Protein Kinase C Zeta As A Drug Target For Arthritis And Other Inflammatory Diseases

LaVallie; Edward R. ;   et al.

Patent Application Summary

U.S. patent application number 12/616484 was filed with the patent office on 2010-03-11 for protein kinase c zeta as a drug target for arthritis and other inflammatory diseases. This patent application is currently assigned to WYETH. Invention is credited to Maya Arai, Lisa A. Collins-Racie, Edward R. LaVallie.

Application Number20100062448 12/616484
Document ID /
Family ID33479253
Filed Date2010-03-11

United States Patent Application 20100062448
Kind Code A1
LaVallie; Edward R. ;   et al. March 11, 2010

PROTEIN KINASE C ZETA AS A DRUG TARGET FOR ARTHRITIS AND OTHER INFLAMMATORY DISEASES

Abstract

The present invention is based on the discovery that .zeta.PKC expression is increased in the tissues of arthritis patients as compared to normal individuals. Accordingly, the present invention provides methods of diagnosing, prognosing, and monitoring the course of arthritis in a patient based on increased .zeta.PKC gene expression in arthritic tissue. The present invention further provides compounds that inhibit the expression of .zeta.PKC for use as remedies in the treatment of arthritis, including, but not limited to, inhibitory polynucleotides and polypeptides, small molecules, and peptide inhibitors. In addition, the present invention provides pharmaceutical formulations and routes of administration for such remedies, as well as methods for assessing their efficacy.


Inventors: LaVallie; Edward R.; (Harvard, MA) ; Collins-Racie; Lisa A.; (Acton, MA) ; Arai; Maya; (Brookline, MA)
Correspondence Address:
    WYETH LLC;PATENT LAW GROUP
    5 GIRALDA FARMS
    MADISON
    NJ
    07940
    US
Assignee: WYETH
Madison
NJ

Family ID: 33479253
Appl. No.: 12/616484
Filed: November 11, 2009

Related U.S. Patent Documents

Application Number Filing Date Patent Number
10842142 May 10, 2004 7638482
12616484
60468987 May 8, 2003
60491274 Jul 31, 2003

Current U.S. Class: 435/6.16 ; 435/15; 435/29; 514/44A; 536/24.5
Current CPC Class: G01N 2333/91215 20130101; G01N 2800/102 20130101; G01N 2500/00 20130101; A61P 19/02 20180101; G01N 33/564 20130101; G01N 33/6872 20130101
Class at Publication: 435/6 ; 435/29; 435/15; 514/44.A; 536/24.5
International Class: C12Q 1/68 20060101 C12Q001/68; C12Q 1/02 20060101 C12Q001/02; C12Q 1/48 20060101 C12Q001/48; A61K 31/7088 20060101 A61K031/7088; C07H 21/02 20060101 C07H021/02

Claims



1. A method for use in the diagnosis of arthritis in a subject comprising the steps of: detecting a test amount of a .zeta.PKC gene product in a sample from the subject; and comparing the test amount with a normal amount of the .zeta.PKC gene product in a control sample, whereby a finding that the test amount is greater than the normal amount provides a positive indication in the diagnosis of arthritis.

2. The method of claim 1, wherein the sample comprises chondrocytes.

3. The method of claim 1, wherein the .zeta.PKC gene product comprises RNA or cDNA.

4. The method of claim 1, wherein the .zeta.PKC gene product is .zeta.PKC polypeptide.

5. A method for use in the prognosis of arthritis in a subject comprising the steps of: detecting a test amount of a .zeta.PKC gene product in a sample from the subject; and comparing the test amount with prognostic amounts of the .zeta.PKC gene product in control samples, whereby a comparison of the test amount with the prognostic amounts provides an indication of the prognosis of arthritis.

6. The method of claim 5, wherein the sample comprises chondrocytes.

7. The method of claim 5, wherein the .zeta.PKC gene product comprises RNA or cDNA.

8. The method of claim 5, wherein the .zeta.PKC gene product is .zeta.PKC polypeptide.

9. A method for use in monitoring the course of arthritis in a subject comprising the steps of: detecting a first test amount of a .zeta.PKC gene product in a sample from the subject at a first time; detecting a second test amount of the .zeta.PKC gene product in a sample from the subject at a second, later time; and comparing the first test amount and the second test amount, whereby an increase in the amount of the .zeta.PKC gene product in the second test amount as compared with the first test amount indicates progression of arthritis, and whereby a decrease in the amount of the PKC gene product in the second test amount as compared with the first test amount indicates remission of arthritis.

10. The method of claim 9, wherein the sample comprises chondrocytes.

11. The method of claim 9, wherein the PKC gene product comprises RNA or cDNA.

12. The method of claim 9, wherein the .zeta.PKC gene product is .zeta.PKC polypeptide.

13. A method for assessing the efficacy of a treatment for arthritis in a subject comprising the steps of: detecting a first test amount of a .zeta.PKC gene product in a sample from the subject prior to treatment; detecting a second test amount of the .zeta.PKC gene product in a sample from the subject after treatment; and comparing the first test amount and the second test amount, whereby a decrease in the amount of the .zeta.PKC gene product in the second test amount as compared with the first test amount indicates that the treatment for arthritis is efficacious.

14. The method of claim 13, wherein the sample comprises chondrocytes.

15. The method of claim 13, wherein the .zeta.PKC gene product comprises RNA or cDNA.

16. The method of claim 13, wherein the .zeta.PKC gene product is .zeta.PKC polypeptide.

17. A method of screening for a compound capable of inhibiting arthritis in a subject comprising the steps of: providing a first sample and a second sample containing equivalent amounts of .zeta.PKC; contacting the first sample with the compound; and determining whether the activity of .zeta.PKC in the first sample is decreased relative to the activity of .zeta.PKC in the second sample not contacted with the compound, whereby a decrease in the activity of .zeta.PKC in the first sample as compared with the second sample indicates that the compound inhibits arthritis in the subject.

18. The method of claim 17, wherein the compound inhibits the activity of PKC in chondrocytes.

19. The method of claim 17, wherein the compound is a small molecule.

20. The method of claim 17, wherein the activity of .zeta.PKC is determined by use of an enzymatic protein kinase assay.

21. The method of claim 17, wherein the activity of .zeta.PKC is determined by use of a chondrocyte pellet assay.

22. The method of claim 17, wherein the activity of .zeta.PKC is determined by use of an assay measuring proteoglycan degradation.

23. The method of claim 17, wherein the activity of .zeta.PKC is determined by use of an assay measuring NF-.kappa.B activity.

24. A method of screening for a compound capable of inhibiting arthritis in a subject comprising the steps of: providing a first sample and a second sample containing equivalent amounts of cells that express .zeta.PKC; contacting the first sample with the compound; and determining whether the expression of .zeta.PKC gene product in the first sample is decreased relative to the expression of .zeta.PKC gene product in the second sample not contacted with the compound, whereby a decrease in the expression of .zeta.PKC gene product in the first sample as compared with the second sample indicates that the compound inhibits arthritis in the subject.

25. The method of claim 24, wherein the compound inhibits the expression of PKC gene product in chondrocytes.

26. The method of claim 24, wherein the compound is a small molecule.

27. The method of claim 24, wherein the expression of .zeta.PKC gene product is determined by use of an enzymatic protein kinase assay.

28. The method of claim 24, wherein the expression of .zeta.PKC gene product is determined by use of a chondrocyte pellet assay.

29. The method of claim 24, wherein the expression of .zeta.PKC gene product is determined by use of an assay measuring proteoglycan degradation.

30. The method of claim 24, wherein the expression of .zeta.PKC gene product is determined by use of an assay measuring NF-.kappa.B activity.

31. A method for the treatment of arthritis in a subject comprising administering to the subject a compound that inhibits the activity of .zeta.PKC in the subject.

32. The method of claim 31, wherein the compound inhibits the activity of PKC in chondrocytes.

33. The method of claim 31, wherein the compound is an antisense polynucleotide.

34. The method of claim 31, wherein the compound is a small molecule.

35. The method of claim 31, wherein the compound is a siRNA molecule.

36. The method of claim 35, wherein the siRNA molecule is selected from the group consisting of siRNA molecules shown in FIG. 1.

37. A method for the treatment of arthritis in a subject comprising administering to the subject a compound that inhibits the expression of .zeta.PKC in the subject.

38. The method of claim 37, wherein the compound inhibits the expression of .zeta.PKC in chondrocytes.

39. The method of claim 37, wherein the compound is an antisense polynucleotide.

40. The method of claim 37, wherein the compound is a small molecule.

41. The method of claim 37, wherein the compound is a siRNA molecule.

42. The method of claim 41, wherein the siRNA molecule is selected from the group consisting of siRNA molecules shown in FIG. 1.

43. A siRNA molecule that inhibits the expression or activity of .zeta.PKC.

44. The siRNA molecule of claim 43, wherein the siRNA molecule is selected from the group consisting of siRNA molecules shown in FIG. 1.
Description



[0001] This application is a continuation of U.S. patent application Ser. No. 10/842,142, filed May 10, 2004, which claims the benefit of U.S. Provisional Application Ser. No. 60/468,987, filed May 8, 2003, and U.S. Provisional Application Ser. No. 60/491,274, filed Jul. 31, 2003, all of which are incorporated herein by reference in their entireties.

BACKGROUND OF THE INVENTION

[0002] 1. Field of the Invention

[0003] The present invention is directed to methods of diagnosing, prognosing, and monitoring the course of arthritis in a subject based on increased protein kinase C zeta (.zeta.PKC) gene expression in arthritic tissue. The present invention further provides compounds that inhibit the expression of .zeta.PKC for use as remedies in the treatment of arthritis.

[0004] 2. Related Background Art

[0005] Protein kinase C zeta (.zeta.PKC) is emerging as an important signal transduction component. There is growing literature suggesting that .zeta.PKC is involved in the NF-.kappa.B and AP-1 pathways. For example, a .zeta.PKC knockout mouse is fully viable but displays a phenotype reminiscent of the tumor necrosis factor (TNF) receptor and lymphotoxin receptor knockouts, with severe impairment of NF-.kappa.B-dependent transcriptional activity (Leitges et al. (2001) Mol. Cell. 8:771-80). Other investigators (Lallena et al. (1999) Mol. Cell. Biol. 19:2180-88) have shown a role for .zeta.PKC in activating I.kappa.B and, thereby, activating NF-.kappa.B.

[0006] NF-.kappa.B activation has been implicated in numerous inflammatory disorders, including asthma, inflammatory bowel disease, and arthritis (reviewed in Roshak et al. (2002) Curr. Opin. Pharmacol. 2:316-21). NF-.kappa.B has been shown to play an essential role in the secretion of various matrix metalloproteinases (MMPs) from various cell types (Bond et al. (1998) FEBS Lett. 435:29-34; Bond et al. (1999) Biochem. Biophys. Res. Commun. 264:561-67; Bond et al. (2001) Cardiovasc. Res. 50:556-65). In arthritis, cytokines such as TNF and interleukin-1 (IL-1) increase the production and synthesis of MMPs and other degradative enzymes above levels that can be naturally controlled, resulting in disease (reviewed in Smith (1999) Front. Biosci. 4:D704; Mort and Billington (2001) Arthritis Res. 3:337-41; Catterall and Cawston (2003) Arthritis Res. Ther. 5:12-24).

[0007] To date, there has been no direct evidence linking .zeta.PKC to arthritis. If PKC were expressed in affected tissues, however, it would help to explain the degradative actions of TNF and IL-1 by transducing the extracellular receptor binding of these factors to the intracellular induction of synthesis of degradative enzymes by NF-.kappa.B. In this regard, inhibitors of .zeta.PKC may block TNF and IL-1 action and serve as treatments for arthritis and other inflammatory diseases. Such .zeta.PKC inhibitors should be more efficacious than traditional cytokine and MMP inhibitors because they should ultimately affect more than just one target (Roshak, supra; Smith, supra). Such .zeta.PKC inhibitors should also be safer than NF-.kappa.B inhibitors because .zeta.PKC is only one of many effectors in the NF-.kappa.B pathway.

