AU2002256518A1 - Inhibitors of receptor activator of NF-kappaB and uses thereof - Google Patents

Inhibitors of receptor activator of NF-kappaB and uses thereof

Info

Publication number
AU2002256518A1
AU2002256518A1 AU2002256518A AU2002256518A AU2002256518A1 AU 2002256518 A1 AU2002256518 A1 AU 2002256518A1 AU 2002256518 A AU2002256518 A AU 2002256518A AU 2002256518 A AU2002256518 A AU 2002256518A AU 2002256518 A1 AU2002256518 A1 AU 2002256518A1
Authority
AU
Australia
Prior art keywords
polypeptide
traf6
group
receptor activator
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2002256518A
Other versions
AU2002256518B2 (en
Inventor
Bharat Aggarwal
Bryant G Darnay
Sujay Singh
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Research Development Foundation
Original Assignee
Research Development Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Research Development Foundation filed Critical Research Development Foundation
Priority claimed from PCT/US2002/014842 external-priority patent/WO2002092623A1/en
Publication of AU2002256518A1 publication Critical patent/AU2002256518A1/en
Application granted granted Critical
Publication of AU2002256518B2 publication Critical patent/AU2002256518B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Description

INHIBITORS OF RECEPTOR ACTIVATOR OF NF-κB AND USES THEREOF
BACKGROUND OF THE INVENTION
Field of the Invention
The present invention relates generally to the fields of cytokine biology and bone diseases. More specifically, the pres ent invention provides inhibitors of receptor activator of NF-κB
(RANK) for therapy of disorders such as diseases associated with bone resorption.
Description of the Related Art Members of the TNF and TNF receptor superfamilies play critical roles in the initiation and regulation of the immune response (1-3). Although members of these families share m an y overlapping biological functions, it appears from gene knockout studies that they have unique features. One such receptor/ligand pair, receptor activator of NF-κB (RANK) and its ligand (RANKL/TRANCE/ODF/OPGL), is critically involved in regulation of bone remodeling and osteoclastogenesis (4). From knockout gene studies in mice, RANKL also functions in lymph node organogenesis and lymphocyte development (5). Furthermore, receptor activator of NF-κB and RANKL are implicated in th e interactions between T cells and dendritic cells during the immu ne response (6).
As indicated by its name, receptor activator of NF-κB stimulates activation of nuclear factor-κB (NF-κB) (7-12), a transcription factor that regulates the expression of a large number of genes that play essential roles in immune a n d inflammatory responses (13). Evidence over the past several years has indicated that some of the TNF receptor family members interact with a family of adapter proteins known as TRAFs, (TNF receptor-associated factors), which participate in activation of th e transcription factor NF-κB and c-Jun N-terminal kinase (JNK) (14).
The TNF receptor-associated factor family consists of six distinct proteins, each of which possesses a C-terminal homologous domain that is critical for self-association and is required for interaction with the receptors. All of the TNF receptor-associated factors, except for TRAF1 and TRAF4, also contain ring and zinc finger motifs in their N-termini, which appear to be utilized for interacting with other signaling molecules. Similar to the trimeric structure of the ligands an d receptors of the TNF family, the C-terminus of the TRAF2 adapts a trimeric structure, as reported for TRAF2 in its interaction with peptides derived from TNF receptor 1 and CD40 (15, 16). This trimeric structure of the TNF receptor-associated factor molecules likely enables them to associate with downstream adap ter proteins .
It has been demonstrated that TRAF2, TRAF5, a n d TRAF6 interact with receptor activator of NF-κB (RANK) and th at receptor activator of NF-κB could activate both the NF-κB and JNK pathways (7). Subsequently, a more detailed analysis of th e interaction of these TNF receptor-associated factors with receptor activator of NF-κB was reported (8). A novel TRAF6-binding motif has been identified in RANK that is distinct from the TRAF2- an d TRAF5- binding domains. A homologous TRAF6-binding motif i n CD40 was described using a combinatorial peptide library approach (17). The TRAF6 binding domain in RANK was sufficient for activation of NF-κB, suggesting that TRAF2 and TRAF5 are not necessary for NF-κB activation. However, it appears that th e TRAF2-binding motif is sufficient for JNK activation, although th e TRAF6-binding domain could also activate JNK, albeit to a les ser extent. Additionally, NIK (NF-κB inducing kinase) was also found to be required for the activation of NF-κB by receptor activator of NF-κB. In addition to TRAF2, TRAF5, and TRAF6, it has b een demonstrated that TRAF1 and TRAF3 also associate with th e carboxy terminus of receptor activator of NF-κB (9-12).
The role played by each TRAF molecule in RANK signal transduction remains elusive. Dominant negative mutants of TRAF2, TRAF5, and TRAF6 have been used to evaluate their role in NF-κB activation by RANK. It appears that all of the dominant negative TRAFs differentially inhibit the activation of NF-κB induced by overexpression of RANK in 293 cells (9, 12). However, inclusion of all dominant negative mutants of TRAF2, TRAF5, an d TRAF6 did not completely eliminate the activation of NF-κB induced by RANK in 293 cells (12). Stimulation of RANK also caused the recruitment of TRAF6, which in turn recruits an d activates c-Src, which appears to be responsible for activation of phosphoinositol-3-kinase and protein kinase B/AKT, a molecule potentially involved in cell survival (18).
Knockout mouse models of RANKL, RANK, an d osteoprotegerin have demonstrated an essential role of thes e molecules in osteoclastogenesis. The biological importance of these molecules is underscored by the induction of severe osteoporosis by targeted disruption of osteoprotegerin and by th e induction of osteopetrosis by targeted disruption of RANKL or b y overexpression of osteoprotegerin (5, 19, 20). Thus, osteoclast formation may be attributed to the relative ratio of RANKL to osteoprotegerin in the microenvironment of bone marrow, a n d alterations in this balance may be a major cause of bone loss i n many metabolic bone disorders.
Similar to RANKL-/- mice, targeted disruption of receptor activator of NF-κB also lead to an osteopetrotic phenotype (21 , 22). Both RANK-/- and RANKL-/- mice exhibited absence of osteoclasts, indicating the essential requirement of these molecules for osteoclastogenesis. Additionally, mice lacking TRAF6 (23-25), c-Src (26), c-Fos (27), or the NF-κB subunits p50/p52 (28, 29) also display an osteopetrotic phenotype. Although these mutant mice have osteoclasts, these cells apparently have defects in bone resorption. Thus, RANKL an d receptor activator of NF-κB as well as their cytoplasmic signaling molecules are required for osteoclastogenesis. Of the TRAF molecules that bind to receptor activator of NF-κB, only TRAF6 appears to be essential for osteoclast differentiation as indicated in mice lacking TRAF6. Thus, th e interaction of receptor activator of NF-κB with TRAF6 may be a unique target for therapeutic intervention, and the ability to disrupt this interaction by a competitive, cell permeable peptide remains to be investigated.
Thus, the prior art is deficient in methods of disrupting the interaction between receptor activator of NF-κB and TRAF6 i n order to inhibit RANKL signaling and osteoclast differentiation induced by RANKL. Such inhibitors would be useful a s therapeutics in bone disorders and cancer associated with increased bone resorption. The present invention fulfills this long-standing need and desire in the art.
SUMMARY OF THE INVENTION
TRAF6 (TNF receptor-associated factor 6) is a critical adapter protein for Receptor Activator of NF-κB (RANK) signaling. The present invention discloses a novel TRAF6 decoy peptide (T6DP) with and without a peptide leader sequence that allows for transversing the cellular membrane. Evidence disclosed herein indicate that the TRAF6 decoy peptide inhibits early events associated with RANKL signaling and RANKL-mediated osteoclast differentiation only when the leader sequence is attached. This data indicates that targeted disruption of interaction betw een Receptor Activator of NF-κB and TRAF6 may prove useful as a therapeutic for metabolic bone disorders, leukemia, arthritis, and metastatic cancer of the bone.
The present invention provides polypeptides th at inhibit signaling mediated by TNF receptor-associated factor 6 (TRAF6), wherein the polypeptides comprise of a TRAF6 binding domain and a leader signal sequence. The present invention is further drawn to methods of inhibiting Receptor Activator of NF- KB Ligand (RANKL)-induced osteoclast differentiation using the polypeptides disclosed herein. In another aspect of the present invention, there is provided a non-peptide analog that mimics the function of th e polypeptide disclosed herein, wherein said non-peptide analog inhibits signaling mediated by TRAF6. The present invention is further drawn to methods of inhibiting Receptor Activator of NF- KB Ligand (RANKL)-induced osteoclast differentiation using a non- peptide analog that mimics the function of the polypeptide disclosed herein.
Other and further aspects, features, and advantages of the present invention will be apparent from the following description of the presently preferred embodiments of th e invention. These embodiments are given for the purpose of disclosure.
BRIEF DESCRIPTION OF THE DRAWINGS
So that the matter in which the above-recited features, advantages and objects of the invention, as well as others which will become clear, are attained and can be understood in detail, more particular descriptions of the invention briefly summarized above may be had by reference to certain embodiments thereof which are illustrated in the appended drawings. These drawings form a part of the specification. It is to be noted, however, th at the appended drawings illustrate preferred embodiments of th e invention and therefore are not to be considered limiting in their scope.
Figure 1 shows the novel TRAF6 binding domain i n
RANK. Figure 1 A shows the consensus sequence of the TRAF6 binding domain. The region within the amino acid sequence of CD40 (residues 230-245) that specifically interacts with TRAF6 (38) was used as the template for alignment of RANK, IRAKI, an d IRAK2. Identical residues are in bold. A consensus motif is shown on the bottom.
Figure IB shows the peptides used in the pres ent study. The hydrophobic domain of Kaposi fibroblast growth factor signal sequence (underlined) (36) was attached to the tw o potential TRAF6 binding domains from murine receptor activator of NF-κB. Additionally, two peptides lacking the leader sequence were synthesized.
Figure 2 shows L-T6Bp-l inhibits RANKL-mediated osteoclast differentiation in RAW264.7 cells. Figure 2A show s RANKL-induces TRAP positive osteoclasts. RAW264.7 cells w ere plated in 12-well plates and stimulated with RANKL (30 ng/ml ) for 4 days. Cells were stained for TRAP essentially as described below. Photographs were taken using a lOx objective lens. Figure 2B shows L-T6DP-1 inhibits osteoclast differentiation. RAW cells were plated as described in Figure 2 A and treated with RANKL in the presence of 1 , 30, or 100 μM of peptides as indicated. On day 4 cells were stained and evaluated as in Figure 2A.
Figure 2C shows L-T6DP-1 inhibits the total numb er of TRAP positive osteoclasts induced by RANKL. RAW264.7 cells were plated in 48-well plates in triplicate and treated with RANKL in the presence of increasing amounts of the indicated peptides . After 4-5 days, the cells were stained for TRAP and the total numbers of osteoclasts were counted as described below.
Figure 3 shows leader TRAF6-binding peptide inhibits normal mouse derived osteoclast differentiation by RANKL and M - CSF. Figure 3 A shows mouse-derived monocytes were plated i n 48-well plates in triplicate and costimulated with murine M-CSF (10 ng/ml) and RANKL (30 ng/ml) in the absence (0) or presence of the indicated peptides. After 6-7 days, the cells were fixed an d stained for TRAP. The numbers of multi-nucleated, TRAP positive osteoclasts were counted. Figure 3B shows LT6DP-1 inhibits osteoclast differentiation of mouse-derived monocytes. Mouse-derived monocytes were plated in 48-well plates and costimulated as in Figure 3A with murine M-CSF (10 ng/ml) and RANKL (30 ng/ml) in the absence (left panel) or presence of the indicated peptides . After 6-7 days, the cells were fixed and stained for TRAP. A representative field from each well was taken at a magnification of lOx.
Figure 4 shows that L-T6DP-1 specifically inhibits RANKL-induced NF-κB activation and TRAF6 binding. Figure 4 A shows inhibition of RANKL-mediated NF-κB activation by L-T6Bp- 1. RAW264.7 cells were plated in 6-well plates and treated with the indicated peptides for 5 h, then treated with RANKL (10 nM) for 15 min. Nuclear and cytoplasmic extracts were prepared and a gel mobility shift assays was performed with 8 μg of nuclear extracts as described below.
Figure 4B shows inhibition of IκB degradation by L- T6DP-1. Cytoplasmic extracts (30 μg) from Figure 4A w ere immunoblotted with anti-IκB as described below. Figure 4C shows L-T6DP-1 inhibits TRAF6 from binding to the receptor activator of NF-κB cytoplasmic domain. Cellular extracts from human 293 cells transfected with either FLAG-TRAF2, -TRAF5, or -TRAF6 were mixed with GST or GST- RANKcd in the absence or presence of 100 μM of the indicated peptides. A GST-pull down assay was performed as described below. The bound TRAFs were visualized by immunoblotting with anti-FLAG antibodies.
Figure 5 shows L-T6DP-1 specifically inhibits RANKL- induced JNK, ERK, and p38 kinase activation. Figure 5 A show s inhibition of RANKL-mediated JNK activation by L-T6DP- 1. RAW264.7 cells were plated in 6-well plates and treated with th e indicated peptides for 6 h, then treated with RANKL (10 nM) for 15 min. Whole cell extracts were prepared and 30 μg of cell lysate was immunoprecipitated with anti-JNKl . In vitro kinase assays were performed using GST-Jun ( 1-79) as the substrate a s described below.
Figure 5B and Figure 5C show inhibition of RANKL- mediated ERK and p38 activation by L-T6DP-1. Whole cell extracts (30 μg) from Figure 5A were separated by 10% SDS-PAGE and electroblotted onto a nitrocellulose membrane. The membranes were first immunoblotted with the indicated phospho-specific antibody and then stripped and re-probed with the indicated antibodies as described below. Figure 6 shows L-T6DP-1 inhibits osteoclast differentiation induced by breast cancer cells. Figure 6A : Breast cancer cells (500/well) were plated in 24-well plates in th e absence or presence of RAW cells (10000/well) and in the absence or presence of L-T6DP-1 or T6DP-1 (100 μM). After 5 days, th e cells were fixed, stained for TRAP, and photographed with a l Ox objective lens.
Figure 6B shows RAW cells were cultured alone or i n the presence of the indicated cells on synthetic bone slides (BD BiCoat Osteologic MultiTest Slides). After 6 days, the cells w ere removed by washing the slide in bleach for 5 minutes and then washing thoroughly with distilled water. The slide was air dried and then photographs were taken with a light microscope (10 x objective lens). The ghosts of osteoclasts (arrow) can be s een where functional osteoclasts destroyed the synthetic bone matrix. Figure 6C shows normal breast epithelial cells
(MCF10A, 1000/well) grown in the presence of RAW and stained for TRAP as indicted in Figure 6A. The right panel includes th e addition of RANKL (100 ng/ml) to the co-culture.
DETAILED DESCRIPTION OF THE INVENTION
Receptor activator of NF-κB (RANK), a recently described member of the TNF receptor superfamily, is expressed primarily by dendritic cells, osteoclast progenitors, activated B a n d T cells and osteoclasts. By binding to its ligand (RANKL), th e receptor activator of NF-κB causes the sequential recruitment of adapter molecules responsible for activation of signaling processes. These pathways lead to activation of protein kinases, which in turn activate transcription factors leading to changes i n gene expression that alter the function of the cell.
Knockout mouse models of RANKL, RANK, an d osteoprotegerin (OPG), a secreted soluble receptor that binds RANKL, have demonstrated an essential role of these molecules i n osteoclastogenesis (i.e., bone remodeling). The biological importance of these molecules is underscored by the induction of severe osteoporosis by targeted disruption of osteoprotegerin an d by the induction of osteoperosis by targeted disruption of RANKL, the receptor activator of NF-κB, or by transgenic expression of osteoprotegerin. These results indicate that osteoclast formation may be attributed to the relative ratio of the receptor activator of NF-κB ligand to osteoprotegerin in the microenvironment of bone marrow, and alterations in this balance may be a major cause of bone loss in many metabolic bone disorders. Hence, RANK/RANKL/osteoprotegerin have a major role in bone diseases and cancer-induced bone destruction that are due to increased osteoclastic activity.
In addition to osteoporosis, recent reports suggest a potential role of these molecules in other diseases including rheumatoid arthritis, giant cell tumor of bone, Paget's disease, and familial expansile osteolysis (due to a mutation in exon 1 of th e receptor activator of NF-κB). A T cell lymphoproliferative disorder has also been identified in which dysregulation of th e receptor activator of NF-κB and RANKL contributes to the survival of malignant T cell clone.
It has been recognized that breast and prostate cancers have the ability to invade and grow as metastases in bone causing osteolytic lesions. In metastatic tumor mouse models i n which the tumor causes increased osteoclastogenesis and bone destruction, systemic administration of osteoprotegerin reduces tumor-mediated bone destruction and pain associated with b one cancer. Thus, development of drugs that target inhibition of th e receptor activator of NF-κB signaling are potential therapeutics for metabolic bone disorders and cancer.
The cytoplasmic domain of receptor activator of NF-κB interacts with TRAF family members, specifically TRAF1, 2, 3, 5 , and 6. Stimulation of the receptor activator of NF-κB activates members of the MAPK family (i.e., JNK, p38, ERK) and IKKs, which lead to activation of transcription factors API and NF-κB. The interactions of TRAF2, TRAF5, and TRAF6 with receptor activator of NF-κB have been reported and it was demonstrated th at receptor activator of NF-κB could activate both the NF-κB and JNK pathways. Subsequently, a novel TRAF6 binding motif w as identified in receptor activator of NF-κB that is distinct from th e TRAF2 and TRAF5 binding domains.
The TRAF6 binding domain in the receptor activator of NF-κB was sufficient for activation of NF-κB, suggesting th at TRAF2 and TRAF5 are not necessary for NF-κB activation. I n support of these findings, TRAF6-deficient mice develop osteopetrosis due to a defect in osteoclastogenesis, which is not found in either the TRAF2- or TRAF5-deficient mice. Since TRAF6 appears to be the critical adapter protein for the receptor activator of NF-κB signaling, the present invention develops a novel TRAF6 decoy peptide (T6DP) with and without a peptide leader sequence that allows for transversing cellular membrane. Evidence disclosed herein indicate that the TRAF6- decoy peptide inhibits RANKL signaling transduction and RANKL- mediated osteoclast differentiation, but only when the leader sequence is attached. These data indicate that targeted disruption of the interaction between receptor activator of NF-κB and TRAF6 would be useful as a therapeutic for metabolic bone disorders, leukemia, multiple myeloma, arthritis, and metastatic cancer of the bone.
The present invention is drawn to polypeptides th at inhibit signaling mediated by TNF receptor-associated factor 6 (TRAF6). These polypeptides comprise a TRAF6 binding domain and a leader signal sequence. A number of approaches may b e utilized by a person having ordinary skill in this art to search for TRAF6 inhibitory polypeptides; two approaches are, for example, screening peptide libraries or synthesizing overlapping peptides from the cytoplasmic domains of RANK or TRAF6. In one embodiment of the present invention, the polypeptide comprises of sequence selected from the group consisting of SEQ ID No. 1 9 and 20. Since TRAF6 also mediates signaling induced by a nu mber of molecules such as IL-1 , LPS, IL-18, and CD40L, the polypeptides claimed herein may inhibit RANKL mediated signaling as well a s signaling induced by these other molecules.
The polypeptides disclosed herein may contain a TRAF6 binding domain derived from CD40, Receptor Activator of NF-κB, IL-1 receptor-associated kinase 1 (IRAKI), IL-1 receptor- associated kinase 2 (IRAK2), IRAK-M or RIP2. Preferably, th e TRAF6 binding domain comprises of sequence selected from th e group consisting of SEQ ID No. 1-18.
The leader signal sequence attached to the TRAF6 binding domain in the polypeptides disclosed herein may b e derived from a number of different proteins. Representative leader signal peptides include Kaposi fibroblast growth factor signal sequence, HIV-1 Tat (48-60), D-amino acid-substituted HIV-1 Tat (48-60), arginine-substituted HIV-1 Tat (48-60) , Drosophila Antennapaedia (43-58), viral RNA binding peptide th at comprises 7 or more arginines, DNA binding peptide th at comprises 7 or more arginines and polyarginine polypeptide th a t has 6 to 8 arginines. These arginine-rich signal sequences that can be used for delivery of exogenous proteins into cells are well known in the art (39). For example, Futaki et al. (39) has reported various arginine-rich peptides that have translocation activities very similar to that of HIV-1 Tat (48-60). These arginine-rich peptides include HIV- 1 Rev (34-50), HTLV-II Rev (4- 16), brome mosaic virus Gag (7-25), flock house virus coat protein (35 -49) , human c-Fos (139- 164), human c-Jun (252-279) and yeast transcription factor GCN4 (231 -252).
The present invention is also drawn to a method of inhibiting Receptor Activator of NF-κB Ligand (RANKL)-induced osteoclast differentiation using the polypeptides disclosed herein, wherein inhibition of interaction between the receptor activator of NF-κB and TRAF6 by these polypeptides result in inhibition of RANKL-induced osteoclast differentiation. This method can b e used to inhibit osteoclast differentiation induced by breast cancer cells. The polypeptide can be applied to the cells by a number of methods well known in the art such as liposomes, viruses or o ther gene delivery vectors. For example, the ProVectin™ protein delivery reagent is a unique lipid-based formulation that allows delivery of the polypeptides disclosed herein or other bioactive molecules into a broad range of cell types.
In another aspect of the present invention, there is also provided a method of inhibiting osteoclast differentiation i n an individual by the polypeptide inhibitors disclosed herein. I n general, the individual would have a disease comprising a metabolic bone disorder, leukemia, arthritis, multiple myeloma, o r metastatic cancer of the bone.
The present invention also provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier an d the polypeptide inhibitors disclosed herein. The phras e "pharmaceutically acceptable" refers to molecular entities an d compositions that do not produce an allergic or similar untoward reaction when administered to a subject. The preparation of a n aqueous composition that contains a protein as an active ingredient is well understood in the art. A person having ordinary skill in this art would readily be able to determine, without undue experimentation, th e appropriate dosages and routes of administration of the active component of the present invention. When used in vivo for therapy, the active composition(s) of the present invention is administered to the patient or an animal in therapeutically effective amounts, i.e., amounts that inhibit RANKL-mediated osteoclast differentiation. See Remington's Pharmaceutical
Science, 17th Ed. ( 1990) Mark Publishing Co., Easton, Penn.; a n d Goodman and Gilman's: The Pharmacological Basis of Therapeutics 8th Ed (1990) Pergamon Press.
The present invention is further drawn to non-peptide analogs that mimic the functions of the polypeptide disclosed herein, wherein these non-peptide analogs inhibit signaling mediated by TRAF6. Low molecular weight, non-peptide molecules that mimic the inhibitory polypeptides disclosed herein can serve as robust tools to help establish the role of TRAF6- mediated signaling in models of physiological an d pathophysiological processes as well as serving as therapeutic agents in their own right. A number of reports have disclosed th e rationale and strategy for the design of low molecular weight, non-peptide molecules that are amenable to high resolution analysis and rapid modification (40-42). The pre s ent invention is also drawn to a method of inhibiting Receptor Activator of NF-κB Ligand (RANKL)-induced osteoclast differentiation using these non-peptide analogs, wherein inhibition of interaction between the receptor activator of NF-κB and TRAF6 by said non-peptide analogs result in inhibition of RANKL-induced osteoclast differentiation. This method can b e used to inhibit osteoclast differentiation induced by breast cancer cells.
The following examples are given for the purpose of illustrating various embodiments of the invention and are not meant to limit the present invention in any fashion. EXAMPLE 1
Reagents. Cell Lines, and Antibodies
The human embryonic kidney 293 cell line and th e mouse macrophage cell line RAW264.7 were obtained from th e American Type Culture Collection (Rockville, MD). The 293 cells were cultured in MEM supplemented with 10% fetal bovine s erum and antibiotics. RAW264.7 cells were cultured in DMEM-F12 supplemented with 10% fetal bovine serum and antibiotics. Monoclonal antibodies to phospho-ERK, p38, and JNK were purchased from New England Biolabs. Goat anti-rabbit IgG- conjugated horseradish peroxidase was purchased from BioRad Laboratories (Hercules, CA). Anti-JNKl and anti-IκB w ere purchased from Santa Cruz Biotechnology (Santa Cruz, CA). Goat anti-mouse IgG-conjugated horseradish peroxidase was purchased from Transduction Laboratories (Lexington, KY). Protein A/G Sepharose beads was purchased from Pierce (Rockford, IL), an d anti-FLAG was purchased from Sigma (St. Louis, MO). Staining for tartrate resistance acid phosphatase (TRAP) positive osteoclasts was performed essentially as described (30) or by using an acid phosphatase kit from Sigma.
EXAMPLE 2
Expression Plasmids
Expression plasmids encoding mouse FLAG-tagged
TRAF5 and TRAF6 (31) were provided by H. Nakano (Juntendo
University, Tokyo, Japan) and FLAG-tagged TRAF2 was provided by J. Ni (Human Genome Sciences, Inc.). Expression vectors and purification of GST-fusion proteins for GST, GST-receptor activator of NF-κB cytoplasmic domain, and GST-Jun (1-79) have b een previously described (7, 8). The expression vector of full-length murine RANKL (also known as TNF-related activation-induced cytokine (TRANCE) (pcDNA3.1 -TRANCE) was provided by Y. Choi (Rockefeller University, New York, NY).
To generate a bacterial expression vector for RANKL, specific 5' and 3' primers with Hindlll and Notl sites, respectively, were used to amplify the DNA which encodes residues 157-316 of RANKL from the pcDNA3-TRANCE template. The PCR product w a s digested with Hindlll/Notl and ligated in-frame with a HA-tag (N-terminal) and a histidine tag (C-terminal) into the expression vector pHB6 (Boerhinger Mannheim). Soluble RANKL w as expressed and purified using Ni-agarose.
EXAMPLE 3
Transient Transfections and Western Blotting
293 cells were plated at 0.6 x 106 cells/well on 6-well plates and transfected the next day as described (8). Total amount of plasmid DNA was kept constant by adding emp ty pCMVFLAGl vector. Cells and the conditioned supernatants w ere harvested 24-36 h after transfection. Lysates were prepared a s described (8). For western blot analysis, whole-cell lysates ( 15 - 30 μg) or proteins from GST-affinity precipitation were separated b y 8.5% SDS-PAGE, electroblotted onto nitrocellulose membranes, an d incubated with the indicated antibodies. The membranes w ere then developed using the enhanced chemiluminescence (ECL) system (Amersham, Chicago, IL).
EXAMPLE 4
In Vitro Osteoclast Differentiation
Primary bone marrow monocytes (BM) or RAW264.7 cells were cultured in 48-well dishes at a density of 1 x 1 05 cells/well or 2 x 103 cells/well, and then treated with the indicated factors at the beginning of the culture and during a medium change on day 3. Osteoclast formation was assessed by counting the total number of multi-nucleated (>3 nuclei), TRAP-positive cells present per well between day 7 and 10 (BM) or on day 5 (RAW264.7) (30).
EXAMPLE 5
GST-RANK Fusion Protein Affinity Binding Assays Equivalent amounts of GST or GST-RANK cytoplasmic domain (GST-RANKcd) fusion protein attached to 20 μl of glutathione agarose beads were mixed with lysates (50 μg) from 293 cells programmed to express the epitope-tagged TRAF protein and the indicated peptides in binding buffer (20 mM TRIS, pH 8, 150 mM NaCl, 1 mM DTT, 2 mM EDTA, and 0.1 % NP-40) an d allowed to rotate for 1 h at 4°C. The beads were collected b y centrifugation, washed three times in binding buffer, and then washed once in low-salt buffer (20 mM TRIS, pH 8, 50 mM NaCl, and 1 mM DTT). Bound proteins were eluted with addition of SDS- sample buffer and boiled. The eluted proteins were subjected to 7.5% SDS-PAGE and western blot analysis was performed with anti-FLAG antibodies.
EXAMPLE 6
Immune Complex Kinase Assays
Lysates were prepared from RAW cells stimulated with RANKL as indicated in the legends to the figures . Approximately 30 μg was then used for immunoprecipitation with indicated antibodies and protein A/G Sepharose beads for 1 h .
Beads were collected by centrifugation, washed three times i n lysis buffer, and then washed two times in low-salt buffer. JNK activity was analyzed using exogenous GST-Jun (1-79) as a substrate as previously described (8). Kinase activity w a s quantitated using a Phospholmager and Imagequant Software
(Molecular Dynamics, Sunnyvale, CA).
EXAMPLE 7
Electrophoretic Mobility Shift Assays (EMSA)
Nuclear extracts were prepared from transfected cells essentially as described (34). Equivalent amounts of nuclear protein were used in an EMS A reaction with 32P-labeled NF-κB oligonucleotide from HIV-LTR as described (34). NF-κB activation was quantitated using a Phospholmager and Imagequant Software. EXAMPLE 8
TRAF6 Binding Domain
A novel TRAF6 binding domain in RANK, which binds to only TRAF6 but not TRAF2 or TRAF5, has been identified previously (8). When transfected in 293 cells, this region of th e receptor activator of NF-κB was sufficient for activation of NF-κB (8). Structure-based sequence alignment of TRAF6-binding sites in human and mouse CD40 and RANK led to the definition of a TRAF6-binding motif PxExx(Ar/Ac) (Ar for aromatic and Ac for acidic residues) (Figure 1A). Careful examination of the RANK sequence indicates that there are three potential TRAF6-binding sites (Figure 1A).
TRAF6 is the only TRAF family member th at participates in the signal transduction of both the TNF receptor superfamily and the interleukin-1 receptor (IL- l R)/Toll-like receptor (TLR) superfamily. The best-characterized TRAF6 signaling pathway for the IL-1R/TLR superfamily involves IRAK, an adapter kinase upstream of TRAF6. Upon receptor stimulation, IRAK becomes oligomerized and interacts with TRAF6 (43). It w as found that full-length IRAK contains three potential TRAF6- binding sites (Figure 1A). Two IRAK homologues, IRAK-2 an d IRAK-M, also contain two and a single potential TRAF6-binding site, respectively (Figure 1A). This is in keeping with th e implicated role of IRAK-2 and IRAK-M in IL-1 signaling and th e role of IRAK-2 in TLR4 signaling. In addition, it was found th at the kinase RIP2, which can activate NF-κB and induce cell death, also contains a putative TRAF6-binding site (Figure 1A). The presence of TRAF6-binding sites in these various molecules suggests that TRAF6 may play a role in mediating multiple signaling cascades.
Delivery of peptides or proteins across cellular membranes can be achieved by covalent attachment of th e peptide to molecules that can freely pass through the membrane (35). For example, the hydrophobic domain of Kaposi fibroblast growth factor signal sequence was attached to the nuclear localization signal from the p50 subunit of NF-κB to allow for translocation across the membrane (36). In the present invention, this hydrophobic sequence was attached to peptides derived from murine RANK, namely L-T6DP-1 which contains the known TRAF6 binding domain and L-T6DP-2 which contains a similar motif (Fig. IB). In addition, both of these peptides were synthesized without the leader sequence, resulting in peptides that were not be able to transverse cellular membrane (Fig. IB).
EXAMPLE 9
TRAF6-decoy Peptides Inhibit RANKL-mediated Osteoclast Differentiation in RAW264.7 Cells
The mouse macrophage cell line RAW264.7 express th e receptor activator of NF-κB on their cell surface and w h en stimulated with the receptor activator of NF-κB ligand differentiate into multi-nucleated, tartrate resistant acid phosphatase (TRAP) positive osteoclasts after 4-5 days (Fig. 2A). To determine whether L-T6DP-1 could inhibit RANKL-mediated osteoclast differentiation, RAW264.7 cells were co-cultured with increasing concentrations of either L-T6DP-1 or T6DP-1 and 3 0 ng/ml RANKL for 4 days. As indicated in Figure 2B, TRAF6decoy peptide without leader sequence failed to block RANKL-mediated osteoclast differentiation; however, treatment with L-T6DP-1 caused a do se- dependent inhibition of osteoclast differentiation. Although th e cells were TRAP positive after treatment with 100 μM L-T6DP- 1 , multi-nucleated osteoclast were not observed (Fig. 2B). Furthermore, treatment of RAW264.7 cells with RANKL in th e presence of either L-T6DP-1 or L-T6DP-2 caused a dose- dependent decrease of TRAP positive osteoclasts (Fig. 2C), although L-T6DP-1 was much more efficient than L-T6DP-2.
EXAMPLE 10
TRAF6decoy Peptides Inhibit RANKL-mediated Osteoclast Differentiation in Bone Marrow-derived Mouse Monocytes
To further support the results obtained from th e RAW264.7 cell line, the ability of these peptides to inhibit RANKL- mediated osteoclast differentiation in primary mouse-derived monocytes was tested. Costimulation of bone marrow -derived monocytes with RANKL and M-CSF cause osteoclast differentiation after 7-10 days, as determined by staining multi-nucleated, TRAP positive osteoclasts (30). Similar to the results with RAW264.7 cells, both L-T6DP- 1 and L-T6DP-2 inhibited the development of TRAP positive osteoclast in a dose-dependent manner and L- T6DP-1 was much more efficient (Fig. 3). TRAF6decoy peptides without the leader sequences failed to inhibit osteoclast differentiation (Fig. 3), indicating that the peptides were not toxic to the cells. Although the cells were TRAP positive after treatment with 30 μM L-T6DP-1 , multi-nucleated osteoclast w ere not observed and at 100 μM L-T6DP-1, no osteoclasts w ere observed (Fig. 3B). Taken together, these results indicate th at interaction of TRAF6 with RANK is essential for RANKL-mediated osteoclast differentiation.
EXAMPLE 11
TRAF6Decov Peptide Inhibits RANKL-mediated NF-kB Activation in RAW264.7 Cells
A mutant form of the receptor activator of NF-κB which contains only the TRAF6 binding domain is sufficient to activate NF-κB (8), and a dominant negative TRAF6 inhibits RANK- mediated NF-κB activation in 293 cells (9). To investigate th e effect of the TRAF6 binding peptides on RANKL-induced NF-κB activation, NF-κB activation was examined in RAW264.7 cells th at activated NF-κB when stimulated with RANKL (37). RANKL- stimulated RAW264.7 cells activated NF-κB as indicated by a gel mobility shift assay (Fig. 4A). NF-κB activation was suppressed i n a dose-dependent manner only by pre-treatment with L-T6DP- 1 (Fig. 4A). The levels of IκBα coincided with the activation an d repression of NF-κB as indicated in Figure 4B. These data indicate that L-T6DP-1 specifically inhibits RANKL-mediated NF-κB activation in RAW264.7 cells. EXAMPLE 12
TRAF6Decov Peptide Specifically Inhibits TRAF6 Binding to th e RANK Cytoplasmic Domain The cytoplasmic domain of RANK interacts with m an y
TRAF molecules, including TRAF1, 2, 3, 5, and 6 (7-9, 11, 12) . While TRAF1, 2, 3, and 5 interact with the c-terminal tail of receptor activator of NF-κB, TRAF6 interacts with a membrane proximal region of the cytoplasmic domain of the receptor activator of NF-κB. To confirm that the TRAF6-binding peptides disclosed herein were specifically inhibiting TRAF6 interaction with RANK, a competitive GST-pull down assay was performed.
Cellular extracts containing FLAG-tagged TRAF2, TRAF5, or TRAF6 were mixed with GST-RANK cytoplasmic domain fusion protein in the presence and absence of either L-T6DP-1 o r L-T6DP-2. If these peptides competed for the TRAF molecules, less FLAG-tagged protein would be observed in the western blots. As shown in Figure 4, neither L-T6DP-1 nor L-T6DP-2 inhibited TRAF2 and TRAF5 binding to the cytoplasmic domain of RANK (Fig. 4C). Only L-T6DP-1 inhibited TRAF6 interaction with th e cytoplasmic domain of RANK (Fig. 4C). These data indicate th at the leader sequence did not interfere with interaction of th e receptor activator of NF-κB with TRAFs and that L-T6DP- 1 specifically inhibited RANK'S interaction with TRAF6. EXAMPLE 13
TRAF6Decov Peptide Specifically Inhibits JNK. ERK. and p38 Kinase Activation bv RANKL in RAW264.7 Cells Stimulation of the receptor activator of NF-κB activates members of the MAPK family including JNK, ERK and p38 kinase. The ability of the TRAF6 binding peptides to inhibit the receptor activator of NF-κB ligand-induced JNK, ERK, and p38 kinase activation was examined in RAW264.7 cells. In vitro JNK kinase assays indicated JNK is activated by RANKL and only treatment with L-T6DP- 1 peptide was capable of inhibiting RANKL-mediated JNK activation (Fig. 5A). Similar to the results with JNK, only L- T6DP- 1 was able to block ERK (Figure 5B) and p38 kinase (Figure 5C) activation induced by the receptor activator of NF-κB ligand in RAW264.7 cells. Taken with the results disclosed above, th e present invention demonstrates that L-T6DP- 1 is able to suppres s the receptor activator of NF-κB ligand-mediated osteoclast differentiation and the receptor activator of NF-κB ligand-initiated early signaling including NF-κB, JNK, ERK, and p38 kinase activation.
EXAMPLE 14
Induction of Osteoclast Formation By Breast Cancer Cells And Inhibition By TRAF6-Decoy Peptide
Breast cancer is the most common female malignancy in the U.S. and is the second leading cause of cancer death i n women. Women with breast cancer are at risk for bone metastases. Five to ten percent of patients with breast cancer initially present with metastatic disease to the bone. Patients with osteolytic bone disease from metastatic breast cancer are a t increased risk for pathologic fractures, bone pain, cord compression and hypercalcemia. Current standard of care for treating bone metastases is bisphosphonate therapy which delays skeletal events but does not completely prevent them. I n addition, not all patients respond to this treatment. While a m ore effective treatment is desired, a further biological and molecular dissection of this disease is required. In fact, recently it w a s demonstrated that osteoprotegerin (OPG) inhibits osteolysis an d decreases tumor burden in nude mouse models injected with breast cancer cells. The ability of breast cancer cells to induce osteoclast formation and the expression of receptor activator of NF-κB / t h e receptor activator of NF-κB ligand/osteoprotegerin in breas t cancer cells are not well defined. Few reports have demonstrated the ability of breast cancer cell lines to influence osteoclast differentiation and function; however, no evidence has b een described for the direct involvement of the receptor activator of NF-κB ligand in this process. As indicated below, there is evidence to support the hypothesis that breast cancer cells directly induce osteoclast differentiation and function in the absence of osteoblast/stromal cells. Through the understanding of th e biological and molecular role of the receptor activator of NF-κB ligand in breast cancer cells in the bone microenvironment an d development of novel inhibitors of osteoclast formation a s described herein, alternative therapeutic approaches o r combination therapy may be developed to treat breast cancer patients with bone metastases.
In the present example, a co-culture assay system w as developed for RAW cells and the osteoblast-like cell (osteosarcoma MG-63) which has been shown to express the receptor activator of NF-κB ligand and cause osteoclast differentiation. The number of MG-63 cells was critical for inducing RAW cells to differentiate into osteoclasts. These observations lead to a direct inverse relationship between the number of RAW cells to MG-63 cells which is required for the formation of osteoclasts. Results using RAW cells co-cultured with MG-63 (data not shown) or with breast cancer cell lines (i.e., T47D and MDA-MB-468) indicated that these breast cancer cells could in fact cause RAW cells to form TRAP"1", multi-nucleated osteoclasts after 4 days (Fig. 6A) similar to RAW cells stimulated with the receptor activator of NF-κB ligand. In addition, when the breast cancer cell lines were grown in tissue culture inserts where they were separated from RAW cells by a membrane, osteoclast still formed, suggesting that direct cell-to- cell contact was not required for osteoclast differentiation (data not shown). When grown on synthetic bone slides, the osteoclasts derived from the co-culture assays were able to cause bone resorption (Fig. 6B).
When L-T6DP-1 was added to these co-culture assays, the ability of the breast cancer cells to induce osteoclast differentiation of RAW cells was abolished, whereas T6DP-1 h ad no effect (Fig. 6A). Normal breast epithelial cells, MCF-10A, failed to induce osteoclast differentiation of RAW; however, osteoclasts did form if exogenous the receptor activator of NF-κB ligand w a s added to these co-cultures (Fig. 6C). Collectively, these data indicate that breast cancer cell lines, but not normal breas t epithelial cells, can directly induce osteoclast differentiation in th e absence of osteoblast/stromal cells and that L-T6DP-1 can inhibit this process.
EXAMPLE 15
Strategy to Discover Non-peptide Analogues that Inhibit RANK- TRAF6 Interaction An ELISA-based method similar to one described previously (J. Biol. Chem., 276: 12235-12240, 2001) can be used to discover small molecules that inhibit RANK-TRAF6 interaction. Briefly, peptides comprising the T6DP are biotinylated, dissolved in TRIS-buffered saline (50 mM TRIS pH 7.5, 150 mM NaCl), an d added to wells in NeutrAvidin-coated 96-well microtiter plates . The plates are shaken overnight at 4°C and then rinsed with TBS followed by TBS-BT (TBS containing 0.1 % bovine serum albumin (BSA) and 0.1 % Tween 20). A solution containing a test small molecule from a library is then added to the well. A solution containing 6x-histidine-tagged TRAF6 (309-522) is then added to each well. The plates are incubated for 1 h at room temperature and then washed 3 times with TBS-BT. An antibody directed against the C-terminus of TRAF6 is then added to the wells a n d the plates are incubated for 1 hour at room temperature, followed by 3 washes with TBS-BT. A secondary antibody consisting of goat anti-mouse alkaline phosphatase is then added to each well and the plates are incubated for 1 hour at room temperature, followed by 3 washes with TBS-BT. The plates are then assayed for alkaline phosphatase activity using a fluorescent plate reader as previously described (Darnay et al., J. Biol. Chem. 274: 7724- 7731 , 1999).
The following references were cited herein:
I . Darnay and Aggarwal. 1999. Ann. Rheum. Dis. 58 Suppl 1 :12. 2. Darnay and Aggarwal. 1997. J Leukoc. Biol. 61 :559.
3. Wallach et al. 1999. Ann. Rev. Immunol. 17:331.
4. Hofbauer et al. 2000. J. Bone Miner. Res. 15:2.13
5. Kong et al. 1999. Nature 397:315.
6. Wong et al. 1997. J Exp. Med. 186:2075. 7. Darnay et al. 1998. J Biol. Chem. 273:20551.
8. Darnay et al. 1999. J Biol. Chem. 274:7724.
9. Galibert et al. 1998. J Biol. Chem. 273:34120.
10. Kim et al. 1999. FEBS Lett. 443:297.
I I . Hsu et al. 1999. Proc. Natl. Acad. Sci. U S A 96:3540. 12. Wong et al. 1998. J Biol. Chem. 273:28355.
13. Pahl. 1999. Oncogene 18:6853.
14. Arch et al. 1998. Genes Dev. 12:2821.
15. McWhirter et al. 1999. Proc. Natl. Acad. Sci. U S A 96:8408.
16. Park et al. 1999. Nature 398:533. 17. Pullen et al. 1999. J Biol. Chem. 274: 14246.
18. Wong et al. 1999. Mol. Cell 4: 1041.
19. Bucay et al. 1998. Genes Dev. 12: 1260.
20. Mizuno et al. 1998. Biochem. Biophys. Res. Commun. 247 : 610. 21. Li et al. 2000. Proc. Natl. Acad. Sci. U S A 97: 1566.
22. Dougall et al. 1999. Genes Dev. 13:2412.
23. Lomaga et al. 1999. Genes Dev. 13: 1015.
24. Naito et al. 1999. Genes Cells 4:353.
25. Kobayashi et al. 2001. EMBO J 20: 1271. 26. Soriano et al. 1991. Cell 64:693.
27. Johnson et al. 1992. Cell 71 :577.
28. Iotsova et al. 1997. Nat. Med. 3: 1285.
29. Franzoso et al. 1997. Genes Dev. 11 :3482. 30. Shevde et al. 2000. Proc. Natl. Acad. Sci. USA 97:7829.
31. Akiba et al. 1998. J Biol. Chem. 273: 13353.
32. Hu et al. 1996. Genes Dev. 10:2251.
33. Yao et al. 1999. J Biol. Chem. 274:2118.
34. Haridas et al. 1998. J Immunol. 160:3152. 35. Schwarze et al. 2000. Trends Cell Biol. 10:290.
36. Yan et al. 2000. J Biol. Chem. 275: 16774.
37. Wei et al. 2001. Endocrinology 142: 1290.
38. Ishida et al. 1996. J Biol. Chem. 271 :28745.
39. Futaki et al. 2001. J Biol. Chem. 276:5836. 40. Horwell. 1995. Trends Biotechnol. 13: 132.
41. Wexler et al. 1992. Am. J. Hypertens. 5:209S.
42. Saragovi et al. 1992. Biotechnology 10:773.
43. Wesche et al. 1999. J Biol. Chem. 274: 19403.
Any patents or publications mentioned in this specification are indicative of the levels of those skilled in the art to which the invention pertains. Further, these patents an d publications are incorporated by reference herein to the s ame extent as if each individual publication is specifically an d individually indicated to be incorporated by reference. One skilled in the art will appreciate readily that th e present invention is well adapted to carry out the objects an d obtain the ends and advantages mentioned, as well as those objects, ends and advantages inherent herein. The present examples, along with the methods, procedures, treatments , molecules, and specific compounds described herein are pres ently representative of preferred embodiments, are exemplary, and are not intended as limitations on the scope of the invention. Changes therein and other uses will occur to those skilled in the art which are encompassed within the spirit of the invention as defined b y the scope of the claims.

Claims (25)

WHAT IS CLAIMED IS:
1. A polypeptide that inhibits signaling mediated by TNF receptor-associated factor 6 (TRAF6), wherein said polypeptide comprises a TRAF6 binding domain and a leader signal sequence.
2. The polypeptide of claim 1, wherein said leader signal sequence comprises a polypeptide selected from the group consisting of Kaposi fibroblast growth factor signal sequence, HIV- 1 Tat (48-60), D-amino acid-substituted HIV-1 Tat (48 -60), arginine-substituted HIV-1 Tat (48-60), Drosophila
Antennapaedia (43-58), viral RNA binding peptide that comprises 7 or more arginines, DNA binding peptide that comprises 7 o r more arginines and polyarginine polypeptide that has 6 to 8 arginines .
3. The polypeptide of claim 2, wherein said viral RNA binding peptide is selected from the group consisting of HIV- 1 Rev (34-50), HTLV-II Rev (4-16), brome mosaic virus Gag ( 7 - 25) and flock house virus coat protein (35-49).
4. The polypeptide of claim 2, wherein said DNA binding peptide is selected from the group consisting of human c - Fos ( 139- 164), human c-Jun (252-279) and yeast transcription factor GCN4 (231-252).
5. The polypeptide of claim 1, wherein said TRAF6 binding domain is a TRAF6 binding domain from a protein selected from the group consisting of CD40, Receptor Activator of NF-κB, IL-1 receptor-associated kinase 1 (IRAKI), IL-1 receptor- associated kinase 2 (IRAK2), IRAK-M and RIP2.
6. The polypeptide of claim 5, wherein said TRAF6 binding domain comprises a sequence selected from the group consisting of SEQ ID No. 1-18.
7. The polypeptide of claim 1, wherein said polypeptide comprises a sequence selected from the group consisting of SEQ ID No. 19 and 20.
8. A method of inhibiting receptor activator of NF- KB ligand (RANKL)-induced osteoclast differentiation, comprising the step of: applying to cells the polypeptide of claim 1, wherein inhibition of interaction between Receptor Activator of NF-κB an d TRAF6 by said polypeptide results in inhibition of the receptor activator of NF-κB ligand-induced osteoclast differentiation.
9. The method of claim 8, wherein said polypeptide is delivered to said cells by a mean selected from the group consisting of liposomes, a virus and a gene delivery vector.
10. The method of claim 8, wherein said osteoclast differentiation is induced by breast cancer cells.
1 1 . A method of inhibiting osteoclast differentiation in an individual in need of such treatment, comprising the step of: applying to said individual the polypeptide of claim 1 , wherein inhibition of interaction between Receptor Activator of NF-κB and TRAF6 by said polypeptide results in inhibition of osteoclast differentiation.
12. The method of claim 11, wherein said individual has a disease selected from the group consisting of metabolic bone disorders, leukemia, multiple myeloma, arthritis, and metastatic cancer of the bone.
13. A pharmaceutical composition comprising th e polypeptide of claim 1 and a pharmaceutically acceptable carrier.
14. A non-peptide analog that mimics the function of a polypeptide comprising a TNF receptor-associated factor 6
(TRAF6) binding domain and a leader signal sequence, wherein said polypeptide inhibits signaling mediated by TRAF6.
15. The non-peptide analog of claim 14, wherein said leader signal sequence comprises a polypeptide selected from th e group consisting of Kaposi fibroblast growth factor signal sequence, HIV-1 Tat (48-60), D-amino acid-substituted HIV-1 Tat (48-60), arginine-substituted HIV-1 Tat (48-60), Drosophila Antennapaedia (43-58), viral RNA binding peptide that comprises 7 or more arginines, DNA binding peptide that comprises 7 o r more arginines and polyarginine polypeptide that has 6 to 8 arginines .
16. The polypeptide of claim 15, wherein said viral RNA binding peptide is selected from the group consisting of HIV- 1 Rev (34-50), HTLV-II Rev (4-16), brome mosaic virus Gag ( 7 - 25) and flock house virus coat protein (35-49).
17. The polypeptide of claim 15, wherein said DNA binding peptide is selected from the group consisting of human c - Fos (139-164), human c-Jun (252-279) and yeast transcription factor GCN4 (231-252).
18. The non-peptide analog of claim 14, wherein said TRAF6 binding domain is a TRAF6 binding domain from a protein selected from the group consisting of CD40, Receptor Activator of NF-KB, IL-1 receptor-associated kinase 1 (IRAKI), IL-1 receptor- associated kinase 2 (IRAK2), IRAK-M and RIP2.
19. The non-peptide analog of claim 18, wherein said TRAF6 binding domain comprises a sequence selected from th e group consisting of SEQ ID No. 1-18.
20. The non-peptide analog of claim 14, wherein said polypeptide comprises a sequence selected from the group consisting of SEQ ID No. 19 and 20.
21 . A method of inhibiting receptor activator of NF-
KB ligand-induced osteoclast differentiation, comprising the s tep of: applying to cells the non-peptide analog of claim 14, wherein inhibition of interaction between Receptor Activator of NF- B and TRAF6 by said non-peptide analog results in inhibition of the receptor activator of NF-κB ligand-induced osteoclast differentiation.
22. The method of claim 21 , wherein said osteoclast differentiation is induced by breast cancer cells.
23. A method of inhibiting osteoclast differentiation in an individual in need of such treatment, comprising the step of: applying to said individual the non-peptide analog of claim 14, wherein inhibition of interaction between Receptor Activator of NF-κB and TRAF6 by said non-peptide analog results in inhibition of osteoclast differentiation.
24. The method of claim 23, wherein said individual has a disease selected from the group consisting of metabolic bone disorders, leukemia, multiple myeloma, arthritis, and metastatic cancer of the bone.
25. A pharmaceutical composition comprising th e non-peptide analog of claim 14 and a pharmaceutically acceptable carrier.
AU2002256518A 2001-05-11 2002-05-10 Inhibitors of receptor activator of NF-kappaB and uses thereof Ceased AU2002256518B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US29042901P 2001-05-11 2001-05-11
US60/290,429 2001-05-11
PCT/US2002/014842 WO2002092623A1 (en) 2001-05-11 2002-05-10 INHIBITORS OF RECEPTOR ACTIVATOR OF NF-κB AND USES THEREOF

Publications (2)

Publication Number Publication Date
AU2002256518A1 true AU2002256518A1 (en) 2003-05-01
AU2002256518B2 AU2002256518B2 (en) 2007-07-19

Family

ID=23115949

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2002256518A Ceased AU2002256518B2 (en) 2001-05-11 2002-05-10 Inhibitors of receptor activator of NF-kappaB and uses thereof

Country Status (7)

Country Link
US (1) US6998383B2 (en)
EP (1) EP1385872A4 (en)
JP (1) JP4603240B2 (en)
AU (1) AU2002256518B2 (en)
CA (1) CA2447431A1 (en)
NZ (1) NZ529314A (en)
WO (1) WO2002092623A1 (en)

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU9303001A (en) 2000-09-22 2002-04-02 Immunex Corp Screening assays for agonists or antagonists of receptor activat or of nf-kb
US20040167072A1 (en) * 2001-05-11 2004-08-26 Aggarwal Bharat B. Inhibitors of receptor activator of NF-kappaB and uses thereof
WO2003090681A2 (en) * 2002-04-24 2003-11-06 Research Development Foundation SYNERGISTIC EFFECTS OF NUCLEAR TRANSCRIPTION FACTOR NF-κB INHIBITORS AND ANTI-NEOPLASTIC AGENTS
WO2004000229A2 (en) * 2002-06-24 2003-12-31 Research Development Foundation Treatment of human multiple myeloma by curcumin
US20050013812A1 (en) * 2003-07-14 2005-01-20 Dow Steven W. Vaccines using pattern recognition receptor-ligand:lipid complexes
US7462646B2 (en) * 2003-08-26 2008-12-09 Research Development Foundation Osteoclastogenesis inhibitors and uses thereof
PL2012773T3 (en) 2006-04-22 2012-12-31 Harbor Biosciences Inc Drugs and uses
WO2008044824A1 (en) * 2006-10-13 2008-04-17 Seoul National University Industry Foundation Antibodies to ip-10 for treating bone diseases with bone destruction
JP5379806B2 (en) * 2007-10-29 2013-12-25 へリックス バイオメディックス インコーポレイテッド Tetrapeptide for protective skin care
TWI573806B (en) 2008-04-17 2017-03-11 巴克斯歐塔公司 Biologically active peptides
EP2437772B1 (en) 2009-06-05 2017-12-06 13Therapeutics, Inc. Immunoregulatory peptides and methods of use
KR101180431B1 (en) * 2010-05-27 2012-09-06 성균관대학교산학협력단 Peptide Inhibiting RANKL-RANK Interaction and Pharmaceutical Composition Comprising the Same
WO2012138879A2 (en) 2011-04-06 2012-10-11 13Therapeutics, Inc. Peptides for the treatment of hearing
US9914770B2 (en) 2013-04-30 2018-03-13 Intas Pharmaceuticals Ltd Cloning, expression and purification method for the preparation of ranibizumab
KR20150050646A (en) * 2013-10-29 2015-05-11 삼성전자주식회사 Fusion peptide and use thereof for cell membrane penetrating

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6271349B1 (en) * 1996-12-23 2001-08-07 Immunex Corporation Receptor activator of NF-κB
EP2003203A1 (en) * 1996-12-23 2008-12-17 Immunex Corporation Ligand for receptor activator of nf-kappa b, ligand is member of TNF superfamily
US5843678A (en) 1997-04-16 1998-12-01 Amgen Inc. Osteoprotegerin binding proteins
ATE517916T1 (en) 1998-05-14 2011-08-15 Immunex Corp METHOD FOR INHIBITING THE EFFECT OF OSTEOCLASTS
US6610820B1 (en) * 1999-10-12 2003-08-26 University Of Lausanne Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US20040167072A1 (en) * 2001-05-11 2004-08-26 Aggarwal Bharat B. Inhibitors of receptor activator of NF-kappaB and uses thereof

Similar Documents

Publication Publication Date Title
US6998383B2 (en) Inhibitors of receptor activator of NF-κB and uses thereof
Fridell et al. Differential activation of the Ras/extracellular-signal-regulated protein kinase pathway is responsible for the biological consequences induced by the Axl receptor tyrosine kinase
US8217138B2 (en) Anti-inflammatory compounds and uses thereof
Veillette et al. Regulation of the enzymatic function of the lymphocyte-specific tyrosine protein kinase p56lck by the non-catalytic SH2 and SH3 domains
US5563039A (en) TNF receptor-associated intracellular signaling proteins and methods of use
AU2002256518A1 (en) Inhibitors of receptor activator of NF-kappaB and uses thereof
AU732793B2 (en) Modulators of TNF receptor associated factor (TRAF), their preparation and use
EP1005540A1 (en) Ikk-beta proteins, nucleic acids and methods
WO2001068666A1 (en) Tcl1 enhances akt kinase activity and mediates its nuclear translocation
US20120329720A1 (en) Anti-inflammatory compounds and uses thereof
DiMaio et al. Virocrine transformation: the intersection between viral transforming proteins and cellular signal transduction pathways
KR100565819B1 (en) Compounds that inhibit interaction between signal-transducing proteins and the glgfpdz/dhr domain and uses thereof
US20040167072A1 (en) Inhibitors of receptor activator of NF-kappaB and uses thereof
CA2172490A1 (en) Receptor recognition factors, protein sequences and methods of use thereof
US6492124B1 (en) Trance activated signal transduction pathways in osteoclasts
US20040109854A1 (en) Novel traf6 inhibiting protein
AU767967B2 (en) Modulators of TNF receptor associated factor (TRAF), their preparation and use
Summy Functional domain contributions to signaling specificity between the non-receptor tyrosine kinases c-Src and c-Yes
Turner Signal transduction by the high affinity receptor for immunoglobulin E, FcϵRI, in the mast cell line RBL-2H3
Turner Signal transduction by the high affinity receptor for immunoglobulin E, FcepsilonRI, in the mast cell line RBL-2H3
Mayer et al. Probing the Tyrosine Phosphorylation State in Breast Cancer by Src Homology 2 Domain Binding
IL126428A (en) Modulators of tnf receptor associated factor (traf), their preparation and use