-Id antibody may also be used as an "immunogen" to induce an immune response in yet another animal, producing a so-called anti- anti-Id antibody. The anti-anti-Id may be epitopically identical to the original mAb which induced the anti-Id. Thus, by using antibodies to the idiotypic determinants of a mAb, it is possible to identify other hybrid clones expressing antibodies of identical specificity.

Accordingly, mAbs generated against RPTPK may be used to induce anti-Id antibodies in suitable animals, such as BALB/c mice. Spleen cells from such immunized mice are used to produce anti-Id hybridomas secreting anti-Id mAbs. Further, the anti-Id mAbs can be coupled to a carrier such as keyhole limpet hemocyanin (KLH) and used to immunize additional BALB/c mice. Sera from these mice will contain anti-anti-Id antibodies that have the binding properties of the original.mAb specific for an RPTPK epitope. The anti- Id mAbs thus have their own idiotypic epitopes, or "idiotopeε" structurally similar to the epitope being evaluated, such as an epitope of RPTPK.

The term "antibody" is also meant to include both intact molecules as well as fragments thereof, such as, for example, Fab and F(ab') 2 , which are capable of binding antigen. Fab and F(ab') 2 fragments lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding than an intact antibody (Wahl et al . , J. Nucl . Med . 24:316-325 (1983)).

It will be appreciated that Fab and F(ab') 2 and other fragments of the antibodies useful in the present invention may be used for the detection and

quantitation of RPTPK according to the methods disclosed herein for intact antibody molecules. . Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab') 2 fragments).

An antibody is said to be "capable of binding" a molecule if it is capable of specifically reacting with the molecule to thereby bind the molecule to the antibody. The term "epitope" is meant to refer to that portion of any molecule capable of being bound by an antibody which can also be recognized by that anti¬ body. Epitopes or "antigenic determinants" usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and have specific three dimensional structural characteristics as well as specific charge characteristics. An "antigen" is a molecule or a portion of a molecule capable of being bound by an antibody which is additionally capable of inducing an animal to produce antibody capable of binding to an epitope of that antigen. An antigen may have one, or more than one epitope.

An antibody is said to be specific for an antigen because it reacts in a highly selective manner, with that antigen and not with the multitude of other antigens which are structurally distinct.

The antibodies or antibody fragments of the present invention may be used to quantitatively or qualitatively detect the presence of cells which express the RPTPK protein. This can be accomplished by immunofluorescence techniques employing a fluorescently labeled antibody (see below) coupled with light microscopic, flow cytometric, or fluorimetric detection. For such methods, the

antibody is preferably specific for an extracellular epitope of RPTPK.

The antibodies (or fragments thereof) useful in the present invention may be employed histologically, as in immunofluorescence or immunoelectron microscopy, for in situ detection of RPTPK. In situ detection may be accomplished by removing a histological specimen from a subject, and providing a labeled antibody or antibody fragment of the present invention to such a specimen, preferably by applying or overlaying the antibody over the specimen. Through the use of such a procedure, it is possible to determine not only the presence of RPTPK but also its distribution in the examined tissue. Using the present invention, those of ordinary skill will readily perceive that any of a wide variety of histological methods (such as staining procedures) can be modified in order to achieve such in situ detection. Such assays for RPTPK typically comprise incubating a biological sample, such as a biological fluid, a tissue extract, freshly harvested cells, or cells which have been incubated in tissue culture, in the presence of a detectably labeled antibody specific for RPTPK, and detecting the antibody by any of a number of techniques well-known in the art.

The biological sample may be incubated with a solid phase support or carrier such as nitrocellulose, or other solid support which is capable of immobilizing cells, cell particles or soluble pro¬ teins. The support may then be washed with suitable buffers followed by treatment with the detectably labeled RPTPκ-specific antibody. The solid phase support may then be washed with the buffer a second time to remove unbound antibody. The amount of bound

label on said solid support may then be detected by conventional means.

By "solid phase support" is intended any support capable of binding antigen or antibodies. Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, and magnetite. The preferred carrier is totally insoluble in the solution in which the assay of the present invention takes place; partially soluble carriers well-known in the art may also be used. The support material may have virtually any possible structural configuration so long as the support-coupled molecule is capable of binding to an antigen or antibody. Thus, the support configuration may be spherical, as in a bead, or cylindrical, as in the inside surface of a test tube, or the external surface of a rod. Alternatively, the surface may be flat such as a sheet, test strip, etc. Preferred supports include polystyrene beads. Those skilled in the art will know many other suitable carriers for binding antibody or antigen, or will be able to ascertain the same by use of routine experimentation. The binding activity of a given lot of anti-RPTPκ antibody may be determined according to well-known methods. Those skilled in the art will be able to determine operative and optimal assay conditions for each determination by employing routine experimentation.

One of the ways in which the RPTPκ-specific anti¬ body can be detectably labeled is by linking the antibody, or a second antibody which binds to the anti-RPTPκ antibody, to an enzyme and use in an enzyme immunoassay (EIA) . This enzyme, in turn, when later exposed to an appropriate substrate, will react with

the substrate in such a manner as to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorimetric or by visual means. Enzymes which can be used to detectably label the antibody include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-5- steroid isomerase, yeast alcohol dehydrogenase, alpha- glycerophosphate dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta- galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase. The detection can be accomplished by colorimetric methods which employ a chromogenic substrate for the enzyme. Detection may also be accomplished by visual comparison of the extent of enzymatic reaction of a substrate in comparison with similarly prepared standards. Detection may be accomplished using any of a variety of other immunoassayε. For example, by radioactively labeling the antibodies or antibody fragments, it iε poεεible to detect RPTPK through the use of a radioimmunoassay (RIA) (see, for example, Work, T.S. et al . , LABORATORY TECHNIQUES AND

BIOCHEMISTRY IN MOLECULAR BIOLOGY , North Holland Publishing Company, New York, 1978, which is incorporated by reference herein) . The radioactive isotope can be detected by such means as the use of a gamma counter or a scintillation counter or by autoradiography.

It is also possible to label the antibody with a fluorescent compound. When the fluorescently labeled antibody is exposed to light of the proper wave length, its presence can then be detected due to fluorescence. Among the most commonly used

fluorescent labelling compounds are fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o- phthaldehyde and fluorescamine. The antibody can also be detectably labeled using fluorescence emitting metals such as - 52 Eu, or others of the lanthanide series. These metals can be attached to the antibody using such metal chelating groups as diethylenetriaminepentaacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA) .

The antibody also can be detectably labeled by coupling it to a chemiluminescent compound. The presence of the chemiluminescent-tagged antibody is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction. Exampleε of particularly useful chemiluminescent labeling compounds are luminol, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester. Likewise, a bioluminescent compound may be used to label the antibody of the present invention. Bioluminescence is a type of chemilumineεcence found in biological systems in which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein iε determined by detecting the presence of luminescence. Important bioluminescent compounds for purposes of labeling are luciferin, luciferase and aequorin.

The antibody molecules of the present invention may be adapted for utilization in an immunometric assay, alεo known as a "two-site" or "sandwich" assay. In a typical immunometric asεay, a quantity of unlabeled antibody (or fragment of antibody) is bound to a solid support and a quantity of detectably labeled soluble antibody is added to permit detection and/or quantitation of the ternary complex formed

between solid-phase antibody, antigen, and labeled antibody.

Typical, and preferred, immunometric assays include "forward" assays in which the antibody bound to the solid phase is first contacted with the sample being tested to extract the antigen from the sample by formation of a binary solid phase antibody-antigen complex. After a suitable incubation period, the solid support is washed to remove the residue of the fluid sample, including unreacted antigen, if any, and then contacted with the solution containing a labeled second antibody (which functions as a "reporter molecule") . After a second incubation period to permit the labeled antibody to complex with the antigen bound to the solid support through the unlabeled antibody, the solid support iε washed a second time to remove the unreacted labeled antibody. In another type of "sandwich" assay, which may also be useful with the antigens of the present invention, the so-called "simultaneous" and "reverse" assays are used. A simultaneous assay involves a single incubation step as the antibody bound to the solid support and labeled antibody are both added to the sample being tested at the same time. After the incubation is completed, the solid support iε waεhed to remove the reεidue of fluid sample and uncomplexed labeled antibody. The presence of labeled antibody associated with the solid εupport iε then determined aε it would be in a conventional "forward" εandwich assay.

In the "reverse" aεsay, stepwise addition first of a solution of labeled antibody to a fluid sample followed by the addition of unlabeled antibody bound to a solid support after a suitable incubation period is utilized. After a second incubation, the solid

phase is washed in conventional fashion to free it of the residue of the sample being tested and the solution of unreacted labeled antibody. The determination of labeled antibody associated with a solid support is then determined as in the "simultaneous" and "forward" asεayε.

The preεence of normally functioning RPTPK in a εubject can also be tested using direct enzymatic assays, for the tyrosine phosphatase activity. Such biochemical measurements can be performed in vitro , using purified enzymeε, allowing precise measurements of enzyme activity, or with membrane preparations, or whole cells, where the net phosphotyrosine level is determined.

In additional embodiments of the present invention, a nucleic acid molecule, prefereably DNA, comprising a sequence encoding an RPTPK protein and methods for expressing the DNA molecule are provided. One of ordinary skill in the art will know how to identify and clone additional RPTP molecules, of human or other mammalian species, which have sequence homology to the RPTPK moleculeε described herein, using the genetic sequences and oligonucleotides of the present invention without undue experimentation. Furthermore, manipulation of the genetic constructε of the preεent invention allow the grafting of a particular ligand-binding receptor domain onto the tranεmembrane and catalytic portions of the RPTPK resulting in chimeric moleculeε. Non-limiting examples of such chimeric moleculeε include RPTPK wherein the receptor portion iε an epidermal growth factor receptor, a fibroblaεt growth factor receptor, and the like. Genetically engineered chimeric recep- torε are known in the art (εee, for example, Riedel, H. et al., Nature 324:628-670 (1986)).

Genetic constructs encoding RPTPK, functional derivative thereof, and chimeric molecules such as those described above, can be used in gene therapy. An abnormal or dysfunctional RPTPK, which results in disease, may be replaced by infusion or implantation of cells of the desired lineage (such as hemopoietic cells, neurons, etc.) transfected with DNA encoding normal RPTPK. Alternatively, or additionally, cells carrying a chimeric RPTPK having a receptor portion which binds a ligand of choice (e.g., EGF) can be used for such gene therapy.

The recombinant DNA molecules of the present invention can be produced through any of a variety of means, such as, for example, DNA or RNA synthesis, or more preferably, by application of recombinant DNA techniques. Techniques for synthesizing such molecules are disclosed by, for example, Wu, R. , et al . (Prog. Nucl . Acid . Res . Molec . Biol . 21:101-141 (1978)), and procedures for constructing recombinant molecules can be found in Sambrook et al . (supra) . Oligonucleotides representing a portion of an RPTPK are useful for screening for the presence of genes encoding such proteinε and for the cloning of an RPTPK gene. Techniqueε for synthesizing such oligonucleotides are discloεed by, for example, Wu, R. , et al . , supra .

