Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
G-BETA-GAMMA REGULATED PHOSPHATIDYLINOSITOL-3' KINASE
Document Type and Number:
WIPO Patent Application WO/1997/049818
Kind Code:
A2
Abstract:
The present invention relates to the discovery, identification and characterization of nucleotides that encode the G protein regulated phosphatidylinositol-3' kinase, a heterodimeric enzyme which produces the intracellular messenger phosphatidylinositol (3,4,5)-triphosphate in response to activation of trimeric G protein-linked receptors. This novel protein, comprised of a catalytic subunit, p120, and a regulatory subunit, p101, is found in cells of hematopoietic origin and is involved in immune system responses which cause inflammation. The presence of p101 subunit is largely responsible of the dramatic stimulation of kinase activity in the presence of activated trimeric G proteins. The invention encompasses p101 and p120 nucleotides, host cells expression systems, p101 and p120 proteins, fusion proteins, polypeptides and peptides, antibodies to these proteins, transgenic animals that express a p101 or p120 transgene, or recombinant knock-out cells and animals that do not express the p101 or p120 gene, antagonists and agonists of the enzyme, and other compounds that modulate p101 or p120 gene expression or enzyme activity that can be used for diagnosis, drug screening, clinical trial monitoring, and/or treatment of inflammatory disorders.

Inventors:
STEPHENS LEN
HAWKINS PHILLIP T
BRASELMANN SYLVIA
Application Number:
PCT/US1997/011219
Publication Date:
December 31, 1997
Filing Date:
June 26, 1997
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ONYX PHARMACEUTICALS (US)
International Classes:
A61K31/00; A61K31/70; A61K31/7088; A61K35/74; A61K38/00; A61K39/395; C12N15/02; A61K45/00; A61K48/00; A61P7/00; A61P9/00; A61P9/10; A61P11/00; A61P11/06; A61P19/00; A61P19/02; A61P25/28; A61P29/00; A61P35/00; A61P37/00; A61P37/08; A61P39/00; A61P39/06; A61P43/00; C07K16/40; C07K19/00; C12N9/12; C12N15/09; C12N15/54; C12P21/02; C12Q1/48; C12Q1/68; (IPC1-7): C12N15/54; C12N9/12; C12N5/10; C12N15/62; C07K16/40; G01N33/50
Domestic Patent References:
WO1996012024A11996-04-25
Other References:
STEPHENS L ET AL: "A heterotrimeric GTPase-regulated isoform of PI3K and the regulation of its potential effectors." PHILOS TRANS R SOC LOND B BIOL SCI, FEB 29 1996, 351 (1336) P211-5, ENGLAND, XP002046082
STEPHENS LR ET AL: "The G beta gamma sensitivity of a PI3K is dependent upon a tightly associated adaptor, p101." CELL, APR 4 1997, 89 (1) P105-14, UNITED STATES, XP002046083
STOYANOV B ET AL: "CLONING AND CHARACTERIZATION OF A G PROTEIN-ACTIVATED HUMAN PHOSPHOINOSITIDE-3 KINASE" SCIENCE, vol. 269, 4 August 1995, pages 690-693, XP000615903
See also references of EP 0939821A2
Download PDF:
Claims:
95WHAT IS CLAIM
1. ED LS: An isolated nucleic acid molecule containing the nucleotide sequence of a plOl gene.
2. An isolated nucleic acid molecule which encodes a plOl regulatory subunit, or a fragment thereof, having a nucleotide sequence that: a) encodes SEQ ID NO:2 or the amino acid sequence encoded by the cDNA contained in the cDNA clone pCMV3mycpl01 as deposited with the ATCC having accession No. 97636; or b) hybridizes under stringent conditions to the nucleotide sequence of (a) or to its complement.
3. An isolated nucleic acid molecule which encodes a plOl regulatory subunit, or a fragment thereof, having a nucleotide sequence that: a) encodes SEQ ID NO: 12; or b) hybridizes under stringent conditions to the nucleotide sequence of (a) or to its complement.
4. An isolated nucleic acid molecule according to Claim 2 or 3, wherein the nucleic acid molecule hybridizes under moderately stringent conditions to the nucleotide sequence of (a) or to its complement, and wherein the nucleic acid molecule encodes a functionally equivalent pi 01 gene product.
5. An isolated nucleic acid molecule according to Claim 2 or 3, wherein the nucleic acid molecule hybridizes under highly stringent conditions to the nucleotide sequence of (a) or to its complement.
6. An isolated nucleic acid molecule according to Claim 2 or 3, wherein the nucleic acid molecule encodes SEQ ID NO:2 or the amino acid sequence encoded by the cDNA contained in the cDNA clone pCMVmycplOl as deposited with the ATCC having accession No. 97636. 96 .
7. An isolated nucleic acid molecule according to Claim 2 or 3, wherein the nucleic acid molecule encodes SEQ ID NO: 12.
8. An isolated nucleotide sequence encoding a polypeptide corresponding to the catalytic subunit interaction domain, or the G/3γ interaction domain, of the p 101 regulatory subunit protein, or a deletion mutant of the plOl regulatory subunit protein in which the catalytic subunit interaction domain, or the Gβγ interaction domain, is deleted.
9. An isolated nucleotide sequence encoding a chimeric protein comprising the polypeptide of any one of Claims 1 through 8 fused to a heterologous polypeptide.
10. The isolated nucleotide sequence of Claim 9 in which the heterologous polypeptide is a Glu tag or a myc epitope tag.
11. A nucleotide vector containing the nucleotide sequence of any one of Claims 1 through 10.
12. An expression vector containing the nucleotide sequence of any one of Claims 1 through 10 in operative association with a nucleotide regulatory sequence that controls expression of the nucleotide sequence in a host cell.
13. The expression vector of Claim 12, in which said regulatory sequence is selected from the group consisting of the cytomegalovirus hCMV immediate early gene promoter, the early or late promoters of SV40 adenovirus, the baculovirus promoter, the lac system, the trp system, the TAC system, the TRC system, the major operator and promoter regions of phage λ, the control regions of fd coat protein, the promoter for 3phosphoglycerate kinase, the promoters of acid phosphatase, and the promoters of the yeast αmating factors. 97 .
14. A host cell that has been genetically engineered to contain the nucleotide sequence any one of Claims 1 through 13.
15. A host cell according to Claim 14, wherein the host cell has been genetically engineered to contain the nucleotide sequence of any one of Claims 1 through 13 in operative association with a nucleotide regulatory sequence that controls expression of the nucleotide sequence in the host cell.
16. The host cell of Claim 15 in which the host cell is a Sf9 cell, a Chinese hamster ovary cell, a COS cell, or a U937 cell.
17. A genetically engineered host cell in which the native plOl gene sequence, or a sequence that encodes the nucleotide sequence of any one of claims 1 through 13, is disrupted so that expression of the native plOl gene product is inhibited or prevented.
18. An isolated Gprotein activated PI3K comprising a plOl regulatory subunit and a pi 20 catalytic subunit.
19. An isolated plOl regulatory subunit protein.
20. An isolated plOl regulatory subunit protein having the amino acid sequence of SEQ ID NO:2, or the amino acid sequence encoded by the cDNA contained in the cDNA clone pCMV3mycpl01 as deposited with the ATCC having accession No. 97636.
21. An isolated plOl regulatory subunit protein having the amino acid sequence of SEQ ID NO: 12.
22. A polypeptide having an amino acid sequence corresponding to the catalytic subunit interaction domain, or the G γ interaction domain, of plOl regulatory subunit protein, or a deletion mutant of the p 101 regulatory subunit 98 protein in which the catalytic subunit interaction domain, or the G/3γ interaction domain, is deleted.
23. A chimeric protein comprising the polypeptide of Claims 19, 20, 21 or 22 fused to a heterologous polypeptide.
24. The chimeric protein of Claim 23 in which the heterologous polypeptide is a Glu tag or a myc epitope tag.
25. An antibody that immunospecifically binds the plOl regulatory subunit protein of Claims 19, 20, 21 or 22.
26. A method for diagnosing an inflammatory response disorder in a mammal, comprising detecting a plOl gene mutation contained in the genome of the mammal.
27. A method for screening compounds useful for the treatment of inflammatory response disorders, comprising contacting a compound with a G protein activated PI3K and a lipid membrane, and assaying the transfer of phosphate to the lipid membrane.
28. The method of Claim 27, in which the G protein activated PI3K is comprised of p 101 and pi 20 and Gβγ.
29. The method of Claim 27, in which the lipid membrane contains PtdIns(4,5)P2.
30. The method of Claim 27, in which the transfer of phosphate to the lipid membrane is assayed by quantitating the transfer of 32Phosphate to a lipid product.
31. A method for screening compounds useful for the treatment of inflammatory response disorders, comprising contacting a compound with a 99 cultured host cell that expresses the plOl gene, and detecting a change in the expression of the plOl gene, a change in activity of the plOl gene product expressed by the cultured cell, a change in production of the second messenger PtdIns(3,4,5)P3, a change in cell adhesion, or a change in production of O2.
32. The method of Claim 31 in which expression of the plOl gene is detected by measuring mRNA transcripts of the plOl gene.
33. The method of Claim 31 in which expression of the plOl gene is detected by measuring plOl regulatory subunit protein.
34. The method of Claim 31 in which PtdIns(3,4,5)P3 levels in the host cell are assayed using anionexchange HPLC.
35. A method for treating an inflammatory response disorder in a mammal, comprising administering a compound to the mammal in an amount sufficient to inhibit activation of a G proteinstimulated PI3K through a plOl regulatory subunit.
36. The method of Claim 35 in which the inflammatory response disorder is selected from the group consisting of arthritis, septic shock, adult respiratory distress syndrome (ARDS), pneumonia, asthma, allergies, reperfusion injury, atherosclerosis, Alzeimer's disease, and cancer.
37. The method of Claim 35 in which the compound is delivered to cells of hematopoietic origin.
38. The method of Claim 35 in which the compound is an antagonist that binds to the p 101 regulatory subunit and inhibits G protein mediated activation of PI3K.
39. The method of Claim 35 in which the compound disrupts the interaction of plOl regulatory subunit with a catalytic subunit of PI3K. 100 .
40. The method of Claim 35 in which the compound is an oligonucleotide construct encoding a signallingincompetent plOl regulatory subunit or a fragment thereof, which oligonucleotide construct is controlled by a regulatory sequence that directs the expression of the signallingincompetent subunit or fragment in target cells in the body.
41. The method of Claim 35 in which the oligonucleotide construct encodes a signallingincompetent deletion mutant of the p 101 regulatory subunit in which the Gβγ interaction domain is deleted.
42. A method for treating an inflammatory response disorder in a mammal, comprising administering a compound to the mammal in an amount sufficient to inhibit expression of the plOl regulatory subunit in vivo.
43. The method of Claim 42 in which the inflammatory response disorder is selected from the group consisting of arthritis, septic shock, adult respiratory distress syndrome (ARDS), pneumonia, asthma, allergies, reperfusion injury, atherosclerosis, Alzeimer's disease, and cancer.
44. The method of Claim 42 in which the compound is delivered to cells of hematopoietic origin.
45. The method of Claim 42 in which the compound is an antisense oligonucleotide that inhibits translation of mRNA transcripts that encode the p 101 regulatory subunit.
46. The method of Claim 42 in which the compound is a ribozyme that inhibits translation of mRNA transcripts that encode the plOl regulatory subunit.
47. The method of Claim 42 in which the compound is an oligonucleotide that forms a triple helix with the regulatory region of the p 101 regulatory subunit gene and inhibits transcription. 101 .
48. The method of Claim 42 in which the compound is a recombinant DNA construct that inactivates the plOl regulatory subunit gene or its regulatory region via targeted homologous recombination.
49. A method for treating an inflammatory response disorder in a mammal, comprising administering a compound to a mammal in an amount sufficient to up regulate expression of a functional pi 01 regulatory subunit in the mammal.
50. The method according to Claim 49 in which the compound is delivered to hematopoietic cells.
51. The method of Claim 49 in which the mammal expresses a defective plOl regulatory subunit and the compound comprises a nucleotide construct encoding a functional plOl regulatory subunit controlled by a regulatory region that directs expression of the functional receptor in target cells in the mammal.
52. The method of Claim 49 in which the mammal expresses a mutant plOl regulatory subunit and the compound comprises a nucleotide construct encoding a wildtype plOl regulatory subunit that corrects the endogenous mutation via targeted homologous recombination.
53. An isolated nucleic acid molecule containing the nucleotide sequence of a pi 20 gene.
54. An isolated nucleic acid molecule which encodes a pl20 catalytic subunit, or a fragment thereof, having a nucleotide sequence that: a) encodes SEQ ID NO:4 or the amino acid sequence encoded by the cDNA contained in the cDNA clone of the pCMV3mycpl20 as deposited with the ATCC having accession No. 97637; or b) hybridizes under stringent conditions to at least the nucleotide sequence of (a) encoding the last 28 amino acids of the pl20 protein, or to its complement.
55. An isolated nucleic acid molecule which encodes a pl20 catalytic subunit, or a fragment thereof, having a nucleotide sequence that: a) encodes SEQ ID NO: 14; or b) hybridizes under stringent conditions to at least the nucleotide sequence of (a) encoding the last 28 amino acids of the pl20 protein, or to its complement.
56. An isolated nucleic acid molecule according to Claim 54 or 55, wherein the nucleic acid molecule hybridizes under highly stringent conditions to at least the nucleotide sequence of (a) encoding the last 28 amino acids of the pi 20 protein, or to its complement.
57. An isolated nucleic acid molecule according to Claim 54, wherein the nucleic acid molecule encodes SEQ ID NO:4 or the amino acid sequence encoded by the cDNA contained in the cDNA clone pCMVmycpl20 as deposited with the ATCC having accession No. 97637.
58. An isolated nucleic acid molecule according to Claim 55, wherein the nucleic acid molecule encodes SEQ ID NO: 14.
59. An expression vector containing the nucleotide sequence of any one of Claims 53 through 58 in operative association with a nucleotide regulatory sequence that controls expression of the nucleotide sequence in a host cell.
60. A host cell that has been genetically engineered to contain the nucleotide sequence of any one of Claims 53 through 58. 103 .
61. A host cell that has been genetically engineered to contain the nucleotide sequence of any one of Claims 1 through 13 and any one of Claims 53 through 58.
62. The host cell of Claim 14 wherein the host cell is part of an animal.
63. The host cell of Claim 60 wherein the host cell is part of an animal.
64. The host cell of Claim 61 wherein the host cell is part of an animal.
65. An isolated nucleic acid molecule which encodes a plOl regulatory subunit, or a fragment thereof, comprising a nucleotide sequence that encodes an amino acid sequence selected from the group consisting of amino acid residues 1160 of SEQ ID NO:2, amino acid residues 161263 of SEQ ID NO:2, amino acid residues 1732 of SEQ ID NO:2; amino acid residues 733877 of SEQ ID NO:2; amino acid residues 1160 of SEQ ID NO: 12, amino acid residues 161 263 of SEQ ID NO: 12, amino acid residues 1735 of SEQ ID NO: 12 and amino acid residues 736880 of SEQ ID NO: 12.
66. An isolated nucleic acid molecule which encodes a pl20 subunit, or a fragment thereof, comprising a nucleotide sequence that encodes an amino acid sequence selected from the group consisting of the amino acid sequence extending from about residue 41 to residue 1102 of SEQ ID NO:4 and the amino acid sequence extending from about residue 41 to residue 1102 of SEQ ID NO: 14.
67. An isolated nucleic acid molecule which encodes a pl20 subunit, or a fragment thereof, comprising a nucleotide sequence that encodes amino acid sequence selected from the group consisting of residues 173302 of SEQ ID NO:4 or amino acid residues 173302 of SEQ ID NO: 14.
Description:
G-beta-gamma Regulated Phosphatidylinositol-3' Kinase

1. INTRODUCTION

The present invention relates to novel G-protein regulated phosphoinositide 3OH-kinase enzymes isolated from cells of hematopoietic lineage which are involved in cellular signal transduction pathways, and to the use of these novel enzymes in the treatment and diagnosis of disease.

2. BACKGROUND OF THE INVENTION Phosphoinositide 30H-kinases (PI3Ks) are a large family of enzymes capable of 3-phosphorylating at least one of the cellular phosphoinositides (Whitman et al. , 1988, Nature 332:644-646; Auger et al. , 1989, Cell 57: 167-175). 3-phosphorylated phosphoinositides are found in all higher eukaryotic cells. A growing body of evidence implicates PI3K and a lipid product of this enzyme, phosphaudylinositol (3,4,5)-triphosphate (hereinafter "PtdIns(3,4,5)P 3 "), as part of a novel and important second messenger system in cellular signal transduction. The components of this novel PtdIns(3,4,5)P 3 -based signalling system appear to be independent of the previously characterized signalling pathway based on inositol phospholipids, in which a phosphoinositidase C (PIC) hydrolyses PtdIns(4,5)P 2 to release the structurally distinct second messengers inositol (l ,4,5)-triphosphate (Ins(l,4,5)P 3 ) and diacylglycerol.

Select extracellular agonists and growth factors will stimulate intracellular PI3K activity and cause the rapid and transient intracellular accumulation of PtdIns(3,4,5)P 3 . Surprisingly, stimulation of a variety of different types of cell surface receptors, including receptor tyrosine kinases, receptors associated with src family non-receptor tyrosine kinases, cytokine growth factors, and G protein coupled receptors will all activate members of the PI3K family. (Reviewed in Stephens et al., 1993, Biochemica et Biophysica Acta, 1 179:27-75). For example, tyrosine kinase receptors which, when activated, result in increased accumulation of PtdIns(3,4,5)P 3 .are the PDGF receptor, the EGF receptor, members of the FGF receptor family, the CSF-1 receptor, the insulin receptor, the IGF-1 receptor, and the NGF receptor. Receptors associated with src family non-receptor tyrosine kinases which stimulate PtdIns(3,4,5)P 3 accumulation are the 11-2 receptor, 11-3 receptor, mlgM receptor, the CD4 receptor, the CD2 receptor, and the CD3/T cell

receptor. Additionally, the cytokine 11-4 receptor and the G protein linked thrombin receptor, ATP receptor, and the fMLP receptor all stimulate the activity of a PI3K, resulting in subsequent PtdIns(3,4,5)P 3 accumulation. Thus, PtdIns(3,4,5)P 3 appears to be a second messenger in extremely diverse signalling pathways.

Support for the proposition that PI3K activity and production of PtdIns(3,4,5)P 3 is a physiological relevant pathway of signal transduction for these diverse receptors is derived, inter alia, from two different lines of experimental evidence: inhibition of PI3K activity by fungal metabolites and observations of direct protein associations. Wortmannin, a fungal metabolite, irreversibly inhibits PI3K activity by binding covalently to the catalytic domain of this enzyme. Inhibition of PI3K activity by wortmannin eliminates the subsequent cellular response to the extracellular factor. For example, neutrophils respond to the chemokine fMet-Leu-Phe (fMLP) by stimulating PI3K and synthesizing PtdIns(3,4,5)P 3 . The synthesis correlates with activation of the respiratory burst involved in neutrophil destruction of invading microorganisms. Treatment of neutrophils with wortmannin prevents the fMLP-induced respiratory burst response. Thelen et al. , 1994, PNAS, USA 91 :4960-4964. Indeed, these experiments with wortmannin, as well as other experimental evidence, shows that PI3K activity in cells of hematopoietic lineage, particularly neutrophils, monocytes, and other types of leukocytes, is involved in many of the non-memory immune responses associated with acute and chronic inflammation.

PI3K enzymes interact directly with, and may be co-purified with, activated forms of several receptor tyrosine kinases. When purified, receptor tyrosine kinase associated PI3K was found to consist of 170-200 kD heterodimers (Otsu et al. , 1991, Cell 65:91-104, Pons et al., 1995, Mol. Cell. BioL 15:4453-4465, Inukai et al. , 1996, J. Biol. Chem. 271:5317-5320) comprising a catalytic subunit and an adaptor (or regulatory) subunit.

Two different homologs of the catalytic subunit, pl lOα and pl lOβ, have been described and cloned. The catalytic subunit, which irreversibly binds wortmannin, tightly associates with one or other members of a small family of highly related regulatory subunits, p55α, p55PlK, p85α and p85/3, to form the 170-200 kD heterodimers. The known regulatory subunits contain a large

collection of protein:protein interaction domains, including two SH2 domains (Cantley et al., 1991, Cell 64:281-302).

The presence of the SH2 domains are thought to be responsible for the binding and stimulation of PI3K heterodimers to activated receptor tyrosine kinases. Activated receptors are phosphorylated at key tyrosine residues within local consensus sequences preferred by the SH2 domains found in the 55-87 kD PI3K adaptors (Songyang et al. , 1993, Cell 72:767-778). Once the PI3K heterodimer binds , it directly activates the PI3K catalytic subunit (although this effect is relatively small in vitro, Carpenter et al. , 1993, J. Biol. Chem. 268:9478- 9483, Backer et al., 1992, EMBO J. 11 :3469-3479) and translocates the cytosolic PI3K to a source of its phospholipid substrate. The combination of these factors leads to a surge in PtdIns(3,4,5)P 3 production. Clearly, these isoforms of PI3Ks (pl00α/pl l0/3/p55α,p55PIK) seem structurally adapted to function as dedicated signal transducers downstream of receptor-regulated tyrosine kinases, very like the way the T- family of PI-PLCs are regulated by receptor-sensitive tyrosine kinases (Lee and Rhee, 1995, Current Biol. 7: 193-189).

However, the pl l0/p85 sub-family of PI3Ks do not seem to be involved in the production of PtdIns(3,4,5)P 3 that can occur as a result of activation of cell surface receptors which utilize heterotrimeric GTPases to transduce their signals (e.g.. fMLP, PAF, ATP, and thrombin). These types of cell surface receptors have been primarily described in cells of hematopoietic origin whose activation is involved inflammatory responses of the immune system. Recent evidence has suggested that a chromatographically distinct form of wortmannin-sensitive PI3K is present in U937 cells and neutrophils that possesses a native, relative molecular mass of about 220 kD (Stephens et al., 1994, Cell 77:83-93). This PI3K activity can be specifically stimulated by G/3γ subunits, but not Gα-GTP subunits. A similar PI3K activity has also been described in an Osteosarcoma cell line (Morris et al., 1995, Mol. Pharm. 48:532-539). Platelets also contain a Gβγ-sensitive PI3K, .although it is unclear whether this is a p85/pl 10 PI3K family member (Thomason et al., 1994, J. Biol. Chem. 269: 16525-16528). It seemed likely that this poorly characterized, GjSγ-sensitive PI3K might be responsible for production of PtdIns(3,4,5)P 3 in response to agonists like ATP, fMLP etc.

Stoyanov et al., (1995) have recently published the cloning and expression of a wortmannin-sensitive PtdIns(4,5)P 2 -selective PI3K, termed pl lOγ, from a human bone marrow cDNA library. pl lOγ was amplified by PCR using primers designed to target potential PI3K's as well as Ptdlns4-kinase's catalytic centers. It is clearly distinct from pl lOα and pi 10/3, as it lacks, for example, an amino- terminus binding domain for a member of the p85 adaptor family. pl lOγ was speculated to be the PI3K activity downstream of hetero trimeric GTPase-linked receptors on the basis of its sensitivity to both Gα-GTP and Gβ -subunits in vitro and its expression in myeloid-derived cells. Nevertheless, this hypothesis left several unresolved questions regarding the earlier biochemical evidence which indicated that the Gβy responsive PI3K was not stimulated by Gα-GTP subunits, and that it possessed a much greater molecular mass of about 220 kD.

The effects of G γ subunits on pl lOγ were suggested to be mediated via a putative NH 2 -terminus pleckstrin homology (PH) domain. However, with the description of an increasing number of G/3γ regulated effectors, mounting evidence suggests that PH domains do not represent a widely used Gβγ binding domain. Recent work, using a panel of relatively small peptides based on the sequence of domains only found in the GjSγ-activated adenylate cyclases (ACs 2 and 4) which specifically block G/3γ activation or inhibition of several effectors, has suggested there may be some grounds for believing G/3γ subunits contain a widely used effector activating domain. Further, regions in different effectors that interact with this effector activating domain show significant sequence similarities. Hence a motif (Gln-X-X-Glu-Arg) within the domain in AC2 highlighted by these peptide studies also appears in regions of potassium channels and β-ARKs already implicated in regulation by Gβγ subunits (Chen et al. , 1995, Science 268: 1166- 1169). However, this motif is not replicated in all proteins known to be regulated by G subunits, and consequently sequence analysis cannot currently predict whether a protein will be regulated by G/ subunits.

Identification of the mechanism by which PI3K activity is activated by cellular agonists which transduce their signals through G protein linked receptors is lacking. It is important to note that the vast majority of agonists which activate the neutrophil respiratory burst involved in the inflammatory response will bind to G- protein-coupled receptors rather than receptor tyrosine kinases. Thus, the

mechanism by which PI3K is regulated in response to these types of chemokines is likely to be very different from regulation by growth factors which signal through tyrosine kinases. The present invention is directed towards resolving this issue by the identification, purification, and cloning of a novel and specific form of PI3K which is activated by βy subunits of trimeric G-proteins.

3. SUMMARY OF THE INVENTION

The present invention relates to the discovery, identification, purification, and cloning of nucleotides that encode the trimeric G protein regulated PI3K, a novel protein that produces accumulation of the second messenger PtdIns(3,4,5)P 3 in response to activation of G protein-linked receptors. This novel G-protein regulated PI3K is comprised of a catalytic subunit, pl20, and a regulatory subunit, plOl. The pl20 catalytic subunit shares partial amino acid sequence homology with the PI3K catalytic subunits pl lOα, pi 10/3, and pl l0γ. p 101 , on the other hand, is a completely new protein with no identifiable homology to any other sequence previously available. In the absence of plOl regulatory subunit, activated G protein subunits induce a mild stimulation of catalytic activity by the pl20 catalytic subunit. However, in the presence of p 101 subunit, the PI3K activity of the catalytic subunit is stimulated over 100 fold by activated G proteins. cDNAs encoding both porcine plOl and p!20 and human plOl and p!20 have been cloned and sequenced, and are described herein. The porcine plOl and p!20 cDNAs encode proteins of about 877 amino acids and about 1102 amino acids, respectively (FIG. 2 and FIG. 4), while the human plOl and p!20 cDNAs encode proteins of about 880 amino acids and about 1 102 amino acids, respectively (FIG. 11 and FIG. 13). Although the amino acid sequences of the pl20 proteins are homologous to those of other known PI3K catalytic subunits, the pi 20 cDNAs described herein diverge from the cDNAs encoding the known PI3K catalytic subunits, particularly at, for example, the carboxyl terminus (amino acid residues 1075 to 1102). The plOl transcript is found primarily in cells of hematopoietic lineage. pl20, and other PI3K catalytic subunit proteins, appear to have a far broader tissue and cell type distribution. Notably, the presence of a trimeric G protein sensitive PI3K activity has only been found in a limited number of cells of hematopoietic

lineage (e.g.. neutrophils, platelets, etc.). Thus, the ability to activate PI3K enzymes in response to stimulation of trimeric G protein linked receptors appears largely dependent on the presence of the plOl subunit.

The invention encompasses the following nucleotides, host cells expressing such nucleotides, and the expression products of such nucleotides: (a) nucleotides that encode mammalian plOl and pl20 proteins, including the porcine plOl and p!20 and the human plOl and pl20. and the plOl and p!20 gene products, including the porcine and human gene products; (b) nucleotides that encode portions of plOl and p!20 that correspond to its functional domains, and the polypeptide products specified by such nucleotide sequences, including but not limited to the plOl nucleotides encoding, for example, the plOl G/3γ interaction domain, or the catalytic subunit associating domain, or amino acid residues from porcine plOl extending from about 1 to 160, 80-120, 161 to 263, 264 to 414, 415 to 565, 566 to 706, 707-832, and/or 833 to 877, or amino acid residues from human plOl extending from about 1 to 160, 80-120, 161 to 263, 264 to 416, 417 to 567, 568 to 709, 710-835, and/or 836 to 880, and p!20 nucleotides that encode the pl20 membrane binding domain, or the regulatory subunit domain, or amino acid residues from about 173 to 302 and 310 to 315; (c) nucleotides that encode mutants of plOl and p!20 in which all or a part of one of the domains is deleted or altered, and the polypeptide products specified by such nucleotide sequences, including but not limited to mutants of plOl wherein the nucleotides encoding the G protein interaction domain, or the catalytic subunit associating domain, or amino acid residues from about 1 to 160, 80-120, 161 to 263, 264 to 414, 415 to 565, 566 to 706, 707-832, and/or 833 to 877 are deleted, and to mutants of p!20 wherein the nucleotides encoding the membrane binding domain, or the regulatory subunit domain, or amino acid residues from about 173 to 302 and 310 to 315 are deleted; (d) nucleotides that encode fusion proteins containing the p 101 protein or one of its domains (e.g.. the G/3γ interaction domain, or the catalytic subunit associating domain, or the domains described by amino acid residues from about 1 to 160, 80-120, 161 to 263, 264 to 414, 415 to 565, 566 to 706, 707-832, and/or 833 to 877), or the pl20 protein or one of its domains (e.g.. the membrane binding domain, or the regulatory subunit domain, or amino acid residues from about 173 to 302 and 310 to 315) fused to another polypeptide.

The invention also encompasses agonists and antagonists of G protein regulated PI3K, including small molecules, large molecules, mutant plOl proteins that compete with native plOl , and antibodies, as well as nucleotide sequences that can be used to inhibit plOl gene expression (e.g.. antisense and ribozyme molecules, and gene or regulatory sequence replacement constructs) or to enhance plOl gene expression (e.g.. expression constructs that place the plOl gene under the control of a strong promoter system), and transgenic cells and animals that express a plOl transgene or "knock-outs" that do not express plOl .

In addition, the present invention encompasses methods and compositions for the diagnostic evaluation, typing and prognosis of immune system disorders, including inflammation, and for the identification of subjects having a predisposition to such conditions. For example, plOl nucleic acid molecules of the invention can be used as diagnostic hybridization probes or as primers for diagnostic PCR analysis for the identification of plOl gene mutations, allelic variations and regulatory defects in the plOl gene. The present invention further provides for diagnostic kits for the practice of such methods.

Further, the present invention also relates to methods for the use of the plOl gene and/or p!20 gene products for the identification of compounds which modulate, J , act as agonists or antagonists, of G protein-regulated PI3K gene expression and or plOl and/or p!20 gene product activity. Such compounds can be used as agents to control immune system disorders and, in particular, as therapeutic agents for the treatment of inflammation disorders such as arthritis, septic shock, adult respiratory distress syndrome (ARDS), pneumonia, asthma, allergies, reperfusion injury, atherosclerosis, Alzheimer's disease, and cancer. Still further, the invention encompasses methods and compositions for the treatment of inflammation disorders such as arthritis, septic shock, adult respiratory distress syndrome (ARDS), pneumonia, asthma, allergies, reperfusion injury, atherosclerosis, Alzheimer's disease, and cancer. Such methods and compositions are capable of modulating the level of plOl gene expression and/or the level of plOl or p!20 gene product activity.

This invention is based, in part, on the suφrising discovery of a G protein stimulated PI3K activity in isolated neutrophils; the purification and characterization of this activity as a heterodimeric PI3K protein comprised of a

plOl subunit and a pi 20 subunit; the identification and cloning of plOl cDNA from libraries prepared from porcine neutrophil mRNA, human monocyte mRNA, and human leukocyte mRNA; the identification and cloning of p!20 cDNA from libraries prepared from porcine neutrophil mRNA and human Leukocyte mRNA; characterization of the novel sequences; and studies of isolated recombinantly expressed plOl and pl20 protein in insect cells, U937 cells, and Cos-7 cells.

3.1 DEFINITIONS

As used herein, the following terms, whether used in the singular or plural, will have the meanings indicated:

G protein-regulated PI3K: refers to a PI3K enzyme whose activity is stimulated by activated trimeric G proteins such as G γ subunits and/or Gα-GTP subunits. plOl: means the regulatory subunit of the G protein-regulated PI3K, also known as the adaptor subunit. plOl includes molecules that are homologous to p 101 or which bind to pi 20 and stimulate PI3K catalytic activity in response to activation of trimeric G proteins. plOl fusion proteins having an N-terminal Glu tag (specifically MEEEEFMPMPMEF) are referred to herein as (EE)101 fusion proteins, while plOl fusion proteins having an N-terminal myc epitope tag are referred to herein as myclOl fusion proteins. plOl nucleotides or coding sequences: means nucleotide sequences encoding the pi 01 regulatory subunit protein, polypeptide or peptide fragments of plOl protein, or plOl fusion proteins. plOl nucleotide sequences encompass DNA, including genomic DNA (e.g. the p 101 gene) or cDNA, or RNA. pl20: means the catalytic subunit of the G protein-regulated PI3K. Polypeptides or peptide fragments of pl20 protein are referred to as pl20 polypeptides or pl20 peptides. Fusions of pl20, or pl20 polypeptides or peptide fragments to an unrelated protein are referred to herein as pl20 fusion proteins. (EE)120 fusion proteins have the N-terminal Glu tag MEEEEFMPMEFSS. Functional equivalents of pl20 refer to a PI3K catalytic subunit protein which binds to the p 101 regulatory subunit with

high affinity in vivo or in vitro. Other functional equivalents of pi 20 are homologous catalytic subunits of a G protein-regulated PI3K, such as pi 17. p!20 nucleotides or coding sequences: means nucleotide sequences encoding the pl20 catalytic subunit protein, polypeptide or peptide fragments of pi 20 protein, or pi 20 fusion proteins. p!20 nucleotide sequences encompass DNA, including genomic DNA (e.g. the pi 20 gene) or cDNA, or RNA.

4. DESCRIPTION OF THE FIGURES Figures 1A, IB, 1C, and ID. Porcine plOl adaptor subunit nucleotide sequence. (SEQ ID NO:l)

Figure 2. Deduced Porcine plOl amino acid sequence. (SEQ ID NO:2)

Tryptic peptides identified by protein sequencing are underlined with a solid line. Figures 3A, 3B and 3C. Porcine p!20 catalytic subunit nucleotide sequence. (SEQ ID NO:3)

Figure 4. Deduced Porcine pl20 catalytic subunit sequence. (SEQ ID NO:4) Tryptic peptides identified by protein sequencing are underlined with a solid line. The region of divergence from the published amino acid sequence of PHO is underscored with a broken line. Figure 5. G ϊγ-sensitive PI3K in neutrophil cytosol is distinct to p85/pll0 PI3Ks. Aliquots of each of the fractions derived from a Q-sepharose chromatographic profile were either Western blotted and probed with ap85a or αp85/3 monoclonal antibodies and visualized with an HRP-linked second antibody (upper panel in Figure 5), or incubated with lOOnM [ 3 H]-17-hydroxy-wortmannin, resolved by SDS-PAGE (6% acrylamide and fluorographed (lower panel in Figure 5). G/3γ-sensitive PI3K activity eluted in fractions 20-24.

Figure 6. Peak B After Final Mini Q Column Purification Contains a Gβγ-sensitive PI3K Activity. The final purification product of the Peak B activity was analyzed after isolation from a Mini Q column in the presence and absence of activated Gβγ subunits (βy), a tyrosine phosphorylated peptide (YP-peptide), wortmannin, and/or GCX J -GDP subunits (α GDP).

Figure 7. Pharmacological and regulatory properties of free pl20 and pl01/pl20 PI3Ks recombinantly expressed in Sf9 cells. Assays contained either

7 nM pl01/pl20 or 36 nM pl20 alone (final concentrations) which were incubated with various reagents; 10 mM NaF and 30 μM A1C1 3 (A/F); 1 μM Gβγ subunits (βy); 2 μM Gα-GDP; 100 nM wortmannin (W) or 50 μM tyrosine phosphorylated peptide (PY) for a total of 15 minutes (at 0°C) prior to starting the assays by adding [γ 32 P]-ATP; [ 2 P]-incorporated into [ 32 P]-PtdIns(3,4,5)P 3 was quantified. The data shown are means (n=2).

Figure 8. plOl can associate with a G/?γ-stimulated PI3K activity in U937 cells. U937 cells were transiently transfected with mammalian expression vectors encoding either (EE)120 or (EE)101 , co-transfected in combination with mammalian expression vectors encoding myclOl , mycl20, or an irrelevant control myc fusion protein, as indicated. A total of 40 μg of vector DNA was used. After co-transfection, the cells lysed, precleaned and immunoprecipitated with protein G sepharose covalently cross-linked to α-(EE) monoclonal antibody as described more fully in the Examples herein. The resulting immunoprecipitates were washed and G / 3γ-activated (1 μM, final concentration) PI3K activity was assayed. The PI3K activity detected in immunoprecipitates from cells transfected with irrelevant (EE)- tagged protein, either with or without G γs was subtracted from the data shown (these were means of 1896 dpm and 2862 dpm in the absence and presence of GjSγs, respectively). Parallel transfections labelled with [ 3<i S]-methionine showed the amount of [ 35 S]-pl01 and [ 35 S]-pl20 in the immunoprecipitates fell by 40% when they were co-transfected (data not shown).

Figure 9. Regulation of pl01/pl20 by G γs in vivo. Cos-7 cells were transfected with mammalian expression vectors encoding G/3γ subunit ("βιy 2 "), plOl (" 101 "), and/or pl20 (" 120"), as indicated. After 48 hours of transient expression, cells from each transfection were either Western blotted to determine "Relative Expression", or assayed for the accumulation of PtdIns(3,4,5)P 3 accumulation (shaded bars). For Western blotting, samples were probed with an α-(myc) monoclonal antibody to quantitate the expression of the various (myc)- tagged proteins. Results are given relative to the expression obtained in the presence of all 4 key vectors; the absolute levels of (myc)-pl20 and (myc)-plθl were very similar and about 10 x greater than that of (myc)-γ 2 . Parallel batches of cells were labelled with [ 32 P]-Pi; after 90 minutes lipids were extracted, deacylated and water-soluble head groups resolved by anion-exchange HPLC and quantitated

by liquid scintillation counting. Data shown are means (n=2) +/- ranges. Data for [ 32 P]-PtdIns(3,4,5)P 3 are above the irrelevant DNA control (972 + 41 dpm).

Figures 10A and 10B. Human plOl adaptor subunit nucleotide sequence. (SEQ ID NO:ll) Figure 11. Deduced Human plOl amino acid sequence. (SEQ ID

NO:12)

Figures 12A .and 12B. Human pl20 catalytic subunit nucleotide sequence. (SEQ ID NO: 13)

Figure 13. Deduced Human pl20 catalytic subunit sequence. (SEQ ID NO:14)

5. DETAILED DESCRIPTION OF THE INVENTION

The present invention relates to the identification, purification, and cloning of a specific form of PI3K which is activated by βy subunits of trimeric G- proteins. plOl, described for the first time herein, is a novel subunit of the G- protein regulated PI3K. Also described herein is the identification, cloning, and correct sequence of pl20, the catalytic subunit of the trimeric G-protein regulated PI3K.

PI3K are enzymes which phosphorylate phosphatidyl inositols at the 3d position to generate the intracellular signaling molecule PtdIns(3,4,5)P 3 . Although it has been shown that POK's are induced in a variety of cell types upon stimulation of tyrosine kinase receptors, PI3K's which are activated by trimeric G- protein linked receptors have only been detected in a limited number of hematopoietic lineage derived cells such as platelets, monocytes, and leukocytes. Interestingly, accumulation of PtdIns(3,4,5)P 3 in these cells is associated with many of the immune responses involved in acute and chronic inflammation. For example, neutrophils are activated by, inter alia, the chemokine fMLP which is released in response to infection by microorganisms. This agonist binds to a pertussis toxin sensitive G protein coupled receptor and activates the neutrophil respiratory burst, resulting in superoxide production and cytotoxicity. The respiratory burst response correlates with a rapid and transient intracellular increase in PI3K activity.

Studies in neutrophils demonstrates that wortmannin, which selectively inhibits the catalytic component of POK's, causes a shutdown of superoxide production. Since the vast majority of agonists which activate this respiratory burst bind to G protein-linked receptors, the G protein regulated PI3K activity represents a candidate for a dominant effector pathway whose inhibition will reduce the destructive cellular effects of inflammation. However, wortmannin is not a clinically appropriate inhibitor of this pathway since wortmannin inhibits all PI3K catalytic activities, including those involved in other cellular pathways such as growth factors. Reported herein is the discovery that the G-protein regulated PI3K is composed of two subunits: the catalytic subunit (pl lθ7, pi 17 or pl20); and the plOl regulatory subunit. The ability of PI3K to respond to cell surface receptors which stimulate the release of G/3γ subunits from activated trimeric G-proteins is largely dependent on the presence of the plOl regulatory subunit. Although catalytic subunits exhibit a small stimulation (approximately 1.7 fold) of PI3K activity in vitro in the presence of Gβγ subunits, addition of p 101 proteins will increase G/3γ stimulation of PI3K activity 100 fold. Neutralization of plOl , removal of plOl, or interference of plOl binding to its binding partners, will render the cells incapable of generating greatly increased levels of the intracellular signal PtdIns(3,4,5)P 3 in response to activated G/3γ subunits. Thus, the limited distribution of the plOl subunit in bone marrow derived cells appears to be the critical factor in the cell type specificity of this response. Furthermore, plOl contains no significant homologies with any other identified sequence. This discovery makes plOl, and the pl01/pl20 complex, the target of choice to inhibit, activate, or modulate G protein-activated PI3K with minimal non-specific effects. The invention also encompasses the use of plOl and pi 20 nucleotides, plOl and pl20 proteins and peptides, as well as antibodies to plOl and pl20 (which can, for example, act as plOl or pl20 agonists or antagonists), antagonists that inhibit G protein-activated PI3K activity or expression, or agonists that activate G protein- activated PI3K activity or increase its expression in the treatment of hematopoietic lineage cell activation disorders, including, but not limited to immune responses associated with acute and chronic inflammation, in animals such as humans. For example, antagonists of G -activated PI3K will be useful in the treatment of

arthritis, including rheumatoid arthritis, septic shock, adult respiratory distress syndrome (ARDS), pneumonia, asthma and other lung conditions, allergies, reperfusion injury, atherosclerosis and other cardiovascular diseases, Alzheimer's disease, and cancer, to name just a few inflammatory disorders. In addition, plOl nucleotides and plOl regulatory subunits, as well as p!20 nucleotides and pl20 catalytic subunits, are useful for the identification of compounds effective in the treatment of hematopoietic lineage cell activation disorders involving G protein- activated PI3Ks.

Further, the invention encompasses the use of plOl and pl20 nucleotides, plOl and pl20 proteins and peptides, as well as antibodies to plOl and pl20 in the diagnosis of hematopoietic lineage cell activation disorders. The diagnosis of a plOl regulatory subunit or pl20 catalytic subunit abnormality in a patient, or an abnormality in the G protein activated PI3K signal transduction pathway, will also assist in devising a proper treatment or therapeutic regimen. In particular, the invention described in the subsections below encompasses plOl regulatory subunit, polypeptides or peptides corresponding to functional domains of the plOl regulatory subunit (e.g.. the catalytic subunit association domain, or the domain which interacts with activated G proteins), mutated, truncated or deleted plOl regulatory subunits (e.g. a plOl regulatory subunit with one or more functional domains or portions thereof deleted), plOl regulatory subunit fusion proteins (e.g. a pi 01 regulatory subunit or a functional domain of plOl regulatory subunit, fused to an unrelated protein or peptide such as an epitope tag, J ., the myc epitope), nucleotide sequences encoding such products, and host cell expression systems that can produce such pi 01 regulatory subunit products. Additionally, the invention encompasses pi 20 catalytic subunit proteins, polypeptides, functional domains of the pi 20 subunit (e.g.. the catalytic domain), mutated, truncated or deleted pl20 subunit proteins, pl20 fusion proteins, nucleotide sequences encoding such products, and host cell expression systems that can produce such pi 20 catalytic subunit products. The invention also encompasses antibodies and anti-idiotypic antibodies

(including Fab fragments), antagonists and agonists of the G protein-activated PI3K, as well as compounds or nucleotide constructs that inhibit expression of the plOl or p!20 gene (transcription factor inhibitors, antisense and ribozyme

molecules, or gene or regulatory sequence replacement constructs), or promote expression of plOl regulatory subunit (e.g.. expression constructs in which plOl coding sequences are operatively associated with expression control elements such as promoters, promoter/enhancers, etc.). The invention also relates to host cells and animals genetically engineered to express the human plOl regulatory subunit (or mutants thereof) or to inhibit or "knock-out" expression of the animal's endogenous plOl regulatory subunit.

Further, the invention particularly encompasses antagonists which prevent the association of plOl regulatory subunits with their binding partners, including pl20 and other PI3K catalytic subunit proteins such as pi 17 and pi 10γ, as well as activated trimeric G protein proteins, including G γ subunits.

The plOl regulatory subunit proteins or peptides, plOl regulatory subunit fusion proteins, plOl nucleotide sequences, antibodies, antagonists and agonists can be useful for the detection of mutant plOl regulatory subunits or inappropriately expressed plOl regulatory subunits for the diagnosis of immune disorders. The plOl and pl20 subunit proteins or peptides, plOl and pl20 subunit fusion proteins, plOl and p!20 nucleotide sequences, host cell expression systems, antibodies, antagonists, agonists and genetically engineered cells and animals can be used for screening for drugs effective in the treatment of such immune disorders. The use of engineered host cells and/or animals may offer an advantage in that such systems allow not only for the identification of compounds that bind to the plOl regulatory subunit, but can also identify compounds that affect the signal transduced by the activated plOl regulatory subunit, specifically, production of the intracellular signaling molecule PtdIns(3,4,5)P 3 . Finally, the plOl regulatory subunit protein products and fusion protein products, antibodies and anti-idiotypic antibodies (including Fab fragments), antagonists or agonists (including compounds that modulate signal transduction which may act on downstream targets in the plOl regulatory subunit signal transduction pathway) can be used for therapy of such diseases. For example, nucleotide constructs encoding functional plOl regulatory subunits, mutant plOl regulatory subunits, as well as antisense and ribozyme molecules can be used in "gene therapy" approaches for the modulation of plOl regulatory subunit expression and/or activity in the treatment of hematopoietic lineage cell activation

disorders. Thus, the invention also encompasses pharmaceutical formulations and methods for treating hematopoietic lineage cell activation disorders.

The invention is based, in part, on the surprising discovery of novel PI3K enzymes in porcine neutrophils. Like other PI3K proteins, these enzymes 3- phosphorylate Ptdlns, PtdIns4P .and PtdIns(4,5)P 2 substrates, and were completely inhibited by 100 nM wortmannin. However, unlike the previously described PI3K p85/pl01 protein complexes, the PI3K activity was stimulated over 100 fold by incubation with G0 subunits. Addition of Gα-GDP subunits could inhibit this G γ activation. Furthermore, the PI3K activity was not stimulated by phosphorylated tyrosine peptides. When purified, two distinct heterodimeric protein complex were identified: a pi 17/plOl complex and a pl20/pl01 complex. Peptide sequencing revealed that the pi 01 protein was identical in each complex. The pl20 and pi 17 proteins were homologous with the exception of the amino terminus (see Examples below). Porcine plOl and p!20 cDNAs were then cloned using degenerate probes based upon the peptide sequence to screen an expression library of cDNAs synthesized from porcine neutrophil mRNA. Human plOl and p!20 were cloned using probes derived from the porcine cDNA sequences to screen human monocyte and leukocyte cDNA libraries. While sequence analysis of the porcine and human subunits revealed that the pl20 proteins are homologous to previously cloned PI3K catalytic subunits (although they diverge significantly at the extreme carboxyl terminus), the plOl proteins are unrelated to any sequence in the databases.

Comparison of the amino acid sequences of the porcine and human homologs of plOl and pi 20 reveals a high degree of conservation between the two species. This high degree of conservation between two mammalian species suggests that a similar degree of conservation probably exists among mammalian species in general. In the same way that the porcine sequences were used to clone their human homologs, one skilled in the art can clone plOl and p!20 homologs from other mammalian species using the disclosed human and/or porcine sequences and the methods described in this specification.

Experiments described herein expressing plOl and/or pl20 fusion proteins in insect cells demonstrated that plOl binds tightly to pl20 in a 1 : 1 molar stoichiometry. Free purified pl20 exhibited PI3K activity which was insensitive to

the presence of Gα subunits and tyrosine phosphorylated peptides, and only mildly stimulated by Gβγ subunits. However, when bound to plOl, the PI3K activity of pl20 was stimulated 100 fold by the presence of G subunits. When plOl was expressed alone in insect cells, plOl did not exhibit PI3K activity. An interesting result occurred when plOl was expressed as a tagged fusion protein in human U937 cells. When this recombinantly expressed porcine plOl was immunoprecipitated via the fusion protein tag, these immunoprecipitants did contain G protein regulated PI3K activity. Coexpression of pi 20 with plOl slightly decreased the amount of PI3K activity that could be immunoprecipitated. These results indicated the human U937 cells contained a PI3K catalytic subunit which could bind to and be activated by the porcine pi 01 regulatory subunit. These results demonstrate that along with their highly conserved amino acid sequences, homologs from different mammalian species resemble one another functionally and structurally. Further, transient transfections were performed in Cos-7 cells, which do not normally stimulate PI3K activity in response to activated G-proteins, with constructs encoding plOl, pl20 and/or Gβγ subunits. Transfection of a construct which expressed pi 20 only produced significant increases in cellular PtdIns(3,4,5)P 3 levels in a G/Sγ dependent fashion when co-expressed in the presence of plOl.

Various aspects of the invention are described in greater detail in the subsections below.

5.1 THE pi 01 AND pi 20 GENES The cDNA sequence (SEQ ID NO: 1) and deduced amino acid sequence

(SEQ ID NO:2) of porcine plOl regulatory subunit are shown in FIGs. 1 and 2, respectively. A relatively common allelic variation occurs at amino acid residue 483 in the open reading frame; a serine may be replaced by a glycine at this position. The cDNA sequence (SEQ ID NO: 11) and deduced amino acid sequence

(SEQ ID NO: 12) of human plOl regulatory subunit are shown in FIGs. 10 and 11, respectively.

The nucleotide sequence encoding the first 733 amino acids of the porcine plOl regulatory subunit, corresponding to the first 736 amino acids of the human subunit, is believed to be sufficient to bind the catalytic subunit. However, without the 145 carboxyl terminal amino acids, this truncated plOl bound to pi 20 will not stimulate PI3K activity in response to G/3γ subunits. Therefore, the catalytic subunit associating domain is believed to be contained within the first 732 amino acids (735 for human), and the carboxyl terminal amino acids 733 to 877 (736 to 880 for human) could be involved in the response to Gβγ subunits.

Other domains of porcine plOl are described by amino acid residues from about 1 to 160, 80-120, 161 to 263, 264 to 414, 415 to 565, 566 to 706, 707-832, and/or 833 to 877. The corresponding domains of human plOl are described by amino acid residues from about 1 to 160, 80-120, 161 to 263, 264 to 416, 417 to 567, 568 to 709, 710-835, and/or 836 to 880. The nucleotide sequences which encode amino acid residues from about 161 to 263 define a pleckstrin homology ("PH") domain (PROSITE:PS50003). PH domains may be involved in binding of proteins to Gβγ subunits (Touhara et al. , 1994, J. Biol. Chem. 269: 10217) and to membrane phospholipids (see Shaw, 1996, Bioessays 18:35-46). Thus, the PH domain of plOl may be involved in both of these events.

The nucleotide sequences encoding amino acids 1 to 160 of plOl may be responsible for binding to the pl20 catalytic subunit. When this region of the plOl protein is analyzed for secondary structure, it is predicted to have a self-contained alternating α-helix/β-sheet structure. Within this structure is found homology to a "WW domain" (Staub et al., 1996, Structure 4:495-499 and TIBS 21: 161-163). This WW domain may bind to a proline-rich domain found within the N-terminus of pl20 protein (residues 310 to 315). Thus, the WW domain of plOl may be involved in mediating the interaction between the regulatory subunit and the catalytic subunit.

The cDNA sequence (SEQ ID NO: 3) and deduced amino acid sequence (SEQ ID NO:4) of porcine pl20 are shown in FIGs. 3 and 4, respectively. The cDNA sequence (SEQ ID NO: 13) and deduced amino acid sequence (SEQ ID NO: 14) of human pl20 are shown in FIGs. 12 and 13, respectively. A cryptic thrombin cleavage site is present after the first approximately 40 amino acid residues. The truncated pl20 protein lacking these approximately 40 amino

terminal residues is still able to bind to plOl , but PI3K activity is reduced approximately 20 to 30%. In other words, a protein having the amino acid sequence of the section of SEQ ID NO:4 (porcine pl20 deduced amino acid sequence) extending from about amino acid residue 41 to residue 1102 retains the functionality of full-length pl20. Based on the high degree of sequence homology between porcine and human pi 20, a protein having the amino acid sequence of the section of SEQ ID NO: 14 (human pl20 deduced amino acid sequence) extending from about amino acid residue 41 to residue 1102 would also retain the functionality of full-length pl20. Although the pl20 protein was highly homologous to the previously cloned pl lOγ protein reported by Stoyanov et al. (1995), the extreme C-terminus of pi 20 diverges from the reported pi IO7 protein at amino acid residue 1075; thus, the last 28 amino acid residues of pl20 have not been published in reports of any homologous protein. As noted above, the nucleotides encoding a proline-rich region including pl20 residues 310 to 315 may be involved in the interaction between pi 20 and plOl . Additionally, the nucleotides encoding pi 20 amino acid residues from about 173 to 302 define a weak PH domain which may be a candidate for membrane binding and/or Gβ subunit interaction of the pl01/pl20 complex.

Data presented in the working examples, infra, demonstrate that the p!20 cDNA encodes the catalytic subunit of the G -activated PI3K. The plOl cDNA encodes a novel regulatory subunit protein which binds to the pi 20 subunit. This heterodimer 3-phosphorylates Ptdlns, PtdIns4P and PtdIns(4,5)P 2 in response to activation of trimeric G-protein linked receptors.

The plOl nucleotide sequences of the invention include: (a) the DNA sequence shown in FIG. 1 or FIG.10 or contained in the cDNA clone pCMV3mycpl01 as deposited with the American Type Culture Collection (ATCC) under accession number 97636; (b) nucleotide sequence that encodes the amino acid sequence shown in FIG. 2 or FIG.11 , or the p 101 regulatory subunit amino acid sequence encoded by the cDNA clone pCMV3mycpl01 as deposited with the ATCC; (c) any nucleotide sequence that hybridizes to the complement of the DNA sequence shown in FIG. 1 or FIG. 10 or contained in the cDNA clone pCMV3mycpl01, as deposited with the ATCC under highly stringent conditions, e.g.. hybridization to filter-bound DNA in 0.5 M NaHP0 4 , 7% sodium dodecyl

sulfate (SDS), 1 mM EDTA at 65°C, and washing in O. lxSSC/0.1 % SDS at 68°C (Ausubel F.M. et al., eds., 1989, Current Protocols in Molecular Biology, Vol. I, Green Publishing Associates, Inc., and John Wiley & sons, Inc. , New York, at p. 2. 10.3) and encodes a functionally equivalent gene product; and (d) any nucleotide sequence that hybridizes to the complement of the DNA sequences that encode the amino acid sequence shown in FIG. 1 or FIG. 10 or contained in the cDNA clone pCMV3mycpl01, as deposited with the ATCC, under less stringent conditions, such as moderately stringent conditions, e.g.. washing in 0.2xSSC/0.1 % SDS at 42°C (Ausubel et al., 1989, supra), yet which still encodes a functionally equivalent plOl gene product. Functional equivalents of the plOl regulatory subunit include naturally occurring plOl regulatory subunit present in other species, and mutant plOl regulatory subunits whether naturally occurring or engineered which retain at least some of the functional activities of p 101 (i.e.. binding to the pl20 or pi 17 catalytic subunit, stimulation of catalytic activity in response to G subunits, and/or interaction with G ?γ subunits). The invention also includes degenerate variants of sequences (a) through (d).

The invention also includes nucleic acid molecules, preferably DNA molecules, that hybridize to, and are therefore the complements of, the nucleotide sequences (a) through (d), in the preceding paragraph. Such hybridization conditions may be highly stringent or less highly stringent, as described above. In instances wherein the nucleic acid molecules are deoxyoligonucleotides ("oligos"), highly stringent conditions may refer, e.g. , to washing in 6xSSC/0.05% sodium pyrophosphate at 37°C (for 14-base oligos), 48°C (for 17-base oligos), 55°C (for 20-base oligos), and 60°C (for 23-base oligos). These nucleic acid molecules may encode or act as plOl antisense molecules, useful, for example, in plOl gene regulation (for and/or as antisense primers in amplification reactions of plOl gene nucleic acid sequences). With respect to plOl gene regulation, such techniques can be used to regulate, for example, inflammatory immune responses. Further, such sequences may be used as part of ribozyme and/or triple helix sequences, also useful for plOl gene regulation. Still further, such molecules may be used as components of diagnostic methods whereby, for example, the presence of a particular plOl allele responsible for causing an inflammatory response, such as arthritis, may be detected.

In addition to the plOl nucleotide sequences described above, full length plOl cDNA or gene sequences present in the same species and/or homologs of the plOl gene present in other species can be identified and readily isolated, without undue experimentation, by molecular biological techniques well known in the art. Experimental evidence described herein indicates that the plOl proteins are conserved in different mammalian species. For example, human plOl was cloned from monocyte and leukocyte libraries using probes derived from the porcine plOl cDNA sequence, and the deduced amino acid sequences of the human and porcine were found to be highly conserved. The structural and functional similarity of the two homologs is evidenced by experiments showing that when porcine plOl was expressed in U937 cells, the porcine plOl bound to a human homolog of the catalytic subunit. Additionally, members of the tyrosine kinase regulated PI3K family of proteins, for example the pl l0/p85 PI3K, are also conserved among different mammalian species. Therefore, homologs of plOl and p!20 can be isolated from a variety of mammalian cells known or suspected to contain a trimeric G-protein regulated PI3K, particularly cells of hematopoietic origin, and more particularly platelets, monocytes, leukocytes, osteoclasts. and neutrophils.

The identification of homologs of plOl in related species can be useful for developing animal model systems more closely related to humans for purposes of drug discovery. For example, expression libraries of cDNAs synthesized from neutrophil mRNA derived from the organism of interest can be screened using labeled catalytic subunit derived from that species, e.g.. a pl20, pi 17, or pl lOγ catalytic subunit fusion protein. Alternatively, such cDNA libraries, or genomic DNA libraries derived from the organism of interest can be screened by hybridization using the nucleotides described herein as hybridization or amplification probes. Furthermore, genes at other genetic loci within the genome that encode proteins which have extensive homology to one or more domains of the plOl gene product can also be identified via similar techniques. In the case of cDNA libraries, such screening techniques can identify clones derived from alternatively spliced transcripts in the same or different species.

Screening can be by filter hybridization, using duplicate filters. The labeled probe can contain at least 15-30 base pairs of the human or porcine plOl nucleotide sequence, as shown in FIGS. 1 and 10, respectively. The hybridization

washing conditions used should be of a lower stringency when the cDNA library is derived from an organism different from the type of organism from which the labeled sequence was derived. With respect to the cloning of a mammalian plOl homolog, using porcine and/or human plOl probes, for example, hybridization can, for example, be performed at 65 °C overnight in Church's buffer (7% SDS, 250 mM NaHPO 4 , 2μM EDTA, 1 % BSA). Washes can be done with 2XSSC, 0.1 % SDS at 65°C and then at O. lXSSC, 0.1 % SDS at 65°C.

Low stringency conditions are well known to those of skill in the art, and will vary predictably depending on the specific organisms from which the library and the labeled sequences are derived. For guidance regarding such conditions see, for example, Sambrook et al. , 1989, Molecular Cloning, A Laboratory Manual, Cold Springs Harbor Press, N.Y. ; and Ausubel et al. , 1989, Current Protocols in Molecular Biology, Green Publishing Associates and Wiley Interscience, N.Y. Alternatively, the labeled plOl nucleotide probe may be used to screen a genomic library derived from the organism of interest, again, using appropriately stringent conditions. The identification and characterization of human genomic clones is helpful for designing diagnostic tests and clinical protocols for treating hematopoietic lineage cell activation disorders in human patients. For example, sequences derived from regions adjacent to the intron/exon boundaries of the human gene can be used to design primers for use in amplification assays to detect mutations within the exons, introns, splice sites (e.g. splice acceptor and/or donor sites), etc., that can be used in diagnostics.

Further, a plOl gene homolog may be isolated from nucleic acid of the organism of interest by performing PCR using two degenerate oligonucleotide primer pools designed on the basis of amino acid sequences within the plOl gene product disclosed herein. The template for the reaction may be cDNA obtained by reverse transcription of mRNA prepared from, for example, human or non-human cell lines or cell types, such as neutrophils, known or suspected to express a plOl gene allele.

The PCR product may be subcloned and sequenced to ensure that the amplified sequences represent the sequences of a plOl gene. The PCR fragment may then be used to isolate a full length cDNA clone by a variety of methods. For

example, the amplified fragment may be labeled and used to screen a cDNA library, such as a bacteriophage cDNA library. Alternatively, the labeled fragment may be used to isolate genomic clones via the screening of a genomic library.

PCR technology may also be utilized to isolate full length cDNA sequences. For example, RNA may be isolated, following standard procedures, from an appropriate cellular source (i.e.. one known, or suspected, to express the plOl gene, such as, for example, neutrophils or other types of leukocytes). A reverse transcription reaction may be performed on the RNA using an oligonucleotide primer specific for the most 5' end of the amplified fragment for the priming of first strand synthesis. The resulting RNA/DNA hybrid may then be "tailed" with guanines using a standard terminal transferase reaction, the hybrid may be digested with RNAase H, and second strand synthesis may then be primed with a poly-C primer. Thus, cDNA sequences upstream of the amplified fragment may easily be isolated. For a review of cloning strategies which may be used, see e.g.. Sambrook et al. , 1989, supra.

The plOl gene sequences may additionally be used to isolate mutant plOl gene alleles. Such mutant alleles may be isolated from individuals either known or proposed to have a genotype which contributes to the symptoms of hematopoietic lineage cell activation disorders such as inflammation. Mutant alleles and mutant allele products may then be utilized in the therapeutic and diagnostic systems described below. Additionally, such plOl gene sequences can be used to detect plOl gene regulatory (e.g.. promoter or promotor/enhancer) defects which can affect hematopoietic lineage cell activation.

A cDNA of a mutant plOl gene may be isolated, for example, by using PCR, a technique which is well known to those of skill in the art. In this case, the first cDNA strand may be synthesized by hybridizing an oligo-dT oligonucleotide to mRNA isolated from cells known or suspected to be expressed in an individual putatively carrying the mutant plOl allele, and by extending the new strand with reverse transcriptase. The second strand of the cDNA is then synthesized using an oligonucleotide that hybridizes specifically to the 5' end of the normal gene. Using these two primers, the product is then amplified via PCR, cloned into a suitable vector, and subjected to DNA sequence analysis through methods well known to those of skill in the art. By comparing the DNA sequence of the mutant plOl

allele to that of the normal plOl allele, the mutation(s) responsible for the loss or alteration of function of the mutant plOl gene product can be ascertained.

Alternatively, a genomic library can be constructed using DNA obtained from an individual suspected of or known to carry the mutant plOl allele, or a cDNA library can be constructed using RNA from a cell type known, or suspected, to express the mutant plOl allele. The normal plOl gene or any suitable fragment thereof may then be labeled and used as a probe to identify the corresponding mutant plOl allele in such libraries. Clones containing the mutant plOl gene sequences may then be purified and subjected to sequence analysis according to methods well known to those of skill in the art.

Additionally, an expression library can be constructed utilizing cDNA synthesized from, for example, RNA isolated from a cell type known, or suspected, to express a mutant plOl allele in an individual suspected of or known to carry such a mutant allele. In this manner, gene products made by the putatively mutant cell type may be expressed and screened using standard antibody screening techniques in conjunction with antibodies raised against the normal plOl gene product, as described, below, in Section 5.3. (For screening techniques, see, for example, Harlow, E. and Lane, eds. , 1988, "Antibodies: A Laboratory Manual", Cold Spring Harbor Press, Cold Spring Harbor.) Additionally, screening can be accomplished by screening with labeled fusion proteins, such as, for example, the (EE)120 or the mycl20 fusion proteins. In cases where a plOl mutation results in an expressed gene product with altered function (e.g.. as a result of a missense or a frameshift mutation), a polyclonal set of antibodies to plOl regulatory subunit are likely to cross-react with the mutant plOl regulatory subunit gene product. Library clones detected via their reaction with such labeled antibodies can be purified and subjected to sequence analysis according to methods well known to those of skill in the art.

The invention also encompasses nucleotide sequences that encode mutant plOl regulatory subunits, peptide fragments of the plOl regulatory subunit, truncated plOl regulatory subunits, and plOl regulatory subunit fusion proteins. These include, but are not limited to nucleotide sequences encoding mutant plOl regulatory subunits described in section 5.2 infra: polypeptides or peptides corresponding to the catalytic binding, or G/3 subunit binding domains of the plOl

regulatory subunit or portions of these domains; truncated plOl regulatory subunits in which one or two of the domains is deleted, or a truncated, nonfunctional plOl regulatory subunit. Nucleotides encoding fusion proteins may include but are not limited to full length plOl regulatory subunit, truncated plOl regulatory subunit or peptide fragments of plOl regulatory subunit fused to an unrelated protein or peptide, such as for example, an epitope tag which aids in purification or detection of the resulting fusion protein; or an enzyme, fluorescent protein, luminescent protein which can be used as a marker.

The invention also encompasses (a) DNA vectors that contain any of the foregoing plOl regulatory subunit coding sequences and/or their complements (i.e.. antisense); (b) DNA expression vectors that contain any of the foregoing plOl regulatory subunit coding sequences operatively associated with a regulatory element that directs the expression of the coding sequences; and (c) genetically engineered host cells that contain any of the foregoing plOl regulatory subunit coding sequences operatively associated with a regulatory element that directs the expression of the coding sequences in the host cell. As used herein, regulatory elements include but are not limited to inducible and non-inducible promoters, enhancers, operators and other elements known to those skilled in the art that drive and regulate expression. Such regulatory elements include but are not limited to the baculovirus promoter, cytomegalovirus hCMV iininediate early gene promoter, the early or late promoters of SV40 adenovirus, the lac system, the trp system, the TAC system, the TRC system, the major operator and promoter regions of phage A, the control regions of fd coat protein, the promoter for 3-phosphoglycerate kinase, the promoters of acid phosphatase, and the promoters of the yeast α-mating factors.

Finally, the invention also encompasses nucleotides encoding pl20 subunit proteins including the newly described carboxyl terminus of this catalytic subunit, deletion variants of pl20 subunit proteins, nucleotides which hybridize to these nucleotides under highly stringent conditions and which encode functionally equivalent products, including the cDNA clone pCMV3mycpl20 deposited with the ATCC under accession number 97637, allelic variants of pl20 (e.g.. mutant alleles or the naturally occurring alleles such as the allelic variation at amino acid residue 483), and equivalent p!20 nucleotides from different organisms isolated as

described above for the plOl nucleotides of the invention. Additionally, the invention encompasses expression vectors and host cells for the recombinant production of pl20.

5.2 plOl AND i 20 PROTEINS AND POLYPEPTIDES plOl regulatory subunit and pl20 catalytic subunit, polypeptides and peptide fragments, mutated, truncated or deleted forms of the plOl regulatory subunit and/or plOl regulatory subunit fusion proteins and the pl20 catalytic subunit and/or pi 20 catalytic subunit fusion proteins can be prepared for a variety of uses, including but not limited to the generation of antibodies, as reagents in diagnostic assays, the identification of other cellular gene products involved in the regulation of hematopoietic lineage cell activation, as reagents in assays for screening for compounds that can be used in the treatment of hematopoietic lineage cell activation disorders, and as pharmaceutical reagents useful in the treatment of hematopoietic lineage cell activation disorders related to G / 3γ-activated PI3K.

FIGS. 2 and 11 show the amino acid sequences of porcine and human plOl regulatory subunit protein, respectively. FIGS. 4 and 13 show the amino acid sequence of porcine and human pl20 catalytic subunit protein, respectively. The broken line on FIG. 4 underscores the region of pi 20 which diverges from the published sequences of the PI3K catalytic subunits pl lOα, pi 10 / 3, and pl lOγ.

The plOl regulatory subunit sequence begins with a methionine in a DNA sequence context consistent with a translation initiation site. The predicted molecular mass of both porcine and human plOl regulatory subunits is 97 kD.

The plOl regulatory subunit amino acid sequences of the invention include the amino acid sequences shown in FIG. 2 (SEQ ID NO:2) or FIG. 4 (SEQ ID NO: 12), or the amino acid sequence encoded by the cDNA clone pCMV3mycpl01, as deposited with the ATCC. Further, plOl regulatory subunits of other species are encompassed by the invention. In fact, any plOl regulatory subunit protein encoded by the plOl nucleotide sequences described above, are within the scope of the invention.

The pi 20 catalytic subunit amino acid sequences of the invention include the cDNA clone pCMV3mycpl20, as deposited with the ATCC. The invention

also encompasses pl20 catalytic subunits of other species, and the pl20 proteins encoded by the p!20 nucleotide sequences described above in the previous section. The invention also encompasses proteins that are functionally equivalent to the plOl regulatory subunit encoded by the nucleotide sequences described above, as judged by any of a number of criteria, including but not limited to the ability to bind catalytic subunit, the binding affinity for catalytic subunit, the ability to stimulate PI3K activity of the catalytic subunit in response to activated trimeric G proteins, the resulting biological effect of catalytic subunit binding and response to activation of trimeric G proteins, e.g.. signal transduction, a change in cellular metabolism (e.g.. generation of PtdIns(3,4,5)P 3 ) or change in phenotype when the plOl regulatory subunit equivalent is present in an appropriate cell type (such as the superoxide burst in neutrophils). Such functionally equivalent plOl regulatory subunit proteins include but are not limited to additions or substitutions of amino acid residues within the amino acid sequence encoded by the plOl nucleotide sequences described, above, but which result in a silent change, thus producing a functionally equivalent gene product. Amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved. For example, nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; positively charged (basic) a ino acids include arginine, lysine, and histidine; and negatively charged (acidic) amino acids include aspartic acid and glutamic acid. Similarly, the invention also encompasses functional equivalents of pl20 protein, as described above.

While random mutations can be made to plOl DNA (using random mutagenesis techniques well known to those skilled in the art) and the resulting mutant plOl regulatory subunits tested for activity, site-directed mutations of the plOl coding sequence can be engineered (using site-directed mutagenesis techniques well known to those skilled in the art) to generate mutant plOl regulatory subunits with increased function, e.g.. higher binding affinity for catalytic subunit, and/or greater signalling capacity; or decreased function, e.g..

lower binding affinity for catalytic subunit, and/or decreased signal transduction capacity.

For example, porcine plOl amino acid sequence may be aligned with that of human plOl regulatory subunit. Mutant plOl regulatory subunits can be engineered so that regions of interspecies identity are maintained, whereas the variable residues are altered, e.g.. by deletion or insertion of an amino acid residue(s) or by substitution of one or more different amino acid residues. Conservative alterations at the variable positions can be engineered in order to produce a mutant plOl regulatory subunit that retains function; e.g.. catalytic subunit binding affinity or activated G protein transduction capability or both. Non-conservative changes can be engineered at these variable positions to alter function, e.g.. catalytic subunit binding affinity or signal transduction capability, or both. Alternatively, where alteration of function is desired, deletion or non- conservative alterations of the conserved regions can be engineered. One of skill in the art may easily test such mutant or deleted plOl regulatory subunits for these alterations in function using the teachings presented herein.

Other mutations to the plOl coding sequence can be made to generate plOl regulatory subunits that are better suited for expression, scale up, etc. in the host cells chosen. For example, the triplet code for each amino acid can be modified to conform more closely to the preferential codon usage of the host cell's translational machinery.

Peptides corresponding to one or more domains (or a portion of a domain) of the plOl regulatory subunit (e.g. , the pl20 binding domain, the G protein interacting domain, or the domains defined by amino acid residues from about 1 to 150, 151 to 300, 301 to 450, 451 to 600, 601-732 (porcine) or 601-735 (human), and 733-877 (porcine) or 736-881 (human)), truncated or deleted plOl regulatory subunits (e.g.. plOl regulatory subunit in which portions of one or more of the above domains are deleted) as well as fusion proteins in which the full length plOl regulatory subunit, a plOl regulatory subunit peptide or truncated plOl regulatory subunit is fused to an unrelated protein are also within the scope of the invention and can be designed on the basis of the plOl nucleotide and plOl regulatory subunit amino acid sequences disclosed in this Section and above. Such fusion proteins include but are not limited to fusions to an epitope tag (such as is

exemplified in the Examples below); or fusions to an enzyme, fluorescent protein, or luminescent protein which provide a marker function.

While the plOl regulatory subunit polypeptides and peptides can be chemically synthesized (e.g.. see Creighton, 1983, Proteins: Structures and Molecular Principles, W.H. Freeman & Co. , N.Y.), large polypeptides derived from the plOl regulatory subunit and the full length plOl regulatory subunit itself may advantageously be produced by recombinant DNA technology using techniques well known in the art for expressing nucleic acid containing plOl gene sequences and/or coding sequences. Such methods can be used to construct expression vectors containing the plOl nucleotide sequences described above and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. See, for example, the techniques described in Sambrook et al. , 1989, supra, and Ausubel et al. , 1989, supra. Alternatively, RNA capable of encoding plOl nucleotide sequences may be chemically synthesized using, for example, synthesizers. See, for example, the techniques described in "Oligonucleotide Synthesis", 1984, Gait, M.J. ed., IRL Press, Oxford, which is incorporated by reference herein in its entirety.

A variety of host-expression vector systems may be utilized to express the plOl nucleotide sequences of the invention. Where the plOl regulatory subunit peptide or polypeptide is a soluble derivative the peptide or polypeptide can be recovered from the culture, he, . , from the host cell in cases where the plOl regulatory subunit peptide or polypeptide is not secreted, and from the culture media in cases where the plOl regulatory subunit peptide or polypeptide is secreted by the cells. However, such engineered host cells themselves may be used in situations where it is important not only to retain the structural and functional characteristics of the plOl regulatory subunit, but to assess biological activity, e.g.. in drug screening assays.

The expression systems that may be used for purposes of the invention include but are not limited to microorganisms such as bacteria (e.g.. E. coli. subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing plOl nucleotide sequences; yeast (e, . Saccharomyces. Pichia) transformed with recombinant yeast expression vectors

containing the plOl nucleotide sequences; insect cell systems infected with recombinant virus expression vectors (e.g.. baculovirus) containing the plOl sequences; plant cell systems infected with recombinant virus expression vectors (e.g.. cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g.. Ti plasmid) containing plOl nucleotide sequences; or mammalian cell systems (e.g.. COS, CHO, BHK, 293, 3T3, U937) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g.. metallothionein promoter) or from mammalian viruses (e.g.. the adenovirus late promoter; the vaccinia virus 7.5K promoter).

In bacterial systems, a number of expression vectors may be advantageously selected depending upon the use intended for the plOl gene product being expressed. For example, when a large quantity of such a protein is to be produced, for the generation of pharmaceutical compositions of plOl regulatory subunit protein or for raising antibodies to the plOl regulatory subunit protein, for example, vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable. Such vectors include, but are not limited, to the E. coli expression vector pUR278 (Ruther et al. , 1983, EMBO J. 2: 1791), in which the plOl coding sequence may be ligated individually into the vector in frame with the lacZ coding region so that a fusion protein is produced; pIN vectors (Inouye & Inouye, 1985, Nucleic Acids Res. 13:3101-3109; Van Heeke & Schuster, 1989, J. Biol. Chem. 264:5503-5509); and the like. pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). If the inserted sequence encodes a relatively small polypeptide (less than 25 kD), such fusion proteins are generally soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione. The pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety. Alternatively, if the resulting fusion protein is insoluble and forms inclusion bodies in the host cell, the inclusion bodies may be purified and the recombinant protein solubilized using techniques well known to one of skill in the art.

In an insect system, Autographa californica nuclear polyhidrosis virus (AcNPV) may be used as a vector to express foreign genes. (E.g.. see Smith et al. , 1983, J. Virol. 46: 584; Smith, U.S. Patent No. 4,215,051). In a specific embodiment described below, Sf9 insect cells are infected with a baculovirus vectors expressing either a 6 x HIS-tagged pl20 construct, or an (EE)-tagged plOl construct.

In mammalian host cells, a number of viral-based expression systems may be utilized. Specific embodiments described more fully below express tagged plOl or pi 20 cDNA sequences using a CMV promoter to transiently express recombinant protein in U937 cells or in Cos-7 cells. Alternatively, retroviral vector systems well known in the art may be used to insert the recombinant expression construct into host cells. For example, retroviral vector systems for transducing hematopoietic cells are described in published PCT applications WO 96/09400 and WO 94/29438. In cases where an adenovirus is used as an expression vector, the plOl nucleotide sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g.. the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g.. region El or E3) will result in a recombinant virus that is viable and capable of expressing the plOl gene product in infected hosts. (E.g.. See Logan & Shenk, 1984, Proc. Natl. Acad. Sci. USA 81 :3655-3659). Specific initiation signals may also be required for efficient translation of inserted plOl nucleotide sequences. These signals include the ATG initiation codon and adjacent sequences. In cases where an entire plOl gene or cDNA, including its own initiation codon and adjacent sequences, is inserted into the appropriate expression vector, no additional translational control signals may be needed. However, in cases where only a portion of the plOl coding sequence is inserted, exogenous translational control signals, including, perhaps, the ATG initiation codon, must be provided. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be

enh.anced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (See Bittner et al. , 1987, Methods in Enzymol. 153:516-544).

In addition, a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g.. glycosylation) and processing (e.g.. cleavage) of protein products may be important for the function of the protein. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed. To this end, eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript may be used. Such mammalian host cells include but are not limited to CHO, VERO, BHK, HeLa, COS, MDCK, 293, 3T3, WI38, and U937 cells.

For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell lines which stably express the plOl sequences described above may be engineered. Rather than using expression vectors which contain viral origins of replication, host cells can be transformed with DNA controlled by appropriate expression control elements (e.g.. promoter, enhancer sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker. Following the introduction of the foreign DNA, engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines which express the plOl gene product. Such engineered cell lines may be particularly useful in screening and evaluation of compounds that affect the endogenous activity of the plOl gene product.

A number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler et al. , 1977, Cell 11 :223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, 1962,

Proc. Natl. Acad. Sci. USA 48:2026), and adenine phosphoribosyltransferase (Lowy et al., 1980, Cell 22:817) genes can be employed in tk " , hgprt " or aprt " cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al., 1980, Natl. Acad. Sci. USA 77:3567; O'Hare et al. , 1981, Proc. Natl. Acad. Sci. USA 78: 1527); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, 1981, Proc. Natl. Acad. Sci. USA 78:2072); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin et al. , 1981, J. Mol. Biol. 150: 1); and hygro, which confers resistance to hygromycin (Santerre et al. , 1984, Gene 30: 147).

The plOl gene products can also be expressed in transgenic animals. Animals of any species, including, but not limited to, mice, rats, rabbits, guinea pigs, pigs, micro-pigs, goats, and non-human primates, e.g.. baboons, monkeys, and chimpanzees may be used to generate plOl transgenic animals. Any technique known in the art may be used to introduce the plOl transgene into animals to produce the founder lines of transgenic animals. Such techniques include, but are not limited to pronuclear microinjection (Hoppe, P.C. and Wagner, T.E., 1989, U.S. Pat. No. 4,873, 191); retrovirus mediated gene transfer into germ lines (Van der Putten et al. , 1985, Proc. Natl. Acad. Sci. , USA 82:6148-6152); gene targeting in embryonic stem cells (Thompson et al. , 1989, Cell 56:313-321); electroporation of embryos (Lo, 1983, Mol Cell. Biol. 3: 1803- 1814); and sperm-mediated gene transfer (Lavitrano et al. , 1989, Cell 57:717-723); etc. For a review of such techniques, see Gordon, 1989, Transgenic Animals, Intl. Rev. Cytol. 115: 171-229, which is incoφorated by reference herein in its entirety. The present invention provides for transgenic animals that carry the plOl transgene in all their cells, as well as animals which carry the transgene in some, but not all their cells, LJ . , mosaic animals. The transgene may be integrated as a single transgene or in concatamers, e.g.. head-to-head tandems or head-to-tail tandems. The transgene may also be selectively introduced into and activated in a particular cell type by following, for example, the teaching of Lasko et al. (Lasko, M. et al. , 1992, Proc. Natl. Acad. Sci. USA 89: 6232-6236). The regulatory sequences required for such a cell-type specific activation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art.

When it is desired that the plOl gene transgene be integrated into the chromosomal site of the endogenous plOl gene, gene targeting is preferred. Briefly, when such a technique is to be utilized, vectors containing some nucleotide sequences homologous to the endogenous plOl gene are designed for the puφose of integrating, via homologous recombination with chromosomal sequences, into and disrupting the function of the nucleotide sequence of the endogenous plOl gene. In this way, the expression of the endogenous plOl gene may also be eliminated by inserting non-functional sequences into the endogenous gene. The transgene may also be selectively introduced into a particular cell type, thus inactivating the endogenous plOl gene in only that cell type, by following, for example, the teaching of Gu et al. (Gu et al., 1994, Science 265: 103-106). The regulatory sequences required for such a cell-type specific inactivation will depend upon the particular cell type of interest, and will be apparent to those of skill in the .art. Once transgenic animals have been generated, the expression of the recombinant plOl gene may be assayed utilizing standard techniques. Initial screening may be accomplished by Southern blot analysis or PCR techniques to analyze animal tissues to assay whether integration of the transgene has taken place. The level of mRNA expression of the transgene in the tissues of the transgenic animals may also be assessed using techniques which include but are not limited to Northern blot analysis of cell type samples obtained from the animal, in situ hybridization analysis, and RT-PCR. Samples of plOl gene-expressing tissue, may also be evaluated immunocytochemically using antibodies specific for the plOl transgene product, as described below.

5.3 ANTIBODIES TO p 101 AND pi 20 PROTEINS

Antibodies that specifically recognize one or more epitopes of p 101 regulatory subunit, or epitopes of conserved variants of plOl regulatory subunit, or peptide fragments of the plOl regulatory subunit are also encompassed by the invention. Also encompassed by the invention are antibodies which recognize one or more epitopes of the pl20 protein, particularly, the novel carboxyl terminus. Such antibodies include but are not limited to polyclonal antibodies, monoclonal antibodies (mAbs), humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab') 2 fragments, fragments produced by a Fab expression library,

anti-idiotypic (anti-Id) antibodies, and epi tope-binding fragments of any of the above.

The antibodies of the invention may be used, for example, in the detection of the p 101 regulatory subunit or pi 20 in a biological sample and may, therefore, be utilized as part of a diagnostic or prognostic technique whereby patients may be tested for abnormal amounts of these proteins. Such antibodies may also be utilized in conjunction with, for example, compound screening schemes, as described, below, in Section 5.5, for the evaluation of the effect of test compounds on expression and/or activity of the plOl or p!20 gene products. Additionally, such antibodies can be used in conjunction with the gene therapy techniques described, below, in Section 5.6, to, for example, evaluate the normal and/or engineered plOl regulatory subunit-expressing cells prior to their introduction into the patient. Such antibodies may additionally be used as a method for the inhibition of abnormal plOl regulatory subunit or pl20 activity. Thus, such antibodies may, therefore, be utilized as part of inflammatory disorder treatment methods.

For the production of antibodies, various host animals may be immunized by injection with the plOl regulatory subunit, a plOl regulatory subunit peptide, truncated plOl regulatory subunit polypeptides, functional equivalents of the p 101 regulatory subunit or mutants of the plOl regulatory subunit. Additionally, host animals may be immunized by injection with pi 20 catalytic subunit or peptides of the pl20 subunit. Such host animals may include but are not limited to rabbits, mice, and rats, to name but a few. Various adjutants may be used to increase the immunological response, depending on the host species, including but not limited to Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, dinitrophenol, and potentially useful human adjutants such as BCG (bacille Calmette-Guerin) and Corynebacterium parvum. Polyclonal antibodies are heterogeneous populations of antibody molecules derived from the sera of the immunized animals.

Monoclonal antibodies, which are homogeneous populations of antibodies to a particular antigen, may be obtained by any technique which provides for the production of antibody molecules by continuous cell lines in culture. These

include, but are not limited to, the hybridoma technique of Kohler and Milstein, (1975, Nature 256:495-497; and U.S. Patent No. 4,376, 1 10), the human B-cell hybridoma technique (Kosbor et al. , 1983, Immunology Today 4:72; Cole et al., 1983, Proc. Natl. Acad. Sci. USA 80:2026-2030), and the EBV-hybridoma technique (Cole et al., 1985, Monoclonal Antibodies And Cancer Therapy, Alan R. Liss, Inc., pp. 77-96). Such antibodies may be of any immunoglobulin class including IgG, IgM, IgE, IgA, IgD and any subclass thereof. The hybridoma producing the mAb of this invention may be cultivated in vitro or in vivo. Production of high titers of mAbs in vivo makes this the presently preferred method of production.

In addition, techniques developed for the production of "chimeric antibodies" (Morrison et al. , 1984, Proc. Natl. Acad. Sci. USA, 81 :6851-6855; Neuberger et al., 1984, Nature, 312:604-608; Takeda et al. , 1985, Nature, 314:452-454) by splicing the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used. A chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a porcine mAb and a human immunoglobulin constant region. Alternatively, techniques described for the production of single chain antibodies (U.S. Patent 4,946,778; Bird, 1988, Science 242:423-426; Huston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883; and Ward et al. , 1989, Nature 334:544-546) can be adapted to produce single chain antibodies against plOl gene products. Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide.

Antibody fragments which recognize specific epitopes may be generated by known techniques. For example, such fragments include but are not limited to: the F(ab') 2 fragments which can be produced by pepsin digestion of the antibody molecule and the Fab fragments which can be generated by reducing the disulfide bridges of the F(ab') 2 fragments. Alternatively, Fab expression libraries may be constructed (Huse et al. , 1989, Science, 246: 1275-1281) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity.

Antibodies to the plOl regulatory subunit or the pl20 catalytic subunit can, in turn, be utilized to generate anti-idiotype antibodies that "mimic" the plOl regulatory subunit or pl20 subunit, respectively, using techniques well known to those skilled in the art. (See, e^, Greenspan & Bona, 1993, FASEB J 7(5):437- 444; and Nissinoff, 1991, J. Immunol. 147(8): 2429-2438). For example antibodies which bind to the plOl regulatory subunit and competitively inhibit the binding of catalytic subunit to the plOl regulatory subunit can be used to generate anti- idiotypes that "mimic" the plOl regulatory subunit and, therefore, bind and neutralize catalytic subunit. Such neutralizing anti-idiotypes or Fab fragments of such anti-idiotypes can be used in therapeutic regimens to neutralize catalytic subunit and reduce inflammation.

5.4 DIAGNOSIS OF HEMATOPOIETIC CELL ACTIVATION DISORDERS A variety of methods can be employed for the diagnostic and prognostic evaluation of hematopoietic lineage cell activation disorders, including inflammatory disorders, and for the identification of subjects having a predisposition to such disorders.

Such methods may, for example, utilize reagents such as the plOl and p!20 nucleotide sequences described above, and plOl regulatory subunit and pl20 antibodies, as described, in Section 5.3. Specifically, such reagents may be used, for example, for: (1) the detection of the presence of plOl or p!20 gene mutations, or the detection of either over- or under-expression of plOl or p!20 mRNA relative to the non-hematopoietic lineage cell activation disorder state; (2) the detection of either an over- or an under-abundance of plOl or p!20 gene product relative to the non-hematopoietic lineage cell activation disorder state; and (3) the detection of perturbations or abnormalities in the signal transduction pathway mediated by p 101 regulatory subunit and pi 20 catalytic subunit.

The methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one specific plOl or p!20 nucleotide sequence or p 101 or pi 20 regulatory subunit antibody reagent described herein, which may be conveniently used, e.g.. in clinical settings, to diagnose patients exhibiting hematopoietic lineage cell activation disorder abnormalities.

For the detection of plOl or p!20 mutations, any nucleated cell can be used as a starting source for genomic nucleic acid. For the detection of plOl or p!20 gene expression or plOl or p!20 gene products, any cell type or tissue in which the plOl or p!20 gene is expressed, such as, for example, neutrophil cells, may be utilized.

Nucleic acid-based detection techniques are described, below, in Section 5.4.1. Peptide detection techniques are described, below, in Section 5.4.2.

5.4.1 DETECTION OF THE plOl GENE AND TRANSCRIPTS

Mutations within the plOl or p!20 genes can be detected by utilizing a number of techniques. Nucleic acid from any nucleated cell can be used as the starting point for such assay techniques, and may be isolated according to standard nucleic acid preparation procedures which are well known to those of skill in the art.

DNA may be used in hybridization or amplification assays of biological samples to detect abnormalities involving gene structure, including point mutations, insertions, deletions and chromosomal rearrangements. Such assays may include, but are not limited to, Southern analyses, single stranded conformational polymoφhism analyses (SSCP), and PCR analyses.

Such diagnostic methods for the detection of plOl or p!20 gene-specific mutations can involve for example, contacting and incubating nucleic acids including recombinant DNA molecules, cloned genes or degenerate variants thereof, obtained from a sample, e.g.. derived from a patient sample or other appropriate cellular source, with one or more labeled nucleic acid reagents including recombinant DNA molecules, cloned genes or degenerate variants thereof, as described above, under conditions favorable for the specific annealing of these reagents to their complementary sequences within the plOl or p!20 gene. Preferably, the lengths of these nucleic acid reagents are at least 15 to 30 nucleotides. After incubation, all non-annealed nucleic acids are removed from the nucleic acid molecule hybrid. The presence of nucleic acids which have hybridized, if any such molecules exist, is then detected. Using such a detection scheme, the nucleic acid from the cell type or tissue of interest can be

immobilized, for example, to a solid support such as a membrane, or a plastic surface such as that on a microtiter plate or polystyrene beads. In this case, after incubation, non-annealed, labeled nucleic acid reagents of the type described above are easily removed. Detection of the remaining, annealed, labeled plOl or p!20 nucleic acid reagents is accomplished using standard techniques well-known to those in the art. The plOl or pi 20 gene sequences to which the nucleic acid reagents have annealed can be compared to the annealing pattern expected from a normal gene sequence in order to determine whether a gene mutation is present. Alternative diagnostic methods for the detection of plOl or p!20 gene specific nucleic acid molecules, in patient samples or other appropriate cell sources, may involve their amplification, e.g.. by PCR (the experimental embodiment set forth in Mullis, K.B. , 1987, U.S. Patent No. 4,683,202), followed by the detection of the amplified molecules using techniques well known to those of skill in the art. The resulting amplified sequences can be compared to those which would be expected if the nucleic acid being amplified contained only normal copies of the plOl or p!20 gene in order to determine whether a gene mutation exists.

Additionally, well-known genotyping techniques can be performed to identify individuals carrying plOl or p!20 gene mutations. Such techniques include, for example, the use of restriction fragment length polymoφhisms

(RFLPs), which involve sequence variations in one of the recognition sites for the specific restriction enzyme used.

The level of plOl or p!20 gene expression can also be assayed by detecting and measuring plOl or p!20 transcription. For example, RNA from a cell type or tissue known, or suspected to express the plOl or p!20 gene, such as hematopoietic lineage cells, especially myeloid cells and platelets, may be isolated and tested utilizing hybridization or PCR techniques such as are described, above. The isolated cells can be derived from cell culture or from a patient. The analysis of cells taken from culture may be a necessary step in the assessment of cells to be used as part of a cell-based gene therapy technique or, alternatively, to test the effect of compounds on the expression of the plOl or p!20 gene. Such analyses may reveal both quantitative and qualitative aspects of the expression pattern of the

plOl or p!20 gene, including activation or inactivation of plOl or p!20 gene expression.

5.4.2 DETECTION OF THE plOl GENE PRODUCTS Antibodies directed against wild type or mutant plOl or p!20 gene products or conserved variants or peptide fragments thereof, which are discussed, above, in Section 5.3, may also be used as hematopoietic lineage cell activation disorder diagnostics and prognostics, as described herein. Such diagnostic methods, may be used to detect abnormalities in the level of plOl or p!20 gene expression, or abnormalities in the structure and/or temporal, tissue, cellular, or subcellular location of the plOl regulatory subunit, and may be performed in vivo or in vitro, such as, for example, on biopsy tissue.

The tissue or cell type to be analyzed will generally include those which are known, or suspected, to contain cells express the plOl or p!20 gene, such as, for example, neutrophil cells which have infiltrated an inflamed tissue. The protein isolation methods employed herein may, for example, be such as those described in Harlow and Lane (Harlow, E. and Lane, D. , 1988, "Antibodies: A Laboratory Manual", Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York), which is incoφorated herein by reference in its entirety. The isolated cells can be derived from cell culture or from a patient. The analysis of cells taken from culture may be a necessary step in the assessment of cells that could be used as part of a cell-based gene therapy technique or, alternatively, to test the effect of compounds on the expression of the plOl or p!20 gene.

For example, antibodies, or fragments of antibodies, such as those described, above, in Section 5.3, useful in the present invention may be used to quantitatively or qualitatively detect the presence of plOl or p!20 gene products or conserved variants or peptide fragments thereof. This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labeled antibody (see below, this Section) coupled with light microscopic, flow cytometric, or fluorimetric detection.

The antibodies (or fragments thereof) or fusion or conjugated proteins useful in the present invention may, additionally, be employed histologically, as in immunofluorescence, immunoelectron microscopy or non-immuno assays, for in

situ detection of plOl or p!20 gene products or conserved variants or peptide fragments thereof, or for catalytic subunit binding (in the case of labeled catalytic subunit fusion protein).

In situ detection may be accomplished by removing a histological specimen from a patient, and applying thereto a labeled antibody or fusion protein of the present invention. The antibody (or fragment) or fusion protein is preferably applied by overlaying the labeled antibody (or fragment) onto a biological sample. Through the use of such a procedure, it is possible to determine not only the presence of the plOl or p!20 gene product, or conserved variants or peptide fragments, 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.

Immunoassays and non-immunoassays for plOl or p!20 gene products or conserved variants or peptide fragments thereof will typically comprise incubating a sample, such as a biological fluid, a tissue extract, freshly harvested cells, or lysates of cells which have been incubated in cell culture, in the presence of a detectably labeled antibody capable of identifying plOl or p!20 gene products or conserved variants or peptide fragments thereof, and detecting the bound antibody by any of a number of techniques well-known in the art.

The biological sample may be brought in contact with and immobilized onto a solid phase support or carrier such as nitrocellulose, or other solid support which is capable of immobilizing cells, cell particles or soluble proteins. The support may then be washed with suitable buffers followed by treatment with the detectably labeled plOl regulatory subunit or pi 20 subunit antibody or fusion protein. The solid phase support may then be washed with the buffer a second time to remove unbound antibody or fusion protein. The amount of bound label on solid support may then be detected by conventional means.

"Solid phase support or carrier" is intended to encompass any support capable of binding an antigen or an antibody. Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite. The nature of the carrier can be either soluble to some extent or insoluble for the

puφoses of the present invention. The support material may have virtually any possible structural configuration so long as the 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 p 101 regulatory subunit or pi 20 subunit antibody or fusion protein 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.

With respect to antibodies, one of the ways in which the antibody can be detectably labeled is by linking the same to an enzyme and use in an enzyme immunoassay (EIA) (Voller, "The Enzyme Linked Immunosorbent Assay

(ELISA)", 1978, Diagnostic Horizons 2: 1-7, Microbiological Associates Quarterly Publication, Walkersville, MD); Voller et al. , 1978, J. Clin. Pathol. 31:507-520; Butler, 1981, Meth. Enzymol. 73:482-523; Maggio (ed.), 1980, Enzyme Immunoassay, CRC Press, Boca Raton, FL,; Ishikawa et al. , (eds.), 1981 , Enzyme Immunoassay, Kgaku Shoin, Tokyo). The enzyme which is bound to the antibody will react with an appropriate substrate, preferably a chromogenic 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 also be accomplished using any of a variety of other immunoassays. For example, by radioactively labeling the antibodies or antibody fragments, it is possible to detect plOl regulatory subunit through the use of a radioimmunoassay (RIA) (see, for example, Weintraub, B., Principles of Radioimmunoassays, Seventh Training Course on Radioligand Assay Techniques, The Endocrine Society, March, 1986, which is incoφorated 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 labeling compounds are fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin and fluorescamine.

The antibody can also be detectably labeled using fluorescence emitting metals such as 152 Eu, or others of the lanthanide series. These metals can be attached to the antibody using such metal chelating groups as diethylenetriaminepentacetic 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. Examples 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 chemiluminescence found in biological systems in, which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence. Important bioluminescent compounds for puφoses of labeling are luciferin, luciferase and aequorin.

5.5 SCREENING ASSAYS FOR COMPOUNDS THAT MODULATE G-PROTEIN ACTIVATED PI3K EXPRESSION OR ACTIVITY

The following assays are designed to identify compounds that interact with

(e.g.. bind to) plOl regulatory subunit or the pi 20 cat ytic subunit, compounds that interact with (e.g.. bind to) intracellular proteins that interact with plOl regulatory subunit and/or the pl20 catalytic subunit, compounds that interfere with the interaction of plOl regulatory subunit with the pl20 catalytic subunit or with other intracellular proteins involved in G protein stimulated PI3K mediated signal transduction, and to compounds which modulate the activity of plOl or p!20 gene (i.e.. modulate the level of plOl or p!20 gene expression) or modulate the level of plOl or pl20. Assays may additionally be utilized which identify compounds which bind to plOl or pi 20 gene regulatory sequences (e.g.. promoter sequences) and which may modulate plOl or p!20 gene expression. See e.g.. Platt, K.A. , 1994, J. Biol. Chem. 269:28558-28562, which is incoφorated herein by reference in its entirety.

The compounds which may be screened in accordance with the invention include but are not limited to peptides, antibodies and fragments thereof, prostaglandins, lipids and other organic compounds (e.g.. teφines, peptidomimetics) that bind to the p 101 regulatory subunit or pi 20 catalytic subunit and either mimic the activity triggered by the natural ligand (i.e.. agonists) or inhibit the activity triggered by the natural ligand (i.e. , antagonists); as well as peptides, antibodies or fragments thereof, and other organic compounds that mimic the p 101 regulatory subunit or the pi 20 catalytic subunit (or a portion thereof) and bind to and "neutralize" natural ligand. Such compounds may include, but are not limited to, peptides such as, for example, soluble peptides, including but not limited to members of random peptide libraries (see, e^, Lam, K.S. et al., 1991, Nature 354:82-84; Houghten, R. et al. , 1991, Nature 354:84-86), and combinatorial chemistry-derived molecular library peptides made of D- and/or L- configuration amino acids, phosphopeptides (including, but not limited to members of random or partially degenerate, directed phosphopeptide libraries; see, e.g. , Songyang, Z. et al., 1993, Cell 72:767-778); antibodies (including, but not limited to, polyclonal, monoclonal, humanized, anti- idiotypic, chimeric or single chain antibodies, and FAb, F(ab') 2 and FAb

expression library fragments, and epitope-binding fragments thereof); and small organic or inorganic molecules.

Other compounds which can be screened in accordance with the invention include but are not limited to small organic molecules that are able to gain entry into an appropriate cell (e.g. , in the neutrophil) and affect the expression of the plOl or p!20 gene or some other gene involved in the pi 01 regulatory subunit signal transduction pathway (e.g.. by interacting with the regulatory region or transcription factors involved in gene expression); or such compounds that affect the activity of the p 101 regulatory subunit, e.g.. by inhibiting or enhancing the binding of plOl to the catalytic subunit of the PI3K or the binding of plOl to some other intracellular factor involved in the plOl regulatory subunit signal transduction pathway, such as, for example, G fy.

Computer modelling and searching technologies permit identification of compounds, or the improvement of already identified compounds, that can modulate plOl regulatory subunit or pl20 catalytic subunit expression or activity. Having identified such a compound or composition, the active sites or regions are identified. Such active sites might typically be the binding partner sites, such as, for example, the interaction domains of the pl20 catalytic subunit with plOl regulatory subunit itself. The active site can be identified using methods known in the .art including, for example, from the amino acid sequences of peptides. from the nucleotide sequences of nucleic acids, or from study of complexes of the relevant compound or composition with its natural ligand. In the latter case, chemical or X-ray crystallographic methods can be used to find the active site by finding where on the factor the complexed ligand is found. Next, the three dimensional geometric structure of the active site is determined. This can be done by known methods, including X-ray crystallography, which can determine a complete molecular structure. On the other hand, solid or liquid phase NMR can be used to determine certain intra-molecular distances. Any other experimental method of structure determination can be used to obtain partial or complete geometric structures. The geometric structures may be measured with a complexed ligand, natural or artificial, which may increase the accuracy of the active site structure determined.

If an incomplete or insufficiently accurate structure is determined, the methods of computer based numerical modelling can be used to complete the structure or improve its accuracy. Any recognized modelling method may be used, including parameterized models specific to particular biopolymers such as proteins or nucleic acids, molecular dynamics models based on computing molecular motions, statistical mechanics models based on thermal ensembles, or combined models. For most types of models, standard molecular force fields, representing the forces between constituent atoms and groups, are necessary, and can be selected from force fields known in physical chemistry. The incomplete or less accurate experimental structures can serve as constraints on the complete and more accurate structures computed by these modeling methods.

Finally, having determined the structure of the active site, either experimentally, by modeling, or by a combination, candidate modulating compounds can be identified by searching databases containing compounds along with information on their molecular structure. Such a search seeks compounds having structures that match the determined active site structure and that interact with the groups defining the active site. Such a search can be manual, but is preferably computer assisted. These compounds found from this search are potential G protein activated PI3K modulating compounds. Alternatively, these methods can be used to identify improved modulating compounds from an already known modulating compound or ligand. The composition of the known compound can be modified and the structural effects of modification can be determined using the experimental and computer modelling methods described above applied to the new composition. The altered structure is then compared to the active site structure of the compound to determine if an improved fit or interaction results. In this manner systematic variations in composition, such as by varying side groups, can be quickly evaluated to obtain modified modulating compounds or ligands of improved specificity or activity. Further experimental and computer modeling methods useful to identify modulating compounds based upon identification of the active sites of pl20 catalytic subunit, plOl regulatory subunit, and related transduction and transcription factors will be apparent to those of skill in the art.

Examples of molecular modeling systems are the CHARMm and QUANTA programs (Polygen Coφoration, Waltham, MA). CHARMm performs the energy minimization and molecular dynamics functions. QUANTA performs the construction, graphic modelling and analysis of molecular structure. QUANTA allows interactive construction, modification, visualization, and analysis of the behavior of molecules with each other.

A number of articles review computer modelling of drugs interactive with specific proteins, such as Rotivinen et al. , 1988, Acta Pharmaceutical Fennica 97: 159-166; Ripka, New Scientist 54-57 (June 16, 1988); McKinaly and Rossmann, 1989, Annu. Rev. Pharmacol. Toxiciol. 29: 1 1 1-122; Perry and Davies, OSAR: Quantitative Structure-Activity Relationships in Drug Design pp. 189-193 (Alan R. Liss, Inc. 1989); Lewis and Dean, 1989, Proc. R. Soc. Lond. 236: 125- 140 and 141-162; and, with respect to a model receptor for nucleic acid components, Askew et al. , 1989, J. Am. Chem. Soc. 1 11 : 1082-1090. Other computer programs that screen and graphically depict chemicals are available from companies such as BioDesign, Inc. (Pasadena, CA.), Allelix, Inc. (Mississauga, Ontario, Canada), and Hypercube, Inc. (Cambridge, Ontario). Although these are primarily designed for application to drugs specific to particular proteins, they can be adapted to design of drugs specific to regions of DNA or RNA, once that region is identified.

Although described above with reference to design and generation of compounds which could alter binding, one could also screen libraries of known compounds, including natural products or synthetic chemicals, and biologically active materials, including proteins, for compounds which are inhibitors or activators.

Compounds identified via assays such as those described herein may be useful, for example, in elaborating the biological function of the plOl or p!20 gene product, and for ameliorating hematopoietic lineage cell activation disorders. Assays for testing the effectiveness of compounds, identified by, for example, techniques such as those described in Section 5.5. 1 through 5.5.3, are discussed, below, in Section 5.5.4.

5.5.1 In vitro SCREENING ASSAYS FOR COMPOUNDS

THAT BIND TO plOl REGULATORY SUBUNIT

In vitro systems may be designed to identify compounds capable of interacting with (e.g. , binding to) plOl regulatory subunit or pl20 catalytic subunit. Compounds identified may be useful, for example, in modulating the activity of wild type and/or mutant plOl or p!20 gene products; may be utilized in screens for identifying compounds that disrupt normal plOl regulatory subunit/catalytic subunit interactions; or may in themselves disrupt such interactions.

The principle of the assays used to identify compounds that bind to the plOl regulatory subunit involves preparing a reaction mixture of the p 101 regulatory subunit and the test compound under conditions and for a time sufficient to allow the two components to interact and bind, thus forming a complex which can be removed and/or detected in the reaction mixture. The plOl regulatory subunit species used can vary depending upon the goal of the screening assay. For example, where agonists of the natural ligand are sought, the full length plOl regulatory subunit, or a fusion protein containing the p 101 regulatory subunit fused to a protein or polypeptide that affords advantages in the assay system (e.g.. labeling, isolation of the resulting complex, etc.) can be utilized.

The screening assays can be conducted in a variety of ways. For example, one method to conduct such an assay would involve anchoring the p 101 regulatory subunit protein, polypeptide, peptide or fusion protein or the test substance onto a solid phase and detecting plOl regulatory subunit/ test compound complexes anchored on the solid phase at the end of the reaction. In one embodiment of such a method, the p 101 regulatory subunit reactant may be anchored onto a solid surface, and the test compound, which is not anchored, may be labeled, either directly or indirectly. In another embodiment of the method, a plOl regulatory subunit protein anchored on the solid phase is complexed with labeled catalytic subunit such as pl20. Then, a test compound could be assayed for its ability to disrupt the association of the pl01/pl20 complex.

In practice, microtiter plates may conveniently be utilized as the solid phase. The anchored component may be immobilized by non-covalent or covalent attachments. Non-covalent attachment may be accomplished by simply coating the

solid surface with a solution of the protein and drying. Alternatively, an immobilized antibody, preferably a monoclonal antibody, specific for the protein to be immobilized may be used to anchor the protein to the solid surface. The surfaces may be prepared in advance and stored. In order to conduct the assay, the nonimmobilized component is added to the coated surface containing the anchored component. After the reaction is complete, unreacted components are removed (e.g. , by washing) under conditions such that any complexes formed will remain immobilized on the solid surface. The detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the previously nonimmobilized component is pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed. Where the previously nonimmobilized component is not pre-labeled, an indirect label can be used to detect complexes anchored on the surface; e.g.. using a labeled antibody specific for the previously nonimmobilized component (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti- Ig antibody).

Alternatively, a reaction can be conducted in a liquid phase, the reaction products separated from unreacted components, and complexes detected; e.g.. using an immobilized antibody specific for plOl regulatory subunit protein, polypeptide, peptide or fusion protein, or the catalytic subunit protein or fusion protein, or the test compound to anchor any complexes formed in solution, and a labeled antibody specific for the other component of the possible complex to detect anchored complexes.

5.5.2 ASSAYS FOR INTRACELLULAR PROTEINS

THAT INTERACT WITH THE plOl OR pl20 PROTEINS

Any method suitable for detecting protein-protein interactions may be employed for identifying intracellular proteins that interact with plOl regulatory subunit and/or the catalytic subunit pi 20. Among the traditional methods which may be employed are co-immunoprecipitation, crosslinking and co-purification through gradients or chromatographic columns of cell lysates or proteins obtained from cell lysates and the plOl regulatory subunit to identify proteins in the lysate

that interact with the plOl regulatory subunit. For these assays, the plOl regulatory subunit component used can be a full length plOl regulatory subunit, or a truncated peptide. Similarly, the component may be a pi 20 catalytic subunit, or a complex of the plOl regulatory subunit with the pl20 catalytic subunit. Once isolated, such an intracellular protein can be identified and can, in turn, be used, in conjunction with standard techniques, to identify proteins with which it interacts. For example, at least a portion of the amino acid sequence of an intracellular protein which interacts with the p 101 regulatory subunit, PI3K (p 101 /p 120 complex), or pl20 catalytic subunit, can be ascertained using techniques well known to those of skill in the art, such as via the Edman degradation technique. (See, e^, Creighton, 1983, "Proteins: Structures and Molecular Principles", W.H. Freeman & Co. , N.Y. , pp.34-49). The amino acid sequence obtained may be used as a guide for the generation of oligonucleotide mixtures that can be used to screen for gene sequences encoding such intracellular proteins. Screening may be accomplished, for example, by standard hybridization or PCR techniques. Techniques for the generation of oligonucleotide mixtures and the screening are well-known. (See, e.g. , Ausubel, supra. , and PCR Protocols: A Guide to Methods and Applications, 1990, Innis, M. et al., eds. Academic Press, Inc., New York). Additionally, methods may be employed which result in the simultaneous identification of genes which encode the intracellular proteins interacting with plOl regulatory subunit and/or the pl20 catalytic subunit and/or the PI3K. These methods include, for example, probing expression, libraries, in a manner similar to the well known technique of antibody probing of λgtl l libraries, using labeled plOl regulatory subunit protein, or a p 101 regulatory subunit polypeptide, peptide or fusion protein, e.g., a plOl regulatory subunit polypeptide or plOl regulatory subunit domain fused to a marker (e.g.. an enzyme, fluor, luminescent protein, or dye), or an Ig-Fc domain.

One method which detects protein interactions in vivo, the two-hybrid system, is described in detail for illustration only and not by way of limitation. One version of this system has been described (Chien et al. , 1991, Proc. Natl. Acad. Sci. USA, 88:9578-9582) and is commercially available from Clontech (Palo Alto, CA).

Briefly, utilizing such a system, plasmids are constructed that encode two hybrid proteins: one plasmid consists of nucleotides encoding the DNA-binding domain of a transcription activator protein fused to a plOl nucleotide sequence encoding plOl regulatory subunit, a plOl regulatory subunit polypeptide, peptide or fusion protein, and the other plasmid consists of nucleotides encoding the transcription activator protein's activation domain fused to a cDNA encoding an unknown protein which has been recombined into this plasmid as part of a cDNA library. The DNA-binding domain fusion plasmid and the cDNA library are transformed into a strain of the yeast Saccharomvces cerevisiae that contains a reporter gene (e.g.. HBS or lacZ) whose regulatory region contains the transcription activator's binding site. Either hybrid protein alone cannot activate transcription of the reporter gene; the DNA-binding domain hybrid cannot because it does not provide activation function, and the activation domain hybrid cannot because it cannot localize to the activator's binding sites. Interaction of the two hybrid proteins reconstitutes the functional activator protein and results in expression of the reporter gene, which is detected by an assay for the reporter gene product.

The two-hybrid system or related methodology may be used to screen activation domain libraries for proteins that interact with the "bait" gene product. By way of example, and not by way of limitation, plOl regulatory subunit may be used as the bait gene product. Total genomic or cDNA sequences are fused to the DNA encoding an activation domain. This library and a plasmid encoding a hybrid of a bait plOl gene product fused to the DNA-binding domain are cotransformed into a yeast reporter strain, and the resulting transformants are screened for those that express the reporter gene. For example, and not by way of limitation, a bait plOl gene sequence, such as the open reading frame of plOl (or a domain of plOl), as depicted in FIG. 1 can be cloned into a vector such that it is translationally fused to the DNA encoding the DNA-binding domain of the GAL4 protein. These colonies are purified and the library plasmids responsible for reporter gene expression are isolated. DNA sequencing is then used to identify the proteins encoded by the library plasmids.

A cDNA library of the cell line from which proteins that interact with bait plOl gene product are to be detected can be made using methods routinely

practiced in the art. According to the particular system described herein, for example, the cDNA fragments can be inserted into a vector such that they are translationally fused to the transcriptional activation domain of GAL4. This library can be co-transfected along with the bait plOl gene-GAL4 fusion plasmid into a yeast strain which contains a lacZ gene driven by a promoter which contains GAL4 activation sequence. A cDNA encoded protein, fused to GAL4 transcriptional activation domain, that interacts with bait plOl gene product will reconstitute an active GAL4 protein and thereby drive expression of the HIS3 gene. Colonies which express HIS3 can be detected by their growth on petri dishes containing semi-solid agar based media lacking histidine. The cDNA can then be purified from these strains, and used to produce and isolate the bait plOl gene- interacting protein using techniques routinely practiced in the art.

5.5.3 ASSAYS FOR COMPOUNDS THAT INTERFERE WITH plOl REGULATORY

SUBUNIT/INTRACELLULAR MACROMOLECULE INTERACTION

The macromolecules that interact with the plOl regulatory subunit are referred to, for puφoses of this discussion, as "binding partners" . These binding partners are likely to be involved in the plOl regulatory subunit signal transduction pathway, and therefore, in the role of p 101 regulatory subunit in hematopoietic lineage cell activation regulation. Known binding partners are catalytic subunits of the PI3K kinase such as pi 20, pi 17, and perhaps certain pi 10 proteins. Other binding partners are likely to be activated trimeric G proteins such as G/3γ subunits, and or lipids. Therefore, it is desirable to identify compounds that interfere with or disrupt the interaction of such binding partners with plOl which may be useful in regulating the activity of the p 101 regulatory subunit and thus control hematopoietic lineage cell activation disorders associated with plOl regulatory subunit activity. The basic principle of the assay systems used to identify compounds that interfere with the interaction between the plOl regulatory subunit and its binding partner or partners involves preparing a reaction mixture containing plOl regulatory subunit protein, polypeptide, peptide or fusion protein as described in Sections 5.5.1 and 5.5.2 above, and the binding partner under conditions and for a

time sufficient to allow the two to interact and bind, thus forming a complex. In order to test a compound for inhibitory activity, the reaction mixture is prepared in the presence and absence of the test compound. The test compound may be initially included in the reaction mixture, or may be added at a time subsequent to the addition of the plOl regulatory subunit moiety and its binding partner. Control reaction mixtures are incubated without the test compound or with a placebo. The formation of any complexes between the p 101 regulatory subunit moiety and the binding partner is then detected. The formation of a complex in the control reaction, but not in the reaction mixture containing the test compound, indicates that the compound interferes with the interaction of the p 101 regulatory subunit and the interactive binding partner. Additionally, complex formation within reaction mixtures containing the test compound and normal plOl regulatory subunit protein may also be compared to complex formation within reaction mixtures containing the test compound and a mutant plOl regulatory subunit. This comparison may be important in those cases wherein it is desirable to identify compounds that disrupt interactions of mutant but not normal plOl regulatory subunits.

The assay for compounds that interfere with the interaction of the p 101 regulatory subunit and binding partners can be conducted in a heterogeneous or homogeneous format. Heterogeneous assays involve anchoring either the plOl regulatory subunit moiety product or the binding partner onto a solid phase and detecting complexes anchored on the solid phase at the end of the reaction. In homogeneous assays, the entire reaction is carried out in a liquid phase. In either approach, the order of addition of reactants can be varied to obtain different information about the compounds being tested. For example, test compounds that interfere with the interaction by competition can be identified by conducting the reaction in the presence of the test substance; L , by adding the test substance to the reaction mixture prior to or simultaneously with the p 101 regulatory subunit moiety and interactive binding partner. Alternatively, test compounds that disrupt preformed complexes, e.g. compounds with higher binding constants that displace one of the components from the complex, can be tested by adding the test compound to the reaction mixture after complexes have been formed. The various formats are described briefly below.

In a heterogeneous assay system, either the p 101 regulatory subunit moiety or the interactive binding partner, is anchored onto a solid surface, while the non- anchored species is labeled, either directly or indirectly. In practice, microtiter plates are conveniently utilized. The anchored species may be immobilized by non-covalent or covalent attachments. Non-covalent attachment may be accomplished simply by coating the solid surface with a solution of the plOl or p!20 gene product or binding partner and drying. Alternatively, an immobilized antibody specific for the species to be anchored may be used to anchor the species to the solid surface. The surfaces may be prepared in advance and stored. In order to conduct the assay, the partner of the immobilized species is exposed to the coated surface with or without the test compound. After the reaction is complete, unreacted components are removed (e.g.. by washing) and any complexes formed will remain immobilized on the solid surface. The detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the non-immobilized species is pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed. Where the non¬ immobilized species is not pre-labeled, an indirect label can be used to detect complexes anchored on the surface; e.g. , using a labeled antibody specific for the initially non- immobilized species (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody). Depending upon the order of addition of reaction components, test compounds which inhibit complex formation or which disrupt preformed complexes can be detected.

Alternatively, the reaction can be conducted in a liquid phase in the presence or absence of the test compound, the reaction products separated from unreacted components, and complexes detected; e.g.. using an immobilized antibody specific for one of the binding components to anchor any complexes formed in solution, and a labeled antibody specific for the other partner to detect anchored complexes. Again, depending upon the order of addition of reactants to the liquid phase, test compounds which inhibit complex or which disrupt preformed complexes can be identified.

In an alternate embodiment of the invention, a homogeneous assay can be used. In this approach, a preformed complex of the plOl regulatory subunit moiety and the interactive binding partner is prepared in which either the plOl

regulatory subunit or its binding partners is labeled, but the signal generated by the label is quenched due to formation of the complex (see, e.g. , U.S. Patent No. 4,109,496 by Rubenstein which utilizes this approach for immunoassays). The addition of a test substance that competes with and displaces one of the species from the preformed complex will result in the generation of a signal above background. In this way, test substances which disrupt plOl regulatory subunit/intracellular binding partner interaction can be identified.

In a particular embodiment, a plOl regulatory subunit fusion can be prepared for immobilization. For example, the plOl regulatory subunit or a peptide fragment, e.g. , corresponding to the CD, can be fused to a glutathione-S- transferase (GST) gene using a fusion vector, such as pGEX-5X-l, in such a manner that its binding activity is maintained in the resulting fusion protein. The interactive binding partner can be purified and used to raise a monoclonal antibody, using methods routinely practiced in the art and described above, in Section 5.3. This antibody can be labeled with the radioactive isotope 125 I, for example, by methods routinely practiced in the art. In a heterogeneous assay, e.g.. the GST- plOl regulatory subunit fusion protein can be anchored to glutathione-agarose beads. The interactive binding partner can then be added in the presence or absence of the test compound in a manner that allows interaction and binding to occur. At the end of the reaction period, unbound material can be washed away, and the labeled monoclonal antibody can be added to the system and allowed to bind to the complexed components. The interaction between the plOl or p!20 gene product and the interactive binding partner can be detected by measuring the amount of radioactivity that remains associated with the glutathione-agarose beads. A successful inhibition of the interaction by the test compound will result in a decrease in measured radioactivity.

Alternatively, the GST-plOl regulatory subunit fusion protein and the interactive binding partner can be mixed together in liquid in the absence of the solid glutathione-agarose beads. The test compound can be added either during or after the species are allowed to interact. This mixture can then be added to the glutathione-agarose beads and unbound material is washed away. Again the extent of inhibition of the p 101 regulatory subunit/binding partner interaction can be

detected by adding the labeled antibody and measuring the radioactivity associated with the beads.

In another embodiment of the invention, these same techniques can be employed using peptide fragments that correspond to the binding domains of the plOl regulatory subunit and/or the interactive or binding partner (in cases where the binding partner is a protein), in place of one or both of the full length proteins. Any number of methods routinely practiced in the art can be used to identify and isolate the binding sites. These methods include, but are not limited to, mutagenesis of the gene encoding one of the proteins and screening for disruption of binding in a co-immunoprecipitation assay. Compensating mutations in the gene encoding the second species in the complex can then be selected. Sequence analysis of the genes encoding the respective proteins will reveal the mutations that correspond to the region of the protein involved in interactive binding. Alternatively, one protein can be anchored to a solid surface using methods described above, and allowed to interact with and bind to its labeled binding partner, which has been treated with a proteolytic enzyme, such as trypsin. After washing, a short, labeled peptide comprising the binding domain may remain associated with the solid material, which can be isolated and identified by amino acid sequencing. Also, once the gene coding for the intracellular binding partner is obtained, short gene segments can be engineered to express peptide fragments of the protein, which can then be tested for binding activity and purified or synthesized.

For example, and not by way of limitation, a plOl gene product can be anchored to a solid material as described, above, by making a GST-plOl regulatory subunit fusion protein and allowing it to bind to glutathione agarose beads. The interactive binding partner can be labeled with a radioactive isotope, such as 35 S, and cleaved with a proteolytic enzyme such as trypsin. Cleavage products can then be added to the anchored GST-plOl fusion protein and allowed to bind. After washing away unbound peptides, labeled bound material, representing the intracellular binding partner binding domain, can be eluted, purified, and analyzed for amino acid sequence by well-known methods. Peptides so identified can be produced synthetically or fused to appropriate facilitative proteins using recombinant DNA technology.

5.5.4 ASSAYS FOR IDENTIFICATION OF

COMPOUNDS THAT AMELIORATE INFLAMMATORY DISORDERS Compounds, including but not limited to binding compounds identified via assay techniques such as those described, above, in Sections 5.5.1 through 5.5.3, can be tested for the ability to ameliorate immune system disorder symptoms, including inflammation. The assays described above can identify compounds which affect plOl regulatory subunit activity (e.g. , compounds that bind to the plOl regulatory subunit, inhibit binding of the natural ligands, and either activate signal transduction (agonists) or block activation (antagonists), and compounds that bind to a natural ligand of the p 101 regulatory subunit and neutralize the ligand activity); or compounds that affect plOl or p!20 gene activity (by affecting plOl or p!20 gene expression, including molecules, e.g. , proteins or small organic molecules, that affect or interfere with splicing events so that expression of the full length or the truncated form of the p 101 regulatory subunit can be modulated). However, it should be noted that the assays described herein can also identify compounds that modulate plOl regulatory subunit signal transduction (e.g.. compounds which affect downstream signaling events, such as inhibitors or enhancers of activities which participate in transducing the PtdIns(4,5)P 3 signal which is generated by catalytic subunit binding to the plOl regulatory subunit). The identification and use of such compounds which affect another step in the p 101 regulatory subunit signal transduction pathway in which the plOl or p!20 gene and/or plOl or p!20 gene product is involved and, by affecting this same pathway may modulate the effect of plOl regulatory subunit on the development of hematopoietic lineage cell activation disorders are within the scope of the invention. Such compounds can be used as part of a therapeutic method for the treatment of hematopoietic lineage cell activation disorders.

The invention encompasses cell-based and animal model-based assays for the identification of compounds exhibiting such an ability to ameliorate hematopoietic lineage cell activation disorder symptoms. Such cell-based assay systems can also be used as the standard to assay for purity and potency of the natural ligand, catalytic subunit, including recombinantly or synthetically produced catalytic subunit and catalytic subunit mutants.

Cell-based systems can be used to identify compounds which may act to ameliorate hematopoietic lineage cell activation disorder symptoms. Such cell systems can include, for example, recombinant or non-recombinant cells, such as cell lines, which express the plOl and/or p!20 gene. For example leukocyte cells, or cell lines derived from leukocyte cells can be used. In addition, expression host cells (e.g.. COS cells, CHO cells, fibroblasts, Sf9 cells) genetically engineered to express a functional pl01/pl20 PI3K and to respond to activation by the natural ligand G/3γ subunits, e.g., as measured by a chemical or phenotypic change, induction of another host cell gene, change in intracellular messenger levels (e.g.. PtdIns(3,4,5)P 3 , etc.), can be used as an end point in the assay.

In utilizing such cell systems, cells may be exposed to a compound suspected of exhibiting an ability to ameliorate hematopoietic lineage cell activation disorder symptoms, at a sufficient concentration and for a time sufficient to elicit such an amelioration of hematopoietic lineage cell activation disorder symptoms in the exposed cells. After exposure, the cells can be assayed to measure alterations in the expression of the plOl or p!20 gene, e.g., by assaying cell lysates for plOl or p!20 mRNA transcripts (e.g. , by Northern analysis) or for plOl or p!20 protein expressed in the cell; compounds which regulate or modulate expression of the plOl or p!20 gene are valuable candidates as therapeutics. Alternatively, the cells are examined to determine whether one or more hematopoietic lineage cell activation disorder-like cellular phenotypes has been altered to resemble a more normal or more wild type phenotype, or a phenotype more likely to produce a lower incidence or severity of disorder symptoms. Still further, the expression and/or activity of components of the signal transduction pathway of which plOl regulatory subunit is a part, or the activity of the plOl regulatory subunit signal transduction pathway itself can be assayed.

For example, after exposure of the cells, cell lysates can be assayed for the presence of increased levels of the second messenger PtdIns(3,4,5)P 3 , compared to lysates derived from unexposed control cells. The ability of a test compound to inhibit production of second messenger in these assay systems indicates that the test compound inhibits signal transduction initiated by p 101 regulatory subunit activation. The cell lysates can be readily assayed using anion-exchange HPLC. Alternatively, levels of superoxide production or O 2 may be assayed by monitoring

chemiluminescence from horseradish-peroxidase catalyzed luminol oxidation as described in Wymann et al., 1987, Anal. Biochem. 165:371-378, incoφorated herein by reference in its entirety. Finally, a change in cellular adhesion of intact cells may be assayed using techniques well known to those of skill in the art. In addition, animal-based hematopoietic lineage cell activation disorder systems, which may include, for example, mice, may be used to identify compounds capable of ameliorating hematopoietic lineage cell activation disorder¬ like symptoms. Such animal models may be used as test systems for the identification of drugs, pharmaceuticals, therapies and interventions which may be effective in treating such disorders. For example, animal models may be exposed to a compound, suspected of exhibiting an ability to ameliorate hematopoietic lineage cell activation disorder symptoms, at a sufficient concentration and for a time sufficient to elicit such an amelioration of hematopoietic lineage cell activation disorder symptoms in the exposed animals. The response of the animals to the exposure may be monitored by assessing the reversal of disorders associated with hematopoietic lineage cell activation disorders such as inflammation. With regard to intervention, any treatments which reverse any aspect of hematopoietic lineage cell activation disorder-like symptoms should be considered as candidates for human hematopoietic lineage cell activation disorder therapeutic intervention. Dosages of test agents may be determined by deriving dose-response curves, as discussed below.

5.6 THE TREATMENT OF DISORDERS ASSOCIATED WITH STIMULATION OF G-PROTEIN ACTIVATED PI3K, INCLUDING INFLAMMATORY DISORDERS

The invention also encompasses methods and compositions for modifying hematopoietic lineage cell activation and treating hematopoietic lineage cell activation disorders, including inflammatory disorders. For example, by decreasing the level of plOl gene expression, and/or plOl regulatory subunit gene activity, and/or downregulating activity of the p 101 regulatory subunit pathway (e.g. , by interfering with the interaction of plOl regulatory subunit with the pl20 catalytic subunit, or by targeting downstream signalling events), the response of leukocyte cells to factors which activate trimeric G protein associated receptors,

such as cytokines, may be reduced and the symptoms of chronic inflammatory diseases ameliorated. Conversely, the response of leukocyte cells to activation of G protein associated receptors may be augmented by increasing plOl regulatory subunit activity. For example, such augmentation may serve to boost the response of the immune system to infections. Different approaches are discussed below.

5.6.1 INHIBITION OF plOl ADAPTOR EXPRESSION OR plOl ADAPTOR ACTIVITY TO REDUCE G PROTEIN ACTIVATED PI3K ACTIVITY AND REDUCE INFLAMMATION

Any method which neutralizes catalytic subunit or inhibits expression of the plOl or p!20 gene (either transcription or translation) can be used to reduce the inflammatory response. Such approaches can be used to treat inflammatory response disorders such as arthritis, including rheumatoid arthritis, septic shock, adult respiratory distress syndrome (ARDS), pneumonia, asthma and other lung conditions, allergies, reperfusion injury, atherosclerosis and other cardiovascular diseases, Alzheimer's disease, and cancer, to name just a few inflammatory disorders.

In one embodiment, immuno therapy can be designed to reduce the level of endogenous plOl or p!20 gene expression, e.g.. using antisense or ribozyme approaches to inhibit or prevent translation of plOl or p!20 mRNA transcripts; triple helix approaches to inhibit transcription of the plOl or p!20 gene; or targeted homologous recombination to inactivate or "knock out" the plOl or p!20 gene or its endogenous promoter. Antisense approaches involve the design of oligonucleotides (either DNA or

RNA) that are complementary to p 101 or pi 20 regulatory subunit mRNA. The antisense oligonucleotides will bind to the complementary plOl or p!20 mRNA transcripts and prevent translation. Absolute complementarity, although preferred, is not required. A sequence "complementary" to a portion of an RNA, as referred to herein, means a sequence having sufficient complementarity to be able to hybridize with the RNA, forming a stable duplex. In the case of double-stranded antisense nucleic acids, a single strand of the duplex DNA may thus be tested, or triplex formation may be assayed. The ability to hybridize will depend on both the degree of complementarity and the length of the antisense nucleic acid. Generally,

the longer the hybridizing nucleic acid, the more base mismatches with an RNA it may contain and still form a stable duplex (or triplex, as the case may be). One skilled in the art can ascertain a tolerable degree of mismatch by use of standard procedures to determine the melting point of the hybridized complex. Oligonucleotides that are complementary to the 5' end of the message, e.g.. the 5' untranslated sequence up to and including the AUG initiation codon, should work most efficiently at inhibiting translation. However, sequences complementary to the 3 ' untranslated sequences of mRNAs have recently shown to be effective at inhibiting translation of mRNAs as well. See generally, Wagner, R., 1994, Nature 372:333-335. Thus, oligonucleotides complementary to either the 5'- or 3'- non- translated, non-coding regions of the plOl or p!20 shown in FIG. 1 and FIG. 3 could be used in an antisense approach to inhibit translation of endogenous plOl or p!20 mRNA. Oligonucleotides complementary to the 5' untranslated region of the mRNA should include the complement of the AUG start codon. Antisense oligonucleotides complementary to mRNA coding regions are less efficient inhibitors of translation but could be used in accordance with the invention. Whether designed to hybridize to the 5'-, 3'- or coding region of p 101 regulatory subunit mRNA, antisense nucleic acids should be at least six nucleotides in length, and are preferably oligonucleotides ranging from 6 to about 50 nucleotides in length. In specific aspects the oligonucleotide is at least 10 nucleotides, at least 17 nucleotides, at least 25 nucleotides or at least 50 nucleotides.

Regardless of the choice of target sequence, it is preferred that in vitro studies are first performed to quantitate the ability of the antisense oligonucleotide to inhibit gene expression. It is preferred that these studies utilize controls that distinguish between antisense gene inhibition and nonspecific biological effects of oligonucleotides. It is also preferred that these studies compare levels of the target RNA or protein with that of an internal control RNA or protein. Additionally, it is envisioned that results obtained using the antisense oligonucleotide are compared with those obtained using a control oligonucleotide. It is preferred that the control oligonucleotide is of approximately the same length as the test oligonucleotide and that the nucleotide sequence of the oligonucleotide differs from the antisense sequence no more than is necessary to prevent specific hybridization to the target sequence.

The oligonucleotides can be DNA or RNA or chimeric mixtures or derivatives or modified versions thereof, single-stranded or double-stranded. The oligonucleotide can be modified at the base moiety, sugar moiety, or phosphate backbone, for example, to improve stability of the molecule, hybridization, etc. The oligonucleotide may include other appended groups such as peptides (ej^, for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, e.g., Letsinger et al. , 1989, Proc. Natl. Acad. Sci. U.S.A. 86:6553-6556; Lemaitre et al. , 1987, Proc. Natl. Acad. Sci. 84:648-652; PCT Publication No. WO88/09810, published December 15, 1988), or hybridization- triggered cleavage agents. (See, e.g. , Krol et al., 1988, BioTechniques 6:958-976) or intercalating agents. (See, e.g. , Zon, 1988, Pharm. Res. 5:539-549). To this end, the oligonucleotide may be conjugated to another molecule, e.g. , a peptide, hybridization triggered cross-linking agent, transport agent, hybridization-triggered cleavage agent, etc. The antisense oligonucleotide may comprise at least one modified base moiety which is selected from the group including but not limited to 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl- 2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D- galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine,

1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta- D-mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxy acetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine. The antisense oligonucleotide may also comprise at least one modified sugar moiety selected from the group including but not limited to arabinose, 2-fluoroarabinose, xylulose, and hexose.

In another embodiment, the antisense oligonucleotide comprises at least one modified phosphate backbone selected from the group consisting of a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phospho triester, and a formacetal or analog thereof.

In yet another embodiment, the antisense oligonucleotide is an α-anomeric oligonucleotide. An α-anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual 3-units, the strands run parallel to each other (Gautier et al. , 1987, Nucl. Acids Res. 15:6625- 6641). The oligonucleotide is a 2 '-O-methylribonucleotide (Inoue et al. , 1987, Nucl. Acids Res. 15:6131-6148), or a chimeric RNA-DNA analogue (Inoue et al., 1987, FEBS Lett. 215:327-330).

Oligonucleotides of the invention may be synthesized by standard methods known in the art, e.g. by use of an automated DNA synthesizer (such as are commercially available from Biosearch, Applied Biosystems, etc.). As examples, phosphorothioate oligonucleotides may be synthesized by the method of Stein et al., 1988, Nucl. Acids Res. 16:3209. Methylphosphonate oligonucleotides can be prepared by use of controlled pore glass polymer supports (Sarin et al. , 1988, Proc. Natl. Acad. Sci. U.S.A. 85:7448-7451). While antisense nucleotides complementary to the p 101 or p!20 coding region sequence could be used, those complementary to the transcribed untranslated region are most preferred.

The antisense molecules should be delivered to cells which express the plOl regulatory subunit in vivo, e.g.. cells of hempatopoetic origin such as platelet, and neutrophils and other leukocytes. A number of methods have been developed for delivering antisense DNA or RNA to cells; e.g.. antisense molecules can be injected directly into the tissue or cell derivation site, or modified antisense molecules, designed to target the desired cells (e.g.. antisense linked to peptides or antibodies that specifically bind receptors or antigens expressed on the target cell surface) can be administered systemically.

However, it is often difficult to achieve intracellular concentrations of the antisense sufficient to suppress translation of endogenous mRNAs. Therefore a preferred approach utilizes a recombinant DNA construct in which the antisense

oligonucleotide is placed under the control of a strong pol III or pol II promoter. The use of such a construct to transfect target cells in the patient will result in the transcription of sufficient amounts of single stranded RNAs that will form complementary base pairs with the endogenous plOl or p!20 transcripts and thereby prevent translation of the plOl or p!20 mRNA. For example, a vector can be introduced in vivo such that it is taken up by a cell and directs the transcription of an antisense RNA. Such a vector can remain episomal or become chromosomally integrated, as long as it can be transcribed to produce the desired antisense RNA. Such vectors can be constructed by recombinant DNA technology methods standard in the art. Vectors can be plasmid, viral, or others known in the art, used for replication and expression in mammalian cells. Expression of the sequence encoding the antisense RNA can be by any promoter known in the art to act in mammalian, preferably human cells. Such promoters can be inducible or constitutive. Such promoters include but are not limited to: the SV40 early promoter region (Beraoist and Chambon, 1981, Nature 290:304-310), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto et al.,

1980, Cell 22:787-797), the heφes thymidine kinase promoter (Wagner et al. ,

1981, Proc. Natl. Acad. Sci. U.S.A. 78: 1441-1445), the regulatory sequences of the metallothionein gene (Brinster et al. , 1982, Nature 296:39-42), etc. Any type of plasmid, cosmid, YAC or viral vector can be used to prepare the recombinant DNA construct which can be introduced directly into the tissue or cell derivation site; e.g., the bone marrow. Alternatively, viral vectors can be used which selectively infect the desired tissue or cell type; (e.g. , viruses which infect cells of hematopoietic lineage), in which case administration may be accomplished by another route (e.g.. systemically).

Ribozyme molecules designed to catalytically cleave plOl or p!20 mRNA transcripts can also be used to prevent translation of plOl or p!20 mRNA and expression of plOl regulatory subunit. (See, e.g.. PCT International Publication WO90/11364, published October 4, 1990; Sarver et al. , 1990, Science 247: 1222- 1225). While ribozymes that cleave mRNA at site specific recognition sequences can be used to destroy plOl or p!20 mRNAs, the use of hammerhead ribozymes is preferred. Hammerhead ribozymes cleave mRNAs at locations dictated by flanking regions that form complementary base pairs with the target mRNA. The

sole requirement is that the target mRNA have the following sequence of two bases: 5'-UG-3'. The construction and production of hammerhead ribozymes is well known in the art and is described more fully in Haseloff and Gerlach, 1988, Nature, 334:585-591. There are hundreds of potential hammerhead ribozyme cleavage sites within the nucleotide sequence of human plOl or p!20 cDNA (FIG. 3). Preferably the ribozyme is engineered so that the cleavage recognition site is located near the 5' end of the plOl or p!20 mRNA; Le ^ , to increase efficiency and minimize the intracellular accumulation of non-functional mRNA transcripts.

The ribozymes of the present invention also include RNA endoribonucleases (hereinafter "Cech-type ribozymes") such as the one which occurs naturally in Tetrahymena Thermophila (known as the IVS, or L-19 IVS RNA) and which has been extensively described by Thomas Cech and collaborators (Zaug et al. , 1984, Science, 224:574-578; Zaug and Cech, 1986, Science, 231:470-475; Zaug et al., 1986, Nature, 324:429-433; published International Patent Application No. WO 88/04300 by University Patents Inc.; Been and Cech, 1986, Cell, 47:207-216). The Cech-type ribozymes have an eight base pair active site which hybridizes to a target RNA sequence whereafter cleavage of the target RNA takes place. The invention encompasses those Cech-type ribozymes which target eight base-pair active site sequences that are present in plOl or p!20. As in the antisense approach, the ribozymes can be composed of modified oligonucleotides (e.g. for improved stability, targeting, etc.) and should be delivered to cells which express the plOl regulatory subunit in vivo, e.g.. neutrophils. A preferred method of delivery involves using a DNA construct "encoding" the ribozyme under the control of a strong constitutive pol III or pol II promoter, so that transfected cells will produce sufficient quantities of the ribozyme to destroy endogenous plOl or p!20 messages and inhibit translation. Because ribozymes unlike antisense molecules, are catalytic, a lower intracellular concentration is required for efficiency.

Endogenous plOl or p!20 gene expression can also be reduced by inactivating or "knocking out" the plOl or p!20 gene or its promoter using targeted homologous recombination. (E.g.. see Smithies et al. , 1985, Nature 317:230-234; Thomas & Capecchi, 1987, Cell 51 :503-512; Thompson et al. , 1989 Cell 5:313-321; each of which is incoφorated by reference herein in its entirety).

For example, a mutant, non-functional plOl regulatory subunit (or a completely unrelated DNA sequence) flanked by DNA homologous to the endogenous plOl or p!20 gene (either the coding regions or regulatory regions of the plOl or p!20 gene) can be used, with or without a selectable marker and/or a negative selectable marker, to transfect cells that express plOl regulatory subunit in vivo. Insertion of the DNA construct, via targeted homologous recombination, results in inactivation of the plOl or p!20 gene. Such approaches are particularly suited in the agricultural field where modifications to ES (embryonic stem) cells can be used to generate animal offspring with an inactive plOl regulatory subunit (e.g.. see Thomas & Capecchi 1987 and Thompson 1989, supra). However this approach can be adapted for use in humans provided the recombinant DNA constructs are directly administered or targeted to the required site in vivo using appropriate viral vectors.

Alternatively, endogenous plOl or p!20 gene expression can be reduced by targeting deoxyribonucleotide sequences complementary to the regulatory region of the plOl or p!20 gene (i.e.. the plOl or p!20 promoter and/or enhancers) to form triple helical structures that prevent transcription of the plOl or p!20 gene in target cells in the body. (See generally, Helene, C. 1991, Anticancer Drug Des. , 6(6):569-84; Helene, C. et al., 1992, Ann, N.Y. Acad. Sci. , 660:27-36; and Maher, L.J., 1992, Bioassays 14(12):807-15).

In yet another embodiment of the invention, the activity of plOl regulatory subunit can be reduced using a "dominant negative" approach to interfere with trimeric G protein activation of PI3K. To this end, constructs which encode defective plOl regulatory subunits can be used in gene therapy approaches to diminish the activity of the plOl regulatory subunit in appropriate target cells. For example, nucleotide sequences that direct host cell expression of plOl regulatory subunits in which the G/3γ interacting domain is deleted or mutated can be introduced into hematopoietic cells (either by in vivo or ex vivo gene therapy methods described above). Alternatively, nucleotide sequences which encode only a functional domain of plOl could be used as an inhibitor of native pl01/pl20 interactions. Alternatively, targeted homologous recombination can be utilized to introduce such deletions or mutations into the subject's endogenous plOl or p!20 gene in the bone marrow. The engineered cells will express non-functional

receptors ( J , a regulatory subunit that is capable of binding the catalytic subunit, but incapable of stimulating the catalytic activity in response to G protein activation). Such engineered cells, i.e. neutrophils or other leukocyte lineages, should demonstrate a diminished response to activation of G protein linked receptors to extracellular chemokines, resulting in reduction of the inflammatory phenotype.

5.6.2 RESTORATION OR INCREASE IN plOl REGULATORY SUBUNIT EXPRESSION OR ACTIVITY TO PROMOTE IMMUNE SYSTEM ACTIVATION

With respect to an increase in the level of normal plOl or p!20 gene expression and/or plOl regulatory subunit gene product activity, plOl or p!20 nucleic acid sequences can be utilized for the treatment of hematopoietic lineage cell activation disorders, including reduced immune system responses to chemokines. Where the cause of the immune system disfunction is a defective plOl regulatory subunit, treatment can be administered, for example, in the form of gene replacement therapy. Specifically, one or more copies of a normal plOl gene or a portion of the plOl gene that directs the production of a plOl gene product exhibiting normal function, may be inserted into the appropriate cells within a patient or animal subject, using vectors which include, but are not limited to adenovirus, adeno-associated virus, retrovirus and heφes virus vectors, in addition to other particles that introduce DNA into cells, such as liposomes.

Because the plOl or p!20 gene is expressed in the hematopoietic lineage cells, including the neutrophils and other leukocytes, such gene replacement therapy techniques should be capable of delivering plOl or p!20 gene sequences to these cell types within patients. Alternatively, targeted homologous recombination can be utilized to correct the defective endogenous plOl or p!20 gene in the appropriate cell type; e.g., bone marrow cells or neutrophils and/or other leukocytes. In animals, targeted homologous recombination can be used to correct the defect in ES cells in order to generate offspring with a corrected trait.

Finally, compounds identified in the assays described above that stimulate, enhance, or modify the signal transduced by activated plOl regulatory subumt, e.g.. by activating downstream signalling proteins in the plOl regulatory subunit

cascade and thereby by-passing the defective plOl regulatory subunit, can be used to achieve immune system stimulation. The formulation and mode of administration will depend upon the physico-chemical properties of the compound.

5.7 PHARMACEUTICAL PREPARATIONS AND METHODS OF ADMINISTRATION

The compounds that are determined to affect plOl or p!20 gene expression or plOl regulatory subunit activity, or the interaction of plOl with any of its binding partners including but not limited to the catalytic subunit, can be administered to a patient at therapeutically effective doses to treat or ameliorate hematopoietic cell activation disorders, including inflammatory response disorders such as arthritis, including rheumatoid arthritis, septic shock, adult respiratory distress syndrome (ARDS), pneumonia, asthma and other lung conditions, allergies, reperfusion injury, atherosclerosis and other cardiovascular diseases, Alzheimer's disease, and cancer. A therapeutically effective dose refers to that amount of the compound sufficient to result in amelioration of symptoms of hematopoietic lineage cell activation disorders.

5.7.1 EFFECTIVE DOSE

Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g.. for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 50 /ED . Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.

The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity. The dosage may vary within this range

depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (i.e.. the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.

5.7.2 FORMULATIONS AND USE

Pharmaceutical compositions for use in accordance with the present invention may be formulated in conventional manner using one or more physiologically acceptable carriers or excipients. Thus, the compounds and their physiologically acceptable salts and solvates may be formulated for administration by inhalation or insufflation (either through the mouth or the nose) or oral, buccal, parenteral or rectal administration.

For oral administration, the pharmaceutical compositions may take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g.. pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g.. lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g.. magnesium stearate, talc or silica); disintegrants (e.g.. potato starch or sodium starch glycolate); or wetting agents (e.g.. sodium lauryl sulphate). The tablets may be coated by methods well known in the art. Liquid preparations for oral administration may take the form of, for example, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g.. sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g.. lecithin or acacia); non-aqueous vehicles (e.g.. almond oil, oily esters, ethyl alcohol or fractionated vegetable oils); and preservatives

(e.g.. methyl or propyl-p-hydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, flavoring, coloring and sweetening agents as appropriate.

Preparations for oral administration may be suitably formulated to give controlled release of the active compound. For buccal administration the compositions may take the form of tablets or lozenges formulated in conventional manner.

For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g.. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e.g. gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.

The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g.. in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.

The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g.. containing conventional suppository bases such as cocoa butter or other glycerides.

In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.

The compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration.

The following examples are presented to illustrate the present invention and to assist one of ordinary skill in making and using the same. The examples are not intended in any way to otherwise limit the scope of the disclosure or the protection granted by Letters patent hereon.

EXAMPLE: Purification and characterization of porcine GøΥ-activated PI3K activities

6.1 MATERIALS & METHODS 6.1.1 PI3K ASSAYS Purified aliquots of Sf9-derived or pig neutrophil cytosol-derived PI3K were diluted in ice-cold 0.12M NaCl, 25 mM HEPES, 1 mM EGTA, 1 mM DTT, 1 mg-ml '1 BSA, 1 % betaine, 0.02% , w/v, Tween-20, pH 7.4, 0°C to an appropriate extent, then 5 μl aliquots were stored on ice until assayed. If the PI3K assays were performed on immunoprecipitates from U937 cells (see following examples) then the PI3K was immobilized on 10 μl of packed protein G Sepharose beads in an ice-cold buffer defined above. 30 μl of a mixture of phospholipids with or without G / 3γs and/or Gas (either GDP-bound or activated) was added to the 5 μl fractions, or 10 μl of beads, and mixed. After 10 minutes on ice, 5-10 μl of last wash buffer, supplemented with MgCl 2 to give a final concentration in the extant assay volume of 3.5 mM, was added and mixed. Six minutes later, 5 μl of last wash buffer was added (to give a final assay volume of 50 μl) containing [-y^P]- ATP (typically 10 μCi assay '1 , Amersham, PB10168) and 3.5 mM MgCl 2 ) tubes were mixed and transferred to a 30°C water bath. Assays were quenched after 15 minute with 500 μl of chloroform: methanol :H 2 0 (29:54: 13. 1 , v/v/v). One μl of 100 mM ATP, 103 μl of 2.4 HCI and 434 μl of chloroform were subsequently added to generate a two phase 'Folch' solvent distribution. After mixing and centrifugation, the lower phases were removed and transferred to clean tubes containing 424 μl of fresh 'upper phase' (methanol: 1M HCI: chloroform; 48:47:3,

v/v/v). After mixing and centrifugation the lower phase was removed to a fresh tube (during the purification of porcine neutrophil PI3Ks, [ 32 P]-lipid product was quantitated at this point with a geiger counter), dried under vacuum, deacylated and resolved by TLC on PEI plates (with 0.6 M HCL; PtdIns(3,4,5)P 3 has an Rf of 0.47) (Stephens et al., 1994, Cell 77:83-93). In some experiments the dried lipid was redissolved in chloroform: methanol/2: l , v/v), applied to a potassium oxalate-impregnated TLC plated and resolved in a mixture of chloroform: acetone: methanol: acetic acid: H 2 O (40: 15: 13: 12: 8, v/v/v/v/v; Traynor-Kaplan et al. , 1988). The lipid/G protein subunit mixtures were prepared as follows:

PtdIns(4,5)P (which was prepared from Folch phosphoinositide fractions by 2 cycles of chromatography on immobilized neomycin) and PtdEtn (Sigma) were dried under vacuum (sufficient to give final concentrations in the assay of 50 μM and 0.5 mM, respectively). In some experiments PtdIns4P (prepared similarly to the PtdIns(4,5)P 2 ) and/or Ptdlns (Sigma) were included. The dried lipid was bath- sonicated (at room temperature) into final wash buffer (above) supplemented with 0.1 % , sodium cholate. After cooling on ice, portions of this dispersed lipid stock were mixed with a mixture, totaling 1 μl assay "1 , of G γ storage buffer (1 % cholate; 50 mM HEPES, pH 7.5, 4°C, 0.1 M NaCl; 1 mM DTT; 0.5 mM EDTA), active G / 3γ, or an equivalent volume of boiled G/3γ from 3-7 mg-ml "1 stocks in storage buffer. In some experiments the 1 μl would include Gα subunits or their storage buffer in which case the Gβγs were premixed with the Gα subunits (either GDP bound or activated; see below) for 10 minutes on ice.

Gα subunits (an equimolar mixture of Gα:o, i, i 2 and i 3 prepared as described in, and stored in the same buffer as the G / 3γ subunits except supplemented with 10 μM GDP) were activated by incubation on ice with 10 mM NaF, 30 μM A1C1 3 (A/F) for 10 mins; assays into which these subunits were diluted also contained A/F. a) Protein Purification Pigs blood (42 batches) was collected directly into anti-coagulant in 21 containers. Within 40 minute of collection the blood/anti-coagulant was mixed with 3% (w/v) polyvinylpyrrolidone (PVP, 360 kD) in isotonic saline (4.2 of blood mixture: 0.8 of PVP). After 35 minutes standing (in 25 containers) the

erythrocytes had settled adequately to allow the supernatant to be siphoned off and centrifuged (8 minutes, 1000 x g) in 1 liter containers. Approximately 28 liters of this primary supernatant was recovered. The sedimented cells were resuspended in Hank's saline such that the final accumulated volume was contained in two 1 liter centrifuge bottles. The cells were sedimented by centrifugation (8 minute 1000 x g). Supernatants were aspirated and the cell pellet was hypotonically shocked (to lyse any residual, contaminating erythrocytes) by the addition of 70 mis of ice-cold H 2 O. After 25-30 seconds of mixing, 77 mis of 10 x Hank's saline (without calcium) was added. After one wash with Hank's saline, the cells were combined into one centrifuge bottle, pelleted and resuspended in 500 mis of ice-cold; 40 mM TRIS, pH 7.5, 4°C; 0.12 M NaCl; 2.5 mM MgCl 2 . Di-isopropylfluorophosphate was added (final concentration 0.5 mM), after 5 minute on ice the cells were pelleted (approximately 80-90 mis packed volume) and resuspended in 300 mis of ice-cold 40 mM TRIS, pH 7.5; 0.1 M NaCl; 2.5 mM MgCl 2 ; 1 mM EGTA; 0.2 mM EDTA and antiproteases I. The cell suspension was sonicated (Heat Systems Probe sonicator, setting 9.25, 4 x 15 seconds with 1 minute mixing, on ice, between each burst) then centrifuged (2000 x g 10 min) to sediment unbroken cells and nuclei (less than 5% of cells remained intact). The supernatant was centrifuged (at 100 000 x g 60 min, 4°C) and the supernatants were decanted, pooled, mixed with EDTA, betaine and DTT (final concentrations of 5 mM, 1 % and 1 mM; respectively) and finally frozen in liquid nitrogen and stored at -80 C C. Cytosolic fractions prepared in this manner typically had a protein concentration of 8 to 9 mg-ml "1 (about 2.5 g protein per preparation). Once the cytosol from the equivalent of 750 liters of blood (18 to 42 preparations, 9 x 10 12 cells, 40 g protein) had been collected and stored at -80°C, they were thawed in three batches separated by 5 hr. From this point onwards all procedures were carried out at 2- 4°C.

The freshly thawed cytosol was supplemented with Tween-20 (0.05% , w/v, final concentration), centrifuged (20 000 x g for 30 min, 2°C) filtered (5 μm cellulose nitrate, 4.5 cms diameter; Whatman) diluted approximately 2.5 x with buffer K (see below) to a final conductivity of 200 μS (at 4°C), then loaded (12.5 ml-min "1 with a peristaltic pump) onto a 800 ml (5 cms diameter) column of fast flow Q Sepharose equilibrated with buffer K. The total volume of diluted cytosol

was approximately 15.5 liters. Once loaded, the column was washed with 1 liter of buffer K then eluted with a 4.5 liter, linear gradient, of 0.1 to 0.6M NaCl in buffer K at 8 ml-min '1 . Fifty 1 ml fractions were collected. The conductivity and Absorbance (at 280 nm) of the column eluate were monitored continuously (and in all subsequent steps). Buffer K had the following composition: 40 mM TRIS/Hl, 1 mM EGTA, 0.2 mM EDTA, 1 % betaine; 0.05% w/v Tween 20, 5 mM β- glycerophosphate pH 7.5 at 4°C, 15 mM /3-mercaptoethanol with 4μgml "1 each of antipain, leupeptin, bestatin, pepstatin A and aprotinin and 0. 1 mM PMSF ('antiproteases IF). This solution, as well as those that follow, was 0.2 μm filtered.

Once the relevant fractions had been identified by PI3K assays, they were pooled and immediately loaded (10 ml-min "1 ) onto a 1.8 1 column (5 cms diameter) of Sephadex-G25-fine, which had been pre-equilibrated with 18 1 buffer L (only last 2 liters with antiproteases II), (buffer L contained: 5 mM β-glycerophosphate, 20 mM KC1, 0.05% w/v Tween 20, 1 % betaine, 0. 1 mM EDTA, 10 mM potassium phosphate pH 7.0 at 4°C, 15 mM / 3-mercaptoethanol plus antiproteases II). The desalted pool from Q sepharose was immediately loaded (5 ml-min 1 ) onto 80 ml of hydroxylapatite (2.6 cms diameter; Macroprep-ceramic, BioRad) equilibrated with 1 liter of buffer M (5 mM / 3-glycerophosphate; 10 mM potassium phosphate, pH 7.0, 4°C, 0.05% w/v Tween 20, 1 % Betaine, 15 mM β- mercaptoethanol) at a flow rate of 10 ml-min "1 , and then with 100 mis of buffer N, (comprised of buffer M supplemented with 0. 1 mM EDTA and antiproteases I) immediately prior to loading the sample. After loading, the column was washed with 100 mis of buffer N and eluted with an 100 ml linear gradient of 0.05 to 0.35 M potassium phosphate in buffer N (4 ml-min 1 ). 25 ml fractions were collected and assayed for Gβγ-stimulated PI3K activity.

Relevant fractions were pooled (typically a total of 100 mis), diluted 3 x with buffer O (to a conductivity of 250 μS, 4°C) and loaded (1.1 ml-min 1 ) onto Heparin Sepharose HR (1.6 cms diameter column that had been pre-equilibrated with 150 mis of buffer O (see below, at 2 ml-min 1 ). After loading, the column was washed with buffer O (30 mis) and eluted with a 140 ml linear gradient of 0.1-0.7 M KC1 in buffer O (flow rate 1 ml-min 1 ), the elute was collected in 5 ml fractions. Buffer O was: 20 mM HEPES, 1 mM EGTA, 0.2 EDTA, 0.05% w/v/

Tween 20, 1.0% butane, 1 mM 0-glycerophosphate pH 7.2, 4°C, 15 MM β- mercaptoethanol, plus antiproteases II.

G / 3γ-stimulated PI3K activity eluted from Heparin sepharose HR in two peaks, designated peaks A and B. Both A and B were further purified by sequential use of the same combination of columns. Peak A was in 15 mis (0.4 M KC1) and was diluted 8 fold into buffer P (see below) to a final conductivity of 200 μS, 4°C), peak B was in 15 mis (0.6 M KC1) and was diluted 10 x into buffer P (to a final conductivity of 200 μS, 4°C). Dilution was immediately prior to loading at 1 ml-min "1 onto a Mono Q 5/5 HR column pre-equilibrated with 20 mis of buffer P. After loading, the column was washed with 5 mis of buffer P. Eluate was collected in 0.5 ml fractions. Buffer P contained: 10 mM Tris, 1 mM EGTA, 0.2 EDTA, 0.05 % w/v Tween 20, 1 % betaine, 1 mM β-glycerophosphate, pH 7.1 , 4°C, 15 mM / 3-mercaptoethanol plus antiproteases II.

The relevant fractions from Mono Q (A) and (B) were pooled independently (both had a total volume of 3 mis) concentrated with an ultrafiltration unit (50 kD cut-off pre-washed with buffer P) to 0.8 mis, centrifuged (10,000 x g for 10 minutes, 0°C) and loaded (0.25 ml-min "1 ) directly onto a high performance size exclusion chromatography column (V. 72 mis, V, 172 mis) pre-equilibrated with buffer Q (see below; 2 liters without antiproteases II, then 150 mis with antiproteases II) 1.5 ml fractions were collected just prior to the V-. Buffer Q contained: 0.17 M KC1, 1 % betaine, 0.05% w/v Tween 20, 1 mM β- glycerophosphate, 1 mM EGTA, 0.2 mM EDTA, 1.5 mM potassium phosphate, 40 mM HEPES, pH 6.9 at 4°C, 15 mM β-mercaptoethanol.

Relevant fractions from A and B were pooled independently (both had a total volume of 6 mis) diluted with buffer R to 24 mis (final conductivity 250 μS, 4°C) and loaded (0.8 ml-min "1 ) onto an acrylic-based cation-exchange HPLC column (2.5 mis volume, BioRad) and eluted with a 25 ml linear gradient of KC1 (0.1 to 0.6 M) in buffer R. The eluate was collected in 1 ml fractions. Buffer R contained: 1 % betaine, 0.05% w/v Tween 20, 1 mM EGTA, 0.2 mM EDTA, 20 mM HEPES, pH 6.8 at 4°C, 15 mM 5-mercaptoethanol plus antiproteases II.

Relevant fractions were pooled (3 mis for (A), 2 mis for (B)), diluted 7 x with buffer S (final conductivity of 180 μS, 4°C) and loaded (0.15 ml-min "1 ); onto a Mini Q column (0.24 mis, operated on a Pharmacia Smart™ system). The

column was washed with 1 ml of buffer S and eluted with a linear gradient of NaCl (0.1 to 0.5 M NaCl) in buffer S at 0.1 ml-min 1 . The elute was collected 75 μl fractions. Buffer S contained: 1 % betaine, 0.05 % w/v Tween 20, 1 mM EGTA, 0.2 mM EDTA, 2 M /3-glycerophosphate, 10 mM TRIS, pH 7.7, 4°C, 1 mM DTT (without antiproteases).

Protein concentrations throughout the purification were was estimated in four ways: (a) with a protein binding dye (BioRad; this was only used on lysates, cytosolic and Q sepharose fractions); (b) by integration of Abs 280 nm peak areas (this was calibrated by using the dye binding assay); (c) proteins on filters were stained with Ponceau S and compared with the staining intensity of defined aliquots of a similarly immobilized standard; and (d) proteins resolved by SDS-PAGE and stained with Coomassie R250 were compared with aliquots of proteins of known concentration run on the same gel.

Final preparation of PI3K (or first stage purified material) were incubated with 100 nM [ 3 H]-17-hydroxy-wortmannin (17.7 Ci mmol "1 , Amersham, custom made), resolved by SDS-PAGE, stained with Coomassie Blue, and photographed. [ 3 H] was then detected fluorographically. 6.2 RESULTS Analysis of porcine neutrophil cytosol by an ion-exchange chromatography showed it contained a G/3γ-activated PI3K activity of similar properties to ones already described in U937 and osteosarcoma cells. Use of [ 3 H]-17-hydroxy- wortmannin as a probe identified a doublet of proteins of apparent size 117 kD and 120 kD which eluted in the fractions containing G/37-activated PI3K activity, and further, that they were at 2-4% of the levels of [ 3 H]-17-hydroxy-wortmannin bound by pl lOα and/or pi 10/3 (see FIGs. 5). This peak of G / 3γ activated PI3K activity was purified further (all figures and tables detail the purification of the preparations of PI3Ks that were ultimately sequenced). During this procedure, it split into two peaks (A and B) both which displayed apparent, native, relative, molecular masses of 220 kD. Once essentially pure, as assessed by Coomassie-stained SDS-PAGE gels, it was clear that both activities co-migrated with two proteins: (A) with proteins of 117 kD (which specifically bound [ 3 H]-17-hydroxy-wortmannin and was assumed therefore to be the catalytic subunit) and 101 kD; and (B) with proteins of 120 kD (which also bound [ H]-17-hydroxy-wortmannin) and 101 kD. This result

indicated that the PI3K activities were pi 17/plOl and pl20/pl01 heterodimers in their native state. In their final forms PI3Ks A and B had been purified approximately 180,000 X and 380,000 X from neutrophil lysates (1 ,000,000,000 X from blood) with 5.5% overall recovery of activity. Table 1 defines the recoveries of protein and PI3K activity through each step).

TABLE 1

Purification of pig leukocyte G-protein βy subunit activated PDK's

Pool of activity Total protein Pool Total Activity Fold in pools Volume in pools Purification

Cytosol 40 g 151 100% 1

Q-Sepharose 1.5 g 40 mis 90% 24 (desalted) (1 -5 g) (450 mis) (124%)

Hydroxylapatite 162 mg 100 mis 125% 309

Heparin 19 mg 15 mis 46% 970 Sepharose Peak A

Peak B 12 mg 15 mis 53% 1769

Mono Q pool A 5.4 mg 3 mis 16% 1187

B 1.4 mg 3 mis 15% 4291

Size exclusion A 0.722 mg 6 mis 16% 8902 (850 μl applied)

B 0.2 mg 6 mis 15.5 % 51038

Cation Exchange 0.13 mg 3 mis 6% 18489

A

B 0.014 mg 2 mis 9.5% 271761

Mini Q pool from A 15 μg 0.225 mis 2.2% 58754 from B 10 μg 0.225 mis 3.1% 174151

These extents of enrichment are consistent with the quantities of [ 3 H]-17- hydroxy-wortmannin bound by these proteins compared to p85/pl 10 family members. All of these proteins are thus considered to be of low abundance.

Purified preparations of PI3Ks A and B were indistinguishable on the basis of their lipid kinase activities. Both preparations were (a) activated over 100 X by

G 3γ subunits, in a Gα-GDP-sensitive fashion, (b) completely inhibited by 100 nM wortmannin (with 5μM ATP in the assays), (c) insensitive, either in the presence or absence of Gβγs, to tyrosine phosphorylated peptides which activate p85/pl l0 family PI3Ks five fold (see FIG. 6), and (d) able to 3-phosphorylate Ptdlns, PtdIns4P and PtdIns(4,5)P 2 (the identity of the products was established by sequential deacylation and deglyceration followed by anion-exchange HPLC analysis of the [ 32 P] -labelled water-soluble products with internal [ 3 H]-labeled standards). Further, the purified preparations of PI3K A and B displayed the lowest apparent Km for the latter substrate (8 and 10 μM for A and B, respectively) utilizing the γ-phosphate of ATP as the phosphate donor (results not shown).

7. EXAMPLE: PEPTIDE SEQUENCING OF PORCINE GB-v-ACTIVATHD

PI3K A AND B

In this example, both porcine PI3K proteins were analyzed by amino acid sequencing. PI3Ks A and B, purified from the equivalent of 40g of cytosolic protein, were Western blotted onto nitrocellulose, stained with Ponceau S, the bands corresponding to all four subunits were excised, treated with trypsin and processed for internal amino acid sequence analysis. 7.1 MATERIALS AND METHODS

Generation of peptides and peptide sequencing. Aliquots of protein for sequencing were Western blotted (in a wet blotter) onto nitrocellulose (0.45 μm pore size BA85; Schleicher and Schuell). The transfer buffer contained 192 mM glycine, 25 mM TRIS and 10% v/v methanol. Prior to assembling the final transfer unit the Whatman No. 1 filter paper supports on the (-) side of gel/filter sandwich, and the gel (1 mm thick), were soaked (2-3 mins) in transfer buffer containing 0.0005% (w/v) SDS. The transfer was for 16 h at a fixed 35 V (0.25 to 0.35 Amps, at 5°C). The filters were stained with 0.1 % Ponceau S in 1 % acetic acid for 1 min, then destained for 1 minute in 1 % acetic acid.

Approximately 85-90% of the protein loaded on the gel was recovered the filter. The bands of interest were excised from the nitrocellulose and processed for internal amino acid sequence analysis as described (Tempst et al. , 1990, Electrophoresis 11:537-552), with modifications (Lui et al. , 1996). Briefly, in situ

proteolytic cleavage was done using 0.5 μg trypsin (Promega, Madison, WI) in 25 μl 100 mM NH 4 HCO 3 (supplemented with 1 % Zwittergent 3-16) at 37°C for 2 hours. The resulting peptide mixture was reduced and S-alkylated with, respectively 0.1 % /3-mercaptoethanol (BioRad, Richmond, CA) and 0.3 % 4-vinyl pyridine (Aldrich, Milwaukee, WI), and fractionated by reversed phase HPLC. An enzyme blank was done on an equally sized strip of nitrocellulose.

HPLC solvents and system configuration were as described (Elicone et al. , 1994), except that an 2.1 mm 214 TP54 Vydac C4 (Separations Group, Hesperia, CA) column was used with gradient elution at a flow rate of 100 μl/min. Identification of Tφ-containing peptides was done by manual ratio analysis of absorbances at 297 and 277 nm, monitored in real time using an Applied Biosystems (Foster City, CA) model 1000S diodarray detector (Erdjument- Bromage et al. , 1994). Fractions were collected by hand, kept on ice for the duration of the run and then stored at -70°C before analysis. Purified peptides were analyzed by combination of automated Edman degradation and matric-assisted laser-desoφtion ionization time-of-flight (MALDI- TOF) mass spectrometry; details about this combined approach, including mass- aided post-chemical sequencing routines can be found elsewhere (Geromanos et al. , 1994, Techniques in Protein Chemistry V 143-150; Elicone et al. , 1994, J. Chromatogr. 676: 121-137; Erdjument-Bromage et al. , 1994, Protein Sci. 3:2435- 2446). After storage, column fractions were supplemented with neat TPA (to give a final concentration of 10%) before loading onto the sequencer disks and mass spectrometer target. Mass analysis (on 2% aliquots) was carried out using a model Voyager RP MALDI-TOF instrument (Vestec/PerSeptive, Framingham, MA) in the linear mode, with a 337 nm output nitrogen laser, an 1.3 m flight tube and α- cyano-4-hydroxy cinnamic acid (premade solution obtained from Linear Sci. , Reno, NV) as the matrix. A 30 kV ion acceleration voltage (grid voltage at 70% , guide wire voltage at 0.1 %) and -2.0 kV multiplier voltage were used. Laser fluence and number of acquisitions were adjusted as judged from optimal deflections of specific maxima, using a TDS 520 Tektronix (Beaverton, OR) digitizing oscilloscope. Mz (mass to charge) spectra were generated from the time-of-flight files using GRAMS (Galactic Ind. , Salem, NH) data analysis software. Every sample was analyzed twice, in the presence and absence of two calibrants (25

femtomoles each of APID and P8930), as described (Geromanos et al. , 1994). Chemical sequencing (on 95 % of the sample) was done using a model 477A instrument from Applied Biosystems (AB). Stepwise liberated PTH-amino acids were identified using an 'on-line' 120A HPLC system (AB) equipped with a PTH C18 (2/1 x 220 mm; 5 micron particle size) column (AB). Instruments and procedures were optimized for femtomole level phenyl thihydantoin amino acid analysis as described (Erdjument-Bromage et al., 1994, Protein Sci. 3:2435-2446; Tempst et al., 1994, Methods CompanionMeth. Enzymol. 6:284-261).

Peptide average isotopic masses were summed from the identified residues (including the presumed ones) using ProComp version 1.2 software (obtained from Dr. P.C. Andrews, University of Michigan, Ann Arbor, MI).

A doubly tyrosine phosphorylated peptide (18 residues) based on the sequence surrounding tyrosines 740 and 751 in the PDGF βR was prepared by the microchemical facility at the Babraham Institute. 7.2 RESULTS

Peptide sequence data immediately resolved several issues regarding the relationships between these proteins. The plOls derived from both PI3Ks A and B were identical and further a relatively common allelic-variant was identified at 483 in the ORF, (marked in fig. 4) such that a serine was replaced by a glycine. pi 17 and pl20 displayed virtually identical tryptic HPLC profiles and all apparently common peptides that were sequenced from both species were identical with the exception of a amino-terminal blocked peptide from pi 17 (see below). Peptide sequence information was then used to design probes for retrieving the nucleotide sequence encoding these proteins.

8. EXAMPLE: Cloning of the cDNAs encoding porcine p!20 and plOl

Degenerate oligonucleotide probes, based on the sequence of a peptide from porcine pl20 and a peptide from porcine plOl were used to screen an oligo-dT- primed, amplified, cDNA library (made from pig neutrophil poly A-selected RNA). Described blow is the cloning and characterization of cDNA's encoding both the plOl and pl20 proteins.

8.1 MATERIALS AND METHODS

We prepared 0.7 mg total RNA from 4.2 x 10 9 pig neutrophils (Chomczynski & Sacchi, 1987, Anal. Biochemistry 162: 156-159). This RNA was used by Stratagene (San Diego, CA) to produce PolyA-selected mRNA from which they prepared oligodT- and random-primed cDNA libraries in λZAPII (approximately 5.4 x 10 "6 and 3.2 x 10 6 primary p.f.u. respectively). Amplified libraries were constructed from approximately 2 x 10 6 original recombinants and these were used to screen for pl20 and plOl cDNAs by standard procedures.

2.5 x 10 6 plaques derived from the oligodT-primed library were screened using a [ 2 P]-labelled oligo based on peptide sequence from pi 20 [CA(T/C) GA(T/C) TT(T/C) ACI CA(A/G) CA(A/G) GTI CA(A/G) GTI AT(T/C/A)

GA(T/C) ATG] (SEQ ID NO:5). Twelve positive clones were identified isolated as Bluescript™ based plasmids and both DBA strands sequenced (using the ABI automatic sequencing facility at the Babraham Institute). The inserts of these plasmids represented a series of overlapping clones with two clones defining a full length ORF encoding all of the peptide sequence derived from pl l7/pl20 tryptic digests (FIG. 4).

0.9 x 10 6 plaques derived from the oligodT-primed library were screened using a [ 32 P] -labelled oligo based on peptide sequence from plOl [GCITA(T/C)ATGGA(A/G)GA(T/C)ATIGA(A/G)GA] (SEQ ID NO:6). 1 positive clone was identified, isolated and sequenced. The 5' end of this clone (DI 1) represented part of the sequence for one of the tryptic peptides. thus identifying it as a partial clone. A further 0.6 x 10 6 plaques from the oligoT-primed library were screened using a [ 32 P]-labelled CIa-1 restriction fragment derived from Dl l . Sixty-six positive clones were identified, 49 of which were isolated and some partially sequenced. These represented a series of overlapping clones all of which contained Dl l sequence but all of which were smaller than Dl l itself. 3.5 x 10 6 plaques from the random-primed library were then screened using a [ 32 P] -labelled Apa-1 restriction fragment derived from Dl l . Ninety-eight positive clones were identified. These clones were re-screened (at the stage of primary plaque isolates) by a PCR-based approach using primers designed against the Bluescript™ vector (either 'forward' and 'reverse' primers) and internal Dl l sequence. This enabled us to identify (independent of orientation) the longest potential N-terminal extensions encoding plOl sequence. The 3 clones giving the largest PCR-fragment

were isolated and sequenced. These represented overlapping clones which, together with Dl l, defined a full length ORF encoding all of the peptide sequence derived from the p 101 tryptic digest (FIG. 1 and FIG. 2) 8.2 RESULTS Two clones defined a full length ORF encoding all of the pi 17/pl20 tryptic peptides (see FIGs. 1-4). Of three potential start methionines, the central one was identified as active (in contrast to the assumption of Stoyanov et al. , 1995, Science 269:690-693) on the basis of the precise match between the measured mass of an amino-terminal blocked pl20-derived peptide and the theoretical masses of amino- terminal peptides that would be derived as a result of initiating translation at each of the three methionines. As such, pl20 has a theoretical size of 126 kD. Comparison of the mass of the amino-terminal blocked peptide produced from pi 17 with the relevant regions of the ainino-teπninal end of pi 20 indicates no precise matches (allowing for usual amino-terminal blocking). Hence pi 17 is unlikely to be a proteolytically or post-translationally modified form of pl20, nor is it likely to result from use of a second translation start point within the pl20 message. However, a cDNA with an ORF encoding pi 17 has not been isolated. The protein and DNA sequences defining pl20 were used to search data bases for similar structures. Similarities with all previously cloned PI3Ks were identified. However, the sequence was nearly identical, allowing for species differences, to pl lOγ (Stoyanov et al. , 1995). The only significant discrepancy between our sequence and that Stoyanov et al. , is found in the extreme COOH terminus. On the basis of primary structure only, the identification of a COOH terminal pleckstrin homology domain in pi 20 could not be confirmed. By utilizing several overlapping fragments, derived from both oligo-dT and random-primed, pig neutrophil-derived, cDNA libraries, a full length ORF encoding all of the peptide sequence derived from pi 01 has been defined. A plOl- derived, amino-terminal blocked peptide was identified; its mass was precisely equivalent to that predicted for an amino-terminal acetylated version of the first 12 residues defined by the predicted start in the ORF described. The predicted relative molecular mass of plOl is 97 kD. Although the protein and DNA sequence data bases were searched for similar structures or sub-structures, no significant matches were identified.

9 EXAMPLE: Cloning of the cDNAs encoding human p!20 and plOl

Radiolabeled PCR products derived from the porcine plOl and pi 20 cDNA sequences were successfully used to screen human cDNA libraries for their human homologs. The cloning and characterization of cDNA's encoding human plOl and pi 20 is described below. 9.1 Human plOl

A human monocyte cDNA library (Clontech, Palo Alto, CA; mRNA source: U937 cell line, PMA treated at 50 ng/ml for 3.5 days) was screened with a radiolabeled PCR product corresponding to bp 887-1287 of the porcine PlOl cDNA sequence, λgtl l phages containing the human cDNA library were plated, as described in the Clontech Lambda Library Protocol Handbook, and transferred to nylon filters. Filter-bound DNA was denatured by autoclaving the filters for one minute, after which the filters were prehybridized for 2 hours at 42 °C in 50% formaldehyde, 5x Denhardts, 5x SSC, 0.05 mg/ml salmon sperm DNA, 0.05M NaPO 4 at pH6.8, ImM sodium pyrophosphate and 1 % SDS. The radiolabeled PCR probe was denatured by boiling for 5 minutes, cooled on ice for 15 minutes, and then added to the filters in the hybridization buffer at a final concentration of 1 million cpm/ml. Hybridization proceeded overnight at 42°C with constant agitation. Filters were then washed once in 2x SSC, 1 % SDS at room temperature for 20 minutes, followed by a wash in 2x SSC, 1 % SDS at 42°C for twenty minutes, and finally a wash at 0.2x SSC, 1 % SDS at 42 °C for 20 minutes. After autoradiography, positive clones were isolated according to the Clontech Library Handbook by picking the area around and including the positive clone and eluting the phage into Phage Dilution Buffer. The phages were then replated at a density allowing for isolation of a single phage plaques, grown overnight, and transferred to nylon filters. The filters were hybridized with the same radioactive PCR probe as above, washed, and subjected to autoradiography. Single, pure, positive phage plaques were picked, the phage eluted into Phage Dilution Buffer and then replated to give a confluent lawn. The plaque purified phage lawn was eluted into Phage Buffer and lambda phage DNA isolated according to the protocol in the Clontech Library Protocol Handbook.

Purified lambda DNA was cut with the restriction enzyme EcoRI, separated on an agarose gel, and the EcoRI DNA insert isolated and recloned into EcoRI

linearized Bluescript KS + . Bluescript plasmids containing these DNA inserts were then sequenced. Five of the independently isolated cDNA clones contained sequences homologous to the porcine plOl cDNA sequence between nucleotide 1137 and the stop codon at nucleotide 3021. Relative to the porcine plOl sequence, the isolated human clones contained an additional 544 bp of 3' untranslated sequence.

Because the porcine sequence is very GC rich at the 5' end, the probability of finding a full length clone in an oligo-dT primed library is low. Therefore, we decided to use a library that was both oligo dT and random primed in order to increase the probability of finding a clone that contained the GC rich 5' end. Using the same radiolaeled PCR product of the porcine plOl sequence and the same methods described above, we screened a human leukocyte 5 '-Stretch Plus cDNA Library (Clontech; mRNA source: normal peripheral blood leukocytes) and isolated several clones, one of which contained the 5' end of the human plOl sequence. This clone included roughly 300 bp upstream of the coding sequence and the first 1612 bp of the coding sequence. A full length cDNA clone containing the entire coding region of the human plOl sequence was then constructed by fusing the 5' end of the leukocyte clone (from a Hindlll site 77 bp upstream of the start codon to a Sad site at position 1262) with the monocyte clone (from the Sad site to an Xbal site at position 3014, 305 bp downstream of the stop codon). The entire sequence of the full-length cDNA clone was determined, and the coded amino acid sequence deduced.

Since this full length clone was derived from two different clones from two different libraries, the existence of the 3' end of the plOl clone in the leukocyte library was verified. Using five different sets of PCR primers, we demonstrated that the 3' end of plOl, which was cloned from the monocyte library, was present in both the monocyte and the leukocyte libraries. 9.2 Human pi 20 The human homolog of porcine pi 20 was cloned by screening a leukocyte cDNA library (Clontech; mRNA source: normal peripheral blood leukocytes) with a radioactively labeled PCR fragment of the porcine sequence from bp 875 to 1315, according to the protocol described above for the cloning of human plOl. We independently isolated two clones containing the 5' end of human pl20,

corresponding to bp 6 to 2414 in the porcine sequence as reported by Stephens et al. The human sequence contains an EcoRI site at bp 2408, and since the cDNA library had been constructed with EcoRI linkers, the full length human sequence had to be contained in two separate clones, one containing the 5' EcoRI fragment, the other containing the 3' EcoRI fragment. The library was therefore rescreened under the same conditions as above with a radioactively labelled PCR fragment from the 3' end (bp 2801 to 3395) of the porcine pl20 sequence. We independently isolated two clones containing the 3' end of the sequence from position 2408 (human sequence) to the beginning of the poly A tail at position 5128. The DNA inserts of the two phages containing the 5' and the 3' EcoRI fragment were isolated and fused at the EcoRI site to give a full length cDNA clone (1-5128) containing the total coding region (84 to 3390) of the human pl20 homolog. The entire sequence of the full-length cDNA clone was determined, and the coded amino acid sequence deduced. 10. EXAMPLE: Expression of porcine p!20 and plOl in insect cells

Recombinant, clonal, baculo-viruses (rbv) harboring either a amino- terminal, 6 x HIS-tagged pl20 (pAcHLT pi 20) cDNA or a amino-terminal, (EE)- tagged plOl cDNA (pAcO-Gl) plOl) were prepared and used to drive production of the above proteins in insect (Sf9) cells. 10.1 MATERIALS AND METHODS

10.1.1 CONSTRUCTION OF EXPRESSION VECTORS

The use of N-terminal PCR and appropriate restriction sites allowed the porcine pl20 and plOl ORFs to be manipulated into a form where they could be inserted in frame into various expression vectors. In each case, the first amino acid encoded after the N-terminal tag was the start methionine. The 3'- untranslated region of the pl20 cDNA was used in full and that of the plOl cDNA truncated at a BamHI site (nucleotide 192, FIG. 1). The vectors used for baculovirus-driven expression in Sf9 cells were pAcHLT (which encodes an N- terminal '6 x His-tag' followed by a thrombin cleavage site, Pharminogen; the pl20 ORF was inserted into the XhoI-EcoRI sites) and pAcO-Gl (which encodes an N-terminal 'EE-tag', ONYX Pharmaceuticals; the plOl ORF was inserted into the EcoRI-NotI sites). All vectors were N-terminally sequenced before use.

10.1.2 Sf9 cell transfections and production of recombinant proteins

Sf9 cells were grown in TNM FH with 1 1 % HI-FBS in a spinner flask and were maintained at between 0.5 and 2 x 10 6 cells ml "1 . Sf9 cells were transfected with Insectin™ (Invitrogen) liposomes with linearized baculo-gold DNA (Pharminogen) and relevant baculo-virus transfer vectors as recommended (Invitrogen). The resulting recombinant baculo viruses were plaque-purified and amplified to yield high-titre viral stocks. The optimal (for production of protein) dilutions were determined for each high-titre stock. pAcO-Gl plOl virus were allowed to infect adherent Sf9 cells for 2.2 days at 27 °C; pAcHLT pi 20 virus were allowed to infect in a spinner culture (usually) for 1.9 days at 27°C. Cells were harvested into ice-cold 0.41 % KC1; 2.66% sucrose; 20 mM MgCl 2 ; 8 mM NaH 2 PO 4 , pH 6.2, 25°C; treated with 1 mM di-isopropylfluorophosphate (5 minutes on ice), and washed once in the harvesting solution. Centrifugally packed aliquots of cells were frozen in liquid N 2 and stored at -80°C.

10.1.3 Purification of Sf9-derived proteins

Porcine pl20 was purified using a metal-ion chelation column (Talon, Clontech). Cell pellets were thawed into 0. 10 M NaCl; 50 mM sodium phosphate, pH 8.0, 4°C; 10 mM Tris.HCl, pH 8.0, 4°C; 1 mM MgCl 2 and antiproteases I (see above) at a ratio of 1 liter of infected Sf9 cell culture into 50 mis of sonication buffer, probe-sonicated (4 x 15 second bursts on ice), and centrifuged (120,000 x g for 40 minutes, 4 C C). The supernatant was removed and pooled (cloudy supernatant at the top of the tube was removed separately, 0.45 μM filtered with a low-protein binding filter and then pooled with the remainder). The cytosolic fraction was supplemented with Tween-20 and betaine (0.05 % , w/v, and 1 %, respectively) then pumped onto a column of Talon resin equilibrated in equivalent buffer (1.2 mis of resin per liter of original infected Sf9 culture at a linear flow velocity of 20 cms hr "1 ). This, and all subsequent steps were carried out at 4°C. The column was sequentially washed (same flow) with 20 column volumes each of: Buffer A, 50 mM sodium phosphate, pH 8.0, 4°C; 10 mM Tris/HCl, pH 8.0, 4°C; 0.15 M NaCl; 1 % betaine; 0.05 % , w/v, Tween-20; buffer B, 1 %, w/v, Triton X-100; 0.15 M NaCl; 50 mM sodium phosphate, pH 8.0, 4°C; 10 mM Tris, pH 8.0, 4°C; 1 % betaine 0.05% , w/v, Tween-20; buffer C, 0.15 M NaCl;

50 mM sodium phosphate, pH 7. 1 , 4°C; 1 % betaine; 0.05 %, w/v, Tween-20; buffer D; 0.15 M NaCl; 30 mM Tris, pH 7.5, 4°C% , 1 % betaine, 0.02% , w/v, Tween-20; 10% , v/v, ethylene glycol; 1 mM MgCl 2 ; and then 8 column volumes buffer E, comprised of buffer D supplemented with 10 MM imidazole (pH 7.5). During the Buffer E wash, 2 ml fractions were collected. Finally, at half the flow used previously, the column was washed with buffer F, which was comprised of buffer D supplemented with 70 mM imidazole (pH 7.5; final concentration). Typically 1 ml fractions were collected. With experience fractions were pooled on the basis of the Abs 280 nm trace (continuously recorded) and supplemented with 1 mM DTT and 1 mM EGTA (final concentrations) immediately. Typically this process yielded 4 mg of pl20 per liter of Sf9 culture. The pl20 prepared in this manner was usually greater than 90% pure. The final pool of pl20 was desalted via a 15 ml column of G-25 superfine equilibrated in buffer G, which was comprised of buffer D supplemented with 1 mM DTT and 1 mM EGTA (final concentrations). 'pl20 blank' preparations used in some experiments were prepared in precise parallel to a normal pl20 preparations except the starting cells were either infected with wild type baculo-virus, or were uninfected. The final fractions derived from these 'blank' preparations were pooled on a 'parallel volume' basis because they contained virtually no protein. The pl20 6 x HIS tag contained a thrombin cleavage recognition motif.

Careful titration with thrombin and analysis with 6% polyacrylamide SDS-gels revealed two thrombin sites, with similar sensitivities to thrombin, both close to the amino-terminal (because an αCOOH-terminal antibody still immunoprecipitated the twice-cut pl20). One site was at the expected location for the site engineered into the tag; the other site was approximately 40-residues in from the amino-terminal (in a region with no favored thrombin recognition sequences). Under optimized conditions (2 U ml "1 , thrombin; 0.2 mg-ml '1 pl20; 4 hours, 4°C, with 1 mM EGTA and 1 mM MgCl 2 ) it was possible to generate preparations of thrombin cleaved pl20 which contained 15 % uncut pl20, 50% cut at the authentic amino- terminal thrombin site and 35 % with an additional approximately 40 residues cleaved. In these experiments thrombin action was terminated by the addition of 100 nM N-acetyl D-Phe Pro-2-amido-5 guanidino butane boronic acid; a potent thrombin inhibitor (Sigma). Throughout this work, preparations of partially

cleaved pl20 were used parallel with totally uncleaved pl20. It was clear that: (A) all three pl20's bound to plOl with very similar affinity, (B) plOl bound to the pl20 mixture was activated by G/3γs to a similar extent to plOl bound to uncleaved pl20 and, (C) both the uncleaved and partially cleaved pl20s were virtually insensitive to G/?γ subunits in the absence of plOl (although complete thrombin cleavage tended to reduce overall pl20 catalytic activity by 20-30% and increase the 1.7 fold apparent activation by G γ in the absence of plOl to about 2.5 fold).

Porcine (EE)-plOl was purified from frozen pellets of Sf9 cells as follows. Cells from 2 liters of infected Sf9 culture were sonicated into 50 mis of 0.12 M NaCl; 1 mM MgCl 2 ; 25 mM HEPES, pH 7.4, 4°C; 1 mM EGTA plus antiproteases I as described above. After centrifugation ( 120,000 Xg, 4°C, 40 minutes), the supernatant was removed (as described above), supplemented with 1 % w/v, Triton X100, 0.4% sodium cholate, 0.4 M NaCl (final concentrations), and mixed with 2 mis of packed protein G sepharose covalently cross-linked to an α-(myc) irrelevant, monoclonal antibody (washed in an equivalent solution). After 30 minutes mixing at 4°C, the beads were sedimented and the supernatant was removed and mixed with 1 ml of protein G Sepharose covalently cross-linked to an α-(EE) monoclonal antibody (equilibrated in an equivalent solution). After 2 hours mixing at 4°C the beads were washed as described below for U937 cell α-(EE) immunoprecipitations, except (a) the washes were in a 20 ml centrifuge tube, and (b) the beads were finally washed 3 x with buffer H, comprised of buffer G with 1 % , w/v, Triton X-100. 'plOl blank' (plOlC) preparations used in some experiments were prepared from either wild-type baculo-virus infected or uninfected cells exactly as described above. pl01/pl20 heterodimers formed in vivo by co-infection of Sf9 cells with both forms of recombinant baculo-virus were purified as described for (EE)-plOl except the immunoprecipitates were washed 4 x with buffer I, 1 % , w/v, Triton X- 100; 0.15M NaCl; 20 mM HEPES, pH 7.4, 4°C; 1 mM EGTA; 2 x with buffer J, comprised of buffer I supplemented with 0.4M NaCl (final concentration), then 3 x with buffer G before being eluted with 1 bed volume of 150 μg ml "1 (final concentration) of (EE)-peptide in buffer G. The (EE)-peptide, amino-terminal- acetylated EYMPTD, has a very high affinity for the α-(EE) monoclonal antibody.

After the beads were incubated with (EE)-peptide, on ice, for 40 minutes, the supernatant was removed. Aliquots of the eluted proteins were diluted and assayed for PI3K activity (see above) or mixed with SDS sample buffer directly.

In in vitro reconstitution experiments, pi 20 preparations in buffer G supplemented with 1 %, w/v, Triton X-100 (now equivalent to buffer H) were mixed with (EE)-plOl (10: 1 molar ratio of protein) still bound to the protein G matrix, mixed for 2 hours (end on end at 4°C), then washed and eluted with (EE)- peptide as described for the purification of pl01/pl20-PI3K reconstituted in vivo, in the Example below. 10.2 RESULTS

Single-step purifications utilizing the tags yielded purified protein preparations as assessed by Coomassie staining; their apparent sizes matched expectation. Further, both were correctly recognized in Western blots by specific, COOH Terminal directed and internal-sequence directed, antipeptide, rabbit polyclonal sera (not shown) indicating their authenticity. pl20 bound tightly, and in 1 : 1 molar stoichiometry, to plOl both (a) in vitro, when both proteins had been independently purified, mixed (with plOl still associated with the protein G matrix used to isolate it) then washed stringently before being eluted with (EE)-peptide or, (b) in vivo, when Sf9 cells were co- infected with both forms of rbv and proteins were purified via the plOl tag (data not shown).

Free, purified pl20 was a PtdIns(4,5)P 2 -selective, wortmannin-sensitive PI3K. G / 3γs had a small and bi-modal effect on free pl20 PI3K activity. An equimolar mixture (final total concentration of 2 μM) of Gα's o, i l 5 i 2 and i 3 bound to GDP and in the presence of aluminum fluoride had no significant effect on free pl20 PI3K activity (this was a preparation of Gas which, when added in a 1.5 fold molar excess, could completely inhibit the effects of G/3γs, on PI3K). Tyrosine phosphorylated peptides able to activate p85/pl l0-PI3K family members also had no detectable effect on pl20 PI3K activity. When bound to plOl (either in vivo or in vitro) pl20's, PI3K activity could be activated greater than 100 x by G/3γ subunits. Tyrosine phosphorylated peptides and Gα-GDP/aluminum fluoride had no effect on G 3γ activated, or basal, pl01/pl20 PI3K activity. In the absence of G 3γs the specific activity of p 120 in a p 101 /pi 20 complex is lower than the

specific activity of free pi 20 but is increased greater than 50x in their presence (see FIG. 7).

Comparison of the specific catalytic activities of pig neutrophil-derived PI3K (B) and the Sf9-derived pl01/pl20 heterodimers, under identical, Gβ - stimulated, assay conditions, showed the Sf9 derived material to have a 2 X higher activity per mg protein. This result is not inconsistent with the likely 'age at assay' of a PI3K preparation derived from circulating neutrophils via a 4 day purification protocol and indicates the bulk of the recombinant PI3K is correctly folded and that any critical post-translational modifications must be in place.

11. EXAMPLE: Expression of porcine plOl and p!20 in mammalian cells

A family of mammalian expression vectors were constructed that enabled transient expression of amino-terminal epitope-tagged forms (either (myc) or (EE)) of porcine plOl and pl20. This example describes the production of purified recombinant plOl and pl20 fusion proteins from mammalian cells, and subsequent analysis of their properties.

11.1 MATERIALS AND METHODS

11.1.1 CELL CULTURE

U937 cells were grown in RPMI 1640 with 10% , v/v, heat-inactivated (HI)- FBS and diluted 4 fold every 2 days. Cos-7 cells were grown in DMEM 10% HI- FBS.

11.1.2 CONSTRUCTION OF EXPRESSION VECTORS The use of N-terminal PCR and appropriate restriction sites allowed the porcine pl20 and plOl ORFs to be manipulated into a form where they could be inserted in frame into various expression vectors (in each case the first amino acid encoded after the N-terminal tag was the start methionine. The 3 '-untranslated resin of the pl20 cDNA was used in full and that of the plOl cDNA truncated at a BamHI site (nucleotide 192, FIG. 1). The vectors used for cytomegalovirus-driven (CMV) expression in mammalian cells were (A) pCMV(EE) (encoding an N- terminal MEEEEFMPMPMEF (SEQ ID NO:7) or MEEEEFMPMEFSS (SEQ ID NO:8) 'EE-tag' for plOl or pl20 expression, respectively) and (B) pCMV (myc) (encoding an N-terminal MEQKLISEEDLEF (SEQ ID NO:9)or

MEQKLISEEDLEFSS (SEQ ID NO: 10) 'myc-tag' for plOl or pl20 expression, respectively). All vectors were N-terminally sequenced before use.

11.1.3 U937 TRANSFECTION PROTOCOLS Exponentially growing U937 cells (diluted 12 hours previously) were washed 2 x with PBS and resuspended in sterile electroporation medium (EM) containing; 30 mM NaCl, 0.12 M KC1, 8.1 mM Na 2 HP0 4 , 1.46 mM KH 2 PO 4 and 5 mM MgCl 2 , at room temperature. Circular plasmid DNA was added in 1 x EM to the cells to produce a 0.5 ml final volume containing 1.4 x 10 7 cells and 40 μg total DNA (usually made up to 40 μg with an expression plasmid with the same promoter and expressing a similarly tagged but irrelevant protein) and were transferred to a cuvette (0.4 cms gap, BioRad). After 15 minutes standing at room temperature the cells were electroporated (1 pulse at 280V and 960 μF, with a BioRad Gene pulser; time constants were typically 18 msec) then placed on ice for a further 8 minutes before being diluted into 5 mis of RPMI, 10% HI-FBS. After standing for 5 minutes, to allow dead cells to clump together, the cells were diluted with 35 ml of RPMI, 10% HI-FBS, supplemented with penicillin and streptomycin, then TPA and ZnCl 2 were added (both of which substantially amplify expression from CMV promoters in U937 cells) to final concentrations of 5 x 10 "8 M and 200 μM, respectively. If the cells were to be labelled with [ 35 S]-methionine (trans- label, ICN) then the RPMI used after the electroporation was methionine-, and leucine-free and contained 2 mM NaHC0 3 and 25 mM HEPES and 10% dialyzed HI-FBS and the cells resuspended in a final volume of 10 mis with 20 μCi/ml [ 35 S]-methionine/leucine (phs TPA and ZnCl 2 , as above). After 12 hours (either with or without [ 35 S]) the cell suspensions were mixed with di-isopropyl fluorophosphate (1 mM final concentration), left for 5 minutes, then collected by centrifugation, washed 1 x with PBS, and lysed for immunoprecipitation. (EE)- epitope tagged proteins were immunoprecipitated from U937 cell lysates as follows. Cells from 1 electroporation were lysed into 1.25 mis of lysis buffer containing 1 %, w/v, Triton X-100, 25 mM HEPES, pH 7.4, 4°C; 1 mM EGTA; 1 mM MgCl 2 ; 0.15 m NaCl and 0.1 mM PMSF, 10 μg ml "1 leupeptin, 10 μgml "1 aprotinin, 10 μgml' 1 antipain 10 μgml "1 pepstatin A and 10 μgml "1 of bestatin (henceforth known as antiproteases I). The lysates were centrifuged (12,000 rpm,

0°C, 15 minutes). The resulting supernatants was removed and mixed with 4M NaCl (0.1 ml), 20% sodium cholate (25 μl) and protein G sepharose (40 μl packed volume) covalently coupled to an irrelevant, isotype-matched, mouse monoclonal antibody (at approx. 5-10 mg-ml "1 sepharose). The supernatants were mixed end over end for 20 minutes, then the beads were sedimented and the supernatant transferred to another tube with protein G sepharose beads (10 μl, packed) covalently crosslinked to α-(EE)-mouse monoclonal antibody (at approx. 5-10 mg- ml "1 sepharose). After 2 hours mixing at 4°C the immunoprecipitates were washed 5 x with 1.0% , w/v, Triton X-100; 0.4% cholate; 0.004% SDS; 0.4 M NaCl; 1 mM EGTA; 25 mM HEPES, pH 7.4, at 0°C; 1 x with 0.5M LiCl, 0.1M Tris, pH 8.0, 4°C; 2 x with 0.12M NaCl; 1 mM EGTA; 25 mM HEPES, pH 7.4, at 0°C. The last buffer wash was supplemented with 1 mM DTT (final wash buffer). If the immunoprecipitates were prepared from [ 3S S]-methionine labelled cells the last wash was omitted and the beads were boiled in SDS sample buffer. Otherwise, the samples were assayed for PI3K activity as described below.

11.1.4 COS-7 CELL TRANSFECTIONS

Exponentially growing Cos-7 cells were trypsinized/replated at about 50-70% confluence 3 hours prior to transfection. At the time of transfection they were again trypsinized, diluted into DMEM 10% FBS, counted, washed 2x in PBS and resuspended in EM (1 x 10 7 per cuvette) mixed with circular plasmid DNA (40 μg total per cuvette, made up of combinations of 10 μg of EXV-(EE)- / 3 ι , 10 μg of EXV-(myc)-r 2 , 10 μg of pCMV-(myc)-pl20 or 10-40 μg of an irrelevant mammalian expression vector), to give a final volume of 500 μl and then transferred to an electroporation cuvette (0.4 cms gap, BioRad). After 10 minutes at room temperature the cells were electroporated (250V, 960 μF), placed on ice for 8 minutes then diluted into DMEM 10% FBS. Aggregated cells were allowed to clump and were avoided as the cells were aliquoted into 6 cm dishes (four from each cuvette).

After 48 hours, the four dishes from each treatment were washed into HEPES -buffered DMEM containing 1 mM NaHC0 3 and 0.2% fatty acid-free BSA. After 10 hours two replicate dishes were harvested for Western blotting (with α- (myc) monoclonal antibody as the 1 ° detection reagent, α-mouse-HRP as the 2° detection system and quantitation by ECL and densiometric scanning). Two dishes

were washed into phosphate-free, DMEM, with 1 mM NaHC0 3 and 0.2% fatty acid-free BSA, then incubated for a further 90 minutes at 37°C with 300 μCi [ 32 P]- Pi per dish (in 4 mis). Media was aspirated and 1 ml of ice-cold 1 M HCI was added. The cells were scraped, removed and the dishes washed with 1.33 mis of methanol. The HCI and methanol washes were pooled with 2.66 mis of chloroform (to yield a 'Folch' two phase, solvent distribution) mixed and centrifuged. The lower phases were removed and mixed with 1.95 mis of fresh upper phase (see above) containing 0.5 mM EDTA and 1 mM tetrabutylammonium sulphate. After mixing and centrifugation, the lower phases were removed, dried down, deacylated and prepared for analysis by anion-exchange HPLC (Stephens et al. , 1991, Nature. Lond. 351:33-39, the disclosure of which is incoφorated herein by reference in its entirety). 11.2 RESULTS When transiently expressed in U937 cells, (EE)-plOl and (EE)-pl20 could be specifically immunoprecipitated from [ 35 S]-methionine labelled cells in approximately equal amounts (allowing for their relative compliment of methionines; 8:25 respectively, data not shown). Stringently washed α-(EE)-pl01 immunoprecipitates contained a wortmannin-sensitive, PtdIns(4,5)P 2 -selective, G γ-sensitive PI3K activity that was absent in controls using either an irrelevant monoclonal antibody for the immunoprecipitation, or a cDNA encoding an (EE)- tagged irrelevant protein which was expressed, as judged by [ 35 S]-methionine labelling, to similar levels as (EE)-plOl.

The activation by G/3γτs could be blocked by preincubation with a 2 fold molar excess of Gα-GDP. Further, the PI3K activity in these plOl immunoprecipitates was insensitive to Gα-GDP/aluminum fluoride. Co- tr.ansfection of (myc)-pl20 with (EE)-plOl did not increase the PI3K activity specifically recovered in α-(EE) immunoprecipitates. Indeed it decreased, probably because the expression of (EE)-pl01 was relatively lower in the presence of (myc)-pl20 expression vectors. In contrast, cells transfected with (EE)-pl20 α- (EE)-immunoprecipitates contained barely detectable PI3K activity either in the presence or absence of G / 3γs. These cells contained comparable moles of [ 35 S]- methionine labelled pl20 as there was (EE)-plOl in α-(EE)-immunoprecipitates from (EE)-plOl transfected cells. Co-transfection with (myc)-plθl resulted in a

substantial increase in, specifically, the G/3γτ-stimulated PI3K activity that could be recovered, despite a fall in the expression of (EE)-pl20 (as judged by [ 35 S]- methionine labelling) (see FIG. 8). This data indicates that U937 cells (human) contain an endogenous PI3K catalytic subunit that can bind to a transiently expressed plOl (pig). When bound to p 101 (pig) that endogenous catalytic subunit displays substantial regulation by G/3γs because all of the pi 20 present in the immunoprecipitates is bound to plOl.

In contrast, in α-(EE) immunoprecipitates from (EE)-pl20 transfected cells much of the pi 20 is unassociated with plOl and hence relatively inactive (compared to that bound to p 101 and in the presence of G/3γs). However, this PI3K activity, when assayed in the presence of G/3γs, is substantially amplified by co-transfection with (myc)-plθl . The alternative explanation for these data-that the pl20 is 'denatured' (although soluble and capable of being immunoprecipitated) unless expressed in the presence of p 101 -is unlikely in view of the data obtained with independently Sf9-purified, derived proteins.

To test whether the pl01/pl20 PI3K could be activated by G/3γs and produce PtdIns(3,4,5)P 3 in vivo, we transiently expressed various combinations of (myc)-γ 2 , (EE)-/3,, (myc)-plθl and (myc)-pl20 in Cos-7 cells and measured their effects on the levels of [ 35 P]-phosphoinositides in cells 48 hours after transfection (see FIG. 9). pl20 only produced significant increases in PtdIns(3,4,5)P 3 and

PtdIns(3,4)P 2 in a / 3,7 2 -dependent fashion in the presence of plOl . This pattern of results could not be explained by changes in the relative expression of the different cDNAs when introduced in combinations (see FIG. 9).

12. DEPOSIT OF CLONES

The following microorganisms or clones were deposited with the American

Type Culture Collection (ATCC), Rockville, Maryland, on the dates indicated and were assigned the indicated accession number: ATCC Date

Clone Access. No. of Deposit pCMV3mycpl01 97636 6/27/96

pCMV3mycpl20 97637 6/27/96

The present invention is not to be limited in scope by the specific embodiments described herein, which are intended as single illustrations of individual aspects of the invention, and functionally equivalent methods and components are within the scope of the invention. Indeed, various modifications of the invention, in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the appended claims.

94.1 -

International Application No: PCT/

MICROORGANISMS

Optional Sheet in connection with the microorganism referred to on page 32-33 , lines 1 -40 of the description

A. IDENTIFICATION OF DEPOSIT '

Further deposits are identified on an additional sheet

Name of depositary institution ' American Type Culture Collection

Address of depositary institution (including postal code and country)

12301 Parklawn Drive Rockville, MD 20852 US

Date of deposit ' June 27, 1 996 Accession Number ' 97636

B. ADDITIONAL INDICATIONS ' (leave blank if not applicable) This information is continued on a separate attached sheet

C. DESIGNATED STATES FOR WHICH INDICATIONS ARE MADE ' ( ,r u. ι»d„_« « «» m »,p««d su∞>

D. SEPARATE FURNISHING OF INDICATIONS ' (leave blank if not applicable)

The indications listed below will be submitted to the International Bureau later ' (Specify the general nature of the indications e g "Accession Number of Deposit")

E. D This sheet was received with the International application when filed (to be checked by die receiving Office)

(Authorized Officer)

D The date of receipt (from the applicant) by the International Bureau "

(Authorized Officer)

Form PCT/RO/1 34 (January 1981 )

94.2

International Application No: PCT/

Form PCT/RO/134 (cont.)

American Type Culture Collection

12301 Parklawn Drive Rockville, MD 20S52 US

Accession No. Date of Deposit 97637 June 27, 1 996