CA2329858A1 - Expression cloning of protein targets for phospholipids - Google Patents

Expression cloning of protein targets for phospholipids Download PDF

Info

Publication number
CA2329858A1
CA2329858A1 CA 2329858 CA2329858A CA2329858A1 CA 2329858 A1 CA2329858 A1 CA 2329858A1 CA 2329858 CA2329858 CA 2329858 CA 2329858 A CA2329858 A CA 2329858A CA 2329858 A1 CA2329858 A1 CA 2329858A1
Authority
CA
Canada
Prior art keywords
lipid
library
protein
moiety
proteins
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA 2329858
Other languages
French (fr)
Inventor
Vikram Rao
Joan Brugge
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Harvard College
Original Assignee
Harvard College
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Harvard College filed Critical Harvard College
Publication of CA2329858A1 publication Critical patent/CA2329858A1/en
Abandoned legal-status Critical Current

Links

Abstract

One aspect of the present invention relates to methods and reagents for identifying proteins or other cellular components (collectively "LBP" or "lipid binding partner"), which bind to lipids such as phospholipids, triacylglycerides, plasmalogens or sphingolipids. In preferred embodiments, the subject method is useful for identifying LBPs that bind to phospholipids such as phosphatidylserines, phosphatidylcholines (also called lecithins), phosphatidylethanolamines, phosphatidylglycerols, phosphatidylinositols, or sphingomyelins. The LBPs can be naturally occurring, such as proteins or fragments of proteins cloned or otherwise derived from cells, or can be artificial, e.g., poypeptides which are selected from random or semi-random polypeptide libraries.

Description

HMV-053.02 ~ 02329858 2000-12-28 Expression Cloning of Protein Targets for Phospholipids Related A,Dplications This application is contiunuation-in-part of USSN 09/---,--- filed 11 December 2000, which in turn claims priority to US Provisional application 60/170,009 ~xled 9 December 2000; the specifications of vsrhich are incorporated by reference herein.
Government Su ~ >, ort This invention was partially funded by NIIi Grant No. CA27951 and CA78773 from the National Cancer Institute; the goven~iment has certain rights to the invention.
Field of the Invention In an embodiment, this invention relates to the field of expression cloning of protein targets for phospholipids and to methods and uses therefor. In another embodiment, the invention relates to methods and reagents for identifying proteins or other cellular components which bind to a lipid moiety, to drug screening assays and to a method of conducting a drug discovery business.
Background of the Invention It is now well recognized that dynamic changes in the phosphorylatiott state of intracellular phosphatidylinositol (PtdIns) play critical roles i» mediating many cellular events. Phosphatidylinositol 3'-kinases (PI 3'-Ks) are a subfamily of PtdIns ltinases that phosphorylate the 3'-OH (D3) position of Ptdlns to create four different Ptdlns derivatives: PtdIas-3-P, PtdIns-3,4-P2, PtdIns-3,5-P2, and Ptdlns-3,4,5-P3.
Nine different isoforms of PI 3'-K have been identified in mammalian cells and they have been grouped into three classes by Domin and Waterfield based on the specific form of PI
that is used as a substrate.
Singly phosphorylated PtdIns-3-P is constitutively expressed in cells and is involved in a variety of events associated with membrane protein trafficking.
While all classes of Pl 3'-Ks can phosphorylate PtdIns to generate this lipid, the majority of PtdIns-3-P is probably produced by Class III PI 3'-Ks which is specific for Ptdlz~ts.
PtdIns-3,4-PZ
and PtdIns-3,4,5-P3 arc generated following stimulation by a wide variety of extracellular stimuli through many diverse classes of receptozs. Class I PI 3'I~s phosphorylate PtdIns-4,5-Pz to generate PtdIns-3,4,5-P3, which can be dephosphorylated to Ptdlns-3,4-P2 by the 5' lipid phosphatase SHIP. Alternate pathways to Ptdlns-3,4-PZ have also been described involving phosphorylation of the 4-position of Ptdlns-3-P by Class II PI 3'K
or an unidentified Ptdins-3-P 4-kinase, but the extent to which these enzymes contribute to PI -HMV-os~.o~
3,4-Pz synthesis in not clear.
Class I PI 3'-Ks play critical roles in many essential cellular processes.
Perhaps most importantly, these kinases regulate cell survival. Inhibition of class I
PI 3'-Ks leads to an induction of programmed cell death ox apoptosis arid constitutive unregulated activation of these enzymes or downstream targets of PtdIns-3,4-P2 and PtdIns-3,4,5-P3 can rescue cells from cell death induced by serum deprivation, loss of matrix attachment, myc expression, a»d other apoptotic stimuli. These kiz~ases also control the activation of marry iuntz~acellular signaling pathways that regulate cell proliferation including Erk/MAPKs, protein translation factors (e.g. eIF-4E), and cyclins /cyclin-dependent kinases. Also, membrane trafficking events regulated by 3'PPIs control receptor internalization. In additiozt, PI 3'-Ks are necessary for glucose transporter recruitment to the plasma membrane and regulation of glycogen synthase kinase 3 and phosphofructokinase, indicating that 3'PPIs are critically involved in insulin-mediated events associated with glucose metabolism. Integrin affinity modulation is also blocked by pharmacological inhibitors of PI 3'-K implicating these kinases in critical events associated with leukocyte traf~ckiua~g and inflammatory responses. PI 3'-K
also plays an important role in regulating cell movement and cytoskeletal rearrangements.
For example, 3'PPIs are necessary for controllieg receptor-induced changes in actin assembly, the formation of lamellipodial protrusions, and cell migration through the small GTP
binding protein Rae.
Because of the central importance o~ PI 3'-Ks in controlling cell proliferation, survival, and motility it is likely that class I PI 3'-Ks and 3'PPI binding proteins play an importazat role in the pathogenesis of cancer. Overexpression of PI 3'-K in chicken cells is sufficient to induce cellular transformation both in vitro and in vivo. PI
3'-K has also been implicated in the induction of Chronic Myelogenous Leukemia (CML) and Acute Lympltocytic Leukemia (ALL) by the $CR-ABL oncogene. ,As might be expected from its importance in cellular motility, PI 3'-K has been shown to play a role in tumor invasion and metastasis in several model systezaas_ ItecentIy, a role for PI
3'-K in human carcinogenesis was demonstz~ated by the evidence that the tumor suppressor PTEN, is a lipid phosphatase which is specific for 3'phosphate of the inositol head-group of and that elimination of the lipid phosphatase activity correlates with the oncogez~ic potential of PTEN mutants found in human cancez~s 3'PPIs.
There are several identilaed protein motifs that bind to 3'PPIs: PH domains, F'YVE domains, SH2 domains, and C2 domains. The pzamary function of these domains, HMV-053.02 each of which is approximately 90 to 120 amiuno acids in size, is believed to be localization of the protein to high local concentrations of 3'PPIs found near active signaling complexes at the cell membrane. However, there is evidence indicating that these domains can regulate protein function as well.
The most diverse and best-cb~aracterized 3'PPI binding dozr~ains are PH
domains which compzase a largo family of binding modules that are known to bind proteins as well as a wide range of lipids. A subset of PH domains binds with a high affinity to PtdIns-3,4-P2 and PtdIns-3,4,5-P3. These PH domains are critical for the function of several signaling proteins including the serine/threonine kiz~ases Akt and PDK1, the tyrosine kinase $tk, and the ARF-GAF Grpl.
L~Y~VE domains are recently characterized domains that contain a zinc finger, associate exclusively with PtdI,r~s-3-P, and are irnportant for vesicle sorting. SH2 domains bind primarily to phosphorylated tyrosines; however, the SH2 domains of PLCy, the Src tyrosine kinase, and the p85 subunit of PI 3'-K can also bind to Ptdlns-3,4,5-P3 with micromolar affinity. C2 domains bind to PtdTns-4,5-P2, Ptdlns-3,4-PZ, and Ptdlns-3,4,5-P3 and the specificity of lipid binding depends upon the local concentration of calcium. C2 domains are found in PKCs, PLA, and in vesicle sorting proteins such as synaptotagmin.
Sun, wa of th Invention The phosphatidylinositol 3-kinase (PI 3'-I~) family of lipid kinases play a critical role in cell proliferation, survival, vesicle trafficking, motility, cytoskeletal rearrangements, and oncogenesis. To identify downstreara effectors of PI 3'-K, we developed a novel scxeen to isolate proteins which bind to the major products of PZ 3'-K r~
phosphatidylinositol-3,4-bisphosphate (Ptdlns-3,4-PZ) and Ptdlns-3,4,5-P~.
This screen uses synthetic analogs of these lipids in conjunction with libraries of proteins that are produced by coupled in vitro transcription/translation reactions_ The feasibility of the screen was initially deianonstrated using avidin-coated beads pre-bound to biotizzylated PtdIns-3,4-T~2 and PtdIns-3,4,S,P3 to specifically isolate the PH domain of the serinelthreonine kinase Akt. We then demonstrated the utility of this technique in isolating novel 3'phosphorylated phosphatidylinositol (3'PPI) binding proteins through the preliminary screening of in vitro transcribed/translated cDNAs from a small pool expression library derived from mouse spleen. Three proteins were isolated that bound >~INIV-os3.o2 speei~cally to 3'PPIs. Two of these proteins have been previously characterized as PIP3$P/p42~P° and the PtdTns-3,4,s-P3-dependent serine/threonine kinase PDK1. The third protein is a novel protean that contains an SH2 domain azzd a PH domain which has a higher specificity for bout PtdIns-3,4,5-P3 and Ptdlns-3,4-P2 than for Ptdlns-4,s-Pz.
Transcripts of this novel gene (called PHISI~ for 3' Phosphoiunosztide Interacting SH2-Containing protein) are present in every tissue analyzed but are most prominently expressed in spleen.
This invention demonstrates the utility of this technique for isolating and characterizing 3'PPI binding protezns and specifically contemplates broad applicability for the isolation of bindiztg domains for other lipid products.
One aspect of the invention pz~ovides a method for identifying a cellular component which binds to a lipid moiety comprisixtg:
a. providing a lipid bait moiety being derivatized to a solid support;
b. contacting the lipid bait moiety with a library of cellular coraponents;
c. ider~txfying those mezobers of the cellular component library which specifically bind to the lipid bait moiety.
1n preferred embodiments, the lipid bait uaoiety is a phospholipid, e.g.
selected from the group consisting of of phosphatidylethanolamines, phosphatidylcholines, phosphatidylserines, phosphatidylglycerols, phosphatidylinositols, polyphosphatidylanositols, and diphosphatidylglycerols. In certain preferred embodiments, the phospholipid is derivatized to the solid support tlu4ugh a cross-linking moiety which is covalently attached to a phosphate head group of the phospholipid.
In other preferred embodiments, the lipid bait moiety a plasrnalogen or a sphingolipid.
In certain preferred embodiments, the library of cellular components is a polypeptide library, e.g., including at least 10 different polypeptides, more preferably at least 100, 1000, or even 10,000 di#fezent proteins. For instance, the polypeptide library can be an expression library, such as derived from replicable genetic display packages. Izx other embodiments, the polypeptide library is a cell lysate or partially puzif~ed protein preparation.
The ide~atity of those members of the cellular component library which specifically bind to the lipid bait uaoiety can eb determined by mass spectroscopy.
Another aspect of the present invention provides a screening assay comprising:
a. providing a reaction mixture including a cellular component identiFed as l3MV-os3.o2 described above fox its ability to specifically bind to the lipid bait moiety;
b, contacting the cellular component with a test compound;
c. determining if the test cuzz~poun~d bxr~ds to the cellular component.
In preferred embodiments, the the assay is repeated for a variegated library of at least 100 different test compounds, even mote preferably at least 100, 1000 or even 10,000 different test compouztds. Exemplary compounds wluicl~ can be screened for activity in the subject assays include peptides, nucleic acids, carbohydrates, small organic molecules, and natural product extract libraries, such as isolated from animals, plants, fungus aad/or microbes In certain preferred embodizaaents, the reaction mixture is a whole cell. In other embodiments, the reaction mixture is a cell lysate or purified protein composition.
In certain embodiments, a test compound which is identified as able to bind to the cellular component is further tested for the ability to inhibit or mimic the activity of a lipid moiety.
Still another aspect of the present invention provides a method of conducting a drug discovery business comprising:
a. providing a lipid bait moiety being derivatized to a solid support;
b. contacting the lipid bait moiety with a library of cellular components;
e. identifying those members of the cellular component library which specifically bind to the lipid bait moiety.
d. providing a reaction mixture including a cellular component identified in step (c) as able to specifically bind to the lipid bait moiety;
e, contacting the cellular component with a test compound;
f. determining if the test compound binds to the cellular component;
g. further tesring those test compound identified in step (f) as able to bind to the cellular component for the ability to inhibit or mimic the activity of a lipid moiety; and h. formulating a pharmaceutical preparation includixag one or more compounds identified in step (g) as able to inhibit or mimic the activity of a lipid moiety.
Xet another aspect of tlae invention pz~ovides a method of conducting a target discovery business comprising:
a. providizxg a lipid bait moiety being derivatized to a solid support;
s HMV-053.02 b, contacting the Iipid bait moiety with a library of cellular components;
c. identifying those members of the cellular component library which specifically bind to the lipid bait moiety.
d. licensing, to a third party, the rights for drug development for a cellulax component identified in step (c) as able to specifically bind to the lipid bait moiety.
Brief Description of the Drawings FIG. 1. Synthesis of biotinylated PIPn probes and structure of dioctanoyl derivatives FIG. 2. The PH domain. of Akt binds specifically to avidin beads pre-bound with biotinylated 3'PPIs. A. 3sS- labeled maltose binding protein (N>BP) or MBP
fused to the PH domain of Akt (M.BP-PH) (lanes 1 and 5) were incubated with avidin beads alone (lanes 2 and G), avidin beads pre-bound with PtdIns-3,4-P2-biotin (lanes 3 and 7), avidin beads pre-bound with PtdIns-3,4,5-P3 -biotin (lanes 4 and 8). Proteins were labeled with 3sS-methionine by in vitro transcriptionltxanslation of 0.5 pg of the respective genes in pCS2(+) and the binding reactions were done as described in "Experimental Procedures".
Truncated species of the M)3P and MBP-PH (eMBP, ~MBP-PH) result from initiation of translation at start sites after the initial AUG codon. $. Avidin beads pre-bound with Ptdlns-3,4-Pz-biotin can speciEcally isolate M13P-PH from a pool of other proteins. 10 ng of MBP-PH DNA and/or ~ pg of DNA from a random pool from the cDNA library were transcribedltranslated in the presence 3sS-methionine and binding reactions were performed as described in "Experimental Procedwres". Total labeled proteins (lanes l, 4, and 7), labeled proteins bound to avidin beads (lanes 2, 5, and 8), labeled proteins bound to avidin beads pre-bound with Ptdlns-3,4-P2-biotin (lanes 3, 6, and 9). C.
PtdIns-3,4,5-P3 az~d PtdIns-3,4~P2 preferentially displace MBP-PH ~rom Ptdlns-3,4,5-P3-biotin.
3sS-labeled MBP-PH was bound to avidin beads coated with Ptdlns-3,4,5-P3-biotin in the presence of the indicated concentrations of PtdTns-4,S-Pi (squares), Ptd~s-3,4-PZ
(circles), Ptdlns-3,4,5-P3 (triangles) and processed as described in ilxperimental Prncedures?. Points represent the mean of two independent experiments.
FIG 3. Isolation of marine isoforms of PDK1 and Is~P3BP/p42~4 from expression library via avidin beads pre-bound with biotinylated 3'PPIs. A. PDK1. Top panel.
SpeeiSc binding of PDK1 in total pool and as single clone. Total 3sS-labeled proteins 1"IMV-053.02 (lanes 1 and 5), labeled proteins bound to avidin beads (lanes 2 and 6), labeled proteins bound to avidin beads pre-bound with Ptdlns-3,4-P2-biotin (lanes 3 and 7), labeled proteins bound to avidin beads pyre-bound with PtdZns-3,4,5-P3 -biotin (lanes 4 and 8).
Bottom panel. Lower diagrams show the protein domain structure of PD1C1 and corresponding cDNA fragment isolated from the expression library. Nucleotide positions of putative stop and start codons (AUG) are indicated. B. PIP38PIp4z~4. ~'op panel.
Specific binding of Pl~'3BPIp42~4 in total pool and as single clone. Total j5S-labeled proteins {lazes 1 and 5), labeled proteins bound to avidin beads (lanes 2 and 6), labeled proteins bound to avidiz~ beads pre-bound with Ptdlns-3,4-Pz-biotin (lanes 3 and 7), labeled proteins bound to avidin beads pre-bound with Ptd,Ins-3,4,5-P3 -biotin {lanes 4 and 8). Bottom panel. Lower diagrams show the protein domain structure of PIP3BPIp42~p4 and corresponding cDNA fragment isolated from the expression library.
Nucleotide positions of putative stop and start codons (AUG) are indicated.
FIG. 4. Isolation of PHISH from expressxrn~ library via avidin beads pre-bound with biotinylated 3'PPIs. A. Specific binding of PHISH in total pool azrd as sxz~gle clone.
Total 35S-labeled proteins (lanes x and 5), labeled proteins bound to avidin beads (lanes 2 and 6), labeled proteins bound to avidin beads pre-bound with PtdIns-3,4-Pz-biotin (lanes 3 and '~), labeled proteins bound to avidin beads pre-bound with PtdIns-3,4,5-P3 -biotin (lanes 4 and 8). B. Ptdlns-3,4,5-P3 and Ptdlns-3,4-Pz preferentially displace PHISH from Ptdlns-3,4,5-P3 -biotin bound to avidity beads. 35S-labeled PHZSH was bound to avidity beads coated with PtdIns-3,4,5-P3-biotin in the presence of the indicated concentrations of Ptdins-4,5-P2 (squares), PtdTns-3,4-Pi {circles), Ptdlns-3,4,5-P3 (firiangles) and processed as described in iExperimental I'roceduresi. Points represent the mean of two independent experiments.
FIG. 5. Nucleotide and amino acid sequence of marine PHISH. Sequence of PHISH isolated via expression elotaing. Putative SH2 domain is outlined in red, predicted tyrosine phosphorylation site is outlined in green, putative PH domain is outlined izt blue.
FIG. 6_ Northern blot of mRNA from various naurine tissues hybridized to prabes derived from PHISH and G,A,PDH. Marine tissue RNA (l0ug per lane) was subjected to agarose gel electrophoresis, transferred to nylon membrane, and hybridized to 3ZP-labeled probes derived from the coding regions of PHISH or GAPDH as described in iExperimental Proceduresl. Lazes: 1-brain, 2-heart, 3-lung, 4-lymph node, 5-spleen, 6-thymus. Arrows represent the mobility of the nnajor (large arrow) and minor (small arrow) products of an in vitro transcription reaction of the PHISH gene isolated from the HMV-053.02 expression library.
Detailed ~escrintion of the lfnveatioa A) Overview One aspect of the present invention relates to methods and reagents fax identifying proteins or other cellular components (collectively "LBP" or "lipid binding partner"), which bind to lipids such as phospholipids, triacylglycerides, plasmalogens or sphingolipids. In preferred embodiments, the subject method is useful for identifying LBPs that bind to phaspholipids such as phosphatidylserines, phosphatidylcholines (also called lecithins), phosphatidylethanolamines, phosphatidylglycerols, phosphatidylinositols, or sphingomyelins. The Ll~~'s can be naturally occurring, such as proteins or fragments of proteins cloned or otherwise dewed from cells, ox can be artificial, e.g., polypeptides which are selected from random ox secni~random polypeptide libraries.
In general, the method of the present invention comprises providing a lipid which includes an "sequestration tag", and contacting the lipid with a structurally diverse (variegated) library of polypeptides or other molecules under conditions wherein binding of lipids to library molecules can occur such the resulting complexes are enriched for library molecules which specifically (as opposed to non-specifically) bind the lipids.
Library molecules which specifically bind to the lipids are isolated from the library, or their identity is otherwise determined, e,g., by the presence of a tag associated with the LBP which is a unique identifier of the LBP. The polypepiide library can be provided as part of a replicable genetic display package, an expression library (especially an intracellular expression library), a synthetic polypeptide library or other form.
In other embodiments, the system can be reversed and a polypeptide can be used to screen a library of structurally diverse lipids to identify lipids which selectively bind to the polypeptide.
Another aspect of the present invention relates to the LBPs which are identified by the subject method. Such zzaolecules can be used as drug screening targets, e.g., for drugs which alter the activity of the LBP {such as its ability to bind a lipid) or which alter the level of the LBP in the cell. lV,loz~eaver, the level of an LBP in a cell can be determined for diagnostic or prognostic purposes.

z~l~v-os3.o2 Where the LBP is a protein, the invention also relates to nucleic acids which encode the protein or a fragment thereof ~'he invention also contemplates nucleic acids which hybridize to the coding sequence fax an LBP, e.g., which may be useful as amplim,ers, probes, primers or antisense.
Another aspect of the present invention relates to antibodies, e.g., monoeolonal, purified and/or recombinant, which are a~unoseleetive for an LBP.
Still another aspect al: the present invention relates to drug screening assays for identifying compoutxds, e.g., such as small organic molecules (1V1W~1000amu) which inhibit or potentiate the activity of an LBP. For instance, the assay can be used to identify compounds which inhibit or potentiate an intrinsic enzymatic activity of an LBP, or the ability of the LBP to bind to otb,er molecules, e.g., to lipids, to proteins, to nucleic acids.
Yet another aspect of the present invention relates to the use the LBPs, or compounds which agozuize or antagonize, as the case may be, the activity of an LBP, far the treatment or prevention of a disorder or unwanted effect mediated by a lipid.
B) De nitions Before further description of the inventia~a, certain terms employed in the specification, examples and appended claims are, for convenience, collected here.
"Fatty acids" are long-chain hydnxarbon molecules containing a carboxylic acid moiety at one end. The numbering o,f carbons in fatty acids begins with the carbon of the carboxytate group. Fatty acids that contain no carbon-carbazt double bonds are termed saturated fatty acids; those that contain double bonds are unsaturated fatty acids. The numeric designations used for fatty acids come from the number of carbon atoms, followed by the number of sites of unsaturation (eg, pahnitie acid is a 1 b-carbon fatty acid with no unsaturation az~d is designated by 16:0). The site of unsaturadan in a fatty acid is iundicated by the symbol D and the number of the first carbon of the double band {e.g.
palmitoleic acid is a I6-carbon fatty acid with on,e site of unsatwration between carbons 9 and 10, and is designated by 16:109).
"Triacylglycerides" are composed of a glycerol backbone to which 3 fatty acids are esterified.
'fhe basic structure of '~hospoIipids" is very similar to that of the triacylglycerides except that C-3 (sn3~f the glycerol bacL~bone is esterihed to phosphoric l3lvw-os3.az acid. The building block of the phospholipids is phosphacidic acid which results when the X substitution in the basic structure shown in the Figure below is a hydrogen atom.
Substitutions include ethanolamine (phosphatidylethanolarnine), choline (phosphatidylcholine, also called Iecithins), serine (pbosphatidylserine), glycerol (phosphatidylglycerol), myo-inositol (plxosphatidylinositol, these compounds can have a variety in the nutnbezs of inositol alcohols that are phosphvrylated generating polyphosphatidylinositols), and phosphatidyIglyeerol (diphosphatidylglycerol more commonly known as cardiolipins), "Plasmalogens" are complex membrazxe lipids that reserrxble phospholipids, principally phosphatidyleholine. The major difference is that the fatty acid at C-1 (snl) of glycerol contains either an O-alkyl ax O-alkenyl ether species. A basic O-alkenyl ether species is shown in the Figure below. Gne of the most potent biological molecules is platelet activating factor (PAF) which is a choline pIasmalogen in which the C-2 (sn2) position of glycerol is esterified with an acetyl group insted of a lvng chain fatty acid.
"Sphingolipids" are corttposed of a backbone of sphingosine which is derived itself from glycerol. Sphingosizze is N-acetylated by a variety of fatty acids generating a family of molecules referred to as ceramides. Sphingolipids predominate in the myelin sheath of nerve fibers. Sphingomyelin is an abundant sphingolipid generated by transfer of the phasphocholine moiety of phosphatidyIcholine to a ceramide, tluus sphingomyelin is a unique form of a phospholipid. The other major class of sphingolipids (besides the sphingorayelins) are the glycosphingolipids generated by substitution of carbohydrates to the snl carbon of the glycerol backbone of a ceramide. There are 4 major classes of glycosphingolipids:
Cerebrosides: contain a single moiety, principally galactose.
Sulfatides: sulfuric acid esters of galactocerebrosides.
Globosides: contain 2 or more sugars.
Gangliosides: similar to globosides except also contain sialic acid.
The term "simultaneously expressing" refers to the expression of a representative population of a polypeptide library, e.g., at least 50 percent, more preferably 7s, 80, 85, 90, 95 or 98 percent of all the different pvlypeptide sequences of a library.
The team "random polypeptide library" refers to a set of random or semi-random polypeptides.

-~S ~ .~~
The language "replicable genetic display package" or "display package"
describes a biological particle which has genetic information providing the particle with the ability to replicate. The package can display a fusion protein iz~cludiz~g a polypeptide derived from the variegated pvlypeptide library. The test polypeptide portion of the fusion protein is presented by the display package in a context which permits the polypeptide to bind to a lipid that is contacted with the display package. The display package will generally be derived froz~ra a system that allows the sampling of very large variegated polypeptide libraries. The display package can be, for example, derived from vegetative bacterial cells, bacterial spores, and bacterial viruses.
The language "differential binding means", as well as "affinity selection" and "afflnxty enrichment", refer to the separation of members of the polypeptide display library based on the differing abilities of polypeptides on the surface of each of the display packages of the library to bind to the lipid lipid. The differential binding of a lipid by test polypeptides of the display can be used in the affinity separation of those polypeptides which specifically bind the lipid from those which do not. For example, the affinity selection protocol can also include a pre- or post-exurichznent step wherein display packages capable of binding "background lipids", e.g., as a negative selection, are removed from the library. Examples of affinity selection means include aflaz~ity chromatography, immunoprecipitation, fluorescence activated cell sorting, agglutination, and plaque lifts. As described below, the affinity chromatography includes bio-panning techniques using either purified, inamabilized lipid proteins or the li~l~e, as well as whole cells.
The phrases "individually selective manner" and "individually selective binding", with respect to binding of a test polypeptide with a lipid, refers to the binding of a polypeptide to a certaiza. protein lipid which binding is specific for, and dependent oz~, the molecular identity of the protein lipid.
The term "solid suppork" refers to a material having a rigid or semi-rigid surface.
Such materials will preferably take the fvz~zn of small beads, pellets, disks, chips, dishes, mufti-well plates, wafers ox the like, although other forms may be used. In some embodiments, at least one su~'ace of the substrate will be substantially flat.
The term "surface" refers to any generally two-dimensional structure on a solid substrate and may have steps, ridges, kinks, tezxaces, and the like without ceasing to be a surface.
In an exemplary embodimexxt of the present invention, the display package is a phage parCicle which comprises a polypeptide fusion coat protein That includes the amino ~tvlv-os3.o2 acid sequence of a test polypepcide. Thus, a library of replieable phage vectors, especially phagemids (as defined herein), encoding a library of palypeptide fusion coat proteins is generated and used to transform suztable host cells. Phage particles formed from the chimeric protein can be separated by affnity selection based on the ability of the polypeptide associated with a particular phage particle to specifically bind a lipid. In a preferred embodiment, each individual phage particle of the library includes a copy of the corresponding phagemid encodizzg the polypeptide fizsion coat protein displayed on the surface of that package. Exezxtplary phage for generating the present variegated polypeptide libraries include M13, fl, fd, Ifl, Ike, Xf, Pfl, Pf3, ~., T4, T7, P2, P4, ~X-174, MS2 sari f2.
The language "fusion protein" and "chimeric protein" are art-recognized terms which are used interchangeably heo~ein, and include contiguous polypeptides comprising a first polypeptide covalently linked via an amide band to one or more amino acid sequences which define polypeptide domains that are foreign to and not substantially homologous with any domain of the first polypeptide. One portion of the fusion pmtein comprises a test polypeptide, e.g., which can be random or semi-random. A
second polypeptide portion of the fusion protein is typically derived from an outer surface protein or display anchor protein which directs the "display package" (as hereafter defined) to associate the test polypeptide with its outer surface. As described below, where the display package is a phage, this anchor protein can be derived from a surface protein native to the generic package, such as a viral coat proteiua. Where the fusion protein comprises a viral coat protein and a test polypeptide, it will be referred to as a "polypeptide fusion coat protein". The fusion protein further comprises a signal sequence, which is a short length of amino acid sequence at the amino terminal end of the fusion protein, that directs at least the portion of the fusion protein including the test polypeptide to be secreted from the cytosol of a cell and localized on the extracellular side of the cell membrane.
Gene constructs encoding fusion proteins are likewise referred to a "chimeric genes" or "fusion genes".
The temp "vector" refers to a DNA, molecule, capable of replication in a host cell, into which a gene can be inserted to construct a recombinant DNA xnalecule.
The terms "phage vector" and "phagemid" are art-recognuized and generally refer to a vector derived by modification of a phage genome, containing an origin of replication for a bacteriophage, and preferably, though optional, an origin (orl~ for a bacterial HMV-05 3 .02 plasmid. The use of phage vectors rather than the phage genome itself provides greater flexibility to vary the ratio of ehiznezic polypeptideleoat protein to wild-type coat protein, as well as supplement the phage genes with additional genes encoding other heterologous polypeptides, such as "auxiliary polypeptides" which znay be useful in the "dual"
polypeptide display constructs described below.
The language "helper phage" describes a phage which is used to infect cells containing a defective phage genome or phage vector and which functions to complement the defect. The defect can be one which results from removal ox inactivation of phage genomic sequence required for pzoduction of phage particles. examples of helper phage are M13K07.
As used herein, a "reporter gene construct" is a nucleic acid that includes a "reporter gene" operatively linked to at least one transcriptional regulatory seduence.
Transcription of the reporter gene is controlled by these sequences to which they are linked.
The term "sequester", as used herein, means to separate, segregate, remove, or bind a lipid complex, e.g., on a solid support. rn preferred embodiuzzez~ts, a lipid complex is sequestered by a solid support such that other non-sequestered LBPs can be removed, e.g., by washing or ocher purification techniques. A lipid complex is "reversibly sequestered" if the process of sequestering the complex on a solid support can be reversed to yield a free complex or free LBP, e.g,, in solution in a reaction mixture.
In preferred embodiments, the process of sequestering a complex, or of reversing the sequestration, or both, occurs under mild cortditivns and in high yield, e.g., greater than at least about 40%
yield.
The term "polymeric support", as used herein, refers to a soluble or insoluble polymer to which a lipid can be covalently bonded (e.g., by through an ester functionality) by reaction with a functio»al group of the polymeric support. Marry suitable polymeric supports arc known, and include soluble polymers such as polyethylene glycols or polyvinyl alcohols, as well as insoluble polymers such as polystyrene resins.
A suitable polyme~c support includes functional groups such as those described below. A
polymeric support is termed "soluble" if a polymer, or a polymer-supported compound, is soluble under the conditions employed- However, in genezal, a soluble polymer can be rendered insoluble under defined conditions. Accordingly, a polymeric support can be soluble under certain conditions and i~x~soluble under other conditions- A
polymeric support is termed "insoluble" if reaction of a lipid with the polymeric support results in an HMV-os3.oz iztsoluble polymer-supported lipid under tl~e conditions employed.
Abbreviations used herein include: AID - ADP ribosylation factor; Btk -Bretons tyrosine kinase; DTT - dxtlxxothreitol; ErkIMAPK - extracellular regulated kinase/mitogez~
activated protein kiz~ase; EST - expressed sequence tag; GAP = GTPase activating protean; GAPDI~ = glyceraldehyde 3-phosphate dehydragenase; GTP - guanosine triphosphate; I-iEPES - (N-[z-hydroxyethyl]piperazine-N'-[2-ethanesulfonic acid); kb kilobase; SAS - sodium dodecyl sulfate; MBP = maltose binding protein; MBP-PI-i =
maltose binding protein Akt P~ domain fusion protein; NP-40 -nonylphenylpolyethylene glycol; 3'PPI - 3' phosphorylated phosphatidylinositols; PAGE - polyacrylamide gel electrophoresis; PCR- polymerise chain reaction; PDK1- phosphoiz~ositide dependent kiztase 1; PH = pleckstrin homology; PI3'-K = phosphatidylinositol 3'-kinase;
PKC
protein kinase C; PLA - phopholipase A; PLCy - phospholipase CY; Ptdlns -phosphatidylinositol; PtdIns-3-P - phosphatidylinositol-3-monophasphate;
PtdIns-3,4-P2 phosphatidylinositol-3,4-bisphosphate; Ptdlns-4,5-Pz - phosphatidyliz~ositol-4,5-bisphosphate; PtdIns-3,4,5-P~ - phosphatidylinositol-3,4,5-trisphosphate; SDS -sodium dodecyl sulfate; SH2 = Src homology 2.
G) F.xemplarv Embodiments o~'Phos~pleolipid Baits As set forth above, in certain embodiments, the subject method can be practiced by utilizing immobilized lipid moieties, such as phospholipids, as the bait for identifying polypeptides and other molecules capable of interacting with, and fonming complexes with the lipid moiety. In certain embodiments, the subject lipid moiety is a phospolipids, such as selected from the group consisting of phosphatidylethanolamines, phosphatidylcholines, phosphatidylserines, phosphatidylglycerals, phosphatidylinositols, polyphosphatxdylinositols, and diphosphatidylglycerols. E~cemplary polyphosphaddylinositols include:
di C16, L-a-D-myo-Phosphatidylxriositol 3-monophosphate di C8, L-a-D-myo-Phosphatidylinositol 3-monophosphate di C 16, L-a-D-myo-Phosphatidyiinositol 3,4-diphosphate di C8, L-a-D-myo-Phosphatidylinositol 3,4-diphosphate di C 15, L-a-D-myo-Phosphatidylinositol 3,4,5-diphosphate di C8, L-a-D-myo-Phosphatidylinositol 3,4,5-diphosphate di C16, L-a-D-myo-Phosphatidylinositol 3,5-diphosphate di C8, L-a-D-xnyo-Phosphataclyliilositol 3,5-ditphosphate di C16, L-a-D-myo-Phosphatidylinositol 4-monophosphate I-1MV-053.02 dl C8, L-a-D-myo-Phosphatidylinositol 4-monophosphate dl C16, L-a-D-myo-Phosphatidylinositol 4,5-diphosphate dl C$, L-a-D-myo-Phosphatidylinositol 4,5-dxphosphate dl C16, L-a-D-myo-Phosphatidylinositol 5-monophosphate dl C8, L-a-D-myo-Phosphatidylinositol 5-moz~ophosphate ~n other embodiments, the subject lipid moiety is a plasmalogen. In still other embodiments, the subject lipid moiety is a sphingolipid, such as may be selected from the goup consisting of cerebrosides, sulfatides, globosides, and gar~gliosides.
In certain preferred embodiments, the subject lipid can be immobilized Qr incorporated into a polymer or other inspluble matrix by, for example, derivativatior~ with one oz~ more of subject lipid moieties derivatized to a solid support, such as glass, silicon, or a polymeric support. The support can be, inter ills, a bead, a chip, a hydrogel, etc.
In certain preferred embodiments, the subject lipid moieties are derivatized by covalent or non-covalent coupling through one or more of its fatty acid side chains, e.g., in order to present at least a portion of its head group. For example, the present invention specx~cally contemplates phosphatidyliz~ositol derivatives represented by the general formula:
Hz P

H -X-R' G-X-R' W~lCreln X, independently for each occurrence, represents O or S;
R, independently for each occurrence, represents hydrogen or -P03;
R' represents, for each occurrence, -CH~HR3-L or -CORD-L;
R3 and R4, independently for each occurrence, represent a C6-C2a alkyl group, e.g., which may be saturated or unsaturated, branched or lxz~ear, substituted or unsubstituted; and L represents a linker, or a linker covalently or non-covalently attached to a solid support.

HMV-053.02 In certain preferred embodiments, X represents O; and L is a Iiz~.ker of 150-1500amu, such a biotin.
In certain embodiments, particularly where zztore than one type of lipid-moiety is used as a bait (e.g., a library of different Iipid moieties), a spatial array o~ lipid baits can be generated, e.g., for libxary versus library scz~eening. For example, libraries of at least different lipid moieties can be tested as baits, and zxtaxe preferably libraries of at least 100 or even 1000 different lipid moieties.
The lipid moiety can be derivatived to the support by any of a number of means.
In the case of phospholipids, the derivatization is preferably through a phosphate head group. As described in the appended examples, biotinylation of the phosphate head group can be used to derivatize the lipid moiety to an avidin-displaying support.
There are a large number of other chemical crass-linking agents which could be used in the present invention are known in the art. For the prese~at invention, the preferred cross-linking agents are heterobifunctionaI cross-linkers, which can be used to link the lipid bait and solid support in a stepwise mawner. Heterobifunctional cross-linkers provide the ability to design more specific coupling methods for conjugatiung the subject moieties, thereby reducing the occurrences of unwanted side reactions such as homo-lipid polymers. A wide variety of heterobifunetional cross-linkers are known in the art. These include: succinimidyl 4-(N-maleimidomethyi) cyclohexane-1-carboxylate (SMCC), m-Malezmidobenzoyk-N- hydroxysuccinimide ester (kl~S); N-succinimidyl (4-iodoacetyl) aminobenzoate (SI:AB), succinimidyl 4-(p-maleimidophenyl) butyrate (SMPB), 1-ethyl-3-(3-dimethylamxnopropyl~arbodxirnide hydrochloride (EDC); 4-succinimidyloxycarbonyl-a-methyl-a-(2-pyridyldithio)-tolune (SMPT), N-succinimidyl 3-(2-pyridyldithio)propionate (SPDP), succinimidyl 6->3-(2-pyridyldithio)propionate!hexanoate (LC-SPDP). Those cross-linking agents having N-hydroxysueeinxmide moieties can be obtaiuaed as the N.-hydroxysulfosuccinimide analogs, which generally have greater water solubility. In addition, those cross-linking agents having disultade bridges within the linking chain can be synthesized instead as the alkyl derivatives so as to reduce the amount of linker cleavage in vivo.
I» addition to the heternbifunctional cross-linkers, there exists a number of other useful cross-linking agents including homobifunetional a~ad photoreactive cross-linkers.
Disucciniznidyl suberate (D55), bismaleimidohexane (BMH) and dimethylpimekiznidate-2 ICI (DMP) are examples of useful ho~naobifunctional cx-oss-linking agents, and bis-~3-(4-azidosalicylamido~thyldisulfide (BASED) and N-succinimidyl-6(4'-azido-2'-I IMV-053.02 nitrophenylamino)hexaxtvate (SANfAIT) are exanraples of useful photoreactive cross-linkers foz use izt this invention. For a review of coupling techztiques which may be applied to the subject lipid moieties, see Means et al. (1990) Bioconjugate Chemistry 1:2-12.
The third component of the heterobifunctional crass-linker is the spacer arm or bridge. The bridge is the structure that connects the two reactive ends. The nrtost apparent attribute of the bridge is its effect on steric hindrance. In some instances, a longer bridge can more easily span the distance necessary to link two complex biomolecules.
For xz~stance, SMPB has a span of 1a.5 angstroms.
D) ExemDlarv Embodiments ofPolvpe~ptide Libraries One goal of the present method is to identify proteins which are bound by the lipid bait. Accordingly, the present invention contemplates that any of a number of methods for trapping protein complexes using non-protein baits can be used. For instance, the proteins which are bound to the lipid bait can be identified by sequencing using mass spectroscopy. This technique can be advazztageous when the source of test proteins is a cell lysate. In other embodiments, the polypeptides ~e associated with a tags) which identi~tes the sequence of the protein, or with the gene which encodes the protein. In still other instance, t'he proteins are provided as part of a spatial array for which the coordinates on the array provides the identity of the protein.
In certain preferred e~mbvdiznents, the polypeptide library is provided as an expression. library. For instance, a library of test pvlypeptides is expressed by a population of display packages to farm a peptide display library. With respect to the display package on which the variegated peptide library is manifest, it will be appreciated from the discussion provided herein that the display package will preferably be able to be (i) genetically altered to encode heterologous peptide, (ii) rraaintained and amplified in culture, (iii) manipulated to display the peptide-containing gene product in a manner pezrrxitting the peptide to intezact with a lipid during an affinity separation step, and (iv) affinity separated while retaining the nucleotide sequence encoding the test pvlypeptide (herein "peptide gene's such that the sequence of the peptide gene can be obtained. In preferned embodizxteztts, the display remains viable after affinity separation.
Ideally, the display package comprises a system that allows the sampling of very large variegated peptide display libraries, rapid sorting after each affinity separation HMV-0s3.02 round, and easy xsolatian of the peptide gene from puni~ed display packages yr further manipulation of that seduenee in the secretion zr~ode. The most attractive candidates for tlxis type of screening are prokaryotic organisms and viruses, as they can be amplified quickly, they are relatively easy to xzxanipulate, and large number of clones can be created.
Preferred display packages include, for example, vegetative bacterial cells, bacterial spores, and most preferably, bacterial viruses (especially DNA viruses).
However, the present invention also contemplates the use of eukaryotic cells, including yeast and their spores, as potential display packages.
In addition to coznnaercially available kits for generating phage display libraries (e.g. the Pharmacia Recombinant Phage Antibody System, catalog no. 27-9400-O1;
and the Stratagene Surfhr4PTM phage display kit, catalog no. 240612), examples of methods and reagents particularly amenable for use in gez~erati~ng the variegated peptide display library of the present invention can be found in, for example, the Ladner et al. U.S. Patent No. 5,223,409; the King et al. International Publication No. WO 92/18619; the Dower et al. International Publication No. WO 91/17271; the Winter et al. International Publication WO 92120791.; the Marklan,d et al. international Publication No. WO 92/15679;
the Breitling et al. International Publication WO 93/01288; the MeCat~'eriy et al.
International Publication No. WO 92101047; the Garrard et al. International Publication No.
WO
92109690;, the L,adner et al. International Publication No. WO 90102$09; Fuchs et a1.
(1991) BiolTechnology 9:1370-1372; Hay et al. (1992) Hum Antibod Hybridornas 3:81-85; Huse et al. (1989) Science 246:1275-1281; GTif~hs et al. (/993),E,I~BOJ
12:725-734;
Hawkins et al. (1992) .T ll~lol Biol 226:889-896; Clackson et al. (1991) Nature 352:624-628; Gram et al. {1992} PNAS 89:3576-3580; Garrad et al. (1991) NiolTechnology 9:1373-1377; Hoogenboom et al. (/991) Nuc Acid Res 19:4133-4137; and Barbas et al.
(1991 ) PNAS 88:'797$-7982. These systems can, with. zraadx~cations described herein, be adapted for use in the subject method.
When the display is based on a bacterial cell, or a phage which is assembled periplasmically, the display means of the package will comprise at least two components.
The first component as a secretion signal which directs the recombinant peptide to be localized on the extracellular side a~ the cell membrane (of the host cell when the display package is a phage). This secretion signal can be selected so as to be cleaved off by a signal peptidase to yield a prneessed, "mature" peptide. The second component is a display anchor protein which directs the display package to associate the test polypeptide HMV-053.02 with its outer surface. As described below, this anchor proteizz can be derived from a surface or coat protein native to the genetic package.
Whets the display package is a bacterial spore, or a phage whose protein coating is assembled intracellularly, a secretion signal directing the peptide to the inner membrane of the host cell is unnecessary. In these cases, the means four arraying the variegated peptide library comprises a dezavative of a spore or phage coat protein amenable for use as a fusion protein.
Irt some instances it may be necessary to introduce an unstructured polypeptide linker region between portions of the chimeric protein, e.g., between the test polypeptide and display polypeptide. 'this linker cart facilitate enhanced flexibility df the chimeric protein allowing the test polypeptide to freely interact with a lipid by reducing steric hindrance between the two fragments, as well as allowing appropriate folding of each portion to occur. The linker can be of natural origin, such as a sequence determined to exist izt random coil between two domains of a protein. Alternatively, the linker can be of synthetic origin. For instance, the sequence (Gly4Ser)3 can be used as a synthetic unstructured linker. Linkers of this type are described in Hustozt et al.
(1988) PNAS
85:4879; and U.S. Patent Nos. 5,091,513 and S,Z58,498. Naturally occurring unstructured linkers of human origin are preferred as they reduce the risk of immunogetticxty.
In the instance wherein the display package is a phage, the cloning site for the test polypeptide gene sequences in the phagemid should be placed so that it does not substantially interfere with normal phage function. One such locus is the intergenic region as described by Zinder and Boske, (1982) Gene 19:1-10.
The number of possible combinations in a peptide library can get large as the length is increased and selection criteria for degenerating at each position is relaxed. To sample as many combinations as possible depends, in part, on the ability to recover large numbers of transforrxtattts. For phage with plasznid-like forms (as ~lamentous phage), eleetrotransformation provides azx efficiency comparable to that of phage-transfeetion with in vitro packaging, in addition to a very high capacity for DNA iun~put.
This allows large amounts of vector DNA to be used to obtain very large numbers of transformants.
The method described by Dower et al. (1988) Nucleic Acids Res., 16:6127-6145, for example, may be used to transform fd-tet derived recombinants at the rate of about 107 transformants/ug of ligated vector into E. coli (such as strain MC1.061.), and libraries may be constructed in fd-tet Bl of up to about 3 x 108 members or more. Increasing DNA

HMV-053.02 input and making modifications to the cloning protocol withizt the ability of the skilled artisan may produce increases of Beater than about 10- fold izt the recovery of transfoz~zz~ants, providiztg libraries of up to 101 or more recombinants_ ,As will be apparent to those skilled in the art, in embodiments wherein high affinity peptides are sought, an important criteria for the present selection method can be that it is able to discriminate between peptides of different affinity far a particular lipid, and preferentially enrich for the peptides of highest affinity. Applying the well k»own principles of peptide affinity and valence (i.e. avidity), it is understood that manipulating the display package to be rendered effectively znonovalent can allow affinity enrichment to be carried out for generally higher binding afi~zaities (i.e. binding constants in the range of 106 to IUI~ M-~) as compared to the broader range of affinities isalable using a multivalent display package. To generate the monovalent display, the natural (i.e. wild-type) form of the surface or coat protein used to anchor the peptide to the display can be added at a high enough level that it almost entirely eliminates inclusion of the peptide fusiazt protein in the display package. Thus, a vast majority of the display packages can be generated to include no more than one copy of the peptide fusion protein (see, for example, Garrad et al. (1991) BiolT'echnology 9:1373-13??). In a preferred ezrtbodiment of a monovalent display library, the library of display packages will comprise no more than 5 to 10% polyvalent displays, and more preferably no more than 2% of the display will be polyvalent , and most preferably, no more than 1% polyvalent display packages in the population. The source of the wild-type anchor protein can be, for example, provided by a copy of the wild-type gene present on the same construct as the peptide fusion protein, or provided by a separate construct altogether. 1"iowever, it will be equally clear that by siz~nilar manipulation, polyvalent displays can be generated to isolate a broader range of binding affinities. Such peptides can be useful, for example, in purification protocols where avidity can be desirable.
l) ,Phages As Display Packages Bacteriophage are attractive prokaryotic-related organisms for use in the subject method. Bacteriophage are exceIleztt candidates for providing a display system of the variegated polypeptide library as there is little or no enzymatic activity associated with intact mature pltage, and because their genes are inactive outside a bacterial host, rendering the zoatuz~ phage particles metabolically inert. In general, the phage surface is a relatively simple structure. Phage can be grown easily in large numbers, they are HMV-OS 3.02 aztaenable to the practical handling involved in many potential mass screening programs, and they carry genetic information for their own synthesis within a small, simple paclCage_ As the polypeptide gene is inserted into the phage genozne, choosing the appropriate phage to be employed in the subject method will generally depend most on whether (i) the genome of the phage allows introduction of tlae polypeptide gene either by tolerating additional genetic material or by having replaceable genetic material; (ix) the virion is capable of packaging the genome after accepting the insertion or substitution of genetic material; and (iii) the display of the polypeptide on the phage surface does not disrupt virion structure sufficiently to interfere with phage propagation.
One concern presented with the use o~ phage is that the morphogenetic pathway of the phage determines the environment in which the polypeptide will have opportunity to fold. Periplasmically assembled phage are preferred as the displayed polypeptides may contain essential disulfides, and such polypeptides may not fold correctly within a cell.
However, in certain embodiments in which the display package forms izttraeellularly (e.g., where ~, phage are used), it has been demonstrated in other instances that disulfide-containing polypeptides can assume proper folding after the phage is released from the cell.
Another concern related to the use of phage, but also pertinent to the use o~
bacterial cells and spores as well, is that multiple infections could generate hybrid displays that carry the gene for one particular test polypeptide yet have two or more different test polypeptides on their surfaces. Therefore, it can be preferable, though optional, to minimize this possibility by infecting cells with phage under conditions resulting in a low multiple-infection.
For a given bacteriophage, the preferred display means is a protein that is present on the phage surface (e.g. a coat protein). Filamentous phage can be described by a helical lattice; isometric phage, by an icosahedral lattice. Each monomer of each major coat protein sits on a lattice point and makes defined interactions with each of its neighbors. Proteins that fit into the lattice by making some, but not all, of the normal lattice contacts are likely to destabilize the virion by aborting formation of the virion as well as by leaving gaps in the virion so that the nucleic acid is not protected. Thus in bacteriophage, unlike the cases of bacteria and spores, it is generally important to retain in the polypeptide fusion proteins those residues of the coat protein that interact with other proteins in the virion. For example, when using the M 13 cpVrII protein, the entire mature protein will generally be retained with the polypeptide fragment being added to HMV-os3.o2 tlae N-tezz~ninus of epVl<Il, while on the other hand it can sufl'zce to xetain only the last 100 carboxy terminal residues (or even fewer) of the M 13 epIII coat protein in the polypeptide fusion protein.
Under the appropriate induction, the test polypeptide library is expressed and exported, as part of the fusion protein, to the bacterial cytoplasm, such as when the ~.
phage is employed. The induction of the fusion proteins) rnay be delayed until some replication of the phage genome, synthesis of some of the phage stzuctural-proteins, and assembly of some phage particles 'has occurred. The assembled protein chains then interact with the phage particles via the binding of the anchor protein on the outer surface of the phage particle. The cells are lysed and the phage bearing the library-encoded test polypeptides {that corresponds to the specific library sequences carried in the DNA of that phage) axe released and isolated ~~ronrt the bacterial debris.
To enrich for and isolate phage which encodes a selected test polypeptide, and thus to ultimately isolate the nucleic acid sequences (the polypeptide gene) themselves, phage harvested from the bacterial debris are af~'xnity purified. As described below, when a test polypeptide which specifically binds a particular lipid is desired, the lipid can be used to retrieve phage displaying the desired test polypeptide. The phage so obtained may then be amplified by infecting into host cells. Additional rounds of affinity enrichment followed by amplification may be employed until the desired level of enrichment is reached.
The enriched polypeptide-phage can also be screened with additional detection-techniques such as expression plaque (or colony) lift (see, e.g., Young and Davis, Science (183) 222:778-782) whereby a Labeled lipid is used as a pmbe.
a) Filameritou.r Phage Filamentous bacteriophages, which include M13, fl, fd, )Cfl, Ilce, Xf, Pfl, and Pf3, are a group of related viruses that infect bacteria. They are termed filamentous because they are long, thin particles comprised of an elongated capsule that envelopes the deoxyribonucleic acid (DNA) that forms the baeteriophage genome. The F pili filamentous bacteriophage (Ff phage) infect only gram-negative bacteria by specifically adsorbing to the tip of F pili, axed include fd, fl and M13.
Compared to othez bacteriophage, filaznentous phage in general are attractive and M13 in particular is especially attractive because: {x) the 3-l~ structure of the virion is known; (ii) the processing of the coat protein is well understood; (iii) the genome is HMV-053.02 expandable; (iv) the genome is small; (v) the sequence of the genome is kn.vwn; (vi) the virior~ is physically resistant to shear, heat, cold, urea, guanidinium chloride, low p~, and high salt; {vii) the phage is a sequez~cxng vector so that sequencing is especially easy;
(viii) antibiotic-resistance genes have been cloned into the genome with predictable results (Hines et al. (1980) Gene 11:207-21$); (ix) it is easily cultured and stored, with no unusual or expensive media requirements for the infected cells, (x) it has a higkt burst size, each iuafected cell yxeldiz~g 100 to 1000 M13 progeny after infection;
and (xi) it is easily harvested and concentrated (Salivar et al. (1964) Virology 24: 359-371 ). The entire life cycle of the filamentous phage M13, a common clozai~ag az~d sequencing vector, is well understood. The genetic structure of M13 is well knvwz~, including the complete sequence {Schaller et al. in The Single-Stranded DNA Phages eds. Denhardt et al. (NY:
CSHL Press, 1978)), the identity and function of the ten genes, and the order of transcription and location of the promoters, as well as the physical structure of the virion (Smith et al. {1985) Science 228:1315-1317; Raschad et al. (1986) Microbiol Dev 50:401-427; Kuhn et al. (1987) Science 23$:1413-1415; Zimmerman et al. {1982) JBiol Chem 257:6529-6536; and Banner et al. (1981) Nature 289;814-816). Because the geno~ane is small (6423 bp), cassette mutagenesis is practical on RF M 13 (Current Protocols in Molecular Biology, eds. ,A,usubel et al. (NX: John Wiley & Sons, 1991 )), as is single-stranded oligonucleotide dixected mutagenesis (Fritz et al. in DNA
Cloning, ed by Glover {Oxford, UK: IRC Press, 1985)). M13 is a plasmid and transformation system in itself, and an ideal sequez~ciz~g vector. M13 cau. be gown vzt Rec-stz~ains o~ E. coll.
The M13 genome is expandable (Messing et al. in 2"he Single-Stranded DNA
Phages, eds Denhardt et al. (NY: CS)EiI. Press, 1978) pages 449-453; and Fritz et al., supra) and M13 does not lyre cells. Extra genes can be inserted into M13 and will be maintained in the viral genome in a stable manner.
The mature capsule or Ff phage is comprised of a coat of eve phage-encoded gene products: cpVBl, the major coat protein product of gene 'VT>I that forms the bulk of the capsule; and four minor coat proteins, cpl~ and cpIV at one end of the capsule and cpV~I
and cpIX at the other end of the capsule. The length of the capsule is formed by 2500 to 3000 copies of cpVIII in an ordered helix array that forms the characteristic filament structure. The gene iII-encoded protein (cp)~T} is typically present in 4 to 6 copies at one end of the capsule az~d serves as the receptor for binding of the phage to its bacterial host in the initial phase of infection. For detailed reviews of Ff phage structure, see Ranched et al., ,~I~TGTOb101. Rev., 50:401-427 (1986); and Model et al.,in The .8acteriophages. Yolurne 1, R. Calendar, Ed., Plenum Press, pp. 375-456 ( 1988).

HMV-OS3.02 The phage particle assembly ixlvolves extrusion of the viral genome through the host cell's membrane. Prior to extrusion, the major coat protein cpVIII and the minor coat protein cpIll are synthesized and transported to the host cell's xnezz~braz~e.
Both cpVnl and cpflT are anchored in the host cell membrane prior to their iztcorporatiozz into the zxxature particle. In addition, tb~e viral genome is produced and coated with cpV
protein. During the extrusion process, cpV-coated genomic DNA is stripped of the cpV coat and simultaneously recoated with the mature coat proteins.
Bath cpIII and cpV3ZI proteins include two domains that provide signals fox assembly of the mature phage particle. The first domain is a Secretion Signal that directs the newly synthesized proteixi to the host cell zzae~onbrane. The secretion signal is located at the amino terminus of the polypeptide and lipids the polypeptide at least to the cell membrane. The second domain is a membrane anchor domain that provides signals for association with the host cell n aembrane and for association with the phage particle during assembly. This second signal for bath cpVI~ and cplLI comprises at least a hydrophobic region for spanning the membrane.
The 50 amino acid mature gene Vl'1I coat protein (cpVll)~ is synthesized as a amino acid precoat (,lto et al. {1979) PNAS 76:1199-1203). cpVIil has been extensively studied as a model membrane protein because it can integrate into lipid bilayers such as the cell txaembrane in an asymmetric orientation with the acidic amino terminus toward the outside and the basic carboxy terminus toward the i;aside of the membrane.
The first 23 amino acids constitute a typical signal-sequence which causes the nascent polypeptide to be inserted into the inner cell membrane. An B. coli signal peptidase {SP-17 recognizes amino acids 18, 21, and 23, and, to a lesser extent, residue 22, and cuts between residues 23 and 24 of the precoat (Kahn et al. (1985) .I. Biol. Chem. 260:15914-15918;
and Kuhzt et al. (1985) d. Biol. Chem. 2b0:15907-15913). After removal of the signal sequexace, the amino terminus of the mature coat is located on the periplasmic side of the inner mem-braxte; the carboxy terminus is on the cytoplasmic side. About 3000 copies of the mature coat protein associate side-by-side in the inner membrane.
The sequence of gene VIII is known, axxd the amino acid sequence can be encoded on a synthetic gene. Mature gene VIII protean znalces up the sheath around the circular ssDNA. The gene VL'.~ protein can be a suitable anchor protein because its location and orientation in the virion are known (Banner et al. (1981) Nature 289:81.4-816). Prefera-bly, the polypeptide is attached to the amino terminus of the mature M13 coat protein to generate the phage display library. As set out above, manipulation of the concentration of HMV-OS 3.02 both the wild-type cpVIII and AbIcpV~ fusion in an infected cell can be utilized to decrease the avidity of the display and thereby enhance the detection of high affinity polypeptides directed to the lipid(s).
Anothez vehicle for displaying the polypeptide is by expressing it as a domain of a chimeric gene containing part or all of gene III, e.g., encoding cpIII. When monovalent displays are required, expressing the polypeptide as a fizsion protein with cpIII can be a preferred embodiment, as manipulation of the ratio of wild-type cps to chimeric cps during formation of the phage particles can b$ readily controlled. This gene encodes one of the minor coat proteins of M13. Genes VI, V1I, and IX also encode minor coat proteins. Each of these minor proteins is present in about 5 copies per virion and is related to morphogenesis or infection. In contrast, the major coat protein is present in more than 2500 copies per vizion. The gene VI, VII, and IX proteins are present at the ends of the virion; these three proteins are not posttranslationally processed (Rasched et al. (1986) Ann ,Rev. Microbiol. 41:507-541). In particular, the single-stranded circular phage DNA associates with about five copies of the gene T1I protein and is then extruded through the patch of membrane-associated coat protein in such a way that the DNA is encased in a helical sheath o~ protein (Webster et al. in The Single-Stranded DNA
Phages, eds Dressier et al. (NY:CSHL Press, 1978).
Manipulation of the sequence of cpIII has demonstrated that the C-terminal 23 amino acid residue stretch of hydrophobic amino acids normally responsible for a membrane anchor function can be altered in a variety of ways and retain the capacity to associate with membranes. Ff phage-based expression vector's were first described in which the cps amino acid residue sequence was modified by insertion of heterologous polypeptide (Parmely et al., Gene (1988) 73:305-318; and Cwirla et al., PNAS
(1990) 87:637$-6382) or an amino acid residue sequence defining a single chain polypeptide domain (McCafferty et al., Science (1990) 348:552-554). It has been demonstrated that insertions into gene III can result in the prodution of novel protein domains on the virion outer surface. (Smith (19$5) Science 228:1315-1317; and de la Cruz et al.
(1988) J. Biol.
Chem. 263:4318-4322). The polypeptide gene may be fused to gene III at the site used by Smith and by de la Cruz et al., at a codon corresponding to another domain bouztdary or to a surface loop of the proteuo, or to the amino terminus of the mature protein.
Generally, the successful cloning strategy utilizing a phage coat protein, such as cpDI of filamentous phage fd, will provide expression of a polypeptide chain fused to the N-terminus of a coat protein (e.g., cpl~ and transport to the inner membrane of the host HNIV-os3.02 where the hydrophobic domain iz~ the C-terminal region of the coat pmtein anchors the fusion protein in the membrane, with the ~1-tezmxnus containing the polypeptide chain protruding into the periplasmic space.
Similar constructions could be made with other filamentous phage. Pf3 is a well known filamentous phage that infects Pseudomonos aerugenosa cells that harbor an IncP-I plasmid. The entire genome has been sequenced ((Luiten et al. (19$5) J.
Virol.
5G:268-276) and the genetic signals involved in replication and assembly are known (Luitezt et al. (19$7) DNA 6:129-137). The major coat protein of PF3 is unusual in having no signal peptide to direct its secretion. The sequence has changed residues ASP-7, ARG-37, LYS-40, and PHE44 which is consistent with the amino terminus being exposed. Thus, to cause a polypeptide to appear on the surface of Pf3, a tripartite gene can be constructed which comprises a signal sequence lsa~own to cause secretion in P.
aerugenosa, fused in-frame to a gene fragment encoding the polypeptide sequence, which is fused in-fi-ame to DNA encoding the mature Pf3 coat protein. optionally, DNA
encoding a flexible linker o~ one to 10 amino acids is introduced between the polypeptide gene fragment and the Pf3 coat-protein gene. This tripartite gene is introduced into Pf3 so that it does not interfere with expression o~any P~'3 genes. Qrtce the signal sequence is cleaved off, the polypeptide is in the periplasm and the mature coat protein acts as an anchor and phage-assembly signal.
b) Bacteriophage ø1~C174 The bacteriophage X174 is a very small icosahedral vizus which has been thoroughly studied by genetics, biochemistry, and electron microscopy (see The Single Stranded DNA ,Phages (eds. Den hardt et al. (NY:CSHL Press, 1978)). Three gene products of X174 are present on the outside of the nnature virioz~: F
(capsid), G (major spike protein, 60 copies per virion), and H (minor spike protein, 12 copies per virion).
The G protein comprises 1.75 amino acids, while H comprises 32$ amino acids.
The F
protein interacts with the single-stranded DNA of the virus. The proteins F, G, and H are translated from a single mRNA in the viral infected cells. As the virus is so tightly constrained because several of its genes overlap, X174 is not typicahy used as a cloning vector due to the fact that it cart accept very little additional DNA.
However, mutations in the viral G gene (encoding the G protein) can be rescued by a copy of the wild-type G
gene carried on a plasmid that is expressed in the same host cell (Chambers et al. (19$2) Nue Acid Res 10:f465-6473). In one embodiment, one or cnot~e stop eodons are introduced into the G gene so that no G protein is produced ~ro~oa the viral genome. The HMV-053.02 variegated polypeptide gene library can then be fused with the nucleic acid sequence of the H gene. An amount of the viral G gene equal to the size of polypeptide gene fragment is eliminated from the X174 genome, such that the size of the genome is ultimately unchanged. Thus, in host cells also transformed with a second plasmid expressing the wild-type G protein, the praductiott of viral panicles from the mutant virus is rescued by the exogenous G protein source. Where it is desirable that only acne test polypeptide be displayed per ~X 174 particle, the second plasmid caz~ fitrthex include one or more copies of the wild-type H protein gene so that a zttix of H and test polypeptide/H
proteins will be predominated by the wild-type H upon incorporation into phage particles.
c) Large DNA Phage Phage such as ~. ax 'f4 have much larger genomes than do M13 or X174, azxd have more complicated 3-D eapsid structures titan 1VI13 or ~PX174, with more coat proteins to choose &om. In embodiments a~ the invention whereby the test palypeptide library is processed az~d assembled into a fuxtctional form and associates with the bacteriophage particles within the cytoplasm of the host cell, bacteriophage ~
and derivatives thereof are examples of suitable vectors. The intracellular morphogenesis of phage ~. can potentially pz~event protean domains that ordinarily contain disulfide bonds from folding correctly. However, variegated libraries expressing a population of functional polypeptides, which include such bonds, have been generated in ~, phage.
(Hose et al. (1989) Science 246:1275-1281; Mullinax et al. {1990) PNAS 87:8095-8099;
and Pearson et al. (1991) PNAS 88:2432-2436). Such strategies take advantage of the rapid construction and eflxcient transformation abilities of ~, phage.
When used for expression of polypeptide sequences (exogenous nucleotide sequences), may be readily inserted into a ~, vector. Pox instance, variegated polypeptide libraries cans be constructed by modification of ~, ZAP TI through use of the multiple cloning site of a ~ ZAP II vector (Hose et al. supra).
ii) Bacterial Cells a~ Dirplay Packages Recombinant polypeptides are able to cross bacterial membranes after the addition of appropriate secretion signal sequences to the N-terminus of the protein (Better et al (1988) Science 240:1041-1043; and Skerra et al. (19$8) Science 240:1038-1041). In addition, recombinant polypeptides have been fused to outer membrane proteins for surface presentation. p'or example, one strategy for displaying poly~peptides on bacterial HMV-p~ 3 .02 cells comprises generating a fusion protein by inserting tl~e polypeptide into cell surface exposed portions of an integral outer membrane protein (Fucks et al. (1991) BiolZ'echnology 9:1370-1372). 1n selecting a bactez'ial cell to serve as the display package, any well-characterized bacterial strain will typically be suitable, provided the bacteria may be gown in culture, engineered to display the test polypeptide library on its surface, and is compatible with the particular affinity selection process practiced iz~ the subject method. Among bacterial cells, the preferred display systems include Salmonella typhirnurium, Bacillus subtilis, Pseudomonas aeruginosa, Yibrio cholerae, Klebsiella pneumonia, Neisseria gonorrhaeae, Neisseria meningitides, Bacteroides nodosus, .~l~oraxella bovis, and especially Escherichia coli. Many bacterial cell surface proteins useful in the present izxvez~tion have been characterized, and works on the localization of these proteins and the methods of determining their structure xz~clude Benz et al. (1988) Ann Rev Microbial 42: 359-393; Balduyck et a1. (1985) Biol Chem Hoppe-Seyler 366:9-14; Ehrmann et al {1990) PNAS 87:7574-7578; Heijne et al. {1990) Protein Engineering 4:109-112; Ladnex et al. C1.S. Patent No. 5,223,409; Ladner et al. W088/06630;
Fucks et al. (1991) Biotechnology 9:1370-1372; and Goward et a1. (1992) TIBS 18:136-140.
To further illustrate, the Lama protein of E coli is a well understood surface protein that can be used to generate a variegated library of test polypeptides on the surl'ace of a bacterial cell (see, for example, Ronco et al. (1990) Biochemfe 72:183-189;
van der Weit et al. (1990) Vaccine 8:269-277; Charabit et al. (1988) Gene 70:181-189;
and Ladner TJ.S. Patent No. 5,222,409). Lama of ~ cvli is a porin for maltose and maltodextrin transport, and serves as the receptor for adsorption of bacteriophages ~, arld K10. Lama is transpot'ted to the outer membrane if a functional N-terminal signal sequence is present (Benson et al. (1984) PNAS 81:3830-3834). As with other cell surface proteins, Lama is synthesized with a typical signal-sequence which is subsequently removed. Thus, the variegated polypeptide gene library can be cloned into the Lama gene such that the resulting library of fusion proteins comprise a portion of Lama sufficient to anchor the protein to the cell membrane with tlae test polypeptide fragnnent ozaented on the extracellular side o;f the nnembrane. Secretion of the extracellular portion of the fusion protein can be facilitated by inclusiozz of the Lama signal sequence, or othex suitable signal sequence, as the N-terminus of the protein.
The E_ coli lama has also been expressed in functional form in S. typhimurium (Harkki et al. (1987) ,Col Gen Genet 209:607-611), Y. cholerae (Harkki et al.
(1986) Microb Pathol 1:283-288), and K. pneumonia (Wehmeier et al. (1989) Mol Gen Genet z8 HMV-053.02 215:529-536), so that one could display a population of test poiypeptides in any of these species as a fusion to E. coli Lama. Moreover, K. ,pneumonia e~cpresses a maltoporin similar to Lama which could also be used. In .P_ aeruginosa, the Dl protein (a honnologue of Lama) can be used (Trios et al. (1988) Biochem Biophys Acta 938:493-496).
Similarly, other bacterial surface proteins, such as PAL, OxnpA, OmpC, OmpF, PhaB, pilin, BtuB, FepA, FhuA, IutA, FecA and FhuE, may be used in place of LanaB as a portion of the display means in a bacterial cell.
In another exemplary embodiment, the fusion protein can be derived using the k'liTrxT"s Random Polypeptide Display Library (Invitragexa). That library is a diverse population of random dodecapolypeptides inserted within the thioredoxin active-site loop inside the dispensable region of the bacterial flagellin gene (fliC). The resultaztt recombinant fusion protein (FLITR?~ is exported and assembled into partially Functional flagella on the bacterial cell surface, displaying the random palypeptide library.
Polypeptides are fused in the nn~iddle of thioredoxin, therefore, both their N-az~d C-termini are anchored by thioredoxin's tertiary structure. This results in the display of a constrained polypeptide. By contrast, phage display proteins are fused to the N-terminus of phage coat proteins in an unconstrained manxaer. The unconstrained molecules possess many degrees of cozifoxroatiaztal Freedom which may result in the lack of proper interaction with the lipid ax~olecule. Without proper interaction, many potential protein-pratein interactions may be missed.
Moreover, phage display is limited by the low expression levels of bacteriophage coat proteins. FliTrxT"t and similar methods can overcome this limitation by using a strong promoter to drive expression of the test polypeptide fusions that arre displayed as multiple copies.
According to the present invention, it is contemplated that the FliTtx vector can be modified to provide, similar to the illustrated vectors of the attached figures, a vector which is differezttially spliced in mammalian cells to yield a secreted, soluble test polypeptide.
iii) Bacterial Spores as Display Packages Bacterial spores also have desirable properties as display package candidates in the subject xxaethod. For example, spores are much more resistant than vegetative bacterial cells or phage to chemical and physical agents, and hence permit the use of a great variety 1~V-053.02 of affinity selection conditions. Also, Bacillus spores neither actively nnetabolize nor alter the proteins on their surface. However, spores have the disadvantage that the molecular mechanisms that tzagger sporulation are less well worked out than is the formation of MX3 or the export of protein to the outer membrane of E. cvli, though such a limitation is not a serious detractant from their use in the present invention.
Bacteria of the genus $acillus form endospores that are extremely resistant to damage by heat, radiation, desiccation, and toxic chemicals (reviewed by Losick et al.
{1986) Ann Rev Genet 20:625--b69). This phenomenon is attributed to extensive intermolecular cross-lxnkang of the coat proteins. In certain embodiments of the subject method, such as those which include relatively harsh affinity separation steps, Bacillus spores can be the preferred display package. Endospores from the genus Bacillus are wore stable than are, for example, exospores from Streptornyces. lv,Iareover, Bacillus subtilis forms spores in 4 to b hours, whereas Streptomyces species may require days or weeks to sporulate. In addition, genetic knowledge and manipulation is much more developed for B, subtilis than for other spore-forming bacteria.
Viable spores that dif>'er only slightly from wild-type are produced in B.
subtilis even if any one of four coat proteins is missing (Donovan et al. (1987) J,tKol Biol 196:1-10). ~areover, plasmid DNA is commonly included in spores, and plasmid encoded proteins have been observed on the surface of Bacillus spores (Debro et al.
(1986) J
Bacteriol 165:258-268). Thus, it can be possible during spozulatio~a to express a gene encoding a chimaric cast protein comprising a polypeptide of the variegated gene library, without interfering materially with spore formation.
To illustrate, several galypeptide components of B. subtilis spore coat (Danavan et al. {x9$7) J Mol Biol 196:1-10) have been characterized. The sequences of two complete coat proteins and amino-terminal fragments of two others have been determined. Fusion of the test polypeptide sequence to cotC or cotD fragments is likely to cause the polypeptide to appear on the spore surface. The genes of each of these spore coat proteins are prefez~red as neither eotC or eotD are post-translationally modified (see Ladner et al. U.S, Patent No. 5,223,409).
iv) Selecting Peptides from the .Dfsplay Mode Upon expression, the variegated polypeptide display is subjected to affinity enrichment in order to select for test poIypeptides which bind preselected lipids. The HMV-053.02 term "affinity separation" or "affrnity enrichment" includes, but is not limited to: ( 1 ) affinity chromatography utilizing immobilized lipids, and (2) precipitation using soluble lipids. In each embodiment, the libxa;ty of display packages are ultimately separated based oz~ the ability of the associated test polypeptide to bind the lipid of interest. See, for example, the Ladner et al. U.S. Patent No. 5,223,409; the l~.ang et al.
International Publication No. WO 92118619; the Dower et al. International Publication Nv. WO
91117271; the Winter et al. lntenn~atxonal Publication WO 92120791; the Markland et al.
International Publication No. WO 92/15679; the Bz~eitling et al. International Publication WO 93/01288; the MeCafferty et al. International Publication No. W4 92101047;
the Garrard et al. International Publication No. WO 92/09b90; and the Ladner et al.
International Publication No. WO 90102809. In most preferred embodiments, the display library will be pre-enriched for peptides specific for the lipid by lxrst contacting the display library with any negative controls or other lipids for which differential binding by the test polypeptide is desired. Subsequently, the non-binding fraction from that pre-treatment step is contacted with the lipid and peptides from the display which are able to specifically bind the lipid are isolated.
With respect to affinity chromatography, it will be generally understood by those skilled in the art that a great number of chromatography techniques can be adapted for use in the present invention, ranging from column chromatography to batch elution, and including ELISA and biopanning techniques. Typically, whexe lipid is or can be immobilized on an insoluble carrier, such as sepharvse or polyacrylamide beads, or, alternatively, the wells of a micrvtitre plate.
The population of display packages is applied to the affinity matrix under conditions compatible with the binding of the test polypeptide to the lipid.
The population is then fractionated by washing with a solute that does not greatly effect specilac binding of polypeptides to the lipid, but which substantially disrupts any non-specific binding of the display package to the lipid or matrix. A certain degree of control can be exerted over the biunding characteristics of the polypeptides recovered from the display library by adjusting the conditions of the binding incubation and subsequent washing.
'fhe temperature, pH, ionic strength, divalent cation concentration, and the volume and duration of the washing can select for polypeptides within a particular range of affnity aztd specificity. Selection based on slow dissociation rate, which is usually predictive of bagh a~fanity, is a very practical route. This may be done either by continued incubation in the presence of a saturating amount of free lipid (if available), or by increasing the HlvlV-053.02 volume, number, and length of the washes. Zn each case, the rebinding of dissociated polypeptide-display package is prevented, axzd with increasing time, display packages of higher and higher affinity are recovered. Moreover, additional modifications of the binding and washing procedures may be applied to find polypeptides with special characteristics. The affinities of some peptides are dependent on ionic strength or cation concentration. 'This is a useful characteristic for peptides to be used in affinity purification of various proteins when gentle conditions fox removing the protein from the peptide are required. Specific examples are polypeptides which depend on Cap for lipid binding activity and which lose or gain binding affinity in the presence of EGTA or other metal ehelating agent. Such peptides may be identified in the recozzibinant polypeptide library by a double screening technique isolating first those that bind the lipid in tlae presence of Cap, and by subsequently identifying those in this group that fail to bind in the presence of EGTA, or vice-versa.
After "washing" to remove nozi-specifically bound display packages, when desired, specifically bound display packages can be eluted by either specific desorption (using excess lipid) ox non-specific desorption (using pH, polazity reducing agents, or chaotropic agents). In preferred embodiments, the elution protocol does not kill the oxganistxi used as the display package such that the enriched population of display packages can be further amplified by reproduction. The list of potential eluants includes salts (such as those in which one of the counter ions is Na+, NH4+, Rb+, S04W, H~E04 citrate, K+, Li+, Cs+, HS04 , CO~2', Ca2+, Sr~'~, Cl-, POa2', HC03-, Mg2~', Ba2+, Br, HP0~2', or acetate), acid, heat, and, when available, soluble forms of the lipid.
Because bacteria continue to metabolize durizig the affinity separation step and are generally more susceptible to damage by harsh conditions, the choice of buffer compoziexats (especially eluates) can be more restricted when the display package is a bacteria rather than for phage or spores. Neutral solutes, such as ethanol, acetone, ether, or urea, are examples of other agents useful for eluting the bound display packages.
In preferred embodiments, afl"inity enriched display packages are iteratively amplified and subjected to further rounds of affinity separation until enrichment of the desired binding activity is detected. In certain embodiments, the specifically bound display packages, especially bacterial cells, need not be eluted per se, but rather, the rz~atrxx bound display packages can be used directly to inoculate a suitable growth media for amplification.

1~V-053.02 Where the display package is a phage particle, the fusion protein generated with tlae coat protein can interfere substantially with the subsequent amplilieation of eluted phage particles, particularly in embodiments wherein the cpIII protein is used as the display anchor. Even though present in only one of the 5-6 tail fibers, some peptide constructs because of their size and/or sequence, may cause severe defects in the infectivity of their carrier phage_ "his causes a loss of phage from the population during reinfection and amplification following each cycle of panning. In one embodiment, the peptide can be derived on the surface of the display package so as to be susceptible to proteolytic cleavage which severs the covalent linkage of at least the target binding sites of the displayed peptide from the remaining package. Eor instance, where the cpI>I coat protein of M13 is employed, such a strategy can be used to obtain infectious phage by treatment with an enzyme which cleaves between the test polypeptide portion and cp)TI
portion of a tail ibex fusion protein (e.g. such as the use of an enterokinase cleavage recognition sequence).
To further minimize problems associated with defective infectivity, DNA
prepared from the eluted phage can be transformed into host cells by electroporation or well known chemical means. The cells are cultivated for a period of tune sufficient for marker expression, and selection is applied as typically done for 1~NA, transformation.
The colonies are arnplihed, and phage harvested for a subsequent rounds) of panning.
ARer isolation of display packages which encode polypeptides having a desired binding specificity for the lipid, the test polypeptides for each of the purred display packages can be tested for biological activity in the secretion mode of the subject method.
(v) C'renerations of Polypeptide Libraries The variegated polypepdde libraries of the subject method can be generated by any of a number of methods, and, though not limited by, preferably exploit recent trends in the preparation of chemical libraries. For instance, chemical synthesis of a degenerate gene sequence can be carried out in an automatic DNA synthesizer, and the synthetic genes then ligated into an appropriate expression vector. The purpose of a degenerate set of genes is to provide, in one mixture, all of the sequences encoding the desired set of potential test sequences. The synthesis of degenerate oligonucleotides is well la~own ire the art (see for example, Narang, SA ( 1983) Tetrahedron 39:3; Itakura et al.
( 1981 ) HMV-053.02 Recombinant DNA, Proc 3rd Cleveland Sympos. Macromolecules, ed. AG Walton, Amstexdatn: Elsevier pp273-289; ltakura et al. (1984) Annu. Rev. Biochem.
53:323;
Itakura et al. (1984) Science 198:1056; Ike et al. (1983) Nucleic Acid Res. 1 X:477. Such techniques have been employed in the directed evolution of other proteins (see, fox example, Scott et al. (1990) Science 249:386-390; Roberts et al. (1992) PNAS
89:2429-2433; Devlin et al. (1990) Science 249: 404-406; Cwirla et al. (1990) PNAS 87:

6382; as well as U.S. Patents Nos. 5,223,409, 5,198,346, and 5,096,815).
As used hezein, "variegated" refers to the fact that a population of peptides is chat-actezized by having a peptide sequence which differ from one member of the library to the next. For example, in a given peptide library of n amino acids in length, the total number of different peptide sequences in the library is given by the product of y1 x y2 X~~~~ vn-1 x vn~ where each on represents the number different amino acid residues occurring at position n of the peptide. In a preferred embodiment of the present invention, the peptide display collectively produces a peptide library including at least 96 to 10~ different peptides, so that diverse peptides may be simultaneously assayed for the ability to interact with the lipid.
In one embodiment, the test polypeptide library is derived to express a combinatorial library of peptides which are not based on any known sequeztce, nor derived from cDNA. That is, the sequences of the library are largely, if not entirely, random. It will be evident that the peptides of the library may range in size from dipeptides to large proteins.
In another embodiment, tlae peptide library is derived to express a combinatorial library o~ peptides which are based at least in part on a known polypeptide sequence or a portion thereof (though preferably not a cDNA library). That is, the sequences of the library is semi-random, being derived by combinatorial mutagenesis of a known sequence{s). See, for example, Ladner et al. PCT publication WO 90/42909;
Garrard et al., PCT publication WO 92/09690; lVlarks et al. (1992) J. Biol. Chem.
267:16007-16010;
Griffths et al. (1993) EMBO J 12:725-734; Clackson et al. (1991) Nature 352:524-628;
and Baxbas et al. (1992) PNAS ~89:a457-4461. Accordingly, polypeptide(s) which are known binding partners far a lipid can be n~utagenized by standard techniques to derive a variegated library of polypeptide sequences which can fiuther be screened far agonists andlor antagonists. The purpose of screening such combinatorial peptide libraries is to generatC, for example, hoxnologs o~ known polypcptidcs which can act as either agonists HMV-053.02 or antagonists, or alternatively, possess novel activities all together. To illustrate, a ligand can be engiz~eezed by the present method to provide more efflciez~t biz~dxz~g or specibcity to a cogitate receptor, yet still retain at least a portion of an activity associated with wild-type ligand. Thus, combinatorially-derived homologs can be generated to have an increased poteztcy relative to a naturally occurring form of the protein.
Likewise, homologs can, be generated by the present approach to act as antagonists, in that they are able to mimic, for exaaxaple, birtdizxg to the lipid, yet not iztduce aray biological response, thereby inhibiting the action of authentic ligand.
Ln pzeferred embodiments, the combinatorial polypeptides are in the range of 3-100 amino acids in length, more preferably at least 5-50, and even more preferably at least 10, 13, 15, 20 or 25 amino acid residues in length. Preferably, the polypeptides of the library are of uniform length. It will be understood that the length of the combinatorial peptide does not reflect any extraneous sequences which may be present in order to facilitate expression, e.g., such as signal sequences yr invazaazxt portivzts of a fusion protein.
The harnessizig of biological systems for the generation of polypeptide diversity is now a well established technique which can be exploited to ge~aerate the peptide libraries of the subject method. The source of diversity is the combinatorial chemical synthesis of mixtures of oligonucleotides. 0ligonucleotide synthesis is a well-characterized chemistry that allows tight control of the composition of the mixtures created.
Degezterate DNA
sequences produced are subsequently placed into an appropriate genetic context for expression as polypeptxdes.
There are two principal ways in which to prepare the required degenerate mixture.
In one method, the DNAs are synthesized a base at a time. When variation is desired at a base position dictated by the genetic code a suitable mixture of nucleotides is reacted with the nascent DNA, rather than the pure nucleotide reagent of conventional polynucleotide syntheses. The second method provides more exact ca~ntral aver the amino acid variatioxx. First, trinucleotide reagents are prepared, each trinucleotide being a codon of one (and anly one) of the amino acids to be featured in the polypeptide library.
When a particular variable residue is to be synthesized, a mixture is made of the appropriate trinucleotides and reacted with the nascent DNA. Once the necessary "degenerate" DN,A is complete, it must be joined with the DNA sequences necessary to assure the expression of the polypeptide, as discussed in more detail below, and the complete DN,A, construct must be introduced into the cell.

H.IvIV-053.02 Whatever the method may be for generating diversity at the codon level, chemical synthesis of a degenerate gene sequence can be carried out xzt an automatic DNA
synthesizer, and the syz~thetac ge~aes can then. be higated into an appropriate gene for expression. The purpose of a degenerate set of genes is to provide, iz~ ox~e mixture, all Qf the sequences encoding the desired set of potential test polypolypeptide sequences. The synthesis of degenerate ohagonucleotides is well known in the art (see for example, Narang, SA (1983) Tetrahedron 39:3; Itakura et al. (1981) Recombinant DNA, .Pros 3rd Cleveland Sympos. Macromolecules, ea. AG Wahton, Amsterdam: Elsevier pp273-289;
Itakura et al. (1984) Annu. Rev. Biochem. 53:323; Itakura et aI. (1984) Science 198:1056;
lke et al. (1983) Nucleic Acid Res. 11:477. Such techniques have been employed in the directed errolution of other proteins (see, for example, Scott et al. ( 1990) Science 249:386-390; Roberts et al. (1992) PNAS 89:2429-2433; Devlin et al. (1990) Science 249: 404-406; Cwirla et al. (1990} PNAS 87: 6378-6382; as well as U.S. Patents Nos.
5,223,409, 5,198,346, and 5,096,815).
E) ExemnliRcation Given, the diversity of cellular responses dependent on the products of PI 3'-K, it is probable that many as yet unidentified proteins bind to and are regulated by PtdLns-3,4,5-P3 and Ptdins-3,4-Pi. Identification of these proteins and the elucidation of their role in cellular signaling will be critical to our understanding of these cellular functions as well as to diseases such as cancer. In order to isolate novel 3'PPI binding proteins we developed a screen using in vitro coupled transc~ptionltranslation technology, izz conjunction with the use of syaathetic biotinylated Ptdins-3,4-Ps and PtdIns-3,4,5-P3 ligands. We show in this report that this screen can isolate 3'PPI binding proteins with high specificity and selectivity ~rom among a vast excess and diversity of other non-speci~c proteins. We report an initial demonstration of the effectiveness of the systenn using the PH domain of the serinelthreonine kinase Akt, a known 3'PPI binder, as a model. Second, we demonstrate the utility of this technique in isolating other 3'PPI
binding proteins by screening a small pool expression library derived from mouse spleen mRNA. Three 3'PPI binding proteins were adenti~ed in this initial screen. Two of these proteins have been previously characterized, PfP3BPIp42~" az~d PDKI, thereby establishing positive controls that the system operates as predicted.
Izoportez~tly, the tlurd protein is a novel protein of unknown fiuactioz~ that contains both a PH
domain and an SH2 domain.

HMV~053.02 EXPERIMENTAL PROCEDURES
Cloning and Expression of Akt PH domain. The PH domain of Akt was cloned as a fusion protein to maltose binding protein (MBP) in the expression vector pMAL2B
(New England Bivlabs). The N-terminal 130 amino acids of murine Aktl in the vector pJ3? (a gift of P. Tsichlis, Fox-Chase Cancer Center) was amplified by PCR
using Pfu polymerase with the coding strand primer Si-cgatcgggatccatggaacag-3e (upstream of the myc tag in p13? and contains a BamHl site) and the z~on coding strand primer Si-ccctgaattctcactgggtga-3' (~rozn Akt amino acid 130 and encodes an EcoRl site).
Both maltose binding protein alone and the MBP - Akt PH domain fusion construct were subeloned into the in vitro translation vector pCS2(+) under the control of tl~e SP6 promoter. The proteins were expressed by transcribing/translating U.Ol-1 ~g of DNA of MBP oz MBP-Akt PH with the Promega TnT Coupled Reticulocyte Lysate System using SP6 RNA polymerase and 35S-methionine (in vivo labelizxg grade, Arnersham) according to the manufacturers protocol.
~nthesis of Biotinylated and Non-~iotinylated PhQSphatidylinositols. All chemucally synthesized probes were prepared using a tztethyl D-glueopyranoside as the chiral starting mate~al fox the inositol head group; different phosphate substitution patterns were elaborated using the Ferrier rearrangement of suitably protected glucose-derived precursors. Di-Cs PtdIns-3,4-P2 and Ptdlns-3,4,5-P3 were prepared by modifications of the synthesis o~ the dipalmitoyl analogs. Biotinylated phosphoinositides were prepared ~roxa the sn-2-aminohexanoyl derivatives of Ptdlns-3,4-P2 and Ptdlns-3,4,5-P3 by condensation of the active ester of biotin with the water-soluble PIP" analog in the presence of a mild base (Figure 1 ). The biotinylated probes, bPtdIns-3,4-Pi and bPtdTns-3,4,5-P3, were purified by ion exchange chromatography and employed as aqueous solutions fox attachment to streptavidin-coated surfaces Binding and Isolation of Radiolabeled In Vitrd Translated Proteins with .8iotinylated Phosphoinositides. Avidim beads (Ultralink immobilized Neutravidin~, Pierce Chenaacal) were washed twice with 5 volumes of wash/binding (WB) buffer (IOmM H.EPES pH=7.4, 150 mM NaCI, 0.5% NP-40, SmM DTT). The beads were then reconstituted in 2x volume of W$ buffer and the biotinylated phosphoinositide was added. Generally, 0.1 ~1 of 100 ~lvl biotinylated lipid was bound per 1 ~1 of packed avidin beads. The biotinylated lipid was incubated with the beads for 1-2 hours at 4°C with gentle agitation and then washed twice with lOx-bead volume of WB buf~'er to remove HMV-053.02 excess ligand. Control beads without biotinylated lipid were prepared in an identical tzaaz~ner but without the addition of the lipid- S u1 of the 35S-labelling reaction containing the in vitro trattscribed/translated protein was then added to the tubes and the protein was allowed to bind for 2 hours at 4°C with gentle agitation. The tubes were then centrifuged briefly, the beads were washed 2x with 0.5 txtl W B buffer, and the bound proteins were eluted by boiling the beads in 20 ~1 Laemmli sample buffer containing 5% 2-mercaptoethanol. The proteins were then separated on a x 3.75% SDS-PAGE gel.
Following electrophoresis the gel was soaked in EztHance (Dupoztt/NEN) For 1 hour and then izt a 7.5% wlv solution of PEG-3350 for 1 hour. The gel was then dried and subjected to autoradiography.
Competition Binding of Isolated Proteins for Non-biotinylated Lipids over Biotinylated Lipidr. Competition binding experiments were performed exactly as described above for standard binding of radiolabeled proteins to biotinylated lipids except that avidin beads which had been pre-bound with the biotinylated dice Ptdlns-3,4,5-P3 or Ptdlns-3,4-P2 were incubated with the in vitro tralaslated proteins in the presence of 1 pM
to 100~.~M of non-biotinylated lipids (diC8 forms of PtdIns-3,4,5-P3, Ptdlns-3,4-PZ, PtdIns-4,5-PZ [Echelon Research Laboratories, Salt Lake City, UtaltJ). Binding was quantitated by measuring the intensities of bands from the autoradiogram or by scintillation counting of the eluted proteins in Laemrnli sample buffer usiatg Opti-fluor (Packard).
Binding curves were modeled by the equation:
bound[ICsol(ICso + C)] x x 00 where % bound represents the quantity of protein bauud to the biotinylated lipid coated beads in the presence of a concentration C of competing lipid. ICSO is the molar excess of competitor C required to reduce the % bound to 50% of its maximal value in the absence of competitor.
In Vitro Translation F.~rpression Cloning. A marine spleen cDNA library containing 2x105 independent clones was divided into 1700 individual small pools. The eDNAs in this library had beezt clotted into the in vitro transcription vector pCS2(+) under control of the SP6 promoter. Individual cDNAs from positive pools were isolated from the other cDNAs of the pool using a 9fi-well format as described pzeviously. The individual cDNAs were sequenced (Harvard Biopolymer Facility) and compared with known sequences by searchixtg GenBank databases.
Northern Blotting. Northeztt blotting was performed as described in Current HMV-053.02 Protocols in Ivlolecular Biology ( 1999). A. 32P-labeled double stranded DNA
probe was made a PCR fragment &oxn the respective gene using random prirraed oligonucleotide synthesis (Prime-a-Gene, Proz~c~ega). Murine tissue RN,A was a gift of Dr. W.
Swat (Harvard Medical School).
RESULTS AND DISCUSSION
Biotinylated forms of Ptdlns-3,4-P~ and .F'tdlns-3,4,5-P3 specifcally bind to the in vitro translated PF~I domain of Akt and can be used for amity isolation. In order to test whether cloning of phvsphonnositide binding proteins through coupled in vitro transcription/translation expression was feasible, we examined whether biotinylated forms of 3'PPIs could be used to affinity isolate a known 3'PPI binding domain that had been prnduced by a coupled in vitro transcription/translation system. We first pre-bound avidin-coated beads with diC~-analogs of Ptdlns-3,4,5-P3 and Ptdlns-3,4-PZ
that had been biotinylated on the co-end of the sn-1-aminohexanoyl derivatives (see Materials and Methods and Figure 1). The beads were then incubated with 35S-methionine-labeled prnteins generated by in vitro transcription/translation of cDNAs encoding either maltose binding pmtein (MBP) or MBP fused to the PH domain of Akt. Both genes were cloned into the in vitro transcription vector pCS2(+) under the control of the SP6 promoter. The PH domain of Akt is known to bind 3'PPIs witb~ b~iglZ affinity and served as a positive control to determine optimal binding conditions and specificity. As shown in Figure 2A, the fusion pzvtein of MBP-Akt PH bound to avidin beads that were pre-bound with the biotinylated forms of either PtdIns-3,4,5-P~ ox Ptdlns-3,4-Pz. The MBP-PH
fusion protein failed to bind to the unmodified avidin beads, and MBP failed to bind to either the avidin beads alone or to the beads pre-bound with the biotinylated phosphoinositides.
These data demonstrate that the biotinylated 3'PPIs can effectively and specifically isolate an in vitro translated form of a phosphoinositide-binding domain.
The goal of this study is to isolate novel 3'PPI binding proteins from among pools of in vitro transeribedltranslated proteins from a small pool expression library. In order to specifically isolate 3'PPI binding proteins fxom among other proteins expressed in the library it is necessary that the system used for screening have a high specificity for 3'PPI
binding proteins and a low background affinity for non-specific binding proteins. The system must also be su~ci,ently sensitive to detect small amounts of a 3'PPI
binding protein in a large background of non-specific binding pxoteiuas. To determine if our system conformed to these criteria we iunvestigated whether biotinylated PtdIns-3,4,5-P3 HNIV-os3.oz azxd Ptdlras-3,4-Pz could specifically isolate the ~P-Akt PH domain fusion protein after having been diluted into a pool from the in vitro translation library.
There are approximately 100 iundependent clones in each pool of cDNA from vur pCS2znouse spleen cDNA library and the total DNA cozzcentration was approximately 1 ~glNl for each pool. Therefore 1.0 ng of the plasmid encoding Iv~P-Akt PH
fusion protein was mixed with 1 ug of DNA of a random pool from the library iz~ order to approximate the amount of 3'PPI binding protein that would be found in a random pool under the conditions of our scxeezt. As is shown in Figure 2B, biotiz~ylated PtdIns-3,4-PZ-beads captured the MBP-Akt PH fusion protein from among the other proteins i»
the pool. However, some proteins in the library, e.g. the 25 kDa protein, bound to the avidin beads in both the presence and absence of lxgand, most likely through a non-specific hydrophobic interaction. These results provided the proof of concept that this approach is feasible for isolation of 3'PPI binding domains.
Fx~nally we examined whether our binding conditions allowed the PH domain of Akt to distinguish between phosphoinositides phosphorylated at the 3' position from phosphoinositides lacking a phosphate at the 3' position. We performed competition binding experiments usi»g unbiotinylated forms of PtdIns-3,4,5-P3, PtdIns-3,4-Ps, and PtdIns-4,5-PZ to compete for biotinylated PtdIns-3,4,5-P3 binding to the MBP-Akt PH
fusion protein (Figure 2C). Ptdlns-3, 4,5-P3 and PtdIns-3,4-PZ displaced the biotin~ylated PtdIns-3,4,5-P3 at concentrations of 1000 to10,00o-fold lower than Ptdlns-4,5-P2 showing that the specificity of the Alst PH domain for the 3' phosphate is preserved under our conditions. The data also shows that PtdIns-3,4,5-P3 binds more tightly to the Akt PH
domain than does PtdTns-3,4-PZ and this is consistexat with two published studies on the a~zaity of the PH domain of Akt for phosphorylated phosphoinositides which rank the order of affinities as Ptdlns-3,4,5-P3~Ptdlns-3,4-P2»Ptdlns-4,5-P2. This result demonstrates that our system preserves the specificity of protein binding to different species of phosphorylated phosphoinositides.
Isolation of the murine isoforms of PDKI and PIP3BPlp42~p' by small pool expression cloning. After an initial screening of 500 pools of the library using the biotinylated phosphoinositides, we isolated three 3'PPT binding proteins. We obserrred a protein of approximately 25 kDa in a single pool which. appeared to bind exclusively to biotinylated PtdIns-3,4,5-P3 (Figure 3A, top panel left). When the cDNA for the protein was isolated from the other cpNAs in the pool, the expressed purified protein was also found to bind to PtdIns-3,4-Pz but with an apparent lower affinity than for PtdIns-3,4,s-P3 HMV-053.02 (Figure 3A, top panel right). Sequencing of the cloned eL7NA revealed that it was identical to the C-terminal 319 amino acids of murine phosphoinositide dependent kinase 1 (P1~K1). PD1C1 is a ubiquitously expressed 559 amino acid (65 kDa) protein which contains an N-terminal serine/threonine kinase domain and a C-terminal PH
domain. In agreement with our results, the PH domain of PDKl has recently been found to bind with a four- fold higher affinity to Ptdlns-3,4,5-P3 than to 1'tdIns-3,4-PZ but with much lower affinity to PtdIns-4,5-PZ. The fragment of PDIC1 that we isolated contains the entire C-terminal PH domain and approximately half of the serine/threonine kinase domain (Figure 3A, bottom panel). However, the f~Zrst 230 nucleotides of the isolated rnKNA are not the sequence of PDKI and the first in-frame AUG codon appears at the end of the kinase domain such that axaly the entire PH domain of PDK1 is translated. The initial 230 non-coding nucleotides could either be due to an alternative splicing of PDKI, or a~n artifact from the construction of the library.
Anothex 3'PPl binding protein identified in our screen was a prntein of approximately 25 kDa that bound tightly to both biotinylated PtdIns-3,4-Pz and Ptdlns-3,4,5-P3 but also had same residual biutading to the avidin beads (Figure 3B, top panel).
Upon isolation and sequencing of the cDNA frarn the pool, the gene was found to encode the C-terminal 210 amino acids of a protein which has >95% amino acid homology to 42 kDa inositol-1,3,4,5-tetrakisphosphate binding proteins isolated from both porcine brain called p42~4, and bovine brain called PIP3BP. PiP3BP/p42~4 is a protein of 374 amino acids which contains an N-terminal zinc-f~ngcr domain which has homology to GTPase activating proteins For the ARF family of small G-proteins, and two tandem PH
domains at the C-terminal end of the protein (Figure 3B, bottom panel). The fragment of the gene that we isolated contains the entire C-terminal PH domain and approximately half of the N-terminal PH domain but contains none of the putative A'RF-GAP domain. The C-tcnninal PH domain ca~atanns a consensus sequence fax high affinity binding of 3'PPIs.
The N-terminal PH domain does not contain this sequence but two studies have shown that it displays some specificity far 3'PPIs.
The function of PIP3BP/p42~° is currently unknown although it is highly expressed in the brain and is thought to play a role in vesicle and membrane transport because of the putative ARF'-GAP domain. Interestingly, a closely related yeast protein called Gcsl has been shown to be an ARF-GAP and is important in proper cytoskeletal organization and actin polymerization in yeast. A recent study has also shown that PIP3BP is localized to the nucleus but translocates to the plasma membrane upon H~J-053.02 activation of PI 3'-~; however, the functional significance of both the nuclear localization and the PI 3'-K dependent translocation is unknown.
Isolation of 1',HISH, a Novel 3 PPI Binddng Protein Containing both ~H and SH2 Domains. We isolated a novel 3'PPI binding protein frarn the library that migrated at approximately 30 kDa and, siznilax to PIP3BP/p42'~4, bound tightly to both biotinylated PtdIns-3,4-Pi and I'tdlns-3,4,5-P3 but not to the avidin beads alone (Figure 4A). In competition binding studies the in vitro transcribedltranslated protein was found to bind with equal affinity to both Ptdlns-3,4,5-P3 and Ptdlns-3,4-P2 but with significantly lower affinity to PtdIns-4,5-P2, thus demonstrating the specificity of the protein for 3'PPIs (Figure 4B).
The genc fragment containing the sequence for the protein was approximately 1.3 kb in length and encoded an open reading fraz~a~e from the beginning of the fragment to a step codon at nucleotide position 927 (Figure 5). A putative ICozak initiator (ATG) codon for methionine is present at position 87 and initiation of translation from this codon is consistent with the observed the size of the translated protein (280 amino acids and 30 kDa). ~Iowevex, since the sequence upstream of this ATG did not contain any in-frame stop codons it was difficult to determine if the isolated gene fragment encoded the entire coding sequence of the gene or if more coding sequences existed upstream.
The amino acid sequence contains coding regions for both an SH2 domain and a C-terminal PH domain. The SH2 domain, which encompasses nucleotides 200 to 470, is most similar to the SH2 domain of the neural adaptor pt~otein Nck (35%
identical, 59%
homologous at the amino acid level). The PH domain, which encompasses nucleotides 590 to 840, is most similar to the PH domain of Akt (39% identical, fi3%
homologous at the amino acid level) and contains the consensus sequence for high affinity biz~di~ng of 3'PPIs. There is also a tyrosine (Y139) located between the SH2 and PH domains which could be phosphorylated in stimulated cells since the sequence surrounding this tyrosine is a putative consensus motif for phosphorylation by tyrosine kinases. 'fhe existence of a putative phosphotyrosine bi~odi~ug SH2 domain, a 3'PPI binding PH domain, and a sequence for phospharylatxoz~ of tyrosine. strongly suggest that this protein plays a rate in cellular signaling. We have nazxaed this protein PHISI~ for 13' Phosphoinositide Interacting SH2-Containing proteini.
In order to determine the tissue distribution of PHISH we performed Northern blots on total RNA from several marine tissues, spleen, brain, heart, lung, thymus, and lymph using a probe derived from nucleotides 87 to 927, the putative coding region of the HMV-053.02 protein. TwQ R~tA species that are approximately 1.2 to 1.4 kb were detected.
These could represent products of alternative spliciztg of the gene. The larger of the two transcripts was detected in all tissues, and the smaller of the two transcripts was highly expressed in spleen and at lower levels in both heart and lung tissue (Figure 6). In vitro transcription of the cDNA for PHISH isolated from the expression library also produced two transcripts that were approximately 1.2 -1.4 kb izx size. The major transcript is very close in size to the larger of the two transcripts present iz~ all marine tissues tested, while the minor product is close in size to the smaller transcript present in heart, lung, card spleen. This data suggests that the gene fragment isolated from the expression cloning library encodes the complete sequence of the gene.
Upon searching the human EST database with the nucleotide sequence of PHISH
we found that PHISH had an 87% rxucleotide sequence identity with that of a human EST
derived from stem cells (locus AF150266)_ This EST encodes a cDNA, that is 1.4 kb long and contains entire coding region of PHISH. The sequences of PHISH and the human EST differ significantly outside of the protein coding sequence. Moreover, three colons upstream from the putative start colon in the human EST are in frame stop colons. The high degree of sequence homology between PHISH and the human EST implies that they encode species-specific homologues of the same protein and that the gene for PHISH
isolated from the expression screen encodes the entire protein.
Since PHISH contains only a PH domain and an SH2 domain, it is possible that PHISH functions as an adaptor protein. The PH doraain could dock PHISH to 3'PPI
generated at membrane receptor complexes and the SH2 domain could recruit phosphotymsine containing proteins) to such complexes. It is interesting to note that Skolnik and coworkers have isolated the PH domain of EST684797 from a homology search of human ESTs for PH domains that would be predicted to bind tightly to 3'PPIs.
They have also shown that the PH domain of ESTb$4797, which is highly homologous to the PH dozraain of PHISH binds tightly and selectively to Ptdlns-3,4-Pz and Ptdlns-3,4,5-P3 but not to Ptdlns-4,5-Pi.
These studies demonstrate that coupled in vitro transcriptiom/translation libraries can be used in conjunction with a#Enity isolation technology using synthetic phosphoinositides to isolate 3'PPI binding domains. Recently, several other methods have been described to isolate and clone 3'PPX binding proteins_ Qne example is the use of resins coupled to phosphorylated inositol phosphates such as IP3, or inositol phosphates linked to a glycerol moiety, to extract proteins from tissue extracts. Using HIV-053.02 these techniques PIP3BP and p42~° were extracted from total brain extracts. However, proteins expressed iz~ Iow abundance are difficult to isolate by this procedure az~d furtHezzz~ore, cDhTA cloz~iUag z~equxres zz~ultiple steps following affinity isolation.
Pxpression cloning of genes from cDNA Iibraries can circumvent some of the limitations of protein affinity isolation techniques. Two expression cloning techni9ues have been recently developed to identify 3'PPI binding proteins. The first involved the screening of a ~.gtl l library with a crude mixture of brain phospholipids that had been phosphorylated in vitro by PI 3'-K. Screening of two marine derived cDNA
libraries (from brain and adipocytes) yielded only oz~e protean that bound tightly to Ptdlns-3,4,5-P3.
This protein, called Grpl (General Receptor for Phosphoinositides), contained a PH
domain and an ARF-GEF domain that catalyzes 3'PPI dependent nucleotide exchange on mammalian AItFs. However, no other 3'PPI dependent binding protein was isolated izt the screen possibly due to improper folding of the proteins.
Another recent expression cloning strategy for isolating 3'PPI binding proteins is the use of a modified yeast two-hybrid system. In this system, genes f~Qrn a mammalian eDNA libzary were fused to a constitutively active Ras. The fusion proteins were then tested for their ability to rescue a temperature sensitive phenotype, due to a defect upstream of Ras, when coexpressed with constitutively active PZ 3'-~,. This system worked well in experiments where constitutively active Ras was fused to PH
domains already lrnown to bind to 3'PPIs. However, when tested with an expression library in yeast only Akty was isolated.
The screen described here is the first to isolate multiple 3'PPI binding targets from an expression library, including a novel protein. The basis for the better effciency of this screen may stem from the fact that in vitro translated proteins are more likely to be properly folded. bo, addition, many internal initiation sites for translation allow for the independent and unoccluded expression of PH and other 3'PPI binding domains.
We also used analogs of PtdIns-3,4-PZ and PtdIns-3,4,5-P3 that are synthetically pure and, as a result of modification in a distal zegiota of the acyl chain, closely resemble the phosphorylated phosphatidylinositols that occur naturally. It has beezt shown that although the main binding of tlae protein is to the inositoI head grnup, the contribution of the glycerol chain and the fatty acid side chains of the phosphatidylinositol are essential far specificity and tight bindiztg to the protein. Our screening technique is not without drawbacks. For example, there can be non-specific binding of proteins to the avidin-coated beads and this non-speciftc binding may obscure the biutding of other 3'PPI

binding proteins with the same electrophoretic mobility. In addition, we isolated only C-terminal PH domains, possibly because our library was made using oligo-dT to prime cDbI,A synthesis from the 3' poly-A tail of mRNAs. In addition, our screen thus far has isolated only strong binders of 3'PPIs indicating that our bindxx~g conditions may be too striz~gez~t to accommodate weaker binding proteins.
In summary, we have described a novel and effective way for isolating and cloning 3'PPl binding proteins from expression libraries. The technique described here has broad applications for the isolation of binding partners for other phosphoinositide polyphosphates or other lipid products.

Claims (16)

1. A method for identifying a cellular component which binds to a lipid moiety comprising:
a- providing a lipid bait moiety being derivatized to a solid support;
b. contacting the lipid bait moiety with a library of cellular components;
c. identifying those members of the cellular component library which specifically bind to the lipid bait moiety.
2. The method of claim 1, wherein the lipid bait moiety is a phospholipid.
3. The method of claim 2, wherein the lipid bait moiety is a phospholipid selected from the group consisting of of phosphatidylethanolamines, phosphatidylcholines, phosphatidylserines, phosphatidylglycerols, phosphatidylinositols, polyphosphatidylinositols, and diphosphatidylglycerols.
4. The method of claim 2, wherein the phospholipid is derivatized to the solid support through a cross-linking moiety which is covalently attached to a phosphate head group of the phospholipid.
5. The method of claim 1, wherein the lipid bait moiety a plasmalogen or a sphingolipid.
6. The method of claim 1, wherein the library of cellular components is a polypeptide library.
7. The method of claim 6, wherein the polypeptide library is an expression library.
8. The method of claim 7, wherein the polypeptide library is derived from replicable genetic display packages.
9. The method of claim 5, wherein the protein library is a cell lysate or partially purified protein preparation.
10. The method of claim 1, 6 or 9, wherein the identity of those members of the cellular component library which specifically bind to the lipid bait moiety is determined by mass spectroscopy
11. A drug screening assay comprising:
a. providing a reaction mixture including a cellular component identified in claim 1 as able to specifically bind to the lipid bait moiety;
b. contacting the cellular component with a test compound;

c. determining if the test compound binds to the cellular component.
12. The method of claim 11, wherein the test compound which is identified as able to bind to the cellular component is further tested for the ability to inhibit or mimic the activity of a lipid moiety.
13. The method of claim 11, wherein the reaction mixture is a whole cell.
14. The method of claim 11, wherein the reaction mixture is a cell lysate or purified protein composition.
15. A method of conducting a drug discovery business comprising:
a. providing a lipid bait moiety being derivatized to a solid support;
b. contacting the lipid bait moiety with a library of cellular components;
c. identifying those members of the cellular component library which specifically bind to the lipid bait moiety.
d. providing a reaction mixture including a cellular component identified in step (c) as able to specifically bind to the lipid bait moiety;
e. contacting the cellular component with a test compound;
f. determining if the test compound binds to the cellular component;
g. further testing those test compound identified in step (f) as able to bind to the cellular component for the ability to inhibit or mimic the activity of a lipid moiety; and h. formulating a pharmaceutical preparation including one or more compounds identified in step (g) as able to inhibit or mimic the activity of a lipid moiety.
16. A method of conducting a target discovery business comprising:
a. providing a lipid bait moiety being derivatized to a solid support;
b. contacting the lipid bait moiety with a library of cellular components;
c. identifying these members of the cellular component library which specifically bind to the lipid bait moiety.
d. licensing, to a third party, the rights for drug development for a cellular component identified in step (c) as able to specifically bind to the lipid bait moiety.
CA 2329858 2000-12-11 2000-12-28 Expression cloning of protein targets for phospholipids Abandoned CA2329858A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US73506500A 2000-12-11 2000-12-11
US09/735,065 2000-12-11

Publications (1)

Publication Number Publication Date
CA2329858A1 true CA2329858A1 (en) 2002-06-11

Family

ID=24954213

Family Applications (1)

Application Number Title Priority Date Filing Date
CA 2329858 Abandoned CA2329858A1 (en) 2000-12-11 2000-12-28 Expression cloning of protein targets for phospholipids

Country Status (1)

Country Link
CA (1) CA2329858A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110279853A (en) * 2012-01-31 2019-09-27 库瑞瓦格股份公司 Pharmaceutical composition comprising polymeric carrier cargo complex and at least one albumen or peptide antigen

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110279853A (en) * 2012-01-31 2019-09-27 库瑞瓦格股份公司 Pharmaceutical composition comprising polymeric carrier cargo complex and at least one albumen or peptide antigen
US11690910B2 (en) 2012-01-31 2023-07-04 CureVac SE Pharmaceutical composition comprising a polymeric carrier cargo complex and at least one protein or peptide antigen

Similar Documents

Publication Publication Date Title
Cassim et al. Plant lipids: Key players of plasma membrane organization and function
US5733731A (en) Peptide library and screening method
US5338665A (en) Peptide library and screening method
AU711141B2 (en) Polypeptides having a functional domain of interest and methods of identifying and using same
Rye et al. Phosphate isosteres in medicinal chemistry
AU761648B2 (en) Methods and reagents for isolating biologically active peptides
US8304211B2 (en) Methods of screening molecular libraries and active molecules identified thereby
Melia et al. Enhancement of phototransduction protein interactions by lipid surfaces
EP0859839A1 (en) ACTIVATION OF p53 PROTEIN
Rapoza et al. The products of gene I and the overlapping in-frame gene XI are required for filamentous phage assembly
Haigh et al. The major coat protein of filamentous bacteriophage f1 specifically pairs in the bacterial cytoplasmic membrane
Groth et al. New results about structure, function and regulation of the chloroplast ATP synthase (CF0CF1)
CA2329858A1 (en) Expression cloning of protein targets for phospholipids
US20020037531A1 (en) Expression cloning of protein targets for phospholipids
US20030194749A1 (en) Wortmannin derivatives as probes of cellular proteins and processes
Caradonna et al. The Nature of Enzymes Involved in Uracil-DNA Repair: Isoform Charac-teristics of Proteins Responsible for Nuclear and Mitochondrial Genomic Integrity 1
FUJII et al. Trans-bilayer movement of added phosphatidylcholine and lysophosphatidylcholine species with various acyl chain lengths in plasma membrane of intact human erythrocytes
JP2001161387A (en) Catalytic domain of human effector cell cycle checkpoint protein kinase chk1, material and methods for identification of inhibitor thereof
Lamers et al. Biochemical studies on the sarcolemmal function in the hypertrophied rabbit heart
US20020098493A1 (en) Drug-amino acids chimeric molecules
AU774332B2 (en) Methods and reagents for identifying synthetic genetic elements
WO2002046213A2 (en) Methods and reagents for isolating angiogenic modulators
KELLEY et al. Phosphorylation of vertebrate smooth muscle and nonmuscle myosin heavy chains in vitro and in intact cells
JP2005095178A (en) Lipid cleavage enzyme
Barbot Characterization of inner mitochondrial membrane architecture

Legal Events

Date Code Title Description
FZDE Dead