Protein moleculeε are fragmented aε with cyanogen bromide, or with proteaεes such aε papain, chymotrypsin, trypsin, etc. (Oike, Y., et al . , J. Biol . Chem . 257:9751-9758 (1982); Liu, C. , et al . , Int . J. Pept . Protein Res . 21:209-215 (1983)). Because the genetic code is degenerate, more than one codon may be used to encode a particular amino acid (Watson, J.D., In: MOLECULAR BIOLOGY OF THE GENE , 4th Ed.,

Benjamin/Cummings Publishing Co., Inc., Menlo Park, CA

(1987)). Using the genetic code, one or more different oligonucleotides can be identified, each of which would be capable of encoding the amino acid. The probability that a particular oligonucleotide will, in fact, constitute the actual XXX-encoding sequence can be estimated by considering abnormal base pairing relationships and the frequency with which a particular codon is actually used (to encode a particular amino acid) in eukaryotic cells. Such "codon usage rules" are disclosed by Lathe, R. , et al., J. Mol . Biol . 183:1-12 (1985). Using such "codon usage rules", a single oligonucleotide, or a set of oligonucleotides, that contains a theoretical "most probable" nucleotide sequence capable of encoding RPTPK is identified.

Although occasionally an amino acid sequence may be encoded by only a single oligonucleotide, frequently the amino acid sequence may be encoded by a ny of a set of similar oligonucleotides.

Importantly, whereaε all of the members of this set contain oligonucleotides which are capable of encoding the peptide fragment and, thus, potentially contain the same oligonucleotide sequence aε the gene which encodes the peptide fragment, only one member of the set contains the nucleotide sequence that iε identical to the nucleotide sequence of the gene. Because this member iε present within the set, and is capable of hybridizing to DNA even in the presence of the other members of the set, it is possible to employ the unfractionated set of oligonucleotides in the same manner in which one would employ a single oligonucleotide to clone the gene that encodes RPTPK. The oligonucleotide, or set of oligonucleotides, containing the theoretical "most probable" sequence capable of encoding the RPTPK fragment is used to

identify the sequence of a complementary oligonucleotide or set of oligonucleotides which is capable of hybridizing to the "most probable" sequence, or set of sequences. An oligonucleotide containing such a complementary sequence can be employed as a probe to identify and isolate the RPTPK gene (Sambrook et a ., supra).

A suitable oligonucleotide, or set of oligonucleotides, capable of encoding a fragment of the RPTPK gene (or complementary to such an oligonucleotide) is identified as above and synthesized, using procedureε well known in the art (Belagaje, R. , et a ., J. Biol . Chem . 254:5765-5780 (1979); Maniatis, T., et a . , In: MOLECULAR MECHANISMS IN THE CONTROL OF GENE EXPRESSION, Nierlich, D.P. , et al . , Eds., Acad. Press, NY (1976); Wu, R. , et al . , Prog. Nucl . Acid Res . Molec . Biol . 21:101-141 (1978); Khorana, R.G., Science 203:614-625 (1979)). DNA synthesis may be achieved using an automated synthesizers. The oligonucleotide probe or set is hybridized by means well known in the art, againεt a DNA or, more preferably, a cDNA preparation derived from cellε which are capable of expressing the RPTPK gene. Techniques of nucleic acid hybridization are discloεed by Sambrook et al . (supra) , and by Hay eε, B.D. , et al . (In: NUCLEIC ACID HYBRIDIZATION, A PRACTICAL APPROACH, IRL Press, Washington, DC (1985)), which referenceε are herein incorporated by reference. Techniqueε εuch as, or similar to, those described above have succesεfully enabled the cloning of geneε for human aldehyde dehydrogenaεeε (Hsu, L.C. et al . , Proc . Natl . Acad . Sci . USA 82:3771-3775 (1985)), fibronectin (Suzuki, S., et al. , EMBO J . 4:2519-2524 (1985)), the human estrogen receptor gene (Walter, P., et a . , Proc . Natl . Acad . Sci . USA

82:7889-7893 (1985)), tissue-type plasminogen activator (Pennica, D., et al . , Nature 301:214-221 (1983)) and human term placental alkaline phosphatase complementary DNA (Kam, W. , et al., Proc. Natl . Acad. Sci . USA 82 : (715-8719 (1985)).

In a alternative way of cloning the RPTPK gene, a library of expression vectorε iε prepared by cloning DNA or, more preferably, cDNA (from a cell capable of expressing RPTPK) into an expression vector. The library is then screened for members capable of expressing a protein which binds to an anti-RPTPκ antibody, and which has a nucleotide sequence that is capable of encoding a polypeptide that has the same amino acid sequence as all or part of RPTP . In this embodiment, DNA, or more preferably cDNA, is extracted and purified from a cell which is capable of expressing RPTPK protein. The purified cDNA is fragmented (by shearing, endonuclease digestion, etc.) to produce a pool of DNA or cDNA fragments. DNA or cDNA fragments from this pool are then cloned into an expression vector in order to produce a genomic or cDNA library of expression vectors whose members each contain a unique cloned DNA or cDNA fragment. An "expression vector" is a vector which (due to the presence of appropriate transcriptional and/or translational control sequences) is capable of expressing a DNA molecule which has been cloned into the vector and of thereby producing a peptide or protein. Expression of the cloned sequenceε occurε when the expression vector is introduced into an appropriate host cell. If a prokaryotic expression vector is employed, then the appropriate host cell would be any prokaryotic cell capable of expressing the cloned sequenceε. If a eukaryotic expression vector is employed, then the appropriate host cell

would be any eukaryotic cell capable of expressing the cloned sequences. Importantly, since eukaryotic DNA may contain intervening sequences, and since such sequences cannot be correctly processed in prokaryotic cells, it is preferable to employ cDNA from a cell which is capable of expressing RPTPK in order to produce a prokaryotic genomic expresεion vector library. Procedureε for preparing cDNA and for producing a genomic library are diεclosed by Sambrook et al . (supra) .

A DNA sequence encoding RPTPK of the present invention, or encoding functional derivatives thereof, may be recombined with vector DNA in accordance with conventional techniques, including blunt-ended or staggered-ended termini for ligation, restriction enzyme digestion to provide appropriate termini, filling in of cohesive endε as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and ligation with appropriate ligases. Techniqueε for such manipulations are disclosed by Sambrook et al., supra , and are well known in the art.

A nucleic acid molecule, such as DNA, is "capable of expressing" a polypeptide if it contains nucleotide sequences which contain transcriptional and translational regulatory information and such sequences are "operably linked" to a polypeptide coding sequence. An operable linkage is a linkage in which the regulatory DNA sequenceε and the coding sequence are connected in such a way as to permit gene expresεion. The preciεe nature of the regulatory regionε needed for gene expreεεion may vary from organiεm to organism, but shall in general include a promoter region which, in prokaryoteε, contains both the promoter (which directs the initiation of RNA transcription) as well as the DNA εequenceε which,

when transcribed into RNA, will signal the initiation of protein synthesis. Such regions will normally include those 5*-non-coding sequences involved with initiation of transcription and translation, such as the TATA box, capping sequence, CAAT sequence, and the like.

If desired, the non-coding region 3' to the coding sequence may be obtained by the above-described methods. This region may be retained for its transcriptional termination regulatory sequences, such aε termination and polyadenylation. Thus, by retaining the 3'-region naturally contiguous to the DNA coding sequence, the transcriptional termination signals may be provided. Where the transcriptional termination εignals are not satisfactorily functional in the host cell used to express the protein, then a 3' region functional in that host cell may be substituted. Two DNA sequences (such as a promoter region sequence and a RPTPK coding sequence) are said to be operably linked if the nature of the linkage between the two DNA sequences does not (1) result in the introduction of a frame-εhift mutation, (2) interfere with the ability of the promoter to regulate the tranεcription of the RPTPK coding εequence. A promoter region iε operably linked to a DNA coding εequence if the promoter is capable of effecting transcription of the coding sequence. Thus, to express the protein, transcriptional and translational εignalε recognized by an appropriate hoεt are neceεsary. In order to be "operably linked" it is not necessary that two sequences be immediately adjacent to one another. A promoter is a double-stranded DNA (or RNA) molecule which is capable of binding to RNA polymerase

and promoting the transcription of an "operably linked" nucleic acid coding sequence. As used herein, a "promoter sequence" is the sequence of the promoter which is found on that strand of the DNA (or RNA) which is transcribed by the RNA polymerase. A "promoter sequence complement" has a sequence which is the complement of the "promoter sequence." Hence, upon extension of a primer DNA or RNA adjacent to a single-stranded "promoter sequence complement" or, of a "promoter sequence," a double-stranded molecule iε created which will contain a functional promoter, if that extension proceeds towards the "promoter sequence" or the "promoter sequence complement." This functional promoter will direct the transcription of a nucleic acid molecule which is operably linked to that strand of the double-stranded molecule which contains the "promoter sequence" (and not that strand of the molecule which contains the "promoter sequence complement") .

Certain RNA polymerases exhibit a high specificity for such promoters. The RNA polymerases of the bacteriophages T7, T3, and SP-6 are especially well characterized, and exhibit high promoter specificity. The promoter sequences which are specific for each of these RNA polymerases also direct the polymerase to transcribe from only one strand of a duplex DNA template. Strand selection is determined by the orientation of the promoter sequence, and determines the direction of tranεcription εince RNA is only polymerized enzymatically by the addition of a nucleotide 5' phosphate to a 3' hydroxyl terminus.

The promoter sequences of the present invention may be either prokaryotic, eukaryotic or viral. Suitable promoters are repressible, or, more preferably, constitutive. Examples of suitable

prokaryotic promoters include promoters capable of recognizing the T4 (Malik, S. et a . , J. Biol . Chem . 263:1174-1181 (1984); Rosenberg, A.H. et al. , Gene 59:191-200 (1987); Shinedling, S. et a ., J. Molec. Biol . 195:471-480 (1987); Hu, M. et a . , Gene 42:21-30 (1986)), T3, Sp6, and T7 (Chamberlin, M. et al. , Nature 228:227-231 (1970); Bailey, J.N. et a . , Proc. Natl . Acad . Sci . USA 80:2814-2818 (1983); Davanloo, P. et al . , Proc. Natl . Acad . Sci . USA 81:2035-2039 (1984)) polymerases; the P R and P L promoters of bacteriophage λ (THE BACTERIOPHAGE LAMBDA, Hershey, A.D., Ed., Cold Spring Harbor Press, Cold Spring Harbor, NY (1973); Lambda II, Hendrix, R.W. , Ed., Cold Spring Harbor Press, Cold Spring Harbor, NY (1980)); the trp, recA, heat shock, and lacZ promoters of E. coli ; the α-amylase (Ulmanen, I., et al., J. Bacteriol . 162:176-182 (1985)) and the σ-28-specific promoters of B . εubtiliε (Gilman, M.Z., et al . , Gene 32:11-20 (1984)); the promoters of the bacteriophages of Bacilluε (Gryczan, T.J., In: THE MOLECULAR BIOLOGY OF THE BACILLI , Academic Press, Inc., NY (1982)); Streptomyces promoters (Ward, J.M., et al . , Mol . Gen . Genet . 203:468-478 (1986)); the int promoter of bacteriophage λ; the Jbla promoter of the β-lactamase gene of pBR322, and the CAT promoter of the chloramphenicol acetyl transferase gene of pPR325, etc. Prokaryotic promoters are reviewed by Glick, B.R., J. Ind . Microbiol . 1:277-282 (1987)); Cenatiempo, Y. (Biochimie 68:505-516 (1986)); Watson, J.D. et al. (supra) ; and Gottesman, S. Ann . Rev . Genet . 18:415-442 (1984)).

Preferred eukaryotic promoters include the promoter of the mouse metallothionein I gene (Hamer, D., et al., J . Mol . Appl . Gen . 1:273-288 (1982)); the TK promoter of Herpes virus (McKnight, S., Cell

31:355-365 (1982)); the SV40 early promoter (Benoist, C, et al . , Nature 290:304-310 (1981)); and the yeaεt gal4 gene promoter (Johnεton, S.A., et al . , Proc . Natl . Acad . Sci . USA 79:6971-6975 (1982); Silver,

P.A., et al., Proc. Natl . Acad. Sci . USA 81:5951-5955 (1984)). All of the above listed references are incorporated by reference herein.

Strong promoters are preferred. Examples of such preferred promoters are those which recognize the T3, SP6 and T7 polymerases, the P L promoter of bacteriophage λ, the recA promoter and the promoter of the mouse metallothionein I gene. A most preferred promoter for eukaryotic expression of RPTPK is an SV40 promoter such as that driving tranεcription in the pLSV vector (Livneh, E. , et al., (1986) J " . Biol . Chem . 261:12490- 12497). The sequences of such polymerase recognition sites are discloεed by Watson, J.D. et al. (εupra) . Having now generally described the invention, the same will be more readily understood through reference to the following example which is provided by way of illustration, and is not intended to be limiting of the present invention, unleεε specified.

EXAMPLE: ISOLATION AND ANALYSIS OF MURINE RPTPK cDNA CLONES

In an effort to identify new PTPases, a mouse brain cDNA library in λgtll was screened under relaxed stringency conditions using aε a probe an oligonucleotide correεponding to the intracellular two tandem PTPaεe homology domainε of human CD45 (Sap et al . , εupra) . Following initial characterization and claεsification of the isolated clones, several εubεequent roundε of εcreening mouse brain libraries at high stringency yielded a set of cDNA fragments

that together encompassed the entire coding sequence for RPTPK. The relationship between the different RPTPK cDNA clones isolated was confirmed by Northern and reverse transcriptase/PCR analyses (see Materials and Methods section and FIG. 2 for details) .

6.1. LIBRARY SCREENING

The original RPTPK cDNA clone was isolated by low- stringency screening of a λgtll mouse brain cDNA library with a probe consisting of the intracellular domain of human CD45, which contains two tandem PTPase domains (see: Sap, J. et a . , 1990 Proc. Natl. Acad. Sci. USA 87:6112-6116, for details). After initial characterization, one of the isolated clones (λ-50, containing a 935 nucleotide fragment with characteristic homology to members of the PTPase family) , was used to rescreen the same library, yielding clones λ-602 and λ-604. Sequence analysis showed that clone λ-602 was identical to λ-604 at both extremities, but was interrupted by a sequence containing stop codons in all three reading frames. Its analysis was therefore diεcontinued, since it is likely to represent an incompletely spliced RNA species. By contrast, λ-604 appeared to contain one PTPase homology domain and an additional 2042 ht. of upstream coding sequence, including a likely membrane- spanning region. In order to obtain a full length RPTPK CDNA, the entire insert of clone 604 was used to screen another (randomly primed) mouse brain cDNA library (Clontech) , leading to the iεolation of two hybridizing cloneε, λ- 35 and λ-37. Clone 35 appeared to overlap with the N- ter inuε of clone 604 and to encompass the translational initiation codon for the RPTPK precursor

protein (see results section) . Initial sequence analysis of clone 37 however revealed no overlap with the clone 604 probe, although it contained a clear 5 additional PTPase homology followed by a stop codon in a position characteristic for the second PTPase domain of a RPTPase. Several controls were used to show that clone 37 corresponds to the bona fide C-terminus of RPTPK. In Northern analysis, clones 37 and 604 l0 recognize identical mRNA species in all mouse tissues examined.

A reverse transcriptase/PCR analysis on mouse liver poly(A)+ RNA using primers corresponding to^ clones 604 and 37, followed by cloning and sequencing,

15 yielded a fragment of the expected size, exactly joining both clones at the same EcoRI site where each isolated cDNA clone ended.

In retrospect, clone 37 was therefore most likely picked up in the screening with the clone 604 fragment

2o due to the existence of an additional small cDNA fragment in the original λ-37 phage isolate that went undetected due to its small size, or by fortuitous crosshybridization between the two PTPaεe homologieε of RPTPK. A schematic summary of the different cDNA

25 cloneε discussed is included in FIG. 2.

6.2. NUCLEOTIDE SEQUENCE DETERMINATION

cDNA fragments were isolated from phage cloneε, 30 εubcloned into Blueεcript cloning vectors and subjected to sequence analysis by the dideoxynucleotide chain termination method (Sequenase, United States Biochemical) using synthetic oligonucleotide primers. All sequenceε were 35 determined on both strands. Sequences were assembled and analyzed using the GCG 7 software package

(Devereux, J. et al . , 1984 Nuc . Acids Res . 12:387- 395) . The assembled RPTPK cDNA nucleotide sequence was submitted to Genbank under accession number L10106.

6.3. SEQUENCE ALIGNMENTS

All DNA and protein data base searches were done with the Genetic Computer Group sequence analysis software package (Devereux et al ., Nucleic Acid Res . 12:387-396 (1989)). The SwissProt and Gene Bank\European Molecular Biology Laboratory data bases were searched with FASTA and TFASTA, respectively (Pearson and Lipman, Proc . Natl . Acad . Sci . 85:2444- 2448 (1988)). Proteins were aligned with the Genetics Computer Group programs, LINEUP, PILEUP, PRETTY and BESTFIT.

6.4. RESULTS AND DISCUSSION

6.4.1. ISOLATION AND SEQUENCE ANALYSIS OF CDNA CLONES ENCODING MURINE RPTPK

The nucleotide sequence of murine RPTPK (SEQ ID NO:3) is shown in FIG. IA - 1H. The complete amino acid sequences of RPTPK (SEQ ID NO:l) is shown in FIG. IA through 1H and in FIG. 3.

The assembled RPTPK cDNA sequence can be divided into a 5 'untranslated region of 1072 base pairs, a single open reading frame of 4374 base pairs and a 3 1 untranεlated region of 388 baεe pairs. The deduced amino acid sequence of the RPTPK precursor protein is shown in FIG. 3. The translational initiation codon is identified by a standard environment for initiation of tranεlation (Kozak, supra) and by the existence of an upstream in-frame stop codon (position -252), and

is followed by a hydrophobic region that may serve as a signal peptide. A second hydrophobic region is found between amino acid residues 753 and 774 and is followed by a series of predominantly basic residues, characteristic of a stop transfer sequence. These features delineate a putative extracellular region of 752 amino acid reεidueε (including the εignal εequence) , and an intracellular portion of 683 amino acidε. The latter containε the tandem repeat of two PTPase homologies typical for most RPTPaseε iεolated so far (Fischer, E.H. et al. , 1991 Science 253:401- 406) .

An intriguing feature of RPTPK is the extended distance between its trans-membrane segment and the start of the first phosphatase homology domain. This region is about 70 residues longer than in all other previously described RPTPases, with the exception of mRPTPμ (Gebbink et al. , εupra) . Interestingly, a variant of RPTPK was found by the present inventors laboratory to contain a similarly-sized insertion in the εame poεition. It iε conceivable that εuch an inεertion generated by alterative splicing might constitute a separate functional unit in RPTPases.

The first approximately 170 amino acidε of RPTPK εhow similarity (26% overall identity) to a region in the Xenopus cell surface protein A5 with features of Ig-like domainε (FIG. 5) . The A5 protein iε thought to function in recognition between input and target neuronε in the visual system (Takagi, S. et al . , 1991 Neuron 7:295-307) .

This first domain is followed by one Ig-like repeat (approximately residues 210 to 270) and four putative fibronectin type Ill-like (FN-III) repeats (residues 296 to 681) . Database searching revealed

clear similarity of these FN-III domains to similar domains in the tyrosine phosphatases R-PTPμ and LAR, the Droεophila R-PTPases DLAR and DPTP10D, and Droεophila neuroglian (Bieber, A.J. et al. 1989. Cell 59:447-460; Gebbink et al . , εupra ; Streuli, M. et al., 1988, εupra ; Streuli, M. et al . , 1989,supra; Tian et al . , εupra ; Yang et al . , εupra) .

Some other features of the extracellular domain of RPTPK are noteworthy. First, it contains the sequence HAV (amino acids 340-342; within the first FN-III repeat) implicated in cell-cell contact in members of the cadherin family (Blaschuk, O.W. et al., 1990 J.Mol.Biol. 211:679-682). Second, the extracellular domain (640-643) contains the sequence RTKR, a consensus cleavage site for the processing endoprotease furin (Hosaka, M. et al., 1991 J. Biol . Chem . 266:12127-12130). Other potential posttranslational modification sites include 12 potential N-linked glycosylation sites, and SG-motifs which are candidates for chondroitin sulfate attachment (residueε 172, 176, 277, 333, 662) (Kjellen, L. et al. 1991 Annu. Rev. Biochem. 60:443- 470) . Overall, the sequence of RPTPK shows a high degree of sequence similarity to mRPTPμ (77% overall similarity at the amino acid level) (Gebbink et al . , εupra) . The sequence identity between this pair of related R-PTPases is highest in the first PTPase homology domain (80% as compared to 74% identity for the second PTPase domain) . This is in contrast to the situation that haε been observed for the relationship between the closely related pairε of R-PTPaεes LAR and HPTP-5, and RPTPβ/HPTP^ and RPTP7 (Kaplan, R. et a . 1990 Proc . Natl . Acad . Sex . USA 87:7000-7004; Krueger, N.X. et al . , 1990 EMBO J. 9:3241-3252; Streuli, M. et

al. , 1988, εupra) . The latter pairs of related R- PTPaseε are more related in their second PTPase homology domains. The sequence of RPTPK is also highly similar to that of PCR fragment PTP 191-33 described by Nishi, M. et al . , 1990 FEBS Lett. 271:178-180.

7. EXAMPLE: EXPRESSION AND TISSUE DISTRIBUTION OF RPTPK

7.1. TISSUE EXPRESSION AND NORTHERN ANALYSIS

Poly(A)+RNA was isolated from adult mouse tisεueε by oligo(dT) selection as described previously (Vennstrom, B. et al . 1982 Cell 28:135-143). Five μg of poly(A)+ RNA per lane were fractionated on formaldehyde-containing 1% agarose gelε, transferred to Nytran membranes, and probed under high stringency conditions with different regions of the RPTPK cDNA. RNA loading and quality was controlled for by ethidium bromide staining.

7.1.1. EXPRESSION OF THE RPTPK PROTEIN

in order to assemble a full-length RPTPK CDNA from the various isolated fragments, a convenient fragment which included the N-terminus was generated from clone 35 by a PCR reaction using the N terminal primer 5•GAGCCGCGGCTCGAGTTAACCGCCATGGATGTGGCGGCCG3 ' (SEQ ID NO:5) and the C-terminal primer

5•GCTCACAGCTAGTTCAGCCC3' (SEQ ID NO:6). This manipulation also removed all of the 5'untranslated sequenceε, while retaining an optimized Kozak consensus sequence for translation initiation (Kozak, M. 1983 Microbiol . Rev . 47:1-45).

The amplified 470 nucleotide product was digeεted with Sac II and PpuM 1, and cloned between the Sac II and PpuM I sites of clone 604, yielding plasmid pKo (the Sac II site being in the polylinker region of the Bluescript cloning vector). The 1.1 kb Eco Rl fragment from clone 37 (containing the C-terminal end of the coding εequence) was then cloned into the unique and corresponding Eco Rl site of pKo in the appropriate orientation, yielding construct pK, containing the fully assembled coding sequence without the 5' untranslated sequences. The modified cDNA was then released as one fragment using Hpa I (N-terminal) and Xho I (C-terminal) , and cloned between the Sma I and Sal I sites of a CMV-enhancer/promoter-driven eukaryotic expression vector.

7.1.2. GENERATION OF ANTISERA SPECIFIC FOR EPITOPES OF RPTPK

Antigenicity of peptides included in the the RPTPK protein waε predicted uεing the Jameson-Wolf algorithm included in the GCG 7 Peptidestructure program (Devereux, J. et al., 1984 Nucl . Acidε Res . 12:387-395). Two peptides were syntheεized. The peptideε were coupled to the protein keyhole limpet hemocyanin by glutaraldehyde croεεlinking and injected into rabbitε at two week intervals (100 pg per injection) .

The first peptide corresponded to a site near the predicted N-terminus of the RPTPK protein (SEQ ID N0:1), εpecifically, reεidueε 60-76, having the εequence SAQEPHYLPPEMPQGST. Immunization with this peptide yielded antiserum 116.

The second peptide corresponded to a region located at the N-terminus of the first PTPase homology in the intracellular region of the RPTPK protein (SEQ

ID NO:l), specifically, residueε 910 to 929 having the sequence SASWDVAKKDQNRAK. Immunization with this peptide yielded antiserum 122) (FIG. 14).

7.1.3. TRANSFECTION, LABELING AND IMMUNOPRECIPITATION

Subconfluent cultures of COS or HeLa cells in 10 cm diameter dishes (as indicated) were tranεfected by the DEAE-dextran or calcium phoεphate technique, respectively. Between 48 and 72 hours after transfection, the cells were metabolically labeled for 2 hours in methionine-free medium containing 50 μCi/ml [ 35 S]-methionine (ICN) . In the pulse-chaεe analysis shown in FIG. 10, cells were labeled with 200 μCi/ml of the isotope. After labeling, cells were washed in PBS and lysed in Triton-X-100 lysiε buffer (50 mM Hepes pH 7.5, 150 μM NaCl, 1.5 mM MgCl 2 , 1 mM EGTA, 10% glycerol, 1% Triton-X-100, 200 μg/ml PMSF, 10 μg/ml Aprotinin, 10 μg/ml Leupeptin) at 4°C.

Cell lysates were incubated at 4°C for 2 hours with Protein A-Sepharose previouεly preincubated with the reεpective anti-RPTPκ antibody. Where indicated, 20 μg of the antigenic peptide was included in the immunoprecipitation reaction aε a control for specificity. Immunoprecipitates were waεhed with high, medium and low salt buffers (Lev, S. et al., 1991 EMBO J. 10:647-654), with the exception of the experiment depicted in FIG. 12 where washing was with HNTG-buffer (20 mM Hepeε pH 7.5, 150 mM NaCl, 10% glycerol, 0.1% Triton-X-100) . Immunoblotting analyses were performed using standard procedures.

7.1.4. PROTEIN TYROSINE PHOSPHATASE ENZYMATIC ASSAY

Phosphatase enzymatic assays were performed with RPTPK protein immunoprecipitated with antiserum 116 (specific for the extracelular domain) from transiently transfected COS cells. The protein A- Sepharose/RPTPK immunoprecipitated complexes were washed 4 times with HNTG, and once with M7.6 buffer (60 mM Triε, pH 7.6, 5 mM EDTA, 10 mM DTT, 50 mM NaCl, 50 μg/ml BSA) . The enzymatic assay was performed essentially aε described (Streuli, M. et a . , 1989 Proc . Natl . Acad . Sci . USA 86:8698-8702). The immune complexes were resuspended in 50 μl M17.6 buffer (containing 1 mM vanadate where indicated) to which had been added 10 μl [ 32 P]tyrosine phosphorylated myelin basic protein (approximately 12 μM) . The [ 32 P]-tyrosine- phosphorylated myelin basic protein had been produced by in vitro phosphorylation using EGF-receptor immunoprecipitated from A431 cells. The reactions were incubated for 15 minuteε at 37°C with shaking, stopped with 750 μl of an acidic stop mix containing activated charcoal, and the amount of releaεed free [ 32 P]-phosphate was meaεured.

7.1.5. ENDOGLYCOSIDASE F TREATMENT

Cultureε of cellε tranεfected with RPTPK cDNA were lyεed in 1% SDS at 100°C for 3 minutes. The total cell lysateε were εonicated 3 timeε at full εpeed, then diluted with diεtilled water to decrease the concentration of SDS to 0.1%. The cell lysateε were incubated at 37°C for 18 hours in the presence of 0.2 unitε endoglycoεidase F (Boehringer- Mannheim), 0.25 M sodium acetate, pH 5.2, 20 mM EDTA, 10 mM β- mercaptoethanol and 0.6% NP-40. The total enzyme- treated lysate was directly loaded onto SDS-PAGE gels,

which were run, transferred to nitrocellulose and blotted with antiserum 116 or antiserum 122 as indicated.

7.1.6. SITE-DIRECTED MUTAGENESIS

In vitro site-directed mutagenesiε waε performed uεing a commercially available kit from Clontech, uεing the manufacturer'ε instructions. An oligonucleotide having the sequence

CTACACCCACATCTAACGAACCGTGAAGCAGGG (SEQ ID NO:7) was used to modify the amino acid sequence RTKR in the cleavage site to the sequence LTNR. Mutagenesiε waε confirmed by direct DNA εequencing.

7.1.7. IN SITU HYBRIDIZATION OF RPTPK cDNA TO RAT TISSUES

Sprague-Dawley ratε were εacrificed by decapitation, and their brainε were removed and εectioned into 20 μm sections in a cryostat. Sections were postfixed in 4% paraformaldehyde in 0.1 M sodium phosphate, pH 7.4, for 20 min.

A 50 base oligonucleotide complementary to residues 1493-1543 of the isolated RPTPK CDNA sequence (SEQ ID NO:3) was uεed as a probe. The oligonucleotide was labeled with [ (α- 35 S]dATP (NEN, DuPont) using terminal deoxynucleotidyl transferase (Boehringer Mannheim) and purified using Sephadex G25 quick spin columns (Boehringer Mannheim) . The specific activity of the labeled probe was from 2 x 10 8 to 5 x 10 8 cpm/μg DNA. Prehybridization and hybridization were carried out in a buffer containing 50% deionized formamide, 4X SCC, IX Denhardt'ε solution, 500 μg/ml denatured salmon sperm DNA, 250 μg/ml yeast tRNA and 10% dextran sulfate.

The tissue sections were incubated in a humidified environment for 14-18 h at 42-46°C in hybridization solution containing the labeled probe and 10 mM dithiothreitol. Specificity controls were performed on adjacent sections by adding to the labeled oligonucleotide a 100-fold excesε of the unlabeled oligonucleotide. After incubation, sectionε were washed in 2 changes of 2X SSC at room temperature 0 for 1 h, then in IX SCC at 50°C for 30 min, 0.5X SCC at 50°C for 30 min, and in 0.5X SCC at room temperature for 10 min. Sections were dehydrated and exposed to X-Omat film for 3 weeks.

5

7.2. RESULTS AND DISCUSSION

7.2.1. EXPRESSION OF RPTPK IN ADULT TISSUES Northern blot analysis on adult mouse tisεueε o (FIG. 6) revealed that RPTPK expreεsion is broad. Two major transcripts of 5.3 and 7.0 kb were detectable at different levels in all examined tissueε except in εpleen and testiε. Particularly high levelε of the 5.3 kb transcript were seen in liver and kidney 5 tissue. An identical pattern was detected using as a probe both an N-terminal and central part of the cDNA. Although the 5.3 kb size is similar to the 5.7 kb described for mRPTP (Gebbink et a . , supra), RPTPK appears to be much more widely expresεed than mRPTPμ. 0 Expression of the latter is virtually reεtricted to lung and, at lower levels, brain and heart.

7.2.2. TRANSIENT EXPRESSION AND ENZYMATIC ACTIVITY OF RPTPK 5

As described above, the RPTPK coding sequence waε cloned into an expression vector under the control of

the CMV enhancer and promoter after manipulation to remove the untranslated leader sequence. The construct was transiently transfected into HeLa cells which were metabolically labeled with [ 35 S]-methionine, lysed and subjected to a radioimmunoprecipitation assay. The antibody probe was an antiserum raised againεt a peptide located in the N-terminus of the protein (residueε 60 to 76) . This antiserum precipitated a protein of about 210 kDa from RPTPK transfected cells, but not from mock transfected cells (transfected with an "empty" expression vector) (FIG. 7) . This immunoprecipitation was blocked by inclusion of the antigenic peptide in the immunoprecipitation reaction (lanes 3 and 6, wherein lane 1 is the left¬ most lane) , but not by inclusion of a heterologous peptide corresponding to the firεt catalytic homology domain of RPTPK.

To confirm that the protein encoded by the RPTPK cDNA had PTPaεe enzymatic activity, immune complexes from transfected cells were incubated in an appropriate buffer with [ 32 P]-tyrosine phosphorylated myelin basic protein as a εubεtrate. As shown in FIG. 8, approximately 3-fold higher PTPase activity was detectable in immune complexes from RPTPK transfected cells as compared to control cells. This PTPase activity could be significantly inhibited by vanadate.

7.2.3. IN SITU HYBRIDIZATION ANALYSIS OF RPTPK EXPRESSION IN THE DEVELOPING AND ADULT CENRAL NERVOUS SYSTEM

The level of expression of RPTPK mRNA was generally higher in the developing than in the adult central nervous syεtem (CNS) . At embryonic day 18 (E18) and at E20, the RPTP(κ) mRNA levels were highest in the cerebral cortex and hippoca pal formation,

followed by the cerebellum, brain stem and spinal cord. In the rest of the embryo, the highest levels were found in the liver, kidney and intestine (FIG. 13A) . At postnatal day 6 (P6) and P8, expression was maximal in the cortex, olfactory bulb and hippocampal formation, especially in the dentate gyrus. and CA3. In the cerebellum, the expression was highest in the granular cell layer, which in this stage of development still occupieε the outermoεt cell layer of the cerebellum (FIG. 13B) .

In the adult rat, expression was lower, but was clearly visible in the olfactory bulb and throughout the cortex, including the pyriform and cingulate cortex. Expresεion of the RPTPK mRNA was also observed in the hippocampal formation. Interestingly, expression in the cerebellum was barely detectable in the adult. This was in marked contrast with the distinct patten and high level of expreεsion observed at P6 and P8, a period of active cerebellar development.

The in situ hybridization studies confirmed the expression of the RPTPK in several organs. In addition, they demonstrated that, in the CNS, RPTPK is expressed in specific areas in a developmentally regulated manner. The levels of RPTPK expression are higher in the actively developing areas, but expression persiεts in the adult, mainly in certain areas of the cortex and in the hippocampal formation. These findings are consistent with the idea that CNS RPTPaseε are actively involved in development and plasticity. Studies on the expression of RPTPs in Droεophila have led to similar suggestions (Tian et al . , εupra ; Yang et al . , supra) .

8. EXAMPLE: CHROMOSOMAL LOCALIZATION OF THE MURINE RPTPK GENE .

The method was essentially as described previously (Sap, J. et al . , 1990 Proc . Natl . Acad . Sci . USA 87:6112-6116; Silver, J., 1985 J. Hered. 76:436-440; Taylor, B. , 1978, In: H.C. Morse, III (ed.), ORIGINS OF INBRED MICE, Academic Press, New York, pp. 423-438; Taylor, B.A. , 1989 In: M.F. Lyon et al., eds, GENETIC VARIANTS AND STRAINS OF THE LABORATORY MOUSE. Oxford Univerεity Press, New York, pp. 773-796) . Southern blotting analysis of Taq I- digeεted mouεe genomic DNA with the 604 RPTPK probe revealed an array of 12 fragmentε that appeared invariant between the inbred strains surveyed, as well as a smaller set of variable bands that were used to define two allelic forms of the gene:

(1) a was defined by the preεence of 1.9, 3.5 and 3.8 kb fragments and was preεent in inbred mouse strains AKR/J, C3H/HeJ, DBA/J, SM/J; and

(2) b was defined by the presence of a 4.1 kb fragment and was present in inbred mouse strains C57BL/6J, 020/A, C57L/J, S R/J, SJL/J, BALB/cJ, STS/A, NZB/B1NJ) .

Analysis of the inheritance pattern of this variant among recombinant inbred strains of mice (Table I) , and comparison of strain distribution patterns thus obtained with those generated previously for other genetic markers, revealed close linkage between RPTPK and two markers of proximal chromosome 10: D10Mit3 ( 2 discordancies among 22 εtrainε typed, indicating a diεtance of 2.6 cM between the loci (0.3 cM to 13.0 cM defined 95% confidence limitε) ; and Ly- 41 (0 discordancies among 30 strains typed, indicating a distance between the loci of < 3.5 cM at 95% confidence) . The gene symbol Ptpk is proposed by the

inventorε, conεiεtent with the symbol Ptpa previously asεigned to RPTPα (Sap et al . , εupra) .

Thiε region of mouεe chromosome 10 contains multiple genes with human horoologueε mapping to chromosome 6q. Based on synteny, this would predict a localization of the human RPTPK homologue to 6q, in contrast to I8pter-qll for human RPTPμ (Gebbink et al . , εupra) .

TABLE I

DNA FRAGMENT LENGTH VARIANT ASSOCIATED WITH THE MODSE RPTPK GENE.

A. Alleles and strain distribution patterns

Allele (Size kb) ) Strains a 1.9 + 3.5 + 3.8 AKR/J, C3H/HeJ, DBA/2J, SM/J

AKXL-5,6,7,8,17,21,25,28,29, 37,38

BXD-1,2,5,14,15,18,21,23,25,

28,32 BXH-2,4,7,8,12,14,19 NXSM-D,L, ,X OXA- ,5,7,8,13 b 4. 1 C57BL/6J, 020/A, C57L/J, SWR/J,SJL/J, BALB/CJ, STS/A,

NZB/B1NJ

AKXL-9, 12,13,14,16,19,24 BXD-6,8,9,11,12,13,16,19,20,

22,24,27,29,30,31 BXH-3,6,9, 10, 11 NXSM-C,E,F,I,N,P,Q,T1,T2,U,Z OXA- 1,2,3,6,9,10,11,12,14

B. Linkage of ptpk to other markers typed in Recombinant Inbred strains

Marker Chr R/N Odds Distance (CM) D10Mit3 10 2/22 0.00941 2.6 (0.3-13.0) Ly-41 10 0/30 <0.00001 0.0 (<3.5)

A) 10 μg quantities of liver or spleen genomic DNA were digested with Taql enzyme and analyzed by Southern blotting with the 604 RPTPK probe as described previously to define two alleles of the ptpk gene and to follow their inheritance in panels of recombinant inbred (Rl) strains of mice.

B) The resulting strain distributions were compared with those previously determined for other loci in these panels of mice. Two matches were found that were unlikely to be due to chance at a 5% confidence level. For each of these, the number of non- matching Rl strains found (R) is shown as a fraction of the total number of Rl strains typed (N) for the two markers, together with the odds of observing that number of non-matches or a smaller one by chance (Blank, R.D. et al . , 1988 Genetics 120:1073-1083), the estimated distance between the marker and ptpk, and the 95% confidence limits for that estimate (Silver,supra; Taylor, 1978, supra ) .

9. EXAMPLE: POSTTRANSLATIONAL

PROTEOLYTIC PROCESSING OF RPTP*

During experiments designed to achieve stable expression of RPTPK in 3T3 cells, the present inventors observed the generation of a product of an unexpected, smaller size as well as the generation of aberrantly-sized products upon transient transfection 0 of COS cells.

The present inventors noted the presence of a proteolytic cleavage signal in the extracellular domain of RPTPK, (RTKR, residues 640 to 643, in the fourth FN-III repeat; FIG. 3) and wished to examine 5 its significance in light of these observations. Thus, additional experiments were performed in COS cells transfected by the DEAE-dextran technique.

In order to detect cleavage products which may o have accumulated, total cell lysates were directly loaded onto SDS-PAGE gels, run in electrophoresis, transferred to nitrocellulose, and immunoblotted with the two different anti-RPTPκ peptide antisera (described above) specific for either the N-terminus 5 or for an epitope near the first PTPase homology domain in the intracellular portion.

In lysates from transfected cells, but not from mock transfected cells, both antisera recognized the same 210 kDa protein described above. The antiserum 0 specific for the N terminus also recognized a smaller 110 kDa protein. The antiserum specific for the cytoplasmic region recognized a smaller 100 kDa protein (FIG. 9) .

The three polypeptides (210, 110 and 100 kDa) 5 were further characterized by subjecting the total cell lysates to endoglycosidase F digestion before

SDS-PAGE and immunoblotting. Such a treatment would be expected mainly to affect the mobility of a protein containing the glycosylated extracellular domain. Following endoglycosidase F treatment, the mobility of the 210 kDa and 110 kDa proteins was significantly reduced, to 160 kDa and 89 kDa respectively. In contrast, the mobility of the 100 kDa band detected with antiserum 122 specific for an epitope in the intracellular domain) was only slightly affected, suggesting that the 100 kDa peptide includes a minor glycosylation component (FIG. 9) .

The above results, as well as pulse-chase analysis shown in FIG. 10, are consistent with the cleavage of a 210 kDa RPTPK precursor protein into an N-terminal 110 kDa product encompassing most of the extracellular domain, and a 100 kDa moiety containing the intracellular portion and about 100 residues of extracellular sequence (FIG. 14) . A consensus site for cleavage by furin, a processing endopeptidase (Hosaka et al . , εupra) , is indeed located 113 amino acids upstream of the start of the transmembrane segment (RTKR, residues 640-643), which would leave one potential N-glycosylation site in the C-terminal cleavage product.

In order to confirm directly that proteolytic cleavage occurred at the RTKR (furin-recognized) site, site-directed mutagenesis was used to mutate this site to LTNR, and the effects of this mutation on the processing of the RPTPK precursor was examined. As shown in FIG. 12, the mutant cDNA gave rise to only a 210 kDa product. These results provide evidence that the RTKR region is the likely proteolytic cleavage signal and site for processing, of the RPTPK proprotein.

The inventors next tested whether the cleavage products were associated. This was accomplished by performing an immunoprecipitation with antiserum 116, 5 specific for the extracellular 110 kDa product, on lysates of cells transfected with the wild type (wt) RPTPK cDNA. Immunoblotting of this precipitate with antiserum 122, specific for an intracellular RPTPK epitope, detected the presence of the 100 kDa C- ° terminal cleavage product in the precipitate (FIG. 12) . This observation strongly suggested that at least a portion of the two RPTPK cleavage products remained associated after cleavage, and that the 100 and 110 kDa species may be considered as subunits of a 5 single complex (FIG. 14) . Experiments with a secreted form of the extracellular domain of RPTPK suggested that this association was not mediated by a disulfide linkage, since no association could be detected using SDS-PAGE under nonreducing conditions. 0

A similar posttranslational processing event has been described for the RPTPase LAR and for the Ng-CAM protein (Burgoon, M. et al . 1992. J. Cell Biol. 112:1017-1029; Streuli, M. et al . , 1992 EMBO J. 5 11:897-907; Yu, Q. et al . , 1992 Oncogene 7:1051-

1057) . In addition, a potential cleavage site exists in the corresponding position in mRPTPμ (Gebbink et al., εupra) . It is therefore likely that proteolytic processing of RPTPs may be a more general phenomenon. 0 Such cleavage, as described above, may allow controlled shedding of the N-terminal 110 kDa subunit, and thus render the membrane-bound 100 kDa form of RPTPK insensitive to modulation by binding of proteins in the cellular environment. Alternatively, shedding 5 might release a soluble species which retains binding activity to the putative RPTPK ligands.

Interestingly, secreted soluble forms of extracellular domains have been described for tyrosine kinase receptors such as the FGF-receptor (Johnson, D.E. et 5 al . , Molec . Cell . Biol . 11:4627-4634 (1991)). However these secreted forms were generated by an alternative splicing mechanism.

10. EXAMPLE: ISOLATION AND ANALYSIS OF 0 HUMAN RPTP* (KCP1 ) cDNA CLONES

10.1. PCR AND cDNA CLONING METHODS

Poly(A)+ RNA was isolated from SK-BR-3 cells 5 (ATCC HTB30) and cDNA synthesized using avian myeloblastosis virus (AMV) reverse transcriptase as described (Sambrook, J. et al . , MOLECULAR CLONING: A LABORATORY MANUAL, 2nd Edition, Cold Spring Harbor Press, Cold Spring Harbor, NY (1989)). Polymerase o chain reaction using a pool of degenerated oligonucleotides based on two highly conserved regions of the PTPase domain (Vogel, W. et al. , Science 259:1611-1614 (1993) was performed under standard conditions, and PCR products were subcloned in 5 Bluescript KS+ vector (Stratagene) . Sequence analysis was done by the dideoxynucleotide chain termination method (Sanger et al., 1977) using Sequenase (United States Biochemical) . a lambda ZAP 11 LIBRARY (Stratagene) from SK-BR-3 poly(A) + RNA was screened Q with a PCR fragment probe under high stringency conditions (Ullrich, A. et al . . 1985, Nature 313:756- 711) .

5

10 . 2 . RESULTS

The complexity of PTPases expressed in the human breast cancer cell line SK-BR-3 was examined by performing a PCR analysis. The primers were degenerate sequences corresponding to conserved sequences within the PTP catalytic domains shared by all identified PTPases (Vogel, . et al . 1993, Science 259:1611-1614) . Sequence analysis of the cloned PCR products revealed the presence of several known PTPases, including PTP1B, LAR, TC-PTP, PTP-S, PTPe, PTPγ, and PTPH1, as well as some novel members of the PTPase family. One of the novel sequences, designated MCP7 ("mammary carcinoma-derived PTPase, clone 7") , was highly represented (18%) in the 121 clones examined. The 2066 bp MCP7 PCR fragment was used to screen a λ ZAP II SK-BR-3 cDNA library at high stringency. Eleven overlapping clones spanning an overall region of approximately 6.1 kb were analyzed, revealing an open reading frame encoding 1444 amino acids, followed by a 3 ' untranslated region of 1.8kb.

The nucleotide sequence of human RPTPc (SEQ ID NO:4) is shown in FIG. 15A-E. The deduced amino acid sequence of MCP7 (SEQ ID NO:2) is also shown in FIG. 15A-E and displays the structural organization of a type II transmembrane PTPase (Fischer et al., 1991, Charbonneau, H. et al . , Annu . Rev . Cell Biol . 8:463- 493 (1992) . The N-terminal hydrophobic stretch of 20-26 amino acids is typical of signal peptides (von Heijne, G., J . Mol . Biol . 184 : 99-105 (1985). A second region consisting of hydrophobic residues is found between positions 755 and 774 and is predicted to be a single α helical transmembrane domain. It is followed by a short region of mainly

basic residues characteristic of a transfer stop sequence ( ickner, .T. et al. , Science 230:400-406 (1985)). The amino-terminal portion of the putative extracellular domain contains a sequence motif, a so called MAM domain, spanning a region of about 170 residues. The MAM structural motif was recently established by comparison of several functionally diverse receptors (including RPTPμ and the A5 protein) and is thought to play a role in cell adhesion

(Beck ann et al . , supra) . This motif is followed by one possible Ig-like domain (residues 207-277) . The remaining extracellular portion contains conserved sequence motifs, indicating that it is composed of four FN-III related domains corresponding to the FN- III-like domains of LAR, PTP/3 and RPTPμ. The extracellular domain contains 12 potential N- glycosylation sites, indicating that MCP7 is highly glycosylated. Interestingly, MCP7 contains the motif RXR/LR (residues 640-643) within the fourth FN-III domain. This motif has been described as the cleavage site for the subtilisin-like endoprotease, furin (Barr, P.J., Cell 66 : 1-3 (1991); Hosaka et al . , supra) . The cytoplasmic part of MCP7 is composed of two PTPase domains containing the conserved amino acid sequences typical of all known PTPases (Saito, H. et al . , Cell Growth Diff . 2:59-65 (1991)) . A particularly intriguing feature is the region linking the transmembrane domain to the amino-terminal PTPase domain, which is nearly twice as large as that of most other receptor-like PTPases. A similar extended distance is shared only by the homologous PTPase, hRPTPμ (FIG. 16A-B, lower lines) . The overall ho ol- °gy between MCP7 and hRPTPμ is 77%, to which the N-

terminal and C-terminal PTPaεe domainε contribute 91% and 86%, respectively (FIG. 16).

10.3. DISCUSSION

The extracellular domain of MCP7 iε composed of one MAM domain, which is a sequence motif spanning about 170 residueε, which waε recently established by comparison of several functionally diverse receptors (including RPTPμ and the A5 protein) and iε thought to play a role in cell adhesion (Beckmann & Bork, 1993, TIBS 18:40). The extracellular domain of MCP7 further includes one Ig-like, and four FN-type Ill-like segments. It therefore shares structural features with some cell adhesion molecules, permitting the classification of MCP7 into the type II PTPase class. MCP7 is highly homologous to mRPTPμ which has a more restricted expression pattern in lung, brain and heart (Gebbink et al . , εupra) . MCP7 is expressed aε a molecule consisting of two noncovalently linked subunits, a structural feature already shown for LAR. A similar procesεing motif waε alεo determined within the extracellular domain of mRPTPμ (RTKR residues 632- 635) , which suggest that thiε structural organization is typical for the family of type II phosphatases. Proteolytic cleavage also occurs in the extracellular domain of the cell adhesion molecule Ng-CAM in a region containing the dibasic processing motif (Burgoon, M.P. et al . , J. Cell . Biol . 112:1017-1029 (1991)). The functional significance of thiε εtructure is not yet clear. For LAR, a shedding of the extracellular E-subunit was observed in a density- dependent manner (Streuli et al . , εupra ) . It iε likely that this shedding is due to a conformational change in the extracellular domain caused by

homophilic or hydrophilic interactions between the moleculeε on the surface of neighboring cells that weakens the interaction between the noncovalently linked subunits. The effect of this shedding on the activity of the PTPase domains within the cellε iε not yet clear, but a modification of the activity of the phoεphataεe or a change in the sensitivity to modifying processes is probable.

11. EXAMPLE: TISSUE DISTRIBUTION OF HUMAN RPTPK

11.1. RNA EXTRACTION AND NORTHERN BLOT ANALYSIS

Total RNA was isolated by the guanidinium isothiocyanate method (Chirgwin et al. , 1979, Biochemistry 18.:5294-5299) from human tiεsue and cultured cells grown to confluency. Poly(A)+ RNA was prepared on an oligo(dT) column (Aviv & Leder, 1972, Proc. Natl. Acad. Sci. USA £9:1408-1412) . 4μg of poly(A)+ RNA waε fractionated on a 1.2% formaldehyde- agaroεe gel and subsequently transferred to nitrocelluloεe filterε (Schleicher & Schuell) .

Hybridization waε performed in 50% formamide, 5x SSC, 50mM NaH 2 P0 4 , pH 6.8, 0.5% SDS, 0.1 mg/ml sonicated salmon sperm DNA, and 5x Denhardt solution at 42°C overnight with 1-3 x 10 6 cpm/ml of 32 P- labeled random- primed DNA (United States Biochemical) . Filters were washed with 2x SSC, 0.1% SDS, and 0.2x SSC, 0.1%SDS at 45°C,and exposed 5 days using an intensifying screen at -80°C.

11.2. RESULTS

Northern blot analysiε revealed a broad tiεεue diεtribution of MCP7 (FIG. 17) . The 6.7 kb tranεcript waε found at elevated levels in lung and colon tissue, and, to a lesser extent, in liver, pancreas, stomach, kidney, and placenta. No tranεcript waε detected in εpleen tissue.

The expression pattern of MCP7 in different mammary carcinoma-derived cell lines is shown in FIG. 18• Although MCP7 expresεion waε observed in all of the cell lines teεted, the quantity of tranεcripts differed significantly. A second transcript with a size of 4.9 kb was also detected in all cell lines displaying a strong signal. Moreover, MDA-MB-435 cells contained a specific l.4kb mRNA that hybridized with the MCP7 probe. It is interesting to note that the intensity of the Northern hybridization signals shown in FIG. 18 correlate with the abnormal over expression of EGF-R and HER2/neu RTKs. Expression of MCP7 was also detected in human melanoma cell lines and some colon-carcinoma derived cell lines.

12. EXAMPLE: TRANSIENT EXPRESSION OF HUMAN RPTPx

12.1. METHODS

MCP7 cDNA was cloned into a cytomegalovirus early promoter-based expresεion plaεmid (pCMV) . The RTK expression plasmidε used were described previously (Vogel, W. et al . . 1993 Science 259:1611-16141. At 30 hours prior transfection, 3xl0 5 cells of human embryonic kidney fibroblast cell line 293 (ATCC CRL 1573), grown in Dulbecco'ε modified Eagle'ε medium (DMEM) which included 4500 mg/1 glucose, 9% fetal calf serum, and 2mM glutamine, were seeded into a well of a six-well dish.

Transfections with CsCl-purified plaεmid DNA were then carried out using the calcium phosphate coprecipitation technique according to the protocol of Chen and Okayama (Chen, C. and Okayawa, H. , 1987, Mol. Cell. Biol. 7:2745-2752) with a total amount of 4μg, which included only 0.2μg expresεion plasmid and complemented with empty vector DNA (Gorman, CM. et al . . 1989, Virology 171:377-385; Lammers, R. et al . . 1990, J. Biol. Chem. 265:16886-16890) . At 16 hours after transfection, cells were washed once and starved with medium containing 0.5% fetal calf serum.

For metabolic labeling, MEM containing Earle'ε salt, but lacking L-methionine, waε used instead of DMEM. [ 3S S] methionine at 40 μCi/ml (1,000 Ci/mmol) was added.

Cells were stimulated with an appropriate ligand for 10 min. Epidermal growth factor (EGF) at 100 ng/ml was used to stimulate cells transfected with EGF-R, HER1/2, EK-R or EP-R. Insulin at 1 μg/ml was used to stimulate cells transfected with IR. SCF at 100 ng/ml was used to stimulate cellε transfected with pl45 c " it . After stimulation, cells were lysed in 200 μl lysis buffer (50mM HEPES, pH7.5; 150mM NaCl, 1.5mM MgCl 2 , ImM EGTA, 10% glycerol, 1% Triton X-100, 2mM phenylmethylsulfonylfluoride, lOμg/ml aprotinin, ImM Na-orthovanadate) . The lysateε were precleared by centrifugation at 125,000 x g for 10 min at 4°C, and 1/10 of the volume of the supernatant was mixed with SDS sample buffer.

Proteins were separated by SDS-PAGE and transferred to nitrocellulose membranes. For detection of phosphotyrosine and protein antigens on immunoblotε, the ECL εyεte (Amersham) in conjunction with horseradish peroxidase-conjugated goat anti-mouse or goat anti-rabbit antibody (Biorad) was uεed. In

order to reprobe with other antibodies, blots were incubated for 1 hour in 67mM Tris-HCl (pH 6.8), 2% SDS, and 0.1% 3-mercaptoethanol at 50°C. For immunoprecipitation, radiolabelled cells were incubated with antiserum at 4°C for 2 hours, washed three times with PBS (15mM NaCl, 3 mM Kcl, 80 mM Na 2 HPO 4 «H 2 0, 1.5mM KH 2 P0 4 , pH 7.4) to remove unbound antibodies, lysed, and precleared by centrifugation. Protein A-sepharose (Pharmacia) in a volume of 20 μl has added and incubated for two hours on a rotating wheel at 4 β C. Precipitates were washed four times with HNTG-buffer (20mM HEPES, ph. 7.5, 150 mM Na CI, 0.1% Triton X-100,. 10* glycerin), SDS-sample buffer added, and SDS-PAGE was performed. X-ray film was then exposed to the dried gel two days.

The polyclonal antiserum, Ab 116, specific for the extracellular domain of murine RPTPK, was raised against a peptide sequence (residueε 60-76) within the extracellular domain of the mouεe homolog of MCP7, and which was perfectly conserved in the human sequence aε deεcribed εupra. in Section 10. The monoclonal antibody specific for phosphotyrosine, 5E.2, was described previously (Fendly, B.M. et al . . 1990, Cane. Res. 50:1550-1558).

12.2. RESULTS

Forty eight hourε after tranεfection of MCP7 cDNA, using a cytomegalovirus promoter-based expresεion vector, into 293 embryonic kidney cellε, radiolabelled cells were incubated with Ab 116. Cells were washed, lyεed, and the antibody-bound material was immunoprecipitated.

MCP7 expression was found on the cell surface only, and appeared as a band having an apparent molecular weight of 185 kDa. The larger size than the calculated molecular weight of 163 kDa was probably due to extenεive glycoεylation of the extracellular domain.

Two additional bands of 97 kDa and 116 kDa were immunoprecipitated (FIG. 19A, lane 1) ; these bands were not detectable in cells tranεfected with a control vector. Such lower molecular weight productε were thought to be cleavage products εince the extracellular domain contains a common cleavage motif (RXR/LR; reεidueε 640-643, FIG. 15A-E) . for proceεεing by the endoproteaεe furin. Theεe productε are εimilar to the cleavage products described above for murine RPTPK. Furthermore, similar procesεing of the extracellular domain of LAR haε been deεcribed (Streuli et al . , εupra) . The 116 kDa fragment, the ct subunit, representε most of the extracellular domain and is highly glycoεylated, aε indicated by the broadneεs of the band upon polyacrylamide gel electrophoreεis analyεiε and itε apparent molecular weight, which exceeded the calculated value, based on the sequence between residing 20 and 639, by 47kD. The 97 kDa fragment, the β subunit, correspondε to an intracellular and tranεmembrane domain and alεo includes a εhort extracellular εegment which is thought to interact with the α εubunit. The relatively minor diεcrepancy between the obεerved 97 kDa molecular weight size and the calculated 91.4 kDa molecular weight of the β subunit can be explained by the presence of only one potential N-glycosylation site. The α and β subunit are believed to form a stable complex, εuch that immunoprecipitation by an antibody

εpecific for the extracellular domain would detect both subunits. To confirm that the 116 kDa band corresponded to the α subunit cleavage product and not merely to a non-specifically cross-reacting εpecieε, lysates from MCP7 cDNA-transfected 293 cells were εubjected to eεtern blots using antiserum 116 specific for an N-terminal epitope. With thiε approach, a band of about 116 kDa aε well as an unprocessed precursor were found (FIG. 19B, lane 1) , neither of which were detected in 293 cells at comparable levels transfected with a control vector (FIG. 19B, lane 2) .

13. EXAMPLE: EXAMINATION OF PTPase

ENZYMATIC ACTIVITY OF HUMAN RPTP/c

To prove that the RPTP/c designated MCP7 is indeed a PTPase enzyme, the above tranεient expression system in 293 cells was used.

Coexpresεion of MCP7 with a panel of different RTKε repreεenting different structural subclasεeε allowed the examination of more phyεiological εubεtrateε for the PTPaεe aε dephoεphorylation targetε than thoεe commonly uεed.

To ensure that the protein localized mainly in the membrane and to avoid an overload of the cell transport syεtem, theεe tranεfection experiments were performed with only small amounts of plasmid compared to the original protocols (Gorman, CM. et al . , Virology 171:377-385 (1989); Lammers, R. et al . , J. Biol . Chem . 265:16886-16890 (1990)) . The receptors tested were mainly chimeric receptors, the respective kinase function of which was under the control of an EGF-R extracellular domain (Lee, J. et al . , EMBO J. 8:167-173 (1989); Herbst, R. et al . , J . Biol . Chem . 266:19908-19916 (1991); Seedorf, K. et al . , J . Biol .

Chem . 266:12424-12431 (1991)) . Human 293 fibroblasts were transfected with equal amounts of expresεion plaεmidε encoding for an RTK and either MCP7 or a control vector. After stimulation with the appropriate ligand for the RTK, cells were lysed, equal aliquots were resolved by SDS PAGE, and the phosphotyrosine level of the receptors was examined by immunoblotting with the anti-phosphotyrosine antibody 5E2 (Fendly, G.M. et al . , Cane . Res . 50:1550-1558 (1990)) .

Co-expresεion of I-R, EGF-R, EP-R, EK-R, and SCF- R/c-kit with MCP7 reεulted in a marked decreaεe in theligand-induced receptor phoεphotyrosine content when compared with control transfections in which MCP7 expression plasmid had been omitted (FIG. 20A, lanes 1 and 9; FIG. 2OB, lanes l, 5, and 9). In contrast, HER1-2 appeared to be a poor substrate of MCP7, since only weak reduction of the ligand-induced phosphorylation state of this chimera was observed (FIG. 20A, lane 5) . Interestingly, the intracellularly localized, incompletely processed precursor forms of I-R, EGF-R and EP-R (FIG. 20A, lanes 2, 4 and 10, 12; 20B, lanes 2, 4), as well aε that of HER 1-2 (FIG. 20A, lanes 6, 8), were efficiently dephosphorylated) , εuggesting that MCP7 was preεent and active in the εa e intracellular compartmentε as the co-expressed RTKs before reaching the cell surface. To verify the above effects and to rule out differences in RTK expresεion levels, the above blots were re-probed with RTK-specific and RPTP/c-specific antibodies. The results indicated that expression levels of the various RTKs were equivalent.

14. EXAMPLE: CORRELATION BETWEEN HUMAN RPTP/c EXPRESSION AND CELL DENSITY

The presence of motifs in the extracellular domain of human RPTP/c that resemble motifs found in proteins involved in cell-cell and cell-extracellular matrix interactions prompted an investigation of the effect on expression level of cell density in culture.

An equal number of SK-BR-3 cells was distributed onto either one, two, or four 15-cm dishes and incubated for two days under standard growth conditions. When harvested after two days, cells seeded at the various starting densities were found to be 100%, 70%, and 40% confluent, respectively. Poly(A)+ RNA was prepared and Northern blot analysis was conducted as described supra , in Section 11.1, using a probe corresponding to the extracellular domain of MCP7. The results indicated that the level of MCP7 transcriptε increased with increased cell density (FIG. 21A) .

To determine whether this effect was unique to SK-BR-3 cells, an identical experiment was performed uεing the colon carcinoma-derived cell line HT 29. Expreεsion of MCP7 mRNA was also found to be density- dependent with these cells (FIG. 19B) .

As a control, the expreεεion of mRNA encoding the enzyme GAPDH waε examined in the above cells at various densitieε. No density dependence of the expression of these transcriptε were obvεerved.

The above reεults support the hypothesis RPTP/c, and other RPTPs of the type II and type III families, are involved in, and modulated by, cell adhesion events (Charbonneau et al . , supra) . PTPases appear to be involved in events leading to growth arrest by cell-cell contact (Klarlund, supra) . The presence of orthovanadate, a potent inhibitor of phosphatase

activity dimishes normal contact inhibition of 3T3 cellε. Furthermore, PTPase activity associated with the membrane fraction of 3T3 cells increased 8 fold when cellε were grown to a higher denεity (Pallen, CJ. et al . , Proc . Natl . Acad. Sci . USA 88:6996-7000 (1991)).

The combination of CAM motifs in the extracellular domain or RPTP/c and the intracellular PTPase activity indicates that RPTPc may act as an important mediator of events asεociated with arrest of cell growth. The structural features of human RPTP/c described above, the density-dependent upregulation or its expression, and its potent activity in dephosphorylating RTKs supports the emerging picture of the pivotal role of RPTP/c in growth arrest through contact inhibition, as well as a role as a tumor suppressor gene.

15. Example: Homophilic Binding by a Receptor Tyrosine Phoεphataεe

The present work investigates whether, similar to

"clasεical" memberε of the CAM family, RPTPaεeε might be capable of homophilic intercellular interaction

(Q. Yu, T. Lenardo, R.A. Weinberg, Oncogene 7, 1051

(1992)). Reaεoning that analyεiε of cell adheεion by the RPTPaεe RPTP/c would be facilitated by itε ectopic expression in a cell line likely to lack conserved ligandε for a mammalian RPTPase, we stably introduced an RPTP/c cDNA into Droεophila S2 cellε. These cells have a very low capacity for spontaneouε aggregation or adheεion, making them an ideal and eεtablished system for such studies (H. Kramer, R.L. Cagan, S.L.

Zipurεky, Nature 352, 207) . Cells transfected with a vector containing the RPTPK cDNA in the sense orientation with respect to the heat-shock protein 70

(hsp 70) promoter of the vector, and induced by brief

heat treatment expressed a protein of 210 kD detectable by immunoblotting with anti-RPTPc antiserum (FIG. 22A) . This protein corresponds to the unprocesεed form of RPTPc seen in mammalian cells (Y.- P. Jiang et al . Mol. Cell. Biol. 13, 2942 (1993)). In addition, after longer expreεεion periods, a protein species of 110 kD also appeared, suggesting that the RPTPc protein may at least, in part, partly be proceεεed in the Droεophila cell line in a manner similar to the way in which it is procesεed in mammalian cellε, i.e.. through proteolytic cleavage by a furin type endoproteaεe (FIG. 22A) (Y.-P. Jiang et al . Mol. Cell. Biol. 13, 2942 (1993)) . A Drosophila furin homolog has recently been described (A.J.M. Roebroek et al., EMBO J. 12, 1853 (1993)).

In order to study whether RPTP/c expresεion may mediate cell-cell aggregation, cells stably transfected with the RPTP/c CDNA in either the sense orientation (senεe cDNA) or the antiεenεe orientation (antiεenεe cDNA) were teεted in an aggregation aεεay. uninduced and heat εhock-induced cells were resuεpended, subjected to rotary shaking to ensure mixing and to avoid adhesion to the vesεel, and were then aεsayed for aggregate formation. The formation of a large number of aggregates consiεting of more than 10 and up to approximately 100 cells was observed in heat-shocked sense cDNA-expresεing cells only, whereaε control cells (i.e.. antisenεe cDNA tranεfected cellε or non-heat shocked cells) remained esεentially single cell suεpenεions (FIG. 22B,C) . Two methods of quantitation, counting of aggregates under the microscope, and determination of super-threshold particleε with a Coulter-counter (FIG. 22D) confirmed this conclusion. The fact that aggregation was incomplete, with a large proportion of RPTP/c

tranεfected cells remaining as single cells throughout the asεay period, is most likely due to the fact that the transfected cell population consiεted of an uncloned pool of cellε preεumably differing in their levelε of RPTP/c expreεεion. Notably, the conditions of the asεay (i.e.. medium, timescale, and εpeed of εhaking) are εimilar to thoεe used to demonstrate the adhesive properties of a number of well established adhesion molecules (H. Kramer, R.L. Cagan, S.L. Zipursky, Nature 352, 207 (1991) ; P.M. Snow, A.J. Bieber, C. Goodman, Cell 59, 313 (1989)) . Therefore, in view of the difficulty of meaεuring binding affinitieε of many cell adhesion molecules which rely on cooperativity, it is likely that the strength of cell-cell-interaction conferred by expression of RPTP/c is comparable to that of established, "classical", cell adhesion molecules.

The above experiments were performed with a full- length RPTP/c cDNA, leaving unclear whether the phosphatase activity of the intracellular domain is required to confer adhesive properties. In several instances, an intact intracellular domain of cell adhesion molecules has in fact been shown to be required for certain aspects of cell-cell interaction (A. Nafaguchi and M. Takeichi, EMBO J. 7, 3679 (1988); S.H. Jaffe et al . , Proc. Natl. Acad. Sci. USA 87, 3589 (1990), R.O. Hynes, Cell 69,111 (1992)) . To test thiε iεεue, a cDNA encoding a mutant protein lacking moεt of the intracellular, catalytic, domain of RPTP/c waε constructed. Fig. 22E shows that such a truncation did not negatively interfere with cell aggregation as measured in this type of asεay. The role of the furin cleavage εite in the extracellular domain of RPTP/c was also tested. Mutation of this site also left the adhesive behavior intact, suggesting that cleavage of

the RPTP/c proprotein (Y.-P. Jiang et al . Mol. Cell. Biol. 13, 2942 (1993)) is not required for induction of cell aggregation.

Cell adhesion molecules have been described which either do (e.g. cadherin family members and integrins) , or do not (e.g. N-CAM, Ng-CAM) require the presence of Ca ++ (G.M. Edelman, Immun. Rev. 100, 11 (1987); A.F. Williams and A.N. Barclay, Annu. Rev. Immunol. 6, 381 (1988); M. Grumet, Curr. Opin. Neurobiol. 1, 370 (1991), R.O. Hynes, Cell 69,111

(1992), B. Geiger and 0. Ayalon, Annu. Rev. Cell Biol. 8 (1992)). The experiments presented in FIG. 22 were performed in the presence of 10 mM Ca ++ in the aggregating cell suεpenεion. Performing a εimilar experiment in the abεence of calcium ions and in the presence of 1 mm EGTA revealed no calcium requirement for RPTP/c mediated cellular aggregation under the conditions of the assay.

The observed aggregation correlating with expression of RPTP/c could be accounted for by either a homophilic binding mechanism, in which cell-cell binding iε mediated by interaction between RPTP/c proteinε on different cells within aggregates, or by binding of the RPTP/c protein to a second cell-surface ligand intrinsic to the parental transfected cellε. It was posεible to diεtinguish between these two hypotheseε by marking different populations of cells with the fluorescent lipophilic dye 1, 1 '-dioctadecyl- 3,3,3 • ,3 'tetramethylindocarbocyanine perchlorate (dil) (j. Schlessinger et al . Science 195, 307 (1977)), and then testing them for their ability to co-aggregate. In these experiments, RPTP/c expressing and non- expressing cells were labeled with dil, mixed with unlabeled cellε of either RPTP/c expressing or non- expreεεing typeε, and the presence of cells of either

type in the aggregates formed was monitored by fluorescence microscopy. The resultε are illustrated in FIG. 23. Strikingly, mixing of unlabeled, RPTP/c positive cells with labeled, RPTP/c negative cells led to the formation of aggregateε conεiεting excluεively of unlabeled cellε. Conversely, when the RPTP/c expressing cells were labeled and allowed to aggregate with unlabeled control cellε, aggregateε conεisted entirely of labeled cellε, demonεtrating that dil labeling doeε not interfere with the aggregation capacity of the transfected cells. Mixing of labeled and unlabeled cellε, both expreεεing RPTP/c, led to the formation of mixed aggregateε consisting of cells of either staining type, thus confirming that both dil stained and unstained cells have the ability to co- aggregate. These resultε εuggest that aggregation of the RPTP/c transfected cells requires the presence of the protein on all cells within the aggregate, implying a homophilic binding mechanism. It waε next determined whether the extracellular domain of RPTPc was able to function by itself as a substrate for attachment of cellε expressing the RPTP/c protein independent of other factors to asεiεt in the adhesion process. A baculovirus expreεεion εyεtem waε uεed to produce a soluble recombinant protein consisting of virtually the entire extracellular domain of the RPTP/c protein, fused to placental alkaline phosphataεe, which εerved aε a tag for purification and detection (J.G. Flanahan and P. Leder, Cell 63, 185 (1990)). Fuεion between the two protein moietieε waε designed to occur precisely before the furin proteolytic cleavage signal in the fourth fibronectin type III repeat in RPTPc (Y.-P. Jiang et al . Mol. Cell. Biol. 13, 2942 (1993)). The purified recombinant protein (K2AP) was uεed to coat

bacteriological Petri dishes, and monitored for its ability to allow attachment of RPTP/c-expressing S2 cells. Only induced, RPTP/c expressing cells showed adhesive behavior to the K2AP coated εurface (FIG. 24A-D; Table II below) .

TABLE I I

Cell S2 control S2 control S2-R-PTP-K S2-R-PTP-X L6 L6R- lype: PTP-x un-induced induced un-induced induced

Protein

K2AP-* - - - + + 4- + +

K2AP-b - - - + + + •+ + +

AP - - - - - -

HER - - - - - -

BSA - - - - - -

Fibro¬ + + + + + + + + + + + + + + nectin poly- n.d. n.d. n.d. n.d. + + + + + - * - lyune

Summary of adhesion data of different cell typeε to surfaceε coated with purified K2AP protein, or other proteinε (-:no cellε attached; + : 50-150 cells; ++ 150-500; +++:>500 n.d. : not determined) K2APa: K2AP protein purified by elution from affinity column at alkaline pH.

K2APb: K2AP protein purified by elution from affinity column using 50 % ethylene glycol. AP: alkaline phosphataεe control protein (J.G. Flanahan and P. Leder, Cell 63, 185 (1990)), corresponding to the tag portion of the K2AP fusion protein.

HER: Human EGF-receptor extracellular domain affinity- purified from a baculovirus expreεεion system (I. Lax et al., J. Biol. Chem. 266, 13828 (1991)). BSA: bovine serum albumin.

L6-R-PTP/C : a clone of L6 cellε stably transfected with the R-PTPc protein.

No attachment occurred to control coated surfaces, which included alkaline phosphatase or the recombinant extracellular domain of human EGF-receptor (I. Lax et al., J. Biol. Chem. 266, 13828 (1991)), also purified by affinity chromatography from a baculovirus expreεεion εyεtem. hereaε the above experimentε were performed in the context of inεect cellε, the effect of RPTP/c protein expression in mammalian cells in a similar cell-to-substrate adhesion assay was also tested. In contrast to parental Drosophila S2 cells, rat L6 myoblast cells, the mammalian cell line used as a recipient for RPTP/c overexpression, already εhows a low level of εpontaneouε adheεion to a K2AP protein coated εurface. However, εtable overexpression of an RPTP/c cDNA in these cells led to a significant (2.7 fold +/-1.0; n=3) increase in adheεive capacity to a surface coated with the recombinant soluble extracellular domain of the RPTP/c protein (FIG. 24A-D) .

15.1 Diεcuεεion Cell-cell contact iε generally conεidered to play a critical role in variouε aspects of malignancy. For example, escape from contact inhibition is a claεεical parameter of tranεformation, and, additionally, many linkε between cell-cell interactions and εuch phenomena as tumor invasion and metastasis are apparent (F. Van Roy and M. Mareel, TICB 2, 163 (1992)). The above data clearly demonstrate that an RPTPase of the LAR-like subfamily (containing a combination of Ig and fibronectin type III domains) is capable of homophilic binding between neighboring cells, leading to the identification of a function for the extracellular domains of such molecules. This

makes it likely that other members of this RPTPase subfamily can behave in a similar fashion, and extends the series of links that have recently emerged between the adhesive properties of cells, and signal transduction pathways involving tyrosine phosphorylation. For instance, adherens junctionε correspond to sites of increased tyrosine phosphorylation and appear to be subject to its control, and reagents directed at integrins or extracellular domains of established CAMs have been shown to elicit changes in cellular tyrosine phosphorylation (J.R. Atashi et al. , Neuron 8, 831 (1992); T. Volberg et al . , EMBO J. 11, 1733 (1992); R.L. Juliano and S. Haskill, J. Cell Biol. 120, 577 (1993)). In addition, reagents directed toward cell adhesion molecules are known to activate a number of second messenger signals (Schuch, U. Lohse, M. Schachner, Neuron 3, 13-20 (1989); P. Doherty, S.V. Ashton, S.E. Moore, F. Walsh, Cell 67, 21 (1991)). The above observation suggests mechanisms by which such signals might be generated. For example, direct cell-cell contact between RPTPases on adjacent cells could lead to local RPTPase oligomerization events affecting either the catalytic activity or localization of RPTPases, which in turn have been suggested to modulate the activity of src-family tyrosine kinases (H. L. Ostergaard et al . , Proc. Natl. Acad. Sci. USA 86, 8959 (1989); T. Mustelin and A. Altman, Oncogene 5, 809 (1989) ; X.M. Zheng, Y. Wang, CJ. Pallen, Nature 359, 336 (1992)). Moreover, the εimilar εtructural and functional propertieε of the extracellular domainε of RPTPaεes and CAMs prompts the speculation that RPTPaεeε may, in addition to εelf- interaction, alεo be capable of interacting heterophilically with other molecules involved in cell

adheεion, whether in cis or in trans (G.M. Edelman, Immun. Rev. 100, 11 (1987); A.F. Williamε and A.N. Barclay, Annu. Rev. Immunol. 6, 381 (1988); M. Grumet, Curr. Opin. Neurobiol. 1, 370 (1991) , R.O. Hyneε, Cell 69,111 (1992), B. Geiger and 0. Ayalon, Annu. Rev. Cell Biol. 8 (1992) , M. Grumet and G.M. Edelman, J. Cell Biol. 106, 487-503 (1988); G.A. Kadmon, A. Kowitz, P. Altevogt, M. Schachner, J. Cell Biol. 110, 193 (1990) ; A.A. Reyeε, R. Akeson, L. Brezina, G.J. Cole, Cell Reg. 1, 567 (1990); P. Sonderegger and F. G. Rathjen, J. Cell Biol. 119, 1387 (1992); M.G. Grumet, A. Flaccus, R.U. Margoliε, J. Cell Biol. 120, 815 (1993)).

The references cited above are all incorporated by reference herein, whether specifically incorporated or not.

Having now fully described this invention, it will be appreciated by thoεe skilled in the art that the same can be performed within a wide range of eguivalent parameterε, concentrationε, and conditionε without departing from the εpirit and εcope of the invention and without undue experimentation. While thiε invention haε been described in connection with specific embodiments thereof, it will be understood that it iε capable of further modificationε. Thiε application iε intended to cover any variationε, uses, or adaptations of the inventions following, in general, the principles of the invention and including εuch departureε from the preεent diεcloεure aε come within known or cuεtomary practice within the art to which the invention pertains and as may be applied to the eεεential features hereinbefore

set forth as follows in the scope of the appended claims.