SUMMARY OF THE INVENTION

[0008] The present invention is based on the discovery that .kappa.PKC expression is increased in the tissues of arthritis patients as compared to normal individuals. The present invention provides compounds that inhibit the expression of .zeta.PKC in arthritic tissue including, but not limited to, inhibitory polynucleotides and polypeptides, small molecules, and peptide inhibitors. The present invention further provides methods of diagnosing, prognosing, and monitoring the course of arthritis based on aberrant .zeta.PKC gene expression in arthritic tissue, as well as therapies for use as remedies for such aberrant expression. In addition, the present invention provides pharmaceutical formulations and routes of administration for such remedies, as well as methods for assessing the efficacy of such remedies.

[0009] In one embodiment, the invention provides a method for use in the diagnosis of arthritis in a subject comprising the steps of detecting a test amount of a .zeta.PKC gene product in a sample from the subject; and comparing the test amount with a normal amount of the .zeta.PKC gene product in a control sample, whereby a finding that the test amount is greater than the normal amount provides a positive indication in the diagnosis of arthritis. In a preferred embodiment, the sample comprises chondrocytes. In some other preferred embodiments, the .zeta.PKC gene product comprises RNA or cDNA, or is .zeta.PKC polypeptide.

[0010] In another embodiment, the invention provides a method for use in the prognosis of arthritis in a subject comprising the steps of detecting a test amount of a .zeta.PKC gene product in a sample from the subject; and comparing the test amount with prognostic amounts of the .zeta.PKC gene product in control samples, whereby a comparison of the test amount with the prognostic amounts provides an indication of the prognosis of arthritis. In a preferred embodiment, the sample comprises chondrocytes. In some other preferred embodiments, the .zeta.PKC gene product comprises RNA or cDNA, or is .zeta.PKC polypeptide.

[0011] In another embodiment, the invention provides a method for use in monitoring the course of arthritis in a subject comprising the steps of detecting a first test amount of a .zeta.PKC gene product in a sample from the subject at a first time; detecting a second test amount of the .zeta.PKC gene product in a sample from the subject at a second, later time; and comparing the first test amount and the second test amount, whereby an increase in the amount of the .zeta.PKC gene product in the second test amount as compared with the first test amount indicates progression of arthritis, and whereby a decrease in the amount of the .zeta.PKC gene product in the second test amount as compared with the first test amount indicates remission of arthritis. In a preferred embodiment, the sample comprises chondrocytes. In some other preferred embodiments, the .zeta.PKC gene product comprises RNA or cDNA, or is .zeta.PKC polypeptide.

[0012] In another embodiment, the invention provides a method for assessing the efficacy of a treatment for arthritis in a subject comprising the steps of detecting a first test amount of a .zeta.PKC gene product in a sample from the subject prior to treatment; detecting a second test amount of the .zeta.PKC gene product in a sample from the subject after treatment; and comparing the first test amount and the second test amount, whereby a decrease in the amount of the .zeta.PKC gene product in the second test amount as compared with the first test amount indicates that the treatment for arthritis is efficacious. In a preferred embodiment, the sample comprises chondrocytes. In some other preferred embodiments, the .zeta.PKC gene product comprises RNA or cDNA, or is .zeta.PKC polypeptide.

[0013] In another embodiment, the invention provides a method of screening for a compound capable of inhibiting arthritis in a subject comprising the steps of providing a first sample and a second sample containing equivalent amounts of .zeta.PKC; contacting the first sample with the compound; and determining whether the activity of .zeta.PKC in the first sample is decreased relative to the activity of .zeta.PKC in the second sample not contacted with the compound, whereby a decrease in the activity of .zeta.PKC in the first sample as compared with the second sample indicates that the compound inhibits arthritis in the subject. In a preferred embodiment, the compound inhibits the activity of .zeta.PKC in chondrocytes. In another preferred embodiment, the compound is a small molecule. In other preferred embodiments, the activity of .zeta.PKC is determined by use of an enzymatic protein kinase assay, a chondrocyte pellet assay, an assay measuring proteoglycan degradation, or an assay measuring NF-.kappa.B activity.

[0014] In another embodiment, the invention provides a method of screening for a compound capable of inhibiting arthritis in a subject comprising the steps of providing a first sample and a second sample containing equivalent amounts of cells that express .zeta.PKC; contacting the first sample with the compound; and determining whether the expression of .zeta.PKC gene product in the first sample is decreased relative to the expression of .zeta.PKC gene product in the second sample not contacted with the compound, whereby a decrease in the expression of .zeta.PKC gene product in the first sample as compared with the second sample indicates that the compound inhibits arthritis in the subject. In a preferred embodiment, the compound inhibits the expression of .zeta.PKC gene product in chondrocytes. In another preferred embodiment, the compound is a small molecule. In other preferred embodiments, the expression of .zeta.PKC gene product is determined by use of an enzymatic protein kinase assay, a chondrocyte pellet assay, an assay measuring proteoglycan degradation, or an assay measuring NF-.kappa.B activity.

[0015] In another embodiment, the invention provides a method for the treatment of arthritis in a subject comprising administering to the subject a compound that inhibits the activity of .zeta.PKC in the subject. In a preferred embodiment, the compound inhibits the activity of .zeta.PKC in chondrocytes. In another preferred embodiment, the compound is an antisense polynucleotide. In another preferred embodiment, the compound is a small molecule. In another preferred embodiment, the compound is a siRNA molecule. In a further preferred embodiment, the siRNA molecule is selected from the group consisting of siRNA molecules shown in FIG. 1.

[0016] In another embodiment, the invention provides a method for the treatment of arthritis in a subject comprising administering to the subject a compound that inhibits the expression of .zeta.PKC in the subject. In a preferred embodiment, the compound inhibits the expression of .zeta.PKC in chondrocytes. In another preferred embodiment, the compound is an antisense polynucleotide. In another preferred embodiment, the compound is a small molecule. In another preferred embodiment, the compound is a siRNA molecule. In a further preferred embodiment, the siRNA molecule is selected from the group consisting of siRNA molecules shown in FIG. 1.

[0017] In another embodiment, the invention provides a siRNA molecule that inhibits the expression or activity of .zeta.PKC. In a preferred embodiment, the siRNA molecule is selected from the group consisting of siRNA molecules shown in FIG. 1.

BRIEF DESCRIPTION OF THE DRAWINGS

[0018] FIG. 1 shows preferred siRNA molecules targeted to human .zeta.PKC mRNA for use in RNAi. Target segments [SEQ ID NOs:9-20; 45-59; 90-109; and 150-154] of the .zeta.PKC transcripts are grouped according to their first two nucleotides (AA, CA, GA, or TA) and are shown in the 5'->3' orientation. "GC Ratio" refers to the percentage of total G+C nucleotides in each target segment; "Position" refers to the nucleotide position in the human .zeta.PKC cDNA (SEQ ID NO:1) immediately preceding the beginning of each target segment. Preferred siRNA molecules (siRNA duplexes) are shown on the right side of the figure. Both the sense strand for each siRNA duplex [SEQ ID NOs:21-32; 60-74; 110-129; and 155-159] and the corresponding antisense strand [SEQ ID NOs:33-44; 75-89; 130-149; and 160-164] are shown in the 5'->3' orientation. For example, the siRNA molecule directed to the first target segment presented in the figure (i.e., SEQ ID NO:9) is the siRNA duplex of the sense and antisense strands identified (i.e., SEQ ID NO:21 and SEQ ID NO:33, respectively).

[0019] FIG. 2 is a graph depicting the effects of the NF-.kappa.B blocker SN50 (300 .mu.g/ml), or its inactive analog SN50M (300 .mu.g/ml), on TNF- or IL-1-mediated proteoglycan degradation in primary bovine chondrocytes in culture. The top panel shows proteoglycan content released in the media (.mu.g/0.5 ml); the bottom panel shows proteoglycan content retained in the cell pellet (.mu.g/ml).

[0020] FIG. 3 is a graph depicting the effects of a myristoylated .zeta.PKC pseudosubstrate peptide (2089) or .zeta.PKC small molecule inhibitor Ro-31-8220 (RO31) on TNF-mediated proteoglycan degradation in primary bovine chondrocytes in culture. The top panel shows proteoglycan content released in the media (.mu.g/0.5 ml); the bottom panel shows proteoglycan content retained in the cell pellet (.mu.g/ml).

[0021] FIG. 4 is a graph depicting the dose-dependent effects of a myristoylated .zeta.PKC pseudosubstrate peptide (2089) on TNF- or IL-1-mediated proteoglycan degradation in primary bovine chondrocytes in culture. The top panel shows proteoglycan content released in the media (.mu.g/0.5 ml); the bottom panel shows proteoglycan content retained in the cell pellet (.mu.g/ml).

[0022] FIG. 5 shows that .zeta.PKC is upregulated in human osteoarthritic articular cartilage. Panel A shows .zeta.PKC mRNA levels using the HG-U95Av2 Affymetrix GeneChip.RTM. Array; panel B shows .zeta.PKC mRNA levels using TaqMan PCR analysis.

[0023] FIG. 6 shows that adenoviral-mediated expression of .zeta.PKC increases proteoglycan degradation. Panel A shows proteoglycan released into the media in the chondrocyte pellet assay in response to overexpression of .zeta.PKC and GFP; panel B shows the effects of stimulation with suboptimal levels of the cytokine TNF.alpha..

[0024] FIG. 7 demonstrates that .zeta.PKC is responsible for TNF.alpha.-mediated proteoglycan release in articular chondrocytes. TNF.alpha. was added (100 ng/ml; denoted by *) to some cultures in the chondrocyte pellet assay. Two inhibitors were added at various doses: bisindolylmaleimide (BIS), a pan-PKC inhibitor; and chelerythrine chloride (CC), a competitive inhibitor of the phorbol ester-binding site that does not inhibit .zeta.PKC. Proteoglycan release into the media is shown on the y-axis as .mu.g/ml.

[0025] FIG. 8 shows the effects of the inhibitors BIS and CC on TNF.alpha.-induced activation of NF-.kappa.B. Activation of NF-.kappa.B was measured in an immortalized human chondrocyte cell line into which a luciferase reporter gene under the control of an NF-.kappa.B response element was introduced; activity (i.e., units on the y-axis) is expressed as "relative luciferase activity."

DETAILED DESCRIPTION OF THE INVENTION

[0026] We have discovered that .zeta.PKC expression is upregulated in the tissues of arthritis patients as compared to normal individuals. The discovery that this enzyme is upregulated in arthritic tissue enables methods for diagnosing arthritis by detecting an increase in .zeta.PKC expression and methods for treating arthritis by downregulating .zeta.PKC expression. In addition, this discovery enables the identification of new .zeta.PKC inhibitors useful in the treatment of arthritis.

Methods for Diagnosing, Prognosing, and Monitoring the Progress of Arthritis

Introduction

[0027] The present invention provides methods for diagnosing arthritis by detecting the upregulation of .zeta.PKC. "Diagnostic" or "diagnosing" means identifying the presence or absence of a pathologic condition. Diagnostic methods involve detecting upregulation of .zeta.PKC by determining a test amount of .zeta.PKC gene product (e.g., mRNA, cDNA, or polypeptide, including fragments thereof) in a biological sample from a subject (human or nonhuman mammal), and comparing the test amount with a normal amount or range (i.e., an amount or range from an individual(s) known not to suffer from arthritis) for the .zeta.PKC gene product. While a particular diagnostic method may not provide a definitive diagnosis of arthritis, it suffices if the method provides a positive indication that aids in diagnosis.

[0028] The present invention also provides methods for prognosing arthritis by detecting the upregulation of .zeta.PKC. "Prognostic" or "prognosing" means predicting the probable development and/or severity of a pathologic condition. Prognostic methods involve determining the test amount of a .zeta.PKC gene product in a biological sample from a subject, and comparing the test amount to a prognostic amount or range (i.e., an amount or range from individuals with varying severities of arthritis) for the .zeta.PKC gene product. Various amounts of the .zeta.PKC gene product in a test sample are consistent with certain prognoses for arthritis. The detection of an amount of .zeta.PKC gene product at a particular prognostic level provides a prognosis for the subject.

[0029] The present invention also provides methods for monitoring the course of arthritis by detecting the upregulation of .zeta.PKC. Monitoring methods involve determining the test amounts of a .zeta.PKC gene product in biological samples taken from a subject at a first and second time, and comparing the amounts. A change in amount of .zeta.PKC gene product between the first and second time indicates a change in the course of arthritis, with a decrease in amount indicating remission of arthritis, and an increase in amount indicating progression of arthritis. Such monitoring assays are also useful for evaluating the efficacy of a particular therapeutic intervention (e.g., disease attenuation vs. reversal) in patients being treated for arthritis.

Biological Sample Collection

[0030] Increased expression of .zeta.PKC can be detected in a variety of biological samples, including cells (e.g., whole cells, cell fractions, and cell extracts) and tissues. Biological samples also include sections of tissue such as biopsies and frozen sections taken for histological purposes. Preferred biological samples include articular cartilage (i.e., chondrocytes), synovium, and synovial fluid.

Normal, Diagnostic, and Prognostic Values

[0031] In the diagnostic and prognostic assays of the present invention, the .zeta.PKC gene product is detected and quantified to yield a test amount. The test amount is then compared to a normal amount or range. An amount above the normal amount or range (e.g., a 30% or greater increase (with p<0.01), or a 100% or greater increase (with p<0.05)) is a positive sign in the diagnosis of arthritis. Particular methods of detection and quantitation of .zeta.PKC gene products are described below.

[0032] Normal amounts or baseline levels of .zeta.PKC gene products can be determined for any particular sample type and population. Generally, baseline (normal) levels of .zeta.PKC protein or mRNA are determined by measuring the amount of .zeta.PKC protein or mRNA in a biological sample type from normal (i.e., healthy) subjects. Alternatively, normal values of .zeta.PKC gene product can be determined by measuring the amount in healthy cells or tissues taken from the same subject from which the diseased (or possibly diseased) test cells or tissues were taken. The amount of .zeta.PKC gene product (either the normal amount or the test amount) can be determined or expressed on a per cell, per total protein, or per volume basis. To determine the cell amount of a sample, one can measure the level of a constitutively expressed gene product or other gene product expressed at known levels in cells of the type from which the biological sample was taken.

[0033] It will be appreciated that the assay methods of the present invention do not necessarily require measurement of absolute values of .zeta.PKC gene product because relative values are sufficient for many applications of these methods. It will also be appreciated that in addition to the quantity or abundance of .zeta.PKC gene products, variant or abnormal .zeta.PKC gene products or their expression patterns (e.g., mutated transcripts, truncated polypeptides) may be identified by comparison to normal gene products and expression patterns.

Assays for .zeta.PKC Gene Products

[0034] The diagnostic, prognostic, and monitoring assays of the present invention involve detecting and quantifying .zeta.PKC gene products in biological samples. .zeta.PKC gene products include, for example, .zeta.PKC mRNA and .zeta.PKC polypeptide, and both can be measured using methods well known to those skilled in the art.

[0035] For example, .zeta.PKC mRNA can be directly detected and quantified using hybridization-based assays, such as Northern hybridization, in situ hybridization, dot and slot blots, and oligonucleotide arrays. Hybridization-based assays refer to assays in which a probe nucleic acid is hybridized to a target nucleic acid. In some formats, the target, the probe, or both are immobilized. The immobilized nucleic acid may be DNA, RNA, or another oligonucleotide or polynucleotide, and may comprise naturally or nonnaturally occurring nucleotides, nucleotide analogs, or backbones. Methods of selecting nucleic acid probe sequences for use in the present invention are based on the nucleic acid sequence of .zeta.PKC and are well known in the art.

[0036] Alternatively, .zeta.PKC mRNA can be amplified before detection and quantitation. Such amplification-based assays are well known in the art and include polymerase chain reaction (PCR), reverse-transcription-PCR(RT-PCR), PCR-enzyme-linked immunosorbent assay (PCR-ELISA), and ligase chain reaction (LCR). Primers and probes for producing and detecting amplified .zeta.PKC gene products (e.g., mRNA or cDNA) may be readily designed and produced without undue experimentation by those of skill in the art based on the nucleic acid sequence of .zeta.PKC. Amplified .zeta.PKC gene products may be directly analyzed, e.g., by gel electrophoresis; by hybridization to a probe nucleic acid; by sequencing; by detection of a fluorescent, phosphorescent, or radioactive signal; or by any of a variety of well-known methods. In addition, methods are known to those of skill in the art for increasing the signal produced by amplification of target nucleic acid sequences. One of skill in the art will recognize that whichever amplification method is used, a variety of quantitative methods known in the art (e.g., quantitative PCR) may be used if quantitation of .zeta.PKC gene products is desired.

[0037] .zeta.PKC polypeptide (or fragments thereof) can be detected and quantified using various well-known enzymatic and immunological assays. Enzymatic assays refer to assays that utilize .zeta.PKC substrates to detect protein kinase activity. Various natural and artificial substrates useful for detecting and quantifying .zeta.PKC activity are known, and include myristoyl alanine-rich C kinase substrate (MARCKS) peptide (Herget et al. (1995) Eur. J. Biochem. 233:448-57), p47phox (Dang et al. (2001) J. Immunol. 166:1206-13), myelin basic protein (Kim et al. (2002) J. Biol. Chem. 277:30375-81), protamine sulfate (McGlynn et al. (1992) J. Cell. Biochem. 49:239-50), nucleolin (Zhou et al. (1997) J. Biol. Chem. 272:31130-37); heterogeneous ribonucleoprotein AI (hnRNPA1) (Municio et al. (1995) J. Biol. Chem. 270:15884-91), .zeta.PKC-derived peptide (Kochs et al. (1993) Eur. J. Biochem. 216:597-606), and .zeta.PKC-derived peptide (Standaert et al. (1999) J. Biol. Chem. 274:14074-78). Numerous enzymatic assay protocols (radioactive and nonradioactive) suitable for detecting and quantifying .zeta.PKC activity are described in the literature and/or are commercially available in kit form from, e.g., PanVera (Madison, Wis.), Promega (Madison, Wis.), Transbio (Baltimore, Md.), Upstate (Waltham, Mass.), and Research & Diagnostic Antibodies (Benicia, Calif.).

[0038] Immunological assays refer to assays that utilize an antibody (e.g., polyclonal, monoclonal, chimeric, humanized, scFv, and fragments thereof) that specifically binds to .zeta.PKC polypeptide (or a fragment thereof). A number of well-established immunological assays suitable for the practice of the present invention are known, and include ELISA, radioimmunoassay (RIA), immunoprecipitation, immunofluorescence, and Western blotting.

[0039] The anti-.zeta.PKC antibodies (preferably anti-mammalian .zeta.PKC; more preferably anti-human .zeta.PKC) to be used in the immunological assays of the present invention are commercially available from, e.g., Sigma-Aldrich (St. Louis, Mo.), Upstate (Waltham, Mass.), and Research Diagnostics (Flanders, N.J.). Alternatively, anti-.zeta.PKC antibodies may be produced by methods well known to those skilled in the art. For example, monoclonal antibodies to .zeta.PKC (preferably mammalian; more preferably human (e.g., GenBank Acc. No. Q05513; SEQ ID NO:2)) can be produced by generation of hybridomas in accordance with known methods. Hybridomas formed in this manner are then screened using standard methods, such as ELISA, to identify one or more hybridomas that produce an antibody that specifically binds to .zeta.PKC. Full-length .zeta.PKC may be used as the immunogen, or, alternatively, antigenic peptide fragments of .zeta.PKC may be used.

[0040] As an alternative to preparing monoclonal antibody-secreting hybridomas, a monoclonal antibody to .zeta.PKC may be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) to thereby isolate immunoglobulin library members that bind to .zeta.PKC. Kits for generating and screening phage display libraries are commercially available from, e.g., Dyax Corp. (Cambridge, Mass.) and Maxim Biotech (South San Francisco, Calif.). Additionally, examples of methods and reagents particularly amenable for use in generating and screening antibody display libraries can be found in the literature.

[0041] Polyclonal sera and antibodies may be produced by immunizing a suitable subject, such as a rabbit, with .zeta.PKC (preferably mammalian; more preferably human) or an antigenic fragment thereof. The antibody titer in the immunized subject may be monitored over time by standard techniques, such as with ELISA, using immobilized marker protein. If desired, the antibody molecules directed against .zeta.PKC may be isolated from the subject or culture media and further purified by well-known techniques, such as protein A chromatography, to obtain an IgG fraction.

[0042] Fragments of antibodies to .zeta.PKC may be produced by cleavage of the antibodies in accordance with methods well known in the art. For example, immunologically active F(ab') and F(ab').sub.2 fragments may be generated by treating the antibodies with an enzyme such as pepsin. Additionally, chimeric, humanized, and single-chain antibodies to .zeta.PKC, comprising both human and nonhuman portions, may be produced using standard recombinant DNA techniques. Humanized antibodies to .zeta.PKC may also be produced using transgenic mice that are incapable of expressing endogenous immunoglobulin heavy and light chain genes, but which can express human heavy and light chain genes.

[0043] In the immunological assays of the present invention, the .zeta.PKC polypeptide is typically detected directly (i.e., the anti-.zeta.PKC antibody is labeled) or indirectly (i.e., a secondary antibody that recognizes the anti-.zeta.PKC antibody is labeled) using a detectable label. The particular label or detectable group used in the assay is usually not critical, as long as it does not significantly interfere with the specific binding of the antibodies used in the assay.

[0044] The immunological assays of the present invention may be competitive or noncompetitive. In competitive assays, the amount of .zeta.PKC in a sample is measured indirectly by measuring the amount of added (exogenous) .zeta.PKC displaced from a capture agent (i.e., an anti-.zeta.PKC antibody) by the .zeta.PKC in the sample. In noncompetitive assays, the amount of .zeta.PKC in a sample is directly measured. In a preferred noncompetitive "sandwich" assay, the capture agent (e.g., a first anti-.zeta.PKC antibody) is bound directly to a solid support (e.g., membrane, microtiter plate, test tube, dipstick, glass or plastic bead) where it is immobilized. The immobilized agent then captures any .zeta.PKC polypeptide present in the sample. The immobilized .zeta.PKC can then be detected using a second labeled anti-.zeta.PKC antibody. Alternatively, the second anti-.zeta.PKC antibody can be detected using a labeled secondary antibody that recognizes the second anti-.zeta.PKC antibody.

Screening Methods for Identifying Compounds that Inhibit .zeta.PKC Expression and/or Activity

Introduction

[0045] The present invention provides methods (also referred to herein as "screening assays") for identifying novel compounds (e.g., small molecules) that inhibit expression of .zeta.PKC in arthritic tissue. In one embodiment, cells that express PKC (either naturally or recombinantly) are contacted with a test compound to determine whether the compound inhibits expression of a .zeta.PKC gene product (e.g., mRNA or polypeptide), with a decrease in expression (as compared to an untreated sample of cells) indicating that the compound inhibits .zeta.PKC in arthritic tissue. Changes in .zeta.PKC gene expression can be determined by any method known in the art or described above. In a preferred embodiment, cells transfected with a reporter construct comprising a marker gene (e.g., luciferase or green fluorescent protein (GFP)) downstream of a NF-.kappa.B binding site are contacted with a test compound to determine whether the compound can inhibit expression of the marker protein when the cells are treated with cytokines. Compounds identified that inhibit .zeta.PKC or marker protein expression are candidates as drugs for the prophylactic and therapeutic treatment of arthritis.

[0046] Alternatively, compounds can be identified that inhibit the kinase activity of .zeta.PKC in vitro using assays described previously. Purified (or partially purified) .zeta.PKC is contacted with a test compound to determine whether the compound inhibits the kinase activity of .zeta.PKC (as compared to an untreated sample of enzyme). Compounds identified that inhibit .zeta.PKC activity could then be tested in in vitro and in vivo models of arthritis. Several in vitro models are described in the Examples below. In vivo models of arthritis include, but are not limited to, the anterior cruciate ligament resection models in the dog and rabbit, and the partial meniscectomy models in the rabbit and mouse. Exemplary methods and assays for directly and indirectly measuring the activity of .zeta.PKC and/or for determining inhibition of the activity of .zeta.PKC include, but are not limited to, enzymatic protein kinase activity assays (as detailed above), chondrocyte pellet assays, assays measuring proteoglycan degradation, and assays measuring NF-.kappa.B activity.

Sources of .zeta.PKC

[0047] The .zeta.PKC (preferably mammalian; more preferably human (e.g., GenBank Acc. No. Q05513; SEQ ID NO:2)) to be used in the screening assays of the current invention are commercially available from, e.g., Sigma-Aldrich, (St. Louis, Mo.), Research Diagnostics (Flanders, N.J.), ProQinase (Freiburg, Germany), and PanVera (Madison, Wis.). Alternatively, .zeta.PKC can be purified or partially purified from various tissues (preferably mammalian; more preferably human), including brain, placenta, testes and lung, using known purification processes such as gel filtration and ion exchange chromatography. Purification may also include affinity chromatography with agents known to bind .zeta.PKC (e.g., anti-.zeta.PKC antibodies). These purification processes may also be used to purify .zeta.PKC from recombinant sources.

[0048] Polynucleotides encoding .zeta.PKC (or enzymatic portions thereof) may be operably linked to an appropriate expression control sequence for recombinant production of .zeta.PKC. The .zeta.PKC polynucleotides are preferably of mammalian origin (e.g., mouse .zeta.PKC cDNA (GenBank Acc. No. M94632); rat .zeta.PKC cDNA (GenBank Acc. No. J04532); rabbit .zeta.PKC cDNA (GenBank Acc. No. U78768)), and more preferably of human origin (e.g., human .zeta.PKC cDNA (GenBank Acc. No. NM.sub.--002744; SEQ ID NO:1)). General methods for expressing these recombinant .zeta.PKC polynucleotides are well known in the art.

[0049] A number of cell lines may act as suitable host cells for recombinant expression of .zeta.PKC. Mammalian host cell lines include, for example, COS cells, CHO cells, 293 cells, A431 cells, 3T3 cells, CV-1 cells, HeLa cells, L cells, BHK21 cells, HL-60 cells, U937 cells, HaK cells, and Jurkat cells, as well as normal diploid cells, cell strains derived from in vitro culture of primary tissue, and primary explants.

[0050] Alternatively, .zeta.PKC (or enzymatic portions thereof) may be recombinantly produced in lower eukaryotes such as yeast or in prokaryotes. Potentially suitable yeast strains include Saccharomyces cerevisiae, Schizosaccharomyces pombe, Kluyveromyces strains, and Candida strains. Potentially suitable bacterial strains include Escherichia coli, Bacillus subtilis, and Salmonella typhimurium. If the polypeptides of the present invention are made in yeast or bacteria, it may be necessary to modify them by, for example, phosphorylation or glycosylation of appropriate sites, in order to obtain functionality. Such covalent attachments may be accomplished using well-known chemical or enzymatic methods.

[0051] .zeta.PKC (or enzymatic portions thereof) may also be recombinantly produced using insect expression vectors, such as baculovirus vectors, and employing an insect cell expression system. Materials and methods for baculovirus/Sf9 expression systems are commercially available in kit form (e.g., the MaxBac.RTM. kit, Invitrogen, Carlsbad, Calif.).

[0052] In order to facilitate purification, .zeta.PKC (or enzymatic portions thereof) may be recombinantly expressed as fusions with proteins such as maltose-binding protein (MBP), glutathione-S-transferase (GST), or thioredoxin (TRX). Kits for expression and purification of such fusion proteins are commercially available from New England BioLabs (Beverly, Mass.), Pharmacia (Piscataway, N.J.), and Invitrogen (Carlsbad, Calif.), respectively. .zeta.PKC can also be tagged with a small epitope and subsequently identified or purified using a specific antibody to the epitope. One such epitope is the FLAG epitope, which is commercially available from Eastman Kodak (New Haven, Conn.).

[0053] .zeta.PKC (or enzymatic portions thereof) may also be produced by known conventional chemical synthesis. Methods for chemically synthesizing polypeptides are well known to those skilled in the art. Such chemically synthetic .zeta.PKC should possess biological properties in common with the naturally produced form, and thus can be employed as a biologically active or immunological substitute for natural .zeta.PKC.

Sources and Screening of Test Compounds

[0054] The test compounds of the present invention may be obtained from a number of sources. For example, combinatorial libraries of molecules are available for screening. Using such libraries, thousands of molecules can be screened for inhibitory activity. Preparation and screening of compounds can be screened as described above or by other methods well known to those of skill in the art. The compounds thus identified can serve as conventional "lead compounds" or can be used as the actual therapeutics.

Methods of Treatment

Introduction

[0055] The present invention provides both prophylactic and therapeutic methods for the treatment of arthritis by inhibiting expression and/or activity of .zeta.PKC. The methods involve contacting cells (either in vitro, in vivo, or ex vivo) with an agent in an amount effective to inhibit expression and/or activity of .zeta.PKC. The agent can be any molecule that inhibits expression and/or activity of .zeta.PKC, including, but not limited to, inhibitory polynucleotides, small molecules, inhibitory protein biologics, and peptide inhibitors.

Inhibitory Polynucleotides

[0056] Decreased expression of .zeta.PKC in an organism afflicted with (or at risk for) arthritis, or in an involved cell from such an organism, may be achieved through the use of various inhibitory polynucleotides, such as antisense polynucleotides and ribozymes, that bind and/or cleave the mRNA transcribed from the .zeta.PKC gene (e.g., Galderisi et al. (1999) J. Cell Physiol. 181:251-57; Sioud (2001) Curr. Mol. Med. 1:575-88).

[0057] The antisense polynucleotides or ribozymes of the invention can be complementary to an entire coding strand of .zeta.PKC, or to a portion thereof. Alternatively, antisense polynucleotides or ribozymes can be complementary to a noncoding region of the coding strand of .zeta.PKC. The antisense polynucleotides or ribozymes can be constructed using chemical synthesis and enzymatic ligation reactions using procedures well known in the art. The nucleoside linkages of chemically synthesized polynucleotides can be modified to enhance their ability to resist nuclease-mediated degradation, as well as to increase their sequence specificity. Such linkage modifications include, but are not limited to, phosphorothioate, methylphosphonate, phosphoroamidate, boranophosphate, morpholino, and peptide nucleic acid (PNA) linkages (Galderisi et al., supra; Heasman (2002) Dev. Biol. 243:209-14; Micklefield (2001) Curr. Med. Chem. 8:1157-79). Alternatively, these molecules can be produced biologically using an expression vector into which a polynucleotide of the present invention has been subcloned in an antisense (i.e., reverse) orientation.

[0058] The inhibitory polynucleotides of the present invention also include triplex-forming oligonucleotides (TFOs) which bind in the major groove of duplex DNA with high specificity and affinity (Knauert and Glazer (2001) Hum. Mol. Genet. 10:2243-51). Expression of .zeta.PKC can be inhibited by targeting TFOs complementary to the regulatory regions of the .zeta.PKC gene (i.e., the promoter and/or enhancer sequences) to form triple helical structures that prevent transcription of the .zeta.PKC gene.

[0059] In a preferred embodiment, the inhibitory polynucleotides of the present invention are short interfering RNA (siRNA) molecules. siRNA molecules are short (preferably 19-25 nucleotides; most preferably 19 or 21 nucleotides), double-stranded RNA molecules that cause sequence-specific degradation of target mRNA. This degradation is known as RNA interference (RNAi) (e.g., Bass (2001) Nature 411:428-29). Originally identified in lower organisms, RNAi has been effectively applied to mammalian cells and has recently been shown to prevent fulminant hepatitis in mice treated with siRNAs targeted to Fas mRNA (Song et al. (2003) Nature Med. 9:347-51). In addition, intrathecally delivered siRNA has recently been reported to block pain responses in two models (agonist-induced pain model and neuropathic pain model) in the rat (Dorn et al. (2004) Nucleic Acids Res. 32(5):e49).

[0060] The siRNA molecules of the present invention can be generated by annealing two complementary single-stranded RNA molecules together (one of which matches a portion of the target mRNA) (Fire et al., U.S. Pat. No. 6,506,559) or through the use of a single hairpin RNA molecule that folds back on itself to produce the requisite double-stranded portion (Yu et al. (2002) Proc. Natl. Acad. Sci. USA 99:6047-52). The siRNA molecules can be chemically synthesized (Elbashir et al. (2001) Nature 411:494-98) or produced by in vitro transcription using single-stranded DNA templates (Yu et al., supra). Alternatively, the siRNA molecules can be produced biologically, either transiently (Yu et al., supra; Sui et al. (2002) Proc. Natl. Acad. Sci. USA 99:5515-20) or stably (Paddison et al. (2002) Proc. Natl. Acad. Sci. USA 99:1443-48), using an expression vector(s) containing the sense and antisense siRNA sequences. Recently, reduction of levels of target mRNA in primary human cells, in an efficient and sequence-specific manner, was demonstrated using adenoviral vectors that express hairpin RNAs, which are further processed into siRNAs (Arts et al. (2003) Genome Res. 13:2325-32).

[0061] The siRNA molecules targeted to the polynucleotides of the present invention can be designed based on criteria well known in the art (e.g., Elbashir et al. (2001) EMBO J. 20:6877-88). For example, the target segment of the target mRNA preferably should begin with AA (most preferred), TA, GA, or CA; the GC ratio of the siRNA molecule preferably should be 45-55%; the siRNA molecule preferably should not contain three of the same nucleotides in a row; the siRNA molecule preferably should not contain seven mixed G/Cs in a row; and the target segment preferably should be in the ORF region of the target mRNA and preferably should be at least 75 by after the initiation ATG and at least 75 by before the stop codon. Based on these criteria, preferred siRNA molecules of the present invention, targeted to human .zeta.PKC mRNA, have been designed and are shown in FIG. 1. Other siRNA molecules targeted to .zeta.PKC mRNAs can be designed by one of ordinary skill in the art using the aforementioned criteria or other known criteria (e.g., Reynolds et al. (2004) Nature Biotechnol. 22:326-30).

Small Molecules

[0062] Decreased expression of .zeta.PKC in an organism afflicted with (or at risk for) arthritis, or in an involved cell from such an organism, may also be achieved through the use of small molecules (usually organic small molecules) that bind to and inhibit the activity of .zeta.PKC. Small molecules known to inhibit the activity of PKC (preferably isoform specific) can be used in the treatment methods of the present invention. Numerous small molecules that inhibit PKC are known in the art (including ones approved for treatment of disease, as well as others in clinical trials), and include both natural (e.g., staurosporine) and artificial (e.g., LY333531) compounds (reviewed in Goekjian and Jirousek (2001) Expert. Opin. Investing. Drugs 10:2117-40; Way et al. (2000) Trends Pharmacol. Sci. 21:181-87, both of which are incorporated by reference herein). These molecules can be used directly or can serve as starting compounds for the development of improved .zeta.PKC inhibitors (preferably isoform specific). Alternatively, novel small molecules (preferably isoform specific) identified by the screening methods described above may be used.

Inhibitory Protein Biologics

[0063] Decreased activity of .zeta.PKC in an organism afflicted with (or at risk for) arthritis, or in an involved cell from such an organism, may also be achieved using protein biologics Inhibitory protein biologics refer to protein molecules having inhibitory biological activity in a cell or organism. Preferred inhibitory protein biologics for use in the treatment methods of the present invention include Par4 and kinase-defective dominant-negative (DN) mutant forms of .zeta.PKC. Par4 is a naturally occurring protein that binds to .zeta.PKC, which serves to inhibit its enzymatic function (Diaz-Meco et al. (1996) Cell 86:777-86). DN mutant forms of .zeta.PKC, such as rat .zeta.PKC with a lysine 281 to tryptophan point mutation (Bandyopadhyay et al. (1997) J. Biol. Chem. 272:2551-58), reduce the activity of endogenous .zeta.PKC by competing for substrate and can be made using well-known site-directed mutagenesis techniques. Any variant of .zeta.PKC that lacks kinase activity but still inhibits .zeta.PKC-mediated signal transduction may be used as a DN mutant. These inhibitory protein biologics may be generated in cells (preferably chondrocytes) in situ using the above-described expression techniques.

Peptide Inhibitors

[0064] Decreased activity of .zeta.PKC in an organism afflicted with (or at risk for) arthritis, or in an involved cell from such an organism, may also be achieved using peptide inhibitors that bind to and inhibit the activity of .zeta.PKC. Peptide inhibitors include peptide pseudosubstrates that prevent .zeta.PKC from interacting with its substrates, as well as peptides that bind to either .zeta.PKC or its substrates and block PKC-mediated phosphorylation. Peptide inhibitors that inhibit .zeta.PKC are known in the literature and include SIYRRGARRWRKL (SEQ ID NO:3), SIYRRGARRWRKLYRAN (SEQ ID NO:4), and RRGARRWRK (SEQ ID NO:5) (e.g., Dang et al., supra; Zhou et al., supra). Preferably these peptide inhibitors are myristoylated (SEQ ID NOs:6, 7, and 8, respectively) to improve cell permeability (e.g., Standaert et al., supra; for SEQ ID NO:6). Myristoylated and nonmyristoylated .zeta.PKC peptide inhibitors can be chemically synthesized and are commercially available from, e.g., Quality Controlled Biochemical (Hopkinton, Mass.) and BioSource International, Inc., USA (Camarillo, Calif.). One can provide a cell (preferably a chondrocyte) with a peptide inhibitor in vitro, in vivo, or ex vivo using the techniques described above.

Administration

[0065] Any of the compounds described herein (preferably a small molecule) can be administered in vivo in the form of a pharmaceutical composition for the treatment of arthritis. The pharmaceutical compositions may be administered by any number of routes, including, but not limited to, oral, nasal, rectal, topical, sublingual, intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intraventricular, intraperitoneal, intraarticular, or transdermal routes. In addition to the active ingredients, the pharmaceutical compositions may contain pharmaceutically acceptable carriers comprising excipients, coatings, and auxiliaries known in the art.

[0066] For any compound, the therapeutically effective dose can be estimated initially either in cell culture or in animal models. The therapeutically effective dose refers to the amount of active ingredient that ameliorates the condition or its symptoms. Therapeutic efficacy and toxicity in cell cultures or animal models may be determined by standard pharmaceutical procedures (e.g., ED50: the dose therapeutically effective in 50% of the population; LD50: the dose lethal to 50% of the population). The dose ratio between therapeutic and toxic effects is the therapeutic index, and can be expressed as the ratio ED50/LD50. Pharmaceutical compositions that exhibit large therapeutic indexes are preferred.

[0067] The data obtained from cell culture and animal models can then be used to formulate a range of dosage for the compound for use in mammals, preferably humans. The dosage of such a compound preferably lies within a range of concentrations that include the ED50 with little to no toxicity. The dosage may vary within this range depending upon the composition form employed and the administration route utilized.

EXAMPLES

[0068] The Examples which follow are set forth to aid in the understanding of the invention but are not intended to, and should not be construed to, limit its scope in any way. The Examples do not include detailed descriptions of conventional methods, such as those employed in the construction of vectors and plasmids, the insertion of genes encoding polypeptides into such vectors and plasmids, the introduction of such vectors and plasmids into host cells, or the expression of polypeptides from such vectors and plasmids in host cells. Such methods, and other conventional methods, are well known to those of ordinary skill in the art.

Example 1

.zeta.PKC Expression is Upregulated in Arthritis

Example 1.1

Experimental Design

[0069] To identify transcripts differentially expressed between arthritic and normal articular cartilage, tissue samples were obtained from arthritis patients with end-stage knee replacement and nonarthritic amputee individuals. The presence or absence of arthritis was confirmed by histology.

Example 1.2

Oligonucleotide Array Hybridization

[0070] The Human Genome U95Av2 (HG-U95Av2) GeneChip.RTM. Array (Affymetrix, Santa Clara, Calif.) was used for expression profiling. The HG-U95Av2 chip contains 25-mer oligonucleotide probes representing .about.12,000 primarily full-length sequences (.about.16 probe pairs/sequence) derived from the human genome. For each probe designed to be perfectly complimentary to a target sequence, a partner probe is generated that is identical except for a single base mismatch in its center. These probe pairs allow for signal quantitation and subtraction of nonspecific noise.

[0071] RNA was extracted from individual articular cartilage tissue, converted to biotinylated cRNA, and fragmented according to the Affymetrix protocol. The fragmented cRNAs were diluted in 1.times.MES buffer containing 100 .mu.g/ml herring sperm DNA and 500 .mu.g/ml acetylated BSA and denatured for 5 min at 99.degree. C. followed immediately by 5 min at 45.degree. C. Insoluble material was removed from the hybridization mixtures by a brief centrifugation, and the hybridization mix was added to each array and incubated at 45.degree. C. for 16 hr with continuous rotation at 60 rpm. After incubation, the hybridization mix was removed and the chips were extensively washed with 6.times.SSPET and stained with SAPE solution as described in the Affymetrix protocol.

Example 1.3

Oligonucleotide Array Data Analysis

[0072] The raw florescent intensity value of each transcript was measured at a resolution of 6 mm with a Hewlett-Packard Gene Array Scanner. GeneChip.RTM. software 3.2 (Affymetrix), which uses an algorithm to determine whether a gene is "present" or "absent," as well as the specific hybridization intensity values or "average differences" of each gene on the array, was used to evaluate the fluorescent data. The average difference for each gene was normalized to frequency values by referral to the average differences of 11 control transcripts of known abundance that were spiked into each hybridization mix according to the procedure of Hill et al. ((2000) Science 290:809-12). The frequency of each gene was calculated and represents a value equal to the total number of individual gene transcripts per 10.sup.6 total transcripts.

[0073] The frequency of each transcript was evaluated, and the transcript was included in the study if it met the following three criteria. First, transcripts which were called "present" by the GeneChip.RTM. software in at least one of the arrays for both arthritis and normal cartilage were included in the analysis. Second, for comparison between arthritis and normal cartilage, a t-test was applied to identify the subset of transcripts that had a significant (p<0.05) increase or decrease in frequency values. Third, average-fold changes in frequency values across the statistically significant subset of transcripts were required to be 2.4-fold or greater. These criteria were established based upon replicate experiments that estimated the intraarray reproducibility.

[0074] Based on these criteria, 602 transcripts were identified that were differentially expressed in arthritic and normal articular cartilage. One such transcript identified was .zeta.PKC.

Example 2

Inhibition of .zeta.PKC Activity Inhibits Extracellular Matrix (ECM) Degradation

Example 2.1

Primary Bovine Chondrocyte Isolation and Culture

[0075] Full-thickness bovine articular cartilage slices were dissected under aseptic conditions, rinsed four times in PBS, and subjected to pronase and collagenase digestion (1 mg/ml pronase (Calbiochem, San Diego, Calif.) for 30 minutes and 1 mg/ml Collagenase P (Roche Diagnostics Corporation, Indianapolis, Ind.) overnight at 37.degree. C. in DME without serum) to isolate chondrocytes embedded in the cartilage extracellular matrix. The digest was filtered through a 70 micron Falcon.TM. cell strainer (BD Biosciences, San Jose, Calif.) and washed twice in DME containing 10% FBS. Typically 2-4.times.10.sup.8 cells were obtained from a calf metacarpophalangeal joint surface. Cells were plated in monolayer in six-well plates at density of 2.times.10.sup.6 cells/well. For pellet culture, cells were resuspended in growth media [HL-1 media (Cambrex Corporation, East Rutherford, N.J.), penicillin+streptomycin, glutamine, 50 ng/ml ascorbate, and 10% FBS] at 1.times.10.sup.6 cells/ml, and 1 ml aliquots of cells were transferred to 15 ml sterile Falcon centrifuge tubes. The cells were centrifuged at 200.times.g for 5 min at 4.degree. C. and the resulting cell pellets were cultured as described previously (Xu et al. (1996) Endocrinology 137:3557-65). Cell media were collected and stored for collagen and proteoglycan assays, and cells were refed with fresh media (3 ml/well, 1 ml/tube) every 3-4 days. Pellet cultures were maintained for 3 weeks, at which time the pellets were harvested and either digested with 0.5 ml of 300 .mu.g/ml papain at 65.degree. C. for 3-6 hrs for dimethylmethylene blue (DMMB) dye assays or prepared for histology.

Example 2.2

Peptide Blocker of NF-.kappa.B Can Inhibit TNF- or IL-1-Mediated Proteoglycan Degradation

[0076] To demonstrate that blocking NF-.kappa.B activity can inhibit proteoglycan degradation in our culture system, primary bovine chondrocytes were cultured with the NF-.kappa.B blocker SN50 for 4 days at a concentration of 300 .mu.g/ml in the presence or absence of 10 ng/ml TNF or 1 ng/ml IL-1. Cells were incubated with the inhibitor for 3 hrs prior to the addition of either TNF or IL-1. SN50 is a peptide that contains the nuclear localization signal of NF-.kappa.B coupled to a stretch of hydrophobic amino acids to facilitate transport across lipid bilayers, and has been shown to block NF-.kappa.B-mediated transcription (e.g., Lin et al. (1995) J. Biol. Chem. 270:14255-58). SN50M, which served as a negative control, is the same peptide with amino acid changes to abolish NF-.kappa.B-blocking activity. SN50 and SN50M are available from, e.g., Biomol Research Laboratories, Inc. (Plymouth Meeting, Pa.).

[0077] As shown in FIG. 2, SN50M was ineffective at preventing TNF- and IL-1-mediated proteoglycan degradation in bovine chondrocytes, as measured by proteoglycan release into the media and decreased recovery in the cell pellet. In contrast, SN50 completely inhibited the cytokine-mediated degradation of proteoglycan. In addition, SN50 even prevented proteoglycan degradation in the absence of cytokine treatment, as compared to SN50M. These results demonstrate that blocking NF-.kappa.B, which controls the cytokine-mediated synthesis of collagenases and aggrecanases, inhibits proteoglycan degradation in primary bovine chondrocytes.

Example 2.3

.zeta.PKC Inhibitors Block TNF-Mediated Proteoglycan Degradation

[0078] To determine whether .zeta.PKC inhibitors can inhibit TNF-mediated proteoglycan degradation, primary bovine chondrocytes were cultured with various concentrations of TNF for 5 days with or without (1) the myristoylated .zeta.PKC peptide pseudosubstrate [herein termed "2089"; synthesized in-house; equivalent to SEQ ID NO:6; available from, e.g., BioSource International, Inc., USA (Camarillo, Calif.)], or (2) the small molecule inhibitor Ro-31-8220 (Sigma-RBI, Natick, Mass.). The inhibitors were added 3 hrs prior to addition of TNF.

[0079] As shown in FIG. 3, TNF at 10 and 100 ng/ml caused significant proteoglycan degradation, as measured by proteoglycan release into the media and decreased recovery in the cell pellet. This proteoglycan degradation was significantly inhibited by both 50 .mu.M 2089 and 10 .mu.M Ro-31-8220 (RO31). In addition, these compounds inhibited proteoglycan degradation even in the absence of TNF, suggesting a blockade of constitutive levels of proteases. Ro-31-8220 at a concentration of 5 .mu.M was relatively ineffective at preventing proteoglycan degradation. These results demonstrate that both 2089 and Ro-31-8220 penetrate the cell membrane of primary chondrocytes and effectively block TNF-mediated proteoglycan degradation. Trypan blue staining of the chondrocytes and lactate assays on the culture media were performed to rule out cytotoxicity as a possible explanation for the results. These experiments confirmed that the compounds do not cause appreciable cytotoxicity at the doses used in these experiments. In addition, control peptides were synthesized and tested in the same assay system to address the possibility that nonspecific effects may be causing the observed results. A nonmyristoylated version of the pseudosubstrate peptide, as well as a "scrambled control" peptide containing the same amino acid content as the pseudosubstrate peptide, but with a scrambled sequence, were tested, and both were found to be ineffective at blocking proteoglycan degradation.

Example 2.4

Myristoylated .zeta.PKC Peptide Pseudosubstrate 2089 Blocks Both TNF- and IL-1-Mediated Proteoglycan Degradation in a Dose-Dependent Manner

[0080] To determine whether myristoylated .zeta.PKC peptide pseudosubstrate 2089 can inhibit cytokine-mediated proteoglycan degradation in a dose-dependent manner, primary bovine chondrocytes were cultured with either 10 ng/ml TNF or 1 ng/ml IL-1 for 4 days after addition of various concentrations of 2089.

[0081] As shown in FIG. 4, 2089 inhibited both TNF- and IL-1-mediated proteoglycan degradation in a dose-dependent manner, with the highest dose (100 .mu.M) completely inhibiting proteoglycan release into the media. Again, increased cell pellet retention of proteoglycan and decreased release of proteoglycan into the media was achieved with 2089 even in the absence of cytokine. Cumulatively, these results indicate that inhibition of .zeta.PKC in chondrocytes inhibits cytokine-mediated proteolytic degradation of proteoglycan. This, along with the fact that the .zeta.PKC knockout mouse has a very benign phenotype (Leitges et al., supra), indicates that inhibition of .zeta.PKC may be a safe, effective treatment for arthritis, as well as other inflammatory diseases.

Example 3

.zeta.PKC mRNA is Upregulated in Human Osteoarthritic (OA) Cartilage

[0082] Transcriptional profiling data on human articular cartilage from osteoarthritic (OA) patients showed a statistically significant increase in .zeta.PKC mRNA as compared with human non-OA cartilage. In panel A of FIG. 5, RNA was extracted from frozen pulverized articular cartilage tissue from clinical samples, and subjected to expression profiling analysis using the HG-U95Av2 chip. Three groups were analyzed: normal (non-OA) cartilage [13 samples]; severe OA cartilage (nonlesional areas) [29 samples]; and severe OA cartilage (lesional areas) [26 samples]. Levels of .zeta.PKC mRNA were elevated in severe OA samples as compared with normal samples. In panel B of FIG. 5, TaqMan.RTM. Q-PCR (Applied Biosystems, Foster City, Calif.) analysis of the same set of samples showed significantly higher levels of .zeta.PKC mRNA in severe OA samples as compared with normal samples; TaqMan.RTM. Q-PCR protocols were conducted according to the manufacturer's instructions.

Example 4

.zeta.PKC Protein is Expressed in Chondrocytes

[0083] An anti-.zeta.PKC antibody (nPKC.zeta.(C-20); Santa Cruz Biotechnology, Inc., Santa Cruz, Calif.) was used to compare production of .zeta.PKC protein in chondrocytes and Jurkat cells. The lysates of bovine chondrocytes and Jurkat cells (human cell line) were prepared by similar methods: cells were washed with cold phosphate-buffered saline and immediately placed in cell lysis buffer (Cell Signaling Technology, Inc., Beverly, Mass.) containing phosphatase inhibitors. Cells were incubated for 5 min on ice, and then centrifuged at 12,000 rpm for 10 min at 4.degree. C. Samples were resolved by 12% SDS-polyacrylamide gel electrophoresis under reducing conditions. A Western blot showed that chondrocytes expressed a substantial amount of .zeta.PKC protein; the same blot did not show appreciable expression of .zeta.PKC protein in Jurkat cells.

Example 5

Adenoviral-Mediated Expression of .zeta.PKC Increases Proteoglycan Degradation

[0084] Primary bovine chondrocytes were isolated and cultured as described above (in pellet format) in Example 2.1. Cells were cultured in 0.5 ml growth media (HL-1) containing 2% FBS prior to the addition of adenovirus (in 15 ml Falcon tubes). Adenovirus vectors containing .zeta.PKC or GFP (green fluorescent protein) were prepared (Alden et al. (1999) Hum. Gene Ther. 10:2245-53), and cultures of chondrocytes were infected immediately following isolation and prior to pelleting. The adenovirus expressing GFP or .zeta.PKC was added directly into the culture at a multiplicity of infection (MOI) of 5000. As seen in panel A of FIG. 6, overexpression of full-length .zeta.PKC in primary bovine chondrocytes in culture (without addition of cytokines) resulted in a modest but statistically significant increase in proteoglycan degradation (as measured by proteoglycan released into the media in the chondrocyte pellet assay) as compared with overexpression of GFP. Medium containing 10% FBS was added to the culture after a 2-hour incubation at 37.degree. C. (in a humidified atmosphere of 5% CO.sub.2). The serum composition was gradually decreased every 3 days (sequentially to 5%, 2.5% and finally to 0% (serum-free) with every feeding of the chondrocyte pellets) to wean the cells from serum. Proteoglycan released into the media represents total proteoglycan released over 25 days.

[0085] In panel B of FIG. 6, proteoglycan was measured in the media over 4 days, with or without addition of cytokines, after cells had been weaned from serum. Addition of suboptimal levels of TNF.alpha. significantly enhanced the amount of proteoglycan released into the media in response to overexpression of .zeta.PKC, as compared with overexpression of GFP or absence of adenovirus infection (FIG. 6, panel B).

Example 6

.zeta.PKC is Responsible for TNF.alpha.-Mediated Proteoglycan Release in Chondrocytes

[0086] Articular bovine chondrocytes were prepared as previously detailed for the pellet assay. As shown in FIG. 7, TNF.alpha. was added (100 ng/ml; bars labeled with *) to some cultures in the chondrocyte pellet assay. Two inhibitors were added at various doses. One inhibitor, bisindolylmaleimide (BIS), is a pan-PKC inhibitor, reported to block the activity of all isoforms of .zeta.PKC, including .zeta.PKC (e.g., Toullec et al. (1991) J. Biol. Chem. 266:15771-81). The other inhibitor, chelerythrine chloride (CC), is a competitive inhibitor of the phorbol ester-binding site. CC competes for the phorbol ester-binding domain of the conventional and novel .zeta.PKC family members and inhibits them; however, as the atypical .zeta.PKCs (e.g., .zeta.PKC; Ca.sup.++-independent and diacylglycerol-independent .zeta.PKCs) lack this binding domain, they are not inhibited by CC (e.g., Herbert et al. (1990) Biochem. Biophys. Res. Commun. 172:993-99). Cytokine (TNF.alpha.)-mediated release of proteoglycan into the media in the chondrocyte pellet assay was blocked by BIS (at 20-40 .mu.M), but was not blocked by CC (FIG. 7), indicating that selective inhibition of .zeta.PKC blocks cytokine-mediated proteoglycan degradation.

Example 7

.zeta.PKC is Responsible for TNF.alpha.-Induced Activation of NF-.kappa.B in Chondrocytes

[0087] Activation of NF-.kappa.B was measured in an immortalized human chondrocyte cell line (C28/I2; see, e.g., Finger et al. (2003) Arthritis Rheum. 48:3395-403; Goldring (1994) J. Clin. Invest. 94:2307-16) into which a luciferase reporter gene under the control of an NF-.kappa.B response element was introduced. The cells were cultured in DMEM/Ham's F12 supplemented with 10% FBS; the cells were split into 96 wells (1.times.10.sup.5 cells/well) and infected with adenovirus expressing NF-.kappa.B luciferase construct (100 MOI) 24 hrs prior to assay. The chondrocytes were incubated with inhibitors in serum-free media 2 hrs prior to the addition of TNF. As shown in FIG. 8, TNF.alpha. was added (1 ng/ml or 10 ng/ml) to some cultures in the chondrocyte pellet assay. BIS (20 .mu.M), a pan-PKC inhibitor, was added to some cultures, and CC (8 .mu.M), a competitive inhibitor of the phorbol ester binding site in some forms of .zeta.PKC (but not the atypical .zeta.PKCs, e.g., .zeta.PKC), was added to other cultures. Cytokine (TNF.alpha.)-mediated activation of NF-.kappa.B was blocked by BIS, but was not blocked by CC, indicating that selective inhibition of .zeta.PKC blocks cytokine-mediated activation of NF-.kappa.B.

Sequence CWU 1

1

16412164DNAHomo sapiensCDS(1)..(1779) 1atg ccc agc agg acc gac ccc aag atg gaa ggg agc ggc ggc cgc gtc 48Met Pro Ser Arg Thr Asp Pro Lys Met Glu Gly Ser Gly Gly Arg Val1 5 10 15cgc ctc aag gcg cat tac ggg ggg gac atc ttc atc acc agc gtg gac 96Arg Leu Lys Ala His Tyr Gly Gly Asp Ile Phe Ile Thr Ser Val Asp 20 25 30gcc gcc acg acc ttc gag gag ctc tgt gag gaa gtg aga gac atg tgt 144Ala Ala Thr Thr Phe Glu Glu Leu Cys Glu Glu Val Arg Asp Met Cys 35 40 45cgt ctg cac cag cag cac ccg ctc acc ctc aag tgg gtg gac agc gaa 192Arg Leu His Gln Gln His Pro Leu Thr Leu Lys Trp Val Asp Ser Glu 50 55 60ggt gac cct tgc acg gtg tcc tcc cag atg gag ctg gaa gag gct ttc 240Gly Asp Pro Cys Thr Val Ser Ser Gln Met Glu Leu Glu Glu Ala Phe65 70 75 80cgc ctg gcc cgt cag tgc agg gat gaa ggc ctc atc att cat gtt ttc 288Arg Leu Ala Arg Gln Cys Arg Asp Glu Gly Leu Ile Ile His Val Phe 85 90 95ccg agc acc cct gag cag cct ggc ctg cca tgt ccg gga gaa gac aaa 336Pro Ser Thr Pro Glu Gln Pro Gly Leu Pro Cys Pro Gly Glu Asp Lys 100 105 110tct atc tac cgc cgg gga gcc aga aga tgg agg aag ctg tac cgt gcc 384Ser Ile Tyr Arg Arg Gly Ala Arg Arg Trp Arg Lys Leu Tyr Arg Ala 115 120 125aac ggc cac ctc ttc caa gcc aag cgc ttt aac agg aga gcg tac tgc 432Asn Gly His Leu Phe Gln Ala Lys Arg Phe Asn Arg Arg Ala Tyr Cys 130 135 140ggt cag tgc agc gag agg ata tgg ggc ctc gcg agg caa ggc tac agg 480Gly Gln Cys Ser Glu Arg Ile Trp Gly Leu Ala Arg Gln Gly Tyr Arg145 150 155 160tgc atc aac tgc aaa ctg ctg gtc cat aag cgc tgc cac ggc ctc gtc 528Cys Ile Asn Cys Lys Leu Leu Val His Lys Arg Cys His Gly Leu Val 165 170 175ccg ctg acc tgc agg aag cat atg gat tct gtc atg cct tcc caa gag 576Pro Leu Thr Cys Arg Lys His Met Asp Ser Val Met Pro Ser Gln Glu 180 185 190cct cca gta gac gac aag aac gag gac gcc gac ctt cct tcc gag gag 624Pro Pro Val Asp Asp Lys Asn Glu Asp Ala Asp Leu Pro Ser Glu Glu 195 200 205aca gat gga att gct tac att tcc tca tcc cgg aag cat gac agc att 672Thr Asp Gly Ile Ala Tyr Ile Ser Ser Ser Arg Lys His Asp Ser Ile 210 215 220aaa gac gac tcg gag gac ctt aag cca gtt atc gat ggg atg gat gga 720Lys Asp Asp Ser Glu Asp Leu Lys Pro Val Ile Asp Gly Met Asp Gly225 230 235 240atc aaa atc tct cag ggg ctt ggg ctg cag gac ttt gac cta atc aga 768Ile Lys Ile Ser Gln Gly Leu Gly Leu Gln Asp Phe Asp Leu Ile Arg 245 250 255gtc atc ggg cgc ggg agc tac gcc aag gtt ctc ctg gtg cgg ttg aag 816Val Ile Gly Arg Gly Ser Tyr Ala Lys Val Leu Leu Val Arg Leu Lys 260 265 270aag aat gac caa att tac gcc atg aaa gtg gtg aag aaa gag ctg gtg 864Lys Asn Asp Gln Ile Tyr Ala Met Lys Val Val Lys Lys Glu Leu Val 275 280 285cat gat gac gag gat att gac tgg gta cag aca gag aag cac gtg ttt 912His Asp Asp Glu Asp Ile Asp Trp Val Gln Thr Glu Lys His Val Phe 290 295 300gag cag gca tcc agc aac ccc ttc ctg gtc gga tta cac tcc tgc ttc 960Glu Gln Ala Ser Ser Asn Pro Phe Leu Val Gly Leu His Ser Cys Phe305 310 315 320cag acg aca agt cgg ttg ttc ctg gtc att gag tac gtc aac ggc ggg 1008Gln Thr Thr Ser Arg Leu Phe Leu Val Ile Glu Tyr Val Asn Gly Gly 325 330 335gac ctg atg ttc cac atg cag agg cag agg aag ctc cct gag gag cac 1056Asp Leu Met Phe His Met Gln Arg Gln Arg Lys Leu Pro Glu Glu His 340 345 350gcc agg ttc tac gcg gcc gag atc tgc atc gcc ctc aac ttc ctg cac 1104Ala Arg Phe Tyr Ala Ala Glu Ile Cys Ile Ala Leu Asn Phe Leu His 355 360 365gag agg ggg atc atc tac agg gac ctg aag ctg gac aac gtc ctc ctg 1152Glu Arg Gly Ile Ile Tyr Arg Asp Leu Lys Leu Asp Asn Val Leu Leu 370 375 380gat gcg gac ggg cac atc aag ctc aca gac tac ggc atg tgc aag gaa 1200Asp Ala Asp Gly His Ile Lys Leu Thr Asp Tyr Gly Met Cys Lys Glu385 390 395 400ggc ctg ggc cct ggt gac aca acg agc act ttc tgc gga acc ccg aat 1248Gly Leu Gly Pro Gly Asp Thr Thr Ser Thr Phe Cys Gly Thr Pro Asn 405 410 415tac atc gcc ccc gaa atc ctg cgg gga gag gag tac ggg ttc agc gtg 1296Tyr Ile Ala Pro Glu Ile Leu Arg Gly Glu Glu Tyr Gly Phe Ser Val 420 425 430gac tgg tgg gcg ctg gga gtc ctc atg ttt gag atg atg gcc ggg cgc 1344Asp Trp Trp Ala Leu Gly Val Leu Met Phe Glu Met Met Ala Gly Arg 435 440 445tcc ccg ttc gac atc atc acc gac aac ccg gac atg aac aca gag gac 1392Ser Pro Phe Asp Ile Ile Thr Asp Asn Pro Asp Met Asn Thr Glu Asp 450 455 460tac ctt ttc caa gtg atc ctg gag aag ccc atc cgg atc ccc cgg ttc 1440Tyr Leu Phe Gln Val Ile Leu Glu Lys Pro Ile Arg Ile Pro Arg Phe465 470 475 480ctg tcc gtc aaa gcc tcc cat gtt tta aaa gga ttt tta aat aag gac 1488Leu Ser Val Lys Ala Ser His Val Leu Lys Gly Phe Leu Asn Lys Asp 485 490 495ccc aaa gag agg ctc ggc tgc cgg cca cag act gga ttt tct gac atc 1536Pro Lys Glu Arg Leu Gly Cys Arg Pro Gln Thr Gly Phe Ser Asp Ile 500 505 510aag tcc cac gcg ttc ttc cgc agc ata gac tgg gac ttg ctg gag aag 1584Lys Ser His Ala Phe Phe Arg Ser Ile Asp Trp Asp Leu Leu Glu Lys 515 520 525aag cag gcg ctc cct cca ttc cag cca cag atc aca gac gac tac ggt 1632Lys Gln Ala Leu Pro Pro Phe Gln Pro Gln Ile Thr Asp Asp Tyr Gly 530 535 540ctg gac aac ttt gac aca cag ttc acc agc gag ccc gtg cag ctg acc 1680Leu Asp Asn Phe Asp Thr Gln Phe Thr Ser Glu Pro Val Gln Leu Thr545 550 555 560cca gac gat gag gat gcc ata aag agg atc gac cag tca gag ttc gaa 1728Pro Asp Asp Glu Asp Ala Ile Lys Arg Ile Asp Gln Ser Glu Phe Glu 565 570 575ggc ttt gag tat atc aac cca tta ttg ctg tcc acc gag gag tcg gtg 1776Gly Phe Glu Tyr Ile Asn Pro Leu Leu Leu Ser Thr Glu Glu Ser Val 580 585 590tga ggccgcgtgc gtctctgtcg tggacacgcg tgattgaccc tttaactgta 1829tccttaacca ccgcatatgc atgccaggct gggcacggct ccgagggcgg ccagggacag 1889acgcttgcgc cgagaccgca gagggaagcg tcagcgggcg ctgctgggag cagaacagtc 1949cctcacacct ggcccggcag gcagcttcgt gctggaggaa cttgctgctg tgcctgcgtc 2009gcggcggatc cgcggggacc ctgccgaggg ggctgtcatg cggtttccaa ggtgcacatt 2069ttccacggaa acagaactcg atgcactgac ctgctccgcc aggaaagtga gcgtgtagcg 2129tcctgaggaa taaaatgttc cgatgaaaaa aaaaa 21642592PRTHomo sapiens 2Met Pro Ser Arg Thr Asp Pro Lys Met Glu Gly Ser Gly Gly Arg Val1 5 10 15Arg Leu Lys Ala His Tyr Gly Gly Asp Ile Phe Ile Thr Ser Val Asp 20 25 30Ala Ala Thr Thr Phe Glu Glu Leu Cys Glu Glu Val Arg Asp Met Cys 35 40 45Arg Leu His Gln Gln His Pro Leu Thr Leu Lys Trp Val Asp Ser Glu 50 55 60Gly Asp Pro Cys Thr Val Ser Ser Gln Met Glu Leu Glu Glu Ala Phe65 70 75 80Arg Leu Ala Arg Gln Cys Arg Asp Glu Gly Leu Ile Ile His Val Phe 85 90 95Pro Ser Thr Pro Glu Gln Pro Gly Leu Pro Cys Pro Gly Glu Asp Lys 100 105 110Ser Ile Tyr Arg Arg Gly Ala Arg Arg Trp Arg Lys Leu Tyr Arg Ala 115 120 125Asn Gly His Leu Phe Gln Ala Lys Arg Phe Asn Arg Arg Ala Tyr Cys 130 135 140Gly Gln Cys Ser Glu Arg Ile Trp Gly Leu Ala Arg Gln Gly Tyr Arg145 150 155 160Cys Ile Asn Cys Lys Leu Leu Val His Lys Arg Cys His Gly Leu Val 165 170 175Pro Leu Thr Cys Arg Lys His Met Asp Ser Val Met Pro Ser Gln Glu 180 185 190Pro Pro Val Asp Asp Lys Asn Glu Asp Ala Asp Leu Pro Ser Glu Glu 195 200 205Thr Asp Gly Ile Ala Tyr Ile Ser Ser Ser Arg Lys His Asp Ser Ile 210 215 220Lys Asp Asp Ser Glu Asp Leu Lys Pro Val Ile Asp Gly Met Asp Gly225 230 235 240Ile Lys Ile Ser Gln Gly Leu Gly Leu Gln Asp Phe Asp Leu Ile Arg 245 250 255Val Ile Gly Arg Gly Ser Tyr Ala Lys Val Leu Leu Val Arg Leu Lys 260 265 270Lys Asn Asp Gln Ile Tyr Ala Met Lys Val Val Lys Lys Glu Leu Val 275 280 285His Asp Asp Glu Asp Ile Asp Trp Val Gln Thr Glu Lys His Val Phe 290 295 300Glu Gln Ala Ser Ser Asn Pro Phe Leu Val Gly Leu His Ser Cys Phe305 310 315 320Gln Thr Thr Ser Arg Leu Phe Leu Val Ile Glu Tyr Val Asn Gly Gly 325 330 335Asp Leu Met Phe His Met Gln Arg Gln Arg Lys Leu Pro Glu Glu His 340 345 350Ala Arg Phe Tyr Ala Ala Glu Ile Cys Ile Ala Leu Asn Phe Leu His 355 360 365Glu Arg Gly Ile Ile Tyr Arg Asp Leu Lys Leu Asp Asn Val Leu Leu 370 375 380Asp Ala Asp Gly His Ile Lys Leu Thr Asp Tyr Gly Met Cys Lys Glu385 390 395 400Gly Leu Gly Pro Gly Asp Thr Thr Ser Thr Phe Cys Gly Thr Pro Asn 405 410 415Tyr Ile Ala Pro Glu Ile Leu Arg Gly Glu Glu Tyr Gly Phe Ser Val 420 425 430Asp Trp Trp Ala Leu Gly Val Leu Met Phe Glu Met Met Ala Gly Arg 435 440 445Ser Pro Phe Asp Ile Ile Thr Asp Asn Pro Asp Met Asn Thr Glu Asp 450 455 460Tyr Leu Phe Gln Val Ile Leu Glu Lys Pro Ile Arg Ile Pro Arg Phe465 470 475 480Leu Ser Val Lys Ala Ser His Val Leu Lys Gly Phe Leu Asn Lys Asp 485 490 495Pro Lys Glu Arg Leu Gly Cys Arg Pro Gln Thr Gly Phe Ser Asp Ile 500 505 510Lys Ser His Ala Phe Phe Arg Ser Ile Asp Trp Asp Leu Leu Glu Lys 515 520 525Lys Gln Ala Leu Pro Pro Phe Gln Pro Gln Ile Thr Asp Asp Tyr Gly 530 535 540Leu Asp Asn Phe Asp Thr Gln Phe Thr Ser Glu Pro Val Gln Leu Thr545 550 555 560Pro Asp Asp Glu Asp Ala Ile Lys Arg Ile Asp Gln Ser Glu Phe Glu 565 570 575Gly Phe Glu Tyr Ile Asn Pro Leu Leu Leu Ser Thr Glu Glu Ser Val 580 585 590313PRTArtificialpeptide inhibitor 3Ser Ile Tyr Arg Arg Gly Ala Arg Arg Trp Arg Lys Leu1 5 10417PRTArtificialpeptide inhibitor 4Ser Ile Tyr Arg Arg Gly Ala Arg Arg Trp Arg Lys Leu Tyr Arg Ala1 5 10 15Asn59PRTArtificialpeptide inhibitor 5Arg Arg Gly Ala Arg Arg Trp Arg Lys1 5613PRTArtificialpeptide inhibitor 6Ser Ile Tyr Arg Arg Gly Ala Arg Arg Trp Arg Lys Leu1 5 10717PRTArtificialpeptide inhibitor 7Ser Ile Tyr Arg Arg Gly Ala Arg Arg Trp Arg Lys Leu Tyr Arg Ala1 5 10 15Asn89PRTArtificialpeptide inhibitor 8Arg Arg Gly Ala Arg Arg Trp Arg Lys1 5921DNAHomo sapiens 9aagtgagaga catgtgtcgt c 211021DNAHomo sapiens 10aagatggagg aagctgtacc g 211121DNAHomo sapiens 11aaggctacag gtgcatcaac t 211221DNAHomo sapiens 12aactgctggt ccataagcgc t 211321DNAHomo sapiens 13aagagcctcc agtagacgac a 211421DNAHomo sapiens 14aagacgactc ggaggacctt a 211521DNAHomo sapiens 15aagagctggt gcatgatgac g 211621DNAHomo sapiens 16aagtcggttg ttcctggtca t 211721DNAHomo sapiens 17aagctcacag actacggcat g 211821DNAHomo sapiens 18aagaggatcg accagtcaga g 211921DNAHomo sapiens 19aactgtatcc ttaaccaccg c 212021DNAHomo sapiens 20aaccaccgca tatgcatgcc a 212121RNAArtificialsiRNA polynucleotide, synthesized 21gugagagaca ugugucgucu u 212221RNAArtificialsiRNA polynucleotide, synthesized 22gauggaggaa gcuguaccgu u 212321RNAArtificialsiRNA polynucleotide, synthesized 23ggcuacaggu gcaucaacuu u 212421RNAArtificialsiRNA polynucleotide, synthesized 24cugcuggucc auaagcgcuu u 212521RNAArtificialsiRNA polynucleotide, synthesized 25gagccuccag uagacgacau u 212621RNAArtificialsiRNA polynucleotide, synthesized 26gacgacucgg aggaccuuau u 212721RNAArtificialsiRNA polynucleotide, synthesized 27gagcuggugc augaugacgu u 212821RNAArtificialsiRNA polynucleotide, synthesized 28gucgguuguu ccuggucauu u 212921RNAArtificialsiRNA polynucleotide, synthesized 29gcucacagac uacggcaugu u 213021RNAArtificialsiRNA polynucleotide, synthesized 30gaggaucgac cagucagagu u 213121RNAArtificialsiRNA polynucleotide, synthesized 31cuguauccuu aaccaccgcu u 213221RNAArtificialsiRNA polynucleotide, synthesized 32ccaccgcaua ugcaugccau u 213321RNAArtificialsiRNA polynucleotide, synthesized 33gacgacacau gucucucacu u 213421RNAArtificialsiRNA polynucleotide, synthesized 34cgguacagcu uccuccaucu u 213521RNAArtificialsiRNA polynucleotide, synthesized 35aguugaugca ccuguagccu u 213621RNAArtificialsiRNA polynucleotide, synthesized 36agcgcuuaug gaccagcagu u 213721RNAArtificialsiRNA polynucleotide, synthesized 37ugucgucuac uggaggcucu u 213821RNAArtificialsiRNA polynucleotide, synthesized 38uaagguccuc cgagucgucu u 213921RNAArtificialsiRNA polynucleotide, synthesized 39cgucaucaug caccagcucu u 214021RNAArtificialsiRNA polynucleotide, synthesized 40augaccagga acaaccgacu u 214121RNAArtificialsiRNA polynucleotide, synthesized 41caugccguag ucugugagcu u 214221RNAArtificialsiRNA polynucleotide, synthesized 42cucugacugg ucgauccucu u 214321RNAArtificialsiRNA polynucleotide, synthesized 43gcggugguua aggauacagu u 214421RNAArtificialsiRNA polynucleotide, synthesized 44uggcaugcau augcgguggu u 214521DNAHomo sapiens 45cagaagatgg aggaagctgt a 214621DNAHomo sapiens 46caaggctaca ggtgcatcaa c 214721DNAHomo sapiens 47cagtagacga caagaacgag g 214821DNAHomo sapiens 48cagacgacaa gtcggttgtt c 214921DNAHomo sapiens 49caagtcggtt gttcctggtc a 215021DNAHomo sapiens 50cacatcaagc tcacagacta c 215121DNAHomo sapiens 51catcaagctc acagactacg g 215221DNAHomo sapiens 52caagctcaca gactacggca t 215321DNAHomo sapiens 53cacagactac ggcatgtgca a 215421DNAHomo sapiens 54catgaacaca gaggactacc t

215521DNAHomo sapiens 55cattccagcc acagatcaca g 215621DNAHomo sapiens 56cacagatcac agacgactac g 215721DNAHomo sapiens 57cagatcacag acgactacgg t 215821DNAHomo sapiens 58cagacgatga ggatgccata a 215921DNAHomo sapiens 59cattattgct gtccaccgag g 216021RNAArtificialsiRNA polynucleotide, synthesized 60gaagauggag gaagcuguau u 216121RNAArtificialsiRNA polynucleotide, synthesized 61aggcuacagg ugcaucaacu u 216221RNAArtificialsiRNA polynucleotide, synthesized 62guagacgaca agaacgaggu u 216321RNAArtificialsiRNA polynucleotide, synthesized 63gacgacaagu cgguuguucu u 216421RNAArtificialsiRNA polynucleotide, synthesized 64agucgguugu uccuggucau u 216521RNAArtificialsiRNA polynucleotide, synthesized 65caucaagcuc acagacuacu u 216621RNAArtificialsiRNA polynucleotide, synthesized 66ucaagcucac agacuacggu u 216721RNAArtificialsiRNA polynucleotide, synthesized 67agcucacaga cuacggcauu u 216821RNAArtificialsiRNA polynucleotide, synthesized 68cagacuacgg caugugcaau u 216921RNAArtificialsiRNA polynucleotide, synthesized 69ugaacacaga ggacuaccuu u 217021RNAArtificialsiRNA polynucleotide, synthesized 70uuccagccac agaucacagu u 217121RNAArtificialsiRNA polynucleotide, synthesized 71cagaucacag acgacuacgu u 217221RNAArtificialsiRNA polynucleotide, synthesized 72gaucacagac gacuacgguu u 217321RNAArtificialsiRNA polynucleotide, synthesized 73gacgaugagg augccauaau u 217421RNAArtificialsiRNA polynucleotide, synthesized 74uuauugcugu ccaccgaggu u 217521RNAArtificialsiRNA polynucleotide, synthesized 75uacagcuucc uccaucuucu u 217621RNAArtificialsiRNA polynucleotide, synthesized 76guugaugcac cuguagccuu u 217721RNAArtificialsiRNA polynucleotide, synthesized 77ccucguucuu gucgucuacu u 217821RNAArtificialsiRNA polynucleotide, synthesized 78gaacaaccga cuugucgucu u 217921RNAArtificialsiRNA polynucleotide, synthesized 79ugaccaggaa caaccgacuu u 218021RNAArtificialsiRNA polynucleotide, synthesized 80guagucugug agcuugaugu u 218121RNAArtificialsiRNA polynucleotide, synthesized 81ccguagucug ugagcuugau u 218221RNAArtificialsiRNA polynucleotide, synthesized 82augccguagu cugugagcuu u 218321RNAArtificialsiRNA polynucleotide, synthesized 83uugcacaugc cguagucugu u 218421RNAArtificialsiRNA polynucleotide, synthesized 84agguaguccu cuguguucau u 218521RNAArtificialsiRNA polynucleotide, synthesized 85cugugaucug uggcuggaau u 218621RNAArtificialsiRNA polynucleotide, synthesized 86cguagucguc ugugaucugu u 218721RNAArtificialsiRNA polynucleotide, synthesized 87accguagucg ucugugaucu u 218821RNAArtificialsiRNA polynucleotide, synthesized 88uuauggcauc cucaucgucu u 218921RNAArtificialsiRNA polynucleotide, synthesized 89ccucggugga cagcaauaau u 219021DNAHomo sapiens 90gagctctgtg aggaagtgag a 219121DNAHomo sapiens 91gaggaagtga gagacatgtg t 219221DNAHomo sapiens 92gaagtgagag acatgtgtcg t 219321DNAHomo sapiens 93gagagacatg tgtcgtctgc a 219421DNAHomo sapiens 94gaagatggag gaagctgtac c 219521DNAHomo sapiens 95gacctgcagg aagcatatgg a 219621DNAHomo sapiens 96gaggagacag atggaattgc t 219721DNAHomo sapiens 97gaggacctta agccagttat c 219821DNAHomo sapiens 98gatgacgagg atattgactg g 219921DNAHomo sapiens 99gattacactc ctgcttccag a 2110021DNAHomo sapiens 100gacgacaagt cggttgttcc t 2110121DNAHomo sapiens 101gacaagtcgg ttgttcctgg t 2110221DNAHomo sapiens 102gacctgatgt tccacatgca g 2110321DNAHomo sapiens 103gatgttccac atgcagaggc a 2110421DNAHomo sapiens 104gactacggca tgtgcaagga a 2110521DNAHomo sapiens 105gacatgaaca cagaggacta c 2110621DNAHomo sapiens 106gacttgctgg agaagaagca g 2110721DNAHomo sapiens 107gatcacagac gactacggtc t 2110821DNAHomo sapiens 108gaggatcgac cagtcagagt t 2110921DNAHomo sapiens 109gatcgaccag tcagagttcg a 2111021RNAArtificialsiRNA polynucleotide, synthesized 110gcucugugag gaagugagau u 2111121RNAArtificialsiRNA polynucleotide, synthesized 111ggaagugaga gacauguguu u 2111221RNAArtificialsiRNA polynucleotide, synthesized 112agugagagac augugucguu u 2111321RNAArtificialsiRNA polynucleotide, synthesized 113gagacaugug ucgucugcau u 2111421RNAArtificialsiRNA polynucleotide, synthesized 114agauggagga agcuguaccu u 2111521RNAArtificialsiRNA polynucleotide, synthesized 115ccugcaggaa gcauauggau u 2111621RNAArtificialsiRNA polynucleotide, synthesized 116ggagacagau ggaauugcuu u 2111721RNAArtificialsiRNA polynucleotide, synthesized 117ggaccuuaag ccaguuaucu u 2111821RNAArtificialsiRNA polynucleotide, synthesized 118ugacgaggau auugacuggu u 2111921RNAArtificialsiRNA polynucleotide, synthesized 119uuacacuccu gcuuccagau u 2112021RNAArtificialsiRNA polynucleotide, synthesized 120cgacaagucg guuguuccuu u 2112121RNAArtificialsiRNA polynucleotide, synthesized 121caagucgguu guuccugguu u 2112221RNAArtificialsiRNA polynucleotide, synthesized 122ccugauguuc cacaugcagu u 2112321RNAArtificialsiRNA polynucleotide, synthesized 123uguuccacau gcagaggcau u 2112421RNAArtificialsiRNA polynucleotide, synthesized 124cuacggcaug ugcaaggaau u 2112521RNAArtificialsiRNA polynucleotide, synthesized 125caugaacaca gaggacuacu u 2112621RNAArtificialsiRNA polynucleotide, synthesized 126cuugcuggag aagaagcagu u 2112721RNAArtificialsiRNA polynucleotide, synthesized 127ucacagacga cuacggucuu u 2112821RNAArtificialsiRNA polynucleotide, synthesized 128ggaucgacca gucagaguuu u 2112921RNAArtificialsiRNA polynucleotide, synthesized 129ucgaccaguc agaguucgau u 2113021RNAArtificialsiRNA polynucleotide, synthesized 130ucucacuucc ucacagagcu u 2113121RNAArtificialsiRNA polynucleotide, synthesized 131acacaugucu cucacuuccu u 2113221RNAArtificialsiRNA polynucleotide, synthesized 132acgacacaug ucucucacuu u 2113321RNAArtificialsiRNA polynucleotide, synthesized 133ugcagacgac acaugucucu u 2113421RNAArtificialsiRNA polynucleotide, synthesized 134gguacagcuu ccuccaucuu u 2113521RNAArtificialsiRNA polynucleotide, synthesized 135uccauaugcu uccugcaggu u 2113621RNAArtificialsiRNA polynucleotide, synthesized 136agcaauucca ucugucuccu u 2113721RNAArtificialsiRNA polynucleotide, synthesized 137gauaacuggc uuaagguccu u 2113821RNAArtificialsiRNA polynucleotide, synthesized 138ccagucaaua uccucgucau u 2113921RNAArtificialsiRNA polynucleotide, synthesized 139ucuggaagca ggaguguaau u 2114021RNAArtificialsiRNA polynucleotide, synthesized 140aggaacaacc gacuugucgu u 2114121RNAArtificialsiRNA polynucleotide, synthesized 141accaggaaca accgacuugu u 2114221RNAArtificialsiRNA polynucleotide, synthesized 142cugcaugugg aacaucaggu u 2114321RNAArtificialsiRNA polynucleotide, synthesized 143ugccucugca uguggaacau u 2114421RNAArtificialsiRNA polynucleotide, synthesized 144uuccuugcac augccguagu u 2114521RNAArtificialsiRNA polynucleotide, synthesized 145guaguccucu guguucaugu u 2114621RNAArtificialsiRNA polynucleotide, synthesized 146cugcuucuuc uccagcaagu u 2114721RNAArtificialsiRNA polynucleotide, synthesized 147agaccguagu cgucugugau u 2114821RNAArtificialsiRNA polynucleotide, synthesized 148aacucugacu ggucgauccu u 2114921RNAArtificialsiRNA polynucleotide, synthesized 149ucgaacucug acuggucgau u 2115021DNAHomo sapiens 150tagacgacaa gaacgaggac g 2115121DNAHomo sapiens 151tacagacaga gaagcacgtg t 2115221DNAHomo sapiens 152tacactcctg cttccagacg a 2115321DNAHomo sapiens 153tattgctgtc caccgaggag t 2115421DNAHomo sapiens 154taaccaccgc atatgcatgc c 2115521RNAArtificialsiRNA polynucleotide, synthesized 155gacgacaaga acgaggacgu u 2115621RNAArtificialsiRNA polynucleotide, synthesized 156cagacagaga agcacguguu u 2115721RNAArtificialsiRNA polynucleotide, synthesized 157cacuccugcu uccagacgau u 2115821RNAArtificialsiRNA polynucleotide, synthesized 158uugcugucca ccgaggaguu u 2115921RNAArtificialsiRNA polynucleotide, synthesized 159accaccgcau augcaugccu u 2116021RNAArtificialsiRNA polynucleotide, synthesized 160cguccucguu cuugucgucu u 2116121RNAArtificialsiRNA polynucleotide, synthesized 161acacgugcuu cucugucugu u 2116221RNAArtificialsiRNA polynucleotide, synthesized 162ucgucuggaa gcaggagugu u 2116321RNAArtificialsiRNA polynucleotide, synthesized 163acuccucggu ggacagcaau u 2116421RNAArtificialsiRNA polynucleotide, synthesized 164ggcaugcaua ugcggugguu u 21

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed