WO2002031512A2 - Molecular interactions in hematopoietic cells - Google Patents

Molecular interactions in hematopoietic cells Download PDF

Info

Publication number
WO2002031512A2
WO2002031512A2 PCT/US2001/032150 US0132150W WO0231512A2 WO 2002031512 A2 WO2002031512 A2 WO 2002031512A2 US 0132150 W US0132150 W US 0132150W WO 0231512 A2 WO0231512 A2 WO 0231512A2
Authority
WO
WIPO (PCT)
Prior art keywords
pdz
binding
ligand
pdz domain
polypeptide
Prior art date
Application number
PCT/US2001/032150
Other languages
French (fr)
Other versions
WO2002031512A3 (en
Inventor
Joshua D. Rabinowitz
Peter S. Lu
Johannes Schweizer
Original Assignee
Arbor Vita Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/688,017 external-priority patent/US6942981B1/en
Application filed by Arbor Vita Corporation filed Critical Arbor Vita Corporation
Priority to AU2002215354A priority Critical patent/AU2002215354A1/en
Publication of WO2002031512A2 publication Critical patent/WO2002031512A2/en
Publication of WO2002031512A3 publication Critical patent/WO2002031512A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70571Assays involving receptors, cell surface antigens or cell surface determinants for neuromediators, e.g. serotonin receptor, dopamine receptor

Definitions

  • the present invention relates to peptides and peptide analogues, and methods for using such compositions to regulate activities of cells of the hematopoietic system.
  • the invention provides methods of modulating metabolism (e.g., activation) of hematopoietic cells (e.g., T cells and B cells) by antagonizing an interaction between a PDZ domain containing protein and a protein that binds a PDZ domain.
  • fusion peptides containing an amino acid sequence corresponding to the carboxyl terminus of a surface receptor expressed by a hematopoietic cell and a transmembrane transporter sequence; such fusion peptides are useful in regulating hematopoietic cells by inhibiting cell activation.
  • PDZ domains of proteins are named after three prototypical proteins: PSD95, Drosophila large disc protein and Zonula Occludin 1 protein (Gomperts et al., 1996, Cell 84:659- 662).
  • PDZ domain-containing proteins are involved in synapse formation by organizing transmembrane neurotransmitter receptors through intracellular interactions.
  • PDZ domains contain the signature sequence GLGF.
  • typical PDZ domain-containing proteins contain three PDZ domains, one SH3 domain and one guanylate kinase domain. Examples of intracellular PDZ domain-containing proteins include LIN-2, LIN-7 and LIN- 10 at the pre-synapse, and PSD95 at the post-synapse.
  • PDZ domains have been shown to bind the carboxyl termini of transmembrane proteins in neuronal cells.
  • Songyang et al. reported that proteins capable of binding PDZ domains contain a carboxyl terminal motif sequence of E-S/T-X-V/I (Songyang et al., 1997, Science 275:73).
  • X-ray crystallography studies have revealed the contact points between the motif sequence and PDZ domains (Doyle et al., 1996, Cell 88:1067-1076). While the interaction between PDZ domains and ion channels in neurons have been studied extensively, such interactions have had limited studies in other biological systems, especially the hematopoietic system.
  • the hematopoietic system is composed of different cell types that perform distinct functions. Many of its diverse function requires coordinated movement of cell surface receptors including ion channels, adhesion surface molecules to coordinate cell-cell interaction, and cytokine receptors. Despite their diverse functional activities, all hematopoietic cells are believed to develop from a multipotent bone marrow hematopoietic stem cell. Such stem cell has been shown to express a surface marker termed CD34. During differentiation, the stem cell gives rise to progenitor cells in each of several specific hematopoietic cell lineages. The progenitor cells then undergo a series of morphological and functional changes to produce mature functionally committed hematopoietic cells.
  • lymphocytes which include T cells, B cells and natural killer (N ) cells
  • monocytes and granulocytes i.e., neutrophils, basophils and eosinophils
  • Lymphocytes, monocytes and granulocytes are collectively referred to as white blood cells or leukocytes.
  • other hematopoietic cells perform functions that are unrelated to the immune system. For example, erythrocytes are involved in gas transport, and cells of the thrombocytic series are involved in blood clotting.
  • T cells and B cells recognize antigens and generate an immune response.
  • T cells recognize antigens by heterodimeric surface receptors termed the T cell receptor (TCR).
  • TCR T cell receptor
  • the TCR is associated with a series of polypeptides collectively referred to as CD3 complex.
  • B cells recognize antigens by surface immunoglobulins (Ig), which are also secretory molecules.
  • Ig surface immunoglobulins
  • co-stimulatory surface receptors have been identified in T cells and B cells, which augment cellular activation during antigen-induced activation.
  • T cell antigen receptor/CD3 complex TCR/CD3
  • other molecules expressed by T cells which mediate an activation signal include but are not limited to, CD2, CD4, CD5, CD6, CD8, CD18, CD27, CD28, CD43, CD45, CD152 (CTLA-4), CD154, MHC class I, MHC class II, CDwl37 (4-1 BB), CDwl 50, and the like (Barclay et al., The Leucocyte . ⁇ ntigen Facts Book, 1997, Second edition, Academic Press; Leucocyte Typing, 1984,
  • T cell surface antigens include antigen-binding antibody derivatives such as variable domains, peptides, superantigens, and their natural ligands such as CD58 (LFA-3) for CD2, HTV gpl20 for CD4, CD27L for CD27,
  • Activation molecules expressed by B cells include but are not limited to, surface Ig, CD18, CD19, CD20, CD21, CD22, CD23, CD40, CD45, CD80, CD86 and ICAMl.
  • natural ligands of these molecules and antibodies directed to them as well as antibody derivatives may be used to deliver an activation signal to B cells.
  • the invention provides a method of modulating a biological function of a cell, e.g., an endothelial cell or hematopoietic cell (such as a a leukocyte, e.g., T cell or B cell), by introducing into the cell an antagonist that inhibits binding of a PDZ protein and a PL protein in the cell, or a agonist that enhances binding of a PDZ protein and a PL protein in the cell.
  • the PL protein is an adhesion protein, an adaptor protein, or an intracellular protein.
  • the PL protein is CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD4, CD97, CD3n, DOCK2, CD34, FceRIb, or FasLigand.
  • the PL protein is characterized by a carboxy-terminal amino acid motif that is X-S-X-A, X-A-D/E-V, X-V/I/L-X*-V, or X-S/T-X-F (where X is any amino acid and X* is any non-aromatic amino acid).
  • the PL protein is expressed by T lymphocytes or B lymphocytes.
  • the PDZ protein is CASK, MPP1 , DLG1 , PSD95, NeDLG, SYNl a, TAX43, LDP, LIM, LIMK, AF6, PTN-4, prILl ⁇ , 41.8, GS12, DVL1, TAX 40, TIAM1, MINT1, K303, TAX2, or KIAA561.
  • the cell is a leukocyte and the biological function is cell activation, cell proliferation, maintenance of cell structure, cell metabolic activity, or cytokine production.
  • the method further includes detecting a change in leukocyte activation.
  • the antagonist is an agent that inhibits the binding of a PL peptide to a PDZ domain polypeptide in an "A" assay, in a "G” assay, or in both an A assay and a G assay.
  • the antagonist can be a polypeptide, such as a polypeptide having at the carboxyterminus at least two residues that are the same as the carboxy-terminal two residues of a PL protein, such as a PL protein is expressed in a hematopoietic or endothelial cell that is an adhesion protein, an adaptor protein, or an intracellular protein.
  • the carboxy-terminal four residues of the polypeptide are the same as the carboxy- terminal four residues of the PL protein.
  • the PL protein has a carboxy- terminal amino acid motif selected from X-S-X-A, X-A-D/E-V, X-V/I/L-X*-V, or X-S/T-X-F, where X is any amino acid and X* is any non-aromatic amino acid.
  • the PL protein is CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD97, CD3n, DOCK2, CD34, FceRIb, or FasLigand.
  • the antagonist is a peptide mimetic of a PL inhibitor sequence peptide.
  • the antagonist is a fusion polypeptide having a PL sequence and transmembrane transporter amino acid sequence (such as HIV tat, Drosophila antenapedia, herpes simplex virus VP22 or anti-DNA CDR 2 and 3).
  • the invention provides a method of determining whether a test compound is an inhibitor of binding between a PDZ protein and a PL protein by contacting a PDZ domain polypeptide having a sequence from the PDZ protein, and a PL peptide under conditions in which they form a complex, in the presence and in the absence of a test compound, and detecting the formation of the complex in the presence and absence of the test compound, where less complex formation in the presence of the test compound than in the absence of the compound indicates that the test compound is an inhibitor of a PDZ protein -PL protein binding.
  • the PL peptide has a sequence that includes the a C-terminal sequence of a PL protein, such as CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD97, CD3n, DOCK2, CD34, FceRIb, or FasLigand.
  • a PL protein such as CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD97, CD3n, DOCK2, CD34, FceRIb, or FasLigand.
  • the PDZ domain polypeptide is a fusion polypeptide.
  • the invention provides a method of determining whether a test compound is an agonist of binding between a PDZ protein and a PL protein by contacting a PDZ domain polypeptide, and a PL peptide under conditions in which they form a complex, in the presence and in the absence of a test compound, and detecting the formation ofthe complex in the presence and absence ofthe test compound, where more complex formation in the presence o the test compound than in the absence ofthe compound indicates that the test compound is an agonist of a PDZ protein -PL protein binding.
  • the invention further provides an inhibitor of binding of a PDZ protein and a PL protein.
  • the inhibitor is characterized in that it reduces binding of a peptide selected from the group consisting of a PL peptide selected from the group consisting of CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD97, CD3n, DOCK2, CD34, FceRIb, and FasLigand and a PDZ domain polypeptide.
  • the inhibitor is a peptide comprising a sequence that is from 3 to about 20 residues of a C-terminal sequence of a PL protein selected from CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD97, CD3n, DOCK2, CD34, FceRIb, and FasLigand; a peptide having a motif X-S-X-A, X-A-D/E-V, X-V/I/L-X*-V, or X-S/T-X-F, (where X is any amino acid and X* is any non-aromatic amino acid); a peptide mimetic,; or a small organic molecule.
  • the invention also provides a pharmaceutical composition containing the inhibitor.
  • the invention also provides a method for treating a disease characterized by leukocyte activation by administering a therapeutically effective amount of an inhibitor of aPL- PDZ interaction.
  • the disease is characterized by an inflammatory or humoral immune response or is an autoimmune disease.
  • the invention further provides a method of reducing inflammation in a subject, by administering an agent that inhibits binding of a PDZ protein and a PL protein, where the PL protein is an adhesion protein, an adaptor protein, or an intracellular protein.
  • the invention also provides use of an inhibitor of the binding of a PDZ protein and a PL protein to inhibit leukocyte activation or to treat a disease mediated by hematopoietic cells, such as a disease is characterized by an inflammatory or humoral immune response.
  • the invention also provides use of an inhibitor ofthe binding of a PDZ protein and a PL protein in the preparation of a medicament for treatment of a disease mediated by hematopoietic cells.
  • the invention also provides a method of modulating a biological function of a hematopoietic cell, comprising introducing into the cell an antagonist that inhibits binding of a PDZ protein and a PL protein in the cell as deduced from Table 2, for example, where the PL protein is DNAM-1 and the PDZ protein is MPP1, MPP2, DLGl, NeDLG, PSD95, LIM, AF6, 41.8 or RGS12, the PL protein is LPAP and the PDZ protein is DLGl or MINTl, or the PL protein is DNAM-1 and the PDZ protein is PSD95 or MPP2.
  • the present invention also relates to peptides and peptide analogues that bind PDZ domains in hematopoietic cells.
  • it relates to fusion peptides and peptide analogues containing a hematopoietic cell surface receptor carboxyl terminal sequence and a transmembrane transporter sequence which facilitates entry ofthe peptides into a target cell.
  • the invention also relates to methods of using such compositions in inhibiting leukocyte activation as measured by cytokine production, cell proliferation, apoptosis and cytotoxicity.
  • the invention provides a method of determining the apparent affinity (Kd) of binding between a PDZ domain and a ligand, by (a) immobilizing a polypeptide comprising the PDZ domain and at least one non-PDZ domain on a surface; (b) contacting the immobilized polypeptide with a plurality of different concentrations of the ligand; (c) determining the amount of binding ofthe ligand to the immobilized polypeptide at each ofthe concentrations of ligand; (d) calculating the apparent affinity of the binding from the binding determined in (c).
  • Kd apparent affinity
  • the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain.
  • the polypeptide comprising the PDZ domain is a fusion protein, for example a GST-PDZ domain fusion protein.
  • the invention provides a method of determining the Ki of an inhibitor or suspected inhibitor of binding between a PDZ domain and a ligand, by (a) immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface; (b) contacting the immobilized polypeptide with a plurality of different mixtures ofthe ligand and inhibitor, wherein the different mixtures comprise a fixed amount of ligand, at least a portion of which is detectably labeled, and different concentrations ofthe inhibitor; (c) determining the amount of ligand bound at the different concentrations of inhibitor; (d) calculating the Ki ofthe inhibitor from the binding determined in (c).
  • the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain.
  • the fixed amount of ligand is between about 0.01 Kd and about 2 Kd.
  • the invention provides a method of identifying an agent that enhances the binding of a PDZ domain to a ligand, by immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface; (b) contacting the immobilized polypeptide with the ligand in the presence of a test agent and determining the amount of ligand bound; and, (c) comparing the amount of ligand bound in the presence of the test agent with the amount of ligand bound by the polypeptide in the absence ofthe test agent, wherein at least twofold greater binding in the presence of the test agent compared to the absence ofthe test agent indicates that the test agent is an agent that enhances the binding of the PDZ domain to the ligand.
  • the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain.
  • the invention provides a method of determining the potency (K enhancer ) °f an enhancer of binding between a PDZ domain and a ligand, by (a) immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface; (b) contacting the immobilized polypeptide with a plurality of different mixtures of the ligand and enhancer, wherein the different mixtures comprise a fixed amount of ligand, at least a portion of which is detectably labeled, and different concentrations ofthe enhancer; (c) determining the amount of ligand bound at the different concentrations of enhancer; (d) calculating the potency (K enhancer ) of the enhancer from the binding determined in (c).
  • the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non- PDZ domain, in an embodiment, the fixed amount of ligand is between about 0.01 Kd and about 0.5 Kd.
  • the invention provides a method of identifying a high specificity interaction between a particular PDZ domain and a ligand known or suspected of binding at least one PDZ domain, by (a) providing a plurality of different immobilized polypeptides, each of said polypeptides comprising a PDZ domain and a non-PDZ domain; (b) determining the affinity of the ligand for each of said polypeptides; (c) comparing the affinity of binding o the ligand to each of said polypeptides.
  • an interaction between the ligand and a particular PDZ domain is deemed to have high specificity when the ligand binds an immobilized polypeptide comprising the particular PDZ domain with at least 2-fold higher affinity than to immobilized polypeptides not comprising the particular PDZ domain (a).
  • the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non- PDZ domain.
  • the invention provides a method for determining the PDZ-PL inhibition profile of a compound by (a) providing (i) a plurality of different immobilized polypeptides, each of said polypeptides comprising a PDZ domain and a non-PDZ domain; (ii) a plurality of corresponding ligands, wherein each ligand binds at least one PDZ domain in (i); (b) contacting each of said immobilized polypeptides in (i) with a corresponding ligand in (ii) in the presence and absence of a test compound; (c) determining for each polypeptide-ligand pair in (b) whether the test compound inhibits binding between the immobilized polypeptide and the corresponding ligand thereby determining the PDZ-PL inhibition profile ofthe test compound.
  • the invention provides an array comprising a plurality of different immobilized polypeptides, each of said polypeptides comprising a PDZ domain and a non-PDZ domain.
  • the array is situated in a plastic multiwell plate.
  • the array has at least 12 different polypeptides comprising at least 12 different PDZ domains, for example, at least 12 different PDZ domains are from PDZs expressed in lymphocytes.
  • the PDZs are selected from those listed in Table 2 or 6.
  • the invention provides an assay device comprising a plurality of different immobilized PDZ-containing proteins organized in an array.
  • the device has at least 25 different PDZ-containing proteins.
  • the invention provides a method for identifying an interaction between a PDZ domain and a PL by contacting a PL to a plurality of PDZ containing polypeptides and detecting binding of at least one PL to a PDZ.
  • the contacting occurs on assay device comprising a plurality of different immobilized PDZ- containing proteins organized in an array.
  • the device has at least 25 different PDZ-containing proteins.
  • an interaction between a PDZ and more than one PL, or between a PL and more than one PDZ, is detected.
  • the invention provides method for identifying a modulator of an interaction between a PDZ and a PL by conducting any ofthe aforementioned assays in the presence and absence of a test compound and detecting a difference in at least one PDZ-PL interaction in the presence and absence ofthe test compound.
  • the modulator is an enhancer of the interaction. In other embodiments, the modulator is an inhibitor of the interaction.
  • FIGURES 1A-1D show the results of exemplary assays in which the binding of biotinylated peptides having a sequence of the carboxyl-terminus ("c-terminus") of various leukocyte proteins to PDZ domains (i.e., GST-PDZ domain fusion proteins) was determined using the "G" assay described infra.
  • the PDZ domains are: PSD95 (Fig. 1 A ); NeDLG (Fig. IB); DLGl (Fig 1C); and 41.8 (Fig. ID). These and other PDZ domain fusion proteins are described infra (e.g., TABLE 2).
  • peptides 1-31 refer to the biotinylated PL peptide used in the assay, and are identified in the Key, infra.
  • “Peptide IDs” are defined in TABLE 3. Key:
  • FIGURES 2A and 2B show the Apparent Affinity Determination for PDZ- Ligand Interactions. Varying concentrations of biotinylated CLASP-2 (Fig. 2 A; TABLE 4) or Fas (Fig. 2B; TABLE 4) C-terminal peptides were reacted with immobilized (plate bound) GST polypeptide or GST-PDZ fusion proteins (GST-DLGl, GST-NeDLG, and GST-PSD95). The binding to GST alone ( ⁇ 0.2 OD units) was subtracted from the binding to the fusion proteins to obtain the signal at each peptide concentration. This signal was then normalized by dividing the signal at each peptide concentration by the maximum signal observed for each peptide-PDZ pair (i.e.
  • FIGURES 3A-3F show inhibition of PDZ - PL peptide interactions.
  • a fixed concentration of biotinylated C-terminal peptide having a sequence based on the C-terminal sequence of a cell surface receptor protein (Clasp 2, CD46, Fas, and KV1.3; see TABLE 4) was bound to immobilized GST polypeptide or the GST-fusion protein indicated at the top left of each frame, in the presence or absence ofthe competitor peptides indicated in the legend of each frame and the level of inhibition determined.
  • the competitor peptides are present at 100 uM; in Figs. 3C-F the competitor is present at the indicated concentration.
  • FIGURES 4A and 4B shows the results of introduction of a Tat-CD3 fusion peptide on T cell activation.
  • Antigen-specific T cell activation was measured by cytokine production.
  • Fusion peptides containing tat and a T cell surface molecule carboxyl terminus inhibited ⁇ -interferon (IFN) production by a T cell line in response to myelin basic protein (MBP) stimulation.
  • IFN ⁇ -interferon
  • MBP myelin basic protein
  • a “fusion protein” or “fusion polypeptide” as used herein refers to a composite protein, i.e., a single contiguous amino acid sequence, made up of two (or more) distinct, heterologous polypeptides which are not normally fused together in a single amino acid sequence.
  • a fusion protein can include a single amino acid sequence that contains two entirely distinct amino acid sequences or two similar or identical polypeptide sequences, provided that these sequences are not normally found together in the same configuration in a single amino acid sequence found in nature.
  • Fusion proteins can generally be prepared using either recombinant nucleic acid methods, i.e., as a result of transcription and translation of a recombinant gene fusion product, which fusion comprises a segment encoding a polypeptide of the invention and a segment encoding a heterologous protein, or by chemical synthesis methods well known in the art.
  • fusion protein construct as used herein is a polynucleotide encoding a fusion protein.
  • PDZ domain refers to protein sequence (i.e., modular protein domain) of approximately 90 amino acids, characterized by homology to the brain synaptic protein PSD-95, the Drosophila septate junction protein Discs-Large (DLG), and the epithelial tight junction protein ZO1 (ZO1). PDZ domains are also known as Discs-Large homology repeats ("DHRs”) and GLGF repeats). PDZ domains generally appear to maintain a core consensus sequence (Doyle, D. A., 1996, Cell 85: 1067-1076).
  • PDZ domains are found in diverse membrane-associated proteins including members ofthe MAGUK family of guanylate kinase homologs, several protein phosphatases and kinases, neuronal nitric oxide synthase, and several dystrophin-associated proteins, collectively known as syntrophins.
  • PDZ domain-containing proteins and PDZ domain sequences are shown in TABLE 3.
  • the term "PDZ domain” also encompasses variants (e.g., naturally occuring variants) of the sequences of TABLE 3 (e.g., polymorphic variants, variants with conservative substitutions, and the like).
  • PDZ domains are substantially identical to those shown in TJ BLE 3, e.g., at least about 70%, at least about 80%, or at least about 90% amino acid residue identity when compared and aligned for maximum correspondence.
  • PDZ protein refers to a naturally occurring protein containing a PDZ domain, e.g., a human protein.
  • exemplary PDZ proteins include CASK, MPPl, DLGl, PSD95, NeDLG, TAX33, SYNla, TAX43, LDP, LIM, LIMKl, LIMK2, MPP2, NOS1 , AF6, PTN-4, prILl ⁇ . 41.8kD, KIAA0559, RGS12, KIAA0316, DVL1, TAX40, TIAM1, MINTl, KIAA0303, CBP, MLNT3, TAX2, KIAA0561.
  • Exemplary PDZ proteins are listed in TABLE 2 and TABLE 3.
  • PDZ-domain polypeptide refers to a polypeptide containing a PDZ domain, such as a fusion protein including a PDZ domain sequence, a naturally occurring PDZ protein, or an isolated PDZ domain peptide.
  • PL protein or "PDZ Ligand protein” refers to a naturally occurring protein that forms a molecular complex with a PDZ-domain, or to a protein whose carboxy-terminus, when expressed separately from the full length protein (e.g., as a peptide fragment of 4-25 residues, e.g., 16 residues), forms such a molecular complex.
  • the molecular complex can be observed in vitro using the "A assay” or "G assay” described infra, or in vivo.
  • Exemplary PL proteins listed in TABLE 2 are demonstrated to bind specific PDZ proteins. This definition is not intended to include anti-PDZ antibodies and the like.
  • a "PL sequence” refers to the amino acid sequence of the C- terminus of a PL protein (e.g., the C-terminal 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 20 or 25 residues) ("C-terminal PL sequence") or to an internal sequence known to bind a PDZ domain (“internal PL sequence).
  • a "PL peptide” is a peptide of having a sequence from, or based on, the sequence of the C-terminus of a PL protein. Exemplary PL peptides (biotinylated) are listed in TABLE 4.
  • a "PL fusion protein” is a fusion protein that has a PL sequence as one domain, typically as the C-terminal domain of the fusion protein.
  • An exemplary PL fusion protein is a tat-PL sequence fusion.
  • PL inhibitor peptide sequence refers to PL peptide an amino acid sequence that (in the form of a peptide or PL fusion protein) inhibits the interaction between a PDZ domain polypeptide and a PL peptide (e.g., in an A assay or a G assay).
  • a "PDZ-domain encoding sequence” means a segment of a polynucleotide encoding a PDZ domain.
  • the polynucleotide is DNA, RNA, single stranded or double stranded.
  • the terms "antagonist” and “inhibitor,” when used in the context of modulating a binding interaction are used interchangeably and refer to an agent that reduces the binding of the, e.g., PL sequence (e.g., PL peptide) and the, e.g., PDZ domain sequence (e.g., PDZ protein, PDZ domain peptide).
  • agonist and “enhancer,” when used in the context of modulating a binding interaction (such as the binding of a PDZ domain sequence to a PL sequence), are used interchangeably and refer to an agent that increases the binding ofthe, e.g.,
  • PL sequence e.g., PL peptide
  • PDZ domain sequence e.g., PDZ protein, PDZ domain peptide
  • peptide mimetic As used herein, the terms “peptide mimetic, " “peptidomimetic,” and “peptide analog” are used interchangeably and refer to a synthetic chemical compound which has substantially the same structural and/or functional characteristics of an PL inhibitory or PL binding peptide ofthe invention.
  • the mimetic can be either entirely composed of synthetic, non- natural analogues of amino acids, or, is a chimeric molecule of partly natural peptide amino acids and partly non-natural analogs of amino acids.
  • the mimetic can also incorporate any amount of natural amino acid conservative substitutions as long as such substitutions also do not substantially alter the mimetic's structure and/or inhibitory or binding activity.
  • a mimetic composition is within the scope of the invention if it is capable of binding to a PDZ domain and/or inhibiting a PL-PDZ interaction.
  • Polypeptide mimetic compositions can contain any combination of nonnatural structural components, which are typically from three stmctural groups: a) residue linkage groups other than the natural amide bond ("peptide bond") linkages; b) non-natural residues in place of naturally occurring amino acid residues; or c) residues which induce secondary structural mimicry, i.e., to induce or stabilize a secondary structure, e.g., a beta turn, gamma turn, beta sheet, alpha helix conformation, and the like.
  • a polypeptide can be characterized as a mimetic when all or some of its residues are joined by chemical means other than natural peptide bonds.
  • DCC dicyclohexylcarbodiimide
  • DIC diisopropylcarbodiimide
  • aminomethylene CH 2 -NH
  • ethylene olefin
  • ether CH 2 -O
  • a polypeptide can also be characterized as a mimetic by containing all or some non-natural residues in place of naturally occurring amino acid residues.
  • Nonnatural residues are well described in the scientific and patent literature; a few exemplary nonnatural compositions useful as mimetics of natural amino acid residues and guidelines are described below.
  • Mimetics of aromatic amino acids can be generated by replacing by, e.g., D- or L- naphylalanine; D- or L- phenylglycine; D- or L-2 thieneylalanine; D- or L-l, -2, 3-, or 4- pyreneylalanine; D- or L-3 thieneylalanine; D- or L-(2-pyridinyl)-alanine; D- orL-(3-pyridinyl)- alanine; D- or L-(2-pyrazinyl)-alanine; D- or L-(4-isopropyl)-phenylglycine; D-(trifluoromethyl)- phenylglycine; D-(trifluoromethyl)-phenylalanine; D-p-fluorophenylalanine; D- or L-p- biphenylphenylalanine; K- or L-p-methoxybiphenylphenyla
  • Aromatic rings of a nonnatural amino acid include, e.g., thiazolyl, thiophenyl, pyrazolyl, benzimidazolyl, naphthyl, furanyl, pyrrolyl, and pyridyl aromatic rings.
  • Mimetics of acidic amino acids can be generated by substitution by, e.g., non- carboxylate amino acids while maintaining a negative charge; (phosphono)alanine; sulfated threonine.
  • Aspartyl or glutamyl can also be converted to asparaginyl and glutaminyl residues by reaction with ammonium ions.
  • Mimetics of basic amino acids can be generated by substitution with, e.g., (in addition to lysine and arginine) the amino acids ornithine, citrulline, or (guanidino)-acetic acid, or (guanidino)alkyl-acetic acid, where alkyl is defined above.
  • Nitrile derivative e.g., containing the CN-moiety in place of COOH
  • Asparaginyl and glutaminyl residues can be deaminated to the corresponding aspartyl or glutamyl residues.
  • Arginine residue mimetics can be generated by reacting arginyl with, e.g., one or more conventional reagents, including, e.g., phenylglyoxal, 2,3-butanedione, 1,2- cyclohexanedione, or ninhydrin, preferably under alkaline conditions.
  • one or more conventional reagents including, e.g., phenylglyoxal, 2,3-butanedione, 1,2- cyclohexanedione, or ninhydrin, preferably under alkaline conditions.
  • Tyrosine residue mimetics can be generated by reacting tyrosyl with, e.g., aromatic diazonium compounds or tetranitromethane.
  • N-acetylimidizol and tetranitromethane can be used to form O-acetyl tyrosyl species and 3-nitro derivatives, respectively.
  • Cysteine residue mimetics can be generated by reacting cysteinyl residues with, e.g., alpha-haloacetates such as 2-chloroacetic acid or chloroacetamide and corresponding amines; to give carboxymethyl or carboxyamidomethyl derivatives.
  • alpha-haloacetates such as 2-chloroacetic acid or chloroacetamide and corresponding amines
  • Cysteine residue mimetics can also be generated by reacting cysteinyl residues with, e.g., bromo-trifluoroacetone, alpha- bromo-beta-(5-imidozoyl) propionic acid; chloroacetyl phosphate, N-alkylmaleimides, 3-nitro-2- pyridyl disulfide; methyl 2-pyridyl disulfide; p-chloromercuribenzoate; 2-chloromercuri-4 nitrophenol; or, chloro-7-nitrobenzo-oxa-l,3-diazole.
  • cysteinyl residues e.g., bromo-trifluoroacetone, alpha- bromo-beta-(5-imidozoyl) propionic acid
  • chloroacetyl phosphate N-alkylmaleimides
  • 3-nitro-2- pyridyl disulfide methyl 2-pyridyl disulfide
  • Lysine mimetics can be generated (and amino terminal residues can be altered) by reacting lysinyl with, e.g., succinic or other carboxylic acid anhydrides. Lysine and other alpha-amino-containing residue mimetics can also be generated by reaction with imidoesters, such as methyl picolinimidate, pyridoxal phosphate, pyridoxal, chloroborohydride, trinitrobenzenesulfonic acid, O-methylisourea, 2,4, pentanedione, and transamidase-catalyzed reactions with glyoxylate.
  • imidoesters such as methyl picolinimidate, pyridoxal phosphate, pyridoxal, chloroborohydride, trinitrobenzenesulfonic acid, O-methylisourea, 2,4, pentanedione, and transamidase-catalyzed reactions with glyoxylate.
  • Mimetics of methionine can be generated by reaction with, e.g., methionine sulfoxide.
  • Mimetics of proline include, e.g., pipecolic acid, thiazohdine carboxylic acid, 3- or
  • Histidine residue mimetics can be generated by reacting histidyl with, e.g., diethylprocarbonate or para- bromophenacyl bromide.
  • mimetics include, e.g., those generated by hydroxylation of proline and lysine; phosphorylation ofthe hydroxyl groups of seryl or threonyl residues; methylation ofthe alpha-amino groups of lysine, arginine and histidine; acetylation of the N-terminal amine; methylation of main chain amide residues or substitution with N-methyl amino acids; or amidation of C-terminal carboxyl groups.
  • a component of a natural polypeptide can also be replaced by an amino acid (or peptidomimetic residue) of the opposite chirality.
  • an amino acid or peptidomimetic residue of the opposite chirality.
  • any amino acid naturally occurring in the L-con figuration (which can also be referred to as the R or S, depending upon the structure ofthe chemical entity) can be replaced with the amino acid of the same chemical structural type or a peptidomimetic, but of the opposite chirality, generally referred to as the D- amino acid, but which can additionally be referred to as the R- or S- form.
  • the mimetics of the invention can also include compositions that contain a structural mimetic residue, particularly a residue that induces or mimics secondary structures, such as a beta turn, beta sheet, alpha helix structures, gamma turns, and the like.
  • a structural mimetic residue particularly a residue that induces or mimics secondary structures, such as a beta turn, beta sheet, alpha helix structures, gamma turns, and the like.
  • substitution of natural amino acid residues with D-amino acids; N-alpha-methyl amino acids; C- alpha-methyl amino acids; or dehydroamino acids within a peptide can induce or stabilize beta turns, gamma turns, beta sheets or alpha helix conformations.
  • Beta turn mimetic structures have been described, e.g., by Nagai (1985) Tet. Lett. 26:647-650; Feigl (1986) J. Amer. Chem. Soc.
  • Beta sheet mimetic structures have been described, e.g., by Smith (1992) J. Amer. Chem. Soc. 114:10672-10674.
  • a type VI beta turn induced by a cis amide surrogate, 1 ,5-disubstituted tetrazol is described by Beusen (1995) Biopolymers 36:181-200.
  • Incorporation of achiral omega-amino acid residues to generate polymethylene units as a substitution for amide bonds is described by Banerjee
  • peptide variants and “conservative amino acid substitutions” refer to peptides that differ from a reference peptide (e.g., a peptide having the sequence of the c arboxy-terminus of a specified PL protein) by substitution of an amino acid residue having similar properties (based on size, polarity, hydrophobicity, and the like).
  • a reference peptide e.g., a peptide having the sequence of the c arboxy-terminus of a specified PL protein
  • substitution of an amino acid residue having similar properties based on size, polarity, hydrophobicity, and the like.
  • amino acids may be generally categorized into three main classes: hydrophilic amino acids, hydrophobic amino acids and cysteine-like amino acids, depending primarily on the characteristics of the amino acid side chain. These main classes may be further divided into subclasses.
  • Hydrophilic amino acids include amino acids having acidic, basic or polar side chains and hydrophobic amino acids include amino acids having aromatic or apolar side chains.
  • Apolar amino acids may be further subdivided to include, among others, aliphatic amino acids.
  • the definitions of the classes of amino acids as used herein are as follows:
  • Hydrophobic Amino Acid refers to an amino acid having a side chain that is uncharged at physiological pH and that is repelled by aqueous solution.
  • genetically encoded hydrophobic amino acids include He, Leu and Val.
  • non-genetically encoded hydrophobic amino acids include t-BuA.
  • Aromatic Amino Acid refers to a hydrophobic amino acid having a side chain containing at least one ring having a conjugated ⁇ -electron system (aromatic group).
  • aromatic group may be further substituted with groups such as alkyl, alkenyl, alkynyl, hydroxyl, sulfanyl, nitro and amino groups, as well as others.
  • genetically encoded aromatic amino acids include Phe, Tyr and Trp.
  • aromatic amino acids include phenylglycine, 2-naphthylalanine, ⁇ -2-thienylalanine, 1,2,3,4- tetrahydroisoquinoline-3-carboxylic acid, 4-chloro-phenylalanine, 2-fluorophenylalanine, 3- fluorophenylalanine and 4-fluorophenylalanine.
  • Apolar Amino Acid refers to a hydrophobic amino acid having a side chain that is generally uncharged at physiological pH and that is not polar. Examples of genetically encoded apolar amino acids include Gly, Pro and Met. Examples of non-encoded apolar amino acids include Cha.
  • Aliphatic Amino Acid refers to an apolar amino acid having a saturated or unsaturated straight chain, branched or cyclic hydrocarbon side chain.
  • genetically encoded aliphatic amino acids include Ala, Leu, Val and lie.
  • non-encoded aliphatic amino acids include Nle.
  • Hydrophilic Amino Acid refers to an amino acid having a side chain that is attracted by aqueous solution.
  • examples of genetically encoded hydrophilic amino acids include Ser and Lys.
  • examples of non-encoded hydrophilic amino acids include Cit and hCys.
  • Acidic Amino Acid refers to a hydrophilic amino acid having a side chain pK value of less than 7. Acidic amino acids typically have negatively charged side chains at physiological pH due to loss of a hydrogen ion. Examples of genetically encoded acidic amino acids include Asp and Glu. "Basic Amino Acid” refers to a hydrophilic amino acid having a side chain pK value of greater than 7. Basic amino acids typically have positively charged side chains at physiological pH due to association with hydronium ion. Examples of genetically encoded basic amino acids include Arg, Lys and His.
  • non-genetically encoded basic amino acids examples include the non-cyclic amino acids omithine, 2,3-diaminopropionic acid, 2,4-diaminobutyric acid and homoarginine.
  • Poly Amino Acid refers to a hydrophilic amino acid having a side chain that is uncharged at physiological pH, but which has a bond in which the pair of electrons shared in common by two atoms is held more closely by one of the atoms.
  • genetically encoded polar amino acids examples include Asx and Glx.
  • non-genetically encoded polar amino acids include citrulline, N-acetyl lysine and methionine sulfoxide.
  • cyste-Like Amino Acid refers to an amino acid having a side chain capable of forming a covalent linkage with a side chain of another amino acid residue, such as a disulfide linkage.
  • cysteine-like amino acids generally have a side chain containing at least one thiol (SH) group.
  • examples of genetically encoded cysteine-like amino acids include Cys.
  • non-genetically encoded cysteine-like amino acids include homocysteine and penicillamine.
  • cysteine has both an aromatic ring and a polar hydroxyl group.
  • cysteine has dual properties and can be included in both the aromatic and polar categories.
  • cysteine in addition to being able to form disulfide linkages, cysteine also has apolar character.
  • cysteine can be used to confer hydrophobicity to a peptide.
  • Certain commonly encountered amino acids which are not genetically encoded of which the peptides and peptide analogues ofthe invention may be composed include, but are not limited to, ⁇ -alanine (b-Ala) and other omega-amino acids such as 3-aminopropionic acid (Dap), 2,3-diaminopropionic acid (Dpr), 4-aminobutyric acid and so forth; ⁇ -aminoisobutyric acid (Aib); ⁇ -aminohexanoic acid (Aha); ⁇ -aminovaleric acid (Ava); N-methylglycine or sarcosine (MeGly); ornithine (Orn); citrulline (Cit); t-butylalanine (t-BuA); t-butylglycine (t-BuG); N-methylisoleucine (Melle); phenylglycine (Phg); cyclohexylalanine (Cha); norleucine (
  • TABLE 1 is for illustrative purposes only and does not purport to be an exhaustive list of amino acid residues which may comprise the peptides and peptide analogues described herein.
  • Other amino acid residues which are useful for making the peptides and peptide analogues described herein can be found, e.g., in Fasman, 1989, CRC Practical Handbook of Biochemistry and Molecular Biology, CRC Press, Inc., and the references cited therein.
  • Amino acids not specifically mentioned herein can be conveniently classified into the above-described categories on the basis of known behavior and/or their characteristic chemical and/or physical properties as compared with amino acids specifically identified.
  • a "detectable label” has the ordinary meaning in the art and refers to an atom (e.g., radionuciide), molecule (e.g., fluorescein), or complex, that is or can be used to detect (e.g., due to a physical or chemical property), indicate the presence of a molecule or to enable binding of another molecule to which it is covalently bound or otherwise associated.
  • label also refers to covalently bound or otherwise associated molecules (e.g., a biomolecule such as an enzyme) that act on a substrate to produce a detectable atom, molecule or complex.
  • Detectable labels suitable for use in the present invention include any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means.
  • Labels useful in the present invention include biotin for staining with labeled streptavidin conjugate, magnetic beads (e.g., DynabeadsTM), fluorescent dyes (e.g., fluorescein, Texas red, rhodamine, green fluorescent protein, enhanced green fluorescent protein, and the like), radiolabels (e.g., 3H, 1251, 35S, 14C, or 32P), enzymes (e.g., hydrolases, particularly phosphatases such as alkaline phosphatase, esterases and glycosidases, or oxidoreductases, particularly peroxidases such as horse radish peroxidase, and others commonly used in ELISAs), substrates, cofactors, inhibitors, chemiluminescent groups, chromogenic agents, and colorimetric labels such as colloidal gold or
  • Patents teaching the use of such labels include U.S. Patent Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241.
  • Means of detecting such labels are well known to those of skill in the art.
  • radiolabels and chemiluminescent labels may be detected using photographic film or scintillation counters
  • fluorescent markers may be detected using a photodetector to detect emitted light (e.g., as in fluorescence-activated cell sorting).
  • Enzymatic labels are typically detected by providing the enzyme with a substrate and detecting the reaction product produced by the action of the enzyme on the substrate, and colorimetric labels are detected by simply visualizing the colored label.
  • a label is any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means.
  • the label may be coupled directly or indirectly to the desired component ofthe assay according to methods well known in the art.
  • Non-radioactive labels are often attached by indirect means.
  • a ligand molecule e.g., biotin
  • the ligand then binds to an anti-ligand (e.g., streptavidin) molecule which is either inherently detectable or covalently bound to a signal generating system, such as a detectable enzyme, a fluorescent compound, or a chemiluminescent compound.
  • a signal generating system such as a detectable enzyme, a fluorescent compound, or a chemiluminescent compound.
  • ligands and anti-ligands can be used.
  • a ligand has a natural anti-ligand, for example, biotin, thyroxine, and cortisol, it can be used in conjunction with the labeled, naturally occurring anti-ligands.
  • any haptenic or antigenic compound can be used in combination with an antibody.
  • the molecules can also be conjugated directly to signal generating compounds, e.g., by conjugation with an enzyme or fluorophore.
  • Means of detecting labels are well known to those of skill in the art.
  • means for detection include a scintillation counter, photographic film as in autoradiography, or storage phosphor imaging.
  • the label is a fluorescent label, it may be detected by exciting the fluorochrome with the appropriate wavelength of light and detecting the resulting fluorescence. The fluorescence may be detected visually, by means of photographic film, by the use of electronic detectors such as charge coupled devices (CCDs) or photomulfipliers and the like.
  • CCDs charge coupled devices
  • enzymatic labels may be detected by providing the appropriate substrates for the enzyme and detecting the resulting reaction product.
  • simple colorimetric labels may be detected by observing the color associated with the label. It will be appreciated that when pairs of fluorophores are used in an assay, it is often preferred that they have distinct emission patterns (wavelengths) so that they can be easily distinguished.
  • the term "substantially identical" in the context of comparing amino acid sequences means that the sequences have at least about 70%, at least about 80%), or at least about 90% amino acid residue identity when compared and aligned for maximum correspondence.
  • An algorithm that is suitable for determining percent sequence identity and sequence similarity is the FASTA algorithm, which is described in Pearson, W.R. & Lipman, D.J., 1988, Proc. Natl. Acad. Sci. U.S.A. 85: 2444. See also W. R. Pearson, 1996, Methods Enzymol. 266: 227-258.
  • hematopoietic cells include leukocytes including lymphocytes (T cells, B cells and NK cells), monocytes, and granulocytes (i.e., neutrophils, basophils and eosinophils), macrophages, dendritic cells, megakaryocytes, reticulocytes, erythrocytes, and CD34 ⁇ stem cells.
  • test compound or “test agent” are used interchangably and refer to a candidate agent that may have enhancer/agonist, or inhibitor/antagonist activity, e.g., inhibiting or enhancing an interaction such as PDZ-PL binding.
  • the candidate agents or test compounds may be any of a large variety of compounds, both naturally occurring and synthetic, organic and inorganic, and including polymers (e.g., oligopeptides, polypeptides, ohgonucleotides, and polynucleotides), small molecules, antibodies (as broadly defined herein), sugars, fatty acids, nucleotides and nucleotide analogs, analogs of naturally occurring structures (e.g., peptide mimetics, nucleic acid analogs, and the like), and numerous other compounds.
  • test agents are prepared from diversity libraries, such as random or combinatorial peptide or non-peptide libraries.
  • libraries are known in the art that can be used, e.g., chemically synthesized libraries, recombinant (e.g., phage display libraries), and in vitro translation-based libraries.
  • chemically synthesized libraries are described in Fodor et al, 1991, Science 251 :767-773; Houghten et al., 1991, Nature 354:84-86; Lam et al., 1991, Nature 354:82-84; Medynski, 1994, Bio/Technology 12:709-710; Gallop et al., 1994, J. Medicinal Chemistry 37(9): 1233- 1251; Ohlmeyer et al., 1993, Proc. Natl. Acad. Sci.
  • phage display libraries are described in Scott and Smith, 1990, Science 249:386-390; Devlin et al, 1990, Science, 249:404-406; Christian, R.B., et al., 1992, J. Mol. Biol. 227:71 1-718); Lenstra, 1992, J. Immunol. Meth. 152:149-157; Kay et al., 1993, Gene 128:59-65; and PCT Publication No. WO 94/18318 dated August 18, 1994.
  • In vitro translation- based libraries include but are not limited to those described in PCT Publication No. WO 91/05058 dated April 18, 1991 ; and Mattheakis et al., 1994, Proc. Natl.
  • a benzodiazepine library (see e.g., Bunin et al., 1994, Proc. Natl. Acad. Sci. USA 91 :4708-4712) can be adapted for use.
  • Peptoid libraries (Simon et al., 1992, Proc. Natl. Acad. Sci. USA 89:9367-9371) can also be used.
  • Another example of a library that can be used, in which the amide functionalities in peptides have been permethylated to generate a chemically transformed combinatorial library, is described by Ostresh et al. (1994, Proc. Natl. Acad.
  • binding refers to binding between two molecules, for example, a ligand and a receptor, characterized by the ability of a molecule (ligand) to associate with another specific molecule (receptor) even in the presence of many other diverse molecules, i.e., to show preferential binding of one molecule for another in a heterogeneous mixture of molecules. Specific binding of a ligand to a receptor is also evidenced by reduced binding of a detectably labeled ligand to the receptor in the presence of excess unlabeled ligand (i.e., a binding competition assay).
  • a "plurality" of PDZ proteins has its usual meaning.
  • the plurality is at least 5, and often at least 25, at least 40 or at least 60 different PDZ proteins.
  • the plurality is selected from the list of PDZ polypeptides listed in Table 2 or Table 6.
  • the plurality of different PDZ proteins are from (i.e., expressed in) a particular specified tissue or a particular class or type of cell.
  • the plurality of different PDZ proteins represents a substantial fraction (e.g., typically at least 50%, more often at least 80%) of all ofthe PDZ proteins known to be, or suspected of being, expressed in the tissue or cell(s), e.g., all of the PDZ proteins known to be present in lymphocytes or hematopoetic cells.
  • the plurality is at least 50%, usually at least 80%, at least 90% or all ofthe PDZ proteins disclosed herein as being expressed in hematopoietic cells (see Tables 2 and 6).
  • the plurality includes at least 1, often at least 2, sometimes at least 5 or at least 10 and sometimes all of the following PDZ proteins: BAl I associated prof, Connector enhancer, DLG5 (pdlg), DVL3, GTPase, Guanin-exchange factor 1, PDZ domain containing prot, KLAA147, KIAA0300, KIAA0380, KIAA0440, KIAA0545, KIAA0807, KIAA0858, KIAA0902, novel serine protease, PDZKl, PICK8, PTN-3, RPIP8, serine protease, 26s subunit p27, hSYNTENTN, TAXI -IP, TAX2-like protein, wwp3, XI 1 prot.
  • a "plurality" may refer to at least 5, at least 10, and often at least 25 PLs such as those specifcally listed herein, or to the classes and percentages set forth supra for PDZ domains. 6.
  • the present inventors have discovered that interactions between PDZ proteins and PL proteins play an important and extensive role in the biological function of hematopoietic cells and other cells involved in the immune response.
  • PDZ-PL interactions were known in the nervous system (i.e., in neurons), their universal importance in hematopoietic cell function, especially in function of T cells and B cells, and their fundamental role in modulation of the immune response has not been recognized.
  • cell adhesion molecules that mediate cell-cell interaction in the hematopoietic system are PDZ-binding proteins (PL proteins) and bind to PDZ proteins.
  • biological function in the context of a cell, refers to a detectable biological activity normally carried out by the cell, e.g., a phenotypic change such as proliferation, cell activation (e.g., T cell activation, B cell activation, T-B cell conjugate formation), cytokine release, degranulation, tyrosine phosphorylation, ion (e.g., calcium) flux, metabolic activity, apoptosis, changes in gene expression, maintenance of cell structure, cell migration, adherance to a substrate, signal transduction, cell-cell interactions, and others described herein or known in the art.
  • a phenotypic change such as proliferation, cell activation (e.g., T cell activation, B cell activation, T-B cell conjugate formation), cytokine release, degranulation, tyrosine phosphorylation, ion (e.g., calcium) flux, metabolic activity, apoptosis, changes in gene expression, maintenance of cell structure, cell migration, adher
  • the present invention relates to peptides, peptide analogues or mimetics, pharmaceutical compositions, and methods of using such compositions to regulate the biological activities of hematopoietic cells, e.g. T cells and B cells, or other cells (e.g., endothelial cells) that necessary for immune function.
  • the invention further relates to methods of using the compositions to modulate hematopoietic cell activation and immune function, as well assays for such inhibitors.
  • TABLE 2 summarizes an extensive analysis of protein interactions in T cells and
  • PDZ proteins the vast majority of which were not previously known to be expressed in immune system cells, are listed in the top row of TABLE 2.
  • the first column ofthe table lists PL proteins. Positions in the matrix denoted by the letter “A” or “G” indicate that an interaction between the PDZ protein and the PL has been detected in novel binding assays (described in detail in, e.g., Section 6.2, infra). A blank cell indicates that no interaction was detected using the assays of the invention.
  • An asterisk (*) denotes a PL-PDZ interaction previously reported in the scientific literature.
  • the PDZ proteins listed in TABLE 2 are naturally occurring proteins containing a PDZ domain.
  • the present invention is particularly directed to the detection and modulation of interactions between PDZ proteins and PL proteins in hematopoietic cells.
  • Exemplary PL proteins are listed in TJABLE 2. Notably, as discussed infra, many of these PL proteins have not previously been recognized as such in any cell system.
  • PL protein classes are known, and the PL proteins described herein can be characterized as (1) "PL adhesion proteins” (2) "PL ion channel proteins” (3) "PL adaptor proteins” (4) "PL intracellular proteins” and (5) "PL cytokine receptor proteins.”
  • an adhesion protein is a cell surface protein involved in cell-cell interaction by direct contact with cell surface molecules (e.g., transmembrane proteins or surface proteins) on a different cell.
  • cell surface molecules e.g., transmembrane proteins or surface proteins
  • the PL adhesion protein localizes at the interface of the two cells and directly contacts a cell surface molecule on the second cell.
  • a cell-cell interface is a region where the plasma membranes of two different cells are in close (generally ⁇ 10 nm, often about 1 nm) apposition.
  • direct molecular contact means interaction of molecules at distances where Van der Walls forces are significant, generally less than about 1 nm.
  • Exemplary PL adhesion proteins include CD6; CD49E (alpha-4); CD49F (a form, alpha ⁇ ); CD138 (syndecan); CLASP-1; CLASP-4; VCAMl; CLASP-2; DNAM-1; CD83; CD44 (long form); CD97; (CD55L); CD3n; DOCK2; CD34; and FceRIb.
  • the PL proteins of the invention are PL adhesion proteins.
  • the invention provides methods and reagents, as detailed herein, for inhibiting interactions between PL adhesion proteins and PDZ proteins to modulate an immune response. In an embodiment, the inhibition or modulation occurs in a hematopoietic cell.
  • the inhibition or modulation occurs in an endothelial cell. In a related embodiment, the inhibition or modulation occurs in an endothelial cell. In a related embodiment, the inhibition or modulation occurs in an epithelial cells, keratinocytes, hepatocytes, cardiac myocytes.
  • an ion channel protein means a transmembrane protein that itself catalyzes the passage of an ion from aqueous solution on one side of a lipid bilayer membrane to aqueous solution on the other side (e.g., by forming a small pore in the membrane).
  • PL ion channel proteins is Kvl .3.
  • the PL proteins of the invention are PL ion channel proteins.
  • the invention provides methods and reagents, as detailed herein, for inhibiting interactions between PL ion channel proteins and PDZ proteins to modulate an immune response. In an embodiment, the inhibition or modulation occurs in a hematopoeitic cell.
  • the inhibition or modulation occurs in an endothelial cell.
  • an intercellular (i.e., cytosolic) protein has the normal meaning in the art and refers to a protein that is not membrane bound, e.g., has no transmembrane domain.
  • the PL proteins of the invention are PL intercellular proteins.
  • Exemplary PL intercellular proteins include Glycophorin C and LPAP.
  • the invention provides methods and reagents, as detailed herein, for inhibiting interactions between PL cytoplasmic proteins and PDZ proteins to modulate an immune response.
  • the inhibition or modulation occurs in a hematopoeitic cell.
  • the inhibition or modulation occurs in an endothelial cell.
  • cytokine receptor has the normal meaning in the art and refers to a membrane protein with an extracellular domain that specifically binds a cytokine.
  • exemplary PL cytokine receptor proteins include CDW125 (IL5R), CDW128A (IL8RA), and
  • the PL proteins of the invention are PL cytokine proteins.
  • the invention provides methods and reagents, as detailed herein, for inhibiting interactions between PL cytokine proteins and PDZ proteins to modulate an immune response.
  • the inhibition or modulation occurs in a hematopoeitic cell.
  • the inhibition or modulation occurs in an endothelial cell.
  • an adaptor protein means a molecule (e.g., protein) that contributes to the formation of a multimolecular complex by binding two or more other biomolecuics.
  • the binding o the two or more other molecules by the adaptor molecule/protein generally involves direct molecular contact between the adaptor protein and each of he two or more other molecules.
  • One exemplary PL adaptor protein is LPAP.
  • the PL proteins of the invention are PL adaptor proteins.
  • the invention provides methods and reagents, as detailed herein, for inhibiting interactions between PL adaptor proteins and PDZ proteins to modulate an immune response. In an embodiment, the inhibition or modulation occurs in a hematopoeitic cell.
  • the inhibition or modulation occurs in an endothelial cell.
  • the PL proteins of the invention are characterized by specific C-terminal (i.e., PL domain) amino acid sequences or amino acid motifs, as described elsewhere in this disclosure.
  • the PL proteins of the invention bind a PDZ protein expressed in T lymphocytes, B lymphocytes, or both T and B lymphocytes. In an embodiment, the PL protein binds a PDZ protein expressed in endothelial cells. In various embodiments, the PL proteins and/or the PDZ protein to which it binds are not expressed in the nervous system (e.g., neurons).
  • the nervous system e.g., neurons
  • the PL protein ofthe invention binds only one PDZ protein listed in TABLE 2. In other embodiment, the PL protein binds 1 to 3, 3 to 5, or more than 5 different PDZ proteins listed in TABLE 2.
  • the PL protein is expressed or upregulated upon cell activation (e.g., in activated B lymphocytes, T lymphocytes) or upon entry into mitosis (e.g., up-regulation in rapidly proliferating cell populations).
  • the PL protein is (i) a protein that mediates immune cell (e.g., hematopoietic cell) activation or migration, (ii) a protein that does not mediate apoptosis in a cell type (iii) a protein that is other than a G-protein coupled seven transmembrane helix receptor, (iv) a protein that is G-protein coupled seven transmembrane helix receptor but not a cytokine receptor or (v) a protein that is not a G-protein coupled seven transmembrane helix receptor and is a cytokine receptor.
  • PDZ proteins are expressed in immune system cells, and play a fundamental biological role in modulation of the immune response.
  • PDZ proteins DLGl and TIAM-I have been previously described to be in T cells.
  • the present inventors discovered, using a BLAST search of the Human EST database and the experiments described infra, that several additional PDZ proteins are present in hematopoietic cells including MPPl, P-DLG, VELI-1, PSD95, syntenin in T cells and CASK, DLGl , DLG2, ZIP KINASE, syntrophin 2, P-dlg, PSD95, and syntenin in B cells.
  • PCR polymerase chain reaction
  • RT reverse transcription
  • PCR, RT-PCR and other methods for analysis and manipulation of nucleic acids are well known and are described generally in Sambrook et al., (1989) MOLECULAR CLONING: A LABORATORY MANUAL, 2ND ED., VOLS.
  • the precipitate was collected by centrifugation at 12000 x g for 15 minutes at 4° C, washed with 75% ethanol, finally recollected by another spin at 12000 x g for 15 minutes at 4° C, air dried and resuspended in an appropriate volume of DEPC treated water.
  • RNA concentration and purity were determined by the measurement of 260/280 nm light absorption by the nucleic acid.
  • the SUPERSCRIPT II reverse transcriptase cDNA kit (GIBCO-BRL; Cat. # 18064-014) was used. RNA input per 200 ⁇ l cDNA reaction sample was 10 ⁇ g. Prior to cDNA synthesis RNA was treated with 1 unit/ ⁇ l DNAse I in 1 10 ⁇ l water at 37° C for 20 minutes. DNase I was then inactivated by a 10 minutes incubation at 70 ° C.
  • Random primer was used for cDNA priming; 10 ⁇ l of random hexamer primer (100 ng/ ⁇ l) was added, samples were heated to 70° C for 5 minutes and chilled on ice. Subsequently 40 ⁇ l SUPERSCRIPT II "first strand" buffer, 20 ⁇ l of 0.1 M DDT, 10 ⁇ l of a 10 mM of mix of deoxynucleotide triphosphates (dATP, dCTP, dGTP, dUTP) and 10 ⁇ l of SUPERSCRIPT II reverse transcriptase were added and cDNA synthesis was done for 45 minutes at 42 ° C. Reavtions were stopped by a 5 minutes incubation at 95 ° C and typically, 2-4 ⁇ l of such cDNA samples were used for PCR.
  • SUPERSCRIPT II "first strand" buffer 20 ⁇ l of 0.1 M DDT, 10 ⁇ l of a 10 mM of mix of deoxynucleotide triphosphates (dATP
  • a portion of the cDNA (typically, 1/5 of a 20 ⁇ l reaction) was used for PCR.
  • PCR was conducted using primers designed to amplify specifically PDZ domain-containing regions of PDZ proteins of interest. Oligonucleotide primers were designed to amplify one or more PDZ-encoding domains.
  • the DNA sequences encoding the various PDZ domains of interest were identified by inspection (i.e., conceptual translation of the PDZ protein cDNA sequences obtained from GenBank, followed by alignment with the PDZ domain amino acid sequence).
  • TABLE 3 shows the PCR primers, the PDZ-encoded domains amplified, and the GenBank accession number of the PDZ-domain containing proteins.
  • the PCR primers included endonuclease restriction sequences at their ends to allow ligation with pGEX-3X cloning vector (Pharmacia, GenBank XXII 3852 ) in frame with glutathione-S transferase (GST).
  • GST glutathione-S transferase
  • mCASK, KIAA0559, PTN-4, and XI 1 beta were detected in B cells, but not T cells.
  • ⁇ ⁇ Gene names and corresponding gene products are provided.
  • cDNA sequences representing the, same gene have several database entries under different accession numbers and names. Accession numbers shown correspond to the gene .name used in this description, and numbering of nucleotides and amino acids correlates to those Genbank entries.
  • Amino acid sequences shown correspond to the cloned DNA portions of PDZ domain containing genes. Linker amino acid sequences (e.g., amino acids encoded by DNA flanking the cloning site ofthe pGEX-3X cloning vector), are in italics
  • sequence analysis ofthe PDZ clones revealed differences to the DNA and/or protein sequence as publish
  • Two complementary assays tenned "A' and "G,”” were developed to detect binding between a PDZ-domain polypeptide and candidate PDZ ligand.
  • binding is detected between a peptide having a sequence corresponding to the C-terminus of a protein anticipated to bind to one or more PDZ domain (i.e. a candidate PL peptide) and a PDZ-domain polypeptide (typically a fusion protein containing a PDZ domain).
  • the candidate, PL peptide is immobilized and binding of a soluble PDZ-domain polypeptide to the immobilized peptide is detected (the "A”' assay is named for the fact that in one embodiment an avidin surface is used to immobilize the peptide).
  • the PDZ-domain polypeptide is immobilized and binding of a soluble PL peptide is detected (The “G” assay is named for the fact that in one embodiment a GST-binding surface is used to immobilize the PDZ-domain polypeptide).
  • Preferred embodiments of these assays are described in detail infra. However, it will be appreciated by ordinarily skilled practitioners that these assays can be modified in numerous ways while remaining useful for the purposes o the present invention.
  • GST-PDZ domain fusion proteins were prepared for use in the assays of the invention.
  • PCR products containing PDZ encoding domains (as described in ⁇ 6.1 supra) were subcloned into an expression vector to permit expression of fusion proteins containing a PDZ domain and a heterologous domain (i.e., a glutathione-S transferase sequence, "GST")- PCR products (i.e., DNA fragments) representing PDZ domain encoding DNA was extracted from agarose gels using the "sephaglas" gel extraction system (Pharmacia) according to the manufacturer's recommendations.
  • GST glutathione-S transferase sequence
  • PCR primers were designed to include endonuclease restriction sites to facilitate ligation of PCR fragments into a GST gene fusion vector (pGEX-3X; Pharmacia, GenBank accession no. XXU 13852) in-frame with the glutathione-S transferase coding sequence.
  • This vector contains a IPTG inducible lacZ promoter.
  • the pGEX-3X vector was linearized using Bam HI and Eco RI or, in some cases, Eco RI or Sma I, as shown in TABLE 3, and dephosphorylated. For most cloning approaches, double digest with Bam HI and Eco RI was performed, so that the ends ofthe PCR fragments to clone were Bam HI and Eco RI.
  • restriction endonuclease combinations used were Bgl II and Eco RI, Bam HI and Mfe I, or Eco RI only, Sma I only, or BamHI only (see TABLE 3).
  • the DNA portion cloned represents the PDZ domains and the cDNA portion located between individual domains. Precise locations of cloned fragments used in the assays are indicated in TABLE 3. DNA linker sequences between the GST portion and the PDZ domain containing DNA portion vary slightly, dependent on which of the above described cloning sites and approaches were used.
  • the amino acid sequence ofthe GST- PDZ fusion protein varies in the linker region between GST and PDZ domain.
  • Protein linkers sequences corresponding to different cloning sites/approaches are shown below. Linker sequences (vector DNA encoded) are bold, PDZ domain containing gene derived sequences are in italics.
  • the PDZ-encoding PCR fragment and linearized pGEX-3X vector were ethanol precipitated and resuspended in 10 ul standard ligation buffer. Ligation was performed for 4-10 hours at 7°C using T4 DNA ligase. It will be understood that some ofthe resulting constructs include very short linker sequences and that, when multiple PDZ domains were cloned, the constructs included some DNA located between individual PDZ domains.
  • the ligation products were transformed in DH5 ⁇ or BL-21 E.coli bacteria strains.
  • Colonies were screened for presence and identity ofthe cloned PDZ domain containing DNA as well as for correct fusion with the glutathione S-transferase encoding DNA portion by PCR and by sequence analysis. Positive clones were tested in a small scale assay for expression of the GST/PDZ domain fusion protein and, if expressing, these clones were subsequently grown up for large scale preparations of GST/PDZ fusion protein. GST-PDZ domain fusion protein was overexpressed following addition of IPTG to the culture medium and purified. Detailed procedure of small scale and large scale fusion protein expression and purification are described in "GST Gene Fusion System" (second edition, revision 2; published by Pharmacia).
  • a small culture (3-5mls) containing a bacterial strain (DH5 , BL21 or JM109) with the fusion protein construct was grown overnight in LB- media at 37°C with the appropriate antibiotic selection (lOOug/ml ampicillin; a.k.a. LB-amp).
  • the overnight culture was poured into a fresh preparation of LB-amp (typically 250-5 OOmls) and grown until the optical density (OD) ofthe culture was between 0.5 and 0.9 (approximately 2.5 hours).
  • IPTG isopropyl ⁇ -D-thiogalactopyranoside
  • LOmM optical density
  • Bacteria were collect by centrifugation (4500 g) and resuspended in Buffer A- (50mM Tris, pH 8.0, 50mM dextrose, ImM EDTA, 200uM phenylmethylsulfonylfluoride). An equal volume of Buffer A+ (Buffer A-, 4mg/ml lysozyme) was added and incubated on ice for 3 min to lyse bacteria. An equal volume of Buffer B (lOmM Tris, pH 8.0, 50mM KC1, ImM EDTA. 0.5% Tween-20, 0.5% NP40 (a.k.a.
  • IGEPAL CA-630 200uM phenylmethylsulfonylfluoride was added and incubated for an additional 20 min.
  • the bacterial cell lysate was centrifuged (x20,000g), and supernatant was added to glutathione sepharose 4B (Pharmacia, cat no. 17-0765- 01) previous swelled (rehydrated) in IX phosphate-buffered saline (PBS).
  • the supernatant- sepharose slurry was poured into a column and washed with at least 20 bed volumes of IX PBS.
  • GST fusion protein was eluted off the glutathione sepharose by applying 0.5-1.0 ml aliquots of 5mM glutathione and collected as separate fractions. Concentrations of fractions were determined using BioRad Protein Assay (cat no. 500-0006) according to manufacturer's specifications. Those fractions containing the highest concentration of fusion protein were pooled and dialyzed against IX PBS/35% glycerol. Fusion proteins were assayed for size and quality by SDS gel electrophoresis (PAGE) as described in "Sambrook.” Fusion protein aliquots were stored at minus 80°C and at minus 20°C.
  • PDZ domains are known to be bound by the C-terminal residues of PDZ-binding proteins.
  • cell surface receptor proteins were identified and peptides having the sequence corresponding to the c-terminus of each protein were synthesized. TABLE 4 lists these proteins, and provides corresponding C-terminal sequences and GenBank accession numbers. "Clasp 1" is described in WO 00/20434 (published 13 April 2000). "Clasp 2" and “Clasp 4" are described in copending applications USSN 09/547276 (attorney docket No. 20054-000200) and 60/196527 (attorney docket No.20054- 000400), both filed April 11 , 2000.
  • Synthetic peptides of defined sequence can be synthesized by any standard resin-based method (see, e.g., U. S. Pat. No. 4,108,846; see also, Caruthers et al, 1980, Nucleic Acids Res. Symp. Ser., 215-223; Horn et al., 1980, Nucleic Acids Res. Symp. Ser., 225-232; Roberge, et al, 1995, Science 269:202).
  • the peptides used in the assays described herein were prepared by the FMOC (see, e.g., Guy and Fields, 1997, Meth. Enz.
  • peptides were labeled with biotin at the amino-terminus by reaction with a four- fold excess of biotin methyl ester in dimethylsulfoxide with a catalytic amount of base.
  • the peptides were cleaved from the resin using a halide containing acid (e.g. trifluoroacetic acid) in the presence of appropriate antioxidants (e.g. ethanedithiol) and excess solvent lyophilized.
  • peptides can be redissolved and purified by reverse phase high performance liquid chromatography (HPLC).
  • HPLC solvent system involves a Vydac C-18 semi-preparative column running at 5 mL per minute with increasing quantities of acetonitrile plus 0.1% trifluoroacetic acid in a base solvent of water plus 0.1% trifluoroacetic acid.
  • MALDI cation-mode mass spectrometry As noted, exemplary biotinylated peptides are provided in TABLE 4.
  • fusion protein are used in the assays and devices of the invention. Methods for constructing and expressing fusion proteins are well known. Fusion proteins generally are described in Ausubel et al., supra, Kroll et al., 1993, DNA Cell.
  • the fusion protein includes a domain to facilitate immobilization of the protein to a solid substrate ("an immobilization domain").
  • the immobilization domain includes an epitope tag (i.e., a sequence recognized by a antibody, typically a monoclonal antibody) such as polyhistidine (Bush et al, 1991, J Biol Chem 266:13811-14), SEAP (Berger et al, 1988, Gene 66:1-10), or Ml and M2 flag (see, e.g, U.S. Pat. Nos.
  • the immobilization domain is a GST coding region.
  • the protein e.g., fusion protein
  • these are residues naturally associated with the PDZ-domain (i.e., in a particular PDZ-protein) but they may include residues of synthetic (e.g., poly(alanine)) or heterologous origin (e.g., spacers of, e.g., between 10 and 300 residues).
  • PDZ domain-containing polypeptide used in the methods ofthe invention e.g., PDZ fusion proteins
  • a vector e.g., plasmid, phage or phagemid
  • an suitable expression system e.g., a prokaryotic, insect, mammalian, or cell free expression system
  • expression of the protein comprises inserting the coding sequence into an appropriate expression vector (i.e., a vector that contains the necessary elements for the transcription and translation of the inserted coding sequence required for the expression system employed, e.g., control elements including enhancers, promoters, transcription terminators, origins of replication, a suitable initiation codon (e.g., methionine), open reading frame, and translational regulatory signals (e.g., a ribosome binding site, a termination codon and a polyadenylation sequence.
  • control elements including enhancers, promoters, transcription terminators, origins of replication, a suitable initiation codon (e.g., methionine), open reading frame, and translational regulatory signals (e.g., a ribosome binding site, a termination codon and a polyadenylation sequence.
  • a suitable initiation codon e.g., methionine
  • open reading frame e.g., open reading frame
  • translational regulatory signals e.g
  • the coding sequence of the fusion protein includes a PDZ domain and an immobilization domain as described elsewhere herein.
  • Polynucleotides encoding the amino acid sequence for each domain can be obtained in a variety of ways known in the art; typically the polynucleotides are obtained by PCR amplification of cloned plasmids, cDNA libraries, and cDNA generated by reverse transcription of RNA, using primers designed based on sequences determined by the practitioner or, more often, publicly available (e.g., through GenBank).
  • the primers include linker regions (e.g., sequences including restriction sites) to facilitate cloning and manipulation in production of the fusion construct.
  • the polynucleotides corresponding to the PDZ and immobilization regions are joined in-frame to produce the fusion protein-encoding sequence.
  • the fusion proteins ofthe invention may be expressed as secreted proteins (e.g., by including the signal sequence encoding DNA in the fusion gene; see, e.g., Lui et al, 1993, PNAS USA, 90:8957-61) or as nonsecreted proteins.
  • the PDZ-containing proteins are immobilized on a solid surface.
  • the substrate to which the polypeptide is bound may in any of a variety of forms, e.g., a microtiter dish, a test tube, a dipstick, a miorocentrifuge tube, a bead, a spinnable disk, and the like.
  • Suitable materials include glass, plastic (e.g., polyethylene, PVC, polypropylene, polystyrene, and the like), protein, paper, carbohydrate, lipip monolayer or supported lipid bilayer, and other solid supports.
  • Other materials that may be employed include ceramics, metals, metalloids, semiconductive materials, cements and the like.
  • the fusion proteins are organized as an array.
  • array refers to an ordered arrangement of immobilized fusion proteins, in which particular different fusion proteins (i.e., having different PDZ domains) are located at different predetermined sites on the substrate. Because the location of particular fusion proteins on the array is known, binding at thai location can be correlated with binding to the PDZ domain situated at that location. Immobilization of fusion proteins on beads (individually or in groups) is another particularly useful approach. In one embodiment, individual fusion proteins are immobilized on beads. In one embodiment, mixtures of distinguishable beads are used.
  • Distinguishable beads are beads that can be separated from each other on the basis of a property such as size, magnetic property, color (e.g., using FACS) or affinity tag (e.g., a bead coated with protein A can be separated from a bead not coated with protein A by using IgG affinity methods). Binding to particular PDZ domain may be determined; similarly, the effect of test compounds (i.e., agonists and antagonists of binding) may be determined. Methods for immobilizing proteins are well known in the art, and include covalent and non-covalent methods.
  • One suitable immobilization method is antibody-mediated immobilization.
  • an antibody specific for the sequence of an "immobilization domain" ofthe PDZ-domain containing protein is itself immobilized on the substrate (e.g., by adsorption).
  • One advantage of this approach is that a single antibody may be adhered to the substrate and used for immobilization of a number of polypeptides (sharing the same immobilization domain).
  • an immobilization domain consisting of poly-histidine (Bush et al, 1991, J.
  • Biol Chem 266: 13811-14 can be bound by an anti-histidine monoclonal antibody (R&D Systems, Minneapolis, MN); an immobilization domain consisting of secreted alkaline phosphatase (“SEAP") (Berger et al, 1988, Gene 66:1-10) can be bound by anti-SEAP (Sigma Chemical Company, St. Louis, MO); an immobilization domain consisting of a FLAG epitope can be bound by anti-FLAG.
  • SEAP secreted alkaline phosphatase
  • SEAP secreted alkaline phosphatase
  • SEAP secreted alkaline phosphatase
  • FLAG epitope can be bound by anti-FLAG.
  • Other ligand-antiligand immobilization methods are also suitable (e.g., an immobilization domain consisting of protein A sequences (Harlow and Lane, 1988, Antibodies A laboratory Manual, Cold Spring Harbor Laboratory; Sigma Chemical Co., St.
  • the immobilization domain is a GST moiety, as described herein.
  • glass and plastic are especially useful substrates.
  • the substrates may be printed with a hydrophobic (e.g., Teflon) mask to form wells.
  • Preprinted glass slides with 3, 10 and 21 wells per 14.5 cm 2 slide "working area" are available from, e.g., SPI Supplies, West Chester, PA; also see U.S.Pat. No. 4,011,350).
  • a large format (12.4 cm x 8.3 cm) glass slide is printed in a 96 well format is used; this format facilitates the use of automated liquid handling equipment and utilization of 96 well format plate readers of various types (fluorescent, colorimetric, scintillation).
  • higher densities may be used (e.g., more than 10 or 100 polypeptides per c ⁇ r). See, e.g., MacBeath et al, 2000, Science 289: 1760-63.
  • antibodies are bound to substrates (e.g., glass substrates) by adsorption.
  • Suitable adsorption conditions are well known in the art and include incubation of 0.5-50ug/ml (e.g., 10 ug/ml) mAb in buffer (e.g., PBS, or 50 to 300 mM Tris, MOPS, HEPES, PIPES, acetate buffers, pHs 6.5 to 8, at 4°C) to 37°C and from lhr to more than 24 hours.
  • buffer e.g., PBS, or 50 to 300 mM Tris, MOPS, HEPES, PIPES, acetate buffers, pHs 6.5 to 8, at 4°C
  • Proteins may be covalently bound or noncovalently attached through nonspecific bonding. If covalent bonding between a the fusion protein and the surface is desired, the surface will usually be polyfunctional or be capable of being polyfunctionalized.
  • Functional groups which may be present on the surface and used for linking can include carboxylic acids, aldehydes, amino groups, cyano groups, ethylenic groups, hydroxyl groups, mercapto groups and the like.
  • the manner of linking a wide variety of compounds to various surfaces is well known and is amply illustrated in the literature.
  • the invention provides an assay in which biotinylated candidate PL peptides are immobilized on an avidin coated surface. The binding of PDZ-domain fusion protein to this surface is then measured.
  • the PDZ-domain fusion protein is a GST/PDZ fusion protein and the assay is carried out as follows:
  • Avidin is bound to a surface, e.g. a protein binding surface.
  • avidin is bound to a polystyrene 96 well plate (e.g., Nunc Polysorb (cat #475094) by addition of 100 uL per well of 20 ug/mL of avidin (Pierce) in phosphate buffered saline without calcium and magnesium, pH 7.4 ("PBS", GibcoBRL) at 4°C for 12 hours.
  • PBS phosphate buffered saline without calcium and magnesium, pH 7.4
  • the plate is then treated to block nonspecific interactions by addition of 200 uL per well of PBS containing 2 g per 100 mL protease-free bovine serum albumin ("PBS/BSA”) for 2 hours at 4°C.
  • PBS/BSA protease-free bovine serum albumin
  • Biotinylated PL peptides (or candidate PL peptides, e.g. see TABLE 4) are immobilized on the surface of wells of the plate by addition of 50 uL per well of 0.4 uM peptide in PBS/BSA for 30 minutes at 4°C.
  • each different peptide is added to at least eight different wells so that multiple measurements (e.g. duplicates and also measurements using different (3ST/PDZ-domain fusion proteins and a GST alone negative control) can be made, and also additional negative control wells are prepared in which no peptide is immobilized.
  • the plate is washed 3 times with PBS.
  • GST/PDZ-domain fusion protein (prepared as described supra) is allowed to react with the surface by addition of 50 uL per well of a solution containing 5 ug/mL GST/PDZ-domain fusion protein in PBS/BSA for 2 hours at 4°C.
  • GST alone i.e. not a fusion protein
  • specified wells generally at least 2 wells (i.e. duplicate measurements) for each immobilized peptide. After the 2 hour reaction, the plate is washed 3 times with PBS to remove unbound fusion protein.
  • the binding of the GST/PDZ-domain fusion protein to the avidin- biotinylated peptide surface can be detected using a variety of methods, and detectors known in the art.
  • 50 uL per well of an anti-GST antibody in PBS/BSA e.g. 2.5 ug/mL of polyclonal goat-anti-GST antibody, Pierce
  • PBS/BSA polyclonal goat-anti-GST antibody, Pierce
  • HRP horseradish peroxidase
  • TMB horseradish peroxidase-conjugated polyclonal rabbit anti-goat inununoglobulin antibody
  • a specific or selective reaction will be at least twice background signal, more typically more than 5 times background, and most typically 10 or more times the background signal.
  • a statistically significant reaction will involve multiple measurements of the reaction with the signal and the background differing by at least two standard errors, more typically four standard errors, and most typically six or more standard errors.
  • a statistical test e.g. a T-test
  • comparing repeated measurements ofthe signal with repeated measurements of the background will result in a p-value ⁇ 0.05, more typically a p-value ⁇ 0.01, and most typically a p-value ⁇ 0.001 or less.
  • the signal from binding of a GST/PDZ-domain fusion protein to an avidin surface not exposed to (i.e. not covered with) the PL peptide is one suitable negative control (sometimes referred to as "B").
  • the signal from binding of GST polypeptide alone (i.e. not a fusion protein) to an avidin-coated surface that has been exposed to (i.e. covered with) the PL peptide is a second suitable negative control (sometimes referred to as "B2"). Because all measurements are done in multiples (i.e.
  • the arithmetic mean (or, equivalently, average) of several measurements is used in determining the binding, and the standard error ofthe mean is used in determining the probable error in the measurement of the binding.
  • the standard error of the mean of N measurements equals the square root of the following: the sum of the squares of the difference between each measurement and the mean, divided by the product of ( f) and (N-l).
  • specific binding ofthe PDZ protein to the plate-bound PL peptide is determined by comparing the mean signal ("mean S") and standard error of the signal (“SE”) for a particular PL-PDZ combination with the mean Bl and/or mean B2.
  • binding was detected to be specific (denoted by an "A" in the matrix) when (1) the, mean S was at least twice the mean Bl and at least twice the mean B2 and (2) the mean S was at least six standard errors (six SE) greater than both the mean Bl and the mean B2.
  • an additional criterion was used to ensure that none ofthe interactions defined as specific arose from a combined tendency of both the particular PDZ fusion protein and PL peptide tested to each give a higher than usual background.
  • This criteria was that (3) the mean S was at least twenty times the product ofthe mean B 1 and the mean B2. The factor twenty times reflects that at least one of Bl and B2 is generally less than 0.1 O.D.
  • the invention provides an assay in which a GST/PDZ fusion protein is immobilized on a surface ("G" assay). The binding of labeled PL peptide (as listed in TABLE 4) to this surface is then measured.
  • the assay is carried out as follows:
  • a PDZ-domain polypeptide is bound to a surface, e.g. a protein binding surface.
  • a GST/PDZ fusion protein containing one or more PDZ domains is bound to a polystyrene 96-well plate.
  • the GST/PDZ fusion protein can be bound to the plate by any of a variety of standard methods known to one of skill in the art, although some care must be taken that the process of binding the fusion protein to the plate does not alter the ligand-binding properties ofthe PDZ domain.
  • the GST/PDZ fusion protein is bound via an anti-GST antibody that is coated onto the 96-well plate. Adequate binding to the plate can be achieved when: a.
  • Biotinylated PL peptides (or candidate PL peptides, e.g. as shown in TABLE 4) are allowed to react with the surface by addition of 50 uL per well of 20 uM solution of the biotinylated peptide in PBS/BSA for 10 minutes at 4°C, followed by an additional 20 minute incubation at 25°C. The plate is washed 3 times with ice cold PBS.
  • the binding of the biotinylated peptide to the GST/PDZ fusion protein surface can be detected using a variety of methods and detectors known to one of skill in the art.
  • 100 uL per well of 0.5 ug/mL streptavidin-horse radish peroxidase (HRP) conjugate dissolved in BSA/PBS is added and allowed to react for 20 minutes at 4°C.
  • the plate is then washed 5 times with 50 mM Tris pH 8.0 containing 0.2% Tween 20, and developed by addition of 100 uL per well of HRP-substrate solution (TMB, Dako) for 20 minutes at room temperature (RT).
  • TMB HRP-substrate solution
  • RT room temperature
  • the reaction ofthe HRP and its substrate is terminated by addition of 100 uL per well of I M sulfuric acid, and the optical density (O.D.) of each well of the plate is read at 450 um.
  • Specific binding of a PL peptide and a PDZ domain polypeptide is determined by comparing the signal from the well(s) in which the PL peptide and PDZ domain polypeptide are combined, with the background signal(s).
  • the background signal is the signal found in the negative control(s).
  • a specific or selective reaction will be at least twice background signal, more typically more than 5 times background, and most typically 10 or more times the background signal.
  • a statistically significant reaction will involve multiple measurements of the reaction with the signal and the background differing by at least two standard errors, more typically four standard errors, and most typically six or more standard errors.
  • a statistical test e.g.
  • the signal from binding of a given PL peptide to immobilized (surface bound) GST polypeptide alone is one suitable negative control (sometimes referred to as "B 1"). Because all measurement are done in multiples (i.e. at least duplicate) the arithmetic mean (or, equivalently, average.) of several measurements is used in determining the binding, and the standard error ofthe mean is used in determining the probable error in the measurement ofthe binding.
  • the standard error of the mean ofN measurements equals the square root ofthe following: the sum of the squares of the difference between each measurement and the mean, divided by the product of (N) and (N-l).
  • specific binding ofthe PDZ protein to the platebound peptide is determined by comparing the mean signal ("mean S") and standard error of the signal ("SE") for a particular PL-PDZ combination with the mean Bl .
  • mean S mean signal
  • SE standard error of the signal
  • the PDZ-PL detection assays can employ a variety of surfaces to bind the PL and PDZ-containing proteins.
  • a surface can be an "assay plate" which is formed from a material (e.g. polystyrene) which optimizes adherence of either the PL protein or PDZ- containing protein thereto.
  • the individual wells of the assay plate will have a high surface area to volume ratio and therefore a suitable shape is a flat bottom well (where the proteins ofthe assays are adherent).
  • Other surfaces include, but are not limited to, polystyrene or glass beads, polystyrene or glass slides, and alike.
  • the assay plate can be a "microtiter" plate.
  • microtiter plate when used herein refers to a multiwell assay plate, e.g., having between about 30 to 200 individual wells, usually 96 wells. Alternatively, high density arrays can be used. Often, the individual wells of the microtiter plate will hold a maximum volume of about 250 ul.
  • the assay plate is a 96 well polystyrene plate (such as that sold by Becton Dickinson Labware, Lincoln Park, N.J.), which allows for automation and high throughput screening. Other surfaces include polystyrene microtiter ELISA plates such as that sold by Nunc Maxisorp, Inter Med, Denmark. Often, about 50 ul to 300 ul, more preferably 100 ul to 200 ul, of an aqueous sample comprising buffers suspended therein will be added to each well of the assay plate.
  • the detectable labels of the invention can be any detectable compound or composition which is conjugated directly or indirectly with a molecule (such as described above).
  • the label can be detectable by itself (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, can catalyze a chemical alteration of a substrate compound or composition which is detectable.
  • the preferred label is an enzymatic one which catalyzes a color change of a non-radioactive color reagent.
  • the label is indirectly conjugated with the antibody.
  • the antibody can be conjugated with biotin and any of the categories of labels mentioned above can be conjugated with avidin, or vice versa (see also "A” and "G” assay above). Biotin binds selectively to avidin and thus, the label can be conjugated with the antibody in this indirect manner. See, Ausubel, supra, for a review of techniques involving biotin-avidin conjugation and similar assays.
  • the antibody is conjugated with a small hapten (e.g. digoxin) and one ofthe different types of labels mentioned above is conjugated with an anti-hapten antibody (e.g. anti-digoxin antibody).
  • an anti-hapten antibody e.g. anti-digoxin antibody
  • washing is meant exposing the solid phase to an aqueous solution (usually a buffer or cell culture media) in such a way that unbound material (e.g., non-adhering cells, non-adhering capture agent, unbound ligand, receptor, receptor construct, cell lysate, or HRP antibody) is removed therefrom.
  • a detergent e.g., Triton X
  • the aqueous washing solution is decanted from the wells of the assay plate following washing. Conveniently, washing can be achieved using an automated washing device.
  • washing steps e.g., between about 1 to 10 washing steps.
  • blocking buffer refers to an aqueous, pH buffered solution containing at least one blocking compound which is able to bind to exposed surfaces of the substrate which are not coated with a PL or PDZ- containing protein.
  • the blocking compound is normally a protein such as bovine serum albumin (BSA), gelatin, casein or milk powder and does not cross-react with any of the reagents in the assay.
  • BSA bovine serum albumin
  • the block buffer is generally provided at a pH between about 7 to 7.5 and suitable buffering agents include phosphate and TRIS.
  • enzyme-substrate combinations can also be utilized in detecting PDZ-PL interactions.
  • enzyme-substrate combinations include, for example: (i) Horseradish peroxidase (HRPO) with hydrogen peroxidase as a substrate, wherein the hydrogen peroxidase oxidizes a dye precursor (e.g. orthophenylene diamine [OPD] or 3,3',5,5'-tetramethyl benzidine hydrochloride [TMB]) (as described above).
  • HRPO Horseradish peroxidase
  • OPD orthophenylene diamine
  • TMB 3,3',5,5'-tetramethyl benzidine hydrochloride
  • alkaline phosphatase (AP) with para-Nitrophenyl phosphate as chromogenic substrate (ii) alkaline phosphatase (AP) with para-Nitrophenyl phosphate as chromogenic substrate.
  • ⁇ -D-galactosidase ( ⁇ D-Gal) with a chromogenic substrate (e.g. p- nitrophenyl- ⁇ -D-galactosidase) or fluorogenic substrate 4-methylumbelliferyl- ⁇ -D- galactosidase.
  • a chromogenic substrate e.g. p- nitrophenyl- ⁇ -D-galactosidase
  • fluorogenic substrate 4-methylumbelliferyl- ⁇ -D- galactosidase
  • TABLE 2 shows the results of assays in which specific binding was detected using the "A” and "G” assays described herein.
  • the top row of the table specifies the source of the PDZ domain used in the GST-PDZ fusion proteins (see TABLE 3).
  • the first column lists the cell surface proteins from which C-terminal peptide sequences were derived and the second column ("code") identifies the peptide used in the assay (see TJ BLE 4).
  • the third column, "Seq” provides the sequence of the four (4) C-terminal residues of the cell surface protein and peptide.
  • A indicates specific binding as detected in the "A” assay.
  • “G” indicates specific binding as shown in the "G” assay.
  • a blank indicates that no specific binding was detected using the "A” or “G” assays.
  • An asterisk (*) indicates that a pairwise interaction between the PDZ protein and the cell surface protein (or subdomains of either) has been described by others.
  • MATRIX to detect binding between PDZ proteins and candidate PL peptides.
  • a number of specific PDZ-PL interactions are identified by the MATRIX and key amino acids and positions important in PDZ binding ("PL motifs") are deduced from these results.
  • the assay can further aid in the rapid discovery and characterization of novel PL proteins and PL motifs to help in rational drug design and synthesis of PL-PDZ interaction inhibitors.
  • PL motifs important in PDZ binding e.g., the C-terminal motifs S/T-X-V/I/L (for DLGl) and Y/F-Y/F-I/L/F for MPPl (see, Doyle et al, 1996, Cell 85, 1067; Songyang et al., 1997, Science 275, 73).
  • the reported motifs are not sufficiently specific (i.e. a large number of proteins meet these criteria yet are not necessarily actual PDZ ligands) and cover only a small number of PDZ proteins (approximately 10).
  • the PRISM MATRIX can be used to determine ligand specificity and to deduce ligand binding motifs for any PDZ protein because it can precisely determine sequences of amino acids that do or do not result in specific PDZ binding.
  • the assay has revealed a significant of new PDZ domain binding motifs (i.e.
  • PL motifs C-terminal sequence of CD6, ISAA; C- terminal sequence of CD49E, TSDA; C- terminal sequence of CD49F, TSDA; C-terminal sequence of Clasp-1, SAEV; C- terminal sequence of CLASP-4, YAEV; C- terminal sequence of CD44, KIGV; C-terminal sequence of IL5R, DSVF; C-terminal sequence of BLR-1, LTTF.
  • PDZ-PL PDZ-PL combinations that result in specific binding interactions
  • negative results i.e. PDZ-PL combinations that do not result in specific binding
  • CD6 the C-terminal sequence of CD6, SAA and the C-terminal sequence of CD49E, SDA bind to the PDZ-domain polypeptide 41.8 while the related C-terminal sequence of CD166, TEA and C- terminal sequence of CD148, YIA do not.
  • Other novel motifs are described in TABLE 5A.
  • X* is any non-aromatic amino acid (any residue other than T, F or W).
  • TABLE 2 is arranged so that both the PDZ/GST fusion proteins and the PL peptide ligands are ordered with structurally similar molecules close to each other.
  • the PDZ domains of each of the GST-PDZ fusion proteins were ranked for amino acid sequence similarity using the CLUSTAL multiple sequence alignment software package. Proteins with greatest sequence similarity are closest to each other in the matrix.
  • the PL peptide ligands were also ordered on the basis of amino acid similarity, but with weight given to residues reported to be important in PDZ binding (Doyle et al., 1996, Cell 85, 1067).
  • peptides were first ordered based on the most C-terminal residue (zero position) in the following amino acid order: G, A, C, S, T, N, Q, D, E, H, K, R, V, I, L, M, P, F, Y, W.
  • the same ranking scheme was then applied to the next most important residue for peptide binding, the -2 position, followed by the —1 position and the -3 position.
  • the GST- PDZ fusions can also be arranged to give additional weight to residues known to be important for ligand binding when aligning the proteins.
  • Regions of the TABLE 1 matrix that are densely filled with significant binding interactions indicate that a particular ligand structure tends to bind to a particular family of PDZ domains.
  • the results indicate that PDZ domains with similar structures bind ligands with similar structures, as indicated by the presence in the matrix of some clusters of many significant binding interactions and other areas of few significant binding interactions.
  • These results reveal a number of structural relationships between different PDZ proteins and their ligands. The understanding of these structural relationships can be used to scan databases for probable ligands of specific PDZ proteins, facilitating the design of inhibitors of such novel interactions and the prediction o the side-effect profile of such inhibitors.
  • DLGl is closely related to two other proteins, PSD95 and NeDLG. Each of these three proteins binds ligands that terminate in the motif S/T/Y - X- V/I/L, as previously described (Songyang et al, 1997, Science 275, 73). Interestingly, if the terminal ligand residue is valine, other previously unidentified residues at the -2 position are compatibile with binding, with the C-terminal sequences AEV and ELV resulting in significant binding events.
  • ligands that bind specifically to KIAA0561 fit the C-terminal motif X* - S/T - D E - V/I/L where X* is any non-aromatic amino acid.
  • one of these ligands also binds to KIAA 0561 's closest relative, TAX2.
  • the two ligands that bind to prIL16 both have a charged amino acid at the -3 position, a hydrophobic amino acid at the - 2 position, and valine at the terminal position.
  • a final interesting example of defining the ligand specificity of a PDZ domain involves PTN-4.
  • X* is any non-aromatic amino acid.
  • the "A” and “G” assays of the invention can be used to determine the "apparent affinity" of binding of a PDZ ligand peptide to a PDZ-domain polypeptide.
  • Apparent affinity is determined based on the concentration of one molecule required to saturate the binding of a second molecule (e.g., the binding of a ligand to a receptor).
  • Two particularly useful approaches for quantitation of apparent affinity of PDZ-ligand binding are provided infra.
  • a GST/PDZ fusion protein, as well as GST alone as a negative control, are bound to a surface (e.g., a 96-well plate) and the surface blocked and washed as described supra for the "G” assay.
  • a surface e.g., a 96-well plate
  • 50 uL per well of a solution of biotinylated PL peptide e.g. as shown in TJABLE 4
  • PBS/BSA e.g. at 0.1 uM, 0.33 uM, 1 uM, 3.3 uM, 10 uM, 33 uM, and 100 uM.
  • the PL peptide is allowed to react with the bound GST/PDZ fusion protein (as well as the GST alone negative control) for 10 minutes at 4 C followed by 20 minutes at 25 C.
  • the plate is washed 3 times with ice cold PBS to remove unbound labeled peptide.
  • the net binding signal is determined by subtracting the binding of the peptide to GST alone from the binding of the peptide to the
  • FIGURE 2 shows vaiying concentrations of biotinylated CLASP-2 (FIG. 2A) or Fas (FIG. 2B). C-terminal peptides reacted with immobilized (plate bound) GST polypeptide or GST/PDZ fusion proteins (GST/DLG1 , GST/NeDLG, and GDT/PSD95) in duplicate.
  • a fixed concentration of a PDZ-domain polypeptide and increasing concentrations of a labeled PL peptide are mixed together in solution and allowed to react.
  • preferred peptide concentrations are 0.1 uM, 1 uM, 10 uM, 100 uM, 1 mM.
  • appropriate reaction times can range from 10 minutes to 2 days at temperatures ranging from 4 C to 37 C.
  • the identical reaction can also be carried out using a non-PDZ domain-containing protein as a control (e.g., if the PDZ- domain polypeptide is fusion protein, the fusion partner can be used).
  • PDZ-ligand complexes can be separated from unbound labeled peptide using a variety of methods known in the art. For example, the complexes can be separated using higb performance size-exclusion chromatography (HPSEC, gel filtration) (Rabinowitz et al., 1998, Immunity 9:699), affinity chromatography(e.g. using glutathione sepharose beads), and affinity absorption (e.g., by binding to an anti-GST-coated plate as described supra).
  • higb performance size-exclusion chromatography HPSEC, gel filtration
  • affinity chromatography e.g. using glutathione sepharose beads
  • affinity absorption e.g., by binding to an anti-GST-coated plate as described supra.
  • the PDZ-ligand complex is detected based on presence ofthe label on the peptide ligand using a variety of methods and detectors known to one of skill in the art. For example, if the label is fluorescein and the separation is achieved using HPSEC, an in-line fluorescence detector can be used. The binding can also be detected as described supra for the G assay. (4) The PDZ-ligand binding signal is plotted as a function of ligand concentration and the plot is fit.
  • Measurement ofthe affinity of a labeled peptide ligand binding to a PDZ-domain polypeptide n is useful because knowledge ofthe affinity (or apparent affinity) of this interaction allows rational design of inhibitors of the interaction with known potency (See EXAMPLE 2).
  • the potency of inhibitors in inhibition would be similar to (i.e. within one-order of magnitude of) the apparent affinity of the labeled peptide ligand binding to the PDZ-domain.
  • the invention provides a method of determining the apparent affinity of binding between a PDZ domain and a ligand by immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface, contacting the immobilized polypeptide with a plurality of different concentrations ofthe ligand, determining the amount of binding ofthe ligand to the immobilized polypeptide at each o the concentrations of ligand, and calculating the apparent affinity of the binding based on that data.
  • the polypeptide comprising the PDZ domain and a non-PDZ domain is a fusion protein.
  • the e.g., fusion protein is GST-PDZ fusion protein, but other polypeptides can also be used (e.g., a fusion protein including a PDZ domain and any of a variety of epitope tags, biotinylation signals and the like) so long as the polypeptide can be immobilized in an orientation that does not abolish the ligand binding properties of the PDZ domain, e.g, by tethering the polypeptide to the surface via the non-PDZ domain via an anti-domain antibody and leaving the PDZ domain as the free end. It was discovered, for example, reacting a PDZ-GST fusion polypeptide directly to a plastic plate provided suboptimal results. The calculation of binding affinity itself can be determined using any suitable equation (e.g., as shown supra; also see Cantor and Schimmel (1980) BIOPHYSICAL CHEMISTRY WH Freeman & Co., San Francisco) or software.
  • any suitable equation e.g., as shown supra; also see Can
  • the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain (e.g., an anti- GST antibody when a GST-PDZ fusion polypeptide is used).
  • an immobilized immunoglobulin that binds the non-PDZ domain e.g., an anti- GST antibody when a GST-PDZ fusion polypeptide is used.
  • the step of contacting the ligand and PDZ-domain polypeptide is carried out under the conditions provided supra in the description o the "G" assay. It will be appreciated that binding assays are conveniently carried out in multiwell plates (e.g., 24-well, 96-well plates, or 384 well plates).
  • the present method has considerable advantages over other methods for measuring binding affinities PDZ-PL affinities, which typically involve contacting varying concentrations of a GST-PDZ fusion protein to a ligand-coated surface.
  • affinity e.g., using immobilized ligand and GST- PDZ protein in solution
  • the assays described supra and other assays can also be used to identify the binding of other molecules (e.g., peptide mimetics, small molecules, and the like) to PDZ domain sequences.
  • other molecules e.g., peptide mimetics, small molecules, and the like
  • combinatorial and other libraries of compounds can be screened, e.g., for molecules that specifically bind to PDZ domains in hematopoietic cells. Screening of libraries can be accomplished by any of a variety of commonly known methods. See, e.g., the following references, which disclose screening of peptide libraries: Parmley and Smith, 1989, Adv. Exp. Med. Biol.
  • screening can be carried out by contacting the library members with a hematopoietic cell PDZ-domain polypeptide immobilized on a solid support (e.g. as described supra in the "G” assay) and harvesting those library members that bind to the protein.
  • a solid support e.g. as described supra in the "G” assay
  • panning techniques are described by way of example in Parmley and Smith, 1988, Gene 73:305-318; Fowlkes et al., 1992, BioTechniques 13:422-427; PCT Publication No. WO 94/18318; and in references cited hereinabove.
  • the two-hybrid system for selecting interacting proteins in yeast can be used to identify molecules that specifically bind to a PDZ domain- containing protein. Furthermore, the identified molecules are further tested for their ability to inhibit transmembrane receptor interactions with a PDZ domain.
  • antagonists of an interaction between a PDZ protein and a PL protein are identified.
  • a modification o the "A" assay described supra is used to identify antagonists.
  • a modification ofthe "G” assay described supra is used to identify antagonists.
  • screening assays are used to detect molecules that specifically bind to PDZ domains in hematopoietic cells.
  • Such molecules are useful as agonists or antagonists of PDZ-protein-mediated cell function (e.g., cell activation, e.g., T cell activation, vesicle transport, cytokine release, growth factors, transcriptional changes, cytoskeletin rearrangement, cell movement, chemotaxis, and the like).
  • cell activation e.g., T cell activation, vesicle transport, cytokine release, growth factors, transcriptional changes, cytoskeletin rearrangement, cell movement, chemotaxis, and the like.
  • such assays are performed to screen for leukocyte activation inhibitors for drug development.
  • the invention thus provides assays to detect molecules that specifically bind to PDZ domain-containing proteins in hematopoietic cells.
  • recombinant cells expressing PDZ domain-encoding nucleic acids can be used to produce PDZ domains in these assays and to screen for molecules that bind to the domains.
  • Molecules are contacted with the PDZ domain (or fragment thereof) under conditions conducive to binding, and then molecules that specifically bind to such domains are identified.
  • Methods that can be used to carry out the foregoing are commonly known in the art.
  • antagonists are identified by conducting the A or G assays in the presence and absence of a known or candidate antagonist. When decreased binding is observed in the presence of a compound, that compound is identified as an antagonist. Increased binding in the presence of a compound signifies that the compound is an agonist.
  • a test compound can be identified as an inhibitor
  • the PL peptide comprises an amino acid sequence substantially identical to the C-terminal sequence of a PL protein (e.g., CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD4, CD97, Neurexin, CD3n, DOCK2, CD34, FceRIb, or FasLigand).
  • a PL protein e.g., CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD4, CD97, Neurexin, CD3n, DOCK2, CD34, FceRIb, or FasLigand.
  • the "G” assay is used in the presence or absence of an candidate inhibitor. In one embodiment, the "A” assay is used in the presense or absence of a candiate inhibitor.
  • one or more PDZ domain- containing GST-fusion proteins are bound to the surface of wells of a 96-well plate as described supra (with appropriate controls including nonfusion GST protein). All fusion proteins are bound in multiple wells so that appropriate controls and statistical analysis can be done.
  • a test compound in BSA/PBS (typically at multiple different concentrations) is added to wells.
  • a detectably labeled (e.g., biotinylated) peptide known to bind to the relevant PDZ domain is added in each of the wells at a final concentration of, e.g., between about 2 uM and about 40 uM, typically 5 uM, 15 uM, or 25 uM.
  • This mixture is then allowed to react with the PDZ fusion protein bound to the surface for 10 minutes at 4°C followed by 20 minutes at 25°C.
  • the surface is washed free of unbound peptide three times with ice cold PBS and the amount of binding of the peptide in the presence and absence ofthe test compound is determined.
  • the level of binding is measured for each set of replica wells (e.g. duplicates) by subtracting the mean GST alone background from the mean ofthe raw measurement of peptide binding in these wells.
  • the A assay is carried out in the presence or absence of an test candidate to identify inhibitors of PL-PDZ interactions.
  • a test compound is determined to be a specific inhibitor of the binding of the PDZ domain (P) and a PL (L) sequence when, at a test compound concentration of less than or equal to 1 mM (e.g., less than or equal to: 500 uM, 100 uM, 10 uM, 1 uM, 100 nM or 1 nM) the binding of P to L in the presence of the test compound less than about 50% of the binding in the absence of the test compound, (in various embodiments, less than about 25%, less than about 10%, or less than about 1%).
  • the net signal of binding of P to L in the presence ofthe test compound plus six (6) times the standard error ofthe signal in the presence ofthe test compound is less than the binding signal in the absence ofthe test compound.
  • assays for an inhibitor are carried out using a single PDZ protein-PL protein pair (e.g., a PDZ domain fusion protein and a PL peptide).
  • the assays are carried out using a plurality of pairs, such as a plurality of different pairs listed in TABLE 2.
  • antagonists can be identified by carrying out a series of assays using a candidate inhibitor and different PL-PDZ pairs (e.g., as shown in the matrix of TABLE 2) and comparing the results of the assays. All such pairwise combinations are contemplated by the invention (e.g., test compound inhibits binding of PL, to PDZ, to a greater degree than it inhibits binding of PL, to PDZ 2 or PL 2 to PDZ 2 ).
  • test compound inhibits binding of PL, to PDZ, to a greater degree than it inhibits binding of PL, to PDZ 2 or PL 2 to PDZ 2 ).
  • inhibitors with different specificities can readily be designed.
  • the Ki (“potency") of an inhibitor of a PDZ-PL interaction can be determined.
  • Ki is a measure of the concentration of an inhibitor required to have a biological effect.
  • administration of an inhibitor of a PDZ-PL interaction in an amount sufficient to result in an intracellular inhibitor concentration of at least between about 1 and about 100 Ki is expected to inhibit the biological response mediated by the target PDZ-PL interaction.
  • the Kd measurement of PDZ-PL binding as determined using the methods supra is used in determining Ki.
  • the invention provides a method of determining the potency (Ki) of an inhibitor or suspected inhibitor of binding between a PDZ domain and a ligand by immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface, contacting the immobilized polypeptide with a plurality of different mixtures of the ligand and inhibitor, wherein the different mixtures comprise a fixed amount of ligand and different concentrations of the inhibitor, determining the amount of ligand bound at the different concentrations of inhibitor, and calculating the Ki ofthe binding based on the amount of ligand bound in the presence of different concentrations of the inhibitor.
  • the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain.
  • This method which is based on the "G” assay described supra, is particularly suited for high-throughput analysis of the Ki for inhibitors of PDZ-ligand interactions. Further, using this method, the inhibition of the PDZ-ligand interaction itself is measured, without distortion of measurements by avidity effects. Typically, at least a portion of the ligand is detectably labeled to permit easy quantitation of ligand binding.
  • the concentration of ligand and concentrations of inhibitor are selected to allow meaningful detection of inhibition.
  • the concentration ofthe ligand whose binding is to be blocked is close to or less than its binding affinity (e.g., preferably less than the 5x Kd ofthe interaction, more preferably less than 2x Kd, most preferably less than lx Kd).
  • the ligand is typically present at a concentration of less than 2 Kd (e.g., between about 0.01 Kd and about 2 Kd) and the concentrations of the test inhibitor typically range from 1 nM to 100 uM (e.g. a 4-fold dilution series with highest concentration 10 uM or 1 mM).
  • the Kd is determined using the assay disclosed supra.
  • an enhancer (sometimes referred to as, augmentor or agonist) of binding between a PDZ domain and a ligand is identified by immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface, contacting the immobilized polypeptide with the ligand in the presence of a test agent and determining the amount of ligand bound, and comparing the amount of ligand bound in the presence ofthe test agent with the amount of ligand bound by the polypeptide in the absence of the test agent.
  • At least two-fold (often at least 5-fold) greater binding in the presence ofthe test agent compared to the absence of the test agent indicates that the test agent is an agent that enhances the binding of the PDZ domain to the ligand.
  • agents that enhance PDZ-ligand interactions are useful for disruption (dysregulation) of biological events requiring normal PDZ-ligand function (e.g., cancer cell division and metastasis, and activation and migration of immune cells).
  • the invention also provides methods for determining the "potency" or "K en , )a ⁇ cer " of an enhancer of a PDZ- ligand interaction.
  • the K,, nhancer of an enhancer of a PDZ-PL interaction can be determined, e.g., using the Kd of PDZ-PL binding as determined using the methods described supra.
  • K enha ⁇ cer is a measure ofthe concentration of an enhancer expected to have a biological effect.
  • administration of an enhancer of a PDZ-PL interaction in an amount sufficient to result in an intracellular inhibitor concentration of at least between about 0.1 and about 100 K e ⁇ l , ⁇ ancer (e.g., between about 0.5 and about 50 K enhancer ) is expected to disrupt the biological response mediated by the target PDZ-PL interaction.
  • the invention provides a method of determining the potency (K e , lhanc( .-) of an enhancer or suspected enhancer of binding between a PDZ domain and a ligand by immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface, contacting the immobilized polypeptide with a plurality of different mixtures ofthe ligand and enhancer, wherein the different mixtures comprise a fixed amount of ligand, at least a portion of which is detectably labeled, and different concentrations ofthe enhancer, determining the amount of ligand bound at the different concentrations of enhancer, and calculating the potency (!£-. convo,,,,,,,-rang-) of the enhancer from the binding based on the amount of ligand bound in the presence of different concentrations ofthe enhancer.
  • the ligand is detectably labeled to permit easy quantitation of ligand binding.
  • This method which is based on the "G” assay described supra, is particularly suited for high-throughput analysis of the K ⁇ ,,,, ⁇ . for enhancers of PDZ-ligand interactions.
  • the concentration of ligand and concentrations of enhancer are selected to allow meaningful detection of enhanced binding.
  • the ligand is typically present at a concentration of between about 0.01 Kd and about 0.5 Kd and the concentrations of the test agent/enhancer typically range from 1 nM to 1 mM (e.g. a 4-fold dilution series with highest concentration 10 uM or 1 mM).
  • the Kd is determined using the assay disclosed supra.
  • the potency ofthe binding can be determined by a variety of standard methods based on the amount of ligand bound in the presence of different concentrations of the enhancer or augmentor. For example, a plot of labeled ligand binding versus enhancer concentration can be fit to the equation:
  • the present invention provides powerful methods for analysis of PDZ-ligand interactions, including high-throughput methods such as the "G” assay and affinity assays described supra.
  • the affinity is determined for a particular ligand and a plurality of PDZ proteins.
  • the plurality is at least 5, and often at least 25, or at least 40 different PDZ proteins.
  • the plurality of different PDZ proteins are from a particular tissue (e.g., central nervous system, spleen, cardiac muscle, kidney) or a particular class or type of cell, (e.g., a hematopoietic cell, a lymphocyte, a neuron) and the like.
  • the plurality of different PDZ proteins represents a substantial fraction (e.g., typically a majority, more often at least 80%) of all ofthe PDZ proteins known to be, or suspected of being, expressed in the tissue or cell(s), e.g., all of the PDZ proteins known to be present in lymphocytes.
  • the plurality is at least 50%, usually at least 80%, at least 90% or all ofthe PDZ proteins disclosed herein as being expressed in hematopoietic cells (see Table 6).
  • the binding of a ligand to the plurality of PDZ proteins is determined. Using this method, it is possible to identify a particular PDZ domain bound with particular specificity by the ligand.
  • the binding may be designated as "specific” if the affinity ofthe ligand to the particular PDZ domain is at least 2-fold that ofthe binding to other PDZ domains in the plurality (e.g., present in that cell type).
  • the binding is deemed "very specific” if the affinity is at least 10-fold higher than to any other PDZ in the plurality or, alternatively, at least 10-fold higher than to at least 90%, more often 95% of the other PDZs in a defined plurality.
  • the binding is deemed “exceedingly specific” if it is at least 100- fold higher.
  • a ligand cound bind to 2 different PDZs with an affinity of 1 uM and to no other PDZs out of a set 40 with an affinty of less than 100 uM. This would constitute specific binding to those 2 PDZs.
  • Similar measures of specifity are used to describe binding of a PDZ to a plurality of PLs. It will be recognized that high specificity PDZ-PL interactions represent potentially more valuable targets for achieving a desired biological effect, the ability of an inhibitor or enhancer to act with high specificity is often desirable. In particular, the most specific PDZ-ligand interactions are also the best therapeutic targets, allowing specific inhibition of the interaction. In an embodiment an interaction between a PDZ and a PL is deemed
  • the invention provides a method of identifying a high specificity interaction between a particular PDZ domain and a ligand known or suspected of binding at least one PDZ domain, by providing a plurality of different immobilized polypeptides, each of said polypeptides comprising a PDZ domain and a non-PDZ domain; determining the affinity ofthe ligand for each of said polypeptides, and comparing the affinity of binding ofthe ligand to each of said polypeptides, wherein an interaction between the ligand and a particular PDZ domain is deemed to have high specificity when the ligand binds an immobilized polypeptide comprising the particular PDZ domain with at least 2-fold higher affinity than to immobilized polypeptides not comprising the particular PDZ domain.
  • the affinity of binding of a specific PDZ domain to a plurality of ligands is determined.
  • the invention provides a method of identifying a high specificity interaction between a PDZ domain and a particular ligand known or suspected of binding at least one PDZ domain, by providing an immobilized polypeptide comprising the PDZ domain and a non-PDZ domain; determining the affinity of each of a plurality of ligands for the polypeptide, and comparing the affinity of binding of each of the ligands to the polypeptide, wherein an interaction between a particular ligand and the PDZ domain is deemed to have high specificity when the ligand binds an immobilized polypeptide comprising the PDZ domain with at least 2-fold higher affinity than other ligands tested.
  • the binding may be designated as "specific” if the affinity ofthe PDZ to the particular PL is at least 2-fold that ofthe binding to other PLs in the plurality (e.g., present in that cell type).
  • the binding is deemed “very specific” if the affinity is at least 10-fold higher than to any other PL in the plurality or, alternatively, at least 10-fold higher than to at least 90%, more often 95% of the other PLs in a defined plurality.
  • the binding is deemed “exceedingly specific” if it is at least 100-fold higher.
  • the plurality is at least 5 different ligands, more often at lease 10.
  • the analysis encompasses all ofthe PDZ proteins expressed in a particular tissue (e.g., spleen) or type or class of cell (e.g., hematopoietic cell, neuron, lymphocyte, B cell, T cell and the like).
  • the analysis encompasses at least about 5, or at least about 10, or at least about 12, or at least about 15 and often at least 50 different polypeptides; or a substantial fraction (e.g., typically a majority, more often at least 80%) of all of the PDZ proteins known to be, or suspected of being, expressed in the tissue or cell(s), e.g., all of the PDZ proteins known to be present in lymphocytes.
  • the plurality is at least 50%, usually at least 80%, at least 90% or all ofthe PDZ proteins disclosed herein as being expressed in hematopoietic cells (see Table 6).
  • the array includes at least one, preferably at least 5 or at least 10 and sometimes all of the following PDZ proteins present in lymphocytes: BAI I associated prot., Connector enhancer, DLG5 (pdlg), DVL3, GTPase, Guanin-exchange factor 1, PDZ domain containing prot, KIAA147, KIAA0300, KIAA0380, KIAA0440, KIAA0545, KIAA0807, KIAA0858, KTAA0902, novel serine protease, PDZKl , PICK8, PTN-3, RPIP8, serine protease, 26s subunit p27, hSYNTENIN, TAXI -IP, TAX2-like protein, wwp3, XI 1 prot.
  • BAI I associated prot. Connector enhancer
  • DLG5 (pdlg) DLG5 (pdlg)
  • DVL3 GTPase
  • simultaneous analysis facilitates, for example, the direct comparison o the effect of an agent (e.g., an potential interaction inhibitor) on the interactions between a substantial portion of PDZs and/or PLs in a tissue or cell.
  • an agent e.g., an potential interaction inhibitor
  • the invention provides an array of immobilized polypeptide comprising the PDZ domain and a non-PDZ domain on a surface.
  • the array comprises at least about 5, or at least about 10, or at least about 12, or at least about 15 and often at least 50 different polypeptides.
  • the different PDZ proteins are from a particular tissue (e.g., central nervous system, speen, cardiac muscle, kidney) or a particular class or type of cell, (e.g., a hematopoietic cell, a lymphocyte, a neuron) and the like.
  • the plurality of different PDZ proteins represents a substantial fraction (e.g., typically a majority, more often at least 80%) of all ofthe PDZ proteins known to be, or suspected of being, expressed in the tissue or cell(s), e.g., all ofthe PDZ proteins known to be present in lymphocytes.
  • the plurality is at least 50%, usually at least 80%, at least 90% or all of the PDZ proteins disclosed herein as being expressed in hematopoietic cells (see Table 6). all of the PDZ proteins known to be present in lymphocytes.
  • the plurality is at least 50%, usually at least 80%, at least 90% or all ofthe PDZ proteins disclosed herein as being expressed in hematopoietic cells (see Table 6).
  • the array includes at least one, preferably at least 5 and sometimes all of the following PDZ proteins present in lymphocytes: BAI I associated prot., Connector enhancer, DLG5 (pdlg), DVL3, GTPase, Guanin-exchange factor 1, PDZ domain containing prot., KIAA147, KIAA0300, KIAA0380, KIAA0440, KIAA0545, KIAA0807, KIAA0858, KIAA0902, novel serine protease, PDZKl, PICK8, PTN-3, RPIP8, serine protease, 26s subunit p27, hSYNTENIN, TAXI -IP, TAX2-like protein, wwp3, XI 1 prot.
  • array refers to an ordered series of of immobilized polypeptides in which the identity of each polypeptide is associated with its location.
  • the plurality of polypeptides are arrayed in a "common” area such that they can be simultaneously exposed to a solution (e.g., containing a ligand or test agent).
  • a solution e.g., containing a ligand or test agent.
  • the plurality of polypeptides can be on a slide, plate or similar surface, which may be plastic, glass, metal, silica, beads or other surface to which proteins can be immobilized.
  • the different immobilized polypeptides are situated in separate areas, such as different wells of multi-well plate (e.g., a 24-well plate, a 96-well plate, a 384 well plate, and the like). It will be recognized that a similar advantage can be obtained by using multiple arrays in tandem.
  • multi-well plate e.g., a 24-well plate, a 96-well plate, a 384 well plate, and the like.
  • the invention provides a method for determining if a test compound inhibits any PDZ-ligand interaction in large set of PDZ-ligand interaction (e.g., some or all of the PDZ-ligands interactions described in Table 2; a majority ofthe PDZ-ligands identified in a particular cell or tissue as described supra (e.g., lymphocytes) and the like.
  • the PDZ domains of interest are expressed as GST-PDZ fusion proteins and immobilized as described herein.
  • a labeled ligand that binds to the domain with a known affinity is identified as described herein.
  • numerous PDZ-PL interactions occur in cells of the hematopoietic system.
  • any known or suspected modulator (e.g., inhibitor) of a PDL-PL interaction(s) it is useful to know which interactions are inhibited (or augmented).
  • an agent that inhibits all PDZ-PL interactions in a cell e.g., a lymphocyte
  • an agent that inhibits only one, or a small number, of specific PDZ-PL interactions e.g., an agent that inhibits only one, or a small number, of specific PDZ-PL interactions.
  • the profile of PDZ interactions inhibited by a particular agent is referred to as the "inhibition profile" for the agent, and is described in detail below.
  • the profile of PDZ interactions enhanced by a particular agent is referred to as the "enhancement profile" for the agent. It will be readily apparent to one of skill guided by the description o the inhibition profile how to determine the enhancement profile for an agent.
  • the present invention provides methods for determining the PDZ interaction (inhibition/enhancement) profile of an agent in a
  • the invention provides a method for determining the PDZ-PL inhibition profile of a compound by providing (i) a plurality of different immobilized polypeptides, each of said polypeptides comprising a PDZ domain and a non-PDZ domain and (ii) a plurality of corresponding ligands, wherein each ligand binds at least one PDZ domain in (i), then contacting each of said immobilized polypeptides in (i) with a corresponding ligand in (ii) in the presence and absence of a test compound, and determining for each polypeptide-ligand pair whether the test compound inhibits binding between the immobilized polypeptide and the corresponding ligand.
  • the plurality is at least 5, and often at least 25, or at least 40 different PDZ proteins.
  • the plurality of different ligands and the plurality of different PDZ proteins are from the same tissue or a particular class or type of cell, e.g., a hematopoietic cell, a lymphocyte, a neuron and the like.
  • the plurality of different PDZs represents a substantial fraction (e.g., at least 80%) of all ofthe PDZs known to be, or suspected of being, expressed in the tissue or cell(s), e.g., all ofthe PDZs known to be present in lymphocytes (for example, at least 80%, at least 90% or all ofthe PDZs disclosed herein as being expressed in hematopoietic cells).
  • the inhibition profile is determined as follows: A plurality (e.g., all known) PDZ domains expressed in a cell (e.g., lymphocytes) are expressed as GST- fusion proteins and immobilized without altering their ligand binding properties as described supra. For each PDZ domain, a labeled ligand that binds to this domain with a known affinity is identified. If the set of PDZ domains expressed in lymphocytes is denoted by ⁇ PI ...Pn ⁇ , any given PDZ domain Pi binds a (labeled) ligand Li with affinity K d i.
  • the "G” assay can be performed as follows in 96- well plates with rows A-H and columns 1-12. Column 1 is coated with PI and washed. The corresponding ligand LI is added to each washed coated well of column 1 at a concentration 0.5 K t jl with (rows B, D, F, H) or without (rows A, C, E, F) between about 1 and about 1000 uM) of test compound X. Column 2 is coated with P2, and L2 (at a concentration 0.5 1 ⁇ 2) is added with or without inhibitor X. Additional PDZ domains and ligands are similarly tested.
  • Compound X is considered to inhibit the binding of Li to Pi if the average signal in the wells of column i containing X is less than half the signal in the equivalent wells of the column lacking X. Thus, in this single assay one determines the full set of lymphocyte PDZs that are inhibited by compound X.
  • the test compound X is a mixture of compounds, such as the product of a combinatorial chemistry synthesis as described supra.
  • the test compound is known to have a desired biological effect, and the assay is used to determine the mechanism of action (i.e., if the biological effect is due to modulating a PDZ-PL interaction). It will be apparent that an agent that modulates only one, or a few PDZ-PL interactions, in a panel (e.g., a panel of all known PDZs lymphocytes, a panel of at least 10, at least 20 or at least 50 PDZ domains) is a more specific modulator than an agent that modulate many or most interactions.
  • an agent that modulates less than 20% of PDZ domains in a panel is deemed a "specific” inhibitor, less than 6% a “very specific” inhibitor, and a single PDZ domain a “maximally specific” inhibitor.
  • compound X may be a composition containing mixture of compounds (e.g., generated using combinatorial chemistry methods) rather than a single compound.
  • the assay above is performed using varying concentrations of the test compound X, rather than fixed concentration. This allows determination ofthe Ki ofthe X for each PDZ as described above.
  • each PDZ Pi with a specific labeled ligand Li
  • a mixture of different labeled ligands is created that such that for every PDZ at least one ofthe ligands in the mixture binds to this PDZ sufficiently to detect the binding in the "G” assay. This mixture is then used for every PDZ domain.
  • compound X is known to have a desired biological effect, but the chemical mechanism by which it has that effect is unknown.
  • the assays ofthe invention can then be used to determine if compound X has its effect by binding to a PDZ domain.
  • PDZ-domain containing proteins are classified in to groups based on their biological function, e.g. into those that regulate chemotaxis versus those that regulate transcription.
  • an optimal inhibitor of a particular function (e.g., including but not limited to an anti-chemotactic agent, an anti-T cell activation agent, cell-cycle control, vesicle transport, apoptosis, etc.) will inhibit multiple PDZ-ligand interactions involved in the function (e.g., chemotaxis, activation) but few other interactions.
  • the assay is used in one embodiment in screening and design of a drug that specifically blocks a particular function.
  • an agent designed to block chemotaxis might be identified because, at a given concentration, the agent inhibits 2 or more PDZs involved in chemotaxis but fewer than 3 other
  • the invention provides a method for identifying an agent that inhibits a first selected PDZ-PL interaction or plurality of interactions but does not inhibit a second selected PDZ-PL interaction or plurality of interactions.
  • the two (or more) sets of interactions can be selected on the basis ofthe known biological function ofthe PDZ proteins, the tissue specificity ofthe PDZ proteins, or any other criteria.
  • the assay can be used to determine effective doses (i.e., drug concentrations) that result in desired biological effects while avoiding undesirable effects.
  • the invention provides a method for determining likely side effects of a therapeutic that inhibits PDZ-ligand interactions.
  • the method entails identifying those target tissues, organs or cell types that express PDZ proteins and ligands that are disrupted by a specified inhibitor. If, at a therapeutic dosage, a drug intended to have an effect in one organ system (e.g., hematopoietic system) disrupts PDZ-PL interactions in a different system (e.g., CNS) it can be predicted that the drug will have effects (“side effects”) on the second system. It will be apparent that the information obtained from this assay will be useful in the rational design and selection of drugs that do not have the side-effect.
  • organ system e.g., hematopoietic system
  • CNS a different system
  • a comprehensive PDZ protein set is obtained.
  • a "perfectly comprehensive" PDZ protein set is defined as the set of all PDZ proteins expressed in the subject animal (e.g., humans).
  • a comprehensive set may be obtained by analysis of, for example, the human genome sequence.
  • a "perfectly comprehensive" set is not required and any reasonably large set of PDZ domain proteins (e.g., the set of all known PDZ proteins; or the set listed in Table 6) will provide valuable information.
  • the method involves some of all ofthe following steps: a) For each PDZ protein, determine the tissues in which it is highly expressed. This can be done experimentally although the information generally will be available in the scientific literature; b) For each PDZ protein (or as many as possible), identify the cognate PL(s) bound by the PDZ protein; c) Determine the Ki at which the test agent inhibits each PDZ-PL interaction, using the methods described supra; d) From this information it is possible to calculate the pattern of PDZ-PL interactions disrupted at various concentrations of the test agent By correlating the set of PDZ-PL interactions disrupted with the expression pattern of the members of that set, it will be possible to identify the tissues likely affected by the agent.
  • Additional steps can also be carried out, including determining whether a specified tissue or cell type is exposed to an agent following a particular route of administration. This can be determined using basis pharmacokinetic methods and principles.
  • the PDZ binding moieties and PDZ protein -PL protein binding antagonists of the invention are used to modulate biological activities or functions of cells (e.g., hematopoietic cells, such as T cells and B cells and the like), endothelial cells, and other immune system cells, as described herein, and for treatment of diseases and conditions in human and nonhuman animals (e.g., experimental models). Exemplary biological acitivities are listed supra.
  • the compounds of the invention are useful for treating (ameliorating symptoms of) a variety of diseases and conditions, including diseases characterized by inflammatory and humoral immune responses, e.g., inflammation, allergy (e.g., systemic anaphylaxis, hypersensitivity responses, drug allergies, insect sting allergies; inflammatory bowel diseases, ulcerative colitis, ileitis and enteritis; psoriasis and inflammatory dermatoses, scleroderma; respiratory allergic diseases such as asthma, allergic rhinitis, hypersensitivity lung diseases, and the like vasculitis, rh incompatibility, transfusion reactions, drug sensitivities, PIH, atopic dermatitis, eczema, rhinnitis; autoimmune diseases, such as arthritis (rheumatoid and psoriatic), multiple sclerosis, systemic lupus erythematosus, insulin-dependent diabetes,
  • inflammatory and humoral immune responses e.g., inflammation, allergy (e
  • PL proteins and PDZ proteins listed in TABLE 2 are well characterized, and one of skill, guided by this disclosure (including the discovery of the interactions between PL proteins and PDZ proteins described herein), will recognize many uses for modulators (e.g., enhancers or inhibitors) of PDZ-PL interactions such as those described in TABLE 2.
  • modulators e.g., enhancers or inhibitors
  • PDZ-PL interactions such as those described in TABLE 2.
  • CD6 binds PDZ protein 41.8.
  • CD6 is expressed on thymocytes, T cells, and B cell chronic lymphocytic leukemias. CD6 plays a role in T cell co-stimulation and CD6 negative T cells are less autoreactive than CD6 positive T cells. Inhibition of CD6 and CD6/41.8 interactions is predicted to reduce the symptoms of graft- versus-host disease (GVHD) or psoriasis.
  • GVHD graft- versus-host disease
  • GVHD graft- versus-host disease
  • GVHD graft- versus-host disease
  • CD6-PDZ interaction inhibitors will improve overall survival of transplantation patients (e.g., leukemia patients).
  • CD49e (ALPHA-4) As shown by the experiments reported herein, the C-terminal end of CD49e binds to the PDZ-domain-containing protein41.8kD.
  • CD49e is a 110 kD transmembrane membrane protein ofthe integrin alpha family (integrin alpha 5). Paired with the integrin beta-1 subunit it forms VLA-5. VLA-5 is expressed predominantly on hematopoietic and lymphoid lineage cells including monocytes, basophils, T cells, and activated B cells. VLA-5 is the receptor for the ubiquitously-expressed adhesion molecule fibronectin. Tissue injury such as myocardial infarction releases soluble fragments of fibronectin.
  • Binding of these soluble fragments to VLA- 5 results in chemotaxis of immune cells including monocytes to the source of fibronectin, as well as down-modulation of VLA-5 expression on these cells.
  • Such ligand-induced down-modulation is a common and required feature of chemotaxic receptors.
  • the 41.8/CD49e interaction is believed to be necessary for proper membrane distribution of CD49e and/or recycling of CD49e such that when it is disrupted, the migration and adherence to fibronectin - containing surfaces is similarly disrupted, resulting in an inability of immune system cells to effectively migrate toward a fibronectin source and adhere to fibronectin-containing surfaces.
  • Such disruption would therefore result in desirable reduced inflammatory processes, including reduced post-myocardial infarction inflammation.
  • Other diseases to be treated include but are not limited to joint inflamation, psoriasis, contact allergy, Crohn's Disease, inflammatory bowel disease, eczema, atopic dermatitis.
  • CD49F VLA-6 ⁇ subunit
  • CD49F binds PDZ protein 41.8.
  • CD49f is known as an integrin subunit that pairs either with the ⁇ l integrin subunit (CD29), forming VLA-6, or with CD 104 ( ⁇ 4 integrin subunit).
  • the integrin supergene family consists of a number of cell surface ⁇ heterodimers important for many different physiologic processes, including embryogenesis, thrombosis, wound healing, tumorigenesis and immune responses. Each ⁇ chain can pair with various ⁇ chains.
  • Both VLA-6 and CD49f/CD104 are widely expressed on epithelia in non- lymphoid tissues. VLA-6 is also expressed on platelets, monocytes, thymocytes and T lymphocytes, with an increased expression on activated and resting memory T cells.
  • Inhibition of interactions between VLA-6 and 41.8 has a number of therapeutic functions such as the prevention and treatment of metastatic cancers, and treatment of overactive immunity.
  • VLA-6 is associated with invasivion of prostrate carcinoma and plays a role in the metastasis of breast cancer. Blockage of VLA-6 function combined with conventional treatment for prostrate cancer, would be a more effective treatment by preventing metastatic disease (see, Cress et al., 1995, Cancer Metastasis R). Blockage of CD49f through PDZ interaction may also treat Rh incompatibility by blunting memory response or in the treatment of keloids.
  • CD 138 is a transmembrane proteoglycan receptor with the extracellular domain functioning as a ligand binding domain for various extracellular matrix components and the intracellular portion functioning to alter cytoskeleton and transduce intracellular signals. CD 138 also binds FGF2 and may be a co-receptor for FGF receptor (Yayon et al., 1991).
  • CD 138 interacts with 41.8kD protein and TIAM1.
  • the c- terminus of CD138 has also been reported to bind the PDZ domains of syntenin and human CASK (Cohen et al., 1998, 1. Cell. Biol. 142:129-138; Grootjans et al., 1997, PNAS 94:13683- 13688; Hsueh et al., 1998, J. Cell. Biol. 142:139-151).
  • CD138 is expressed in pre-B cells, immature B cells, plasma cells, neural cells, the basolateral surface of epithelial cells, embryonic mesenchymal cells, vascular smooth muscle cells, endothelial cells and neural cells but not mature circulating B cells.
  • CD138 The interaction between CD 138 and the PDZ domains ofthe 41 ,8kD protein and TIAM1 proteins is believed to be necessary for the proper distribution of CD 138 on the cell surface. Disruption of the interaction by administration of an effective amount of an antagonist is expected to interfere with the migration and adherence of cells to the extracellular matrix, resulting in reduced inflammatory and humoral immune responses. Inhibition of CD138 may be used to treat without limitation diseases such as post-myocardial infarction inflamatory damage, joint injury, rheumatoid arthritis, vasculitis, drug reaction, scleraderma, SLE, Hashimoto thyroiditis, Goodpasture's syndrome, juvenile insulin-dependent diabetes, psoriasis.
  • diseases such as post-myocardial infarction inflamatory damage, joint injury, rheumatoid arthritis, vasculitis, drug reaction, scleraderma, SLE, Hashimoto thyroiditis, Goodpasture's syndrome, juvenile insulin-dependent diabetes,
  • CD98 interacts with MPP2.
  • CD98 is expressed at high levels on monocytes and at low levels on T cells, B cells, splenocytes, NK cells, and granulocytes.
  • CD98 plays roles in adhesion, fusion and is a L-type amino acid transporter.
  • CD98 is also involved in virus-mediated cell fusion (e.g. paramyoviruses: parainfluenza virus type 2, Newcastle disease virus, and rubulaviruses)and antagonism of CD98 function is expected to treat )viral infections and limit viral spread.
  • CD98 inhibitors can be an antiviral agent for, but not limited to paramyovirus, parainfluenza, Newcastle disease and rubula. Other roles include treatment for acute leukemias.
  • CLASP-1 interacts with DLGl, PSD95, and NeDLG.
  • CLASP-1 is a member of a superfamily of immune-cell associated proteins with similar motifs (see PCT/US99/22996 published as WO 00/20434).
  • CLASP-1 functions in the maintenance of the immune synapse.
  • the CLASP-1 transcript is present in lymphoid organs and neural tissue, and the protein is expressed by T and B lymphocytes and macrophages in the MOMA-1 subregion of the marginal zone of the spleen, an area known to be important in T:B cell interaction.
  • CLASP-1 staining of individual T and B cells exhibits a preactivation structural polarity, being organized as a "ball” or “cap” structure in B cells, and forming a "ring", "ball” or “cap” structure in T cells. The placement of these structures is adjacent to the microtubule-organizing center ("MTOC").
  • MTOC microtubule-organizing center
  • CLASP-1 antibody staining indicates that CLASP-1 is at the interface of T-B cell conjugates that are fully committed to differentiation. Antibodies to the extracellular domain of CLASP-1 also block T-B cell conjugate formation and T cell activation.
  • Antagonism of CLASP-1 function is expected to interfere with immune responses (e.g., T and B cell activation), signal transduction, cell-cell interactions, and membrane organization.
  • Diseases to be treatment by CLASP- 1 agonists/antagonists include, but is not limited to, rheumaotoid arthritis, juvenile diabetes, organ rejection, graft-versus-host disease, scleroderma, multiple sclerosis.
  • CLASP-4 interacts with DLGl , PSD95, NeDLG, LDP, AF6, 41.8, and MINTl.
  • CLASP-4 is a member of a superfamily of immune-cell associated proteins with similar motifs (see copending U.S.Pat. Application 60/196,527 filed April 11, 2000).
  • the CLASP-4 protein is expressed primarily in peripheral blood lymphocytes. Inhibition of the interaction of CLASP-4 and PDZ domains will interfere with immune responses (e.g., T and B cell activation), signal transduction, cell-cell interactions, and membrane organization.
  • rheumaotoid arthritis include, but is not limited to, rheumaotoid arthritis, juvenile diabetes, organ rejection, graft-versus-host disease, scleroderma, multiple sclerosis, acute leukemias, leukemic blast crisis, post-infarction inflamation (cardiac, etc.), atherosclerosis.
  • VCAM-1 The vascular cell adhesion molecule- 1 (VCAM-1, CD106) is predominantly expressed by vascular endothelium (i.e., endothelial cells) but has been detected in macrophages, dendritic cells, bone marrow-derived cells, fibroblasts, cortical thymic epithelial cells, vascular smooth muscle cells, myoblasts and myotubes.
  • VCAM-1 mediates adhesion through interacting with an integrin ligand, VLA-4, which is expressed by lymphocytes, monocytes and eosinophils.
  • VCAM-1 and VLA-4 The interaction between VCAM-1 and VLA-4 is important for activation, flattening and extravasation of VLA-4 expressing cells when the endothelium itself has become activated due to inflammation or injury (Salomon et al., 1997, Blood 89:2461-2471; St-Pierre et al., 1996, Eur. J Immunol. 26:2050-2055; Bell et al., 1995, Int. Immunol. 7:1861-1871).
  • VCAM-1 is a ligand for the PDZ domains of MPPl, DLGl, NeDLG 1, LDP, 41.8 protein, TIAM1 and K303. These interactions are believed to mediate the function of endothelial cell interactions with integrin expressing leukocytes. When the PDZ-PL interactions are disrupted, the adherence of leukocytes to the endothelium will similarly be disrupted, resulting in, e.g., reduction of inflammation.
  • VCAM-1 binding to PDZ proteins is useful for reducing abnormal VCAM-1 inflammatory responses and associated pathologies such as (but not limited to) renal allograft rejection, insulin-dependent diabetes, rheumatoid arthritis, post-myocardial infarction complications and systemic lupus erythematosus (Pasloske et al., 1994, Ann Rev Med 45:283; Ockenhouse et al., 1992, J Exp. Med. 176: 1 183; Solezk et al., 1997, Kidney Int. 51 : 1476; Tedla, et al., 1999, Clin. Exp. Immunol.
  • CLASP-2 interacts with PSD-95, NeDLG, and DLGl .
  • CLASP-2 is a member of a superfamily of immune-cell associated proteins with similar motifs (see copending U.S. Pat. 09/547,276 filed April 11, 2000; WO 00/10158 filed April 11, 2000; WO 00/10156 filed April 11, 2000).
  • the CLASP-2 transcript is present most strongly in placenta followed by lung, kidney and heart and the protein is expressed in T and B cells, and kidney epithelial cells.
  • CLASP-2 Inhibition o the interaction of CLASP-2 and PDZ domains will interfere with CLASP-2 function resulting in interference with T and B cell function (e.g., T and B cell activation), signal transduction, cell-cell interactions, and membrane organization.
  • T and B cell function e.g., T and B cell activation
  • signal transduction e.g., T and B cell interactions
  • cell-cell interactions e.g., T and B cell activation
  • membrane organization e.g., T and B cell activation
  • blocking CLASP-2 function or expression can selectively block immune responses in the heart (for example, to selectively stop immune response in the heart compartment, e.g., following cardiac transplant rejection or post-MI inflammation, without compromising immunity elsewhere).
  • Other diseases to be treatment by CLASP-1 agonists/antagonists include, but is not limited to, rheumaotoid arthritis, juvenile diabetes, organ rejection, graft-versus-host disease, scleroderma, multiple sclerosis.
  • CD95 Fas/Apo-1
  • Fas ligand Fas ligand
  • CD95 binds the PDZ domains
  • Kvl .3 binds DLGl, PSD95, NeDLG, LIMK, 41.8, RGS12, DVLl, and MINTl .
  • Kvl.3 is a Shaker-related channel protein that is involved in modulating the membrane potential of T lymphocytes (Lewis and Cahalan, 1995, Ann. Rev. Immunol.
  • T cell signaling can be a useful therapeutic drug to treat, but not limited to, organ transplantation, graft-versus-host disease, Crohn's Disease, Ulcerative colitis, inflammatory bowel disease, rheumatoid arthritis, osteoarthritis, multiple sclerosis, scleroderma, mixed connective tissue disease.
  • DNAM-1 binds several PDZ proteins, including MPP2, DLGl ,
  • DNAM-1 is associated with Fyn constitutively but required the presence of pervanadate (a tyrosine phosphatase inhibitor) (see Shibuya, et al.
  • DNAM-1 Upon stimulation with anti-CD3 (or DNAM-1), DNAM-1 is phosphorylated at Serine 329 (Shibuya, et al. 1998, J. Immun. 161:1671-1676.) and associates with LFA-1. Furthermore, Fyn becomes associated with DNAM-1 independent of pervanadate. Fyn phosphorylates DNAM-1 at Y 322, but does not require Y322 to continue binding to DNAM-1. Since DNAM-1 itself does not have a SH3 binding domain but only has the Src phosphorylation site at Y322, an adaptor molecule must be present to bridge DNAM-1 and Fyn.
  • DLGl has been described in the literature to be present in T cells (Hanada, et al. 1997. J Biol Chem 272:26899), but does not bind Fyn.
  • PSD95 does not have a SH3 binding site.
  • SH3 binding domains including but not limited to NeDLG, RGS12, MPP2 and can fulfill this function.
  • These adaptor PDZ listed supra binds to DNAM-1 through its PDZ domain and simultaneously binds to the SH3 domain of Fyn through its proline-rich sequences just N-terminal to the PDZ domain.
  • the Y139 is a candidate phosphorylation site to control association of Fyn to DNAM-1 and the adaptor PDZ.
  • inhibition of PDZ association with DNAM-1 using the reagents of the invention will inhibit Fyn association with DNAM-1 and the subsequent Y322 phosphorylation and activation of cytotoxic T cells.
  • Diseases that can be treated include but are not limited to Crohn's Disease, multiple sclerosis, ulcerative colitis, inflammatory bowel disease, graft-versus-host, juvenile diabetes, Hashimoto's disease.
  • CD83 is a transmembrane glycoprotein, expressed predominantly on activated dendritic cells (DCs), Langerhans cells in the skin, with some weak expression detected on activated peripheral lymphocytes, and interdigitating reticulum cells within the T cell zones of lymphoid organs (Zhou and Tedder, 1995, J. Immunol. 154:3821-3835; Zhou et al, 1992, J. Immunol. 149:735-742).
  • CD83 is up-regulated de novo upon activation of an immature DCs, and is the major discriminating marker and characteristic for activated, mature DCs (Czerniecki et al, 1997, J. Immunol. 159:3823-3837).
  • DCs function as antigen presenting cells (APCs).
  • CD83 Upregulation and expression of CD83 thus appears to be required for DCs to mature and function as APCs.
  • the CD83 binds to the PDZ domains of DLGl , PSD95, and NeDLG. These interactions between CD83 and PDZ domains, and between CD83 and DLGl , PSD95, and NeDLG are believed to be important for proper distribution and recycling of CD83.
  • Disruption of CD83 and PDZ proteins with agonists and antagonist can be used to treat, but not limited to, psoriasis, cancers, allergies, autoimmune diseases such as multiple sclerosis, system lupus ery hematosis.
  • CD44 phagocytic glycoprotein 1, lymphocyte homing receptor, p85 and HCAM
  • CD44 is single pass transmembrane protein that has several different isoforms due to alternative splicing. It has a broad pattern of expression being detected on both hematopoietic and non-hematopoietic cell types including epithelial, endothelial, mesenchymal and neuronal cells.
  • CD44H is a major isoform that is expressed in lymphoid, myeloid and erythroid cells
  • CD44 is a receptor for hyaluronate (HA), which is a constituent of the extracellular matrix
  • CD44 functions as an adhesion molecule on the surface of leukocytes and erythrocytes that binds HA polymers in the ECM, and it can also act as a signaling receptor when HA becomes soluble during inflammatory reactions or tissue damage.
  • the cytoplasmic region of CD44 has been shown to bind or be associated with the actin cytoskeleton through interactions with spectrin and members of the ERM (ezrin, radixin, and meosin) family (reviewed in Lesley et al., 1993; Bajorath, 2000, Proteins 39:103-111).
  • CD44 is associated with the non-receptor tyrosine kinase p56Lck (Taher et al., 1996, J Biol. Chem. 271 :2863-2867. CD44 has been shown to be a co-stimulatory molecule with CD3/TCR engagement to activate T cells (reviewed in Aruffo, 1996, J. Clin. Invest. 98:2191-2192).
  • CD44 is a ligand for the PDZ domain contained in MPPl, prIL-16 and MINTl . It is believed that the interactions of CD44 with PDZ domains, and between CD44 with MPPl, prIL-16 and MINTl function in maintenance of leukocyte structure and in leukocyte signaling. Thus, when a CD44-PDZ interaction is disrupted, CD44 will fail to transduce proper intracellular signals, and maintain proper distribution of CD44 on the surface, which will prevent adhesion of leukocytes to the endothelium during inflammation and tissue damage.
  • agonists/antagonists of this interaction will thus result in, but not limited to, reduced inflammatory responses during tissue ischemia and cell lysis (e.g., rhabdomyosis), vascular insufficiencies (e.g. frostbite), psoriasis, eczema, graft-versus-host disease, granuloma annulare, scleroderma.
  • CD97 binds the PDZ domains of DLGl and 41.8.
  • CD97 is a 79.7 kD seven-span transmembrane protein expressed on granulocytes and monocytes and at low levels on resting T cells and B cells.
  • T or B cell activation expression levels of CD97 in T cells and B cells increases rapidly (Eichler et al., 1994, Scand. J. Immunol. 39, 111-115; Pickl et al, 1995, Leukocyte Typing :1151-1 153).
  • CD97 confers adhesion to lymphocytes and to erythrocytes. According to the present invention, the interaction of CD97 with DLGl and the interaction of CD97 with DLGl and the interaction of CD97 with DLGl and the interaction of CD97 with DLGl and the interaction of CD97 with DLGl and the interaction of CD97 with DLGl and the interaction of CD97 with DLGl and the interaction of CD97 with DLGl and the interaction of CD97 with DLG
  • 41.8 kd protein can be altered to interfere with proper membrane distribution of CD97 and/or recycling of CD97. Such modulation will affect CD97 dependent adherence of cells with therapeutic benefit.
  • agonists and antagonists of CD97-PDZ protein interaction can be used to treat rheumatoid arthritis, osteoarthritis, Crohn's Disease, Ulcerative colitis, psoriasis.
  • Glycophorin C interacts with the PDZ domains of human DLG, PSD95, NeDLG, MMP2, AF6, 41.8, and MINTl (with Mint-1 described previously).
  • Glycophorin C is an integral membrane protein expressed in erythroid cells, thymus, stomach, breast, adult and fetal liver, monocytes, T and B cells (Le Van Kim et al., 1989, J. Biol. Chem. 264:20407-20414) and is known for its role in human erythrocytes where it interacts with MPPl and protein 4.1 to regulate the shape, integrity and mechanical stability of red cells (Marfatia et al., 1997, J. Biol. Chem, 272:24191-24197; Reid et al., 1987, 5/ootf 69:1068-1072.).
  • Glycophorin C and PDZ proteins DLG, PSD95, NeDLG, MMP2, AF6, 41.8, and MINTl are believed necessary for maintenance ofthe physical integrity of cells in which they are expressed. Modulation of GC-PDZ interactions will alter with the function of these and can be utilized to treat, but not limited to, polycythemia vera, spherocytosis.
  • CDW128A binds to the PDZ domains of DLGl and NeDLG.
  • IL-8RA CDwl28A
  • IL-8RB CDwl28B
  • CDwl28A and CDwl28B both bind IL-8 with the same affinity but only CDwl28B, binds three other IL-8-related CXC chemokines: melanoma growth-stimulating activity (GRO/MGSA), neutrophil-activating peptide 2 (NAP-2) and ENA-78.
  • GRO/MGSA melanoma growth-stimulating activity
  • NAP-2 neutrophil-activating peptide 2
  • ENA-78 ENA-78
  • CDwl28A is expressed on all granulocytes, a subset of T cells, monocytes, endothelial cells, keratinocytes, erythrocytes, and melanoma cells.
  • IL-8 induces chemotaxis of neutrophils, basophils, and T lymphocytes and increases neutrophil and monocyte adhesion to endothelial cells.
  • the binding of IL-8 to IL8RA induces a transient increase in intracellular calcium levels, activation of phospholipase D, a respiratory burst of neutrophils and chemotaxis. This pro-inflammatory response is effective in normal immune responses.
  • Inhibitors of CDwl28A are useful for treatment of psoriasis, rheumatoid arthritis, polyarthritis, and for control of angiogenesis-dependent disorders such as melanomas and breast cancer.
  • CD3- ⁇ is a splice variant of CD3 zeta and a component ofthe CD3/TCR complex, which is required for antigen recognition, signal transduction and activation of T cells (Weiss and Liftman, 1994, Cell 76:263-274.). See, Barclay et al., 1997, The Leucocyte antigen facts book, 2nd Ed, Academic Press. As shown by experiments reported herein, the C-terminal region of CD3- ⁇ is a ligand for the PDZ domains of MINTl, 41.8 protein, DLGl, and PSD95.
  • CD3- ⁇ with PDZ domains and of CD3- ⁇ with MTNT1, 41.8 protein, DLGl, and PSD95 are believed to be important activation of T cells, which is required for all cellular immune responses. Modulation of this interaction by agonists and antogonists can be used to treat, but is not limited to, acute and chronic allograft rejection, multiple sclerosis, graft-versus- host disease, rheumatoid arthritis.
  • LPAP (CD45-AP, LSM-1) LPAP is a transmembrane protein expressed on resting and activated T- and B- cells. LPAP has been shown to bind to CD45, a protein that is part of the T-cell receptor complex and has been found to co-localize with CD4, CD2 and Thy-1. LPAP has also been co- immune precipitated with p56(lck) and ZAP-70. The actual function of LPAP is unknown, but it has been suggested that it is an assembly molecule for the CD45 complex.
  • LPAP binds to DLG-1 and MINT-1.
  • DLG-1 and MINT-1 are both expressed in T-cells. It has also been shown that DLG-1 co-precipitates with p56(lck) in T-cells.
  • the assays described herein also demonstrated that DLG-1 binds to CD95 and KV1.3, and binding of MINT-1 to KV1.3. All of these molecules are involved in signaling by the TCR.
  • LPAP is believed to function in organizing the signaling by CD45 in T-cell activation, possibly by recruiting p56(lck) as a substrate for CD45. Blocking the function of CD45 has been shown to severely impair the T-cell response.
  • Inhibiting the interaction between LPAP and PDZ proteins is expected to alter the CD45-mediated path from the rest ofthe immune response.
  • Agonists and antagonists of PL - PDZ binding can be used to treat, but is not limited to, rheumatoid arthritis, transplant rejection, multiple sclerosis, scleroderma, graft-versus-host disease.
  • T Complement membrane cofactor protein (MCP)
  • CD46 is a membrane protein expressed on all nucleated cells, but not on erythrocytes. CD46 is a member o the regulator of complement activation protein family. Its primary function is the protection of cells from complement attack by inactivating membrane deposited C3b/C4b complement (Liszewski, et al., 1999, Adv. Immunol. 61 :201-283). CD46 exists in more than 8 isoforms that are generated by differential splicing, with molecular weights ranging from 45 to 70 kD. In addition to the above function, CD46 also serves as the receptor for the measles virus and for other pathogenic microorganisms (e.g.
  • CD46 also appears to be over-expressed on certain tumors (Jurianz et al., 1999, Mol Immunol 36:929-939) thus rendering tumor cells insensitive to the action of complement. See, Barclay et al., (1997) The Leucocyte antigen facts book, 2ed, Academic Press.
  • CD46 binds DLGl, PSD95 and Ne-DLG. This interaction is believed to be necessary for proper membrane distribution of CD46 and/or recycling of CD46. Alteration of the CD46-PDZ interaction can reduce the ability of measles virus and other pathogens to enter cells, renders CD46-expressing tumors susceptible to attack by complement.
  • the administration of CD46-PDZ interaction agonists and antagonists is useful for the treatment of , but not limited to, cancers and viral infectious diseases.
  • CDwl28B binds to the PDZ domains of DLGl, NeDLG, PSD95, and 41.8 in the assays described supra.
  • IL-8 receptor IL- 8RA
  • IL-8RB CDwl28B
  • CDwl28A and CDwl28B both binds IL-8 with equal affinity but only CDwl28B, also binds three other IL-8- related CXC chemokines: melanoma growth-stimulating activity (GRO/MGSA), neutrophil- activating peptide 2 (NAP-2) and ENA-78. See, e.g., Ahuja, SK and Murphy, PM. 1996. J Biol Chem 271 :20545-50.
  • CDwl28B is expressed on all granulocytes, a subset of T cells, monocytes, endothelial cells, keratinocytes, erythrocytes, and melanoma cells.
  • IL-8 induces chemotaxis of neutrophils, basophils, and T lymphocytes but diminished relative to IL8RA and increases neutrophil and monocyte adhesion to endothelial cells.
  • the binding of IL-8A to its receptor induces a transient increase in intracellular calcium levelsand granule release but does not induce activation of phospholipase D or a respiratory burst in neutrophils. This pro-inflammatory response is effective in normal immune responses.
  • Inhibitors of CDwl28B are useful for treatment of psoriasis, rheumatoid arthritis, polyarthritis, and for control of angiogenesis- dependent disorders such as melanomas and breast cancer.
  • DOCK2 The DOCK family are a group of morphogenetic transmembrane proteins that interacts with cytoskeleton to affect cell shape changes. Members of this new family include Drosophila myoblast city (mbc), DOCK180, DOCK2, DOCK3, CED5, KIAA0209 and CLASP.
  • DOCK1 or DOCK180 is the human homolog of C. elegans gene, CED5 which is involved in the engulfment and phagacytosis by macrophages of apoptotic cells.
  • DOCK2 is found in peripheral blood lymphocytes and can convert a flatten cell into a rounded morphology upon transfection (Nagase, et. al. 1996. DNA Res 3:321-329, Nishihara, H. 1999. Hokkaido Igaku Zasshi 74:157).
  • DOCK2 is a PL and binds to PDZ proteins.
  • DOCK2 complexes with A,B,C to control cell shape in preparation for transit of lymphocytes for vascular circulation.
  • Modulation of DOCK2 by agonists and antagonists of its PDZ protein interaction can be used to treat, but is not limited to, acute leukemia, blast crisis, post-myocardial infarction inflammation, post-traumatic inflammation.
  • CD34 binds DLGl, PSD95, and NeDLG.
  • CD34 is expressed on a small subpopulation of bone marrow cells which includes hematopoietic stem cells. CD34 is also present on bone marrow stomal cells and on endothelial cells.
  • the selectins CD62L (L- selectin) and CD62E (E-selectin) bind CD34.
  • CD34 mediates attachment and rolling of leukocytes.
  • the hematopoietic stem cell properties of CD34 include myeloid differentiation of stem cells. Modulation of the CD34-PDZ interaction with agonists and/or antagonists can be used to treat, but is not limited to, myelodysplasia, leukemias, post-traumatic inflammation, post- myocardial infarction inflammation.
  • the high affinity receptor for human IgE is composed of an ⁇ , ⁇ , and disulfide-linked ⁇ homodimer.
  • the ⁇ -chain binds the Fc portion of IgE, whereas the ⁇ -chain serves to amplify signals that are transduced through the ⁇ -chain homodimer.
  • the Fc ⁇ R ⁇ l is a PDZ ligand and is a member of the CD20/ Fc ⁇ R ⁇ l receptor family. As is shown supra, Fc ⁇ R ⁇ l binds MINTl.
  • Fc ⁇ RI on basophils and mast cells plays a central role in the initiation of allergic responses. Signaling through the Fc ⁇ RI begins by crosslinking of a multivalent allergen bound to IgE. The result is vesicular degranulation, release of histamine, leukotrienes and pro-inflammatory cytokines (IL-6 and TNF ⁇ ), factors responsible for the symptoms of immediate hypersensitivity.
  • IL-6 and TNF ⁇ pro-inflammatory cytokines
  • Alteration of signaling by targeting PL/PDZ interaction with agonists and antagonists can be used to treat, but is not limited to, asthma, atopic dermatitis, eczema, drug reaction, mastocytosis, urticaria, eosinophilia myalgia syndrome (Turner, H., et. al., 1999, Nature 402 SUPP:B24).
  • FasL CD95 (Fas/Apo-1) and Fas ligand (FasL) are a receptor-ligand pair critically involved in lymphocyte homeostasis and peripheral tolerance. Binding of Fas by its ligand results in apoptotic cell death, an important major mechanism for safe clearance of unwanted cell during resolution of the acute inflammatory response. FasL is mainly restricted to activated T lymphocytes and is rapidly induced. Fas ligand is frequently up-regulated in breast cancer, as compared with normal breast epithelial cells and benign breast disease. As is shown supra, FasL binds the KIAA0561 PDZ domain.
  • the PDZ-PL modifiers are useful for treatment of, but not limited to, tumors, e.g., tumors unresponsive to conventional chemotherapy.
  • CDW125 binds PTN-4 and RGS12.
  • CDW125 is an IL-5 receptor expressed on eosinophils and basophils. IL5 promotes growth and differentiation of eosinophil precursors and actives mature eosinophils (Takatsu et al 1994, Adv. Immunol. 57:145- 190).
  • the secreted form of CDwl25 has antagonistic properties and is able to inhibit IL-5- induced eosinophil proliferation and differentiation.
  • Modulation of CDW125 binding to PDZ domains may be used to treat, but is not limited to, asthma, atopic dermatitis, eczema, drug reaction, urticaria, mastocytosis, eosinophilia.
  • BLR-1 Burkitt's lymphoma receptor-1
  • BLR-1 is a transmembrane receptor detected primarily on B cells, and shown to be upregulated in stimulated T cells (Dobner et al., 1992, EurJ. Immunol. 22:2795-2799.; Flynn et al., 1998, J. Exp. Med. 188:297-304). BLR-1 functions in chemotaxis of B and T cells into follicles of secondary lymphoid organs (e.g. spleen) for proper development and selection toward antigens (Forster et al., 1996, Cell 87: 1037-1047.
  • secondary lymphoid organs e.g. spleen
  • BLC B-lymphocyte chemoattractant
  • BLR-1 binds to the PDZ domain containing protein MINTl .
  • MINTl protein containing protein
  • BLR-1 and MINTl the interaction between BLR-1 and MINTl, and BLR-1 and PDZ domains is necessary for the proper distribution and signaling of BLR-1 on the cell surface.
  • the chemotactic abilities of lymphoid cells expressing BLR-1 is similarly disrupted.
  • Such a disruption results in a reduced immune response, interference with the ability of lymphocytes to properly circulate and develop responses to antigen.
  • Agonists and antagonists of this interaction can be used to treat, but is not limited to, systemic lupus erythematosus, scleroderma and other autoimmune diseases.
  • CD4 is a co-receptor with the T cell receptor (TCR) involved in antigen recognition. Both CD4 and TCR belong to the immunoglobulin supergene family. T cell activation is enhanced by increasing the avidity of T cells for effector and target cells.
  • the cytoplasmic domain is involved in signal transduction and association with the tyrosine kinase p56' clc .
  • CD4 is expressed on most fhymocytes, two-thirds of peripheral blood T lymphocytes, monocytes and macrophages.
  • HIV-1 Human immunodeficiency virus type-1 infects cells by membrane fusion mediated by its envelope glycoproteins (gpl20-gp41) and is triggered by the interaction of CD4 and a chemokine co-receptor, CCR5 or CXCR4.
  • Modulation of CD4-PDZ inhibitors with agonists and antagonists can be used to treat, but is not limited to, HIV infection immediately after exposure to HIV, rheumatoid arthritis, multiple sclerosis, scleroderma, systemic lupus erythematosis, psoriasis.
  • the antagonists of the invention may be disrupted or inhibited by the administration of inhibitors or antagonists.
  • Inhibitors can be identified using screening assays described herein.
  • the motifs disclosed herein are used to design inhibitors.
  • the antagonists of the invention have a structure (e.g., peptide sequence) based on the C-terminal residues of PL-domain proteins listed in TABLE 2.
  • the antagonists ofthe invention have a structure (e.g., peptide sequence) based on a PL motif disclosed herein.
  • the PDZ/PL antagonists and antagonists ofthe invention may be any of a large variety of compounds, both naturally occurring and synthetic, organic and inorganic, and including polymers (e.g., oligopeptides, polypeptides, ohgonucleotides, and polynucleotides), small molecules, antibodies, sugars, fatty acids, nucleotides and nucleotide analogs, analogs of naturally occurring structures (e.g., peptide mimetics, nucleic acid analogs, and the like), and numerous other compounds.
  • polymers e.g., oligopeptides, polypeptides, ohgonucleotides, and polynucleotides
  • small molecules antibodies, sugars, fatty acids, nucleotides and nucleotide analogs, analogs of naturally occurring structures (e.g., peptide mimetics, nucleic acid analogs, and the like), and numerous other compounds.
  • PDZ-PL interaction agonists can also
  • the peptides and peptide mimetics or analogues o the invention contain an amino add sequence that binds a PDZ domain in hematopoietic cells such as T cells and B cells, or otherwise inhibits the association of PL proteins and PDZ proteins.
  • the antagonists comprise a peptide that has a sequence corresponding to the carboxy-terminal sequence of a PL protein listed in TJ BLE 2, e.g., a peptide listed TABLE 4.
  • the peptide comprises at least the C-terminal four (4) residues ofthe PL protein, and often the inhibitory peptide comprises more than four residues (e.g., at least five, six, seven, eight, nine, ten, twelve or fifteen residues) from the PL protein C-terminus.
  • the C-terminal domains of specific surface receptors expressed by hematopoietic system and endothelial cells may themselves be used as inhibitors, and may be used as the basis for rational design of non-peptide inhibitors. See Section 6.6, infra.
  • the antagonist is a fusion protein comprising such a sequence.
  • Fusion proteins containing a transmembrane transporter amino acid sequence are particularly useful. See, e.g. Section 6.5.2, infra.
  • the inhibitor is conserved variant of the PL C-te ⁇ ninal protein sequence having inhibitory activity. See, e.g. Section 6.5.3, infra.
  • the antagonist is a peptide mimetic of a PL C-terminal sequence. See, e.g. Section 6.5.4, infra.
  • the inhibitor is a small molecule (i.e., having a molecular weight less than 1 kD). See, e.g. Section 6.5.5, infra.
  • the antagonists comprise a peptide that has a sequence of a PL protein oarboxy-terminus listed in TABLE 2.
  • the peptide comprises at least the C-terminal two (2) residues ofthe PL protein, and typically, the inhibitory peptide comprises more than two residues (e.g, at least three, four, five, six, seven, eight, nine, ten, twelve or fifteen residues) from the PL protein C-terminus. Most often, the residues shared by the inhibitory peptide with the PL protein are found at the C-terminus of the peptide.
  • the sequence is internal.
  • the inhibitory peptide comprises residues from a PL sequence that is near, but not at the c-terminus of a PL protein (see,Gee et al., 1998, J Biological Chem. 273:21980-87).
  • the "core PDZ motif sequence" of a hematopoietic cell surface receptor at its C-terminus contains the last four amino acids, this sequence may be used to target PDZ domains in hematopoietic cells.
  • the four amino acid core of a PDZ motif sequence may contain additional amino acids at its amino terminus to further increase its binding affinity and/or stability.
  • the PDZ motif sequence peptide can be from four amino acids up to 15 amino acids. It is preferred that the length of the sequence to be 6-10 amino acids. More preferably, the PDZ motif sequence contains 8 amino acids. Additional amino acids at the amino terminal end of the core sequence may be derived from the natural sequence in each hematopoietic cell surface receptor or a synthetic linker. The additional amino acids may also be conservatively substituted. When the third residue from the C-terminus is S, T or Y, this residue may be phosphorylated prior to the use ofthe peptide.
  • the peptide and nonpeptide inhibitors ofthe are small, e.g., fewer than ten amino acid residues in length if a peptide.
  • a limited number of ligand amino acids directly contact the PDZ domain (generally less than eight) (Kozlov et al., 2000, Biochemistry 39, 2572; Doyle et al., 1996, Cell 85, 1067) and that peptides as short as the C-terminal three amino acids often retain similar binding properties to longer (> 15) amino acids peptides (Yanagisawa et al., 1997, J. Biol. Chem. 272, 8539).
  • FIGURES 3 -H show the use of peptides to inhibit PL-PDZ interactions using the G assay described supra.
  • the inhibiton assays were carried out using GST fusion proteins containing PDZ domains from DLGl or PSD95 (see supra and TJABLE 3). Binding of biotinylated PL peptides for Clasp 2, CD46, Fas, or KV1.3 (as listed in TABLE 4) was determined in the presence of various competitor peptides (at a concentration of 100 uM) or in the absence of a competitor (equalized as 100% binding).
  • the competitor peptides were 8-mers peptides having the sequence of C-terminus of Clasp 2 (MTSSSSVV), CD46 (REVKFTSL), or Fas (RNEIQSLV), a unlabeled 19-mer having the sequence of c-terminus of KV1.3 (i.e., non-biotinylated AA33L as listed in TABLE 3), or a peptide having the sequence of residues 64-76 of hemoglobin (Vidal et al., 1999, J. Immunol. 163, 4811), i.e., an unrelated competitor.
  • FIGURES 3C-F show similar assays using shorter peptides to inhibit (e.g., a 3- mer and a 5-mer).
  • Figures 3C-E show binding of biotinylated PL peptides for Clasp 2, CD46, Fas, or KV1.3, at the indicated concentration (as listed in TABLE 3) to GST fusion proteins containing PDZ domains from NeDLG, DLGl, or PSD95 in the absence or presence of 1 mM 3-mer peptide having the sequence ofthe C-terminus of Clasp 2 (SVV).
  • FIGURE 3F shows the effect on binding of a 5-mer CD49E peptide (ATSDA) to GST fusion proteins containing a PDZ domain from 41.8Kd
  • Peptide Variants Having identified PDZ binding peptides and PDZ-PL interaction inhibitory sequences, variations of these sequences can be made and the resulting peptide variants can be tested for PDZ domain binding or PDZ-PL inhibitory activity.
  • the variants have the same or a different ability to bind a PDZ domain as the parent peptide.
  • amino acid substitutions are conservative, i.e., the amino acid residues are replaced with other amino acid residues having physical and/or chemical properties similar to the residues they are replacing.
  • conservative amino acid substitutions are those wherein an amino acid is replaced with another amino acid encompassed within the same designated class.
  • peptide mimetics can be prepared using routine methods, and the inhibitory activity ofthe mimetics can be confirmed using the assays ofthe invention.
  • the antagonist is a peptide mimetic of a PL C-terminal sequence.
  • individual synthetic residues and polypeptides incorporating mimetics can be synthesized using a variety of procedures and methodologies, which are well described in the scientific and patent literature, e.g., Organic Syntheses Collective Nolumes, Gilman et al. (Eds) John Wiley & Sons, Inc., ⁇ Y.
  • Polypeptides incorporating mimetics can also be made using solid phase synthetic procedures, as described, e.g., by Di Marchi, et al., U.S. Pat. No. 5,422,426.
  • Mimetics o the invention can also be synthesized using combinatorial methodologies.
  • Various techniques for generation of peptide and peptidomimetic libraries are well known, and include, e.g., multipin, tea bag, and split-couple-mix techniques; see, e.g., al-Obeidi (1998) Mol. Biotechnol. 9:205-223; Hruby (1997) Curr. Opin. Chem. Biol. 1 :114-119; Ostergaard (1997) Mol. Divers. 3:17-27; Ostresh (1996) Methods Enzymol. 267:220-234.
  • the inhibitor is a small molecule (i.e., having a molecular weight less than 1 kD).
  • Methods for screening small molecules are well known in the art and include those described supra at Section 6.4.
  • the following sections describe specific surface receptors expressed by different cell types in the hematopoietic and immune response system.
  • the C-termini of these receptors are used as inhibitors, or serve as the basis for designing PDZ motif sequence peptides, variants, fusion proteins, peptidomimetics, and small molecules for use in inhibiting PDZ-PL interactions.
  • the peptides are tested in an assay ofthe invention for inhibitory or modulator/ activity (also see, TABLE 4, and discussion supra).
  • T Cell Surface Receptors A number of surface receptors expressed by T cells contain a PDZ motif sequence
  • PL sequence (PL sequence). These molecules include CD3 ⁇ , CD4, CD6, CD38, CD49e, CD49f, CD53,
  • the C-terminal core sequence of CD3 is SSQL (SEQ ID NO:4).
  • SSSQL SEQ ID NO:5
  • SSSSQL SEQ ID NO:6
  • PSSSSQL PSSSSQL
  • PPSSSSQL PPSSSSQL
  • the C-terminal core sequence of CD4 is CSPI (SEQ ID NO:9).
  • TCSPI SEQ ID NO: 10
  • KTCSPI SEQ ID NO:l l
  • QKTCSPI SEQ ID NO:12
  • FQKTCSPI SEQ ID NO:13
  • the C-terminal core sequence of CD6 is ISAA (SEQ ID NO: 14).
  • DISAA SEQ ID NO: 15
  • DDISAA SEQ ID NO: 16
  • YDDISAA SEQ ID NO: 17
  • DYDDISAA SEQ ID NO: 18
  • the C-terminal core sequence of CD38 is TSEI (SEQ ID NO: 19).
  • CTSEI SEQ ID NO:20
  • SCTSEI SEQ ID NO:21
  • SSCTSEI SEQ ID NO:22
  • DSSCTSEI SEQ ID NO:23
  • the C-terminal core sequence of CD49e is TSDA (SEQ ID NO:24).
  • ATSDA SEQ ID NO:25
  • PATSDA SEQ ID NO:26
  • PP ATSDA SEQ ID NO:27
  • KPPATSDA SEQ ID NO:28
  • the C-terminal core sequence of CD49f is TSDA (SEQ ID NO:29).
  • TSDA SEQ ID NO:29
  • LTSDA SEQ ID NO:30
  • RLTSDA SEQ ID NO:31
  • ERLTSDA SEQ ID NO:32
  • KERLTSDA SEQ ID NO:33
  • the C-terminal core sequence of CD53 is TIGL (SEQ ID NO:34).
  • QTIGL (SEQ ID NO:35), SQTIGL (SEQ ID NO:36), TSQTIGL (SEQ ID NO:37), and KTSQTIGL (SEQ ID NO:38) may also be used to target a PDZ domain-containing protein in T cells.
  • the C-terminal core sequence of CD83 is TELV (SEQ. ID. NO: 248).
  • KTELV (SEQ. ID. NO: 249), HKTELV (SEQ. ID. NO: 250), PHKTELV (SEQ. ID. NO: 251), and TPHKTELV (SEQ. ID.
  • CD90 The C-terminal core sequence of CD90 is FMSL (SEQ ID NO:39).
  • DFMSL SEQ ID NO:40
  • TDFMSL SEQ ID NO:41
  • ATDFMSL SEQ ID NO:42
  • QATDFMSL SEQ ID NO:43
  • the C-terminal core sequence of CD95 is QSLV (SEQ ID NO:44).
  • QSLV SEQ ID NO:44
  • IQSLV SEQ ID NO:45
  • EIQSLV SEQ ID NO:46
  • NEIQSLV SEQ ID NO:47
  • RNEIQSLV SEQ ID NO:48
  • the C-terminal core sequence of CD97 is ESGI (SEQ ID NO:49).
  • SESGI SEQ ID NO:50
  • ASESGI SEQ ID NO:51
  • RASESGI SEQ ID NO:52
  • LRASESGI SEQ ID NO:53
  • the C-terminal core sequence of CD98 is PYAA (SEQ ID NO:54).
  • FPYJAA SEQ ID NO:55
  • RFPYAA SEQ ID NO:56
  • LRFPYAA SEQ ID NO:57
  • LLRFPYAA SEQ ID NO:58
  • the C-terminal core sequence of CDwl37 is GCEL (SEQ ID NO:59).
  • GGCEL SEQ ID NO:60
  • EGGCEL SEQ ID NO:61
  • EEGGCEL SEQ ID NO:62
  • EEEGGCEL SEQ ID NO:63
  • the C-terminal core sequence of CD 166 is KTEA (SEQ ID NO:64).
  • HKTEA SEQ ID NO:65
  • NHKTEA SEQ ID NO:66
  • NNHKTEA SEQ ID NO:67
  • ENNHKTEA SEQ ID NO:68
  • the C-terminal core sequence of CDwl28 is SSNL (SEQ ID NO:69).
  • VSSNL SEQ ID NO:70
  • NVSSNL SEQ ID NO:71
  • VNVSSNL SEQ ID NO:72
  • SVNVSSNL SEQ ID NO:73
  • the C-terminal core sequence of DNAM-1 is KTRV (SEQ ID NO:74).
  • PKTRV SEQ ID NO:75
  • RPKTRV SEQ ID NO:76
  • RRPKTRV SEQ ID NO:77
  • SRRPKTRV SEQ ID NO:78
  • the C-terminal core sequence of FasL is LYKL (SEQ ID NO:79).
  • GLYKL (SEQ ID NO:80), FGLYKL (SEQ ID NO:81), FFGLYKL (SEQ ID NO:82), and TFFGLYKL (SEQ ID NO:83) may also be used to target a PDZ domain-containing protein in T cells.
  • the C-terminal core sequence of LPAP is VTAL (SEQ ID NO:84).
  • VTAL SEQ ID NO:84
  • HVTAL SEQ ID NO:85
  • LHVTAL SEQ ID NO:86
  • GLHVTAL SEQ ID NO:87
  • QGLHVTAL SEQ ID NO:88
  • the C-terminal core sequence of CLASP-1 is SAQV (SEQ. ID. NO: 218).
  • SSAQV SEQ. ID. NO: 219
  • SSSAQV SEQ. ID. NO: 220
  • ISSSAQV SEQ. ID. NO: 221
  • SISSSAQV SEQ. ID. NO: 222
  • the C-terminal core sequence of CLASP-2 is SSVV (SEQ. ID. NO: 223).
  • SSSVV SEQ. ID. NO: 224
  • SSSSVV SEQ. ID. NO: 225
  • TSSSSVV SEQ. ID. NO: 226
  • MTSSSS W SEQ. ID. NO: 227)
  • the C-terminal core sequence of CLASP-4 is YAEV (SEQ. ID. NO: 228).
  • RYAEV SEQ. ID. NO: 229)
  • PRYAEV SEQ. ID. NO: 230
  • SPRYAEV SEQ. ID. NO: 231
  • GSPRYAEV SEQ. ID. NO: 232
  • FTDV SEQ. ID. NO: 238
  • IFTDV SEQ. ID. NO: 239
  • KIFTDV SEQ. ID. NO: 240
  • KKIFTDV SEQ. ID. NO: 241
  • IK IFTDV SEQ. ID. NO: 242
  • the C-terminal core sequence of DOCK2 is STDL (SEQ. ID. NO: 243).
  • LSTDL SEQ. ID. NO: 244
  • SLSTDL SEQ. ID. NO: 245
  • DSLSTDL SEQ. ID. NO: 246
  • PDSLSTDL SEQ. ID. NO: 247
  • B Cell Surface Receptors A number of surface receptors expressed by B cells contain a PDZ domain motif sequence. These molecules include, but are not limited to, CD38, CD53, CD95, CD97, CD98, CDwl37, CD138, CDwl25 (IL5R), DNAM-1 , LPAP, Syndecan-2 (Barclay et al., 1997, The Leucocyte Antigen Facts Book, second edition, Academic Press) and BLR-1.
  • the specific motif sequences of CD38, CD53, CD83, CD95, CD97, CD98, CDwl37, DNAM-1, DOCK2, LPAP, CLASP-1, CLASP-2 and CLASP-4 have been described in the preceding paragraphs.
  • the C-terminal core sequence of CD138 is EFYA (SEQ ID NO:89).
  • EEFYA SEQ ID NO:90
  • QEEFYA SEQ ID NO:91
  • KQEEFYA SEQ ID NO:92
  • TKQEEFYA SEQ ID NO:93
  • the C-terminal core sequence of CDwl25 is DSVF (SEQ ID NO:94).
  • EDSVF SEQ ID NO:95
  • LEDSVF SEQ ID NO:96
  • TLEDSVF SEQ ID NO:97
  • ETLEDSVF SEQ ID NO:98
  • the C-terminal core sequence of Syndecan-2 is EFYA (SEQ. ID. NO: 258).
  • KEFYA SEQ. ID. NO: 259
  • TKEFYA SEQ. ID. NO: 260
  • PTKEFYA SEQ. ID. NO: 261
  • APTKEFYA SEQ. ID. NO: 262
  • the C-terminal core sequence of BLR-1 is LTTF (SEQ. ID. NO: 253).
  • SLTTF SEQ. ID. NO: 254
  • TSLTTF SEQ. ID. NO: 255
  • ATSLTTF SEQ. ID. NO: 256
  • NATSLTTF SEQ. ID. NO: 257
  • PDZ motif sequences of Natural Killer Cell Surface Receptors A number of surface receptors expressed by NK cells contain a PDZ domain motif sequence. These molecules include, but are not limited to CD38, CD56, CD98 and DNAM-1. The specific motif sequences of CD38, CD98 and DNAM-1 have been described in the preceding paragraphs.
  • the C-terminal core sequence of CD56 is ESKA (SEQ ID NO:99).
  • NESKA SEQ ID NO: 100
  • ENESKA SEQ ID NO: 101
  • KENESKA SEQ ID NO: 102
  • TKENESKA SEQ ID NO: 103
  • PDZ motif sequences of Monocyte Surface Receptors A number of surface receptors expressed by cells of the monocytic lineage (monocytes and macrophages) contain a PDZ domain motif sequence. These molecules include, but are not limited to CD38, CD44, CD46, CD49e, CD49f, CD53, CD61 , CD95, CD97, CD98, CD148, CDwl28, CDwl37, Ly-6, DNAM-1 and Fc ⁇ RI ⁇ .
  • the specific motif sequences of CD38, CD49e, CD49f, CD53, CD95, CD97, CD98, CDwl28, CDwl37, DNAM-1, Galectin 3 (Mac-2) and Mannose receptor have been described in the preceding paragraphs.
  • the C-terminal core sequence of CD44 is KIGV (SEQ ID NO: 104).
  • MKIGV SEQ ID NO: 105
  • DMKIGV SEQ ID NO: 106
  • VDMKIGV SEQ ID NO: 107
  • NYDMKIGV SEQ ID NO: 108
  • the C-terminal core sequence of CD46 is FTSL (SEQ ID NO: 109).
  • KFTSL SEQ ID NO: 110
  • VKFTSL SEQ ID NOT 11
  • EVKFTSL SEQ ID NO: 112
  • REVKFTSL SEQ ID NOT 13
  • the C-terminal core sequence of CD61 is KSLV (SEQ ID NOT 14).
  • LKSLV SEQ ID NO: 115
  • FLKSLV SEQ ID NO: 116
  • RFLKSLV SEQ ID NO: 117
  • GRFLKSLV SEQ ID NOT 18
  • the C-terminal core sequence of CD148 is GYIA (SEQ ID NOT 19).
  • GYIA SEQ ID NO: 120
  • TNGYIA SEQ ID NO: 121
  • KTNGYIA SEQ ID NO: 122
  • GKTNGYIA SEQ ID NO: 123
  • the C-terminal core sequence of Ly-6 is QTLL (SEQ ID NO: 124).
  • LQTLL SEQ ID NO: 125
  • LLQTLL SEQ ID NOT26
  • VLLQTLL SEQ ID NOT27
  • SNLLQTLL SEQ ID ⁇ OT28
  • the C-terminal core sequence of Fc ⁇ RI ⁇ is PIDL (SEQ ID NO: 129).
  • PPIDL SEQ ID NO: 130
  • SPPIDL SEQ ID NO: 131
  • MSPPIDL SEQ ID NO: 132
  • EMSPPIDL SEQ ID NO: 133
  • the C-terminal core sequence of Galectin 3 is YTMI (SEQ ID NO: 134).
  • SYTMI SEQ ID NO: 135)
  • ASYTMI SEQ ID NO: 136
  • SASYTMI SEQ ID NO:137
  • TSASYTMI SEQ ID NOT 38
  • the C-terminal core sequence of mannose receptor is HSVI (SEQ ID NO: 139).
  • EHSVI SEQ ID NO: 140
  • NEHSVI SEQ ID NO: 141
  • QNEHSVI SEQ ID NOT 42
  • EQNEHSVI SEQ ID NO: 143
  • a number of surface receptors expressed by granulocytes contain a PDZ domain motif sequence. These molecules include, but are not limited to CD53, CD95, CD97, CD98, CD148, CDwl25, CDwl28, Fc ⁇ RI ⁇ and G-CSFR. The specific motif sequences of most of these molecules have been described in the preceding paragraphs.
  • the C-terminal core sequence of G-CSFR is TSVL (SEQ ID NOT44).
  • ITSVL SEQ ID NOT 45
  • PITSVL SEQ ID NOT46
  • FPITSVL SEQ ID NOT47
  • LFPITSVL SEQ ID NO.T48
  • endothelial cells While endothelial cells are not hematopoietic cells, they closely interact with the hematopoietic system as they form the lining of blood vessels. As such, endothelial cells come in contact with the cells ofthe hematopoietic system. Thus, the ability to regulate endothelial cell function provides for indirect regulation of hematopoietic cells.
  • a number of surface receptors expressed by endothelial cells contain a PDZ domain motif sequence. These molecules include, but are not limited to CD34, CD46, CD66b, CD66c, CD 105, CD106, CD62e (E-selectin) and VCAMl .
  • the C-terminal core sequence of CD34 is DTEL (SEQ ID NO: 149). When naturally-occurring residues are added to the core sequence, ADTEL (SEQ ID NO: 150), VADTEL (SEQ ID NO: 151), VVADTEL (SEQ ID NO: 152) and HVVADTEL (SEQ ID NO: 153) may also be used to target a PDZ domain-containing protein in endothelial cells.
  • the C-terminal core sequence of CD66b and CD66c is VALI (SEQ ID NOT 54).
  • RVALI SEQ ID NO: 155
  • ARVALI SEQ ID NO: 156
  • LARVALI SEQ ID NO: 15
  • VLARVALI SEQ ID NO: 158
  • the C-terminal core sequence of CD 105 is SSMA (SEQ ID NO: 159).
  • TSSMA (SEQ ID NO: 160), STSSMA (SEQ ID NOT 61), CSTSSMA (SEQ ID NOT 62) and PCSTSSMA (SEQ ID NO: 162) may also be used to target a PDZ domain-containing protein in endothelial cells.
  • the C-terminal core sequence of CD 106 is KSKV (SEQ ID NO: 163).
  • QKSKV (SEQ ID NO: 164), AQKSKV (SEQ ID NO: 165), EAQKSKV (SEQ ID NO: 166) and VEAQKSKV (SEQ ID NO: 167) may also be used to target a PDZ domain-containing protein in endothelial cells.
  • the C-terminal core sequence of CD62e is SYIL (SEQ ID NO: 168).
  • PSYIL SEQ ID NOT 69
  • KPSYIL SEQ ID NOT 70
  • QKPSYIL SEQ ID NO: 171
  • YQKPSYIL SEQ ID NOT 72
  • the C-terminal core sequence of VCAMl is KSKV (SEQ. ID. NO: 233).
  • QKSKV SEQ. ID. NO: 234
  • AQKSKV SEQ. ID. NO: 235
  • EAQKSKV SEQ. ID. NO: 236
  • VEAQKSKV SEQ. ID. NO: 237
  • Fc ⁇ RI ⁇ , CDwl25, CDwl28 and IL-8RB are transmembrane receptors expressed by mast cells, basophils and eosinophils. These receptors play a role in the activation of these cells to result in degranulation and histamine release in allergic reactions.
  • the C-terminal core sequence of Fc ⁇ RI ⁇ is PIDL (SEQ ID NO:4).
  • PPIDL SEQ ID NO:5
  • SPPIDL SEQ ID NO:6
  • MSPPIDL SEQ ID NO:7
  • EMSPPIDL SEQ ID NO:8
  • the residue E may be substituted with G to increase its binding affinity.
  • the C-terminal core sequence of CDwl25 is DSVF (SEQ ID NO: 9).
  • EDSVF SEQ ID NO: 10
  • LEDSVF LEDSVF
  • the C-terminal core sequence of CDwl28 is SSNL (SEQ ID NOT4).
  • VSSNL SEQ ID NO: 15
  • NNSSNL SEQ ID NO: 16
  • VNVSSNL SEQ ID NO: 17
  • SVNVSSNL SEQ ID NO: 18
  • the C-terminal core sequence of IL-8RB is STTL (SEQ ID NO: 19).
  • TSTTL SEQ ID NO:20
  • HTSTTL SEQ ID NO:21
  • GHTSTTL SEQ ID NO:22
  • SGHTSTTL SEQ ID NO:23
  • the C-terminal core sequence of NMD A is ESDV (SEQ. ID. NO: 263).
  • IESDV SEQ. ID. NO: 264
  • SIESDV SEQ. ID. NO: 265
  • PSIESDV SEQ. ID. NO: 266
  • MPSIESDV SEQ. ID. NO: 267
  • EYYV EYYV
  • KEYYV SEQ. ID. NO: 269
  • DKEYYV SEQ. ID. NO: 270
  • KDKEYYV SEQ. ID. NO: 271
  • NKDKEYYV SEQ. ID. NO: 272
  • the C-terminal core sequence of Glycophorin C is EYFI (SEQ. ID. NO: 273).
  • EYFI SEQ. ID. NO: 273
  • KEYFI SEQ. ID. NO: 274
  • RKEYFI SEQ. ID. NO: 275
  • SRKEYFI SEQ. ID. NO: 276
  • SSRKEYFI SEQ. ID. NO: 277
  • the C-terminal core sequence of CD148 is KTIA (SEQ. ID. NO: 278).
  • GKTIA SEQ. ID. NO: 279
  • FGKTIA SEQ. ID. NO: 280
  • TFGKTIA SEQ. ID. NO: 281
  • TTFGKTIA SEQ. ID. NO:
  • PDZ domain-containing protein may also be used to target a PDZ domain-containing protein in epithelial or myeloid cells.
  • the peptides of the invention or analogues thereof may be prepared using virtually any art-known technique for the preparation of peptides and peptide analogues.
  • the peptides may be prepared in linear form using conventional solution or solid phase peptide syntheses and cleaved from the resin followed by purification procedures (Creighton, 1983, Protein Structures And Molecular Principles, W.H. Freeman and Co., N.Y.). Suitable procedures for synthesizing the peptides described herein are well known in the art.
  • the composition of the synthetic peptides may be confirmed by amino acid analysis or sequencing (e.g., the Edman degradation procedure and mass spectroscopy).
  • analogues and derivatives of the peptides can be chemically synthesized.
  • the linkage between each amino acid of the peptides of the invention may be an amide, a substituted amide or an isostere of amide.
  • Nonclassical amino acids or chemical amino acid analogues can be introduced as a substitution or addition into the sequence.
  • Non-classical amino acids include, but are not limited to, the D-isomers ofthe common amino acids, ⁇ -amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, ⁇ -Abu, ⁇ -Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, ⁇ -alanine, fluoro-amino acids, designer amino acids such as ⁇ -methyl amino acids, C ⁇ -methyl amino acids, N ⁇ -methyl amino acids, and amino acid analogues in general.
  • the amino acid can be D (dextrorotary) or L (levorotary
  • the peptide is composed entirely of gene-encoded amino acids, or a portion of it is so composed, the peptide or the relevant portion may also be synthesized using conventional recombinant genetic engineering techniques.
  • a polynucleotide sequence encoding a linear form of the peptide is inserted into an appropriate expression vehicle, i.e., a vector which contains the necessary elements for the transcription and translation of the inserted coding sequence, or in the case of an RNA viral vector, the necessary elements for replication and translation.
  • the expression vehicle is then transfected into a suitable target cell which will express the peptide.
  • the expressed peptide is then isolated by procedures well-established in the art. Methods for recombinant protein and peptide production are well known in the art (see, e.g., Maniatis et al, 1989, Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y.; and Ausubel et al., 1989, Current
  • host-expression vector systems may be utilized to express the peptides described herein. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage DNA or plasmid DNA expression vectors containing an appropriate coding sequence; yeast or filamentous fungi transformed with recombinant yeast or fungi expression vectors containing an appropriate coding sequence; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing an appropriate coding sequence; plant cell systems infected with recombinant vims expression vectors (e.g., cauliflower mosaic virus or tobacco mosaic virus) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing an appropriate coding sequence; or animal cell systems.
  • microorganisms such as bacteria transformed with recombinant bacteriophage DNA or plasmid DNA expression vectors containing an appropriate coding sequence; yeast or filamentous fungi transformed with
  • the expression elements of the expression systems vary in their strength and specificities.
  • any of a number of suitable transcription and translation elements may be used in the expression vector.
  • inducible promoters such as pL of bacteriophage ⁇ , plac, ptrp, ptac (ptrp-lac hybrid promoter) and the like may be used;
  • promoters such as the baculovirus polyhedron promoter may be used;
  • promoters derived from the genome of plant cells e.g., heat shock promoters; the promoter for the small subunit of RUBISCO; the promoter for the chlorophyll a/b binding protein
  • plant viruses e.g., the 35S RNA promoter of CaMV; the coat protein promoter of TMV
  • sequences encoding the peptides of the invention may be driven by any of a number of promoters.
  • viral promoters such as the 35S RNA and 19S RNA promoters of CaMV (Brisson et al., 1984, Nature 310:511-514), or the coat protein promoter of TMV (Takamatsu et al., 1987, EMBO J. 3:17-311) may be used; alternatively, plant promoters such as the small subunit of RUBISCO (Coruzzi et al, 1984, EMBO J.
  • Autographa californica nuclear polyhidrosis virus (AcNPV) is used as a vector to express the foreign genes.
  • the virus grows in Spodoptera frugiperda cells.
  • a coding sequence may be cloned into non-essential regions (for example the polyhedron gene) of the virus and placed under control of an AcNPV promoter (for example, the polyhedron promoter). Successful insertion of a coding sequence will result in inactivation ofthe polyhedron gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat coded for by the polyhedron gene).
  • recombinant viruses are then used to infect Spodoptera frugiperda cells in which the inserted gene is expressed, (e.g., see Smith et al., 1983, J. Virol. 46:584; Smith, U.S. Patent No. 4,215,051). Further examples of this expression system may be found in Current Protocols in Molecular Biology, Vol. 2, Ausubel et al, eds., Greene Publish. Assoc. & Wiley Interscience. In mammalian host cells, a number of viral based expression systems may be utilized.
  • a coding sequence may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence.
  • This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region ofthe viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing peptide in infected hosts, (e.g., See Logan & Shenk, 1984, Proc. Natl. Acad. Sci. USA 81:3655- 3659).
  • the vaccinia 7.5 K promoter may be used, (see, e.g., Mackett et al, 1982, Proc. Natl. Acad. Sci. USA 79:7415-7419; Mackett et al, 1984, J. Virol. 49:857-864; Panicali et al, 1982, Proc. Natl. Acad. Sci. USA 79:4927-4931).
  • Other expression systems for producing linear peptides ofthe invention will be apparent to those having skill in the art.
  • the peptides and peptide analogues ofthe invention can be purified by art-known techniques such as high performance liquid chromatography, ion exchange chromatography, gel electrophoresis, affinity chromatography and the like.
  • the actual conditions used to purify a particular peptide or analogue will depend, in part, on factors such as net charge, hydrophobicity, hydrophilicity, etc., and will be apparent to those having skill in the art.
  • the purified peptides can be identified by assays based on their physical or functional properties, including radioactive labeling followed by gel electrophoresis, radioimmuno-assays, ELISA, bioassays, and the like.
  • any antibody which specifically binds the peptides or peptide analogues may be used.
  • various host animals including but not limited to rabbits, mice, rats, etc., may be immunized by injection with a peptide.
  • the peptide may be attached to a suitable carrier, such as BSA or KLH, by means of a side chain functional group or linkers attached to a side chain functional group.
  • adjuvants may be used to increase the immunological response, depending on the host species, including but not limited to Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, dinitrophenol, and potentially useful human adjuvants such as BCG (bacilli Calmette-Guerin) and Corynebacterium parvum.
  • BCG Bacilli Calmette-Guerin
  • Corynebacterium parvum bacilli Calmette-Guerin
  • Monoclonal antibodies to a peptide may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include but are not limited to the hybridoma technique originally described by Koehler and Milstein, 1975, Nature 256:495-497, the human B-cell hybridoma technique, Kosbor et al, 1983, Immunology Today 4:72; Cote et al, 1983, Proc. Natl. Acad. Sci. U.S.A. 80:2026-2030 and the EBV-hybridoma technique (Cole et al, 1985, Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96 (1985)).
  • Antibody fragments which contain deletions of specific binding sites may be generated by known techniques.
  • such fragments include but are not limited to F(ab') 2 fragments, which can be produced by pepsin digestion ofthe antibody molecule and Fab fragments, which can be generated by reducing the disulfide bridges of the F(ab') 2 fragments.
  • Fab expression libraries may be constructed (Huse et al, 1989, Science 246:1275- 1281) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity for the peptide of interest.
  • the antibody or antibody fragment specific for the desired peptide can be attached, for example, to agarose, and the antibody-agarose complex is used in immunochromatography to purify peptides of the invention. See, Scopes, 1984, Protein
  • the PDZ domain binding and PDZ-PL inhibitory compounds ofthe present invention are useful in regulating diverse activities of hematopoietic cells (e.g., T cells and B cells) and other cells. involved in the immune response.
  • hematopoietic cells e.g., T cells and B cells
  • the compounds ofthe invention are used to inhibit leukocyte activation, which is manifested in measurable events including but not limited to, cytokine production, cell adhesion, expansion of cell numbers, apoptosis and cytotoxicity.
  • the compounds of the invention may be used to treat diverse conditions associated with undesirable leukocyte activation, including but not limited to, acute and chronic inflammation, graft-versus-host disease, transplantation rejection, hypersensitivities and autoimmunity such as multiple sclerosis, rheumatoid arthritis, peridontal disease, systemic lupus erythematosis, juvenile diabetes mellitis, non-insulin-dependent diabetes, and allergies, and other conditions listed herein (see, e.g., Section 6.4, supra).
  • the invention also relates to methods of using such compositions in modulating leukocyte activation as measured by, for example, cytotoxicity, cytokine production, cell proliferation, and apoptosis.
  • Assays for activation are well known.
  • PDZ/PL interaction antagonists can be evaluated in the following: (1) cytotoxic T lymphocytes can be incubated with radioactively labeled target cells and the antigen-specific lysis of these target cells detected by the release of radioactivity, (2) helper T lymphocytes can be incubated with antigens and antigen presenting cells and the synthesis and secretion of cytokines measured by standard methods (Windhagen A; et al., 1995, Immunity 2(4): 373-80), (3) antigen presenting cells can be incubated with whole protein antigen and the presentation of that antigen on MHC detected by either T lymphocyte activation assays or biophysical methods (Harding et al., 1989, Proc.
  • mast cells can be incubated with reagents that cross-link their Fc-epsilon receptors and histamine release measured by enzyme immunoassay (Siraganian, et al., 1983, TIPS 4: 432-437).
  • the effect of PDZ/PL interaction antagonists on products of leukocyte activation in either a model organism (e.g., mouse) or a human patient can also be evaluated by various methods that are well known.
  • a model organism e.g., mouse
  • the production of antibodies in response to vaccination can be readily detected by standard methods currently used in clinical laboratories, e.g., an ELISA;
  • the migration of immune cells to sites of inflammation can be detected by scratching the surface of skin and placing a sterile container to capture the migrating cells over scratch site (Peters et al., 1988, Blood 72: 1310-5);
  • the proliferation of peripheral blood mononuclear cells in response to mitogens or mixed lymphocyte reaction can be measured using 3 H-thymidine;
  • the phagocytic capacity of granulocytes, macrophages, and other phagocytes in PBMCs can be measured by placing PMBCs in wells together with labeled particles (Peters et al., 1988); and
  • human peripheral blood mononuclear cells PBMC
  • human T cell clones e.g., Jurkat E6, ATCC TIB- 152
  • EBV-transformed B cell clones e.g., 9D10, ATCC CRL-8752
  • antigen-specific T cell clones or lines can be used to examine PDZ/PL interaction antagonists in vitro. Inhibition of activation of these cells or cell lines can be used for the evaluation of potential PDZ/PL interaction antagonists.
  • Standard methods by which hematopoietic cells are stimulated to undergo activation characteristic of an immune response are, for example:
  • MBP-specific antigen-specific T cell clones and hybridomas
  • B-cells can be mixed with antigen specific T-cells in the presence of antigen to generate an immune response. This can be performed in the presence or absence of PDZ/PL interaction antagonists to as
  • TCR cross-linking T cell receptor
  • receptor activation molecules e.g., TCR, CD3, or CD2
  • co-stimulator molecules for example anti-CD28
  • activation molecules e.g., TCR, CD3, or CD2
  • co-stimulator molecules for example anti-CD28
  • PHA phytohemagglutinin
  • pharmacological agents that activate protein kinase C e.g., phorbol esters
  • increase cytoplasmic Ca 24" e.g., ionomycin
  • Non-specific B cell activation 1) application of antibodies against cell surface molecules such as IgM, CD20, or CD21. 2) Lipopolysaccharide (LPS), phorbol esters, calcium ionophores and ionomycin can also be used to by-pass receptor triggering.
  • LPS Lipopolysaccharide
  • MLR Mixed lymphocyte reaction
  • a standard approach is to generate tetanus toxin-specific T cells from a donor that has recently been boosted with tetanus toxin.
  • Major histocompatability complex- (MHC-) matched antigen presenting cells and a source of tetanus toxin are used to maintain antigen specificity of the cell line or T cell clone (Lanzavecchia, A., et al, 1983, Eur. J. Immun. 13: 733-738).
  • Assay Quantitation The assays described above can be quantitated by a variety of well known quantitation methods. For example:
  • Tyrosine phosphorylation of early response proteins such as HS1, PLC-r, ZAP- 76, and Vav is an early biochemical event following leukocyte activation.
  • the tyrosine phosphorylated proteins can be detected by Western blot using antibodies against phosphorylated tyrosine residues. Tyrosine phosphorylation of these early response proteins can be used as a standard assay for leukocyte activation (J. Biol. Chem., 1997, 272(23): 14562-14570). Any change in the phosphorylation pattern of these or related proteins when immune responses are generated in the presence of potential PDZ/PL interaction antagonists is indicative of a potential PDZ/PL interaction antagonists.
  • (B) Intracellular Calcium Flux The kinetics of intracellular Ca 2+ concentrations are measured over time after stimulation of cells preloaded with a calcium sensitive dye. Upon binding Ca 21' the indicator dye (e.g., Fluor-4 (Molecular Probes)), exhibits an increase in fluorescence level using flow cytometry, solution fluorometry, and confocal microscopy. Any change in the level or timing of calcium flux when immune responses are generated in the presence of PDZ/PL interaction antagonists is indicative of an inhibition of this response.
  • the indicator dye e.g., Fluor-4 (Molecular Probes)
  • lymphocyte activation marker levels such as CD69, IL-2R, MHC class II, B7, and TCR are commonly measured with fluorescently labeled antibodies using flow cytometry. All antibodies are commercially available. Any change in the expression levels of lymphocyte activation markers when immune responses are generated in the presence ofthe PDZ/PL interaction antagonists is indicative of an inhibition of this response.
  • MTS [5-(3-carboxymethoxyphenyl)-2-(4,5-dimethylthiazolyl)-3(4- sulfo ⁇ henyl)tetrazolium, inner salt] is a colorimetric method for determining the number of viable cells in proliferation or cytotoxicity assays (Barltrop, J.A. et al., 1991, Bioorg. & Med. Chem. Lett. 1 : 611). 1-5 days after lymphocyte activation, MTS tetrazolium compound, Owen's reagent, is bioreduced by cells into a colored formazan product that is soluble in tissue culture media. Color intensity is read at 490 nm minus 650 nm using a microplate reader.
  • BrdU-pulsed cells are labeled with an enzyme-conjugated anti-BrdU antibody (Gratzner, H.G., 1982, Science 218: 474-475.).
  • a colorimetric, soluble substrate is used to visualize proliferating cells that have incorporated BrdU. Reaction is stopped with sulfuric acid and plate is read at 450 nm using a microplate reader. Any statistically significant increase or decrease in color intensity of the PDZ/PL interaction antagonist-treated sample, as compared to control sample (no treatment), suggest an effect of the PDZ/PL interaction antagonist on biological function.
  • F Apoptosis by Annexin V
  • IL-2, IL-3, IFN- ⁇ and other cytokine ELISA Assays are available for mouse, rat, and human (Endogen, Inc. and BioSource). Cytokine production is measured using a standard two-antibody sandwich ELISA protocol as described by the manufacturer. The presence of horseradish peroxidase is detected with 3, 3'5, 5' tertamethyl benziidine (TMB) substrate and the reaction is stopped with sulfuric acid. The absorbency at 450 nm is measured using a microplate reader.
  • TMB 3, 3'5, 5' tertamethyl benziidine
  • T cell activation requires the import of nuclear factor of activated T cells (NF- AT) to the nucleus.
  • NF-AT nuclear factor of activated T cells
  • This translocation of NF-AT can be visualized by immunostaining with anti- NF-AT antibody (Cell 1998, 93: 851-861). Therefore, NF-AT nuclear translocation has been used to assay T cell activation.
  • NF-AT/luciferase reporter assays have been used as a standard measurement of T cell activation (MCB 1996, 12: 7151-7160). Any statistically significant increase or decrease in the nuclear translocation of NF-AT brought about by the PDZ/PL interaction antagonist-treated sample, as compared to control sample (no treatment), suggest an effect of the PDZ/PL interaction antagonist on biological function.
  • the compounds of the invention may be tested in different dosages, formulations and route of administration in a cardiac transplant mouse model to optimize their ability to inhibit rejection responses to solid organ transplants (Fulmer et al., 1963, Am. J. Anat. 1 13:273; Jockusch et al., 1983, Exp. Neurol. 81 :749).
  • the compounds of the inventions may be used to inhibit PDZ domain interactions with CD3, CD4, CD6 and CDwl37.
  • the compounds of the invention may be used to inhibit PDZ domain interactions with CD53 and CD138 in B cells.
  • the compounds of the invention may be used to inhibit PDZ domain interactions with Fc ⁇ RI ⁇ , CDwl25 and CDwl28.
  • a PDZ motif sequence (PL sequence) of CD95 may be used to induce apoptosis of lymphomas.
  • the PDZ-PL antagonists ofthe invention are introduced into a cell to modulate (i.e., increase or decrease) a biological function or activity ofthe cell.
  • Many small organic molecules readily cross the cell membranes (or can be modified by one of skill using routine methods to increase the ability of compounds to enter cells, e.g., by reducing or eliminating charge, increasing lipophilicity, conjugating the molecule to a moiety targeting a cell surface receptor such that after interacting with the receptor).
  • Methods for introducing larger molecules e.g., peptides and fusion proteins are also well known, including, e.g., injection, liposome-mediated fusion, application of a hydrogel, conjugation to a targeting moiety conjugate endocytozed by the cell, electroporation, and the like).
  • the antagonist or agent is a fusion polypeptide or derivatized polypeptide.
  • a fusion or derivatized protein may include a targeting moiety that increases the ability ofthe polypeptide to traverse a cell membrane or causes the polypeptide to be delivered to a specified cell type (e.g., liver cells or tumor cells) preferentially or cell compartment (e.g., nuclear compartment) preferentially.
  • targeting moieties include lipid tails, amino acid sequences such as antennapoedia peptide or a nuclear localization signal (NLS; e.g., Xenopus nucleoplasmin Robbins et al., 1991, Cell 64:615).
  • a peptide sequence or peptide analog determined to inhibit a PDZ domain-PL protein binding, in an assay of the invention is introduced into a cell by linking the sequence to an amino acid sequence that facilitates its transport through the plasma membrane (a "transmembrane transporter sequence").
  • the peptides of the invention may be used directly or fused to a transmembrane transporter sequence to facilitate their entry into cells.
  • each peptide may be fused with a heterologous peptide at its amino terminus directly or by using a flexible polylinker such as the pentamer G-G-G-G-S (SEQ ID NOT) repeated 1 to 3 times.
  • linker has been used in constructing single chain antibodies (scFv) by being inserted between V H and V L (Bird et al., 1988, Science 242:423-426; Huston et al., 1988, Proc. Natl. Acad. Sci. U.S.A. 85:5979-5883).
  • the linker is designed to enable the correct interaction between two beta-sheets forming the variable region ofthe single chain antibody.
  • Other linkers which may be used include Glu-Gly- Lys-Ser-Ser-Gly-Ser-Gly-Ser-Glu-Ser-Lys-Val-Asp (SEQ ID NO:2) (Chaudhary et al., 1990,
  • transmembrane transporter peptides examples include, but are not limited to, tat derived from HIV (Vives et al, 1997, J. Biol. Chem. 272:16010; Nagahara et al, 1998, Nat. Med. 4:1449), antennapedia from Drosophila (Derossi et al., 1994, J Biol. Chem.
  • a truncated HIV tat peptide having the sequence of GYGRKKRRQRRRG (SEQ ID NO: 173) is used. It is preferred that a transmembrane transporter sequence is fused to a hematopoietic cell surface receptor carboxyl terminal sequence at its amino-terminus with or without a linker. Generally, the C-terminus of a PDZ motif sequence (PL sequence) must be free in order to interact with a PDZ domain.
  • the transmembrane transporter sequence may be used in whole or in part as long as it is capable of facilitating entry ofthe peptide into a cell.
  • a hematopoietic cell surface receptor C-terminal sequence may be used alone when it is delivered in a manner that allows its entry into cells in the absence of a transmembrane transporter sequence.
  • the peptide may be delivered in a liposome formulation or using a gene therapy approach by delivering a coding sequence for the PDZ motif alone or as a fusion molecule into a target cell.
  • the compounds ofthe ofthe invention may also be administered via liposomes, which serve to target the conjugates to a particular tissue, such as lymphoid tissue, or targeted selectively to infected cells, as well as increase the half-life of the peptide composition.
  • Liposomes include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phosphohpid dispersions, lamellar layers and the like.
  • the peptide to be delivered is incorporated as part of a liposome, alone or in conjunction with a molecule which binds to, e.g., a receptor prevalent among lymphoid cells, such as monoclonal antibodies which bind to the CD45 antigen, or with other therapeutic or immunogenic compositions.
  • liposomes filled with a desired peptide or conjugate ofthe invention can be directed to the site of lymphoid cells, where the liposomes then deliver the selected inhibitor compositions.
  • Liposomes for use in the invention are formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol. The selection of lipids is generally guided by consideration of, e.g., liposome size, acid lability and stability of the liposomes in the blood stream. A variety of methods are available for preparing liposomes, as described in, e.g., Szoka et al., Ann. Rev. Biophys. Bioeng. 9:467 (1980), U.S. Pat.
  • a ligand to be incorporated into the liposome can include, e.g., antibodies or fragments thereof specific for cell surface determinants of the desired immune system cells.
  • a liposome suspension containing a peptide or conjugate may be administered intravenously, locally, topically, etc. in a dose which varies according to, inter alia, the manner of administration, the conjugate being delivered, and the stage of the disease being treated.
  • the peptide may be linked to a cell-specific targeting moiety, which include but are not limited to, ligands for diverse leukocyte surface molecules such as growth factors, hormones and cytokines, as well as antibodies or antigen-binding fragments thereof. Since a large number of cell surface receptors have been identified in leukocytes, ligands or antibodies specific for these receptors may be used as cell-specific targeting moieties.
  • a cell-specific targeting moiety include but are not limited to, ligands for diverse leukocyte surface molecules such as growth factors, hormones and cytokines, as well as antibodies or antigen-binding fragments thereof. Since a large number of cell surface receptors have been identified in leukocytes, ligands or antibodies specific for these receptors may be used as cell-specific targeting moieties.
  • interleukin-2, B7-1 (CD80), B7-2 (CD86) and CD40 or peptide fragments thereof may be used to specifically target activated T cells (The Leucocyte Antigen Facts Book, 1997, Barclay et al. (eds.), Academic Press).
  • CD28, CTLA-4 and CD40L or peptide fragments thereof may be used to specifically target B cells.
  • Fc domains may be used to target certain Fc receptor- expressing cells such as monocytes.
  • Antibodies are the most versatile cell-specific targeting moieties because they can be generated against any cell surface antigen. Monoclonal antibodies have been generated against leukocyte lineage-specific markers such as certain CD antigens. Antibody variable region genes can be readily isolated from hybridoma cells by methods well known in the art. However, since antibodies are assembled between two heavy chains and two light chains, it is preferred that a scFv be used as a cell-specific targeting moiety in the present invention. Such scFv are comprised of V H and V L domains linked into a single polypeptide chain by a flexible linker peptide.
  • the PDZ motif sequence may be linked to a transmembrane transporter sequence and a cell-specific targeting moiety to produce a tri-fusion molecule. This molecule can bind to a leukocyte surface molecule, passes through the membrane and targets PDZ domains. Alternatively, a PDZ motif sequence (PL sequence) may be linked to a cell- specific targeting moiety that binds to a surface molecule that internalizes the fusion peptide. In an other approach, microspheres of artificial polymers of mixed amino acids
  • a polynucleotide encoding a surface receptor C-terminal peptide may be useful in the treatment of various leukocyte activation-associated abnormal conditions. By introducing gene sequences into cells, gene therapy can be used to treat conditions in which leukocytes are activated to result in deleterious consequences.
  • a polynucleotide that encodes a PL sequence peptide of the invention is introduced into a cell where it is expressed.
  • the expressed peptide then inhibits the interaction of PDZ proteins and PL proteins in the cell.
  • the polypeptides of the invention are expressed in a cell by introducing a nucleic acid (e.g., a DNA expression vector or mRNA) encoding the desired protein or peptide into the cell.
  • a nucleic acid e.g., a DNA expression vector or mRNA
  • Expression may be either constitutive or inducible depending on the vector and choice of promoter. Methods for introduction and expression of nucleic acids into a cell are well known in the art and described herein.
  • nucleic acids comprising a sequence encoding a peptide disclosed herein, are administered to a human subject.
  • the nucleic acid produces its encoded product that mediates a therapeutic effect by inhibiting leukocyte activation. Any ofthe methods for gene therapy available in the art can be used according to the present invention. Exemplary methods are described below.
  • the therapeutic composition comprises a coding sequence that is part of an expression vector.
  • a nucleic acid has a promoter operably linked to the coding sequence, said promoter being inducible or constitutive, and, optionally, tissue-specific.
  • a nucleic acid molecule is used in which the coding sequence and any other desired sequences are flanked by regions that promote homologous recombination at a desired site in the genome, thus providing for intrachromosomal expression of the nucleic acid (Koller and Smithies, 1989, Proc. Natl. Acad. Sci. USA 86:8932-8935; Zijlstra et al., 1989, Nature 342:435-438).
  • nucleic acid into a patient may be either direct, in which case the patient is directly exposed to the nucleic acid or nucleic acid-carrying vector, or indirect, in which case, cells are first transformed with the nucleic acid in vitro, then transplanted into the patient. These two approaches are known, respectively, as in vivo or ex vivo gene therapy.
  • the nucleic acid is directly administered in vivo, where it is expressed to produce the encoded product.
  • a nucleic acid- ligand complex can be formed in which the ligand comprises a fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation.
  • the nucleic acid can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor (see, e.g., PCT Publications WO 92/06180 dated April 16, 1992; WO 92/22635 dated December 23, 1992; WO92/20316 dated November 26, 1992; WO93/14188 dated July 22, 1993; WO 93/20221 dated October 14, 1993).
  • the nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination (Koller and Smithies, 1989, Proc. Natl. Acad. Sci. USA 86:8932- 8935; Zijlstra et al., 1989, Nature 342:435-438).
  • adenoviruses as viral vectors can be used in gene therapy. Adenoviruses have the advantage of being capable of infecting non- dividing cells (Kozarsky and Wilson, 1993, Current Opinion in Genetics and Development 3:499-503).
  • adenoviruses in gene therapy can be found in Rosenfeld et al., 1991 , Science 252:431-434; Rosenfeld et al., 1992, Cell 68: 143-155; and Mastrangeli et al., 1993, J. Clin. Invest. 91 :225-234.
  • adenoviral vectors with modified tropism may be used for cell specific targeting (WO98/40508).
  • Adeno-associated virus (AAV) has also been proposed for use in gene therapy (Walsh et al., 1993, Proc. Soc. Exp. Biol. Med. 204:289-300).
  • retroviral vectors see Miller et al., 1993, Meth.
  • Enzymol. 217:581- 599) have been modified to delete retroviral sequences that are not necessary for packaging of the viral genome and integration into host cell DNA.
  • the coding sequence to be used in gene therapy is cloned into the vector, which facilitates delivery ofthe gene into a patient. More detail about retroviral vectors can be found in Boesen et al., 1994, Biotherapy 6:291-302, which describes the use of a retroviral vector to deliver the mdrl gene to hematopoietic stem cells in order to make the stem cells more resistant to chemotherapy. Other references illustrating the use of retroviral vectors in gene therapy are: Clowes et al., 1994, J. Clin. Invest.
  • Another approach to gene therapy involves transferring a gene to cells in tissue culture.
  • the method of transfer includes the transfer of a selectable marker to the cells.
  • the cells are then placed under selection to isolate those cells that have taken up and are expressing the transferred gene. Those cells are then delivered to a patient.
  • the nucleic acid is introduced into a cell prior to administration in vivo ofthe resulting recombinant cell.
  • Such introduction can be carried out by any method known in the art, including but not limited to transfection, electroporation, lipofection, microinjection, infection with a viral or bacteriophage vector containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcell-mediated gene transfer, spheroplast fusion, etc.
  • Numerous techniques are known in the art for the introduction of foreign genes into cells (see e.g., Loeffler and Behr, 1993, Meth. Enzymol. 217:599-618; Cohen et al., 1993, Meth. Enzymol. 217:618-644; Cline, 1985, Pharmac. Ther.
  • the technique should provide for the stable transfer o the nucleic acid to the cell, so that the nucleic acid is expressible by the cell and preferably heritable and expressible by its cell progeny.
  • the cell used for gene therapy is autologous to the patient.
  • the nucleic acid to be introduced for purposes of gene therapy comprises an inducible promoter operably linked to the coding sequence, such that expression of the nucleic acid is controllable by controlling the presence or absence of the appropriate inducer of transcription.
  • Ohgonucleotides such as anti-sense RNA and DNA molecules, and ribozymes that function to inhibit the translation of a leukocyte surface receptor mRNA, especially its C- terminus are also within the scope of the invention.
  • Anti-sense RNA and DNA molecules act to directly block the translation of mRNA by binding to targeted mRNA and preventing protein translation.
  • antisense DNA oligodeoxyribonucleotides derived from the translation initiation site, e.g., between -10 and +10 regions of a nucleotide sequence, are preferred.
  • the antisense oligonucleotide may comprise at least one modified base moiety which is selected from the group including, but not limited to, 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine,
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization ofthe ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
  • engineered hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of leukocyte surface receptor RNA sequences.
  • ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites which include the following sequences, GUA, GUU and GUC. Once identified, short RNA sequences of between 15 and 20 ribonucleotides corresponding to the region ofthe target gene containing the cleavage site may be evaluated for predicted structural features such as secondary structure that may render the oligonucleotide sequence unsuitable. The suitability of candidate targets may also be evaluated by testing their accessibility to hybridization with complementary ohgonucleotides, using ribonuclease protection assays.
  • RNA molecules and DNA molecules and ribozymes ofthe invention may be prepared by any method known in the art for the synthesis of nucleic acid molecules. These include techniques for chemically synthesizing oligodeoxyribonucleotides well known in the art such as for example solid phase phosphoramidite chemical synthesis.
  • RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the RNA molecule. Such DNA sequences may be incorporated into a wide variety of vectors which contain suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters.
  • antisense cDNA constructs that synthesize antisense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably into cell lines.
  • DNA molecules may be introduced as a means of increasing intracellular stability and half-life. Possible modifications include, but are not limited to, the addition of flanking sequences of ribo- or deoxy- nucleotides to the 5' and/or 3' ends of the molecule or the use of phosphorothioate or 2' O-methyl rather than phosphodiesterase linkages within the oligodeoxyribonucleotide backbone.
  • compositions may be administered to a subject per se or in the form of a sterile composition or a pharmaceutical composition.
  • Pharmaceutical compositions comprising the compounds of the invention may be manufactured by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • Pharmaceutical compositions may be formulated in conventional manner using one or more physiologically acceptable carriers, diluents, excipients or auxiliaries that facilitate processing of the active peptides or peptide analogues into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the compounds ofthe invention may be formulated as solutions, gels, ointments, creams, suspensions, etc. as are well-known in the art.
  • Systemic formulations include those designed for administration by injection, e.g. subcutaneous, intravenous, intramuscular, intrathecal or intraperitoneal injection, as well as those designed for transdermal, transmucosal, oral or pulmonary administration.
  • the compounds of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • the solution may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the compounds may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. This route of administration may be used to deliver the compounds to the nasal cavity.
  • the compounds can be readily formulated by combining the active peptides or peptide analogues with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds ofthe invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • suitable excipients include fillers such as sugars, such as lactose, sucrose, mannitol and sorbitol; cellulose preparations such as maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP); granulating agents; and binding agents.
  • disintegrating agents may be added, such as the cross-linked polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • solid dosage forms may be sugar-coated or enteric-coated using standard techniques.
  • suitable carriers, excipients or diluents include water, glycols, oils, alcohols, etc. Additionally, flavoring agents, preservatives, coloring agents and the like may be added.
  • the compounds may take the form of tablets, lozenges, etc. formulated in conventional manner.
  • the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of e.g. gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compounds may also be formulated in rectal or vaginal compositions such as suppositories
  • the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • suitable polymeric or hydrophobic materials for example as an emulsion in an acceptable oil
  • ion exchange resins for example as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • other pharmaceutical delivery systems may be employed.
  • Liposomes and emulsions are well known examples of delivery vehicles that may be used to deliver peptides and peptide analogues of the invention.
  • Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity.
  • the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid polymers containing the therapeutic agent.
  • sustained-release materials have been established and are well known by those skilled in the art.
  • Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability ofthe therapeutic reagent, additional strategies for protein stabilization may be employed.
  • the compounds ofthe invention may contain charged side chains or termini, they may be included in any o the above-described formulations as the free acids or bases or as pharmaceutically acceptable salts.
  • Pharmaceutically acceptable salts are those salts which substantially retain the biologic activity ofthe free bases and which are prepared by reaction with inorganic acids. Pharmaceutical salts tend to be more soluble in aqueous and other protic solvents than are the corresponding free base forms.
  • the compounds ofthe invention will generally be used in an amount effective to achieve the intended purpose.
  • the compounds ofthe invention or pharmaceutical compositions thereof are administered or applied in a therapeutically effective amount.
  • therapeutically effective amount is meant an amount effective ameliorate or prevent the symptoms, or prolong the survival of, the patient being treated. Determination of a therapeutically effective amount is well within the capabilities of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • An "inhibitory amount” or “inhibitory concentration" of a PL-PDZ binding inhibitor is an amount that reduces binding by at least about 40%, preferably at least about 50%, often at least about 70%, and even as much as at least about 90%.
  • Binding can as measured in vitro (e.g., in an A assay or G assay) or in situ.
  • a therapeutically effective dose can be estimated initially from in vitro assays.
  • a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC 50 as determined in cell culture (i.e., the concentration of test compound that inhibits 50% of leukocyte surface receptor-PDZ domain- containing protein interactions). Such information can be used to more accurately determine useful doses in humans.
  • Initial dosages can also be estimated from in vivo data, e.g., animal models, using techniques that are well known in the art. One having ordinary skill in the art could readily optimize administration to humans based on animal data.
  • Dosage amount and interval may be adjusted individually to provide plasma levels ofthe compounds that are sufficient to maintain therapeutic effect.
  • Usual patient dosages for administration by injection range from about 0.1 to 5 mg/kg/day, preferably from about 0.5 to 1 mg/kg/day.
  • Therapeutically effective serum levels may be achieved by administering multiple doses each day.
  • the effective local concentration ofthe compounds may not be related to plasma concentration.
  • One having skill in the art will be able to optimize therapeutically effective local dosages without undue experimentation.
  • the amount of compound administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • the therapy may be repeated intermittently while symptoms detectable or even when they are not detectable.
  • the therapy may be provided alone or in combination with other drugs.
  • the drugs that may be used in combination with the compounds of the invention include, but are not limited to, steroid and non-steroid anti-inflammatory agents.
  • a therapeutically effective dose of the compounds described herein will provide therapeutic benefit without causing substantial toxicity.
  • Toxicity of the compounds described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the LD 50 (the dose lethal to 50% of the population) or the LD 100 (the dose lethal to 100% of the population). The dose ratio between toxic and therapeutic effect is the therapeutic index. Compounds which exhibit high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in formulating a dosage range that is not toxic for use in human.
  • the dosage o the compounds described herein lies preferably within a range of circulating concentrations that include the effective dose with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view ofthe patient's condition. (See, e.g., Fingl et al, 1975, In: The Pharmacological Basis of Therapeutics, Ch.l, p.l).
  • Tat-CD3 carboxyl terminus fusion peptide (GYGRKKRRQRRRGPPSSSSGL, SEQ ID NO: 174); Tat- CLASP1 carboxyl terminus fusion peptide, (GYGRKKRRQRRRGSISSSAEV, SEQ ID NOT 75); Tat-CLASP2 carboxyl terminus fusion peptide, (GYGRKKRRQRRRGMTSSSSVV,
  • RPMI 1640 media supplemented with 10% fetal calf serum (HyClone), 2 mM glutamine, 10 mM Hepes, 100 U/ml penicillin, 100 ⁇ g/ml streptomycin, 0.1 mM non-essential amino acids, 1 mM sodium pyruvate, and 50 ⁇ M beta mercaptoethanol.
  • T Cell Stimulation Assay Supernatants were assayed for cytokine production following activation of T cell lines. Mouse T cell lines were stimulated using two different methods, either with antigen and antigen presenting cells or anti-mouse CD3.
  • Antigen-specific mouse T cells BR4.2, were activated with the N-terminal 16 amino acid sequences of myelin basic protein (MBPAcl-16) and syngenic mouse splenocytes in 96-well plates. Mitomycin C-treated antigen presenting cells, 2 x 10 5 B 10.BR, were added to each row of serially diluted MBPAcl-16 ranging from 0 to 200 ⁇ M. Next, 10 ⁇ M Tat- peptides or media alone was added to each row. Finally, 2 x 10 4 MBPAcl-16-specific T cell, pre-loaded with 10 ⁇ M Tat-peptides (see above), were added to all wells (Rabinowitz et al.,1997,. Proc. Natl.
  • Antibody against mouse CD3 (Pharmigen #145-2C11) was coated overnight at 4°C using 96-well flat bottom Elisa plates at a final concentration of 0.5 ⁇ g/ml, diluted in PBS. Just prior to use, plates were washed three times with 200 ⁇ l/well PBS to remove excess anti- CD3. To ensure that cells were given sufficient time to transduce Tat-peptides before activation, T cells (5x10 5 cells/ml) were pre-treated with or without 10 ⁇ M Tat-peptides for two hours at 5% CO2, 37°C and then diluted in media with or without 10 ⁇ M Tat-peptides to a final concentration of 2x10 4 cells per well in a final volume of 200 ⁇ l. Cells were then treated as described above.
  • IFN ⁇ was measured from cell supernatants, described above, at ambient temperature using the Endogen, Inc. ELISA protocol 3. Briefly, 96-well, flat bottom, high binding ELISA plates were preincubated overnight with coating antibody (MM700). Plates were washed with 50 mM TRIS, 0.2% tween-20, pH 8 and they blocked for one hour with PBS plus 2% BSA. Washed plates were then incubated one hour with 25 ⁇ l of cell supernatant and 25 ⁇ l blocking buffer, or with 50 ⁇ l IFN ⁇ standard. The presence of IFN ⁇ was detected with a biotin- labeled anti-mouse IFN ⁇ monoclonal antibody (MM700B, Endogen, Inc.,).
  • MM700B biotin- labeled anti-mouse IFN ⁇ monoclonal antibody
  • Quantitative amounts of detection antibody are revealed with horseradishperoxidase-conjugated streptavidin.
  • the absorbance at 450 nm was measured using a microtiter plate reader (Thermo Max, Molecular Devices) and the concentration of unknown IFN ⁇ from cell supernatants was calculated from a standard curve generated by Softmax Pro. software (Molecular Devices).
  • FIGURE 4A shows that the Tat-CD3 fusion peptide inhibits T cell activation mediated by peptide:MHC as compared to controls of Tat-peptide alone or no peptide.
  • FIGURE 4B shows that Tat-CLASP2 carboxyl terminus fusion peptide inhibited T cell activation mediated by monoclonal anti-CD3 as compared to Tat-peptide alone. Tat-CLASPl fusion peptide did not inhibit T cell activation in this experiment.
  • peptides containing potential inhibitory sequences can be transported into T cells through transporter peptide such as Tat to disrupt surface receptor organization mediated by PDZ proteins. Disruption of PDZ-mediated surface receptor organization leads to blockage of T cell activation in response to antigen.
  • a GST/DLG1 fusion protein See TABLE 3
  • a biotin-labeled peptide corresponding to the C-terminal 20 amino acids of the CLASP-2 protein, peptide AA2L See TABLE 3.
  • 21 uM unlabeled CLASP-2 peptide should be able to block the binding of a given ligand to DLGl by approximately 50%, assuming that the given ligand (1) binds to the same site(s) on DLGl as Qasp ⁇ 2 and (2) is not added at sufficient concentration to reduce significantly the binding ofthe CLASP-2 peptide (i.e. cannot out-compete the CLASP-2 peptide).
  • an effective inhibitor of DLGl -ligand binding i.e. the eight amino acid CLASP-2 peptide MTSSSSVV
  • a known potency range order of magnitude 21 uM

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The invention provides reagents and methods for inhibiting or enhancing interactions between proteins in hematopoietic cells and other cells involved in the mediation of an immune response. Reagents and methods provided are useful for treatment of a variety of diseases and conditions mediated by immune system cells.

Description

MOLECULAR INTERACTIONS IN HEMATOPOIETIC CELLS
REFERENCE TO RELATED APPLICATIONS This application claims priority to U.S. patent applications serial nos. 09/570118,
09/570364, 09/569525 (all filed May 12, 2000), 60/196,460, 60/196,528, and 60/196,527, 60/196267, 09/547,276 (all filed April 11, 2000), serial no. 60/182,296 (filed February 14, 2000); serial no. 60/176,195 (filed January 14, 2000); serial no. 60/170,453 (filed December 13, 1999); serial no. 60/162,498 (filed October 29, 1999); serial no. 60/160,860 (filed October 21, 1999); and serial nos. 60/134,1 8; 60/134,117; and 60/134,114 (all filed May 14, 1999); the disclosures of each of which are incorporated herein in their entirety.
1. FIELD OF THE INVENTION
The present invention relates to peptides and peptide analogues, and methods for using such compositions to regulate activities of cells of the hematopoietic system. In one aspect, the invention provides methods of modulating metabolism (e.g., activation) of hematopoietic cells (e.g., T cells and B cells) by antagonizing an interaction between a PDZ domain containing protein and a protein that binds a PDZ domain. In one aspect, it relates to fusion peptides containing an amino acid sequence corresponding to the carboxyl terminus of a surface receptor expressed by a hematopoietic cell and a transmembrane transporter sequence; such fusion peptides are useful in regulating hematopoietic cells by inhibiting cell activation.
2. BACKGROUND OF THE INVENTION
PDZ domains of proteins are named after three prototypical proteins: PSD95, Drosophila large disc protein and Zonula Occludin 1 protein (Gomperts et al., 1996, Cell 84:659- 662). PDZ domain-containing proteins are involved in synapse formation by organizing transmembrane neurotransmitter receptors through intracellular interactions. PDZ domains contain the signature sequence GLGF. In the nervous system, typical PDZ domain-containing proteins contain three PDZ domains, one SH3 domain and one guanylate kinase domain. Examples of intracellular PDZ domain-containing proteins include LIN-2, LIN-7 and LIN- 10 at the pre-synapse, and PSD95 at the post-synapse. PDZ domains have been shown to bind the carboxyl termini of transmembrane proteins in neuronal cells. Songyang et al. reported that proteins capable of binding PDZ domains contain a carboxyl terminal motif sequence of E-S/T-X-V/I (Songyang et al., 1997, Science 275:73). X-ray crystallography studies have revealed the contact points between the motif sequence and PDZ domains (Doyle et al., 1996, Cell 88:1067-1076). While the interaction between PDZ domains and ion channels in neurons have been studied extensively, such interactions have had limited studies in other biological systems, especially the hematopoietic system.
The hematopoietic system is composed of different cell types that perform distinct functions. Many of its diverse function requires coordinated movement of cell surface receptors including ion channels, adhesion surface molecules to coordinate cell-cell interaction, and cytokine receptors. Despite their diverse functional activities, all hematopoietic cells are believed to develop from a multipotent bone marrow hematopoietic stem cell. Such stem cell has been shown to express a surface marker termed CD34. During differentiation, the stem cell gives rise to progenitor cells in each of several specific hematopoietic cell lineages. The progenitor cells then undergo a series of morphological and functional changes to produce mature functionally committed hematopoietic cells.
Among the functions performed by hematopoietic cells, certain cell types are involved exclusively in immunity. For example, lymphocytes, which include T cells, B cells and natural killer (N ) cells, are effectors in immune responses. Monocytes and granulocytes (i.e., neutrophils, basophils and eosinophils) play a role in non-specific forms of defense. Lymphocytes, monocytes and granulocytes are collectively referred to as white blood cells or leukocytes. On the other hand, other hematopoietic cells perform functions that are unrelated to the immune system. For example, erythrocytes are involved in gas transport, and cells of the thrombocytic series are involved in blood clotting.
T cells and B cells recognize antigens and generate an immune response. T cells recognize antigens by heterodimeric surface receptors termed the T cell receptor (TCR). The TCR is associated with a series of polypeptides collectively referred to as CD3 complex. B cells recognize antigens by surface immunoglobulins (Ig), which are also secretory molecules. In addition, a large number of co-stimulatory surface receptors have been identified in T cells and B cells, which augment cellular activation during antigen-induced activation. In addition to the T cell antigen receptor/CD3 complex (TCR/CD3), other molecules expressed by T cells which mediate an activation signal, include but are not limited to, CD2, CD4, CD5, CD6, CD8, CD18, CD27, CD28, CD43, CD45, CD152 (CTLA-4), CD154, MHC class I, MHC class II, CDwl37 (4-1 BB), CDwl 50, and the like (Barclay et al., The Leucocyte .\ntigen Facts Book, 1997, Second edition, Academic Press; Leucocyte Typing, 1984,
Bernard et al. (eds.), Springer-Verlag; Leukocyte Typing II, 1986, Reinherz et al. (eds.),
Springer-Verlag; Leukocyte Typing III, 1987, McMichael (ed.), Oxford University Press;
Leukocyte Typing IV, 1989, Knapp et al. (eds.), Oxford University Press; CD Antigens, 1996, VI Internal Workshop and Conference on Human Leukocyte Differentiation Antigens. http://www.ncbi.nlm. nih.gov/prow). Cell surface antigens that work together with TCR/CD3 are often referred to as co-receptors in the art.
Specific antibodies have been generated against all ofthe aforementioned T cell surface antigens. Other molecules that bind to the aforementioned T cell surface receptors include antigen-binding antibody derivatives such as variable domains, peptides, superantigens, and their natural ligands such as CD58 (LFA-3) for CD2, HTV gpl20 for CD4, CD27L for CD27,
CD80 or CD86 for CD28 or CD152, ICAM1, ICAM2 and ICAM3 for CDUa/CD18, 4-1BBL for CDwl37.
Activation molecules expressed by B cells, include but are not limited to, surface Ig, CD18, CD19, CD20, CD21, CD22, CD23, CD40, CD45, CD80, CD86 and ICAMl.
Similarly, natural ligands of these molecules and antibodies directed to them as well as antibody derivatives may be used to deliver an activation signal to B cells.
However, prior to the present invention, it was not known that signal transduction following stimulation of any leukocyte receptor was mediated by receptor interactions with PDZ domain-containing proteins. Therefore, it was not even contemplated in the art that an interference of leukocyte surface receptor/PDZ domain interactions could regulate leukocyte activation.
3. SUMMARY OF THE INVENTION
In one aspect, the invention provides a method of modulating a biological function of a cell, e.g., an endothelial cell or hematopoietic cell (such as a a leukocyte, e.g., T cell or B cell), by introducing into the cell an antagonist that inhibits binding of a PDZ protein and a PL protein in the cell, or a agonist that enhances binding of a PDZ protein and a PL protein in the cell. In various embodiments the PL protein is an adhesion protein, an adaptor protein, or an intracellular protein. In embodiments it is CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD4, CD97, CD3n, DOCK2, CD34, FceRIb, or FasLigand. In an embodiment the PL protein is characterized by a carboxy-terminal amino acid motif that is X-S-X-A, X-A-D/E-V, X-V/I/L-X*-V, or X-S/T-X-F (where X is any amino acid and X* is any non-aromatic amino acid). In embodiments, the PL protein is expressed by T lymphocytes or B lymphocytes. In some embodiments of this method, the PDZ protein is CASK, MPP1 , DLG1 , PSD95, NeDLG, SYNl a, TAX43, LDP, LIM, LIMK, AF6, PTN-4, prILlδ, 41.8, GS12, DVL1, TAX 40, TIAM1, MINT1, K303, TAX2, or KIAA561.
In some embodiments, the cell is a leukocyte and the biological function is cell activation, cell proliferation, maintenance of cell structure, cell metabolic activity, or cytokine production. In some embodiments, the method further includes detecting a change in leukocyte activation.
In preferred embodiments, the antagonist is an agent that inhibits the binding of a PL peptide to a PDZ domain polypeptide in an "A" assay, in a "G" assay, or in both an A assay and a G assay. The antagonist can be a polypeptide, such as a polypeptide having at the carboxyterminus at least two residues that are the same as the carboxy-terminal two residues of a PL protein, such as a PL protein is expressed in a hematopoietic or endothelial cell that is an adhesion protein, an adaptor protein, or an intracellular protein. In an embodiment, at least the carboxy-terminal four residues of the polypeptide are the same as the carboxy- terminal four residues of the PL protein. In an embodiment, the PL protein has a carboxy- terminal amino acid motif selected from X-S-X-A, X-A-D/E-V, X-V/I/L-X*-V, or X-S/T-X-F, where X is any amino acid and X* is any non-aromatic amino acid. In embodiment, the PL protein is CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD97, CD3n, DOCK2, CD34, FceRIb, or FasLigand. In a related aspect, the antagonist is a peptide mimetic of a PL inhibitor sequence peptide. In another related aspect the antagonist is a fusion polypeptide having a PL sequence and transmembrane transporter amino acid sequence (such as HIV tat, Drosophila antenapedia, herpes simplex virus VP22 or anti-DNA CDR 2 and 3).
In another aspect, the invention provides a method of determining whether a test compound is an inhibitor of binding between a PDZ protein and a PL protein by contacting a PDZ domain polypeptide having a sequence from the PDZ protein, and a PL peptide under conditions in which they form a complex, in the presence and in the absence of a test compound, and detecting the formation of the complex in the presence and absence of the test compound, where less complex formation in the presence of the test compound than in the absence of the compound indicates that the test compound is an inhibitor of a PDZ protein -PL protein binding. The PL peptide has a sequence that includes the a C-terminal sequence of a PL protein, such as CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD97, CD3n, DOCK2, CD34, FceRIb, or FasLigand. In some embodiments, the PDZ domain polypeptide is a fusion polypeptide.
In a related aspect, the invention provides a method of determining whether a test compound is an agonist of binding between a PDZ protein and a PL protein by contacting a PDZ domain polypeptide, and a PL peptide under conditions in which they form a complex, in the presence and in the absence of a test compound, and detecting the formation ofthe complex in the presence and absence ofthe test compound, where more complex formation in the presence o the test compound than in the absence ofthe compound indicates that the test compound is an agonist of a PDZ protein -PL protein binding.
The invention further provides an inhibitor of binding of a PDZ protein and a PL protein. In an embodiment, the inhibitor is characterized in that it reduces binding of a peptide selected from the group consisting of a PL peptide selected from the group consisting of CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD97, CD3n, DOCK2, CD34, FceRIb, and FasLigand and a PDZ domain polypeptide. In various embodiments, the inhibitor is a peptide comprising a sequence that is from 3 to about 20 residues of a C-terminal sequence of a PL protein selected from CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD97, CD3n, DOCK2, CD34, FceRIb, and FasLigand; a peptide having a motif X-S-X-A, X-A-D/E-V, X-V/I/L-X*-V, or X-S/T-X-F, (where X is any amino acid and X* is any non-aromatic amino acid); a peptide mimetic,; or a small organic molecule. The invention also provides a pharmaceutical composition containing the inhibitor.
The invention also provides a method for treating a disease characterized by leukocyte activation by administering a therapeutically effective amount of an inhibitor of aPL- PDZ interaction. In embodiments, the disease is characterized by an inflammatory or humoral immune response or is an autoimmune disease. The invention further provides a method of reducing inflammation in a subject, by administering an agent that inhibits binding of a PDZ protein and a PL protein, where the PL protein is an adhesion protein, an adaptor protein, or an intracellular protein.
The invention also provides use of an inhibitor of the binding of a PDZ protein and a PL protein to inhibit leukocyte activation or to treat a disease mediated by hematopoietic cells, such as a disease is characterized by an inflammatory or humoral immune response. The invention also provides use of an inhibitor ofthe binding of a PDZ protein and a PL protein in the preparation of a medicament for treatment of a disease mediated by hematopoietic cells. The invention also provides a method of modulating a biological function of a hematopoietic cell, comprising introducing into the cell an antagonist that inhibits binding of a PDZ protein and a PL protein in the cell as deduced from Table 2, for example, where the PL protein is DNAM-1 and the PDZ protein is MPP1, MPP2, DLGl, NeDLG, PSD95, LIM, AF6, 41.8 or RGS12, the PL protein is LPAP and the PDZ protein is DLGl or MINTl, or the PL protein is DNAM-1 and the PDZ protein is PSD95 or MPP2.
The present invention also relates to peptides and peptide analogues that bind PDZ domains in hematopoietic cells. In particular, it relates to fusion peptides and peptide analogues containing a hematopoietic cell surface receptor carboxyl terminal sequence and a transmembrane transporter sequence which facilitates entry ofthe peptides into a target cell. The invention also relates to methods of using such compositions in inhibiting leukocyte activation as measured by cytokine production, cell proliferation, apoptosis and cytotoxicity.
It is an object ofthe invention to administer a therapeutically effective amount of the aforementioned fusion peptides or peptide analogues as pharmaceutical compositions to a subject to inhibit undesirable leukocyte-mediated events.
It is also an object of the invention to administer a therapeutically effective amount of the aforementioned fusion peptides or peptide analogues as pharmaceutical compositions to a subject to treat an autoimmune disorder or to prevent transplantation rejection of a solid organ transplant. In one aspect, the invention provides a method of determining the apparent affinity (Kd) of binding between a PDZ domain and a ligand, by (a) immobilizing a polypeptide comprising the PDZ domain and at least one non-PDZ domain on a surface; (b) contacting the immobilized polypeptide with a plurality of different concentrations of the ligand; (c) determining the amount of binding ofthe ligand to the immobilized polypeptide at each ofthe concentrations of ligand; (d) calculating the apparent affinity of the binding from the binding determined in (c). In an embodiment, the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain. In an embodiment, the polypeptide comprising the PDZ domain is a fusion protein, for example a GST-PDZ domain fusion protein. In one aspect, the invention provides a method of determining the Ki of an inhibitor or suspected inhibitor of binding between a PDZ domain and a ligand, by (a) immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface; (b) contacting the immobilized polypeptide with a plurality of different mixtures ofthe ligand and inhibitor, wherein the different mixtures comprise a fixed amount of ligand, at least a portion of which is detectably labeled, and different concentrations ofthe inhibitor; (c) determining the amount of ligand bound at the different concentrations of inhibitor; (d) calculating the Ki ofthe inhibitor from the binding determined in (c). In an embodiment, the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain. In an embodiment, the fixed amount of ligand is between about 0.01 Kd and about 2 Kd.
In another aspect, the invention provides a method of identifying an agent that enhances the binding of a PDZ domain to a ligand, by immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface; (b) contacting the immobilized polypeptide with the ligand in the presence of a test agent and determining the amount of ligand bound; and, (c) comparing the amount of ligand bound in the presence of the test agent with the amount of ligand bound by the polypeptide in the absence ofthe test agent, wherein at least twofold greater binding in the presence of the test agent compared to the absence ofthe test agent indicates that the test agent is an agent that enhances the binding of the PDZ domain to the ligand. In an embodiment, the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain.
In another aspect, the invention provides a method of determining the potency (Kenhancer) °f an enhancer of binding between a PDZ domain and a ligand, by (a) immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface; (b) contacting the immobilized polypeptide with a plurality of different mixtures of the ligand and enhancer, wherein the different mixtures comprise a fixed amount of ligand, at least a portion of which is detectably labeled, and different concentrations ofthe enhancer; (c) determining the amount of ligand bound at the different concentrations of enhancer; (d) calculating the potency (Kenhancer) of the enhancer from the binding determined in (c). In an embodiment, the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non- PDZ domain, in an embodiment, the fixed amount of ligand is between about 0.01 Kd and about 0.5 Kd.
In another aspect, the invention provides a method of identifying a high specificity interaction between a particular PDZ domain and a ligand known or suspected of binding at least one PDZ domain, by (a) providing a plurality of different immobilized polypeptides, each of said polypeptides comprising a PDZ domain and a non-PDZ domain; (b) determining the affinity of the ligand for each of said polypeptides; (c) comparing the affinity of binding o the ligand to each of said polypeptides. An interaction between the ligand and a particular PDZ domain is deemed to have high specificity when the ligand binds an immobilized polypeptide comprising the particular PDZ domain with at least 2-fold higher affinity than to immobilized polypeptides not comprising the particular PDZ domain (a). In an embodiment, the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non- PDZ domain.
In another aspect, the invention provides a method for determining the PDZ-PL inhibition profile of a compound by (a) providing (i) a plurality of different immobilized polypeptides, each of said polypeptides comprising a PDZ domain and a non-PDZ domain; (ii) a plurality of corresponding ligands, wherein each ligand binds at least one PDZ domain in (i); (b) contacting each of said immobilized polypeptides in (i) with a corresponding ligand in (ii) in the presence and absence of a test compound; (c) determining for each polypeptide-ligand pair in (b) whether the test compound inhibits binding between the immobilized polypeptide and the corresponding ligand thereby determining the PDZ-PL inhibition profile ofthe test compound.
In another aspect, the invention provides an array comprising a plurality of different immobilized polypeptides, each of said polypeptides comprising a PDZ domain and a non-PDZ domain.. In an embodiment, the array is situated in a plastic multiwell plate. In an embodiment, the array has at least 12 different polypeptides comprising at least 12 different PDZ domains, for example, at least 12 different PDZ domains are from PDZs expressed in lymphocytes. In an embodiment, the PDZs are selected from those listed in Table 2 or 6.
In an aspect, the invention provides an assay device comprising a plurality of different immobilized PDZ-containing proteins organized in an array. In one embodiment, the device has at least 25 different PDZ-containing proteins.
In a further aspect, the invention provides a method for identifying an interaction between a PDZ domain and a PL by contacting a PL to a plurality of PDZ containing polypeptides and detecting binding of at least one PL to a PDZ. In an embodiment, the contacting occurs on assay device comprising a plurality of different immobilized PDZ- containing proteins organized in an array. In one embodiment, the device has at least 25 different PDZ-containing proteins. In embodiments, an interaction between a PDZ and more than one PL, or between a PL and more than one PDZ, is detected.
In a related aspect, the invention provides method for identifying a modulator of an interaction between a PDZ and a PL by conducting any ofthe aforementioned assays in the presence and absence of a test compound and detecting a difference in at least one PDZ-PL interaction in the presence and absence ofthe test compound. In embodiments, the the modulator is an enhancer of the interaction. In other embodiments, the modulator is an inhibitor of the interaction.
4. BRIEF DESCRIPTION OF THE DRAWINGS
FIGURES 1A-1D show the results of exemplary assays in which the binding of biotinylated peptides having a sequence of the carboxyl-terminus ("c-terminus") of various leukocyte proteins to PDZ domains (i.e., GST-PDZ domain fusion proteins) was determined using the "G" assay described infra. The PDZ domains are: PSD95 (Fig. 1 A ); NeDLG (Fig. IB); DLGl (Fig 1C); and 41.8 (Fig. ID). These and other PDZ domain fusion proteins are described infra (e.g., TABLE 2). In the figure, peptides 1-31 refer to the biotinylated PL peptide used in the assay, and are identified in the Key, infra. "Peptide IDs" are defined in TABLE 3. Key:
Figure imgf000012_0001
Figure imgf000012_0002
FIGURES 2A and 2B show the Apparent Affinity Determination for PDZ- Ligand Interactions. Varying concentrations of biotinylated CLASP-2 (Fig. 2 A; TABLE 4) or Fas (Fig. 2B; TABLE 4) C-terminal peptides were reacted with immobilized (plate bound) GST polypeptide or GST-PDZ fusion proteins (GST-DLGl, GST-NeDLG, and GST-PSD95). The binding to GST alone (< 0.2 OD units) was subtracted from the binding to the fusion proteins to obtain the signal at each peptide concentration. This signal was then normalized by dividing the signal at each peptide concentration by the maximum signal observed for each peptide-PDZ pair (i.e. the signal obtained at 30 uM Clasp 2 peptide or 100 uM Fas peptide; 0.4 - 1.0 OD units for Clasp 2 and 1.2 - 2.0 OD units for Fas). The normalized signals were then plotted and fit to a saturation binding curve, yielding an apparent affinity of 21 uM for DLGl -Clasp 2 interaction, 7.5 uM for NeDLG-Clasp 2 interaction, 45 uM for PSD95-Clasp 2 interaction, 54 uM for DLGl - Fas interaction, 54 uM for NeDLG-Fas interaction, and 85 uM forPSD95-Fas interaction. Data are means of duplicate data points, with standard errors between duplicate data points < 20%.
FIGURES 3A-3F show inhibition of PDZ - PL peptide interactions. A fixed concentration of biotinylated C-terminal peptide having a sequence based on the C-terminal sequence of a cell surface receptor protein (Clasp 2, CD46, Fas, and KV1.3; see TABLE 4) was bound to immobilized GST polypeptide or the GST-fusion protein indicated at the top left of each frame, in the presence or absence ofthe competitor peptides indicated in the legend of each frame and the level of inhibition determined. Fig. 3 A- DLGl; Fig. 3B- PSD95; Fig. 3C NeDLG; Fig. 3D - DLGl, Fig. 3E, PSD95; Fig. F - 41.8. In Fig. 3A-B the competitor peptides are present at 100 uM; in Figs. 3C-F the competitor is present at the indicated concentration.
FIGURES 4A and 4B shows the results of introduction of a Tat-CD3 fusion peptide on T cell activation. Antigen-specific T cell activation was measured by cytokine production. Fusion peptides containing tat and a T cell surface molecule carboxyl terminus inhibited γ-interferon (IFN) production by a T cell line in response to myelin basic protein (MBP) stimulation. The level of inhibition was determined by first subtracting the binding ofthe labeled peptide to GST alone from the binding to the fusion protein and dividing by the signal in the absence of competitor peptide.
Tables
Table 1 Amino Acid Classification
Table 2 Protein-Ligand Pairs
Table 3 PDZ Domains
Table 3A Note on Table 3
Table 4 PL Peptides
Table 5A&B Exemplary PL Motifs
Table 6 PDZ Domain-Containing Genes Expressed in T Cells and B Cells
5. DEFINITIONS
5.1 A "fusion protein" or "fusion polypeptide" as used herein refers to a composite protein, i.e., a single contiguous amino acid sequence, made up of two (or more) distinct, heterologous polypeptides which are not normally fused together in a single amino acid sequence. Thus, a fusion protein can include a single amino acid sequence that contains two entirely distinct amino acid sequences or two similar or identical polypeptide sequences, provided that these sequences are not normally found together in the same configuration in a single amino acid sequence found in nature. Fusion proteins can generally be prepared using either recombinant nucleic acid methods, i.e., as a result of transcription and translation of a recombinant gene fusion product, which fusion comprises a segment encoding a polypeptide of the invention and a segment encoding a heterologous protein, or by chemical synthesis methods well known in the art.
5.2 A "fusion protein construct" as used herein is a polynucleotide encoding a fusion protein. 5.3 As used herein, the term "PDZ domain" refers to protein sequence (i.e., modular protein domain) of approximately 90 amino acids, characterized by homology to the brain synaptic protein PSD-95, the Drosophila septate junction protein Discs-Large (DLG), and the epithelial tight junction protein ZO1 (ZO1). PDZ domains are also known as Discs-Large homology repeats ("DHRs") and GLGF repeats). PDZ domains generally appear to maintain a core consensus sequence (Doyle, D. A., 1996, Cell 85: 1067-1076).
PDZ domains are found in diverse membrane-associated proteins including members ofthe MAGUK family of guanylate kinase homologs, several protein phosphatases and kinases, neuronal nitric oxide synthase, and several dystrophin-associated proteins, collectively known as syntrophins.
Exemplary PDZ domain-containing proteins and PDZ domain sequences are shown in TABLE 3. The term "PDZ domain" also encompasses variants (e.g., naturally occuring variants) of the sequences of TABLE 3 (e.g., polymorphic variants, variants with conservative substitutions, and the like). Typically, PDZ domains are substantially identical to those shown in TJ BLE 3, e.g., at least about 70%, at least about 80%, or at least about 90% amino acid residue identity when compared and aligned for maximum correspondence.
5.4 As used herein, the term "PDZ protein" refers to a naturally occurring protein containing a PDZ domain, e.g., a human protein. Exemplary PDZ proteins include CASK, MPPl, DLGl, PSD95, NeDLG, TAX33, SYNla, TAX43, LDP, LIM, LIMKl, LIMK2, MPP2, NOS1 , AF6, PTN-4, prILlβ. 41.8kD, KIAA0559, RGS12, KIAA0316, DVL1, TAX40, TIAM1, MINTl, KIAA0303, CBP, MLNT3, TAX2, KIAA0561. Exemplary PDZ proteins are listed in TABLE 2 and TABLE 3.
5.5 As used herein, the term "PDZ-domain polypeptide" refers to a polypeptide containing a PDZ domain, such as a fusion protein including a PDZ domain sequence, a naturally occurring PDZ protein, or an isolated PDZ domain peptide.
5.6 As used herein, the term "PL protein" or "PDZ Ligand protein" refers to a naturally occurring protein that forms a molecular complex with a PDZ-domain, or to a protein whose carboxy-terminus, when expressed separately from the full length protein (e.g., as a peptide fragment of 4-25 residues, e.g., 16 residues), forms such a molecular complex. The molecular complex can be observed in vitro using the "A assay" or "G assay" described infra, or in vivo. Exemplary PL proteins listed in TABLE 2 are demonstrated to bind specific PDZ proteins. This definition is not intended to include anti-PDZ antibodies and the like.
5.7 As used herein, a "PL sequence" refers to the amino acid sequence of the C- terminus of a PL protein (e.g., the C-terminal 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 20 or 25 residues) ("C-terminal PL sequence") or to an internal sequence known to bind a PDZ domain ("internal PL sequence)..
5.8 As used herein, a "PL peptide" is a peptide of having a sequence from, or based on, the sequence of the C-terminus of a PL protein. Exemplary PL peptides (biotinylated) are listed in TABLE 4.
5.9 As used herein, a "PL fusion protein" is a fusion protein that has a PL sequence as one domain, typically as the C-terminal domain of the fusion protein. An exemplary PL fusion protein is a tat-PL sequence fusion.
5.10 As used herein, the term "PL inhibitor peptide sequence" refers to PL peptide an amino acid sequence that (in the form of a peptide or PL fusion protein) inhibits the interaction between a PDZ domain polypeptide and a PL peptide (e.g., in an A assay or a G assay).
5.11 As used herein, a "PDZ-domain encoding sequence" means a segment of a polynucleotide encoding a PDZ domain. In various embodiments, the polynucleotide is DNA, RNA, single stranded or double stranded.
5.12 As used herein, the terms "antagonist" and "inhibitor," when used in the context of modulating a binding interaction (such as the binding of a PDZ domain sequence to a PL sequence), are used interchangeably and refer to an agent that reduces the binding of the, e.g., PL sequence (e.g., PL peptide) and the, e.g., PDZ domain sequence (e.g., PDZ protein, PDZ domain peptide). 5.13 As used herein, the terms "agonist" and "enhancer," when used in the context of modulating a binding interaction (such as the binding of a PDZ domain sequence to a PL sequence), are used interchangeably and refer to an agent that increases the binding ofthe, e.g.,
PL sequence (e.g., PL peptide) and the, e.g., PDZ domain sequence (e.g., PDZ protein, PDZ domain peptide).
5.14 As used herein, the terms "peptide mimetic, " "peptidomimetic," and "peptide analog" are used interchangeably and refer to a synthetic chemical compound which has substantially the same structural and/or functional characteristics of an PL inhibitory or PL binding peptide ofthe invention. The mimetic can be either entirely composed of synthetic, non- natural analogues of amino acids, or, is a chimeric molecule of partly natural peptide amino acids and partly non-natural analogs of amino acids. The mimetic can also incorporate any amount of natural amino acid conservative substitutions as long as such substitutions also do not substantially alter the mimetic's structure and/or inhibitory or binding activity. As with polypeptides of the invention which are conservative variants, routine experimentation will determine whether a mimetic is within the scope of the invention, i.e., that its structure and/or function is not substantially altered. Thus, a mimetic composition is within the scope of the invention if it is capable of binding to a PDZ domain and/or inhibiting a PL-PDZ interaction.
Polypeptide mimetic compositions can contain any combination of nonnatural structural components, which are typically from three stmctural groups: a) residue linkage groups other than the natural amide bond ("peptide bond") linkages; b) non-natural residues in place of naturally occurring amino acid residues; or c) residues which induce secondary structural mimicry, i.e., to induce or stabilize a secondary structure, e.g., a beta turn, gamma turn, beta sheet, alpha helix conformation, and the like. A polypeptide can be characterized as a mimetic when all or some of its residues are joined by chemical means other than natural peptide bonds. Individual peptidomimetic residues can be joined by peptide bonds, other chemical bonds or coupling means, such as, e.g., glutaraldehyde, N-hydroxysuccinimide esters, bi functional maleimides, N,N=- dicyclohexylcarbodiimide (DCC) or N,N=-diisopropylcarbodiimide (DIC). Linking groups that can be an alternative to the traditional amide bond ("peptide bond") linkages include, e.g., ketomethylene (e.g., -C(=O)-CH,- for -C(=O)-NH-), aminomethylene (CH2-NH), ethylene, olefin (CH=CH), ether (CH2-O), thioether (CH2-S), tetrazole (CN4-), thiazole, retroamide, thioamide, or ester (see, e.g., Spatola (1983) in Chemistry and
Figure imgf000017_0001
Acids, Peptides and Proteins, Vol. 7, pp 267-357, A Peptide Backbone Modifications, Marcell Dekker, NY).
A polypeptide can also be characterized as a mimetic by containing all or some non-natural residues in place of naturally occurring amino acid residues. Nonnatural residues are well described in the scientific and patent literature; a few exemplary nonnatural compositions useful as mimetics of natural amino acid residues and guidelines are described below.
Mimetics of aromatic amino acids can be generated by replacing by, e.g., D- or L- naphylalanine; D- or L- phenylglycine; D- or L-2 thieneylalanine; D- or L-l, -2, 3-, or 4- pyreneylalanine; D- or L-3 thieneylalanine; D- or L-(2-pyridinyl)-alanine; D- orL-(3-pyridinyl)- alanine; D- or L-(2-pyrazinyl)-alanine; D- or L-(4-isopropyl)-phenylglycine; D-(trifluoromethyl)- phenylglycine; D-(trifluoromethyl)-phenylalanine; D-p-fluorophenylalanine; D- or L-p- biphenylphenylalanine; K- or L-p-methoxybiphenylphenylalanine; D- or L-2- indole(alkyl)alanines; and, D- or L-alkylainines, where alkyl can be substituted or unsubstituted methyl, ethyl, propyl, hexyl, butyl, pentyl, isopropyl, iso-butyl, sec-isotyl, iso-pentyl, or a non- acidic amino acids. Aromatic rings of a nonnatural amino acid include, e.g., thiazolyl, thiophenyl, pyrazolyl, benzimidazolyl, naphthyl, furanyl, pyrrolyl, and pyridyl aromatic rings. Mimetics of acidic amino acids can be generated by substitution by, e.g., non- carboxylate amino acids while maintaining a negative charge; (phosphono)alanine; sulfated threonine. Carboxyl side groups (e.g., aspartyl or glutamyl) can also be selectively modified by reaction with carbodiimides (R=-N-C-N-R=) such as, e.g., l -cyclohexyl-3(2-morpholinyl-(4- ethyl) carbodiimide or l-ethyl-3(4-azonia- 4,4- dimetholpentyl) carbodiimide. Aspartyl or glutamyl can also be converted to asparaginyl and glutaminyl residues by reaction with ammonium ions.
Mimetics of basic amino acids can be generated by substitution with, e.g., (in addition to lysine and arginine) the amino acids ornithine, citrulline, or (guanidino)-acetic acid, or (guanidino)alkyl-acetic acid, where alkyl is defined above. Nitrile derivative (e.g., containing the CN-moiety in place of COOH) can be substituted for asparagine or glutamine. Asparaginyl and glutaminyl residues can be deaminated to the corresponding aspartyl or glutamyl residues.
Arginine residue mimetics can be generated by reacting arginyl with, e.g., one or more conventional reagents, including, e.g., phenylglyoxal, 2,3-butanedione, 1,2- cyclohexanedione, or ninhydrin, preferably under alkaline conditions.
Tyrosine residue mimetics can be generated by reacting tyrosyl with, e.g., aromatic diazonium compounds or tetranitromethane. N-acetylimidizol and tetranitromethane can be used to form O-acetyl tyrosyl species and 3-nitro derivatives, respectively.
Cysteine residue mimetics can be generated by reacting cysteinyl residues with, e.g., alpha-haloacetates such as 2-chloroacetic acid or chloroacetamide and corresponding amines; to give carboxymethyl or carboxyamidomethyl derivatives. Cysteine residue mimetics can also be generated by reacting cysteinyl residues with, e.g., bromo-trifluoroacetone, alpha- bromo-beta-(5-imidozoyl) propionic acid; chloroacetyl phosphate, N-alkylmaleimides, 3-nitro-2- pyridyl disulfide; methyl 2-pyridyl disulfide; p-chloromercuribenzoate; 2-chloromercuri-4 nitrophenol; or, chloro-7-nitrobenzo-oxa-l,3-diazole.
Lysine mimetics can be generated (and amino terminal residues can be altered) by reacting lysinyl with, e.g., succinic or other carboxylic acid anhydrides. Lysine and other alpha-amino-containing residue mimetics can also be generated by reaction with imidoesters, such as methyl picolinimidate, pyridoxal phosphate, pyridoxal, chloroborohydride, trinitrobenzenesulfonic acid, O-methylisourea, 2,4, pentanedione, and transamidase-catalyzed reactions with glyoxylate.
Mimetics of methionine can be generated by reaction with, e.g., methionine sulfoxide. Mimetics of proline include, e.g., pipecolic acid, thiazohdine carboxylic acid, 3- or
4- hydroxy proline, dehydroproline, 3- or 4-methylproline, or 3,3,-dimethylproline. Histidine residue mimetics can be generated by reacting histidyl with, e.g., diethylprocarbonate or para- bromophenacyl bromide.
Other mimetics include, e.g., those generated by hydroxylation of proline and lysine; phosphorylation ofthe hydroxyl groups of seryl or threonyl residues; methylation ofthe alpha-amino groups of lysine, arginine and histidine; acetylation of the N-terminal amine; methylation of main chain amide residues or substitution with N-methyl amino acids; or amidation of C-terminal carboxyl groups.
A component of a natural polypeptide (e.g., a PL polypeptide or PDZ polypetide) can also be replaced by an amino acid (or peptidomimetic residue) of the opposite chirality. Thus, any amino acid naturally occurring in the L-con figuration (which can also be referred to as the R or S, depending upon the structure ofthe chemical entity) can be replaced with the amino acid of the same chemical structural type or a peptidomimetic, but of the opposite chirality, generally referred to as the D- amino acid, but which can additionally be referred to as the R- or S- form. The mimetics of the invention can also include compositions that contain a structural mimetic residue, particularly a residue that induces or mimics secondary structures, such as a beta turn, beta sheet, alpha helix structures, gamma turns, and the like. For example, substitution of natural amino acid residues with D-amino acids; N-alpha-methyl amino acids; C- alpha-methyl amino acids; or dehydroamino acids within a peptide can induce or stabilize beta turns, gamma turns, beta sheets or alpha helix conformations. Beta turn mimetic structures have been described, e.g., by Nagai (1985) Tet. Lett. 26:647-650; Feigl (1986) J. Amer. Chem. Soc. 108: 181-182; Kahn (1988) J. Amer. Chem. Soc. 110:1638-1639; Kemp (1988) Tet. Lett. 29:5057-5060; Kahn (1988) J. Molec. Recognition 1 :75-79. Beta sheet mimetic structures have been described, e.g., by Smith (1992) J. Amer. Chem. Soc. 114:10672-10674. For example, a type VI beta turn induced by a cis amide surrogate, 1 ,5-disubstituted tetrazol, is described by Beusen (1995) Biopolymers 36:181-200. Incorporation of achiral omega-amino acid residues to generate polymethylene units as a substitution for amide bonds is described by Banerjee
(1996) Biopolymers 39:769-777. Secondary structures of polypeptides can be analyzed by, e.g., high-field 1H NMR or 2D NMR spectroscopy, see, e.g., Higgins (1997) J. Pept. Res.
50:421-435. See also, Hruby (1997) Biopolymers 43:219-266, Balaji, et al., U.S. Pat. No. 5,612,895.
5.15 As used herein, "peptide variants" and "conservative amino acid substitutions" refer to peptides that differ from a reference peptide (e.g., a peptide having the sequence of the c arboxy-terminus of a specified PL protein) by substitution of an amino acid residue having similar properties (based on size, polarity, hydrophobicity, and the like). Thus, insofar as the compounds that are encompassed within the scope of the invention are partially defined in terms of amino acid residues of designated classes, the amino acids may be generally categorized into three main classes: hydrophilic amino acids, hydrophobic amino acids and cysteine-like amino acids, depending primarily on the characteristics of the amino acid side chain. These main classes may be further divided into subclasses. Hydrophilic amino acids include amino acids having acidic, basic or polar side chains and hydrophobic amino acids include amino acids having aromatic or apolar side chains. Apolar amino acids may be further subdivided to include, among others, aliphatic amino acids. The definitions of the classes of amino acids as used herein are as follows:
"Hydrophobic Amino Acid" refers to an amino acid having a side chain that is uncharged at physiological pH and that is repelled by aqueous solution. Examples of genetically encoded hydrophobic amino acids include He, Leu and Val. Examples of non-genetically encoded hydrophobic amino acids include t-BuA.
"Aromatic Amino Acid" refers to a hydrophobic amino acid having a side chain containing at least one ring having a conjugated π-electron system (aromatic group). The aromatic group may be further substituted with groups such as alkyl, alkenyl, alkynyl, hydroxyl, sulfanyl, nitro and amino groups, as well as others. Examples of genetically encoded aromatic amino acids include Phe, Tyr and Trp. Commonly encountered non-genetically encoded aromatic amino acids include phenylglycine, 2-naphthylalanine, β-2-thienylalanine, 1,2,3,4- tetrahydroisoquinoline-3-carboxylic acid, 4-chloro-phenylalanine, 2-fluorophenylalanine, 3- fluorophenylalanine and 4-fluorophenylalanine. "Apolar Amino Acid" refers to a hydrophobic amino acid having a side chain that is generally uncharged at physiological pH and that is not polar. Examples of genetically encoded apolar amino acids include Gly, Pro and Met. Examples of non-encoded apolar amino acids include Cha.
"Aliphatic Amino Acid" refers to an apolar amino acid having a saturated or unsaturated straight chain, branched or cyclic hydrocarbon side chain. Examples of genetically encoded aliphatic amino acids include Ala, Leu, Val and lie. Examples of non-encoded aliphatic amino acids include Nle.
"Hydrophilic Amino Acid" refers to an amino acid having a side chain that is attracted by aqueous solution. Examples of genetically encoded hydrophilic amino acids include Ser and Lys. Examples of non-encoded hydrophilic amino acids include Cit and hCys.
"Acidic Amino Acid" refers to a hydrophilic amino acid having a side chain pK value of less than 7. Acidic amino acids typically have negatively charged side chains at physiological pH due to loss of a hydrogen ion. Examples of genetically encoded acidic amino acids include Asp and Glu. "Basic Amino Acid" refers to a hydrophilic amino acid having a side chain pK value of greater than 7. Basic amino acids typically have positively charged side chains at physiological pH due to association with hydronium ion. Examples of genetically encoded basic amino acids include Arg, Lys and His. Examples of non-genetically encoded basic amino acids include the non-cyclic amino acids omithine, 2,3-diaminopropionic acid, 2,4-diaminobutyric acid and homoarginine. "Polar Amino Acid" refers to a hydrophilic amino acid having a side chain that is uncharged at physiological pH, but which has a bond in which the pair of electrons shared in common by two atoms is held more closely by one of the atoms. Examples of genetically encoded polar amino acids include Asx and Glx. Examples of non-genetically encoded polar amino acids include citrulline, N-acetyl lysine and methionine sulfoxide. "Cysteine-Like Amino Acid" refers to an amino acid having a side chain capable of forming a covalent linkage with a side chain of another amino acid residue, such as a disulfide linkage. Typically, cysteine-like amino acids generally have a side chain containing at least one thiol (SH) group. Examples of genetically encoded cysteine-like amino acids include Cys. Examples of non-genetically encoded cysteine-like amino acids include homocysteine and penicillamine.
As will be appreciated by those having skill in the art, the above classification are not absolute — several amino acids exhibit more than one characteristic property, and can therefore be included in more than one category. For example, tyrosine has both an aromatic ring and a polar hydroxyl group. Thus, tyrosine has dual properties and can be included in both the aromatic and polar categories. Similarly, in addition to being able to form disulfide linkages, cysteine also has apolar character. Thus, while not strictly classified as a hydrophobic or apolar amino acid, in many instances cysteine can be used to confer hydrophobicity to a peptide.
Certain commonly encountered amino acids which are not genetically encoded of which the peptides and peptide analogues ofthe invention may be composed include, but are not limited to, β-alanine (b-Ala) and other omega-amino acids such as 3-aminopropionic acid (Dap), 2,3-diaminopropionic acid (Dpr), 4-aminobutyric acid and so forth; α-aminoisobutyric acid (Aib); ε-aminohexanoic acid (Aha); δ-aminovaleric acid (Ava); N-methylglycine or sarcosine (MeGly); ornithine (Orn); citrulline (Cit); t-butylalanine (t-BuA); t-butylglycine (t-BuG); N-methylisoleucine (Melle); phenylglycine (Phg); cyclohexylalanine (Cha); norleucine (Nle); 2-naphthylalanine (2-Nal); 4-chlorophenylalanine (Phe(4-Cl)); 2-fluorophenylalanine (Phe(2-F)); 3-fluorophenylalanine (Phe(3-F)); 4-fluorophenylalanine (Phe(4-F)); penicillamine (Pen); l,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (Tic); β-2-thienylalanine (Thi); methionine sulfoxide (MSO); homoarginine (hArg); N-acetyl lysine (AcLys); 2,3-diaminobutyric acid (Dab); 2,3-diaminobutyric acid (Dbu); p-aminophenylalanine (Phe(pNH2)); N-methyl valine (Me Val); homocysteine (hCys) and homoserine (hSer). These amino acids also fall conveniently into the categories defined above.
The classifications ofthe above-described genetically encoded and non-encoded amino acids are summarized in TABLE 1, below. It is to be understood that TABLE 1 is for illustrative purposes only and does not purport to be an exhaustive list of amino acid residues which may comprise the peptides and peptide analogues described herein. Other amino acid residues which are useful for making the peptides and peptide analogues described herein can be found, e.g., in Fasman, 1989, CRC Practical Handbook of Biochemistry and Molecular Biology, CRC Press, Inc., and the references cited therein. Amino acids not specifically mentioned herein can be conveniently classified into the above-described categories on the basis of known behavior and/or their characteristic chemical and/or physical properties as compared with amino acids specifically identified.
TABLE 1
Classification Genetically Encoded Genetically Non-Encoded
Hydrophobic
Aromatic F, Y, W Phg, Nal, Thi, Tic, Phe(4-Cl), Phe(2-F), Phe(3-F), Phe(4-F), Pyridyt Ala, Benzothienyl Ala Apolar M, G, P
Aliphatic A, V, L, I t-BuA, t-BuG, Melle, Nle, MeVal, Cha, bAla, MeGly, Aib
Hydrophilic
Acidic D, E
Basic H, K, R Dpr, Om, hArg, Phe(p-NH2), DBU, A2BU
Polar Q, N, S, T, Y Cit, AcLys, MSO, hSer
Cysteine-Like C Pen, hCys, p-methyl Cys
5.16 As used herein, a "detectable label" has the ordinary meaning in the art and refers to an atom (e.g., radionuciide), molecule (e.g., fluorescein), or complex, that is or can be used to detect (e.g., due to a physical or chemical property), indicate the presence of a molecule or to enable binding of another molecule to which it is covalently bound or otherwise associated. The term "label" also refers to covalently bound or otherwise associated molecules (e.g., a biomolecule such as an enzyme) that act on a substrate to produce a detectable atom, molecule or complex. Detectable labels suitable for use in the present invention include any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Labels useful in the present invention include biotin for staining with labeled streptavidin conjugate, magnetic beads (e.g., DynabeadsTM), fluorescent dyes (e.g., fluorescein, Texas red, rhodamine, green fluorescent protein, enhanced green fluorescent protein, and the like), radiolabels (e.g., 3H, 1251, 35S, 14C, or 32P), enzymes ( e.g., hydrolases, particularly phosphatases such as alkaline phosphatase, esterases and glycosidases, or oxidoreductases, particularly peroxidases such as horse radish peroxidase, and others commonly used in ELISAs), substrates, cofactors, inhibitors, chemiluminescent groups, chromogenic agents, and colorimetric labels such as colloidal gold or colored glass or plastic (e.g., polystyrene, polypropylene, latex, etc.) beads. Patents teaching the use of such labels include U.S. Patent Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241. Means of detecting such labels are well known to those of skill in the art. Thus, for example, radiolabels and chemiluminescent labels may be detected using photographic film or scintillation counters, fluorescent markers may be detected using a photodetector to detect emitted light (e.g., as in fluorescence-activated cell sorting). Enzymatic labels are typically detected by providing the enzyme with a substrate and detecting the reaction product produced by the action of the enzyme on the substrate, and colorimetric labels are detected by simply visualizing the colored label. Thus, a label is any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. The label may be coupled directly or indirectly to the desired component ofthe assay according to methods well known in the art. Non-radioactive labels are often attached by indirect means. Generally, a ligand molecule (e.g., biotin) is covalently bound to the molecule. The ligand then binds to an anti-ligand (e.g., streptavidin) molecule which is either inherently detectable or covalently bound to a signal generating system, such as a detectable enzyme, a fluorescent compound, or a chemiluminescent compound. A number of ligands and anti-ligands can be used. Where a ligand has a natural anti-ligand, for example, biotin, thyroxine, and cortisol, it can be used in conjunction with the labeled, naturally occurring anti-ligands. Alternatively, any haptenic or antigenic compound can be used in combination with an antibody. The molecules can also be conjugated directly to signal generating compounds, e.g., by conjugation with an enzyme or fluorophore. Means of detecting labels are well known to those of skill in the art. Thus, for example, where the label is a radioactive label, means for detection include a scintillation counter, photographic film as in autoradiography, or storage phosphor imaging. Where the label is a fluorescent label, it may be detected by exciting the fluorochrome with the appropriate wavelength of light and detecting the resulting fluorescence. The fluorescence may be detected visually, by means of photographic film, by the use of electronic detectors such as charge coupled devices (CCDs) or photomulfipliers and the like. Similarly, enzymatic labels may be detected by providing the appropriate substrates for the enzyme and detecting the resulting reaction product. Also, simple colorimetric labels may be detected by observing the color associated with the label. It will be appreciated that when pairs of fluorophores are used in an assay, it is often preferred that they have distinct emission patterns (wavelengths) so that they can be easily distinguished.
5. 17 As used herein, the term "substantially identical" in the context of comparing amino acid sequences, means that the sequences have at least about 70%, at least about 80%), or at least about 90% amino acid residue identity when compared and aligned for maximum correspondence. An algorithm that is suitable for determining percent sequence identity and sequence similarity is the FASTA algorithm, which is described in Pearson, W.R. & Lipman, D.J., 1988, Proc. Natl. Acad. Sci. U.S.A. 85: 2444. See also W. R. Pearson, 1996, Methods Enzymol. 266: 227-258. Preferred parameters used in a FASTA alignment of DNA sequences to calculate percent identity are optimized, BL50 Matrix 15: -5, k-tuple = 2; joining penalty = 40, optimization = 28; gap penalty -12, gap length penalty =-2; and width = 16.
5.18 As used herein, "hematopoietic cells" include leukocytes including lymphocytes (T cells, B cells and NK cells), monocytes, and granulocytes (i.e., neutrophils, basophils and eosinophils), macrophages, dendritic cells, megakaryocytes, reticulocytes, erythrocytes, and CD34÷ stem cells.
?? 5.19 As used herein, the terms "test compound" or "test agent" are used interchangably and refer to a candidate agent that may have enhancer/agonist, or inhibitor/antagonist activity, e.g., inhibiting or enhancing an interaction such as PDZ-PL binding. The candidate agents or test compounds may be any of a large variety of compounds, both naturally occurring and synthetic, organic and inorganic, and including polymers (e.g., oligopeptides, polypeptides, ohgonucleotides, and polynucleotides), small molecules, antibodies (as broadly defined herein), sugars, fatty acids, nucleotides and nucleotide analogs, analogs of naturally occurring structures (e.g., peptide mimetics, nucleic acid analogs, and the like), and numerous other compounds. In certain embodiment, test agents are prepared from diversity libraries, such as random or combinatorial peptide or non-peptide libraries. Many libraries are known in the art that can be used, e.g., chemically synthesized libraries, recombinant (e.g., phage display libraries), and in vitro translation-based libraries. Examples of chemically synthesized libraries are described in Fodor et al, 1991, Science 251 :767-773; Houghten et al., 1991, Nature 354:84-86; Lam et al., 1991, Nature 354:82-84; Medynski, 1994, Bio/Technology 12:709-710; Gallop et al., 1994, J. Medicinal Chemistry 37(9): 1233- 1251; Ohlmeyer et al., 1993, Proc. Natl. Acad. Sci. USA 90:10922-10926; Erb et al, 1994, Proc. Natl. Acad. Sci. USA 91 :11422-11426; Houghten et al., 1992, Biotechniques 13:412; Jayawickreme et al., 1994, Proc. Natl. Acad. Sci. USA 91 :1614-1618; Salmon et al., 1993, Proc. Natl. Acad. Sci. USA 90:11708-11712; PCT Publication No. WO 93/20242; and Brenner and Lerner, 1992, Proc. Natl. Acad. Sci. USA 89:5381-5383. Examples of phage display libraries are described in Scott and Smith, 1990, Science 249:386-390; Devlin et al, 1990, Science, 249:404-406; Christian, R.B., et al., 1992, J. Mol. Biol. 227:71 1-718); Lenstra, 1992, J. Immunol. Meth. 152:149-157; Kay et al., 1993, Gene 128:59-65; and PCT Publication No. WO 94/18318 dated August 18, 1994. In vitro translation- based libraries include but are not limited to those described in PCT Publication No. WO 91/05058 dated April 18, 1991 ; and Mattheakis et al., 1994, Proc. Natl. Acad. Sci. USA 91:9022-9026. By way of examples of nonpeptide libraries, a benzodiazepine library (see e.g., Bunin et al., 1994, Proc. Natl. Acad. Sci. USA 91 :4708-4712) can be adapted for use. Peptoid libraries (Simon et al., 1992, Proc. Natl. Acad. Sci. USA 89:9367-9371) can also be used. Another example of a library that can be used, in which the amide functionalities in peptides have been permethylated to generate a chemically transformed combinatorial library, is described by Ostresh et al. (1994, Proc. Natl. Acad. Sci. USA 91 : 1 1138-11142). 5.20 The term "specific binding" refers to binding between two molecules, for example, a ligand and a receptor, characterized by the ability of a molecule (ligand) to associate with another specific molecule (receptor) even in the presence of many other diverse molecules, i.e., to show preferential binding of one molecule for another in a heterogeneous mixture of molecules. Specific binding of a ligand to a receptor is also evidenced by reduced binding of a detectably labeled ligand to the receptor in the presence of excess unlabeled ligand (i.e., a binding competition assay).
5.21 As used herein, a "plurality" of PDZ proteins (or corresponding PDZ domains or PDZ fusion polypeptides) has its usual meaning. In some embodiments, the plurality is at least 5, and often at least 25, at least 40 or at least 60 different PDZ proteins. In some embodiments, the plurality is selected from the list of PDZ polypeptides listed in Table 2 or Table 6. In some embodiments, the plurality of different PDZ proteins are from (i.e., expressed in) a particular specified tissue or a particular class or type of cell. In some embodiments, the plurality of different PDZ proteins represents a substantial fraction (e.g., typically at least 50%, more often at least 80%) of all ofthe PDZ proteins known to be, or suspected of being, expressed in the tissue or cell(s), e.g., all of the PDZ proteins known to be present in lymphocytes or hematopoetic cells. In some embodiments, the plurality is at least 50%, usually at least 80%, at least 90% or all ofthe PDZ proteins disclosed herein as being expressed in hematopoietic cells (see Tables 2 and 6). I an embodiment, the plurality includes at least 1, often at least 2, sometimes at least 5 or at least 10 and sometimes all of the following PDZ proteins: BAl I associated prof, Connector enhancer, DLG5 (pdlg), DVL3, GTPase, Guanin-exchange factor 1, PDZ domain containing prot, KLAA147, KIAA0300, KIAA0380, KIAA0440, KIAA0545, KIAA0807, KIAA0858, KIAA0902, novel serine protease, PDZKl, PICK8, PTN-3, RPIP8, serine protease, 26s subunit p27, hSYNTENTN, TAXI -IP, TAX2-like protein, wwp3, XI 1 prot. beta, ZO1. When referring to PL ligands or corresponding PL proteins (e.g., corresponding to those listed in Table 2, Table 4, Table 5, or elsewhere herein) a "plurality" may refer to at least 5, at least 10, and often at least 25 PLs such as those specifcally listed herein, or to the classes and percentages set forth supra for PDZ domains. 6. DETAILED DESCRIPTION OF THE INVENTION
The present inventors have discovered that interactions between PDZ proteins and PL proteins play an important and extensive role in the biological function of hematopoietic cells and other cells involved in the immune response. Although PDZ-PL interactions were known in the nervous system (i.e., in neurons), their universal importance in hematopoietic cell function, especially in function of T cells and B cells, and their fundamental role in modulation of the immune response has not been recognized. In particular, the present inventors have surprisingly discovered that cell adhesion molecules that mediate cell-cell interaction in the hematopoietic system are PDZ-binding proteins (PL proteins) and bind to PDZ proteins. The inventors have identified numerous interactions between PDZ proteins and PL proteins present in immune system cells, and the invention provides reagents and methods for affecting biological function in the immune system by inhibiting these interactions. As used herein, the term "biological function" in the context of a cell, refers to a detectable biological activity normally carried out by the cell, e.g., a phenotypic change such as proliferation, cell activation (e.g., T cell activation, B cell activation, T-B cell conjugate formation), cytokine release, degranulation, tyrosine phosphorylation, ion (e.g., calcium) flux, metabolic activity, apoptosis, changes in gene expression, maintenance of cell structure, cell migration, adherance to a substrate, signal transduction, cell-cell interactions, and others described herein or known in the art.
In one aspect, the present invention relates to peptides, peptide analogues or mimetics, pharmaceutical compositions, and methods of using such compositions to regulate the biological activities of hematopoietic cells, e.g. T cells and B cells, or other cells (e.g., endothelial cells) that necessary for immune function. The invention further relates to methods of using the compositions to modulate hematopoietic cell activation and immune function, as well as assays for such inhibitors. TABLE 2 summarizes an extensive analysis of protein interactions in T cells and
B cells. PDZ proteins, the vast majority of which were not previously known to be expressed in immune system cells, are listed in the top row of TABLE 2. The first column ofthe table lists PL proteins. Positions in the matrix denoted by the letter "A" or "G" indicate that an interaction between the PDZ protein and the PL has been detected in novel binding assays (described in detail in, e.g., Section 6.2, infra). A blank cell indicates that no interaction was detected using the assays of the invention. An asterisk (*) denotes a PL-PDZ interaction previously reported in the scientific literature. PDZ-LIGAND/PDZ INTERACTION TABLE 2 SUMMARY
Figure imgf000028_0001
Intβcuc iαna dB3crib»d la th« scionti-ie literature PDZ-LIGAND/PDZ INTERACTION TABLEΪ2 . SUMMARY CONTINUED
Figure imgf000029_0001
Intenctioc-- es ribed In t « sci.antJ.fic literature As discussed in detail herein, the PDZ proteins listed in TABLE 2 are naturally occurring proteins containing a PDZ domain. The present invention is particularly directed to the detection and modulation of interactions between PDZ proteins and PL proteins in hematopoietic cells. Exemplary PL proteins are listed in TJABLE 2. Notably, as discussed infra, many of these PL proteins have not previously been recognized as such in any cell system. A variety of PL protein classes are known, and the PL proteins described herein can be characterized as (1) "PL adhesion proteins" (2) "PL ion channel proteins" (3) "PL adaptor proteins" (4) "PL intracellular proteins" and (5) "PL cytokine receptor proteins."
As used herein, an adhesion protein is a cell surface protein involved in cell-cell interaction by direct contact with cell surface molecules (e.g., transmembrane proteins or surface proteins) on a different cell. Thus, when a cell expressing a PL adhesion protein contacts an appropriate other cell, the PL adhesion protein localizes at the interface of the two cells and directly contacts a cell surface molecule on the second cell. A cell-cell interface is a region where the plasma membranes of two different cells are in close (generally <10 nm, often about 1 nm) apposition. Typically, direct molecular contact means interaction of molecules at distances where Van der Walls forces are significant, generally less than about 1 nm. Exemplary PL adhesion proteins include CD6; CD49E (alpha-4); CD49F (a form, alphaβ); CD138 (syndecan); CLASP-1; CLASP-4; VCAMl; CLASP-2; DNAM-1; CD83; CD44 (long form); CD97; (CD55L); CD3n; DOCK2; CD34; and FceRIb. Thus, in one embodiment, the PL proteins of the invention are PL adhesion proteins. In an embodiment, the invention provides methods and reagents, as detailed herein, for inhibiting interactions between PL adhesion proteins and PDZ proteins to modulate an immune response. In an embodiment, the inhibition or modulation occurs in a hematopoietic cell. In a related embodiment, the inhibition or modulation occurs in an endothelial cell. In a related embodiment, the inhibition or modulation occurs in an endothelial cell. In a related embodiment, the inhibition or modulation occurs in an epithelial cells, keratinocytes, hepatocytes, cardiac myocytes.
As used herein, an ion channel protein means a transmembrane protein that itself catalyzes the passage of an ion from aqueous solution on one side of a lipid bilayer membrane to aqueous solution on the other side (e.g., by forming a small pore in the membrane). One exemplary PL ion channel proteins is Kvl .3. Thus, in one embodiment, the PL proteins of the invention are PL ion channel proteins. In an embodiment, the invention provides methods and reagents, as detailed herein, for inhibiting interactions between PL ion channel proteins and PDZ proteins to modulate an immune response. In an embodiment, the inhibition or modulation occurs in a hematopoeitic cell. In a related embodiment, the inhibition or modulation occurs in an endothelial cell. As used herein, an intercellular (i.e., cytosolic) protein has the normal meaning in the art and refers to a protein that is not membrane bound, e.g., has no transmembrane domain. Thus, in one embodiment, the PL proteins of the invention are PL intercellular proteins. Exemplary PL intercellular proteins include Glycophorin C and LPAP. In an embodiment, the invention provides methods and reagents, as detailed herein, for inhibiting interactions between PL cytoplasmic proteins and PDZ proteins to modulate an immune response. In an embodiment, the inhibition or modulation occurs in a hematopoeitic cell. In a related embodiment, the inhibition or modulation occurs in an endothelial cell.
As used herein a cytokine receptor has the normal meaning in the art and refers to a membrane protein with an extracellular domain that specifically binds a cytokine. Exemplary PL cytokine receptor proteins include CDW125 (IL5R), CDW128A (IL8RA), and
BRL-1. Thus, in one embodiment, the PL proteins of the invention are PL cytokine proteins.
In an embodiment, the invention provides methods and reagents, as detailed herein, for inhibiting interactions between PL cytokine proteins and PDZ proteins to modulate an immune response. In an embodiment, the inhibition or modulation occurs in a hematopoeitic cell. In a related embodiment, the inhibition or modulation occurs in an endothelial cell.
As used herein, an adaptor protein means a molecule (e.g., protein) that contributes to the formation of a multimolecular complex by binding two or more other biomolecuics. The binding o the two or more other molecules by the adaptor molecule/protein generally involves direct molecular contact between the adaptor protein and each of he two or more other molecules.. One exemplary PL adaptor protein is LPAP. Thus, in one embodiment, the PL proteins of the invention are PL adaptor proteins. In an embodiment, the invention provides methods and reagents, as detailed herein, for inhibiting interactions between PL adaptor proteins and PDZ proteins to modulate an immune response. In an embodiment, the inhibition or modulation occurs in a hematopoeitic cell. In a related embodiment, the inhibition or modulation occurs in an endothelial cell. In various embodiments, the PL proteins of the invention are characterized by specific C-terminal (i.e., PL domain) amino acid sequences or amino acid motifs, as described elsewhere in this disclosure.
In various embodiments of the invention, the PL proteins of the invention bind a PDZ protein expressed in T lymphocytes, B lymphocytes, or both T and B lymphocytes. In an embodiment, the PL protein binds a PDZ protein expressed in endothelial cells. In various embodiments, the PL proteins and/or the PDZ protein to which it binds are not expressed in the nervous system (e.g., neurons).
In various embodiments ofthe invention, the PL protein ofthe invention binds only one PDZ protein listed in TABLE 2. In other embodiment, the PL protein binds 1 to 3, 3 to 5, or more than 5 different PDZ proteins listed in TABLE 2.
In various embodiments of the invention, the PL protein is expressed or upregulated upon cell activation (e.g., in activated B lymphocytes, T lymphocytes) or upon entry into mitosis (e.g., up-regulation in rapidly proliferating cell populations). In various embodiments of the invention, the PL protein is (i) a protein that mediates immune cell (e.g., hematopoietic cell) activation or migration, (ii) a protein that does not mediate apoptosis in a cell type (iii) a protein that is other than a G-protein coupled seven transmembrane helix receptor, (iv) a protein that is G-protein coupled seven transmembrane helix receptor but not a cytokine receptor or (v) a protein that is not a G-protein coupled seven transmembrane helix receptor and is a cytokine receptor.
6.1 Detection of PDZ Domain-Containing Proteins Expressed in Hematopoietic Cells
As noted supra, the present inventors surprisingly discovered that numerous PDZ proteins are expressed in immune system cells, and play a fundamental biological role in modulation of the immune response. PDZ proteins DLGl and TIAM-I have been previously described to be in T cells. The present inventors discovered, using a BLAST search of the Human EST database and the experiments described infra, that several additional PDZ proteins are present in hematopoietic cells including MPPl, P-DLG, VELI-1, PSD95, syntenin in T cells and CASK, DLGl , DLG2, ZIP KINASE, syntrophin 2, P-dlg, PSD95, and syntenin in B cells. To determine the full extent of PDZ proteins' involvement in hematopoietic function, the inventors embarked on a systematic investigation of PDZ proteins in T and B cells. A comprehensive list of PDZ domain-containing proteins was retrieved from the Sanger Centre database (Pfam) searching for the keyword, PDZ. The corresponding cDNA sequences were retrieved from GenBank using the NCBI "entrez" database (hereinafter, "GenBank PDZ protein cDNA sequences"). The DNA portion encoding PDZ domains was identified by alignment of cDNA and protein sequence using CLUSTALW. Based on the DNA/protein alignment information, primers encompassing the PDZ domains were designed. The expression of certain PDZ-containing proteins in immune cells was detected by polymerase chain reaction ("PCR") amplification of cDNAs obtained by reverse transcription ("RT") of immune cell derived RNA (i.e., "RT-PCR"). PCR, RT-PCR and other methods for analysis and manipulation of nucleic acids are well known and are described generally in Sambrook et al., (1989) MOLECULAR CLONING: A LABORATORY MANUAL, 2ND ED., VOLS. 1-3, Cold Spring Harbor Laboratory hereinafter, "Sambrook"); and Ausubel et al., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, Greene Publishing and Wiley-Interscience, New York (1997), as supplemented through January 1999 (hereinafter "Ausubel"). In the experiments summarized in TABLE 2, T-cells (Jurkat E6 cell line) and B- cells (MV 4-11 cell line) were tested for expression of specific PDZ domain containing genes by RT-PCR. RNA was prepared using the "trizol" RNA preparation kit (GIBCO-BRL; Cat. # 15596-018) according to the manufacturer's recommendations. Briefly, 1-5 x 107 lymphoblasts were harvested by centrifugation at 200 x g for 10 minutes at 20° C. Cells were resuspended in 100 μl PBS buffer and 1 ml of TRIZOL reagent was added per 5 x 106 cells. The cells resuspension was mixed and after 5 minutes incubation at room temperature (RT), chloroform was added at 0.2 ml per ml TRIZOL. The resuspension was vigorously shaken and incubated for 3 more minutes at RT. Samples were then centrifuged at 12000 x g for 15 minutes at 4 ° C, the aqueous phase was recovered and RNA was precipitated with 2-propanol. The precipitate was collected by centrifugation at 12000 x g for 15 minutes at 4° C, washed with 75% ethanol, finally recollected by another spin at 12000 x g for 15 minutes at 4° C, air dried and resuspended in an appropriate volume of DEPC treated water.
RNA concentration and purity were determined by the measurement of 260/280 nm light absorption by the nucleic acid. For cDNA synthesis, the SUPERSCRIPT II reverse transcriptase cDNA kit (GIBCO-BRL; Cat. # 18064-014) was used. RNA input per 200 μl cDNA reaction sample was 10 μg. Prior to cDNA synthesis RNA was treated with 1 unit/μl DNAse I in 1 10 μl water at 37° C for 20 minutes. DNase I was then inactivated by a 10 minutes incubation at 70 ° C. Random primer was used for cDNA priming; 10 μl of random hexamer primer (100 ng/μl) was added, samples were heated to 70° C for 5 minutes and chilled on ice. Subsequently 40 μl SUPERSCRIPT II "first strand" buffer, 20 μl of 0.1 M DDT, 10 μl of a 10 mM of mix of deoxynucleotide triphosphates (dATP, dCTP, dGTP, dUTP) and 10 μl of SUPERSCRIPT II reverse transcriptase were added and cDNA synthesis was done for 45 minutes at 42 ° C. Reavtions were stopped by a 5 minutes incubation at 95 ° C and typically, 2-4 μl of such cDNA samples were used for PCR.
A portion of the cDNA (typically, 1/5 of a 20 μl reaction) was used for PCR. PCR was conducted using primers designed to amplify specifically PDZ domain-containing regions of PDZ proteins of interest. Oligonucleotide primers were designed to amplify one or more PDZ-encoding domains. The DNA sequences encoding the various PDZ domains of interest were identified by inspection (i.e., conceptual translation of the PDZ protein cDNA sequences obtained from GenBank, followed by alignment with the PDZ domain amino acid sequence). TABLE 3 shows the PCR primers, the PDZ-encoded domains amplified, and the GenBank accession number of the PDZ-domain containing proteins. To facilitate subsequent cloning of PDZ domains, the PCR primers included endonuclease restriction sequences at their ends to allow ligation with pGEX-3X cloning vector (Pharmacia, GenBank XXII 3852 ) in frame with glutathione-S transferase (GST). TABLE 3 lists proteins detected in the aforementioned assays. The results showed that PDZ proteins are widely utilized in T and B cells in both lineage specific as well as lineage independent manner. INADL2/3 (PDZ dom.), KIAA0316, and 26s subunit p27 were detected in T cells, but not B cells. mCASK, KIAA0559, PTN-4, and XI 1 beta were detected in B cells, but not T cells. AF6, BAII associated prot., Cytohesin bind. Prot., DLGl, DLG5 (pdlg), DVLl, DVL3, GTPase, hypoth. 41.8 kd, KIAA147, KIAA0300, KIAA0303, KIAA0380, KIAA0440, KIAA0545, KIAA0561, LIMKl , LIM 2, LIM domain prot, LIM protein, MINTl, MINT3, MPPl, MPP2, NE-DLG, NOS1, novel serine protease, PTN-3, prIL 16, PSD95, RGS12, serine protease, SYNTENIN, SYNTR 1 alpha, TAXI, TAX2, TAX33, TAX40, Tax43 (SYN, Betal), TIAM w p3, and XI 1 prot. were detected in both T cells and B cells. o
TABLE 3 PDZ DOMAINS o
Key: υ α. Gene names and corresponding gene products are provided. In some cases, cDNA sequences representing the, same gene have several database entries under different accession numbers and names. Accession numbers shown correspond to the gene .name used in this description, and numbering of nucleotides and amino acids correlates to those Genbank entries. Amino acid sequences shown correspond to the cloned DNA portions of PDZ domain containing genes. Linker amino acid sequences (e.g., amino acids encoded by DNA flanking the cloning site ofthe pGEX-3X cloning vector), are in italics
Figure imgf000035_0001
o O
Figure imgf000036_0001
PSD95 human post- U83192 AA387-724; Bam HI / | 8PSF 11PSR synaptic Eco RI density PDZ domains 1-3 (of 3) 5'-
Figure imgf000037_0001
protein 95 TCGGATCCTT TCGGAATTC
LEGEGEMEYEEITLERGNSGLGF GAGGGGGAGA TATACTCTTC
SIAGGTDNPHIGDDPSIFITKII TGGA-3' GG-3'
PGGAAAQDGRLRVNDSILFVNEV
DVREVTHSAAVEALKEAGSIVRL N1150-1173 N2191-2168 VMRRKPPAEKVMEIKLIKGPKG
LGFSIAGGVGNQHIPGDNSIYVT
KIIEGGAAHKDGRLQ1GDKILAV
NSVGLEDVMHEDAVAALKNTYDV
VYLKVAKPSNAYLSDSYAPPDIT
TSYSQHLDNEISHSSYLGTDYPT
AMTPTSPRRYSPVAKDLLGEEDI
PREPRRIVIHRGSTGLGFNIVGG
EDGEGIFISFILAGGPADLSGEL
RKGDQILSVNGVDLRNASHEQAA lALKNAGQTVTIIAQYKPEFIV
IΛ rt o O
©
© in
Figure imgf000038_0002
Figure imgf000038_0003
Figure imgf000038_0001
©
©
o rt
Figure imgf000039_0001
Figure imgf000039_0002
rt rt IΛ © o
©
C5
© t/1
H U rt
Figure imgf000040_0001
LIM Human LIM AF061258 AA29-112; Bam HI / 182LF 183LR protein PDZ domain 1 (of 1) Eco RI
5'- 5'- iSNYSVSLVGPAPWGFRLQGGKD TTAGGATCCT CTTGAATTCA FNMPLTISSLKDGGKAAQANVRI GAGCAAGTAC CAGATGCTCT GDVVLSIDGINAQGMTHLEAQNK AGTGTGTCAC TGCAGAGTC- IKGCTGSLNMTLQRASC -3' 3'
N86-115 N350-320
LIMKl human LIM NM_ AA194-291; SMA I 52LIFP ■ 53LIRP domain 002314 PDZ domain 1 (of 1) kinase 1 5'- 5'-
ΓVTLVSIPASSHGKRGLSVSIDP CTGCCCGGGA TCGCCCGGGT PHGPPGCGTEHSHTVRVQGVDPG CCGTCACCCT ATGCTCGAGG
IΛ CMSPDVKNSIHVGDRILEINGTP GGTGTCC-3' TC-3'
^H IRNVPLDEIDLLIQETSRLLQLT
© IEH DPGIHRD N570-597 N874-851 O
©
©
rt
Figure imgf000041_0001
Figure imgf000041_0002
rt rt IΛ
©
O
©
JO
©
o rt
Figure imgf000042_0001
Figure imgf000042_0002
©
(O
©
U rt
Figure imgf000043_0001
^H (O
© O
©
CO
©
u rt
rt rt IΛ
Figure imgf000044_0001
O
Figure imgf000045_0001
Figure imgf000045_0002
< tr*
Sr T3 TJ cn α sr
O H O CD μ- c
3 O H cQ cnn 3 O rt 0) 3 sr CD
CD li CD C CDD S3
O H- H- S3 <
S3 rt rt CD
H
CD
o o <τ> o
Figure imgf000046_0001
P3 03 O Cu 0 3 C M
S3 t-3 t-3 cn CO S3 NO -3 cn t-3 1-3 Cn Cn
O 0 O CD en 1 cn r cn α cn cn UJ o o > α o < rt M >
1 s cn cn t-3 t-3 < on
0 H3 frj CD o π
1 0 1-3 cn pα n o n CO n O
4-. O > 0 ~α cn > > 1 o o CO cn en t-3 en > o
Figure imgf000046_0002
©
JO
©
& CJ rt
Figure imgf000047_0001
rt rt IΛ JO
© O
©
JO
©
r υt
Figure imgf000048_0001
Figure imgf000048_0002
^H JO
© O
©
JO
©
& CJ rt
Figure imgf000049_0001
Figure imgf000049_0002
rt rt IΛ JO
© O
Figure imgf000049_0003
©
JO
©
1*1
& CJ rt
Figure imgf000050_0001
©
JO
© tn *Note concerning TABLE 3
In several cases, sequence analysis ofthe PDZ clones revealed differences to the DNA and/or protein sequence as publish
CJ rt in the databases, summarized in TABLE 3A.
TABLE 3A
Figure imgf000051_0002
( ) = s ent mutat on, oes not effect the AA sequence;
(**) = MINTl is the same as Xlla. The database entry for XI la shows'the same sequence as our actual construct with regard to
2386 of he MINTl GenBank entry.
(***)= Nucleotide ("N") and amino acid ("AA") annotations correspond to the numbering as found in the GenBank files.
Figure imgf000051_0001
6.2 Assays for Detection of Interactions Between PDZ-Domain Polypeptides and Candidate PDZ Ligand proteins (PL proteins)
Two complementary assays, tenned "A' and "G,"" were developed to detect binding between a PDZ-domain polypeptide and candidate PDZ ligand. In each of the two different assays, binding, is detected between a peptide having a sequence corresponding to the C-terminus of a protein anticipated to bind to one or more PDZ domain (i.e. a candidate PL peptide) and a PDZ-domain polypeptide (typically a fusion protein containing a PDZ domain). In the "A" assay, the candidate, PL peptide is immobilized and binding of a soluble PDZ-domain polypeptide to the immobilized peptide is detected (the "A"' assay is named for the fact that in one embodiment an avidin surface is used to immobilize the peptide). In the "G" assay, the PDZ-domain polypeptide is immobilized and binding of a soluble PL peptide is detected (The "G" assay is named for the fact that in one embodiment a GST-binding surface is used to immobilize the PDZ-domain polypeptide). Preferred embodiments of these assays are described in detail infra. However, it will be appreciated by ordinarily skilled practitioners that these assays can be modified in numerous ways while remaining useful for the purposes o the present invention.
6.2.1 Production of Fusion Proteins Containing PDZ-Domains
GST-PDZ domain fusion proteins were prepared for use in the assays of the invention. PCR products containing PDZ encoding domains (as described in §6.1 supra) were subcloned into an expression vector to permit expression of fusion proteins containing a PDZ domain and a heterologous domain (i.e., a glutathione-S transferase sequence, "GST")- PCR products (i.e., DNA fragments) representing PDZ domain encoding DNA was extracted from agarose gels using the "sephaglas" gel extraction system (Pharmacia) according to the manufacturer's recommendations. As noted supra, PCR primers were designed to include endonuclease restriction sites to facilitate ligation of PCR fragments into a GST gene fusion vector (pGEX-3X; Pharmacia, GenBank accession no. XXU 13852) in-frame with the glutathione-S transferase coding sequence. This vector contains a IPTG inducible lacZ promoter. The pGEX-3X vector was linearized using Bam HI and Eco RI or, in some cases, Eco RI or Sma I, as shown in TABLE 3, and dephosphorylated. For most cloning approaches, double digest with Bam HI and Eco RI was performed, so that the ends ofthe PCR fragments to clone were Bam HI and Eco RI. In some cases, restriction endonuclease combinations used were Bgl II and Eco RI, Bam HI and Mfe I, or Eco RI only, Sma I only, or BamHI only (see TABLE 3). When more than one PDZ domain was cloned, the DNA portion cloned represents the PDZ domains and the cDNA portion located between individual domains. Precise locations of cloned fragments used in the assays are indicated in TABLE 3. DNA linker sequences between the GST portion and the PDZ domain containing DNA portion vary slightly, dependent on which of the above described cloning sites and approaches were used. As a consequence, the amino acid sequence ofthe GST- PDZ fusion protein varies in the linker region between GST and PDZ domain. Protein linkers sequences corresponding to different cloning sites/approaches are shown below. Linker sequences (vector DNA encoded) are bold, PDZ domain containing gene derived sequences are in italics.
1) GST — BamHl/BamHI — PDZ domain insert Gly~Ue — PDZ domain insert
2) GST— Bamm/Bglll— PDZ domain insert Gly — He — PDZ domain insert
3) GSΥ—EcoRl/EcoI—PDZ domain insert Gly — lie — Pro — Gly— Asn — PDZ domain insert
4) GSTSmal/Smal— PDZ domain insert Gly — He — Pro — PDZ domain insert
The PDZ-encoding PCR fragment and linearized pGEX-3X vector were ethanol precipitated and resuspended in 10 ul standard ligation buffer. Ligation was performed for 4-10 hours at 7°C using T4 DNA ligase. It will be understood that some ofthe resulting constructs include very short linker sequences and that, when multiple PDZ domains were cloned, the constructs included some DNA located between individual PDZ domains. The ligation products were transformed in DH5α or BL-21 E.coli bacteria strains.
Colonies were screened for presence and identity ofthe cloned PDZ domain containing DNA as well as for correct fusion with the glutathione S-transferase encoding DNA portion by PCR and by sequence analysis. Positive clones were tested in a small scale assay for expression of the GST/PDZ domain fusion protein and, if expressing, these clones were subsequently grown up for large scale preparations of GST/PDZ fusion protein. GST-PDZ domain fusion protein was overexpressed following addition of IPTG to the culture medium and purified. Detailed procedure of small scale and large scale fusion protein expression and purification are described in "GST Gene Fusion System" (second edition, revision 2; published by Pharmacia). In brief, a small culture (3-5mls) containing a bacterial strain (DH5 , BL21 or JM109) with the fusion protein construct was grown overnight in LB- media at 37°C with the appropriate antibiotic selection (lOOug/ml ampicillin; a.k.a. LB-amp). The overnight culture was poured into a fresh preparation of LB-amp (typically 250-5 OOmls) and grown until the optical density (OD) ofthe culture was between 0.5 and 0.9 (approximately 2.5 hours). IPTG (isopropyl β-D-thiogalactopyranoside) was added to a final concentration of LOmM to induce production of GST fusion protein, and culture was grown an additional 1.5-2.5 hours. Bacteria were collect by centrifugation (4500 g) and resuspended in Buffer A- (50mM Tris, pH 8.0, 50mM dextrose, ImM EDTA, 200uM phenylmethylsulfonylfluoride). An equal volume of Buffer A+ (Buffer A-, 4mg/ml lysozyme) was added and incubated on ice for 3 min to lyse bacteria. An equal volume of Buffer B (lOmM Tris, pH 8.0, 50mM KC1, ImM EDTA. 0.5% Tween-20, 0.5% NP40 (a.k.a. IGEPAL CA-630), 200uM phenylmethylsulfonylfluoride) was added and incubated for an additional 20 min. The bacterial cell lysate was centrifuged (x20,000g), and supernatant was added to glutathione sepharose 4B (Pharmacia, cat no. 17-0765- 01) previous swelled (rehydrated) in IX phosphate-buffered saline (PBS). The supernatant- sepharose slurry was poured into a column and washed with at least 20 bed volumes of IX PBS. GST fusion protein was eluted off the glutathione sepharose by applying 0.5-1.0 ml aliquots of 5mM glutathione and collected as separate fractions. Concentrations of fractions were determined using BioRad Protein Assay (cat no. 500-0006) according to manufacturer's specifications. Those fractions containing the highest concentration of fusion protein were pooled and dialyzed against IX PBS/35% glycerol. Fusion proteins were assayed for size and quality by SDS gel electrophoresis (PAGE) as described in "Sambrook." Fusion protein aliquots were stored at minus 80°C and at minus 20°C.
6.2.2 Identification of Candidate PL Proteins and Synthesis of Peptides
In some non-hematopoietic cells (e.g., neurons, epithelial cells), certain PDZ domains are known to be bound by the C-terminal residues of PDZ-binding proteins. To identify PL proteins that function in hematopoietic and endothelial cells, cell surface receptor proteins were identified and peptides having the sequence corresponding to the c-terminus of each protein were synthesized. TABLE 4 lists these proteins, and provides corresponding C-terminal sequences and GenBank accession numbers. "Clasp 1" is described in WO 00/20434 (published 13 April 2000). "Clasp 2" and "Clasp 4" are described in copending applications USSN 09/547276 (attorney docket No. 20054-000200) and 60/196527 (attorney docket No.20054- 000400), both filed April 11 , 2000.
Synthetic peptides of defined sequence (e.g., corresponding to the carboxyl- termini of the indicated proteins) can be synthesized by any standard resin-based method (see, e.g., U. S. Pat. No. 4,108,846; see also, Caruthers et al, 1980, Nucleic Acids Res. Symp. Ser., 215-223; Horn et al., 1980, Nucleic Acids Res. Symp. Ser., 225-232; Roberge, et al, 1995, Science 269:202). The peptides used in the assays described herein were prepared by the FMOC (see, e.g., Guy and Fields, 1997, Meth. Enz. 289:67-83; Wellings and Atherton, 1997, Meth. Enz.289:44-67). In some cases (e.g., for use in the A and G assays of the invention), peptides were labeled with biotin at the amino-terminus by reaction with a four- fold excess of biotin methyl ester in dimethylsulfoxide with a catalytic amount of base. The peptides were cleaved from the resin using a halide containing acid (e.g. trifluoroacetic acid) in the presence of appropriate antioxidants (e.g. ethanedithiol) and excess solvent lyophilized.
Following lyophilization, peptides can be redissolved and purified by reverse phase high performance liquid chromatography (HPLC). One appropriate HPLC solvent system involves a Vydac C-18 semi-preparative column running at 5 mL per minute with increasing quantities of acetonitrile plus 0.1% trifluoroacetic acid in a base solvent of water plus 0.1% trifluoroacetic acid. After HPLC purification, the identities of the peptides are confirmed by MALDI cation-mode mass spectrometry. As noted, exemplary biotinylated peptides are provided in TABLE 4.
6.2.3 Detecting PDZ-PL Interactions
Based on the determination that immune system cells contain both many PDZ proteins and similarly many candidate PL proteins, it was apparent to the inventors that characterization ofthe specific PDZ-PL interactions among these proteins would require reliable and rapid assays for such interactions. A variety of assay formats known in the art can be used to select ligands that are specifically reactive with a particular protein. For example, solid-phase ELISA immunoassays, immunoprecipitation, Biacore, and Western blot assays can be used to identify peptides that specifically bind PDZ-domain polypeptides. As discussed supra, two different, complementary assays were developed to detect PDZ-PL interactions. In each, one binding partner of a PDZ-PL pair is immobilized, and the ability ofthe second binding partner to bind is determined. These assays, which are described infra, can be readily used to screen for hundreds to thousand of potential PDZ-ligand interactions in a few hours. Thus these assays can be used to identify yet more novel PDZ-PL interactions in hematopoietic cells. In addition, they can be used to identify antagonists of PDZ-PL interactions (see infra). In various embodiments, fusion protein are used in the assays and devices of the invention. Methods for constructing and expressing fusion proteins are well known. Fusion proteins generally are described in Ausubel et al., supra, Kroll et al., 1993, DNA Cell. Biol. 12:441, and Imai et al., 1997, Cell 91 :521-30. Usually, the fusion protein includes a domain to facilitate immobilization of the protein to a solid substrate ("an immobilization domain"). Often, the immobilization domain includes an epitope tag (i.e., a sequence recognized by a antibody, typically a monoclonal antibody) such as polyhistidine (Bush et al, 1991, J Biol Chem 266:13811-14), SEAP (Berger et al, 1988, Gene 66:1-10), or Ml and M2 flag (see, e.g, U.S. Pat. Nos. 5,011 ,912; 4,851,341 ; 4,703,004; 4,782,137). In an embodiment, the immobilization domain is a GST coding region. It will be recognized that, in addition to the PDZ-domain and the particular residues bound by an immobilized antibody, protein A, or otherwise contacted with the surface, the protein (e.g., fusion protein), will contain additional residues. In some embodiments these are residues naturally associated with the PDZ-domain (i.e., in a particular PDZ-protein) but they may include residues of synthetic (e.g., poly(alanine)) or heterologous origin (e.g., spacers of, e.g., between 10 and 300 residues).
PDZ domain-containing polypeptide used in the methods ofthe invention (e.g., PDZ fusion proteins) of the invention are typically made by (1) constructing a vector (e.g., plasmid, phage or phagemid) comprising a polynucleotide sequence encoding the desired polypeptide, (2) introducing the vector into an suitable expression system (e.g., a prokaryotic, insect, mammalian, or cell free expression system), (3) expressing the fusion protein and (4) optionally purifying the fusion protein. (1 ) In one embodiment, expression of the protein comprises inserting the coding sequence into an appropriate expression vector (i.e., a vector that contains the necessary elements for the transcription and translation of the inserted coding sequence required for the expression system employed, e.g., control elements including enhancers, promoters, transcription terminators, origins of replication, a suitable initiation codon (e.g., methionine), open reading frame, and translational regulatory signals (e.g., a ribosome binding site, a termination codon and a polyadenylation sequence. Depending on the vector system and host utilized, any number of suitable transcription and translation elements, including constitutive and inducible promoters, can be used.
The coding sequence of the fusion protein includes a PDZ domain and an immobilization domain as described elsewhere herein. Polynucleotides encoding the amino acid sequence for each domain can be obtained in a variety of ways known in the art; typically the polynucleotides are obtained by PCR amplification of cloned plasmids, cDNA libraries, and cDNA generated by reverse transcription of RNA, using primers designed based on sequences determined by the practitioner or, more often, publicly available (e.g., through GenBank). The primers include linker regions (e.g., sequences including restriction sites) to facilitate cloning and manipulation in production of the fusion construct. The polynucleotides corresponding to the PDZ and immobilization regions are joined in-frame to produce the fusion protein-encoding sequence.
The fusion proteins ofthe invention may be expressed as secreted proteins (e.g., by including the signal sequence encoding DNA in the fusion gene; see, e.g., Lui et al, 1993, PNAS USA, 90:8957-61) or as nonsecreted proteins.
In some embodiments, the PDZ-containing proteins are immobilized on a solid surface. The substrate to which the polypeptide is bound may in any of a variety of forms, e.g., a microtiter dish, a test tube, a dipstick, a miorocentrifuge tube, a bead, a spinnable disk, and the like. Suitable materials include glass, plastic (e.g., polyethylene, PVC, polypropylene, polystyrene, and the like), protein, paper, carbohydrate, lipip monolayer or supported lipid bilayer, and other solid supports. Other materials that may be employed include ceramics, metals, metalloids, semiconductive materials, cements and the like.
In some embodiments, the fusion proteins are organized as an array. The term "array," as used herein, refers to an ordered arrangement of immobilized fusion proteins, in which particular different fusion proteins (i.e., having different PDZ domains) are located at different predetermined sites on the substrate. Because the location of particular fusion proteins on the array is known, binding at thai location can be correlated with binding to the PDZ domain situated at that location. Immobilization of fusion proteins on beads (individually or in groups) is another particularly useful approach. In one embodiment, individual fusion proteins are immobilized on beads. In one embodiment, mixtures of distinguishable beads are used. Distinguishable beads are beads that can be separated from each other on the basis of a property such as size, magnetic property, color (e.g., using FACS) or affinity tag (e.g., a bead coated with protein A can be separated from a bead not coated with protein A by using IgG affinity methods). Binding to particular PDZ domain may be determined; similarly, the effect of test compounds (i.e., agonists and antagonists of binding) may be determined. Methods for immobilizing proteins are well known in the art, and include covalent and non-covalent methods.
One suitable immobilization method is antibody-mediated immobilization. According to this method, an antibody specific for the sequence of an "immobilization domain" ofthe PDZ-domain containing protein is itself immobilized on the substrate (e.g., by adsorption). One advantage of this approach is that a single antibody may be adhered to the substrate and used for immobilization of a number of polypeptides (sharing the same immobilization domain). For example, an immobilization domain consisting of poly-histidine (Bush et al, 1991, J. Biol Chem 266: 13811-14) can be bound by an anti-histidine monoclonal antibody (R&D Systems, Minneapolis, MN); an immobilization domain consisting of secreted alkaline phosphatase ("SEAP") (Berger et al, 1988, Gene 66:1-10) can be bound by anti-SEAP (Sigma Chemical Company, St. Louis, MO); an immobilization domain consisting of a FLAG epitope can be bound by anti-FLAG. Other ligand-antiligand immobilization methods are also suitable (e.g., an immobilization domain consisting of protein A sequences (Harlow and Lane, 1988, Antibodies A laboratory Manual, Cold Spring Harbor Laboratory; Sigma Chemical Co., St. Louis, MO) can be bound by IgG; and an immobilization domain consisting of strepavidin can be bound by biotin (Harlow & Lane, supra; Sigma Chemical Co., St. Louis, MO). In a preferred embodiment, the immobilization domain is a GST moiety, as described herein.
When antibody-mediated immobilization methods are used, glass and plastic are especially useful substrates. The substrates may be printed with a hydrophobic (e.g., Teflon) mask to form wells. Preprinted glass slides with 3, 10 and 21 wells per 14.5 cm2 slide "working area" are available from, e.g., SPI Supplies, West Chester, PA; also see U.S.Pat. No. 4,011,350). In certain applications, a large format (12.4 cm x 8.3 cm) glass slide is printed in a 96 well format is used; this format facilitates the use of automated liquid handling equipment and utilization of 96 well format plate readers of various types (fluorescent, colorimetric, scintillation). However, higher densities may be used (e.g., more than 10 or 100 polypeptides per cπr). See, e.g., MacBeath et al, 2000, Science 289: 1760-63.
Typically, antibodies are bound to substrates (e.g., glass substrates) by adsorption. Suitable adsorption conditions are well known in the art and include incubation of 0.5-50ug/ml (e.g., 10 ug/ml) mAb in buffer (e.g., PBS, or 50 to 300 mM Tris, MOPS, HEPES, PIPES, acetate buffers, pHs 6.5 to 8, at 4°C) to 37°C and from lhr to more than 24 hours. Proteins may be covalently bound or noncovalently attached through nonspecific bonding. If covalent bonding between a the fusion protein and the surface is desired, the surface will usually be polyfunctional or be capable of being polyfunctionalized. Functional groups which may be present on the surface and used for linking can include carboxylic acids, aldehydes, amino groups, cyano groups, ethylenic groups, hydroxyl groups, mercapto groups and the like. The manner of linking a wide variety of compounds to various surfaces is well known and is amply illustrated in the literature.
6.2.3.1 "A Assay'TJetection of PDZ-Ligand Binding Using Immobilized PL Peptide. In one aspect, the invention provides an assay in which biotinylated candidate PL peptides are immobilized on an avidin coated surface. The binding of PDZ-domain fusion protein to this surface is then measured. In a preferred embodiment, the PDZ-domain fusion protein is a GST/PDZ fusion protein and the assay is carried out as follows:
(1) Avidin is bound to a surface, e.g. a protein binding surface. In one embodiment, avidin is bound to a polystyrene 96 well plate (e.g., Nunc Polysorb (cat #475094) by addition of 100 uL per well of 20 ug/mL of avidin (Pierce) in phosphate buffered saline without calcium and magnesium, pH 7.4 ("PBS", GibcoBRL) at 4°C for 12 hours. The plate is then treated to block nonspecific interactions by addition of 200 uL per well of PBS containing 2 g per 100 mL protease-free bovine serum albumin ("PBS/BSA") for 2 hours at 4°C. The plate is then washed 3 times with PBS by repeatedly adding 200 uL per well of PBS to each well of the, plate and then dumping the contents ofthe plate into a waste container and tapping the plate gently on a dry surface.
(2) Biotinylated PL peptides (or candidate PL peptides, e.g. see TABLE 4) are immobilized on the surface of wells of the plate by addition of 50 uL per well of 0.4 uM peptide in PBS/BSA for 30 minutes at 4°C. Usually, each different peptide is added to at least eight different wells so that multiple measurements (e.g. duplicates and also measurements using different (3ST/PDZ-domain fusion proteins and a GST alone negative control) can be made, and also additional negative control wells are prepared in which no peptide is immobilized. Following immobilization ofthe PL peptide on the surface, the plate is washed 3 times with PBS.
(3) GST/PDZ-domain fusion protein (prepared as described supra) is allowed to react with the surface by addition of 50 uL per well of a solution containing 5 ug/mL GST/PDZ-domain fusion protein in PBS/BSA for 2 hours at 4°C. As a negative control, GST alone (i.e. not a fusion protein) is added to specified wells, generally at least 2 wells (i.e. duplicate measurements) for each immobilized peptide. After the 2 hour reaction, the plate is washed 3 times with PBS to remove unbound fusion protein.
(4) The binding of the GST/PDZ-domain fusion protein to the avidin- biotinylated peptide surface can be detected using a variety of methods, and detectors known in the art. In one embodiment, 50 uL per well of an anti-GST antibody in PBS/BSA (e.g. 2.5 ug/mL of polyclonal goat-anti-GST antibody, Pierce) is added to the plate and allowed to react for 20 minutes at 4°C. The plate is washed 3 times with PBS and a second, delectably labeled antibody is added. In one embodiment, 50 uL per well of 2.5 ug/mL of horseradish peroxidase (HRP)-conjugated polyclonal rabbit anti-goat inununoglobulin antibody is added to the plate and allowed to react for 20 minutes at 4°C. The plate is washed 5 times with 50 mM Tris pH 8.0 containing 0.2% Tween 20, and developed by addition of 100 uL per well of HRP-substrate solution (TMB, Dako) for 20 minutes at room temperature (RT). The reaction ofthe HRP and its substrate is terminated by the addition of 100 uL per well of 1M sulfuric acid and the optical density (O.D.) of each well ofthe plate is read at 450 nm. (5) Specific binding of a PL peptide and a PDZ-domain polypeptide is detected by comparing the signal from the well(s) in which the PL peptide and PDZ domain polypeptide are combined with the background signal(s). The background signal is the signal found in the negative controls. Typically a specific or selective reaction will be at least twice background signal, more typically more than 5 times background, and most typically 10 or more times the background signal. In addition, a statistically significant reaction will involve multiple measurements of the reaction with the signal and the background differing by at least two standard errors, more typically four standard errors, and most typically six or more standard errors. Correspondingly, a statistical test (e.g. a T-test) comparing repeated measurements ofthe signal with repeated measurements of the background will result in a p-value < 0.05, more typically a p-value < 0.01, and most typically a p-value < 0.001 or less.
As noted, in an embodiment of the "A" assay, the signal from binding of a GST/PDZ-domain fusion protein to an avidin surface not exposed to (i.e. not covered with) the PL peptide is one suitable negative control (sometimes referred to as "B"). The signal from binding of GST polypeptide alone (i.e. not a fusion protein) to an avidin-coated surface that has been exposed to (i.e. covered with) the PL peptide is a second suitable negative control (sometimes referred to as "B2"). Because all measurements are done in multiples (i.e. at least duplicate) the arithmetic mean (or, equivalently, average) of several measurements is used in determining the binding, and the standard error ofthe mean is used in determining the probable error in the measurement of the binding. The standard error of the mean of N measurements equals the square root of the following: the sum of the squares of the difference between each measurement and the mean, divided by the product of ( f) and (N-l). Thus, in one embodiment, specific binding ofthe PDZ protein to the plate-bound PL peptide is determined by comparing the mean signal ("mean S") and standard error of the signal ("SE") for a particular PL-PDZ combination with the mean Bl and/or mean B2. In TABLE 2, binding was detected to be specific (denoted by an "A" in the matrix) when (1) the, mean S was at least twice the mean Bl and at least twice the mean B2 and (2) the mean S was at least six standard errors (six SE) greater than both the mean Bl and the mean B2. In addition, in the experiments summarized in TABLE 2, an additional criterion was used to ensure that none ofthe interactions defined as specific arose from a combined tendency of both the particular PDZ fusion protein and PL peptide tested to each give a higher than usual background. This criteria was that (3) the mean S was at least twenty times the product ofthe mean B 1 and the mean B2. The factor twenty times reflects that at least one of Bl and B2 is generally less than 0.1 O.D. units, and therefore twenty times the product ofthe mean Bl and the mean B2 is generally less than twice the mean Bl and twice the mean B2, making criteria (3) less stringent than criteria (1). Only in a few cases where the mean B 1 and the mean B2 are both greater than 0.1 O.D. units (i.e. both the particular PDZ fusion protein and PL peptide tested tend to give a higher than usual background) is criteria (3) more stringent than criteria (1).
Figure imgf000063_0001
6.2.3.2 "G Assay" - Detection of PDZ-Ligand Binding Using Immobilized PDZ- Domain Fusion Polypeptide
In one aspect, the invention provides an assay in which a GST/PDZ fusion protein is immobilized on a surface ("G" assay). The binding of labeled PL peptide (as listed in TABLE 4) to this surface is then measured. In a preferred embodiment, the assay is carried out as follows:
(1) A PDZ-domain polypeptide is bound to a surface, e.g. a protein binding surface. In a preferred embodiment, a GST/PDZ fusion protein containing one or more PDZ domains is bound to a polystyrene 96-well plate. The GST/PDZ fusion protein can be bound to the plate by any of a variety of standard methods known to one of skill in the art, although some care must be taken that the process of binding the fusion protein to the plate does not alter the ligand-binding properties ofthe PDZ domain. In one embodiment, the GST/PDZ fusion protein is bound via an anti-GST antibody that is coated onto the 96-well plate. Adequate binding to the plate can be achieved when: a. 100 uL per well of 5 ug/mL goat anti-GST polyclonal antibody (Pierce) in PBS is added to a polystyrene 96-well plate (e.g., Nunc Polysorb) at 4°C for 12 hours. b. The plate is blocked by addition of 200 uL per well of PBS/BSA for 2 hours at 4°C. c. The plate is washed 3 times with PBS. d. 50 uL per well of 5 ug/mL GST/PDZ fusion protein) or, as a negative control, GST polypeptide alone (i.e. not a fusion protein) in PBS/BSA is added to the plate for 2 hours at 4°C. e. the plate is again washed 3 times with PBS.
(2) Biotinylated PL peptides (or candidate PL peptides, e.g. as shown in TABLE 4) are allowed to react with the surface by addition of 50 uL per well of 20 uM solution of the biotinylated peptide in PBS/BSA for 10 minutes at 4°C, followed by an additional 20 minute incubation at 25°C. The plate is washed 3 times with ice cold PBS. (3) The binding of the biotinylated peptide to the GST/PDZ fusion protein surface can be detected using a variety of methods and detectors known to one of skill in the art.
In one embodiment, 100 uL per well of 0.5 ug/mL streptavidin-horse radish peroxidase (HRP) conjugate dissolved in BSA/PBS is added and allowed to react for 20 minutes at 4°C. The plate is then washed 5 times with 50 mM Tris pH 8.0 containing 0.2% Tween 20, and developed by addition of 100 uL per well of HRP-substrate solution (TMB, Dako) for 20 minutes at room temperature (RT). The reaction ofthe HRP and its substrate is terminated by addition of 100 uL per well of I M sulfuric acid, and the optical density (O.D.) of each well of the plate is read at 450 um.
(4) Specific binding of a PL peptide and a PDZ domain polypeptide is determined by comparing the signal from the well(s) in which the PL peptide and PDZ domain polypeptide are combined, with the background signal(s). The background signal is the signal found in the negative control(s). Typically a specific or selective reaction will be at least twice background signal, more typically more than 5 times background, and most typically 10 or more times the background signal. In addition, a statistically significant reaction will involve multiple measurements of the reaction with the signal and the background differing by at least two standard errors, more typically four standard errors, and most typically six or more standard errors. Correspondingly, a statistical test (e.g. a T-test) comparing repeated measurements ofthe signal with -repeated measurements of the background will result in a p-value < 0.05, more typically a p-value < 0.01, and most typically a p-value < 0.001 or less. As noted, in an embodiment of the "G" assay, the signal from binding of a given PL peptide to immobilized (surface bound) GST polypeptide alone is one suitable negative control (sometimes referred to as "B 1"). Because all measurement are done in multiples (i.e. at least duplicate) the arithmetic mean (or, equivalently, average.) of several measurements is used in determining the binding, and the standard error ofthe mean is used in determining the probable error in the measurement ofthe binding. The standard error of the mean ofN measurements equals the square root ofthe following: the sum of the squares of the difference between each measurement and the mean, divided by the product of (N) and (N-l). Thus, in one embodiment, specific binding ofthe PDZ protein to the platebound peptide is determined by comparing the mean signal ("mean S") and standard error of the signal ("SE") for a particular PL-PDZ combination with the mean Bl . In experiments summarized in TABLE 2, binding was determined to be specific (denoted by a "G" in the matrix) when (1) the mean S was at least twice the mean Bl and (2) the mean S was at least six standard errors (six SE) greater than the mean B l . Results of exemplary "G" assays are shown in Figures 1A-1D.
6.2.4 Assay Variations
As discussed supra, it will be appreciated that many of the steps in the above- described assays can be varied, for example, various substrates can be used for binding the PL and PDZ-containing proteins; different types of PDZ containing fusion proteins can be used; different labels for detecting PDZ/PL interactions can be employed; and different ways of detection can be used.
The PDZ-PL detection assays can employ a variety of surfaces to bind the PL and PDZ-containing proteins. For example, a surface can be an "assay plate" which is formed from a material (e.g. polystyrene) which optimizes adherence of either the PL protein or PDZ- containing protein thereto. Generally, the individual wells of the assay plate will have a high surface area to volume ratio and therefore a suitable shape is a flat bottom well (where the proteins ofthe assays are adherent). Other surfaces include, but are not limited to, polystyrene or glass beads, polystyrene or glass slides, and alike.
For example, the assay plate can be a "microtiter" plate. The term "microtiter" plate when used herein refers to a multiwell assay plate, e.g., having between about 30 to 200 individual wells, usually 96 wells. Alternatively, high density arrays can be used. Often, the individual wells of the microtiter plate will hold a maximum volume of about 250 ul. Conveniently, the assay plate is a 96 well polystyrene plate (such as that sold by Becton Dickinson Labware, Lincoln Park, N.J.), which allows for automation and high throughput screening. Other surfaces include polystyrene microtiter ELISA plates such as that sold by Nunc Maxisorp, Inter Med, Denmark. Often, about 50 ul to 300 ul, more preferably 100 ul to 200 ul, of an aqueous sample comprising buffers suspended therein will be added to each well of the assay plate.
The detectable labels of the invention can be any detectable compound or composition which is conjugated directly or indirectly with a molecule (such as described above). The label can be detectable by itself (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, can catalyze a chemical alteration of a substrate compound or composition which is detectable. The preferred label is an enzymatic one which catalyzes a color change of a non-radioactive color reagent.
Sometimes, the label is indirectly conjugated with the antibody. One of skill is aware of various techniques for indirect conjugation. For example, the antibody can be conjugated with biotin and any of the categories of labels mentioned above can be conjugated with avidin, or vice versa (see also "A" and "G" assay above). Biotin binds selectively to avidin and thus, the label can be conjugated with the antibody in this indirect manner. See, Ausubel, supra, for a review of techniques involving biotin-avidin conjugation and similar assays. Alternatively, to achieve indirect conjugation o the label with the antibody, the antibody is conjugated with a small hapten (e.g. digoxin) and one ofthe different types of labels mentioned above is conjugated with an anti-hapten antibody (e.g. anti-digoxin antibody). Thus, indirect conjugation of the label with the antibody can be achieved.
Assay variations can include different washing steps. By "washing" is meant exposing the solid phase to an aqueous solution (usually a buffer or cell culture media) in such a way that unbound material (e.g., non-adhering cells, non-adhering capture agent, unbound ligand, receptor, receptor construct, cell lysate, or HRP antibody) is removed therefrom. To reduce background noise, it is convenient to include a detergent (e.g., Triton X) in the washing solution. Usually, the aqueous washing solution is decanted from the wells of the assay plate following washing. Conveniently, washing can be achieved using an automated washing device.
Sometimes, several washing steps (e.g., between about 1 to 10 washing steps) can be required.
Various buffers can also be used in PDZ-PL detection assays. For example, various blocking buffers can be used to reduce assay background. The term "blocking buffer" refers to an aqueous, pH buffered solution containing at least one blocking compound which is able to bind to exposed surfaces of the substrate which are not coated with a PL or PDZ- containing protein. The blocking compound is normally a protein such as bovine serum albumin (BSA), gelatin, casein or milk powder and does not cross-react with any of the reagents in the assay. The block buffer is generally provided at a pH between about 7 to 7.5 and suitable buffering agents include phosphate and TRIS. Various enzyme-substrate combinations can also be utilized in detecting PDZ-PL interactions. Examples of enzyme-substrate combinations include, for example: (i) Horseradish peroxidase (HRPO) with hydrogen peroxidase as a substrate, wherein the hydrogen peroxidase oxidizes a dye precursor (e.g. orthophenylene diamine [OPD] or 3,3',5,5'-tetramethyl benzidine hydrochloride [TMB]) (as described above).
(ii) alkaline phosphatase (AP) with para-Nitrophenyl phosphate as chromogenic substrate.
(iii) β-D-galactosidase (β D-Gal) with a chromogenic substrate (e.g. p- nitrophenyl- β-D-galactosidase) or fluorogenic substrate 4-methylumbelliferyl- β-D- galactosidase.
Numerous other enzyme-substrate combinations are available to those skilled in the art. For a general review of these, see U.S. Pat. Nos. 4,275,149 and 4,318,980, both of which are herein incorporated by reference.
Further, it will be appreciated that, although, for convenience, the present discussion primarily refers antagonists of PDZ-PL interactions, agonists of PDZ-PL interactions can be identified using the methods disclosed herein or readily apparent variations thereof.
6.2.5 Results of PDL-PL Interaction Assays
TABLE 2, supra, shows the results of assays in which specific binding was detected using the "A" and "G" assays described herein. The top row of the table specifies the source of the PDZ domain used in the GST-PDZ fusion proteins (see TABLE 3). The first column lists the cell surface proteins from which C-terminal peptide sequences were derived and the second column ("code") identifies the peptide used in the assay (see TJ BLE 4). The third column, "Seq" provides the sequence of the four (4) C-terminal residues of the cell surface protein and peptide. In the matrix, "A" indicates specific binding as detected in the "A" assay. "G" indicates specific binding as shown in the "G" assay. A blank indicates that no specific binding was detected using the "A" or "G" assays. An asterisk (*) indicates that a pairwise interaction between the PDZ protein and the cell surface protein (or subdomains of either) has been described by others.
6.2.5.1 New PL Motifs As noted supra, TABLE 2 shows the results of assays (referred to as "PRISM
MATRIX") to detect binding between PDZ proteins and candidate PL peptides. A number of specific PDZ-PL interactions are identified by the MATRIX and key amino acids and positions important in PDZ binding ("PL motifs") are deduced from these results. Not only is the MATRIX useful to catalog comprehensively PDZ-PL binding combinations, the assay can further aid in the rapid discovery and characterization of novel PL proteins and PL motifs to help in rational drug design and synthesis of PL-PDZ interaction inhibitors. Other investigators have reported certain PL motifs important in PDZ binding, e.g., the C-terminal motifs S/T-X-V/I/L (for DLGl) and Y/F-Y/F-I/L/F for MPPl (see, Doyle et al, 1996, Cell 85, 1067; Songyang et al., 1997, Science 275, 73). However, the reported motifs are not sufficiently specific (i.e. a large number of proteins meet these criteria yet are not necessarily actual PDZ ligands) and cover only a small number of PDZ proteins (approximately 10). The PRISM MATRIX can be used to determine ligand specificity and to deduce ligand binding motifs for any PDZ protein because it can precisely determine sequences of amino acids that do or do not result in specific PDZ binding. In addition, the assay has revealed a significant of new PDZ domain binding motifs (i.e. PL motifs): C-terminal sequence of CD6, ISAA; C- terminal sequence of CD49E, TSDA; C- terminal sequence of CD49F, TSDA; C-terminal sequence of Clasp-1, SAEV; C- terminal sequence of CLASP-4, YAEV; C- terminal sequence of CD44, KIGV; C-terminal sequence of IL5R, DSVF; C-terminal sequence of BLR-1, LTTF. Identification of these novel PL sequences allows the definition of novel PL motifs (See TABLE 5A, infra). The specificity with which these novel motifs are defined is enhanced by the fact that the MATRIX reports both positive results (i.e. PDZ-PL) combinations that result in specific binding interactions) and negative results (i.e. PDZ-PL combinations that do not result in specific binding). For example, the C-terminal sequence of CD6, SAA and the C-terminal sequence of CD49E, SDA bind to the PDZ-domain polypeptide 41.8 while the related C-terminal sequence of CD166, TEA and C- terminal sequence of CD148, YIA do not. This identifies the novel PL motif (Motif 1 , infra) of polypeptides terminating in alanine with serine at the -2 position and excludes polypeptides with threonine and tyrosine at the -2 position. This motif is therefore more specific than most previously identified motifs. Other novel motifs are described in TABLE 5A.
TABLE 5A
Position: -3 -2 -1 C- terminal
Motif 1 X S X A Motif 2 X A D/E V
Motif 3 X V/I/L X* V
Motif 4 X S/T X F
X* is any non-aromatic amino acid (any residue other than T, F or W).
6.2.5.2 Binding Clusters
TABLE 2 is arranged so that both the PDZ/GST fusion proteins and the PL peptide ligands are ordered with structurally similar molecules close to each other. In preparing TABLE 2 and carrying out the experiments used to create this table, the PDZ domains of each of the GST-PDZ fusion proteins were ranked for amino acid sequence similarity using the CLUSTAL multiple sequence alignment software package. Proteins with greatest sequence similarity are closest to each other in the matrix. The PL peptide ligands were also ordered on the basis of amino acid similarity, but with weight given to residues reported to be important in PDZ binding (Doyle et al., 1996, Cell 85, 1067). In particular, peptides were first ordered based on the most C-terminal residue (zero position) in the following amino acid order: G, A, C, S, T, N, Q, D, E, H, K, R, V, I, L, M, P, F, Y, W. Among peptides with identical C-termini, the same ranking scheme was then applied to the next most important residue for peptide binding, the -2 position, followed by the —1 position and the -3 position. (In an alternative approach, the GST- PDZ fusions can also be arranged to give additional weight to residues known to be important for ligand binding when aligning the proteins.)
Regions of the TABLE 1 matrix that are densely filled with significant binding interactions indicate that a particular ligand structure tends to bind to a particular family of PDZ domains. The results indicate that PDZ domains with similar structures bind ligands with similar structures, as indicated by the presence in the matrix of some clusters of many significant binding interactions and other areas of few significant binding interactions. These results reveal a number of structural relationships between different PDZ proteins and their ligands. The understanding of these structural relationships can be used to scan databases for probable ligands of specific PDZ proteins, facilitating the design of inhibitors of such novel interactions and the prediction o the side-effect profile of such inhibitors.
The most striking example of a cluster OGCUΓS in columns 1 — 5 of the matrix (PDZ domains from CASK, MPPl, DLGl, PSD95, and NeDLG). In our assays, CASK bound significantly to only one o the tested ligands, neurexin (this interaction was previously identified in neuronal cells). MPPl , the closest relative to CASK in the ordering system used, bound a set of five different ligands, including neurexin. All ofthe MPPl ligands identified are structurally similar in that they terminate in valine (V) and have a charged amino acid (E or K) at the -3 position from the C-terminus ((C-terminal motif: E/K - X - X - V)
Out ofthe five MPPl ligands identified, four of these also bind to DLGl, the next most similar protein to CASK and MPPl . DLGl is closely related to two other proteins, PSD95 and NeDLG. Each of these three proteins binds ligands that terminate in the motif S/T/Y - X- V/I/L, as previously described (Songyang et al, 1997, Science 275, 73). Interestingly, if the terminal ligand residue is valine, other previously unidentified residues at the -2 position are compatibile with binding, with the C-terminal sequences AEV and ELV resulting in significant binding events. While this reflects a previously unrecognized flexibility in PDZ-ligand binding, the specificity ofthe interactions detected in our assays is reflected in the large areas at the top and bottom of columns 1 — 5 that contain no significant binding event. These areas reflect that none of the potential ligands terminating in residues other than V/I/L bound significantly to CASK, MPPl, DLGl, PSD95, or NeDLG.
(With respect to MPPl, it worth noting that Glycophorin C, a known ligand of MPPl, Marfatia et al., 1997, 1. Biol. Chem. 272, 24191, did not bind significantly to MPPl in our tests, but does terminate in isoleucine, an amino acids very similar to valine, and does have a charged residue at the -3 position o the C-terminus.)
Other smaller clusters on the matrix also provide valuable information about the ligand specificity of certain PDZ domains. For example, five ligands bind significantly in both the "G" and the "A" assays to 41.8 kD protein, a previously unstudied PDZ domain-containing protein. All of these ligands terminate in A, V, I, or L, and four of five have serine (S) at the -2 position, defining the C-terminal ligand motif S - X - A/V/I/L for this PDZ domain. Similarly, two of three ligands that bind specifically to KIAA0561 fit the C-terminal motif X* - S/T - D E - V/I/L where X* is any non-aromatic amino acid. Interestingly, one of these ligands also binds to KIAA 0561 's closest relative, TAX2. Likewise, the two ligands that bind to prIL16 both have a charged amino acid at the -3 position, a hydrophobic amino acid at the - 2 position, and valine at the terminal position. A final interesting example of defining the ligand specificity of a PDZ domain involves PTN-4. Although the two ligands that bind significantly to PTN-4 are not closely related at their C-terminal 2 residues, both fit the C- terminal motif D/E -S-X- V/I/L/F/Y. These motifs characterizing the ligand specificity of particular PDZ proteins are summarized in TABLE 5B. Such motifs allow searches of databases for novel ligands (PL proteins) binding to these particular PDZ proteins. Knowledge o the PL proteins that bind to these PDZ proteins allows the design of inhibitors of these interactions, using the methods described herein, infra. In addition, knowledge ofthe set of PL proteins that bind to a particular PDZ protein can be used to predict the utility and the side effects of compounds that target this PDZ protein.
TAI JLE 5B
PDZ -3 -2 -1 0
41.8 kD X X X A/V/I/L
KIAA 0561 X* S/T D/E V/I/L
TAX 2 X s D/E V
PPJL16 D/E/K R V/I/L/F/Y X V
PTN4 D/E s X V/I/L/F/Y
X* is any non-aromatic amino acid.
6.3 Measurement of PDZ-Ligand Binding Affinity
The "A" and "G" assays of the invention can be used to determine the "apparent affinity" of binding of a PDZ ligand peptide to a PDZ-domain polypeptide. Apparent affinity is determined based on the concentration of one molecule required to saturate the binding of a second molecule (e.g., the binding of a ligand to a receptor). Two particularly useful approaches for quantitation of apparent affinity of PDZ-ligand binding are provided infra.
(1 ) A GST/PDZ fusion protein, as well as GST alone as a negative control, are bound to a surface (e.g., a 96-well plate) and the surface blocked and washed as described supra for the "G" assay. (2) 50 uL per well of a solution of biotinylated PL peptide (e.g. as shown in TJABLE 4) is added to the surface in increasing concentrations in PBS/BSA (e.g. at 0.1 uM, 0.33 uM, 1 uM, 3.3 uM, 10 uM, 33 uM, and 100 uM). In one embodiment, the PL peptide is allowed to react with the bound GST/PDZ fusion protein (as well as the GST alone negative control) for 10 minutes at 4 C followed by 20 minutes at 25 C. The plate is washed 3 times with ice cold PBS to remove unbound labeled peptide.
(3) The binding of the PL peptide to the immobilized PDZ-domain polypeptide is detected as described supra for the "G" assay.
(4) For each concentration of peptide, the net binding signal is determined by subtracting the binding of the peptide to GST alone from the binding of the peptide to the
GST/PDZ fusion protein. The net binding signal is then plotted as a function of ligand concentration and the plot is fit (e.g. by using the Kaleidagraph software package curve fitting algorithm) to the following equation, where "Signal[ltgand]" is the net binding signal at PL peptide concentration "[ligand]," "Kd" is the apparent affinity of the binding event, and "Saturation Binding" is a constant determined by the curve fitting algorithm to optimize the fit to the experimental data:
Signalti;gaildj = Saturation Binding x ([ligand] / ([ligand] + Kd))
For reliable application of the above equation it is necessary that the highest peptide ligand concentration successfully tested experimentally be greater than, or at least similar to, the calculated Kd (equivalently, the maximum observed binding should be similar to the calculated saturation binding). In cases where satisfying the above criteria proves difficult, an alternative approach (infra) can be used.
The results obtained when using approach 1 are demonstrated in FIGURES 2A and 2B. FIGURE 2 shows vaiying concentrations of biotinylated CLASP-2 (FIG. 2A) or Fas (FIG. 2B). C-terminal peptides reacted with immobilized (plate bound) GST polypeptide or GST/PDZ fusion proteins (GST/DLG1 , GST/NeDLG, and GDT/PSD95) in duplicate. The signals were normalized, plotted and fit to a saturation binding curve, yielding an apparent affinity of 21 uM for DLG1-CLASP-2 interaction, 7.5 uM for NeDLG-CLASP-2 interaction, 45 uM for PSD95-CLASP-2 interaction, and 54 uM for DLG 1 -Fas interaction, 54 uM for NeDLG- Fas interaction, and 85 uM for PSD95-Fas interaction. Approach 2:
(1) A fixed concentration of a PDZ-domain polypeptide and increasing concentrations of a labeled PL peptide (labeled with, for example, biotin or fluorescein, see TABLE 4 for representative peptide amino acid sequences) are mixed together in solution and allowed to react. In one embodiment, preferred peptide concentrations are 0.1 uM, 1 uM, 10 uM, 100 uM, 1 mM. In various embodiments, appropriate reaction times can range from 10 minutes to 2 days at temperatures ranging from 4 C to 37 C. In some embodiments, the identical reaction can also be carried out using a non-PDZ domain-containing protein as a control (e.g., if the PDZ- domain polypeptide is fusion protein, the fusion partner can be used). (2) PDZ-ligand complexes can be separated from unbound labeled peptide using a variety of methods known in the art. For example, the complexes can be separated using higb performance size-exclusion chromatography (HPSEC, gel filtration) (Rabinowitz et al., 1998, Immunity 9:699), affinity chromatography(e.g. using glutathione sepharose beads), and affinity absorption (e.g., by binding to an anti-GST-coated plate as described supra). (3) The PDZ-ligand complex is detected based on presence ofthe label on the peptide ligand using a variety of methods and detectors known to one of skill in the art. For example, if the label is fluorescein and the separation is achieved using HPSEC, an in-line fluorescence detector can be used. The binding can also be detected as described supra for the G assay. (4) The PDZ-ligand binding signal is plotted as a function of ligand concentration and the plot is fit. (e.g., by using the Kaleidagraph software package curve fitting algorithm) to the following equation, where "SignaltIigaild]" is the binding signal at PL peptide concentration "[ligand]," "Kd" is the apparent affinity ofthe binding event, and "Saturation Binding" is a constant determined by the curve fitting algorithm to optimize the fit to the experimental data:
SignaltLlj,aπd] = Saturation Binding x ([ligand] / ([ligand + Kd])
Measurement ofthe affinity of a labeled peptide ligand binding to a PDZ-domain polypeptide n is useful because knowledge ofthe affinity (or apparent affinity) of this interaction allows rational design of inhibitors of the interaction with known potency (See EXAMPLE 2). The potency of inhibitors in inhibition would be similar to (i.e. within one-order of magnitude of) the apparent affinity of the labeled peptide ligand binding to the PDZ-domain.
Thus, in one aspect, the invention provides a method of determining the apparent affinity of binding between a PDZ domain and a ligand by immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface, contacting the immobilized polypeptide with a plurality of different concentrations ofthe ligand, determining the amount of binding ofthe ligand to the immobilized polypeptide at each o the concentrations of ligand, and calculating the apparent affinity of the binding based on that data. Typically, the polypeptide comprising the PDZ domain and a non-PDZ domain is a fusion protein. In one embodiment, the e.g., fusion protein is GST-PDZ fusion protein, but other polypeptides can also be used (e.g., a fusion protein including a PDZ domain and any of a variety of epitope tags, biotinylation signals and the like) so long as the polypeptide can be immobilized in an orientation that does not abolish the ligand binding properties of the PDZ domain, e.g, by tethering the polypeptide to the surface via the non-PDZ domain via an anti-domain antibody and leaving the PDZ domain as the free end. It was discovered, for example, reacting a PDZ-GST fusion polypeptide directly to a plastic plate provided suboptimal results. The calculation of binding affinity itself can be determined using any suitable equation (e.g., as shown supra; also see Cantor and Schimmel (1980) BIOPHYSICAL CHEMISTRY WH Freeman & Co., San Francisco) or software.
Thus, in a preferred embodiment, the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain (e.g., an anti- GST antibody when a GST-PDZ fusion polypeptide is used). In a preferred embodiment, the step of contacting the ligand and PDZ-domain polypeptide is carried out under the conditions provided supra in the description o the "G" assay. It will be appreciated that binding assays are conveniently carried out in multiwell plates (e.g., 24-well, 96-well plates, or 384 well plates). The present method has considerable advantages over other methods for measuring binding affinities PDZ-PL affinities, which typically involve contacting varying concentrations of a GST-PDZ fusion protein to a ligand-coated surface. For example, some previously described methods for determining affinity (e.g., using immobilized ligand and GST- PDZ protein in solution) did not account for oligomerization state of the fusion proteins used, resulting in potential errors of more than an order of magnitude. 6.4 Assays to Identify Novel PDZ Domain Binding Moieties and to Identify Inhibitors of PDZ Protein-PL Protein Binding
Although described supra primarily in terms of identifying interactions between PDZ-domain polypeptides and PL proteins, the assays described supra and other assays can also be used to identify the binding of other molecules (e.g., peptide mimetics, small molecules, and the like) to PDZ domain sequences. For example, using the assays disclosed herein, combinatorial and other libraries of compounds can be screened, e.g., for molecules that specifically bind to PDZ domains in hematopoietic cells. Screening of libraries can be accomplished by any of a variety of commonly known methods. See, e.g., the following references, which disclose screening of peptide libraries: Parmley and Smith, 1989, Adv. Exp. Med. Biol. 251:215-218; Scott and Smith, 1990, Science 249:386-390; Fowlkes et al., 1992; BioTechniques 13:422-427; Oldenburg et al., 1992, Proc. Natl. Acad. Sci. USA 89:5393-5397; Yu et al., 1994, Cell 76:933-945; Staudt et al., 1988, Science 241:577-580; Bock et al., 1992, Nature 355:564-566; Tuerk et al., 1992, Proc. Natl. Acad. Sci. USA 89:6988-6992; Ellington et al., 1992, Nature 355:850-852; U.S. Patent No. 5,096,815, U.S. Patent No. 5,223,409, and U.S. Patent No. 5,198,346, all to Ladner et al.; Rebar and Pabo, 1993, Science 263:671-673; and PCT Publication No. WO 94/18318.
In a specific embodiment, screening can be carried out by contacting the library members with a hematopoietic cell PDZ-domain polypeptide immobilized on a solid support (e.g. as described supra in the "G" assay) and harvesting those library members that bind to the protein. Examples of such screening methods, termed "panning" techniques are described by way of example in Parmley and Smith, 1988, Gene 73:305-318; Fowlkes et al., 1992, BioTechniques 13:422-427; PCT Publication No. WO 94/18318; and in references cited hereinabove. In another embodiment, the two-hybrid system for selecting interacting proteins in yeast (Fields and Song, 1989, Nature 340:245-246; Chien et al., 1991, Proc. Natl. Acad. Sci. USA 88:9578-9582) can be used to identify molecules that specifically bind to a PDZ domain- containing protein. Furthermore, the identified molecules are further tested for their ability to inhibit transmembrane receptor interactions with a PDZ domain. In one aspect of the invention, antagonists of an interaction between a PDZ protein and a PL protein are identified. In one embodiment, a modification o the "A" assay described supra is used to identify antagonists. In one embodiment, a modification ofthe "G" assay described supra is used to identify antagonists.
In one embodiment, screening assays are used to detect molecules that specifically bind to PDZ domains in hematopoietic cells. Such molecules are useful as agonists or antagonists of PDZ-protein-mediated cell function (e.g., cell activation, e.g., T cell activation, vesicle transport, cytokine release, growth factors, transcriptional changes, cytoskeletin rearrangement, cell movement, chemotaxis, and the like). In one embodiment, such assays are performed to screen for leukocyte activation inhibitors for drug development. The invention thus provides assays to detect molecules that specifically bind to PDZ domain-containing proteins in hematopoietic cells. For example, recombinant cells expressing PDZ domain-encoding nucleic acids can be used to produce PDZ domains in these assays and to screen for molecules that bind to the domains. Molecules are contacted with the PDZ domain (or fragment thereof) under conditions conducive to binding, and then molecules that specifically bind to such domains are identified. Methods that can be used to carry out the foregoing are commonly known in the art. It will be appreciated by the ordinarily skilled practitioner that, in one embodiment, antagonists are identified by conducting the A or G assays in the presence and absence of a known or candidate antagonist. When decreased binding is observed in the presence of a compound, that compound is identified as an antagonist. Increased binding in the presence of a compound signifies that the compound is an agonist. For example, in one assay, a test compound can be identified as an inhibitor
(antagonist) of binding between a PDZ protein and a PL protein by contacting a PDZ domain polypeptide and a PL peptide in the presence and absence ofthe test compound, under conditions in which they would (but for the presence ofthe test compound) form a complex, and detecting the formation of the complex in the presence and absence of the test compound. It will be appreciated that less complex formation in the presence of the test compound than in the absence ofthe compound indicates that the test compound is an inhibitor of a PDZ protein -PL protein binding. In various embodiments, the PL peptide comprises an amino acid sequence substantially identical to the C-terminal sequence of a PL protein (e.g., CD6, CD49E, CD49F, CD138, Clasp-1, Clasp-4, VCAMl, Clasp-2, CD95, DNAM-1, CD83, CD44, CD4, CD97, Neurexin, CD3n, DOCK2, CD34, FceRIb, or FasLigand).
In one embodiment, the "G" assay is used in the presence or absence of an candidate inhibitor. In one embodiment, the "A" assay is used in the presense or absence of a candiate inhibitor.
In one embodiment (in which a G assay is used), one or more PDZ domain- containing GST-fusion proteins are bound to the surface of wells of a 96-well plate as described supra (with appropriate controls including nonfusion GST protein). All fusion proteins are bound in multiple wells so that appropriate controls and statistical analysis can be done. A test compound in BSA/PBS (typically at multiple different concentrations) is added to wells. Immediately thereafter, 30 uL of a detectably labeled (e.g., biotinylated) peptide known to bind to the relevant PDZ domain (see, e.g., TABLE 2) is added in each of the wells at a final concentration of, e.g., between about 2 uM and about 40 uM, typically 5 uM, 15 uM, or 25 uM. This mixture is then allowed to react with the PDZ fusion protein bound to the surface for 10 minutes at 4°C followed by 20 minutes at 25°C. The surface is washed free of unbound peptide three times with ice cold PBS and the amount of binding of the peptide in the presence and absence ofthe test compound is determined. Usually, the level of binding is measured for each set of replica wells (e.g. duplicates) by subtracting the mean GST alone background from the mean ofthe raw measurement of peptide binding in these wells.
In an alternative embodiment, the A assay is carried out in the presence or absence of an test candidate to identify inhibitors of PL-PDZ interactions.
In one embodiment, a test compound is determined to be a specific inhibitor of the binding of the PDZ domain (P) and a PL (L) sequence when, at a test compound concentration of less than or equal to 1 mM (e.g., less than or equal to: 500 uM, 100 uM, 10 uM, 1 uM, 100 nM or 1 nM) the binding of P to L in the presence of the test compound less than about 50% of the binding in the absence of the test compound, (in various embodiments, less than about 25%, less than about 10%, or less than about 1%). Preferably, the net signal of binding of P to L in the presence ofthe test compound plus six (6) times the standard error ofthe signal in the presence ofthe test compound is less than the binding signal in the absence ofthe test compound.
In one embodiment, assays for an inhibitor are carried out using a single PDZ protein-PL protein pair (e.g., a PDZ domain fusion protein and a PL peptide). In a related embodiment, the assays are carried out using a plurality of pairs, such as a plurality of different pairs listed in TABLE 2. In some embodiments, it is desirable to identify compounds that, at a given concentration, inhibit the binding of one PL-PDZ pair, but do not inhibit (or inhibit to a lesser degree) the binding of a specified second PL-PDZ pair. These antagonists can be identified by carrying out a series of assays using a candidate inhibitor and different PL-PDZ pairs (e.g., as shown in the matrix of TABLE 2) and comparing the results of the assays. All such pairwise combinations are contemplated by the invention (e.g., test compound inhibits binding of PL, to PDZ, to a greater degree than it inhibits binding of PL, to PDZ2 or PL2 to PDZ2). Importantly, it will be appreciated that, based on the data provided in TABLE 2 and disclosed herein (and additional data that can be generated using the methods described herein) inhibitors with different specificities can readily be designed.
For example, according to the invention, the Ki ("potency") of an inhibitor of a PDZ-PL interaction can be determined. Ki is a measure of the concentration of an inhibitor required to have a biological effect. For example, administration of an inhibitor of a PDZ-PL interaction in an amount sufficient to result in an intracellular inhibitor concentration of at least between about 1 and about 100 Ki is expected to inhibit the biological response mediated by the target PDZ-PL interaction. In one aspect of the invention, the Kd measurement of PDZ-PL binding as determined using the methods supra is used in determining Ki.
Thus, in one aspect, the invention provides a method of determining the potency (Ki) of an inhibitor or suspected inhibitor of binding between a PDZ domain and a ligand by immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface, contacting the immobilized polypeptide with a plurality of different mixtures of the ligand and inhibitor, wherein the different mixtures comprise a fixed amount of ligand and different concentrations of the inhibitor, determining the amount of ligand bound at the different concentrations of inhibitor, and calculating the Ki ofthe binding based on the amount of ligand bound in the presence of different concentrations of the inhibitor. In an embodiment, the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain. This method, which is based on the "G" assay described supra, is particularly suited for high-throughput analysis of the Ki for inhibitors of PDZ-ligand interactions. Further, using this method, the inhibition of the PDZ-ligand interaction itself is measured, without distortion of measurements by avidity effects. Typically, at least a portion of the ligand is detectably labeled to permit easy quantitation of ligand binding.
It will be appreciated that the concentration of ligand and concentrations of inhibitor are selected to allow meaningful detection of inhibition. Thus, the concentration ofthe ligand whose binding is to be blocked is close to or less than its binding affinity (e.g., preferably less than the 5x Kd ofthe interaction, more preferably less than 2x Kd, most preferably less than lx Kd). Thus, the ligand is typically present at a concentration of less than 2 Kd (e.g., between about 0.01 Kd and about 2 Kd) and the concentrations of the test inhibitor typically range from 1 nM to 100 uM (e.g. a 4-fold dilution series with highest concentration 10 uM or 1 mM). In a preferred embodiment, the Kd is determined using the assay disclosed supra.
The Ki ofthe binding can be calculated by any of a variety of methods routinely used in the art, based on the amount of ligand bound in the presence of different concentrations of the inhibitor, in an illustrative embodiment, for example, a plot of labeled ligand binding versus inhibitor concentration is fit to the equation: Slπhib, = S0*Ki/([I]+Ki) where Sinhibitor is the signal of labeled ligand binding to immobilized PDZ domain in the presence of inhibitor at concentration [I] and S0 is the signal in the absence of inhibitor (i.e., [I] = 0). Typically [I] is expressed as a molar concentration.
In another aspect of the invention, an enhancer (sometimes referred to as, augmentor or agonist) of binding between a PDZ domain and a ligand is identified by immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface, contacting the immobilized polypeptide with the ligand in the presence of a test agent and determining the amount of ligand bound, and comparing the amount of ligand bound in the presence ofthe test agent with the amount of ligand bound by the polypeptide in the absence of the test agent. At least two-fold (often at least 5-fold) greater binding in the presence ofthe test agent compared to the absence of the test agent indicates that the test agent is an agent that enhances the binding of the PDZ domain to the ligand. As noted supra, agents that enhance PDZ-ligand interactions are useful for disruption (dysregulation) of biological events requiring normal PDZ-ligand function (e.g., cancer cell division and metastasis, and activation and migration of immune cells). The invention also provides methods for determining the "potency" or "Ken,)aπcer" of an enhancer of a PDZ- ligand interaction. For example, according to the invention, the K,,nhancer of an enhancer of a PDZ-PL interaction can be determined, e.g., using the Kd of PDZ-PL binding as determined using the methods described supra. Kenhaιιcer is a measure ofthe concentration of an enhancer expected to have a biological effect. For example, administration of an enhancer of a PDZ-PL interaction in an amount sufficient to result in an intracellular inhibitor concentration of at least between about 0.1 and about 100 Keιl,ιancer (e.g., between about 0.5 and about 50 Kenhancer) is expected to disrupt the biological response mediated by the target PDZ-PL interaction.
Thus, in one aspect the invention provides a method of determining the potency (Ke,lhanc(.-) of an enhancer or suspected enhancer of binding between a PDZ domain and a ligand by immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface, contacting the immobilized polypeptide with a plurality of different mixtures ofthe ligand and enhancer, wherein the different mixtures comprise a fixed amount of ligand, at least a portion of which is detectably labeled, and different concentrations ofthe enhancer, determining the amount of ligand bound at the different concentrations of enhancer, and calculating the potency (!£-.„,,,,,,-„-) of the enhancer from the binding based on the amount of ligand bound in the presence of different concentrations ofthe enhancer. Typically, at least a portion ofthe ligand is detectably labeled to permit easy quantitation of ligand binding. This method, which is based on the "G" assay described supra, is particularly suited for high-throughput analysis of the K^,,,,^^. for enhancers of PDZ-ligand interactions.
It will be appreciated that the concentration of ligand and concentrations of enhancer are selected to allow meaningful detection of enhanced binding. Thus, the ligand is typically present at a concentration of between about 0.01 Kd and about 0.5 Kd and the concentrations of the test agent/enhancer typically range from 1 nM to 1 mM (e.g. a 4-fold dilution series with highest concentration 10 uM or 1 mM). In a preferred embodiment, the Kd is determined using the assay disclosed supra.
The potency ofthe binding can be determined by a variety of standard methods based on the amount of ligand bound in the presence of different concentrations of the enhancer or augmentor. For example, a plot of labeled ligand binding versus enhancer concentration can be fit to the equation:
S([E]) = S(0) + (S(0)*(Dmhancer-l)*[E]/([E]+ Keπlιa cer) where "Kenhallcer" is the potency ofthe augmenting compound, and "Denhancer" is the fold-increase in binding ofthe labeled ligand obtained with addition of saturating amounts ofthe enhancing compound, [E] is the concentration of the enhancer. It will be understood that saturating amounts are the amount of enhancer such that further addition does not significantly increase the binding signal. Knowledge of "K,hancer" is useful because it describes a concentration of the augmenting compound in a target cell that will result in a biological effect due to dysregulation ofthe PDZ-PL interaction. Typical therapeutic concentrations are between about 0.1 and about
Figure imgf000082_0001
Global Analysis of PDZ-PL Interactions
As described supra, the present invention provides powerful methods for analysis of PDZ-ligand interactions, including high-throughput methods such as the "G" assay and affinity assays described supra. In one embodiment ofthe invention, the affinity is determined for a particular ligand and a plurality of PDZ proteins. Typically the plurality is at least 5, and often at least 25, or at least 40 different PDZ proteins. In a preferred embodiment, the plurality of different PDZ proteins are from a particular tissue (e.g., central nervous system, spleen, cardiac muscle, kidney) or a particular class or type of cell, (e.g., a hematopoietic cell, a lymphocyte, a neuron) and the like. In a most preferred embodiment, the plurality of different PDZ proteins represents a substantial fraction (e.g., typically a majority, more often at least 80%) of all ofthe PDZ proteins known to be, or suspected of being, expressed in the tissue or cell(s), e.g., all of the PDZ proteins known to be present in lymphocytes. In an embodiment, the plurality is at least 50%, usually at least 80%, at least 90% or all ofthe PDZ proteins disclosed herein as being expressed in hematopoietic cells (see Table 6).
In one embodiment of the invention, the binding of a ligand to the plurality of PDZ proteins is determined. Using this method, it is possible to identify a particular PDZ domain bound with particular specificity by the ligand. The binding may be designated as "specific" if the affinity ofthe ligand to the particular PDZ domain is at least 2-fold that ofthe binding to other PDZ domains in the plurality (e.g., present in that cell type). The binding is deemed "very specific" if the affinity is at least 10-fold higher than to any other PDZ in the plurality or, alternatively, at least 10-fold higher than to at least 90%, more often 95% of the other PDZs in a defined plurality. Similarly, the binding is deemed "exceedingly specific" if it is at least 100- fold higher. For example, a ligand cound bind to 2 different PDZs with an affinity of 1 uM and to no other PDZs out of a set 40 with an affinty of less than 100 uM. This would constitute specific binding to those 2 PDZs. Similar measures of specifity are used to describe binding of a PDZ to a plurality of PLs. It will be recognized that high specificity PDZ-PL interactions represent potentially more valuable targets for achieving a desired biological effect, the ability of an inhibitor or enhancer to act with high specificity is often desirable. In particular, the most specific PDZ-ligand interactions are also the best therapeutic targets, allowing specific inhibition of the interaction. In an embodiment an interaction between a PDZ and a PL is deemed
Thus, in one embodiment, the invention provides a method of identifying a high specificity interaction between a particular PDZ domain and a ligand known or suspected of binding at least one PDZ domain, by providing a plurality of different immobilized polypeptides, each of said polypeptides comprising a PDZ domain and a non-PDZ domain; determining the affinity ofthe ligand for each of said polypeptides, and comparing the affinity of binding ofthe ligand to each of said polypeptides, wherein an interaction between the ligand and a particular PDZ domain is deemed to have high specificity when the ligand binds an immobilized polypeptide comprising the particular PDZ domain with at least 2-fold higher affinity than to immobilized polypeptides not comprising the particular PDZ domain. In a related aspect, the affinity of binding of a specific PDZ domain to a plurality of ligands (or suspected ligands) is determined. For example, in one embodiment, the invention provides a method of identifying a high specificity interaction between a PDZ domain and a particular ligand known or suspected of binding at least one PDZ domain, by providing an immobilized polypeptide comprising the PDZ domain and a non-PDZ domain; determining the affinity of each of a plurality of ligands for the polypeptide, and comparing the affinity of binding of each of the ligands to the polypeptide, wherein an interaction between a particular ligand and the PDZ domain is deemed to have high specificity when the ligand binds an immobilized polypeptide comprising the PDZ domain with at least 2-fold higher affinity than other ligands tested. Thus, the binding may be designated as "specific" if the affinity ofthe PDZ to the particular PL is at least 2-fold that ofthe binding to other PLs in the plurality (e.g., present in that cell type). The binding is deemed "very specific" if the affinity is at least 10-fold higher than to any other PL in the plurality or, alternatively, at least 10-fold higher than to at least 90%, more often 95% of the other PLs in a defined plurality. Similarly, the binding is deemed "exceedingly specific" if it is at least 100-fold higher. Typically the plurality is at least 5 different ligands, more often at lease 10.
TABLE 6 PDZ Domain-Containing Genes Expressed in T Cells and B Cells
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Use of Array for Global Predictions
One discovery ofthe present inventors relates to the important and extensive roles played by interactions between PDZ proteins and PL proteins, particularly in the biological function of hematopoietic cells and other cells involved in the immune response. Further, it has been discovered that valuable information can be ascertained by analysis (e.g., simultaneous analysis) of a large number of PDZ-PL interactions. In a most preferred embodiment, the analysis encompasses all ofthe PDZ proteins expressed in a particular tissue (e.g., spleen) or type or class of cell (e.g., hematopoietic cell, neuron, lymphocyte, B cell, T cell and the like). Alternatively, the analysis encompasses at least about 5, or at least about 10, or at least about 12, or at least about 15 and often at least 50 different polypeptides; or a substantial fraction (e.g., typically a majority, more often at least 80%) of all of the PDZ proteins known to be, or suspected of being, expressed in the tissue or cell(s), e.g., all of the PDZ proteins known to be present in lymphocytes. In an embodiment, the plurality is at least 50%, usually at least 80%, at least 90% or all ofthe PDZ proteins disclosed herein as being expressed in hematopoietic cells (see Table 6).
In an embodiment the array includes at least one, preferably at least 5 or at least 10 and sometimes all of the following PDZ proteins present in lymphocytes: BAI I associated prot., Connector enhancer, DLG5 (pdlg), DVL3, GTPase, Guanin-exchange factor 1, PDZ domain containing prot, KIAA147, KIAA0300, KIAA0380, KIAA0440, KIAA0545, KIAA0807, KIAA0858, KTAA0902, novel serine protease, PDZKl , PICK8, PTN-3, RPIP8, serine protease, 26s subunit p27, hSYNTENIN, TAXI -IP, TAX2-like protein, wwp3, XI 1 prot. beta, ZO1. It will be apparent from this disclosure that analysis ofthe relatively large number of different interactions preferably takes place simultaneously. In this context, "simultaneously" means that the analysis of several different PDZ-PL interactions (or the effect of a test agent on such interactions) is assessed means together (e.g., the same day or same hour). Typically the analysis is carried out in a highthroughput (e.g., robotic) fashion. One advantage of this method of simultaneous analysis is that it permits rigorous comparison of multiple different PDZ-PL interactions. For example, as explained in detail elsewhere herein, simultaneous analysis (and use of the arrays described infra) facilitates, for example, the direct comparison o the effect of an agent (e.g., an potential interaction inhibitor) on the interactions between a substantial portion of PDZs and/or PLs in a tissue or cell.
Accordingly, in one aspect, the invention provides an array of immobilized polypeptide comprising the PDZ domain and a non-PDZ domain on a surface. Typically, the array comprises at least about 5, or at least about 10, or at least about 12, or at least about 15 and often at least 50 different polypeptides. In one preferred embodiment, the different PDZ proteins are from a particular tissue (e.g., central nervous system, speen, cardiac muscle, kidney) or a particular class or type of cell, (e.g., a hematopoietic cell, a lymphocyte, a neuron) and the like.
In a most preferred embodiment, the plurality of different PDZ proteins represents a substantial fraction (e.g., typically a majority, more often at least 80%) of all ofthe PDZ proteins known to be, or suspected of being, expressed in the tissue or cell(s), e.g., all ofthe PDZ proteins known to be present in lymphocytes. In an embodiment, the plurality is at least 50%, usually at least 80%, at least 90% or all of the PDZ proteins disclosed herein as being expressed in hematopoietic cells (see Table 6). all of the PDZ proteins known to be present in lymphocytes. In an embodiment, the plurality is at least 50%, usually at least 80%, at least 90% or all ofthe PDZ proteins disclosed herein as being expressed in hematopoietic cells (see Table 6).
In an embodiment the array includes at least one, preferably at least 5 and sometimes all of the following PDZ proteins present in lymphocytes: BAI I associated prot., Connector enhancer, DLG5 (pdlg), DVL3, GTPase, Guanin-exchange factor 1, PDZ domain containing prot., KIAA147, KIAA0300, KIAA0380, KIAA0440, KIAA0545, KIAA0807, KIAA0858, KIAA0902, novel serine protease, PDZKl, PICK8, PTN-3, RPIP8, serine protease, 26s subunit p27, hSYNTENIN, TAXI -IP, TAX2-like protein, wwp3, XI 1 prot. beta, ZO1. In this context, "array" refers to an ordered series of of immobilized polypeptides in which the identity of each polypeptide is associated with its location. In some embodiments the plurality of polypeptides are arrayed in a "common" area such that they can be simultaneously exposed to a solution (e.g., containing a ligand or test agent). For example, the plurality of polypeptides can be on a slide, plate or similar surface, which may be plastic, glass, metal, silica, beads or other surface to which proteins can be immobilized. In a different embodiment, the different immobilized polypeptides are situated in separate areas, such as different wells of multi-well plate (e.g., a 24-well plate, a 96-well plate, a 384 well plate, and the like). It will be recognized that a similar advantage can be obtained by using multiple arrays in tandem.
a) Analysis of PDZ-PL Inhibition Profile
In one aspect, the invention provides a method for determining if a test compound inhibits any PDZ-ligand interaction in large set of PDZ-ligand interaction (e.g., some or all of the PDZ-ligands interactions described in Table 2; a majority ofthe PDZ-ligands identified in a particular cell or tissue as described supra (e.g., lymphocytes) and the like. in one embodiment, the PDZ domains of interest are expressed as GST-PDZ fusion proteins and immobilized as described herein. For each PDZ domain, a labeled ligand that binds to the domain with a known affinity is identified as described herein. As disclosed herein, numerous PDZ-PL interactions occur in cells of the hematopoietic system. For any known or suspected modulator (e.g., inhibitor) of a PDL-PL interaction(s), it is useful to know which interactions are inhibited (or augmented). For example, an agent that inhibits all PDZ-PL interactions in a cell (e.g., a lymphocyte) will have different uses than an agent that inhibits only one, or a small number, of specific PDZ-PL interactions. The profile of PDZ interactions inhibited by a particular agent is referred to as the "inhibition profile" for the agent, and is described in detail below. The profile of PDZ interactions enhanced by a particular agent is referred to as the "enhancement profile" for the agent. It will be readily apparent to one of skill guided by the description o the inhibition profile how to determine the enhancement profile for an agent. The present invention provides methods for determining the PDZ interaction (inhibition/enhancement) profile of an agent in a single assay.
In one aspect, the invention provides a method for determining the PDZ-PL inhibition profile of a compound by providing (i) a plurality of different immobilized polypeptides, each of said polypeptides comprising a PDZ domain and a non-PDZ domain and (ii) a plurality of corresponding ligands, wherein each ligand binds at least one PDZ domain in (i), then contacting each of said immobilized polypeptides in (i) with a corresponding ligand in (ii) in the presence and absence of a test compound, and determining for each polypeptide-ligand pair whether the test compound inhibits binding between the immobilized polypeptide and the corresponding ligand.
Typically the plurality is at least 5, and often at least 25, or at least 40 different PDZ proteins. In a preferred embodiment, the plurality of different ligands and the plurality of different PDZ proteins are from the same tissue or a particular class or type of cell, e.g., a hematopoietic cell, a lymphocyte, a neuron and the like. In a most preferred embodiment, the plurality of different PDZs represents a substantial fraction (e.g., at least 80%) of all ofthe PDZs known to be, or suspected of being, expressed in the tissue or cell(s), e.g., all ofthe PDZs known to be present in lymphocytes (for example, at least 80%, at least 90% or all ofthe PDZs disclosed herein as being expressed in hematopoietic cells).
In one embodiment, the inhibition profile is determined as follows: A plurality (e.g., all known) PDZ domains expressed in a cell (e.g., lymphocytes) are expressed as GST- fusion proteins and immobilized without altering their ligand binding properties as described supra. For each PDZ domain, a labeled ligand that binds to this domain with a known affinity is identified. If the set of PDZ domains expressed in lymphocytes is denoted by {PI ...Pn}, any given PDZ domain Pi binds a (labeled) ligand Li with affinity Kdi. To determine the inhibition profile for a test agent "compound X" the "G" assay (supra) can be performed as follows in 96- well plates with rows A-H and columns 1-12. Column 1 is coated with PI and washed. The corresponding ligand LI is added to each washed coated well of column 1 at a concentration 0.5 Ktjl with (rows B, D, F, H) or without (rows A, C, E, F) between about 1 and about 1000 uM) of test compound X. Column 2 is coated with P2, and L2 (at a concentration 0.5 1^2) is added with or without inhibitor X. Additional PDZ domains and ligands are similarly tested.
Compound X is considered to inhibit the binding of Li to Pi if the average signal in the wells of column i containing X is less than half the signal in the equivalent wells of the column lacking X. Thus, in this single assay one determines the full set of lymphocyte PDZs that are inhibited by compound X.
In some embodiments, the test compound X is a mixture of compounds, such as the product of a combinatorial chemistry synthesis as described supra. In some embodiments, the test compound is known to have a desired biological effect, and the assay is used to determine the mechanism of action (i.e., if the biological effect is due to modulating a PDZ-PL interaction). It will be apparent that an agent that modulates only one, or a few PDZ-PL interactions, in a panel (e.g., a panel of all known PDZs lymphocytes, a panel of at least 10, at least 20 or at least 50 PDZ domains) is a more specific modulator than an agent that modulate many or most interactions. Typically, an agent that modulates less than 20% of PDZ domains in a panel (e.g., Table 2) is deemed a "specific" inhibitor, less than 6% a "very specific" inhibitor, and a single PDZ domain a "maximally specific" inhibitor.
It will also be appreciated that "compound X" may be a composition containing mixture of compounds (e.g., generated using combinatorial chemistry methods) rather than a single compound.
Several variations of this assay are contemplated: In some alternative embodiments, the assay above is performed using varying concentrations of the test compound X, rather than fixed concentration. This allows determination ofthe Ki ofthe X for each PDZ as described above.
In an alternative embodiment, instead of pairing each PDZ Pi with a specific labeled ligand Li, a mixture of different labeled ligands is created that such that for every PDZ at least one ofthe ligands in the mixture binds to this PDZ sufficiently to detect the binding in the "G" assay. This mixture is then used for every PDZ domain.
In one embodiment, compound X is known to have a desired biological effect, but the chemical mechanism by which it has that effect is unknown. The assays ofthe invention can then be used to determine if compound X has its effect by binding to a PDZ domain. In one embodiment, PDZ-domain containing proteins are classified in to groups based on their biological function, e.g. into those that regulate chemotaxis versus those that regulate transcription. An optimal inhibitor of a particular function (e.g., including but not limited to an anti-chemotactic agent, an anti-T cell activation agent, cell-cycle control, vesicle transport, apoptosis, etc.) will inhibit multiple PDZ-ligand interactions involved in the function (e.g., chemotaxis, activation) but few other interactions. Thus, the assay is used in one embodiment in screening and design of a drug that specifically blocks a particular function. For example, an agent designed to block chemotaxis might be identified because, at a given concentration, the agent inhibits 2 or more PDZs involved in chemotaxis but fewer than 3 other
PDZs, or that inhibits PDZs involved in chemotaxis with a Ki > 10-fold better than for other PDZs. Thus, the invention provides a method for identifying an agent that inhibits a first selected PDZ-PL interaction or plurality of interactions but does not inhibit a second selected PDZ-PL interaction or plurality of interactions. The two (or more) sets of interactions can be selected on the basis ofthe known biological function ofthe PDZ proteins, the tissue specificity ofthe PDZ proteins, or any other criteria. Moreover, the assay can be used to determine effective doses (i.e., drug concentrations) that result in desired biological effects while avoiding undesirable effects.
b) Side Effects of PDZ-PL Modulator Interactions
In a related embodiment, the invention provides a method for determining likely side effects of a therapeutic that inhibits PDZ-ligand interactions. The method entails identifying those target tissues, organs or cell types that express PDZ proteins and ligands that are disrupted by a specified inhibitor. If, at a therapeutic dosage, a drug intended to have an effect in one organ system (e.g., hematopoietic system) disrupts PDZ-PL interactions in a different system (e.g., CNS) it can be predicted that the drug will have effects ("side effects") on the second system. It will be apparent that the information obtained from this assay will be useful in the rational design and selection of drugs that do not have the side-effect.
In one embodiment, for example, a comprehensive PDZ protein set is obtained. A "perfectly comprehensive" PDZ protein set is defined as the set of all PDZ proteins expressed in the subject animal (e.g., humans). A comprehensive set may be obtained by analysis of, for example, the human genome sequence. However, a "perfectly comprehensive" set is not required and any reasonably large set of PDZ domain proteins (e.g., the set of all known PDZ proteins; or the set listed in Table 6) will provide valuable information.
In one embodiment, the method involves some of all ofthe following steps: a) For each PDZ protein, determine the tissues in which it is highly expressed. This can be done experimentally although the information generally will be available in the scientific literature; b) For each PDZ protein (or as many as possible), identify the cognate PL(s) bound by the PDZ protein; c) Determine the Ki at which the test agent inhibits each PDZ-PL interaction, using the methods described supra; d) From this information it is possible to calculate the pattern of PDZ-PL interactions disrupted at various concentrations of the test agent By correlating the set of PDZ-PL interactions disrupted with the expression pattern of the members of that set, it will be possible to identify the tissues likely affected by the agent.
Additional steps can also be carried out, including determining whether a specified tissue or cell type is exposed to an agent following a particular route of administration. This can be determined using basis pharmacokinetic methods and principles.
Modulation of Activities
The PDZ binding moieties and PDZ protein -PL protein binding antagonists of the invention are used to modulate biological activities or functions of cells (e.g., hematopoietic cells, such as T cells and B cells and the like), endothelial cells, and other immune system cells, as described herein, and for treatment of diseases and conditions in human and nonhuman animals (e.g., experimental models). Exemplary biological acitivities are listed supra.
When administered to patients, the compounds of the invention (e.g., PL-PDZ interaction inhibitors) are useful for treating (ameliorating symptoms of) a variety of diseases and conditions, including diseases characterized by inflammatory and humoral immune responses, e.g., inflammation, allergy (e.g., systemic anaphylaxis, hypersensitivity responses, drug allergies, insect sting allergies; inflammatory bowel diseases, ulcerative colitis, ileitis and enteritis; psoriasis and inflammatory dermatoses, scleroderma; respiratory allergic diseases such as asthma, allergic rhinitis, hypersensitivity lung diseases, and the like vasculitis, rh incompatibility, transfusion reactions, drug sensitivities, PIH, atopic dermatitis, eczema, rhinnitis; autoimmune diseases, such as arthritis (rheumatoid and psoriatic), multiple sclerosis, systemic lupus erythematosus, insulin-dependent diabetes, glomerulonephritis, scleroderma, MCTD, IDDM, Hashimoto thyroiditis, Goodpasture syndrome, psoriasis and the like, osteoarthritis, polyarthritis, graft rejection (e.g., allograft rejection, e.g., renal allograft rejection, graft-vs-host disease, transplantation rejection (cardiac, kidney, lung, liver, small bowel, cornea, pancreas, cadaver, autologous, bone marrow, xenotransplantation)), atherosclerosis, angiogenesis-dependent disorders, cancers (e.g., melanomas and breast cancer, prostrate cancer, leukemias, lymphomas, metastatic disease), infectious diseases (e.g., viral infection, such as HIV, measles, parainfluenza, virus-mediated cell fusion,), ischemia (e.g., post-myocardial infarction complications, joint injury, kidney, scleroderma).
The PL proteins and PDZ proteins listed in TABLE 2 are well characterized, and one of skill, guided by this disclosure (including the discovery of the interactions between PL proteins and PDZ proteins described herein), will recognize many uses for modulators (e.g., enhancers or inhibitors) of PDZ-PL interactions such as those described in TABLE 2. To further assist the reader, a discussion of the characteristics of selected PL proteins (and their function) is provided infra. It will be recognized that this discussion is not comprehensive and is not intended to limit the invention in any way. Moreover, nothing in this section should be construed as an intention by the inventors to be limited to a particular mechanism of action.
A. CD6
As shown supra, CD6 binds PDZ protein 41.8. CD6 is expressed on thymocytes, T cells, and B cell chronic lymphocytic leukemias. CD6 plays a role in T cell co-stimulation and CD6 negative T cells are less autoreactive than CD6 positive T cells. Inhibition of CD6 and CD6/41.8 interactions is predicted to reduce the symptoms of graft- versus-host disease (GVHD) or psoriasis. Thus, in one embodiment ofthe invention, GVHD is reduced in a patient receiving donor bone marrow cells by pre-treating the cells with an effective amount of an antagonist. In combination with post-transplantation immunosuppressive therapy such as FK506, Cellcept, or cyclosporin, CD6-PDZ interaction inhibitors will improve overall survival of transplantation patients (e.g., leukemia patients).
B. CD49e (ALPHA-4) As shown by the experiments reported herein, the C-terminal end of CD49e binds to the PDZ-domain-containing protein41.8kD. CD49e is a 110 kD transmembrane membrane protein ofthe integrin alpha family (integrin alpha 5). Paired with the integrin beta-1 subunit it forms VLA-5. VLA-5 is expressed predominantly on hematopoietic and lymphoid lineage cells including monocytes, basophils, T cells, and activated B cells. VLA-5 is the receptor for the ubiquitously-expressed adhesion molecule fibronectin. Tissue injury such as myocardial infarction releases soluble fragments of fibronectin. Binding of these soluble fragments to VLA- 5 results in chemotaxis of immune cells including monocytes to the source of fibronectin, as well as down-modulation of VLA-5 expression on these cells. Such ligand-induced down-modulation is a common and required feature of chemotaxic receptors. Once immune cells migrate fully to the source of fibronectin, adhesion to the fibronectin surface is enhanced by fibronectin- VLA-5 interaction. Without intending to be bound by a particular mechanism, the 41.8/CD49e interaction is believed to be necessary for proper membrane distribution of CD49e and/or recycling of CD49e such that when it is disrupted, the migration and adherence to fibronectin - containing surfaces is similarly disrupted, resulting in an inability of immune system cells to effectively migrate toward a fibronectin source and adhere to fibronectin-containing surfaces. Such disruption would therefore result in desirable reduced inflammatory processes, including reduced post-myocardial infarction inflammation. Other diseases to be treated include but are not limited to joint inflamation, psoriasis, contact allergy, Crohn's Disease, inflammatory bowel disease, eczema, atopic dermatitis.
C. CD49F (VLA-6 α subunit)
As shown supra, CD49F binds PDZ protein 41.8. CD49f is known as an integrin subunit that pairs either with the βl integrin subunit (CD29), forming VLA-6, or with CD 104 (β4 integrin subunit). The integrin supergene family consists of a number of cell surface αβ heterodimers important for many different physiologic processes, including embryogenesis, thrombosis, wound healing, tumorigenesis and immune responses. Each β chain can pair with various α chains. Both VLA-6 and CD49f/CD104 are widely expressed on epithelia in non- lymphoid tissues. VLA-6 is also expressed on platelets, monocytes, thymocytes and T lymphocytes, with an increased expression on activated and resting memory T cells.
Inhibition of interactions between VLA-6 and 41.8 has a number of therapeutic functions such as the prevention and treatment of metastatic cancers, and treatment of overactive immunity. For example, VLA-6 is associated with invasivion of prostrate carcinoma and plays a role in the metastasis of breast cancer. Blockage of VLA-6 function combined with conventional treatment for prostrate cancer, would be a more effective treatment by preventing metastatic disease (see, Cress et al., 1995, Cancer Metastasis R). Blockage of CD49f through PDZ interaction may also treat Rh incompatibility by blunting memory response or in the treatment of keloids.
D. CD138 (syndecan-l)
CD 138 is a transmembrane proteoglycan receptor with the extracellular domain functioning as a ligand binding domain for various extracellular matrix components and the intracellular portion functioning to alter cytoskeleton and transduce intracellular signals. CD 138 also binds FGF2 and may be a co-receptor for FGF receptor (Yayon et al., 1991).
As shown supra, CD 138 interacts with 41.8kD protein and TIAM1. The c- terminus of CD138 has also been reported to bind the PDZ domains of syntenin and human CASK (Cohen et al., 1998, 1. Cell. Biol. 142:129-138; Grootjans et al., 1997, PNAS 94:13683- 13688; Hsueh et al., 1998, J. Cell. Biol. 142:139-151).CD138 is expressed in pre-B cells, immature B cells, plasma cells, neural cells, the basolateral surface of epithelial cells, embryonic mesenchymal cells, vascular smooth muscle cells, endothelial cells and neural cells but not mature circulating B cells. The interaction between CD 138 and the PDZ domains ofthe 41 ,8kD protein and TIAM1 proteins is believed to be necessary for the proper distribution of CD 138 on the cell surface. Disruption of the interaction by administration of an effective amount of an antagonist is expected to interfere with the migration and adherence of cells to the extracellular matrix, resulting in reduced inflammatory and humoral immune responses. Inhibition of CD138 may be used to treat without limitation diseases such as post-myocardial infarction inflamatory damage, joint injury, rheumatoid arthritis, vasculitis, drug reaction, scleraderma, SLE, Hashimoto thyroiditis, Goodpasture's syndrome, juvenile insulin-dependent diabetes, psoriasis.
E. CD98
As shown supra, CD98 interacts with MPP2. CD98 is expressed at high levels on monocytes and at low levels on T cells, B cells, splenocytes, NK cells, and granulocytes. CD98 plays roles in adhesion, fusion and is a L-type amino acid transporter. CD98 is also involved in virus-mediated cell fusion (e.g. paramyoviruses: parainfluenza virus type 2, Newcastle disease virus, and rubulaviruses)and antagonism of CD98 function is expected to treat )viral infections and limit viral spread. CD98 inhibitors can be an antiviral agent for, but not limited to paramyovirus, parainfluenza, Newcastle disease and rubula. Other roles include treatment for acute leukemias.
F. CLASP-1
As shown supra, CLASP-1 interacts with DLGl, PSD95, and NeDLG. CLASP-1 is a member of a superfamily of immune-cell associated proteins with similar motifs (see PCT/US99/22996 published as WO 00/20434). CLASP-1 functions in the maintenance of the immune synapse. The CLASP-1 transcript is present in lymphoid organs and neural tissue, and the protein is expressed by T and B lymphocytes and macrophages in the MOMA-1 subregion of the marginal zone of the spleen, an area known to be important in T:B cell interaction. CLASP-1 staining of individual T and B cells exhibits a preactivation structural polarity, being organized as a "ball" or "cap" structure in B cells, and forming a "ring", "ball" or "cap" structure in T cells. The placement of these structures is adjacent to the microtubule-organizing center ("MTOC"). CLASP-1 antibody staining indicates that CLASP-1 is at the interface of T-B cell conjugates that are fully committed to differentiation. Antibodies to the extracellular domain of CLASP-1 also block T-B cell conjugate formation and T cell activation.
Antagonism of CLASP-1 function is expected to interfere with immune responses (e.g., T and B cell activation), signal transduction, cell-cell interactions, and membrane organization. Diseases to be treatment by CLASP- 1 agonists/antagonists include, but is not limited to, rheumaotoid arthritis, juvenile diabetes, organ rejection, graft-versus-host disease, scleroderma, multiple sclerosis.
G. CLASP-4
As shown supra, CLASP-4 interacts with DLGl , PSD95, NeDLG, LDP, AF6, 41.8, and MINTl. CLASP-4 is a member of a superfamily of immune-cell associated proteins with similar motifs (see copending U.S.Pat. Application 60/196,527 filed April 11, 2000). The CLASP-4 protein is expressed primarily in peripheral blood lymphocytes. Inhibition of the interaction of CLASP-4 and PDZ domains will interfere with immune responses (e.g., T and B cell activation), signal transduction, cell-cell interactions, and membrane organization. Diesease to be treated bu CLASP-4 agonists/antagonists include, but is not limited to, rheumaotoid arthritis, juvenile diabetes, organ rejection, graft-versus-host disease, scleroderma, multiple sclerosis, acute leukemias, leukemic blast crisis, post-infarction inflamation (cardiac, etc.), atherosclerosis.
H. VCAMl
The vascular cell adhesion molecule- 1 (VCAM-1, CD106) is predominantly expressed by vascular endothelium (i.e.,, endothelial cells) but has been detected in macrophages, dendritic cells, bone marrow-derived cells, fibroblasts, cortical thymic epithelial cells, vascular smooth muscle cells, myoblasts and myotubes. VCAM-1 mediates adhesion through interacting with an integrin ligand, VLA-4, which is expressed by lymphocytes, monocytes and eosinophils. The interaction between VCAM-1 and VLA-4 is important for activation, flattening and extravasation of VLA-4 expressing cells when the endothelium itself has become activated due to inflammation or injury (Salomon et al., 1997, Blood 89:2461-2471; St-Pierre et al., 1996, Eur. J Immunol. 26:2050-2055; Bell et al., 1995, Int. Immunol. 7:1861-1871).
As discussed supra, the C-terminal region of VCAM-1 is a ligand for the PDZ domains of MPPl, DLGl, NeDLG 1, LDP, 41.8 protein, TIAM1 and K303. These interactions are believed to mediate the function of endothelial cell interactions with integrin expressing leukocytes. When the PDZ-PL interactions are disrupted, the adherence of leukocytes to the endothelium will similarly be disrupted, resulting in, e.g., reduction of inflammation. Thus, inhibition of VCAM-1 binding to PDZ proteins is useful for reducing abnormal VCAM-1 inflammatory responses and associated pathologies such as (but not limited to) renal allograft rejection, insulin-dependent diabetes, rheumatoid arthritis, post-myocardial infarction complications and systemic lupus erythematosus (Pasloske et al., 1994, Ann Rev Med 45:283; Ockenhouse et al., 1992, J Exp. Med. 176: 1 183; Solezk et al., 1997, Kidney Int. 51 : 1476; Tedla, et al., 1999, Clin. Exp. Immunol. 117:92-99; Kusterer et al., 1999, Exp Clin Endocrinol Diabetes 107:S102-107; Bonomini et al., 1998, Nephron 79:399; Suassuna et al., 1994, Kidney Int. 46:443; Ferri et al., 1999, Hypertension 34:568).
I. CLASP-2
As shown supra, CLASP-2 interacts with PSD-95, NeDLG, and DLGl . CLASP-
2 is a member of a superfamily of immune-cell associated proteins with similar motifs (see copending U.S. Pat. 09/547,276 filed April 11, 2000; WO 00/10158 filed April 11, 2000; WO 00/10156 filed April 11, 2000). The CLASP-2 transcript is present most strongly in placenta followed by lung, kidney and heart and the protein is expressed in T and B cells, and kidney epithelial cells.
Inhibition o the interaction of CLASP-2 and PDZ domains will interfere with CLASP-2 function resulting in interference with T and B cell function (e.g., T and B cell activation), signal transduction, cell-cell interactions, and membrane organization. In addition, since CLASP-2 is present in heart, blocking CLASP-2 function or expression can selectively block immune responses in the heart (for example, to selectively stop immune response in the heart compartment, e.g., following cardiac transplant rejection or post-MI inflammation, without compromising immunity elsewhere). Other diseases to be treatment by CLASP-1 agonists/antagonists include, but is not limited to, rheumaotoid arthritis, juvenile diabetes, organ rejection, graft-versus-host disease, scleroderma, multiple sclerosis.
J. CD95 (Apo-1/Fas)
CD95 (Fas/Apo-1) and Fas ligand (FasL) are a receptor-ligand pair involved in lymphocyte homeostasis and peripheral tolerance. Binding of Fas by its ligand results in apoptotic cell death, an important major mechanism for safe clearance of unwanted cell during resolution ofthe acute inflammatory response. As is shown supra, CD95 binds the PDZ domains
DLGl, PSD95, NeDLG, and 41.8. Inhibition ofthe interaction of CD95 and PDZ domains.
K. KV1.3 (Shaker Type Kvl.3 Potassium Channel)
As shown supra, Kvl .3 binds DLGl, PSD95, NeDLG, LIMK, 41.8, RGS12, DVLl, and MINTl . Kvl.3 is a Shaker-related channel protein that is involved in modulating the membrane potential of T lymphocytes (Lewis and Cahalan, 1995, Ann. Rev. Immunol.
13:623). Inhibition ofthe Kvl.3 channel chronically depolarizes the T cell membrane, reduces calcium entry via calcium-activated release calcium channels in the plasma membrane, and consequently inhibits the calcium-signaling pathway essential for lymphocyte activation. Hanada et al., reported that Kvl .3 is associated with DLGl and PSD95 in Jurkat T cells (J. Biol.
Chem. 1997, 272:26899). Administration of Kvl .3-PDZ protein agonist/antagonists will disrupt
T cell signaling and can be a useful therapeutic drug to treat, but not limited to, organ transplantation, graft-versus-host disease, Crohn's Disease, Ulcerative colitis, inflammatory bowel disease, rheumatoid arthritis, osteoarthritis, multiple sclerosis, scleroderma, mixed connective tissue disease.
L. DNAM-1
As shown supra, DNAM-1 binds several PDZ proteins, including MPP2, DLGl ,
PSD95, NeDLG, LIM, AF6, 41.8 and RGS 12. DNAM-1 is associated with Fyn constitutively but required the presence of pervanadate (a tyrosine phosphatase inhibitor) (see Shibuya, et al.
1999, Immunity. 1 1 :615-623). Upon stimulation with anti-CD3 (or DNAM-1), DNAM-1 is phosphorylated at Serine 329 (Shibuya, et al. 1998, J. Immun. 161:1671-1676.) and associates with LFA-1. Furthermore, Fyn becomes associated with DNAM-1 independent of pervanadate. Fyn phosphorylates DNAM-1 at Y 322, but does not require Y322 to continue binding to DNAM-1. Since DNAM-1 itself does not have a SH3 binding domain but only has the Src phosphorylation site at Y322, an adaptor molecule must be present to bridge DNAM-1 and Fyn. DLGl has been described in the literature to be present in T cells (Hanada, et al. 1997. J Biol Chem 272:26899), but does not bind Fyn. PSD95 does not have a SH3 binding site. However, several of the other PDZ proteins do have SH3 binding domains including but not limited to NeDLG, RGS12, MPP2 and can fulfill this function. These adaptor PDZ listed supra binds to DNAM-1 through its PDZ domain and simultaneously binds to the SH3 domain of Fyn through its proline-rich sequences just N-terminal to the PDZ domain. The Y139 is a candidate phosphorylation site to control association of Fyn to DNAM-1 and the adaptor PDZ.
Based on this analysis, inhibition of PDZ association with DNAM-1 using the reagents of the invention will inhibit Fyn association with DNAM-1 and the subsequent Y322 phosphorylation and activation of cytotoxic T cells. Diseases that can be treated include but are not limited to Crohn's Disease, multiple sclerosis, ulcerative colitis, inflammatory bowel disease, graft-versus-host, juvenile diabetes, Hashimoto's disease.
M. CD83 (HB15)
CD83 is a transmembrane glycoprotein, expressed predominantly on activated dendritic cells (DCs), Langerhans cells in the skin, with some weak expression detected on activated peripheral lymphocytes, and interdigitating reticulum cells within the T cell zones of lymphoid organs (Zhou and Tedder, 1995, J. Immunol. 154:3821-3835; Zhou et al, 1992, J. Immunol. 149:735-742). CD83 is up-regulated de novo upon activation of an immature DCs, and is the major discriminating marker and characteristic for activated, mature DCs (Czerniecki et al, 1997, J. Immunol. 159:3823-3837). DCs function as antigen presenting cells (APCs).
Upregulation and expression of CD83 thus appears to be required for DCs to mature and function as APCs. As shown by experiments described supra, the CD83 binds to the PDZ domains of DLGl , PSD95, and NeDLG. These interactions between CD83 and PDZ domains, and between CD83 and DLGl , PSD95, and NeDLG are believed to be important for proper distribution and recycling of CD83. Disruption of CD83 and PDZ proteins with agonists and antagonist can be used to treat, but not limited to, psoriasis, cancers, allergies, autoimmune diseases such as multiple sclerosis, system lupus ery hematosis.
N. CD44 (phagocytic glycoprotein 1, lymphocyte homing receptor, p85 and HCAM)
CD44 is single pass transmembrane protein that has several different isoforms due to alternative splicing. It has a broad pattern of expression being detected on both hematopoietic and non-hematopoietic cell types including epithelial, endothelial, mesenchymal and neuronal cells. CD44H is a major isoform that is expressed in lymphoid, myeloid and erythroid cells
(reviewed in Barclay et al., 1997, The Leukocyte Antigen Facts Book, 2ed, Academic Press).
CD44 is a receptor for hyaluronate (HA), which is a constituent of the extracellular matrix
(ECM). In the immune system, CD44 functions as an adhesion molecule on the surface of leukocytes and erythrocytes that binds HA polymers in the ECM, and it can also act as a signaling receptor when HA becomes soluble during inflammatory reactions or tissue damage. The cytoplasmic region of CD44 has been shown to bind or be associated with the actin cytoskeleton through interactions with spectrin and members of the ERM (ezrin, radixin, and meosin) family (reviewed in Lesley et al., 1993; Bajorath, 2000, Proteins 39:103-111). Additionally, CD44 is associated with the non-receptor tyrosine kinase p56Lck (Taher et al., 1996, J Biol. Chem. 271 :2863-2867. CD44 has been shown to be a co-stimulatory molecule with CD3/TCR engagement to activate T cells (reviewed in Aruffo, 1996, J. Clin. Invest. 98:2191-2192).
As described supra by experiments reported herein, the C-terminus of CD44 is a ligand for the PDZ domain contained in MPPl, prIL-16 and MINTl . It is believed that the interactions of CD44 with PDZ domains, and between CD44 with MPPl, prIL-16 and MINTl function in maintenance of leukocyte structure and in leukocyte signaling. Thus, when a CD44-PDZ interaction is disrupted, CD44 will fail to transduce proper intracellular signals, and maintain proper distribution of CD44 on the surface, which will prevent adhesion of leukocytes to the endothelium during inflammation and tissue damage. Administration of agonists/antagonists of this interaction will thus result in, but not limited to, reduced inflammatory responses during tissue ischemia and cell lysis (e.g., rhabdomyosis), vascular insufficiencies (e.g. frostbite), psoriasis, eczema, graft-versus-host disease, granuloma annulare, scleroderma.
O. CD97 (CD55)
As discussed supra, CD97 binds the PDZ domains of DLGl and 41.8. CD97 is a 79.7 kD seven-span transmembrane protein expressed on granulocytes and monocytes and at low levels on resting T cells and B cells. Upon T or B cell activation expression levels of CD97 in T cells and B cells increases rapidly (Eichler et al., 1994, Scand. J. Immunol. 39, 111-115; Pickl et al, 1995, Leukocyte Typing :1151-1 153). When expressed on COS cells, CD97 confers adhesion to lymphocytes and to erythrocytes. According to the present invention, the interaction of CD97 with DLGl and the
41.8 kd protein can be altered to interfere with proper membrane distribution of CD97 and/or recycling of CD97. Such modulation will affect CD97 dependent adherence of cells with therapeutic benefit. Without being limited, agonists and antagonists of CD97-PDZ protein interaction can be used to treat rheumatoid arthritis, osteoarthritis, Crohn's Disease, Ulcerative colitis, psoriasis.
P. Glycophorin C (GC)
As is shown supra, the c-terminus of Glycophorin C (GC) interacts with the PDZ domains of human DLG, PSD95, NeDLG, MMP2, AF6, 41.8, and MINTl (with Mint-1 described previously). Glycophorin C is an integral membrane protein expressed in erythroid cells, thymus, stomach, breast, adult and fetal liver, monocytes, T and B cells (Le Van Kim et al., 1989, J. Biol. Chem. 264:20407-20414) and is known for its role in human erythrocytes where it interacts with MPPl and protein 4.1 to regulate the shape, integrity and mechanical stability of red cells (Marfatia et al., 1997, J. Biol. Chem, 272:24191-24197; Reid et al., 1987, 5/ootf 69:1068-1072.).
Interactions between Glycophorin C and PDZ proteins DLG, PSD95, NeDLG, MMP2, AF6, 41.8, and MINTl are believed necessary for maintenance ofthe physical integrity of cells in which they are expressed. Modulation of GC-PDZ interactions will alter with the function of these and can be utilized to treat, but not limited to, polycythemia vera, spherocytosis.
CDwϊ28A (IL8RA) As is described supra, CDW128A binds to the PDZ domains of DLGl and NeDLG. There are two forms for the IL-8 receptor, IL-8RA (CDwl28A) and IL-8RB (CDwl28B) both of which are members of the G protein-coupled receptor superfamily and chemokine receptor branch of rhodopsin family. CDwl28A and CDwl28B both bind IL-8 with the same affinity but only CDwl28B, binds three other IL-8-related CXC chemokines: melanoma growth-stimulating activity (GRO/MGSA), neutrophil-activating peptide 2 (NAP-2) and ENA-78. See, e.g., Ahuja, SK. And Murphy, PM. 1996. J Biol Chem 271 :20545-50.
CDwl28A is expressed on all granulocytes, a subset of T cells, monocytes, endothelial cells, keratinocytes, erythrocytes, and melanoma cells. IL-8 induces chemotaxis of neutrophils, basophils, and T lymphocytes and increases neutrophil and monocyte adhesion to endothelial cells. The binding of IL-8 to IL8RA induces a transient increase in intracellular calcium levels, activation of phospholipase D, a respiratory burst of neutrophils and chemotaxis. This pro-inflammatory response is effective in normal immune responses. Inhibitors of CDwl28A are useful for treatment of psoriasis, rheumatoid arthritis, polyarthritis, and for control of angiogenesis-dependent disorders such as melanomas and breast cancer.
(R) CD3-eta(η)
CD3-η is a splice variant of CD3 zeta and a component ofthe CD3/TCR complex, which is required for antigen recognition, signal transduction and activation of T cells (Weiss and Liftman, 1994, Cell 76:263-274.). See, Barclay et al., 1997, The Leucocyte antigen facts book, 2nd Ed, Academic Press. As shown by experiments reported herein, the C-terminal region of CD3-η is a ligand for the PDZ domains of MINTl, 41.8 protein, DLGl, and PSD95. The interactions of CD3-η with PDZ domains, and of CD3-η with MTNT1, 41.8 protein, DLGl, and PSD95 are believed to be important activation of T cells, which is required for all cellular immune responses. Modulation of this interaction by agonists and antogonists can be used to treat, but is not limited to, acute and chronic allograft rejection, multiple sclerosis, graft-versus- host disease, rheumatoid arthritis.
(S) LPAP (CD45-AP, LSM-1) LPAP is a transmembrane protein expressed on resting and activated T- and B- cells. LPAP has been shown to bind to CD45, a protein that is part of the T-cell receptor complex and has been found to co-localize with CD4, CD2 and Thy-1. LPAP has also been co- immune precipitated with p56(lck) and ZAP-70. The actual function of LPAP is unknown, but it has been suggested that it is an assembly molecule for the CD45 complex.
As shown supra, LPAP binds to DLG-1 and MINT-1. Notably, DLG-1 and MINT-1 are both expressed in T-cells. It has also been shown that DLG-1 co-precipitates with p56(lck) in T-cells. The assays described herein also demonstrated that DLG-1 binds to CD95 and KV1.3, and binding of MINT-1 to KV1.3. All of these molecules are involved in signaling by the TCR. LPAP is believed to function in organizing the signaling by CD45 in T-cell activation, possibly by recruiting p56(lck) as a substrate for CD45. Blocking the function of CD45 has been shown to severely impair the T-cell response. Inhibiting the interaction between LPAP and PDZ proteins is expected to alter the CD45-mediated path from the rest ofthe immune response. Agonists and antagonists of PL - PDZ binding can be used to treat, but is not limited to, rheumatoid arthritis, transplant rejection, multiple sclerosis, scleroderma, graft-versus-host disease.
(T) CD46 (Complement membrane cofactor protein (MCP))
CD46 is a membrane protein expressed on all nucleated cells, but not on erythrocytes. CD46 is a member o the regulator of complement activation protein family. Its primary function is the protection of cells from complement attack by inactivating membrane deposited C3b/C4b complement (Liszewski, et al., 1999, Adv. Immunol. 61 :201-283). CD46 exists in more than 8 isoforms that are generated by differential splicing, with molecular weights ranging from 45 to 70 kD. In addition to the above function, CD46 also serves as the receptor for the measles virus and for other pathogenic microorganisms (e.g. Streptococcus pyogenes) (Manchester et al, 1994, Proc. Natl Acad Sci. USA 91 :2161; Okada et al., 1995, Proc. Natl Acad. Sci. USA 92:2489-2493.). CD46 also appears to be over-expressed on certain tumors (Jurianz et al., 1999, Mol Immunol 36:929-939) thus rendering tumor cells insensitive to the action of complement. See, Barclay et al., (1997) The Leucocyte antigen facts book, 2ed, Academic Press.
As shown supra, CD46 binds DLGl, PSD95 and Ne-DLG. This interaction is believed to be necessary for proper membrane distribution of CD46 and/or recycling of CD46. Alteration of the CD46-PDZ interaction can reduce the ability of measles virus and other pathogens to enter cells, renders CD46-expressing tumors susceptible to attack by complement. The administration of CD46-PDZ interaction agonists and antagonists is useful for the treatment of , but not limited to, cancers and viral infectious diseases.
(U) CDwl28B
As is described supra, CDwl28B binds to the PDZ domains of DLGl, NeDLG, PSD95, and 41.8 in the assays described supra. There are two forms for the IL-8 receptor, IL- 8RA (CDwl28A) and IL-8RB (CDwl28B) both of which are members ofthe G protein-coupled receptor superfamily and chemokine receptor branch of rhodopsin family. CDwl28A and CDwl28B both binds IL-8 with equal affinity but only CDwl28B, also binds three other IL-8- related CXC chemokines: melanoma growth-stimulating activity (GRO/MGSA), neutrophil- activating peptide 2 (NAP-2) and ENA-78. See, e.g., Ahuja, SK and Murphy, PM. 1996. J Biol Chem 271 :20545-50.
CDwl28B is expressed on all granulocytes, a subset of T cells, monocytes, endothelial cells, keratinocytes, erythrocytes, and melanoma cells. IL-8 induces chemotaxis of neutrophils, basophils, and T lymphocytes but diminished relative to IL8RA and increases neutrophil and monocyte adhesion to endothelial cells. The binding of IL-8A to its receptor induces a transient increase in intracellular calcium levelsand granule release but does not induce activation of phospholipase D or a respiratory burst in neutrophils. This pro-inflammatory response is effective in normal immune responses. Inhibitors of CDwl28B are useful for treatment of psoriasis, rheumatoid arthritis, polyarthritis, and for control of angiogenesis- dependent disorders such as melanomas and breast cancer.
(V) DOCK2 The DOCK family are a group of morphogenetic transmembrane proteins that interacts with cytoskeleton to affect cell shape changes. Members of this new family include Drosophila myoblast city (mbc), DOCK180, DOCK2, DOCK3, CED5, KIAA0209 and CLASP. The prototypical molecule, DOCK1 or DOCK180 is the human homolog of C. elegans gene, CED5 which is involved in the engulfment and phagacytosis by macrophages of apoptotic cells. DOCK2 is found in peripheral blood lymphocytes and can convert a flatten cell into a rounded morphology upon transfection (Nagase, et. al. 1996. DNA Res 3:321-329, Nishihara, H. 1999. Hokkaido Igaku Zasshi 74:157).
As shown supra, DOCK2 is a PL and binds to PDZ proteins. Using PDZ proteins as an adaptor, DOCK2 complexes with A,B,C to control cell shape in preparation for transit of lymphocytes for vascular circulation. Modulation of DOCK2 by agonists and antagonists of its PDZ protein interaction can be used to treat, but is not limited to, acute leukemia, blast crisis, post-myocardial infarction inflammation, post-traumatic inflammation.
W. CD34
As is shown supra, CD34 binds DLGl, PSD95, and NeDLG. CD34 is expressed on a small subpopulation of bone marrow cells which includes hematopoietic stem cells. CD34 is also present on bone marrow stomal cells and on endothelial cells. The selectins CD62L (L- selectin) and CD62E (E-selectin) bind CD34. CD34 mediates attachment and rolling of leukocytes. The hematopoietic stem cell properties of CD34 include myeloid differentiation of stem cells. Modulation of the CD34-PDZ interaction with agonists and/or antagonists can be used to treat, but is not limited to, myelodysplasia, leukemias, post-traumatic inflammation, post- myocardial infarction inflammation.
X. Fc epsilon receptor beta I chain (FcεRβl)
The high affinity receptor for human IgE, FcεRI, is composed of an α, β, and disulfide-linked γ homodimer. The α-chain binds the Fc portion of IgE, whereas the β-chain serves to amplify signals that are transduced through the γ-chain homodimer. Both αβγ2 tetramer and αγ2 trimer complexes exist, but the β-chain amplifies the signal 5- to 7-fold, as measured by Syk activation and calcium mobilization. Additionally, the FcεRβl is a PDZ ligand and is a member of the CD20/ FcεRβl receptor family. As is shown supra, FcεRβl binds MINTl.
As the high-affinity receptor for IgE, FcβRI on basophils and mast cells plays a central role in the initiation of allergic responses. Signaling through the FcβRI begins by crosslinking of a multivalent allergen bound to IgE. The result is vesicular degranulation, release of histamine, leukotrienes and pro-inflammatory cytokines (IL-6 and TNFα), factors responsible for the symptoms of immediate hypersensitivity. Alteration of signaling by targeting PL/PDZ interaction with agonists and antagonists can be used to treat, but is not limited to, asthma, atopic dermatitis, eczema, drug reaction, mastocytosis, urticaria, eosinophilia myalgia syndrome (Turner, H., et. al., 1999, Nature 402 SUPP:B24).
Y. FAS Ligand (FasL) CD95 (Fas/Apo-1) and Fas ligand (FasL) are a receptor-ligand pair critically involved in lymphocyte homeostasis and peripheral tolerance. Binding of Fas by its ligand results in apoptotic cell death, an important major mechanism for safe clearance of unwanted cell during resolution of the acute inflammatory response. FasL is mainly restricted to activated T lymphocytes and is rapidly induced. Fas ligand is frequently up-regulated in breast cancer, as compared with normal breast epithelial cells and benign breast disease. As is shown supra, FasL binds the KIAA0561 PDZ domain. The PDZ-PL modifiers are useful for treatment of, but not limited to, tumors, e.g., tumors unresponsive to conventional chemotherapy.
Z. CDW125 (IL5R) As is shown supra, CDW125 binds PTN-4 and RGS12. CDW125 is an IL-5 receptor expressed on eosinophils and basophils. IL5 promotes growth and differentiation of eosinophil precursors and actives mature eosinophils (Takatsu et al 1994, Adv. Immunol. 57:145- 190). The secreted form of CDwl25 has antagonistic properties and is able to inhibit IL-5- induced eosinophil proliferation and differentiation. Modulation of CDW125 binding to PDZ domains may be used to treat, but is not limited to, asthma, atopic dermatitis, eczema, drug reaction, urticaria, mastocytosis, eosinophilia.
AA. Burkitt's lymphoma receptor-1 (BLR-1; CXCR5)
BLR-1 is a transmembrane receptor detected primarily on B cells, and shown to be upregulated in stimulated T cells (Dobner et al., 1992, EurJ. Immunol. 22:2795-2799.; Flynn et al., 1998, J. Exp. Med. 188:297-304). BLR-1 functions in chemotaxis of B and T cells into follicles of secondary lymphoid organs (e.g. spleen) for proper development and selection toward antigens (Forster et al., 1996, Cell 87: 1037-1047. Its ligand is B-lymphocyte chemoattractant (BLC), which is strongly expressed in the follicles of Peyer's patches, spleen and lymph nodes (Gunn et al., 1998, Nature 391 :799-803). Consistent with its chemotactic role is the demonstration that BLR-1 expression is downregulated in developed, activated B cells (plasma cells) to prevent them from being retained in follicles (Forster et al., 1994, Cell Mol Biol 40:381- 387), and blr -/- B cells fail to migrate to B cell follicles (Forster et al., 1996).
As shown by experiments reported herein, the C-terminal end of BLR-1 binds to the PDZ domain containing protein MINTl . Without intending to be bound by a particular mechanism, the interaction between BLR-1 and MINTl, and BLR-1 and PDZ domains is necessary for the proper distribution and signaling of BLR-1 on the cell surface. When this interaction is disrupted, the chemotactic abilities of lymphoid cells expressing BLR-1 is similarly disrupted. Such a disruption results in a reduced immune response, interference with the ability of lymphocytes to properly circulate and develop responses to antigen. Agonists and antagonists of this interaction can be used to treat, but is not limited to, systemic lupus erythematosus, scleroderma and other autoimmune diseases.
BB. CD4
CD4 is a co-receptor with the T cell receptor (TCR) involved in antigen recognition. Both CD4 and TCR belong to the immunoglobulin supergene family. T cell activation is enhanced by increasing the avidity of T cells for effector and target cells. The cytoplasmic domain is involved in signal transduction and association with the tyrosine kinase p56'clc. CD4 is expressed on most fhymocytes, two-thirds of peripheral blood T lymphocytes, monocytes and macrophages. Human immunodeficiency virus type-1 (HIV-1) infects cells by membrane fusion mediated by its envelope glycoproteins (gpl20-gp41) and is triggered by the interaction of CD4 and a chemokine co-receptor, CCR5 or CXCR4. Modulation of CD4-PDZ inhibitors with agonists and antagonists can be used to treat, but is not limited to, HIV infection immediately after exposure to HIV, rheumatoid arthritis, multiple sclerosis, scleroderma, systemic lupus erythematosis, psoriasis.
6.5 Agonists and Antagonists of PDZ-PL Interactions
As described herein, interactions between PDZ proteins and PL proteins in cells
(e.g., hematopoietic cells, e.g., T cells and B cells) may be disrupted or inhibited by the administration of inhibitors or antagonists. Inhibitors can be identified using screening assays described herein. In embodiment, the motifs disclosed herein are used to design inhibitors. Sn some embodiments, the antagonists of the invention have a structure (e.g., peptide sequence) based on the C-terminal residues of PL-domain proteins listed in TABLE 2. In some embodiments, the antagonists ofthe invention have a structure (e.g., peptide sequence) based on a PL motif disclosed herein. The PDZ/PL antagonists and antagonists ofthe invention may be any of a large variety of compounds, both naturally occurring and synthetic, organic and inorganic, and including polymers (e.g., oligopeptides, polypeptides, ohgonucleotides, and polynucleotides), small molecules, antibodies, sugars, fatty acids, nucleotides and nucleotide analogs, analogs of naturally occurring structures (e.g., peptide mimetics, nucleic acid analogs, and the like), and numerous other compounds. Although, for convenience, the present discussion primarily refers antagonists of PDZ-PL interactions, it will be recognized that PDZ-PL interaction agonists can also be use in the methods disclosed herein.
In one aspect, the peptides and peptide mimetics or analogues o the invention contain an amino add sequence that binds a PDZ domain in hematopoietic cells such as T cells and B cells, or otherwise inhibits the association of PL proteins and PDZ proteins. In one embodiment, the antagonists comprise a peptide that has a sequence corresponding to the carboxy-terminal sequence of a PL protein listed in TJ BLE 2, e.g., a peptide listed TABLE 4. Typically, the peptide comprises at least the C-terminal four (4) residues ofthe PL protein, and often the inhibitory peptide comprises more than four residues (e.g., at least five, six, seven, eight, nine, ten, twelve or fifteen residues) from the PL protein C-terminus. See, e.g. Section 6.5.1, infra. Moreover, the C-terminal domains of specific surface receptors expressed by hematopoietic system and endothelial cells may themselves be used as inhibitors, and may be used as the basis for rational design of non-peptide inhibitors. See Section 6.6, infra.
In some embodiments, the antagonist is a fusion protein comprising such a sequence. Fusion proteins containing a transmembrane transporter amino acid sequence are particularly useful. See, e.g. Section 6.5.2, infra.
In some embodiments, the inhibitor is conserved variant of the PL C-teπninal protein sequence having inhibitory activity. See, e.g. Section 6.5.3, infra.
In some embodiments, the antagonist is a peptide mimetic of a PL C-terminal sequence. See, e.g. Section 6.5.4, infra. In some embodiments, the inhibitor is a small molecule (i.e., having a molecular weight less than 1 kD). See, e.g. Section 6.5.5, infra.
6.5.1 Peptide Antagonists
In one embodiment, the antagonists comprise a peptide that has a sequence of a PL protein oarboxy-terminus listed in TABLE 2. The peptide comprises at least the C-terminal two (2) residues ofthe PL protein, and typically, the inhibitory peptide comprises more than two residues (e.g, at least three, four, five, six, seven, eight, nine, ten, twelve or fifteen residues) from the PL protein C-terminus. Most often, the residues shared by the inhibitory peptide with the PL protein are found at the C-terminus of the peptide. However, in some embodiments, the sequence is internal. Similarly, in some cases, the inhibitory peptide comprises residues from a PL sequence that is near, but not at the c-terminus of a PL protein (see,Gee et al., 1998, J Biological Chem. 273:21980-87).
For example, the "core PDZ motif sequence" of a hematopoietic cell surface receptor at its C-terminus contains the last four amino acids, this sequence may be used to target PDZ domains in hematopoietic cells. The four amino acid core of a PDZ motif sequence may contain additional amino acids at its amino terminus to further increase its binding affinity and/or stability. In one embodiment, the PDZ motif sequence peptide can be from four amino acids up to 15 amino acids. It is preferred that the length of the sequence to be 6-10 amino acids. More preferably, the PDZ motif sequence contains 8 amino acids. Additional amino acids at the amino terminal end of the core sequence may be derived from the natural sequence in each hematopoietic cell surface receptor or a synthetic linker. The additional amino acids may also be conservatively substituted. When the third residue from the C-terminus is S, T or Y, this residue may be phosphorylated prior to the use ofthe peptide.
In some embodiments, the peptide and nonpeptide inhibitors ofthe are small, e.g., fewer than ten amino acid residues in length if a peptide. Further, it is reported that a limited number of ligand amino acids directly contact the PDZ domain (generally less than eight) (Kozlov et al., 2000, Biochemistry 39, 2572; Doyle et al., 1996, Cell 85, 1067) and that peptides as short as the C-terminal three amino acids often retain similar binding properties to longer (> 15) amino acids peptides (Yanagisawa et al., 1997, J. Biol. Chem. 272, 8539). FIGURES 3 -H show the use of peptides to inhibit PL-PDZ interactions using the G assay described supra. In FIGURE 3A and B, the inhibiton assays were carried out using GST fusion proteins containing PDZ domains from DLGl or PSD95 (see supra and TJABLE 3). Binding of biotinylated PL peptides for Clasp 2, CD46, Fas, or KV1.3 (as listed in TABLE 4) was determined in the presence of various competitor peptides (at a concentration of 100 uM) or in the absence of a competitor (equalized as 100% binding). The competitor peptides were 8-mers peptides having the sequence of C-terminus of Clasp 2 (MTSSSSVV), CD46 (REVKFTSL), or Fas (RNEIQSLV), a unlabeled 19-mer having the sequence of c-terminus of KV1.3 (i.e., non-biotinylated AA33L as listed in TABLE 3), or a peptide having the sequence of residues 64-76 of hemoglobin (Vidal et al., 1999, J. Immunol. 163, 4811), i.e., an unrelated competitor. The binding of biotinylated peptide (10 uM for Fas and KV1.3, 20 uM for Clasp 2 and CD46) to GST alone was subtracted from the binding to the fusion proteins to obtain the net signal for each experimental condition. This net signal was then normalized by dividing by the signal in the absence of competitor peptide and the data were plotted. Error bars indicated the standard deviation of duplicate measurements. Specific inhibition of Clasp 2 PL-DLG PDZ binding was observed with the CLASP-2 8-mer, the CD46 8-mer, the FAS-8-mer, and the KV13 peptide, but not in the absence of peptide or using an unrelated peptide.
FIGURES 3C-F show similar assays using shorter peptides to inhibit (e.g., a 3- mer and a 5-mer). Figures 3C-E show binding of biotinylated PL peptides for Clasp 2, CD46, Fas, or KV1.3, at the indicated concentration (as listed in TABLE 3) to GST fusion proteins containing PDZ domains from NeDLG, DLGl, or PSD95 in the absence or presence of 1 mM 3-mer peptide having the sequence ofthe C-terminus of Clasp 2 (SVV). (Table 3). FIGURE 3F shows the effect on binding of a 5-mer CD49E peptide (ATSDA) to GST fusion proteins containing a PDZ domain from 41.8Kd
6.5.2 Peptide Variants Having identified PDZ binding peptides and PDZ-PL interaction inhibitory sequences, variations of these sequences can be made and the resulting peptide variants can be tested for PDZ domain binding or PDZ-PL inhibitory activity. In embodiments, the variants have the same or a different ability to bind a PDZ domain as the parent peptide. Typically, such amino acid substitutions are conservative, i.e., the amino acid residues are replaced with other amino acid residues having physical and/or chemical properties similar to the residues they are replacing. Preferably, conservative amino acid substitutions are those wherein an amino acid is replaced with another amino acid encompassed within the same designated class.
6.5.3 Peptide Mimetics
Having identified PDZ binding peptides and PDZ-PL interaction inhibitory sequences, peptide mimetics can be prepared using routine methods, and the inhibitory activity ofthe mimetics can be confirmed using the assays ofthe invention. Thus, in some embodiments, the antagonist is a peptide mimetic of a PL C-terminal sequence. The skilled artisan will recognize that individual synthetic residues and polypeptides incorporating mimetics can be synthesized using a variety of procedures and methodologies, which are well described in the scientific and patent literature, e.g., Organic Syntheses Collective Nolumes, Gilman et al. (Eds) John Wiley & Sons, Inc., ΝY. Polypeptides incorporating mimetics can also be made using solid phase synthetic procedures, as described, e.g., by Di Marchi, et al., U.S. Pat. No. 5,422,426. Mimetics o the invention can also be synthesized using combinatorial methodologies. Various techniques for generation of peptide and peptidomimetic libraries are well known, and include, e.g., multipin, tea bag, and split-couple-mix techniques; see, e.g., al-Obeidi (1998) Mol. Biotechnol. 9:205-223; Hruby (1997) Curr. Opin. Chem. Biol. 1 :114-119; Ostergaard (1997) Mol. Divers. 3:17-27; Ostresh (1996) Methods Enzymol. 267:220-234.
6.5.4 Small Molecules
In some embodiments, the inhibitor is a small molecule (i.e., having a molecular weight less than 1 kD). Methods for screening small molecules are well known in the art and include those described supra at Section 6.4.
6.6 Cell Surface Receptors and PDZ-Domain Binding Sequences
The following sections describe specific surface receptors expressed by different cell types in the hematopoietic and immune response system. The C-termini of these receptors are used as inhibitors, or serve as the basis for designing PDZ motif sequence peptides, variants, fusion proteins, peptidomimetics, and small molecules for use in inhibiting PDZ-PL interactions.
In a preferred embodiment, the peptides are tested in an assay ofthe invention for inhibitory or modulator/ activity (also see, TABLE 4, and discussion supra).
6.6.1 PDZ Motif Sequences of T Cell Surface Receptors A number of surface receptors expressed by T cells contain a PDZ motif sequence
(PL sequence). These molecules include CD3η, CD4, CD6, CD38, CD49e, CD49f, CD53,
CD83, CD90, CD95, CD97, CD98, CDwl37 (41BB), CD166, CDwl28 (IL8 R), DNAM-1, Fas ligand (FasL) and LPAP (Barclay et al., 1997, The Leucocyte Antigen Facts Book, second edition, Academic Press), CLASP-1, CLASP-2, CLASP-4, KV1.3, and DOCK2.
The C-terminal core sequence of CD3 is SSQL (SEQ ID NO:4). When naturally- occurring residues are added to the core sequence, SSSQL (SEQ ID NO:5), SSSSQL (SEQ ID NO:6), PSSSSQL (SEQ ID NO:7), and PPSSSSQL (SEQ ID NO:8) may also be used to target a PDZ domain-containing protein in T cells. The C-terminal core sequence of CD4 is CSPI (SEQ ID NO:9). When naturally- occurring residues are added to the core sequence, TCSPI (SEQ ID NO: 10), KTCSPI (SEQ ID NO:l l), QKTCSPI (SEQ ID NO:12), and FQKTCSPI (SEQ ID NO:13) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of CD6 is ISAA (SEQ ID NO: 14). When naturally-occurring residues are added to the core sequence, DISAA (SEQ ID NO: 15), DDISAA (SEQ ID NO: 16), YDDISAA (SEQ ID NO: 17), and DYDDISAA (SEQ ID NO: 18) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of CD38 is TSEI (SEQ ID NO: 19). When naturally-occurring residues are added to the core sequence, CTSEI (SEQ ID NO:20), SCTSEI (SEQ ID NO:21), SSCTSEI (SEQ ID NO:22), and DSSCTSEI (SEQ ID NO:23) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of CD49e is TSDA (SEQ ID NO:24). When naturally-occurring residues are added to the core sequence, ATSDA (SEQ ID NO:25), PATSDA (SEQ ID NO:26), PP ATSDA (SEQ ID NO:27), and KPPATSDA (SEQ ID NO:28) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of CD49f is TSDA (SEQ ID NO:29). When naturally-occurring residues are added to the core sequence, LTSDA (SEQ ID NO:30), RLTSDA (SEQ ID NO:31), ERLTSDA (SEQ ID NO:32), and KERLTSDA (SEQ ID NO:33) may also be used to target a PDZ domain-containing protein in T cells. The C-terminal core sequence of CD53 is TIGL (SEQ ID NO:34). When naturally-occurring residues are added to the core sequence, QTIGL (SEQ ID NO:35), SQTIGL (SEQ ID NO:36), TSQTIGL (SEQ ID NO:37), and KTSQTIGL (SEQ ID NO:38) may also be used to target a PDZ domain-containing protein in T cells. The C-terminal core sequence of CD83 is TELV (SEQ. ID. NO: 248). When naturally-occurring residues are added to the core sequence, KTELV (SEQ. ID. NO: 249), HKTELV (SEQ. ID. NO: 250), PHKTELV (SEQ. ID. NO: 251), and TPHKTELV (SEQ. ID. NO: 252) may also be used to target a PDZ domain-containing protein in T cells. The C-terminal core sequence of CD90 is FMSL (SEQ ID NO:39). When naturally-occurring residues are added to the core sequence, DFMSL (SEQ ID NO:40), TDFMSL (SEQ ID NO:41), ATDFMSL (SEQ ID NO:42), and QATDFMSL (SEQ ID NO:43) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of CD95 is QSLV (SEQ ID NO:44). When naturally-occurring residues are added to the core sequence, IQSLV (SEQ ID NO:45), EIQSLV (SEQ ID NO:46), NEIQSLV (SEQ ID NO:47), and RNEIQSLV (SEQ ID NO:48) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of CD97 is ESGI (SEQ ID NO:49). When naturally-occurring residues are added to the core sequence, SESGI (SEQ ID NO:50), ASESGI (SEQ ID NO:51), RASESGI (SEQ ID NO:52), and LRASESGI (SEQ ID NO:53) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of CD98 is PYAA (SEQ ID NO:54). When naturally-occurring residues are added to the core sequence, FPYJAA (SEQ ID NO:55), RFPYAA (SEQ ID NO:56), LRFPYAA (SEQ ID NO:57), and LLRFPYAA (SEQ ID NO:58) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of CDwl37 is GCEL (SEQ ID NO:59). When naturally-occurring residues are added to the core sequence, GGCEL (SEQ ID NO:60), EGGCEL (SEQ ID NO:61), EEGGCEL (SEQ ID NO:62), and EEEGGCEL (SEQ ID NO:63) may also be used to target a PDZ domain-containing protein in T cells. The C-terminal core sequence of CD 166 is KTEA (SEQ ID NO:64). When naturally-occurring residues are added to the core sequence, HKTEA (SEQ ID NO:65), NHKTEA (SEQ ID NO:66), NNHKTEA (SEQ ID NO:67), and ENNHKTEA (SEQ ID NO:68) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of CDwl28 is SSNL (SEQ ID NO:69). When naturally-occurring residues are added to the core sequence, VSSNL (SEQ ID NO:70), NVSSNL (SEQ ID NO:71), VNVSSNL (SEQ ID NO:72), and SVNVSSNL (SEQ ID NO:73) may also be used to target a PDZ domain-containing protein in T cells. The C-terminal core sequence of DNAM-1 is KTRV (SEQ ID NO:74). When naturally-occurring residues are added to the core sequence, PKTRV (SEQ ID NO:75), RPKTRV (SEQ ID NO:76), RRPKTRV (SEQ ID NO:77), and SRRPKTRV (SEQ ID NO:78) may also be used to target a PDZ domain-containing protein in T cells. The C-terminal core sequence of FasL is LYKL (SEQ ID NO:79). When naturally-occurring residues are added to the core sequence, GLYKL (SEQ ID NO:80), FGLYKL (SEQ ID NO:81), FFGLYKL (SEQ ID NO:82), and TFFGLYKL (SEQ ID NO:83) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of LPAP is VTAL (SEQ ID NO:84). When naturally-occurring residues are added to the core sequence, HVTAL (SEQ ID NO:85), LHVTAL (SEQ ID NO:86), GLHVTAL (SEQ ID NO:87), and QGLHVTAL (SEQ ID NO:88) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of CLASP-1 is SAQV (SEQ. ID. NO: 218). When naturally-occurring residues are added to the core sequence, SSAQV (SEQ. ID. NO: 219), SSSAQV (SEQ. ID. NO: 220), ISSSAQV (SEQ. ID. NO: 221), and SISSSAQV (SEQ. ID. NO: 222) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of CLASP-2 is SSVV (SEQ. ID. NO: 223). When naturally-occurring residues are added to the core sequence, SSSVV (SEQ. ID. NO: 224), SSSSVV (SEQ. ID. NO: 225), TSSSSVV (SEQ. ID. NO: 226), and MTSSSS W (SEQ. ID. NO: 227) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of CLASP-4 is YAEV (SEQ. ID. NO: 228). When naturally-occurring residues are added to the core sequence, RYAEV (SEQ. ID. NO: 229), PRYAEV (SEQ. ID. NO: 230), SPRYAEV (SEQ. ID. NO: 231), and GSPRYAEV (SEQ. ID. NO: 232) may also be used to target a PDZ domain-containing protein in T cells. The C-terminal core sequence of KV1.3 is FTDV (SEQ. ID. NO: 238). When naturally-occurring residues are added to the core sequence, IFTDV (SEQ. ID. NO: 239), KIFTDV (SEQ. ID. NO: 240), KKIFTDV (SEQ. ID. NO: 241), and IK IFTDV (SEQ. ID. NO: 242) may also be used to target a PDZ domain-containing protein in T cells.
The C-terminal core sequence of DOCK2 is STDL (SEQ. ID. NO: 243). When naturally-occurring residues are added to the core sequence, LSTDL (SEQ. ID. NO: 244), SLSTDL (SEQ. ID. NO: 245), DSLSTDL (SEQ. ID. NO: 246), and PDSLSTDL (SEQ. ID. NO: 247) may also be used to target a PDZ domain-containing protein in T cells.
6.6.2 PDZ Motif Sequences of B Cell Surface Receptors A number of surface receptors expressed by B cells contain a PDZ domain motif sequence. These molecules include, but are not limited to, CD38, CD53, CD95, CD97, CD98, CDwl37, CD138, CDwl25 (IL5R), DNAM-1 , LPAP, Syndecan-2 (Barclay et al., 1997, The Leucocyte Antigen Facts Book, second edition, Academic Press) and BLR-1. The specific motif sequences of CD38, CD53, CD83, CD95, CD97, CD98, CDwl37, DNAM-1, DOCK2, LPAP, CLASP-1, CLASP-2 and CLASP-4 have been described in the preceding paragraphs.
The C-terminal core sequence of CD138 is EFYA (SEQ ID NO:89). When naturally-occurring residues are added to the core sequence, EEFYA (SEQ ID NO:90), QEEFYA (SEQ ID NO:91), KQEEFYA (SEQ ID NO:92), and TKQEEFYA (SEQ ID NO:93) may also be used to target a PDZ domain-containing protein in B cells.
The C-terminal core sequence of CDwl25 is DSVF (SEQ ID NO:94). When naturally-occurring residues are added to the core sequence, EDSVF (SEQ ID NO:95), LEDSVF (SEQ ID NO:96), TLEDSVF (SEQ ID NO:97), and ETLEDSVF (SEQ ID NO:98) may also be used to target a PDZ domain-containing protein in B cells. The C-terminal core sequence of Syndecan-2 is EFYA (SEQ. ID. NO: 258).
When naturally-occurring residues are added to the core sequence, KEFYA (SEQ. ID. NO: 259), TKEFYA (SEQ. ID. NO: 260), PTKEFYA (SEQ. ID. NO: 261), and APTKEFYA (SEQ. ID. NO: 262) may also be used to target a PDZ domain-containing protein in B cells.
The C-terminal core sequence of BLR-1 is LTTF (SEQ. ID. NO: 253). When naturally-occurring residues are added to the core sequence, SLTTF (SEQ. ID. NO: 254), TSLTTF (SEQ. ID. NO: 255), ATSLTTF (SEQ. ID. NO: 256), and NATSLTTF (SEQ. ID. NO: 257) may also be used to target a PDZ domain-containing protein in B cells.
6.6.3 PDZ motif sequences of Natural Killer Cell Surface Receptors A number of surface receptors expressed by NK cells contain a PDZ domain motif sequence. These molecules include, but are not limited to CD38, CD56, CD98 and DNAM-1. The specific motif sequences of CD38, CD98 and DNAM-1 have been described in the preceding paragraphs.
The C-terminal core sequence of CD56 is ESKA (SEQ ID NO:99). When naturally-occurring residues are added to the core sequence, NESKA (SEQ ID NO: 100), ENESKA (SEQ ID NO: 101 ), KENESKA (SEQ ID NO: 102), and TKENESKA (SEQ ID NO: 103) may also be used to target a PDZ domain-containing protein in NK cells.
6.6.4 PDZ motif sequences of Monocyte Surface Receptors A number of surface receptors expressed by cells of the monocytic lineage (monocytes and macrophages) contain a PDZ domain motif sequence. These molecules include, but are not limited to CD38, CD44, CD46, CD49e, CD49f, CD53, CD61 , CD95, CD97, CD98, CD148, CDwl28, CDwl37, Ly-6, DNAM-1 and FcεRIβ. The specific motif sequences of CD38, CD49e, CD49f, CD53, CD95, CD97, CD98, CDwl28, CDwl37, DNAM-1, Galectin 3 (Mac-2) and Mannose receptor have been described in the preceding paragraphs.
The C-terminal core sequence of CD44 is KIGV (SEQ ID NO: 104). When naturally-occurring residues are added to the core sequence, MKIGV (SEQ ID NO: 105), DMKIGV (SEQ ID NO: 106), VDMKIGV (SEQ ID NO: 107) and NYDMKIGV (SEQ ID NO: 108) may also be used to target a PDZ domain-containing protein in monocytes.
The C-terminal core sequence of CD46 is FTSL (SEQ ID NO: 109). When naturally-occurring residues are added to the core sequence, KFTSL (SEQ ID NO: 110), VKFTSL (SEQ ID NOT 11), EVKFTSL (SEQ ID NO: 112) and REVKFTSL (SEQ ID NOT 13) may also be used to target a PDZ domain-containing protein in monocytes. The C-terminal core sequence of CD61 is KSLV (SEQ ID NOT 14). When naturally-occurring residues are added to the core sequence, LKSLV (SEQ ID NO: 115), FLKSLV (SEQ ID NO: 116), RFLKSLV (SEQ ID NO: 117) and GRFLKSLV (SEQ ID NOT 18) may also be used to target a PDZ domain-containing protein in monocytes.
The C-terminal core sequence of CD148 is GYIA (SEQ ID NOT 19). When naturally-occurring residues are added to the core sequence, NGYIA (SEQ ID NO: 120), TNGYIA (SEQ ID NO: 121), KTNGYIA (SEQ ID NO: 122) and GKTNGYIA (SEQ ID NO: 123) may also be used to target a PDZ domain-containing protein in monocytes.
The C-terminal core sequence of Ly-6 is QTLL (SEQ ID NO: 124). When naturally-occurring residues are added to the core sequence, LQTLL (SEQ ID NO: 125), LLQTLL (SEQ ID NOT26), VLLQTLL (SEQ ID NOT27) and SNLLQTLL (SEQ ID ΝOT28) may also be used to target a PDZ domain-containing protein in monocytes.
The C-terminal core sequence of FcεRIβ is PIDL (SEQ ID NO: 129). When naturally-occurring residues are added to the core sequence, PPIDL (SEQ ID NO: 130), SPPIDL (SEQ ID NO: 131), MSPPIDL (SEQ ID NO: 132) and EMSPPIDL (SEQ ID NO: 133) may also be used to target a PDZ domain-containing protein in monocytes.
The C-terminal core sequence of Galectin 3 is YTMI (SEQ ID NO: 134). When naturally-occurring residues are added to the core sequence, SYTMI (SEQ ID NO: 135), ASYTMI (SEQ ID NO: 136), SASYTMI (SEQ ID NO:137) and TSASYTMI (SEQ ID NOT 38) may also be used to target a PDZ domain-containing protein in monocytes.
The C-terminal core sequence of mannose receptor is HSVI (SEQ ID NO: 139). When naturally-occurring residues are added to the core sequence, EHSVI (SEQ ID NO: 140), NEHSVI (SEQ ID NO: 141), QNEHSVI (SEQ ID NOT 42) and EQNEHSVI (SEQ ID NO: 143) may also be used to target a PDZ domain-containing protein in monocytes.
6.6.5 PDZ motif sequences of Granulocvte Surface Receptors
A number of surface receptors expressed by granulocytes contain a PDZ domain motif sequence. These molecules include, but are not limited to CD53, CD95, CD97, CD98, CD148, CDwl25, CDwl28, FcεRIβ and G-CSFR. The specific motif sequences of most of these molecules have been described in the preceding paragraphs.
The C-terminal core sequence of G-CSFR is TSVL (SEQ ID NOT44). When naturally-occurring residues are added to the core sequence, ITSVL (SEQ ID NOT 45), PITSVL (SEQ ID NOT46), FPITSVL (SEQ ID NOT47) and LFPITSVL (SEQ ID NO.T48) may also be used to target a PDZ domain-containing protein in monocytes.
6.6.6 PDZ motif sequences of Endothelial Cell Surface Receptors
While endothelial cells are not hematopoietic cells, they closely interact with the hematopoietic system as they form the lining of blood vessels. As such, endothelial cells come in contact with the cells ofthe hematopoietic system. Thus, the ability to regulate endothelial cell function provides for indirect regulation of hematopoietic cells.
A number of surface receptors expressed by endothelial cells contain a PDZ domain motif sequence. These molecules include, but are not limited to CD34, CD46, CD66b, CD66c, CD 105, CD106, CD62e (E-selectin) and VCAMl . The C-terminal core sequence of CD34 is DTEL (SEQ ID NO: 149). When naturally-occurring residues are added to the core sequence, ADTEL (SEQ ID NO: 150), VADTEL (SEQ ID NO: 151), VVADTEL (SEQ ID NO: 152) and HVVADTEL (SEQ ID NO: 153) may also be used to target a PDZ domain-containing protein in endothelial cells.
The C-terminal core sequence of CD66b and CD66c is VALI (SEQ ID NOT 54). When naturally-occurring residues are added to the core sequence, RVALI (SEQ ID NO: 155), ARVALI (SEQ ID NO: 156), LARVALI (SEQ ID NO: 157) and VLARVALI (SEQ ID NO: 158) may also be used to target a PDZ domain-containing protein in endothelial cells. The C-terminal core sequence of CD 105 is SSMA (SEQ ID NO: 159). When naturally-occurring residues are added to the core sequence, TSSMA (SEQ ID NO: 160), STSSMA (SEQ ID NOT 61), CSTSSMA (SEQ ID NOT 62) and PCSTSSMA (SEQ ID NO: 162) may also be used to target a PDZ domain-containing protein in endothelial cells. The C-terminal core sequence of CD 106 is KSKV (SEQ ID NO: 163). When naturally-occurring residues are added to the core sequence, QKSKV (SEQ ID NO: 164), AQKSKV (SEQ ID NO: 165), EAQKSKV (SEQ ID NO: 166) and VEAQKSKV (SEQ ID NO: 167) may also be used to target a PDZ domain-containing protein in endothelial cells.
The C-terminal core sequence of CD62e is SYIL (SEQ ID NO: 168). When naturally-occurring residues are added to the core sequence, PSYIL (SEQ ID NOT 69), KPSYIL (SEQ ID NOT 70), QKPSYIL (SEQ ID NO: 171) and YQKPSYIL (SEQ ID NOT 72) may also be used to target a PDZ domain-containing protein in endothelial cells.
The C-terminal core sequence of VCAMl is KSKV (SEQ. ID. NO: 233). When naturally-occurring residues are added to the core sequence, QKSKV (SEQ. ID. NO: 234), AQKSKV (SEQ. ID. NO: 235), EAQKSKV (SEQ. ID. NO: 236), and VEAQKSKV (SEQ. ID. NO: 237) may also be used to target a PDZ domain-containing protein in endothelial cells.
6.6.7 Mast Cell, Basophils and Eosinophil Cell Surface Receptors
FcεRIβ, CDwl25, CDwl28 and IL-8RB are transmembrane receptors expressed by mast cells, basophils and eosinophils. These receptors play a role in the activation of these cells to result in degranulation and histamine release in allergic reactions. The C-terminal core sequence of FcεRIβ is PIDL (SEQ ID NO:4). When naturally-occurring residues are added to the core sequence, PPIDL (SEQ ID NO:5), SPPIDL (SEQ ID NO:6), MSPPIDL (SEQ ID NO:7) and EMSPPIDL (SEQ ID NO:8) may also be used to target a PDZ domain-containing protein in mast cells. In addition, the residue E may be substituted with G to increase its binding affinity.
The C-terminal core sequence of CDwl25 is DSVF (SEQ ID NO: 9). When naturally-occurring residues are added to the core sequence, EDSVF (SEQ ID NO: 10), LEDSVF
(SEQ ID NOT 1), TLEDSVF (SEQ ID NO: 12), and ETLEDSVF (SEQ ID NOT 3) may also be used to target a PDZ domain-containing protein in mast cells. The C-terminal core sequence of CDwl28 is SSNL (SEQ ID NOT4). When naturally-occurring residues are added to the core sequence, VSSNL (SEQ ID NO: 15), NNSSNL (SEQ ID NO: 16), VNVSSNL (SEQ ID NO: 17), and SVNVSSNL (SEQ ID NO: 18) may also be used to target a PDZ domain-containing protein in mast cells. The C-terminal core sequence of IL-8RB is STTL (SEQ ID NO: 19). When naturally-occurring residues are added to the core sequence TSTTL (SEQ ID NO:20), HTSTTL (SEQ ID NO:21), GHTSTTL (SEQ ID NO:22) and SGHTSTTL (SEQ ID NO:23) may also be used to target a PDZ domain-containing protein in mast cells.
6.6.8 Other PDZ motif sequences
The C-terminal core sequence of NMD A is ESDV (SEQ. ID. NO: 263). When naturally-occurring residues are added to the core sequence, IESDV (SEQ. ID. NO: 264), SIESDV (SEQ. ID. NO: 265), PSIESDV (SEQ. ID. NO: 266), and MPSIESDV (SEQ. ID. NO: 267) may also be used to target a PDZ domain-containing protein in neuronal cells. The C-terminal core sequence of neurexin is EYYV (SEQ. ID. NO: 268). When naturally-occurring residues are added to the core sequence, KEYYV (SEQ. ID. NO: 269), DKEYYV (SEQ. ID. NO: 270), KDKEYYV (SEQ. ID. NO: 271), and NKDKEYYV (SEQ. ID. NO: 272) may also be used to target a PDZ domain-containing protein in neuronal cells.
The C-terminal core sequence of Glycophorin C is EYFI (SEQ. ID. NO: 273). When naturally-occurring residues are added to the core sequence, KEYFI (SEQ. ID. NO: 274), RKEYFI (SEQ. ID. NO: 275), SRKEYFI (SEQ. ID. NO: 276), and SSRKEYFI (SEQ. ID. NO: 277) may also be used to target a PDZ domain-containing protein.
The C-terminal core sequence of CD148 is KTIA (SEQ. ID. NO: 278). When naturally-occurring residues are added to the core sequence, GKTIA (SEQ. ID. NO: 279), FGKTIA (SEQ. ID. NO: 280), TFGKTIA (SEQ. ID. NO: 281), and TTFGKTIA (SEQ. ID. NO:
282) may also be used to target a PDZ domain-containing protein in epithelial or myeloid cells.
6.7. Preparation of Peptides
6.7.1. Chemical Synthesis The peptides of the invention or analogues thereof, may be prepared using virtually any art-known technique for the preparation of peptides and peptide analogues. For example, the peptides may be prepared in linear form using conventional solution or solid phase peptide syntheses and cleaved from the resin followed by purification procedures (Creighton, 1983, Protein Structures And Molecular Principles, W.H. Freeman and Co., N.Y.). Suitable procedures for synthesizing the peptides described herein are well known in the art. The composition of the synthetic peptides may be confirmed by amino acid analysis or sequencing (e.g., the Edman degradation procedure and mass spectroscopy).
In addition, analogues and derivatives of the peptides can be chemically synthesized. The linkage between each amino acid of the peptides of the invention may be an amide, a substituted amide or an isostere of amide. Nonclassical amino acids or chemical amino acid analogues can be introduced as a substitution or addition into the sequence. Non-classical amino acids include, but are not limited to, the D-isomers ofthe common amino acids, α-amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, γ-Abu, ε-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, β-alanine, fluoro-amino acids, designer amino acids such as β-methyl amino acids, Cα-methyl amino acids, Nα-methyl amino acids, and amino acid analogues in general. Furthermore, the amino acid can be D (dextrorotary) or L (levorotary).
6.7.2. Recombinant Synthesis If the peptide is composed entirely of gene-encoded amino acids, or a portion of it is so composed, the peptide or the relevant portion may also be synthesized using conventional recombinant genetic engineering techniques. For recombinant production, a polynucleotide sequence encoding a linear form of the peptide is inserted into an appropriate expression vehicle, i.e., a vector which contains the necessary elements for the transcription and translation of the inserted coding sequence, or in the case of an RNA viral vector, the necessary elements for replication and translation. The expression vehicle is then transfected into a suitable target cell which will express the peptide. Depending on the expression system used, the expressed peptide is then isolated by procedures well-established in the art. Methods for recombinant protein and peptide production are well known in the art (see, e.g., Maniatis et al, 1989, Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y.; and Ausubel et al., 1989, Current
Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, N.Y.).
A variety of host-expression vector systems may be utilized to express the peptides described herein. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage DNA or plasmid DNA expression vectors containing an appropriate coding sequence; yeast or filamentous fungi transformed with recombinant yeast or fungi expression vectors containing an appropriate coding sequence; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing an appropriate coding sequence; plant cell systems infected with recombinant vims expression vectors (e.g., cauliflower mosaic virus or tobacco mosaic virus) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing an appropriate coding sequence; or animal cell systems.
The expression elements of the expression systems vary in their strength and specificities. Depending on the host/vector system utilized, any of a number of suitable transcription and translation elements, including constitutive and inducible promoters, may be used in the expression vector. For example, when cloning in bacterial systems, inducible promoters such as pL of bacteriophage λ, plac, ptrp, ptac (ptrp-lac hybrid promoter) and the like may be used; when cloning in insect cell systems, promoters such as the baculovirus polyhedron promoter may be used; when cloning in plant cell systems, promoters derived from the genome of plant cells (e.g., heat shock promoters; the promoter for the small subunit of RUBISCO; the promoter for the chlorophyll a/b binding protein) or from plant viruses (e.g., the 35S RNA promoter of CaMV; the coat protein promoter of TMV) may be used; when cloning in mammalian cell systems, promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5 K promoter) may be used; when generating cell lines that contain multiple copies of expression product, SV40-, BPV- and EBV-based vectors may be used with an appropriate selectable marker.
In cases where plant expression vectors are used, the expression of sequences encoding the peptides of the invention may be driven by any of a number of promoters. For example, viral promoters such as the 35S RNA and 19S RNA promoters of CaMV (Brisson et al., 1984, Nature 310:511-514), or the coat protein promoter of TMV (Takamatsu et al., 1987, EMBO J. 6:307-311) may be used; alternatively, plant promoters such as the small subunit of RUBISCO (Coruzzi et al, 1984, EMBO J. 3: 1671-1680; Broglie et al, 1984, Science 224:838- 843) or heat shock promoters, e.g., soybean hspl7.5-E or hspl7.3-B (Gurley et al, 1986, Mol. Cell. Biol. 6:559-565) may be used. These constructs can be introduced into planleukocytes using Ti plasmids, Ri plasmids, plant virus vectors, direct DNA transformation, microinjeotion, electroporation, etc. For reviews of such techniques see, e.g., Weissbach & Weissbach, 1988, Methods for Plant Molecular Biology, Academic Press, NY, Section VIII, pp. 421-463; and Grierson & Corey, 1988, Plant Molecular Biology, 2d Ed., Blackie, London, Ch. 7-9.
In one insect expression system that may be used to produce the peptides of the invention, Autographa californica nuclear polyhidrosis virus (AcNPV) is used as a vector to express the foreign genes. The virus grows in Spodoptera frugiperda cells. A coding sequence may be cloned into non-essential regions (for example the polyhedron gene) of the virus and placed under control of an AcNPV promoter (for example, the polyhedron promoter). Successful insertion of a coding sequence will result in inactivation ofthe polyhedron gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat coded for by the polyhedron gene). These recombinant viruses are then used to infect Spodoptera frugiperda cells in which the inserted gene is expressed, (e.g., see Smith et al., 1983, J. Virol. 46:584; Smith, U.S. Patent No. 4,215,051). Further examples of this expression system may be found in Current Protocols in Molecular Biology, Vol. 2, Ausubel et al, eds., Greene Publish. Assoc. & Wiley Interscience. In mammalian host cells, a number of viral based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, a coding sequence may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region ofthe viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing peptide in infected hosts, (e.g., See Logan & Shenk, 1984, Proc. Natl. Acad. Sci. USA 81:3655- 3659). Alternatively, the vaccinia 7.5 K promoter may be used, (see, e.g., Mackett et al, 1982, Proc. Natl. Acad. Sci. USA 79:7415-7419; Mackett et al, 1984, J. Virol. 49:857-864; Panicali et al, 1982, Proc. Natl. Acad. Sci. USA 79:4927-4931). Other expression systems for producing linear peptides ofthe invention will be apparent to those having skill in the art.
6.7.3. Purification of the Peptides and Peptide Analogues
The peptides and peptide analogues ofthe invention can be purified by art-known techniques such as high performance liquid chromatography, ion exchange chromatography, gel electrophoresis, affinity chromatography and the like. The actual conditions used to purify a particular peptide or analogue will depend, in part, on factors such as net charge, hydrophobicity, hydrophilicity, etc., and will be apparent to those having skill in the art. The purified peptides can be identified by assays based on their physical or functional properties, including radioactive labeling followed by gel electrophoresis, radioimmuno-assays, ELISA, bioassays, and the like. For affinity chromatography purification, any antibody which specifically binds the peptides or peptide analogues may be used. For the production of antibodies, various host animals, including but not limited to rabbits, mice, rats, etc., may be immunized by injection with a peptide. The peptide may be attached to a suitable carrier, such as BSA or KLH, by means of a side chain functional group or linkers attached to a side chain functional group. Various adjuvants may be used to increase the immunological response, depending on the host species, including but not limited to Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, dinitrophenol, and potentially useful human adjuvants such as BCG (bacilli Calmette-Guerin) and Corynebacterium parvum.
Monoclonal antibodies to a peptide may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include but are not limited to the hybridoma technique originally described by Koehler and Milstein, 1975, Nature 256:495-497, the human B-cell hybridoma technique, Kosbor et al, 1983, Immunology Today 4:72; Cote et al, 1983, Proc. Natl. Acad. Sci. U.S.A. 80:2026-2030 and the EBV-hybridoma technique (Cole et al, 1985, Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96 (1985)). In addition, techniques developed for the production of "chimeric antibodies" (Morrison et al, 1984, Proc. Natl. Acad. Sci. U.S.A. 81 :6851-6855; Neuberger et al, 1984, Nature 312:604-608; Takeda et al., 1985, Nature 314:452-454) by splicing the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used. Alternatively, techniques described for the production of single chain antibodies (U.S. Patent No. 4,946,778) can be adapted to produce peptide-specific single chain antibodies.
Antibody fragments which contain deletions of specific binding sites may be generated by known techniques. For example, such fragments include but are not limited to F(ab')2 fragments, which can be produced by pepsin digestion ofthe antibody molecule and Fab fragments, which can be generated by reducing the disulfide bridges of the F(ab')2 fragments. Alternatively, Fab expression libraries may be constructed (Huse et al, 1989, Science 246:1275- 1281) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity for the peptide of interest.
The antibody or antibody fragment specific for the desired peptide can be attached, for example, to agarose, and the antibody-agarose complex is used in immunochromatography to purify peptides of the invention. See, Scopes, 1984, Protein
Purification: Principles and Practice, Springer-Verlag New York, Inc., NY, Livingstone, 1974,
Methods Enzymology: Immunoaffinity Chromatography of Proteins 34:723-731.
6.8. Uses of PDZ Domain Binding and Antagonist Compounds
In one aspect ofthe invention, the PDZ domain binding and PDZ-PL inhibitory compounds ofthe present invention are useful in regulating diverse activities of hematopoietic cells (e.g., T cells and B cells) and other cells. involved in the immune response.
In one embodiment ofthe invention, the compounds ofthe invention are used to inhibit leukocyte activation, which is manifested in measurable events including but not limited to, cytokine production, cell adhesion, expansion of cell numbers, apoptosis and cytotoxicity. As a corollary, the compounds of the invention may be used to treat diverse conditions associated with undesirable leukocyte activation, including but not limited to, acute and chronic inflammation, graft-versus-host disease, transplantation rejection, hypersensitivities and autoimmunity such as multiple sclerosis, rheumatoid arthritis, peridontal disease, systemic lupus erythematosis, juvenile diabetes mellitis, non-insulin-dependent diabetes, and allergies, and other conditions listed herein (see, e.g., Section 6.4, supra).
Thus, the invention also relates to methods of using such compositions in modulating leukocyte activation as measured by, for example, cytotoxicity, cytokine production, cell proliferation, and apoptosis. Assays for activation are well known. For example, PDZ/PL interaction antagonists can be evaluated in the following: (1) cytotoxic T lymphocytes can be incubated with radioactively labeled target cells and the antigen-specific lysis of these target cells detected by the release of radioactivity, (2) helper T lymphocytes can be incubated with antigens and antigen presenting cells and the synthesis and secretion of cytokines measured by standard methods (Windhagen A; et al., 1995, Immunity 2(4): 373-80), (3) antigen presenting cells can be incubated with whole protein antigen and the presentation of that antigen on MHC detected by either T lymphocyte activation assays or biophysical methods (Harding et al., 1989, Proc. Natl. Acad. Sci., 86: 4230-4), (4) mast cells can be incubated with reagents that cross-link their Fc-epsilon receptors and histamine release measured by enzyme immunoassay (Siraganian, et al., 1983, TIPS 4: 432-437).
Similarly, the effect of PDZ/PL interaction antagonists on products of leukocyte activation in either a model organism (e.g., mouse) or a human patient can also be evaluated by various methods that are well known. For example, (1) the production of antibodies in response to vaccination can be readily detected by standard methods currently used in clinical laboratories, e.g., an ELISA; (2) the migration of immune cells to sites of inflammation can be detected by scratching the surface of skin and placing a sterile container to capture the migrating cells over scratch site (Peters et al., 1988, Blood 72: 1310-5); (3) the proliferation of peripheral blood mononuclear cells in response to mitogens or mixed lymphocyte reaction can be measured using 3H-thymidine; (4) the phagocytic capacity of granulocytes, macrophages, and other phagocytes in PBMCs can be measured by placing PMBCs in wells together with labeled particles (Peters et al., 1988); and (5) the differentiation of immune system cells can be measured by labeling PBMCs with antibodies to CD molecules such as CD4 and CD8 and measuring the fraction of the PBMCs expressing these markers.
In one exemplary assay, human peripheral blood mononuclear cells (PBMC), human T cell clones (e.g., Jurkat E6, ATCC TIB- 152), EBV-transformed B cell clones (e.g., 9D10, ATCC CRL-8752), antigen-specific T cell clones or lines can be used to examine PDZ/PL interaction antagonists in vitro. Inhibition of activation of these cells or cell lines can be used for the evaluation of potential PDZ/PL interaction antagonists.
Standard methods by which hematopoietic cells are stimulated to undergo activation characteristic of an immune response are, for example:
A) Antigen specific stimulation of immune responses. Either pre-immunized or naϊve mouse splenocytes can be generated by standard procedures. In addition, antigen-specific T cell clones and hybridomas (e.g., MBP-specific), and numerous B cell lymphoma cell lines (e.g., CH27), have been previously characterized and are available for the assays discussed below. Antigen specific splenocytes or B-cells can be mixed with antigen specific T-cells in the presence of antigen to generate an immune response. This can be performed in the presence or absence of PDZ/PL interaction antagonists to assay whether PDZ/PL interaction antagonists modulate the immune response infra. B) Non-specific T cell activation. The following methods can be used to activate T cells in the absence of antigen: 1) cross-linking T cell receptor (TCR) by addition of antibodies against receptor activation molecules (e.g., TCR, CD3, or CD2) together with antibodies against co-stimulator molecules, for example anti-CD28; 2) activating cell surface receptors in a non- specific fashion using lectins such as concanavalin A (con A) and phytohemagglutinin (PHA); 3) mimicking cell surface receptor-mediated activation using pharmacological agents that activate protein kinase C (e.g., phorbol esters) and increase cytoplasmic Ca24" (e.g., ionomycin).
C) Non-specific B cell activation: 1) application of antibodies against cell surface molecules such as IgM, CD20, or CD21. 2) Lipopolysaccharide (LPS), phorbol esters, calcium ionophores and ionomycin can also be used to by-pass receptor triggering.
D) Mixed lymphocyte reaction (MLR). Mix donor PBMC with recipient PBMC to activate lymphocytes by presentation of mismatched tissue antigens, which occurs in all cases except identical twins.
E) Generation of a specific T cell clone or line that recognizes a particular antigen. A standard approach is to generate tetanus toxin-specific T cells from a donor that has recently been boosted with tetanus toxin. Major histocompatability complex- (MHC-) matched antigen presenting cells and a source of tetanus toxin are used to maintain antigen specificity of the cell line or T cell clone (Lanzavecchia, A., et al, 1983, Eur. J. Immun. 13: 733-738). Assay Quantitation The assays described above can be quantitated by a variety of well known quantitation methods. For example:
(A) Tyrosine phosphorylation
Tyrosine phosphorylation of early response proteins such as HS1, PLC-r, ZAP- 76, and Vav is an early biochemical event following leukocyte activation. The tyrosine phosphorylated proteins can be detected by Western blot using antibodies against phosphorylated tyrosine residues. Tyrosine phosphorylation of these early response proteins can be used as a standard assay for leukocyte activation (J. Biol. Chem., 1997, 272(23): 14562-14570). Any change in the phosphorylation pattern of these or related proteins when immune responses are generated in the presence of potential PDZ/PL interaction antagonists is indicative of a potential PDZ/PL interaction antagonists.
(B) Intracellular Calcium Flux The kinetics of intracellular Ca2+ concentrations are measured over time after stimulation of cells preloaded with a calcium sensitive dye. Upon binding Ca21' the indicator dye (e.g., Fluor-4 (Molecular Probes)), exhibits an increase in fluorescence level using flow cytometry, solution fluorometry, and confocal microscopy. Any change in the level or timing of calcium flux when immune responses are generated in the presence of PDZ/PL interaction antagonists is indicative of an inhibition of this response.
(C) Regulation of early activation markers
Increased and diminished expression/regulation of early lymphocyte activation marker levels such as CD69, IL-2R, MHC class II, B7, and TCR are commonly measured with fluorescently labeled antibodies using flow cytometry. All antibodies are commercially available. Any change in the expression levels of lymphocyte activation markers when immune responses are generated in the presence ofthe PDZ/PL interaction antagonists is indicative of an inhibition of this response.
(D) Increased metabolic activity/acid release Activation of most known signal transduction pathways trigger increases in acidic metabolites. This reproducible biological event is measured as the rate of acid release using a microphysiometer (Molecular Devices), and is used as an early activation marker when comparing the treatment of cells with potential biological therapeutics (McConnell, H.M. et al., 1992, Science 257: 1906-1912 and McConnell, H.M., 1995, Proc. Natl. Acad. Sci. 92: 2750- 2754). Any statistically significant increase or decrease in acid release ofthe PDZ PL interaction antagonist-treated sample, as compared to control sample (no treatment), suggest an effect ofthe PDZ/PL interaction antagonist on biological function.
(E) Cell proliferation/cell viability assays (1) 3H-thimidine incorporation Exposure of lymphocytes to antigen or mitogen in vitro induces DNA synthesis and cellular proliferation. The measurement of mitotic activity by 3H-fhimidine incorporation into newly synthesized DNA is one of the most frequently used assays to quantitative T cell activation. Depending on the cell population and form of stimulation used to activate the T cells, mitotic activity can be measured within 24-72 hrs. in vitro, post 3H-thimidine pulse (Mishell, B. B. and S. M. Shiigi, 1980, Selected Methods in Cellular Immunology, W. H. Freeman and Company and Dutton, R. W. and Pearce, J. D., 1962, Nature 194: 93). Any statistically significant increase or decrease in CPM of the PDZ/PL interaction antagonist-treated sample, as compared to control sample (no treatment), suggest and effect of the PDZ/PL interaction antagonist on biological function.
(2) MTS [5-(3-carboxymethoxyphenyl)-2-(4,5-dimethylthiazolyl)-3(4- sulfo ^henyl)tetrazolium, inner salt] is a colorimetric method for determining the number of viable cells in proliferation or cytotoxicity assays (Barltrop, J.A. et al., 1991, Bioorg. & Med. Chem. Lett. 1 : 611). 1-5 days after lymphocyte activation, MTS tetrazolium compound, Owen's reagent, is bioreduced by cells into a colored formazan product that is soluble in tissue culture media. Color intensity is read at 490 nm minus 650 nm using a microplate reader. Any statistically significant increase or decrease in color intensity of the PDZ/PL interaction antagonist-treated sample, as compared to control sample (no treatment), can suggest an effect ofthe PDZ/PL interaction antagonist on biological function (Mosmann, T., 1983, J. Immunol. Methods 65: 55 and Barltrop, J.A. et al. (1991)).
(3) Bromodeoxyuridine (BrdU), a thymidine analogue, readily incorporates into cells undergoing DNA synthesis. BrdU-pulsed cells are labeled with an enzyme-conjugated anti-BrdU antibody (Gratzner, H.G., 1982, Science 218: 474-475.). A colorimetric, soluble substrate is used to visualize proliferating cells that have incorporated BrdU. Reaction is stopped with sulfuric acid and plate is read at 450 nm using a microplate reader. Any statistically significant increase or decrease in color intensity of the PDZ/PL interaction antagonist-treated sample, as compared to control sample (no treatment), suggest an effect of the PDZ/PL interaction antagonist on biological function. (F) Apoptosis by Annexin V
Programmed cell death or apoptosis is an early event in a cascade of catabolic reactions leading to cell death. A lose in the integrity of the cell membrane allows for the binding of fluorescently conjugated phosphatidylserine. Stained cells can be measured by fluorescence microscopy and flow cytometry (Vermes, I., 1995, J. Immunol. Methods. 180: 39- 52). In one embodiment, any statistically significant increase or decrease in apoptotic cell number ofthe PDZ/PL interaction antagonist-treated sample, as compared to control sample (no treatment), suggest an effect of the PDZ/PL interaction antagonist on biological function. For evaluating apoptosis in situ, assays for evaluating cell death in tissue samples can also be used in vivo studies. (G) Quantitation of cytokine production
Cell supematants harvested after cell stimulation for 16-48 hrs are stored at -80°C until assayed or directly tested for cytokine production. Multiple cytokine assays can be performed on each sample. IL-2, IL-3, IFN-γ and other cytokine ELISA Assays are available for mouse, rat, and human (Endogen, Inc. and BioSource). Cytokine production is measured using a standard two-antibody sandwich ELISA protocol as described by the manufacturer. The presence of horseradish peroxidase is detected with 3, 3'5, 5' tertamethyl benziidine (TMB) substrate and the reaction is stopped with sulfuric acid. The absorbency at 450 nm is measured using a microplate reader. Any statistically significant increase or decrease in color intensity of the PDZ/PL interaction antagonist-treated sample, as compared to control sample (no treatment), suggest an effect ofthe PDZ/PL interaction antagonist on biological function. See also Example 1, infra. Detection of intracellular cytokines using anti-cytokine antibodies provides the additional advantage of measuring cytokines fore mixed cell populations. This allows for phenotyping measuring frequency of cytokine producing cell types in suspension or in tissues. (H) NF-AT can be visualized by Immunostaining
T cell activation requires the import of nuclear factor of activated T cells (NF- AT) to the nucleus. This translocation of NF-AT can be visualized by immunostaining with anti- NF-AT antibody (Cell 1998, 93: 851-861). Therefore, NF-AT nuclear translocation has been used to assay T cell activation. Similarly, NF-AT/luciferase reporter assays have been used as a standard measurement of T cell activation (MCB 1996, 12: 7151-7160). Any statistically significant increase or decrease in the nuclear translocation of NF-AT brought about by the PDZ/PL interaction antagonist-treated sample, as compared to control sample (no treatment), suggest an effect of the PDZ/PL interaction antagonist on biological function. In order to optimize the use of the peptides and peptide analogues disclosed herein in a human subject, various animal models may be used to define certain clinical parameters. For example, the compounds of the invention may be tested in different dosages, formulations and route of administration in a cardiac transplant mouse model to optimize their ability to inhibit rejection responses to solid organ transplants (Fulmer et al., 1963, Am. J. Anat. 1 13:273; Jockusch et al., 1983, Exp. Neurol. 81 :749). In situations where inhibition of a T cell response is desired, the compounds of the inventions may be used to inhibit PDZ domain interactions with CD3, CD4, CD6 and CDwl37. In addition, the compounds of the invention may be used to inhibit PDZ domain interactions with CD53 and CD138 in B cells. In order to inhibit IgE-mediated allergic reactions, the compounds of the invention may be used to inhibit PDZ domain interactions with FcεRIβ, CDwl25 and CDwl28. Furthermore, a PDZ motif sequence (PL sequence) of CD95 may be used to induce apoptosis of lymphomas.
(I) Inflammatory Mediator Release Assays
Assays are well known in the art for inflammatory mediator release to access the effect of compounds or treatments IgE-mediated degranulation. See, e.g. Berger et al., 1997, Measuring Cell Degranulation e.g., Ch 19.6 Immunology Method Manual. Academic Press, Ltd.
1436-1440 and Siraganian, 1983, Histamine Secretion from Mast Cells and Basophil. TIPS
4:432-437, both incorporated by reference herein.
6.9. Formulation and Route of Administration
6.9.1 Introduction of Agonists or Antagonists (e.g.. Peptides and Fusion Proteins) into
Cells
In one aspect, the PDZ-PL antagonists ofthe invention are introduced into a cell to modulate (i.e., increase or decrease) a biological function or activity ofthe cell. Many small organic molecules readily cross the cell membranes (or can be modified by one of skill using routine methods to increase the ability of compounds to enter cells, e.g., by reducing or eliminating charge, increasing lipophilicity, conjugating the molecule to a moiety targeting a cell surface receptor such that after interacting with the receptor). Methods for introducing larger molecules, e.g., peptides and fusion proteins are also well known, including, e.g., injection, liposome-mediated fusion, application of a hydrogel, conjugation to a targeting moiety conjugate endocytozed by the cell, electroporation, and the like).
In one embodiment, the antagonist or agent is a fusion polypeptide or derivatized polypeptide. A fusion or derivatized protein may include a targeting moiety that increases the ability ofthe polypeptide to traverse a cell membrane or causes the polypeptide to be delivered to a specified cell type (e.g., liver cells or tumor cells) preferentially or cell compartment (e.g., nuclear compartment) preferentially. Examples of targeting moieties include lipid tails, amino acid sequences such as antennapoedia peptide or a nuclear localization signal (NLS; e.g., Xenopus nucleoplasmin Robbins et al., 1991, Cell 64:615).
In one embodiment of the invention, a peptide sequence or peptide analog determined to inhibit a PDZ domain-PL protein binding, in an assay of the invention is introduced into a cell by linking the sequence to an amino acid sequence that facilitates its transport through the plasma membrane (a "transmembrane transporter sequence"). The peptides of the invention may be used directly or fused to a transmembrane transporter sequence to facilitate their entry into cells. In the case of such a fusion peptide, each peptide may be fused with a heterologous peptide at its amino terminus directly or by using a flexible polylinker such as the pentamer G-G-G-G-S (SEQ ID NOT) repeated 1 to 3 times. Such linker has been used in constructing single chain antibodies (scFv) by being inserted between VH and VL (Bird et al., 1988, Science 242:423-426; Huston et al., 1988, Proc. Natl. Acad. Sci. U.S.A. 85:5979-5883). The linker is designed to enable the correct interaction between two beta-sheets forming the variable region ofthe single chain antibody. Other linkers which may be used include Glu-Gly- Lys-Ser-Ser-Gly-Ser-Gly-Ser-Glu-Ser-Lys-Val-Asp (SEQ ID NO:2) (Chaudhary et al., 1990,
Proc. Natl. Acad. Sci. U.S.A. 87:1066-1070) and Lys-Glu-Ser-Gly-Ser-Val-Ser-Ser-Glu-Gln-
Leu-Ala-Gln-Phe-Arg-Ser-Leu-Asp (SEQ ID NO:3) (Bird et al., 1988, Science 242:423-426).
A number of peptide sequences have been described in the art as capable of facilitating the entry of a peptide linked to these sequences into a cell through the plasma membrane (Derossi et al., 1998, Trends in Cell Biol. 8:84). For the purpose of this invention, such peptides are collectively referred to as transmembrane transporter peptides. Examples of these peptide include, but are not limited to, tat derived from HIV (Vives et al, 1997, J. Biol. Chem. 272:16010; Nagahara et al, 1998, Nat. Med. 4:1449), antennapedia from Drosophila (Derossi et al., 1994, J Biol. Chem. 261 :10444), VP22 from herpes simplex virus (Elliot and D'Hare, 1997, Cell 88:223-233), complementarity-determining regions (CDR) 2 and 3 of anti- DΝA antibodies (Avrameas et al., 1998, Proc. Natl Acad. Sci. U.S.A., 95:5601-5606), 70 KDa heat shock protein (Fujihara, 1999, EMBO J. 18:411-419) and transportan (Pooga et al., 1998, FASEB J. 12:67-77). In a preferred embodiment of the invention, a truncated HIV tat peptide having the sequence of GYGRKKRRQRRRG (SEQ ID NO: 173) is used. It is preferred that a transmembrane transporter sequence is fused to a hematopoietic cell surface receptor carboxyl terminal sequence at its amino-terminus with or without a linker. Generally, the C-terminus of a PDZ motif sequence (PL sequence) must be free in order to interact with a PDZ domain. The transmembrane transporter sequence may be used in whole or in part as long as it is capable of facilitating entry ofthe peptide into a cell.
In an alternate embodiment ofthe invention, a hematopoietic cell surface receptor C-terminal sequence may be used alone when it is delivered in a manner that allows its entry into cells in the absence of a transmembrane transporter sequence. For example, the peptide may be delivered in a liposome formulation or using a gene therapy approach by delivering a coding sequence for the PDZ motif alone or as a fusion molecule into a target cell.
The compounds ofthe ofthe invention may also be administered via liposomes, which serve to target the conjugates to a particular tissue, such as lymphoid tissue, or targeted selectively to infected cells, as well as increase the half-life of the peptide composition. Liposomes include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phosphohpid dispersions, lamellar layers and the like. In these preparations the peptide to be delivered is incorporated as part of a liposome, alone or in conjunction with a molecule which binds to, e.g., a receptor prevalent among lymphoid cells, such as monoclonal antibodies which bind to the CD45 antigen, or with other therapeutic or immunogenic compositions. Thus, liposomes filled with a desired peptide or conjugate ofthe invention can be directed to the site of lymphoid cells, where the liposomes then deliver the selected inhibitor compositions. Liposomes for use in the invention are formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol. The selection of lipids is generally guided by consideration of, e.g., liposome size, acid lability and stability of the liposomes in the blood stream. A variety of methods are available for preparing liposomes, as described in, e.g., Szoka et al., Ann. Rev. Biophys. Bioeng. 9:467 (1980), U.S. Pat. Nos. 4,235,871, 4,501,728 and 4,837,028. The targeting of liposomes using a variety of targeting agents is well known in the art (see, e.g., U.S. Patent Nos. 4,957,773 and 4,603,044). For targeting to the immune cells, a ligand to be incorporated into the liposome can include, e.g., antibodies or fragments thereof specific for cell surface determinants of the desired immune system cells. A liposome suspension containing a peptide or conjugate may be administered intravenously, locally, topically, etc. in a dose which varies according to, inter alia, the manner of administration, the conjugate being delivered, and the stage of the disease being treated. In order to specifically deliver a PDZ motif sequence (PL sequence) peptide into a specific cell type, the peptide may be linked to a cell-specific targeting moiety, which include but are not limited to, ligands for diverse leukocyte surface molecules such as growth factors, hormones and cytokines, as well as antibodies or antigen-binding fragments thereof. Since a large number of cell surface receptors have been identified in leukocytes, ligands or antibodies specific for these receptors may be used as cell-specific targeting moieties. For example, interleukin-2, B7-1 (CD80), B7-2 (CD86) and CD40 or peptide fragments thereof may be used to specifically target activated T cells (The Leucocyte Antigen Facts Book, 1997, Barclay et al. (eds.), Academic Press). CD28, CTLA-4 and CD40L or peptide fragments thereof may be used to specifically target B cells. Furthermore, Fc domains may be used to target certain Fc receptor- expressing cells such as monocytes.
.Antibodies are the most versatile cell-specific targeting moieties because they can be generated against any cell surface antigen. Monoclonal antibodies have been generated against leukocyte lineage-specific markers such as certain CD antigens. Antibody variable region genes can be readily isolated from hybridoma cells by methods well known in the art. However, since antibodies are assembled between two heavy chains and two light chains, it is preferred that a scFv be used as a cell-specific targeting moiety in the present invention. Such scFv are comprised of VH and VL domains linked into a single polypeptide chain by a flexible linker peptide. The PDZ motif sequence (PL sequence) may be linked to a transmembrane transporter sequence and a cell-specific targeting moiety to produce a tri-fusion molecule. This molecule can bind to a leukocyte surface molecule, passes through the membrane and targets PDZ domains. Alternatively, a PDZ motif sequence (PL sequence) may be linked to a cell- specific targeting moiety that binds to a surface molecule that internalizes the fusion peptide. In an other approach, microspheres of artificial polymers of mixed amino acids
(proteinoids) have been used to deliver pharmaceuticals. For example, U.S. Pat. No. 4,925,673 describes drug-containing proteinoid microsphere carriers as well as methods for their preparation and use. These proteinoid microspheres are useful for the delivery of a number of active agents. Also see, U.S. Patent Nos. 5,907,030 and 6,033,884, which are incorporated herein by reference. 6.9.2 Introduction of Polynucleotides into Cells
A polynucleotide encoding a surface receptor C-terminal peptide may be useful in the treatment of various leukocyte activation-associated abnormal conditions. By introducing gene sequences into cells, gene therapy can be used to treat conditions in which leukocytes are activated to result in deleterious consequences. In one embodiment, a polynucleotide that encodes a PL sequence peptide of the invention is introduced into a cell where it is expressed.
The expressed peptide then inhibits the interaction of PDZ proteins and PL proteins in the cell.
Thus, in one embodiment, the polypeptides of the invention are expressed in a cell by introducing a nucleic acid (e.g., a DNA expression vector or mRNA) encoding the desired protein or peptide into the cell. Expression may be either constitutive or inducible depending on the vector and choice of promoter. Methods for introduction and expression of nucleic acids into a cell are well known in the art and described herein.
In a specific embodiment, nucleic acids comprising a sequence encoding a peptide disclosed herein, are administered to a human subject. In this embodiment of the invention, the nucleic acid produces its encoded product that mediates a therapeutic effect by inhibiting leukocyte activation. Any ofthe methods for gene therapy available in the art can be used according to the present invention. Exemplary methods are described below.
For general reviews ofthe methods of gene therapy, see Goldspiel et al., 1993, Clinical Pharmacy 12:488-505; Wu and Wu, 1991, Biotherapy 3:87-95; Tolstoshev, 1993, Ann. Rev. Pharmacol. Toxicol. 32:573-596; Mulligan, 1993, Science 260:926-932; and Morgan and Anderson, 1993, Ann. Rev. Biochem. 62:191-217; May, 1993, TIBTECH 11 (5): 155-215. Methods commonly known in the art of recombinant DNA technology which can be used are described in Ausubel et al. (eds.), 1993, Current Protocols in Molecular Biology, John Wiley & Sons, NY; and Kriegler, 1990, Gene Transfer and Expression, A Laboratory Manual, Stockton Press, NY.
In a preferred embodiment of the invention, the therapeutic composition comprises a coding sequence that is part of an expression vector. In particular, such a nucleic acid has a promoter operably linked to the coding sequence, said promoter being inducible or constitutive, and, optionally, tissue-specific. In another specific embodiment, a nucleic acid molecule is used in which the coding sequence and any other desired sequences are flanked by regions that promote homologous recombination at a desired site in the genome, thus providing for intrachromosomal expression of the nucleic acid (Koller and Smithies, 1989, Proc. Natl. Acad. Sci. USA 86:8932-8935; Zijlstra et al., 1989, Nature 342:435-438). Delivery ofthe nucleic acid into a patient may be either direct, in which case the patient is directly exposed to the nucleic acid or nucleic acid-carrying vector, or indirect, in which case, cells are first transformed with the nucleic acid in vitro, then transplanted into the patient. These two approaches are known, respectively, as in vivo or ex vivo gene therapy. In a specific embodiment, the nucleic acid is directly administered in vivo, where it is expressed to produce the encoded product. This can be accomplished by any methods known in the art, e.g., by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by infection using a defective or attenuated retroviral or other viral vector (see U.S. Patent No. 4,980,286), by direct injection of naked DNA, by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), by coating with lipids or cell-surface receptors or transfecting agents, by encapsulation in liposomes, microparticles, or microcapsules, by administering it in linkage to a peptide which is known to enter the nucleus, or by administering it in linkage to a ligand subject to receptor-mediated endocytosis (see e.g., Wu and Wu, 1987, J. Biol. Chem. 262:4429-4432) which can be used to target cell types specifically expressing the receptors. In another embodiment, a nucleic acid- ligand complex can be formed in which the ligand comprises a fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation. In yet another embodiment, the nucleic acid can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor (see, e.g., PCT Publications WO 92/06180 dated April 16, 1992; WO 92/22635 dated December 23, 1992; WO92/20316 dated November 26, 1992; WO93/14188 dated July 22, 1993; WO 93/20221 dated October 14, 1993). Alternatively, the nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination (Koller and Smithies, 1989, Proc. Natl. Acad. Sci. USA 86:8932- 8935; Zijlstra et al., 1989, Nature 342:435-438). In a preferred embodiment of the invention, adenoviruses as viral vectors can be used in gene therapy. Adenoviruses have the advantage of being capable of infecting non- dividing cells (Kozarsky and Wilson, 1993, Current Opinion in Genetics and Development 3:499-503). Other instances of the use of adenoviruses in gene therapy can be found in Rosenfeld et al., 1991 , Science 252:431-434; Rosenfeld et al., 1992, Cell 68: 143-155; and Mastrangeli et al., 1993, J. Clin. Invest. 91 :225-234. Furthermore, adenoviral vectors with modified tropism may be used for cell specific targeting (WO98/40508). Adeno-associated virus (AAV) has also been proposed for use in gene therapy (Walsh et al., 1993, Proc. Soc. Exp. Biol. Med. 204:289-300). In addition, retroviral vectors (see Miller et al., 1993, Meth. Enzymol. 217:581- 599) have been modified to delete retroviral sequences that are not necessary for packaging of the viral genome and integration into host cell DNA. The coding sequence to be used in gene therapy is cloned into the vector, which facilitates delivery ofthe gene into a patient. More detail about retroviral vectors can be found in Boesen et al., 1994, Biotherapy 6:291-302, which describes the use of a retroviral vector to deliver the mdrl gene to hematopoietic stem cells in order to make the stem cells more resistant to chemotherapy. Other references illustrating the use of retroviral vectors in gene therapy are: Clowes et al., 1994, J. Clin. Invest. 93:644-651; Kiem et al., 1994, Blood 83:1467-1473; Salmons and Gunzberg, 1993, Human Gene Therapy 4:129-141 ; and Grossman and Wilson, 1993, Curr. Opin. in Genetics and Devel. 3:1 10-114. Another approach to gene therapy involves transferring a gene to cells in tissue culture. Usually, the method of transfer includes the transfer of a selectable marker to the cells. The cells are then placed under selection to isolate those cells that have taken up and are expressing the transferred gene. Those cells are then delivered to a patient. In this embodiment, the nucleic acid is introduced into a cell prior to administration in vivo ofthe resulting recombinant cell. Such introduction can be carried out by any method known in the art, including but not limited to transfection, electroporation, lipofection, microinjection, infection with a viral or bacteriophage vector containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcell-mediated gene transfer, spheroplast fusion, etc. Numerous techniques are known in the art for the introduction of foreign genes into cells (see e.g., Loeffler and Behr, 1993, Meth. Enzymol. 217:599-618; Cohen et al., 1993, Meth. Enzymol. 217:618-644; Cline, 1985, Pharmac. Ther. 29:69-92) and may be used in accordance with the present invention, provided that the necessary developmental and physiological functions ofthe recipient cells are not disrupted. The technique should provide for the stable transfer o the nucleic acid to the cell, so that the nucleic acid is expressible by the cell and preferably heritable and expressible by its cell progeny. In a preferred embodiment, the cell used for gene therapy is autologous to the patient.
In a specific embodiment, the nucleic acid to be introduced for purposes of gene therapy comprises an inducible promoter operably linked to the coding sequence, such that expression of the nucleic acid is controllable by controlling the presence or absence of the appropriate inducer of transcription.
Ohgonucleotides such as anti-sense RNA and DNA molecules, and ribozymes that function to inhibit the translation of a leukocyte surface receptor mRNA, especially its C- terminus are also within the scope of the invention. Anti-sense RNA and DNA molecules act to directly block the translation of mRNA by binding to targeted mRNA and preventing protein translation. In regard to antisense DNA, oligodeoxyribonucleotides derived from the translation initiation site, e.g., between -10 and +10 regions of a nucleotide sequence, are preferred. The antisense oligonucleotide may comprise at least one modified base moiety which is selected from the group including, but not limited to, 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine,
5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine,
5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta- D-mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6- isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil- 5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino- 3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine.
Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. The mechanism of ribozyme action involves sequence specific hybridization ofthe ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage. Within the scope of the invention are engineered hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of leukocyte surface receptor RNA sequences.
Specific ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites which include the following sequences, GUA, GUU and GUC. Once identified, short RNA sequences of between 15 and 20 ribonucleotides corresponding to the region ofthe target gene containing the cleavage site may be evaluated for predicted structural features such as secondary structure that may render the oligonucleotide sequence unsuitable. The suitability of candidate targets may also be evaluated by testing their accessibility to hybridization with complementary ohgonucleotides, using ribonuclease protection assays.
The anti-sense RNA and DNA molecules and ribozymes ofthe invention may be prepared by any method known in the art for the synthesis of nucleic acid molecules. These include techniques for chemically synthesizing oligodeoxyribonucleotides well known in the art such as for example solid phase phosphoramidite chemical synthesis. Alternatively, RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the RNA molecule. Such DNA sequences may be incorporated into a wide variety of vectors which contain suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters. Alternatively, antisense cDNA constructs that synthesize antisense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably into cell lines.
Various modifications to the DNA molecules may be introduced as a means of increasing intracellular stability and half-life. Possible modifications include, but are not limited to, the addition of flanking sequences of ribo- or deoxy- nucleotides to the 5' and/or 3' ends of the molecule or the use of phosphorothioate or 2' O-methyl rather than phosphodiesterase linkages within the oligodeoxyribonucleotide backbone.
6.9.3 Other Pharmaceutical Compositions The compounds of the invention, may be administered to a subject per se or in the form of a sterile composition or a pharmaceutical composition. Pharmaceutical compositions comprising the compounds of the invention may be manufactured by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes. Pharmaceutical compositions may be formulated in conventional manner using one or more physiologically acceptable carriers, diluents, excipients or auxiliaries that facilitate processing of the active peptides or peptide analogues into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
For topical administration the compounds ofthe invention may be formulated as solutions, gels, ointments, creams, suspensions, etc. as are well-known in the art.
Systemic formulations include those designed for administration by injection, e.g. subcutaneous, intravenous, intramuscular, intrathecal or intraperitoneal injection, as well as those designed for transdermal, transmucosal, oral or pulmonary administration.
For injection, the compounds of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. The solution may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the compounds may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. This route of administration may be used to deliver the compounds to the nasal cavity.
For oral administration, the compounds can be readily formulated by combining the active peptides or peptide analogues with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds ofthe invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. For oral solid formulations such as, for example, powders, capsules and tablets, suitable excipients include fillers such as sugars, such as lactose, sucrose, mannitol and sorbitol; cellulose preparations such as maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP); granulating agents; and binding agents. If desired, disintegrating agents may be added, such as the cross-linked polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
If desired, solid dosage forms may be sugar-coated or enteric-coated using standard techniques.
For oral liquid preparations such as, for example, suspensions, elixirs and solutions, suitable carriers, excipients or diluents include water, glycols, oils, alcohols, etc. Additionally, flavoring agents, preservatives, coloring agents and the like may be added.
For buccal administration, the compounds may take the form of tablets, lozenges, etc. formulated in conventional manner.
For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e.g. gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch. The compounds may also be formulated in rectal or vaginal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. Alternatively, other pharmaceutical delivery systems may be employed.
Liposomes and emulsions are well known examples of delivery vehicles that may be used to deliver peptides and peptide analogues of the invention. Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity. Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid polymers containing the therapeutic agent. Various of sustained-release materials have been established and are well known by those skilled in the art.
Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability ofthe therapeutic reagent, additional strategies for protein stabilization may be employed. As the compounds ofthe invention may contain charged side chains or termini, they may be included in any o the above-described formulations as the free acids or bases or as pharmaceutically acceptable salts. Pharmaceutically acceptable salts are those salts which substantially retain the biologic activity ofthe free bases and which are prepared by reaction with inorganic acids. Pharmaceutical salts tend to be more soluble in aqueous and other protic solvents than are the corresponding free base forms.
6.10. Effective Dosages
The compounds ofthe invention will generally be used in an amount effective to achieve the intended purpose. For use to inhibit leukocyte activation-associated disorders, the compounds ofthe invention or pharmaceutical compositions thereof, are administered or applied in a therapeutically effective amount. By therapeutically effective amount is meant an amount effective ameliorate or prevent the symptoms, or prolong the survival of, the patient being treated. Determination of a therapeutically effective amount is well within the capabilities of those skilled in the art, especially in light of the detailed disclosure provided herein. An "inhibitory amount" or "inhibitory concentration" of a PL-PDZ binding inhibitor is an amount that reduces binding by at least about 40%, preferably at least about 50%, often at least about 70%, and even as much as at least about 90%. Binding can as measured in vitro (e.g., in an A assay or G assay) or in situ. For systemic administration, a therapeutically effective dose can be estimated initially from in vitro assays. For example, a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture (i.e., the concentration of test compound that inhibits 50% of leukocyte surface receptor-PDZ domain- containing protein interactions). Such information can be used to more accurately determine useful doses in humans.
Initial dosages can also be estimated from in vivo data, e.g., animal models, using techniques that are well known in the art. One having ordinary skill in the art could readily optimize administration to humans based on animal data.
Dosage amount and interval may be adjusted individually to provide plasma levels ofthe compounds that are sufficient to maintain therapeutic effect. Usual patient dosages for administration by injection range from about 0.1 to 5 mg/kg/day, preferably from about 0.5 to 1 mg/kg/day. Therapeutically effective serum levels may be achieved by administering multiple doses each day.
In cases of local administration or selective uptake, the effective local concentration ofthe compounds may not be related to plasma concentration. One having skill in the art will be able to optimize therapeutically effective local dosages without undue experimentation.
The amount of compound administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
The therapy may be repeated intermittently while symptoms detectable or even when they are not detectable. The therapy may be provided alone or in combination with other drugs. In the case of conditions associated with leukocyte activation such as transplantation rejection and autoimmunity, the drugs that may be used in combination with the compounds of the invention include, but are not limited to, steroid and non-steroid anti-inflammatory agents.
6.10.1 Toxicity
Preferably, a therapeutically effective dose of the compounds described herein will provide therapeutic benefit without causing substantial toxicity. Toxicity of the compounds described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the LD50 (the dose lethal to 50% of the population) or the LD100 (the dose lethal to 100% of the population). The dose ratio between toxic and therapeutic effect is the therapeutic index. Compounds which exhibit high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in formulating a dosage range that is not toxic for use in human. The dosage o the compounds described herein lies preferably within a range of circulating concentrations that include the effective dose with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view ofthe patient's condition. (See, e.g., Fingl et al, 1975, In: The Pharmacological Basis of Therapeutics, Ch.l, p.l).
6.11. EXAMPLE 1 : TAT-T CELL SURFACE RECEPTOR CARBOXYL TERMINUS FUSION PEPTIDES INHIBITED T CELL ACTIVATION 6.11.1. Materials And Methods
6.11.1.1. Peptide Synthesis
All peptides were chemically synthesized by standard procedures. The Tat-CD3 carboxyl terminus fusion peptide, (GYGRKKRRQRRRGPPSSSSGL, SEQ ID NO: 174); Tat- CLASP1 carboxyl terminus fusion peptide, (GYGRKKRRQRRRGSISSSAEV, SEQ ID NOT 75); Tat-CLASP2 carboxyl terminus fusion peptide, (GYGRKKRRQRRRGMTSSSSVV,
SEQ ID NO: 176); and Tat peptide, (GYGRKKRRQRRRG, SEQ ID NO: ); were dissolved at 1 mM in PBS, pH 7, or dH2O. Stock MBPAcl-16 peptide, (AcASQKRPSQRHGSKYLA), was dissolved at 5 mM. All peptides were aliquoted and stored at -80°C until tested.
6.1 1.1.2 Cell Cultures
Cells were maintained and tested in RPMI 1640 media supplemented with 10% fetal calf serum (HyClone), 2 mM glutamine, 10 mM Hepes, 100 U/ml penicillin, 100 μg/ml streptomycin, 0.1 mM non-essential amino acids, 1 mM sodium pyruvate, and 50 μM beta mercaptoethanol.
6.1 1.1.3 T Cell Stimulation Assay Supernatants were assayed for cytokine production following activation of T cell lines. Mouse T cell lines were stimulated using two different methods, either with antigen and antigen presenting cells or anti-mouse CD3.
Antigen-specific mouse T cells, BR4.2, were activated with the N-terminal 16 amino acid sequences of myelin basic protein (MBPAcl-16) and syngenic mouse splenocytes in 96-well plates. Mitomycin C-treated antigen presenting cells, 2 x 105 B 10.BR, were added to each row of serially diluted MBPAcl-16 ranging from 0 to 200 μM. Next, 10 μM Tat- peptides or media alone was added to each row. Finally, 2 x 104 MBPAcl-16-specific T cell, pre-loaded with 10 μM Tat-peptides (see above), were added to all wells (Rabinowitz et al.,1997,. Proc. Natl. Acad. Sci. U.S.A., 94:8702-8707). Cells were activated during an overnight incubation at 5% CO2, 37°C. Cell supernatant was collected and stored at -80°C until assayed for cytokine production. The final volume was 200 μl/well.
Antibody against mouse CD3 (Pharmigen #145-2C11) was coated overnight at 4°C using 96-well flat bottom Elisa plates at a final concentration of 0.5 μg/ml, diluted in PBS. Just prior to use, plates were washed three times with 200 μl/well PBS to remove excess anti- CD3. To ensure that cells were given sufficient time to transduce Tat-peptides before activation, T cells (5x105 cells/ml) were pre-treated with or without 10 μM Tat-peptides for two hours at 5% CO2, 37°C and then diluted in media with or without 10 μM Tat-peptides to a final concentration of 2x104 cells per well in a final volume of 200 μl. Cells were then treated as described above.
6.11.1.4 Cytokine ELISA
IFNγ was measured from cell supernatants, described above, at ambient temperature using the Endogen, Inc. ELISA protocol 3. Briefly, 96-well, flat bottom, high binding ELISA plates were preincubated overnight with coating antibody (MM700). Plates were washed with 50 mM TRIS, 0.2% tween-20, pH 8 and they blocked for one hour with PBS plus 2% BSA. Washed plates were then incubated one hour with 25 μl of cell supernatant and 25 μl blocking buffer, or with 50 μl IFNγ standard. The presence of IFNγ was detected with a biotin- labeled anti-mouse IFNγ monoclonal antibody (MM700B, Endogen, Inc.,). Quantitative amounts of detection antibody are revealed with horseradishperoxidase-conjugated streptavidin. The enzymatic, color, substrate for HRP, tetramethylbenzidine (TMB), was developed for up to 30 minutes and stopped with 1.0 M H2SO4. The absorbance at 450 nm was measured using a microtiter plate reader (Thermo Max, Molecular Devices) and the concentration of unknown IFNγ from cell supernatants was calculated from a standard curve generated by Softmax Pro. software (Molecular Devices).
6.1 1.2 Results
Peptides containing Tat transporter sequences linked to C-terminal sequences of various PLs were testing for their ability to inhibit T cell activation. FIGURE 4A shows that the Tat-CD3 fusion peptide inhibits T cell activation mediated by peptide:MHC as compared to controls of Tat-peptide alone or no peptide. FIGURE 4B shows that Tat-CLASP2 carboxyl terminus fusion peptide inhibited T cell activation mediated by monoclonal anti-CD3 as compared to Tat-peptide alone. Tat-CLASPl fusion peptide did not inhibit T cell activation in this experiment. These results indicate that peptides containing potential inhibitory sequences can be transported into T cells through transporter peptide such as Tat to disrupt surface receptor organization mediated by PDZ proteins. Disruption of PDZ-mediated surface receptor organization leads to blockage of T cell activation in response to antigen.
6.12. EXAMPLE 2: DESIGN OF AN INHIBITOR OF DLG 1-LIGAND BINDING WITH GREATER THAN 100 uM POTENCY
A GST/DLG1 fusion protein (See TABLE 3) and a biotin-labeled peptide corresponding to the C-terminal 20 amino acids of the CLASP-2 protein, peptide AA2L (See
TABLE 4), were synthesized and purified by standard techniques well known in the art as described supra. This PDZ-ligand combination was then shown to bind specifically using both the "A" assay and the "G" assay (See TABLE 2). Once specific binding was demonstrated, the apparent affinity of the binding interaction was determined using Approach 1 of the section entitled "Measurement of PDZ-ligand binding affinity" (see FIGURE 2A). The measured apparent affinity was 21 uM. This implies that 21 uM labeled CLASP-2 peptide AA2L filled 50% of the binding sites for CLASP-2 on DLGl . Thus, 21 uM unlabeled CLASP-2 peptide should be able to block the binding of a given ligand to DLGl by approximately 50%, assuming that the given ligand (1) binds to the same site(s) on DLGl as Qasp~2 and (2) is not added at sufficient concentration to reduce significantly the binding ofthe CLASP-2 peptide (i.e. cannot out-compete the CLASP-2 peptide).
To detect such inhibition, it was necessary to synthesize an analogue of the CLASP2peptide AA2L that (1) retained similar DLGl binding properties and (2) would not itself generate a signal in the assay selected to measure inhibition. Because most molecular interactions between PDZ proteins and their ligands involve only the C-terminal 6 amino acids ofthe ligand, an eight amino acid variant of the CLASP-2 peptide, MTSSSSVV, was anticipated to retain similar DLGl binding properties as the 20 amino acid AA2L CLASP-2 peptide. This eight amino acid CLASP-2 peptide (lacking a functional label) was therefore synthesized and purified by standard techniques as described supra. When 100 uM of the (functionally unlabeled) eight amino acid CLASP-2 peptide and 20 uM of the biotin-labeled AA2L CLASP-2 peptide were added simultaneously to DLGl in a variant o the "G" assay (described supra), the binding ofthe labeled AA2L CLASP-2 peptide was, as predicted, inhibited by greater than 50% (FIGURE 3A). An analogous experiment in which the labeled AA2L CLASP-2 peptide was replaced with another labeled DLGl ligand, labeled AAI3L Fas peptide demonstrated similar inhibition by the eight amino acid CLASP-2 peptide (FIGURE 3A). Thus, an effective inhibitor of DLGl -ligand binding (i.e. the eight amino acid CLASP-2 peptide MTSSSSVV) with a known potency range (order of magnitude 21 uM) was designed based on knowledge ofthe affinity, 21 uM, with which a particular labeled ligand, the CLASP-2 peptide AA2L, bound to DLGl .
The present invention is not to be limited in scope by the exemplified embodiments which are intended as illustrations of single aspects of the invention and any sequences which are functionally equivalent are within the scope of the invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope ofthe appended claims.
All publications cited herein are incorporated by reference in their entirety and for all purposes.

Claims

WHAT IS CLAIMED IS
1. A method of determining the apparent affinity (Kd) of binding between a PDZ domain and a ligand, comprising
(a) immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface;
(b) contacting the immobilized polypeptide with a plurality of different concentrations ofthe ligand;
(c) determining the amount of binding ofthe ligand to the immobilized polypeptide at each o the concentrations of ligand; (d) calculating the apparent affinity ofthe binding from the binding determined in (c).
2. The method of claim 1, wherein the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain.
3. The method of claim 1, wherein the polypeptide comprising the PDZ domain is a fusion protein.
4. The method of claim 3, wherein the fusion protein is a GST-PDZ domain fusion protein.
5. A method of determining the Ki of an inhibitor or suspected inhibitor of binding between a PDZ domain and a ligand, comprising
(a) immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface; (b) contacting the immobilized polypeptide with a plurality of different mixtures of the ligand and inhibitor, wherein the different mixtures comprise a fixed amount of ligand, at least a portion of which is detectably labeled, and different concentrations of the inhibitor;
(c) determining the amount of ligand bound at the different concentrations of inhibitor; (d) calculating the Ki of the inhibitor from the binding determined in (c).
6. The method of claim 5 wherein the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain.
7. The method of claim 6 wherein the fixed amount of ligand is between about 0.01 Kd and about 2 Kd.
8. A method of identifying an agent that enhances the binding of a PDZ domain to a ligand, comprising
(a) immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface;
(b) contacting the immobilized polypeptide with the ligand in the presence of a test agent and determining the amount of ligand bound; and,
(c) comparing the amount of ligand bound in the presence ofthe test agent with the amount of ligand bound by the polypeptide in the absence of the test agent, wherein at least two-fold greater binding in the presence ofthe test agent compared to the absence ofthe test agent indicates that the test agent is an agent that enhances the binding ofthe PDZ domain to the ligand.
9. The method of claim 8 wherein the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain.
10. A method of determining the potency (K-,,,,,^-) of an enhancer of binding between a PDZ domain and a ligand, comprising
(a) immobilizing a polypeptide comprising the PDZ domain and a non-PDZ domain on a surface;
(b) contacting the immobilized polypeptide with a plurality of different mixtures of the ligand and enhancer, wherein the different mixtures comprise a fixed amount of ligand, at least a portion of which is detectably labeled, and different concentrations ofthe enhancer;
(c) determining the amount of ligand bound at the different concentrations of enhancer: (d) calculating the potency (K,,n|iancer) ofthe enhancer from the binding determined in (c).
1 1. The method of claim 10 wherein the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain.
12. The method of claim 11 wherein the fixed amount of ligand is between about 0.01 Kd and about 0.5 Kd.
13. A method of identifying a high specificity interaction between a particular PDZ domain and a ligand known or suspected of binding at least one PDZ domain, comprising:
(a) providing a plurality of different immobilized polypeptides, each of said polypeptides comprising a PDZ domain and a non-PDZ domain;
(b) determining the affinity ofthe ligand for each of said polypeptides;
(c) comparing the affinity of binding ofthe ligand to each of said polypeptides, wherein an interaction between the ligand and a particular PDZ domain is deemed to have high specificity when the ligand binds an immobilized polypeptide comprising the particular PDZ domain with at least 2-fold higher affinity than to immobilized polypeptides not comprising the particular PDZ domain (a).
14. The method of claim 13 wherein the polypeptide is immobilized by binding the polypeptide to an immobilized immunoglobulin that binds the non-PDZ domain.
15. A method for determining the PDZ-PL inhibition profile of a compound comprising: (a) providing
(i) a plurality of different immobilized polypeptides, each of said polypeptides comprising a PDZ domain and a non-PDZ domain;
(ii) a plurality of corresponding ligands, wherein each ligand binds at least one PDZ domain in (i); (b) contacting each of said immobilized polypeptides in (i) with a corresponding ligand in (ii) in the presence and absence of a test compound; (c) determining for each polypeptide-ligand pair in (b) whether the test compound inhibits binding between the immobilized polypeptide and the corresponding ligand thereby determining the PDZ-PL inhibition profile of the test compound.
16. An array comprising a plurality of different immobilized polypeptides, each of said polypeptides comprising a PDZ domain and a non-PDZ domain.
17. The array of claim 16 wherein the array is situated in a plastic multiwell plate.
18. The array of claim 16 comprising at least 12 different polypeptides comprising at least 12 different PDZ domains.
19. The array of claim 18 wherein the at least 12 different PDZ domains are from PDZs expressed in lymphocytes.
20. The array of claim 19 wherein said PDZs are listed in Table 6.
21. An assay device comprising a plurality of different immobilized PDZ-containing proteins organized in an array.
22. The device of claim 21 comprising at least 25 different PDZ-containing proteins.
23. A method for identifying an interaction between a PDZ domain and a PL comprising contacting a PL to a plurality of PDZ containing polypeptides and detecting binding of at least one PL to a PDZ.
24. The method of claim 23 wherein the contacting occurs on the assay device of claim 1.
25. The method of claim 23 wherein an interaction between a PDZ and more than one
PL is detected.
26. The method of claim 23 wherein an interaction between a PL and more than one PDZ is detected.
27. A method for identifying a modulator of an interaction between a PDZ and a PL comprising carrying out the method of claims 23 in the presence and absence of a test compound and detecting a difference in at least one PDZ-PL interaction in the presence and absence o the test compound.
28. A method for identifying a modulator of an interaction between a PDZ and a PL comprising carrying out the method of claim 24 in the presence and absence of a test compound and detecting a difference in at least one PDZ-PL interaction in the presence and absence ofthe test compound.
29. The method of claim 27 wherein the modulator is an enhancer ofthe interaction.
30. The method of claim 27 wherein the modulator is an inhibitor ofthe interaction.
PCT/US2001/032150 2000-10-13 2001-10-11 Molecular interactions in hematopoietic cells WO2002031512A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002215354A AU2002215354A1 (en) 2000-10-13 2001-10-11 Molecular interactions in hematopoietic cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US09/688,017 US6942981B1 (en) 1999-05-14 2000-10-13 Method of determining interactions with PDZ-domain polypeptides
US09/688,017 2000-10-13

Publications (2)

Publication Number Publication Date
WO2002031512A2 true WO2002031512A2 (en) 2002-04-18
WO2002031512A3 WO2002031512A3 (en) 2003-03-27

Family

ID=24762770

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/032150 WO2002031512A2 (en) 2000-10-13 2001-10-11 Molecular interactions in hematopoietic cells

Country Status (2)

Country Link
AU (1) AU2002215354A1 (en)
WO (1) WO2002031512A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6942981B1 (en) 1999-05-14 2005-09-13 Arbor Vita Corporation Method of determining interactions with PDZ-domain polypeptides
EP1897888A1 (en) * 2006-09-05 2008-03-12 AplaGen GmbH Peptides binding the TPO receptor
WO2008028645A1 (en) * 2006-09-05 2008-03-13 Aplagen Gmbh Peptides binding the tpo receptor
US7745109B2 (en) 2000-12-22 2010-06-29 Dana-Farber Cancer Insitute, Inc. Regulation of cell growth by MUC1
US7871784B2 (en) 2007-02-02 2011-01-18 Dana-Farber Cancer Institute, Inc. Methods and compositions relating to the regulation of apoptosis by MUC1 and BH3-containing proapoptotic proteins
US7972870B2 (en) 2007-02-02 2011-07-05 Dana-Farber Cancer Institute, Inc. Methods and compositions relating to the regulation of MUC1 by HSF1 and STAT3
US8129506B2 (en) 2003-10-24 2012-03-06 Genzyme Corporation Modulation of the interaction of MUC1 with MUC1 ligands

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000069898A2 (en) * 1999-05-14 2000-11-23 Arbor Vita Corporation Molecular interactions in allergy cells
WO2000069897A2 (en) * 1999-05-14 2000-11-23 Arbor Vita Corporation Modulation of glgf motif (dhr/pdz) containing proteins interaction with cd3n in t lymphocytes

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000069898A2 (en) * 1999-05-14 2000-11-23 Arbor Vita Corporation Molecular interactions in allergy cells
WO2000069897A2 (en) * 1999-05-14 2000-11-23 Arbor Vita Corporation Modulation of glgf motif (dhr/pdz) containing proteins interaction with cd3n in t lymphocytes

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
GEE STEPHEN H ET AL: "Cyclic peptides as non-carboxyl-terminal ligands of syntrophin PDZ domains" JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY OF BIOLOGICAL CHEMISTS, BALTIMORE, MD, US, vol. 273, no. 34, 21 August 1998 (1998-08-21), pages 21980-21987, XP002155720 ISSN: 0021-9258 *
KOBAYASHI ICHIRO ET AL: "Identification of an autoimmune enteropathy-related 75-kilodalton antigen" GASTROENTEROLOGY, SAUNDERS, PHILADELPHIA, PA,, US, vol. 117, no. 4, October 1999 (1999-10), pages 823-830, XP002154870 ISSN: 0016-5085 *
MAEKAWA KAZUHIKO ET AL: "Association of protein-tyrosine phosphatase PTP-BAS with the transcription-factor-inhibitory protein IkappaBalpha through interaction between the PDZ1 domain and ankyrin repeats" BIOCHEMICAL JOURNAL, PORTLAND PRESS, LONDON, GB, vol. 337, no. 2, 15 January 1999 (1999-01-15), pages 179-184, XP002154869 ISSN: 0264-6021 *
SARAS J ET AL: "PDZ domains bind carboxy-terminal sequences of target proteins" TIBS TRENDS IN BIOCHEMICAL SCIENCES, ELSEVIER PUBLICATION, CAMBRIDGE, EN, vol. 21, no. 12, 1 December 1996 (1996-12-01), pages 455-458, XP004063028 ISSN: 0968-0004 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6942981B1 (en) 1999-05-14 2005-09-13 Arbor Vita Corporation Method of determining interactions with PDZ-domain polypeptides
US7432065B2 (en) 1999-05-14 2008-10-07 Arbor Vita Corporation Methods of identifying modulators of interaction between PDZ domain proteins and PL proteins
US7514224B2 (en) 1999-05-14 2009-04-07 Arbor Vita Corporation Arrays of PDZ domain polypeptides
US7745109B2 (en) 2000-12-22 2010-06-29 Dana-Farber Cancer Insitute, Inc. Regulation of cell growth by MUC1
US8129506B2 (en) 2003-10-24 2012-03-06 Genzyme Corporation Modulation of the interaction of MUC1 with MUC1 ligands
EP1897888A1 (en) * 2006-09-05 2008-03-12 AplaGen GmbH Peptides binding the TPO receptor
WO2008028645A1 (en) * 2006-09-05 2008-03-13 Aplagen Gmbh Peptides binding the tpo receptor
US7871784B2 (en) 2007-02-02 2011-01-18 Dana-Farber Cancer Institute, Inc. Methods and compositions relating to the regulation of apoptosis by MUC1 and BH3-containing proapoptotic proteins
US7972870B2 (en) 2007-02-02 2011-07-05 Dana-Farber Cancer Institute, Inc. Methods and compositions relating to the regulation of MUC1 by HSF1 and STAT3

Also Published As

Publication number Publication date
AU2002215354A1 (en) 2002-04-22
WO2002031512A3 (en) 2003-03-27

Similar Documents

Publication Publication Date Title
US7432065B2 (en) Methods of identifying modulators of interaction between PDZ domain proteins and PL proteins
US20120093873A1 (en) Molecular Interactions in Hematopoietic Cells
ES2436624T3 (en) Peptides or peptide analogs for modulation of the binding of a PDZ protein and a PL protein
US20090075377A1 (en) Molecular interactions in cells
US7858322B2 (en) Method of determining inhibition of binding to TRPM7 protein
EP1387692A2 (en) Molecular interactions in hematopoietic cells
EP1443951A2 (en) Molecular interactions in cells
US20110039758A1 (en) Molecular Interactions in Neurons
EP1578365A2 (en) Molecular interactions in neurons
US20050019841A1 (en) Modulation of signaling pathways
US20050282743A1 (en) Molecular interactions in cells
US20020169283A1 (en) Clasp-7 transmembrane protein
WO2002031512A2 (en) Molecular interactions in hematopoietic cells
EP2385124B1 (en) Peptides or peptide analogues for modulating the binding of a PDZ protein and a PL protein
US7919584B1 (en) Methods and compositions for diagnosis and treatment of asthma
WO2002031117A2 (en) Clasp-2 transmembrane proteins
US20030103992A1 (en) Clasp membrane proteins
WO2001042297A9 (en) Clasp-3 transmembrane protein
WO2001085908A2 (en) Clasp-1 transmembrane protein
US20020068302A1 (en) Clasp-4 transmembrane protein
WO2001042294A2 (en) Clasp-4 transmembrane protein

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PH PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP