CA2325354A1 - Highly active forms of interferon regulatory factor proteins - Google Patents

Highly active forms of interferon regulatory factor proteins Download PDF

Info

Publication number
CA2325354A1
CA2325354A1 CA002325354A CA2325354A CA2325354A1 CA 2325354 A1 CA2325354 A1 CA 2325354A1 CA 002325354 A CA002325354 A CA 002325354A CA 2325354 A CA2325354 A CA 2325354A CA 2325354 A1 CA2325354 A1 CA 2325354A1
Authority
CA
Canada
Prior art keywords
irf
ser
modified
protein
regulatory factor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002325354A
Other languages
French (fr)
Inventor
John Hiscott
Rongtuan Lin
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.)
Sir Mortimer B Davis Jewish General Hospita
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from CA002234588A external-priority patent/CA2234588A1/en
Application filed by Individual filed Critical Individual
Priority to CA002325354A priority Critical patent/CA2325354A1/en
Publication of CA2325354A1 publication Critical patent/CA2325354A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Abstract

The present invention relates to IRF proteins that have been modified in the carboxy-terminus domain (transactivation domain) by modification of serine and/or threonine sites. Modification may be achieved by phosphorylation of serine and/or threonine, or by replacement of serine and/or threonine residues with residues having acidic side-chains, preferably carboxylic acid-containing side-chains, such as aspartic acid or glutamic acid residues. Such modified proteins may be mutants of IRF-3 and IRF-7, including chimeric proteins having portions of both IRF-3 and IRF-7, and post-translationally modified (phosphorylated) IRF-3 protein, the phosphorylation being induced by Sendai virus infection. More specifically, the present invention provides a modified interferon regulatory factor (IRF) protein, the protein comprising at least one modified serine or threonine phosphoacceptor site in the carboxy-terminus domain, preferably wherein cytokine gene activation by the modified IRF is increased relative to cytokine gene activation by a corresponding wild type IRF protein. The invention also provides for pharmaceutical compositions containing IRF protein, and uses of the protein, nucleotide sequence encoding it, and pharmaceutical compositions containing it.

Description

HIGHLY ACTIVE FORMS OF INTERFERON REGULATORY FACTOR PROTEINS
BACKGROUND OF THE INVENTION
Interferons (IFNs) are a large family of multifunctional secreted proteins involved in antiviral defence, cell growth regulation and immune activation (63).
Virus infection induces the transcription and synthesis of multiple IFN genes (33,52,63); newly synthesized IFN interacts with neighbouring cells through cell surface receptors and the JAK-STAT signalling pathway, resulting in the induction of over 30 new cellular proteins that mediate the diverse functions of the IFNs (17,35,39,58). Among the many virus- and IFN-inducible proteins are the growing family of IRF
transcription factors, the Interferon Regulatory Factors (IRFs) .
IRF-1 and IRF-2 are the best characterized members of this family, originally identified by studies of the transcriptional regulation of the human IFN-Q gene (22,23,30,47). Their discovery preceded the recent expansion of this group of IFN-responsive proteins which now include seven other members: IRF-3, IRF-4/Pip/ICSAT, IRF-5, IRF-6, IRF-7, ISGF3~y/p48 and ICSBP (48). Structurally, the Myb oncoproteins share homology with the IRF family, although its relationship to the IFN system is unclear (62). Recent evidence also demonstrates the presence of virally encoded analogue of cellular IRFs - vIRF in the genome of human herpes virus 8 (HHVB) (55).
The presence of IRF-like binding sites in the promoter region of the IFNA and IFNB genes implicated the IRF
factors as essential mediators of the induction of IFN genes.
The original results of Harada et al. (30,32) indicated that IFN gene induction was activated by IRF-1, while the related IRF-2 factor suppressed IFN expression. However, the essential role of IRF-1 and IRF-2 in the regulation of IFNA and IFNB gene expression has become controversial with the observation that mice containing homozygous deletion of IRF-1 or IRF-2, or fibroblasts derived from these mice, induced IFNA and IFNB gene expression after virus infection to the same level as the wild-type mice or cells (44).
On the other hand, IRF-1 was shown to have an important role in the antiviral effects of IFNs (44,54). IRF-1 binds to the ISRE element present in many IFN-inducible gene promoters and activates expression of some of these genes (54).
However, activation of ISG genes by IFNA and IFNB was shown to be mediated generally by the multiprotein ISGF3 complex (31,36,38). The binding of this complex to DNA is mediated by another member of the IRF family, ISGF3~y/p48, which in IFN-treated cells interacts with phosphorylated STAT1 and STAT2 transcription factors forming the heterotrimeric complex ISGF3 (8,39,62). The homozygous deletion of p48 in mice abolished the sensitivity of these mice to the antiviral effects of IFNs, and virus-infected macrophages from p48-/- mice showed an impaired induction of IFNA and IFNB genes (31).
Several other members of the IRF family have been identified. The ICSBP gene is expressed exclusively in the cells of the immune system (18,64) and its expression can be enhanced by IFN~y. ICSBP was shown to form a complex with IRF-1 and inhibit the transactivating activity of IRF-1 (9,59). The homozygous deletion of ICSBP in mice leads to defects in myeloid cell lineage development and chronic myelogous leukemia (34). Another lymphoid specific Pip/LSIRF/IRF-4 was identified (19,43,66) that interacts with phosphorylated PU.1, a member of the Ets family of transcription factors (15). The Pip/PU.1 heterodimer can bind to the immunoglobulin light chain enhancer and function as a B cell specific transcriptional activator.
Expression of Pip/LSIRF was induced by antigenic stimulation but not by IFN, and Pip/LSIRF/IRF-4 -/- mice failed to develop mature T and B cells (46). A novel member of the IRF family was recently identified by its ability to bind to an ISRE-like element in the promoter region of the Qp gene of EBV (69).
Another unique member of the human IRF family, IRF-3 was characterized recently (2). The IRF-3 gene encodes a 55-kDa protein which is expressed constitutively in all tissues. IRF-3 was originally identified as a member of the IRF family based on homology with other IRF family members and on binding to the ISRE of the ISG15 promoter. The relative levels of IRF-3 mRNA do not change in virus-infected or IFN-treated cells. Recombinant IRF-3 binds to the ISRE element of the IFN-induced gene ISG-15 and stimulates this promoter in transient expression assays. In previous studies, it has been shown that IRF-3 binds to the IE and PRDIII regions of the IFNA
and IFNB promoters respectively, but has different effects on their transcriptional activity (56). While the induction of the IFNA4 promoter activated by IRF-1 or virus infection was inhibited in the presence of IRF-3, the fusion protein containing the IRF-3 DNA binding domain and the RelA(p65) transactivation domain effectively activated both IFNA and IFNB
promoters. In contrast, co-expression of IRF-3 and RelA
plasmids transactivated the IFNB gene promoter, but not the promoter of the IFNA4 gene {56).
Most of the IRF family members so far identified appear to have specific and critical functions in lymphoid cells and/or their action is related to the signalling pathway induced by IFN or viruses. Interestingly, there is recent evidence indicating that the IRF(s) may also play a role in the transcriptional activation of viral promoters. The Qp promoter region of the EBV-encoded gene EBNA-1 contains an ISRE-like element that is responsive to the IRF-1 and IRF-2 as well as to IFN-a. Using a yeast one-hybrid screen technique, a new factor was recently isolated that binds specifically to the Qp ISRE.
The amino acid sequence of this protein is identical to the IRF-7 protein present in the Genbank database ((69); accession number U73036). By homology search of the HGF ETS cDNA library the Pitha group has also identified a novel IRF whose sequence is identical to that of IRF-7. At the amino acid level, IRF-7 shows highest homology to IRF-3. Several open reading frames (ORFs) of IRF-7 have been identified. Pagano's group found three shorter ORFs, listed in the database as IRF-7A, B and C
((69), accession nos. U53830, U53831 and U53832, respectively).
A new IRF-7 isoform, IRF-7H, was recently identified by Pitha's group ((70), accession number AF076494).
In vitro translated IRF-7 encodes a protein of 68 kDa (69, 72). Interestingly, while in vitro translated IRF-7 protein binds effectively to the Qp ISRE, it doesn't seem to affect transcription of Qp-driven reporter constructs in a transient transcription assay (72). In contrast to IRF-3, IRF-7 expression is not generally constitutive but can be effectively induced by IFN-a in fibroblast cells, B-cells and other cells of lymphoid origin (70, 71). Like IRF-3, in uninfected cells, IRF-3 is present mainly in the cytoplasm, virus infection induced phosphorylation of IRF-7, resulting in cytoplasmic to nuclear translocation of phosphorylated IRF-7 and activated gene transcription (70, 71). Recent studies indicate that virus-stimulated phosphorylation of IRF-3 results in the activation of IFNa4 and IFN~i gene transcription in murine cells. Once produced and secreted from the infected cell, IFNa4 and IFN~i subsequently feed back on cells through the IFN receptor, stimulate the Jak-STAT pathway and lead to the IFN-responsive activation of another member of the IRF
family - IRF-7; up-regulation of IRF-7 production then mediates the induction of non-IFNa4 gene expression (71).
SUI~iARY OF THE INVENTION
The present invention relates to IRF proteins that have been modified in the carboxy-terminus domain (transactivation domain)by modification of serine and/or threonine sites. Modification may be achieved by phosphorylation of serine and/or threonine, or by replacement of serine and/or threonine residues with residues having acidic side-chains, preferably carboxylic acid-containing side-chains, such as aspartic acid or glutamic acid residues. Such modified proteins may be mutants of IRF-3 and IRF-7, including chimeric proteins having portions of both IRF-3 and IRF-7, and post-translationally modified (phosphorylated) IRF-3 protein, the phosphorylation being induced by Sendai virus infection.
More specifically, the present invention provides a modified interferon regulatory factor (IRF) protein, the protein comprising at least one modified serine or threonine .:. ...: .: .. .:
_ s: .. : . . . : . . . a-~:
~so23-19 : : :. . . ... . . ::. : ..
~ : ~ ~ ~ ~ . . . . : r .
~ ~ ~ ~ , . . .
~ - .. ... .. .. . .. .:
phosphoacceptor site in the carboxy-terminus domain, preferably wherein cytokine gene activation by the modified IRF is increased relative to cytokine gene activation by a corresponding wild type IRF protein.
5 The present invention also provides nucleotide sequences which encode a protein of the invention as described above. Such nucleotide sequences may, for example, be used to modify a target cell of an organism.
The present invention also provides a pharmaceutical composition comprising an effective amount of the interferon regulatory factor (IRF) protein according to the invention, together with a pharmaceutically acceptable carrier, for the treatment of a viral infection, for example, an influenza infection, a herpes infection, a hepatitis infection or an HIv infection. ~ ' The present invention also provides a commercial package containing the IRF protein or pharmaceutical composition according to the invention, together with instructions for its use for the treatment of cancer or of a.
viral infection, for example, an influenza infection, a herpes infection, a hepatitis infection or an HIV infection.
The present invention further provides use of the interferon regulatory factor (IRF) protein according to the invention to activate a cytokine gene, preferably wherein the cytokine gene is an interferon gene or a chemokine gene.
DESCRIPTION OF THE FIGURES
Figure 1. Sendai virus infection induces IRF-3 degradation. IRF-3 expression plasmid CMVBL-IRF3 (lanes 1 and 2) or CMVHL vector alone (lanes 3 and 4), both at 5 ~g were transiently transfected into 293 cells by the calcium AMENDED SHEET

05-06-2000 . CA 02325354 2000-10-10 ",. ; , , " " CA 009900314 .. : .. . :. . ..
76023-19 ~ . i - ~~ . . ..... . ::... : .. s . : . . . . : . . .~ v . .
. . . . . . . . .' . . : .
. . : .. ... .. .. . .. .c . . - Sa .
phosphate method. At ~24h post. transfection, . cel.ls were infected with Sendai virus for 16h (lanes 2 and- 4). or left uninfected (lanes 1 and 3) . Whole cell extracts (20 .fig) were 'prepared and analyzed by immunoblotting with anti-IRF-3' antibody. .
- Figure 2. .Sendai virus induced phosphorylat~ion and degradation of IRF-3 protein. A) rtTA-IRF-3 cells, selected as described in Example,.were induced to express IRF-3 by doxycycline treatment for 24h. At 24h after Dox addition, cells were infected with Sendai virus for 4, 8, 12, 16, 20, or - ' 24h~~.(lanes 2-7) or were left uninfected (lane 1) . IRF-3 protein -was detected in whole cell -extracts ~ ( 10 fig) ~ by .
.- immunoblot.~ Two forms of IRF-3 were detected, designated as - form I and form II. B)-At 24h post ~Dox induction, rtTA-IRF-3 cells were infected with Sendai virus for 16 hours (lanes 4-8) or were left uninfected (lanes 1-3). Whole cell extracts from untreated ~ . .
AMENDED SHEET
cells (20 ~,g) or Sendai virus infected cells (60 ~.g) were incubated with 0.3 units of potato acidic phosphatase (PPA, lanes 2, 3, 7 and 8) or 5 units of calf intestinal alkaline phosphatase (CIP, lanes 4 and 5) in the absence (lanes 1, 2, 4, 6 and 7) or presence of phosphatase inhibitors (lanes 3, 5 and 8). Phosphorylated IRF-3 protein appears as a distinct band in immunoblots, migrating more slowly than IRF-3 forms I and II.
Figure 3. Analysis of IRF-3 deletion mutants in Sendai virus induced phosphorylation.
(A) Schematic representation of four IRF-3 deletions.
Thick solid lines and thin dashed lines indicate included and excluded sequences, respectively. The N-terminal IRF homology domain, the nuclear export signal (NES) and C-terminal IRF
association domain are indicated.
(B) Expression plasmids (5 ~,g each) encoding wild type and deletion mutants of IRF-3 (as indicated above the lanes) were transiently transfected into 293 cells; at 24h post transfection, cells were infected with Sendai virus for 16h (lanes 2, 4, 6, 8, and 10) or left uninfected (lanes 1, 3, 5, 7, and 9). Whola cell extracts (20 fig) were prepared from infected and control cells and analyzed by immunoblotting for IRF-3 forms I and II and for the presence of phosphorylated IRF-3 (P-IRF-3) with anti-IRF-3 antibody.
Figure 4. Analysis of IRF-3 point mutations in Sendai virus induced phosphorylation.
(A) Schematic representation of IRF-3 point mutations.
Thick solid lines and thin dashed lines indicate included and excluded sequences, respectively. The N-terminal IRF homology domain, the Nes element and C-terminal IRF association domain are indicated. Amino acids residues from 382 to 414 and from 141 to 147 are shown. The amino acids targeted for alanine or aspartic acid substitution are shown in large print. The point mutations are indicated below the sequence: (2A: 5396A/S398A;
3A: S402A/T404A/S405A; 5A: S396A/S398A/S402A/T404A/S405A); 5D
S396D/S398D/S402D/T404D/S405D; J2A: S385A/S386A; NES:
S145A/S146A).
(B) Expression plasmids (5 ~.g each) encoding wild type and point mutants of IRF-3 (as indicated above the lanes) were transiently transfected into 293 cells; at 24h post transfection, cells were infected with Sendai virus for 16h (lanes 2, 4, 6, 8, 10, 12, 14, 16 and 18) or left uninfected (lanes 1, 3, 5, 7, 9, 11, 13, 15 and 17). Whole cell extracts (20 ~.g) were prepared from infected and control cells and analyzed by immunoblotting for IRF-3 forms I and II and for the presence of phosphorylated IRF-3 {P-IRF-3) with anti-IRF-3 antibody.
Figure 5. Virus dependent cytoplasmic-nuclear translocation of IRF-3.
The subcellular localization of the GFP-IRF-3 (A and B), GFP-IRF-3(5A) (C and D), GFP-IRF-3(5D) (E and F) and GFP-IRF-3{NES) {G and H) was analyzed in uninfected (A, C, E, and G) and Sendai virus infected COS-7 cells at 16h after infection. GFP fluorescence was analyzed in living cells with a Leica fluorescence microscope using 40x objective.
Figure 6. Transactivation of PRDI/PRDIII and ISRE
containing promoters by IRF-3.
293 cells were transfected with IFN~i-CAT {A and B) or ISG15-CAT
(C) reporter plasmids and the various expression plasmids as indicated below the bar graph. CAT activity was analyzed at 48h post-transfection with 100 ~.g (IFN~i-CAT) or 10 ~g {ISG15-CAT) of total protein extract for 1-2h at 37°C.
Relative CAT activity was measured as fold activation (relative to the basal level of reporter gene in the presence of CMV-B1 vector alone after normalization with co-transfected ~i-Gal activity); the values represent the average of three experiments with variability shown in the error bar.
Figure 7. IRF-3 inducible expression of RANTES gene.
(A) Stimulation of RANTES gene transcription in virus-infected and IRF-3{5D)-expressing cells. The rtTA, IRF-3 and IRF-3(5D) cells were cultured in the presence or absence of Dox as indicated. After 30 hours, cells were either left untreated, infected with Sendai virus (80HAU/ml) for 16 hours, or treated with IFN-a//3 (100 IU/ml). The neutralizing antibody for type I IFN {Sigma) was added at the time of Dox addition.
Total RNA was isolated from each sample and analyzed by RPA
using the hCK5 kit (Pharmingen).
(B) Repression of virus-induced RANTES gene transcription by a dominant-negative form of IRF-3. The rtTA- and IRF-3(~N)-expressing cells were either left untrated or infected with Sendai virus (80 HAU/ml) for 16 hours. Total RNA
was isolated from each sample and analyzed by RPA.
(C) The kinetics of RANTES expression induced by IRF-3 (5D). Total RNA from IRF-3(5D)-expressing cells was isolated from each sample after Dox addition and analyzed by RPA.
(D) Cell culture supernatants were analyzed for the presence of RANTES protein by an ELISA performed as specified by the manufacturer (Biosource International).
Figure 8. Stabilization of IRF-3 by proteasome inhibitors.
IRF-3 t1N (09-133) (B) or IRF-3 ~N2A (C) expression plasmids were transiently transfected into 293 cells; at 24h post transfection, cells were infected with Sendai virus and treated for 12h with calpain inhibitor I (100 ~.M, lanes 2 and 5) or MG132 proteasome inhibitor (40 ~.M, lanes 3 and 6).
Ethanol, the solvent for calpain inhibitor I and MG132, was added to the cells as control (lanes 1 and 4). Endogenous (A) and transfected (B and C) IRF-3 proteins were detected in whole cell extracts (20 ~.g) by immunoblot.
Figure 9. IRF-3 interacts with CHP in virus infected cells.
(A) Schematic representation of CBP, illustrating the domains, involved in interaction with host or viral proteins (modified from (28)) and the myc-tagged CBP proteins (CBP1, CBP2, CBP3) used for immunoprecipitation.
(B) 293 cells were transfected with wild type and deletion mutants of iRF-3 expression plasmid (5 ug, as indicated above the lanes) or left untransfected (lanes 1 and 8). At 24h after transfection, cells were infected with Sendai virus for 16h (lanes 1, 3-8, and 10-13) or left uninfected (lanes 1 and 9).
Whole cell extracts (300 fig, except lane 1, which was 600 fig) were immunoprecipitated with anti-CBP antibody A22 (lanes 1-6) or with preimmune serum (lane 7). The immunoprecipitated complexes (lanes 1-7) or 30 ~g whole cell extracts (lanes 8-13) were run on 5~ SDS-PAGE and subsequently probed with anti-IRF-3 antibody.
(C) 293 cells were co-transfected with myc-tagged CBP
expression plasmids (as indicated above the lanes) and IRF-3 ON
(D9-133) expression plasmid. At 24h after transfection, cells were infected with Sendai virus (lanes 2, 4 and 6) or left uninfected (lanes 1, 3 and 5). Whole cell extracts (300 were immunoprecipitated with monoclonal anti-myc-tag antibody 9E10. The immunoprecipitated complexes were run on 5~ SDS-PAGE
and different forms of IRF-3 in the precipitates were analyzed by immunoblotting with anti-IRF-3 antibody.
(D) Whole cell extracts (30 fig) from (C) were also analyzed directly for the expression of myc-tagged CBP proteins by immunoblotting using anti-myc antibody 9E10.
Figure 10. The cDNA sequence encoding IRF-3(5D), together with the amino acid sequence of IRF-3(5D).
Figure 11. Transactivation study as described in Figure 6, using the IFN~i-CAT reporter plasmid to indicate the activity of various forms of IRF-3 and IRF-7 and binary mixtures thereof.
Figure 12. The cDNA sequence encoding IRF-7A(2D), together with the amino acid sequence of IRF-7A(2D).
Figure 13. The cDNA sequence encoding the IRF-7(1-246)/IRF-3(5D)(132-427) chimeric protein, together with the amino acid sequence of the IRF-7(1-246)/IRF-3(5D)(132-427) chimeric protein.
Figure 14. Transactivation study as described in Figure 6, using the IFN/3-CAT reporter plasmid to indicate the relative activity of various forms of IRF-3 and IRF-7, binary mixtures thereof and the chimeric protein IRF-7(1-246)/IRF-3(132-427) (IRF-7N-IRF-3(5D}C in Figure 14).
DETAILED DESCRIPTION OF THE INVENTION
As used herein, the term "nucleotide sequence" means a DNA or RNA molecule or sequence, and can include, for example, a cDNA, genomic DNA, or synthetic DNA sequence, a structural gene or a fragment thereof, or an mRNA sequence, that encodes an active or functional polypeptide.
Two DNA, RNA or polypeptide sequences are 5 "substantially homologous" or "structurally equivalent" when there is at least about 85~ (preferably at least about 90%, more preferably at least about 95~) identity between the nucleotides or amino acids over a defined length of the molecule. DNA sequences that are substantially homologous can 10 be identified in a Southern hybridization experiment under, for example, stringent conditions, as defined for that particular system. Appropriate hybridization conditions are within the knowledge of a person skilled in the art. See, for example, Maniatis et al., Molecular Cloning, A Laboratory Manual. Cold Spring Harbour Laboratory, New York (1982); Brown, T. A., Gene Cloning: An Introduction (2nd Ed.) Chapman & Hall, London (1990) .
The results disclosed herein show that phosphorylation represents an important post-translational modification of IRF-3 leading to cytoplasmic-to-nuclear translocation of phosphorylated IRF-3, stimulation of DNA
binding and transcriptional activity, association of IRF-3 with the transcriptional co-activator CBP/p300, and ultimately proteasome mediated degradation.
More specifically, the results disclosed herein show that, following Sendai virus infection, IRF-3 may be post-translationally modified by protein phosphorylation at multiple serine and threonine residues, located in the carboxy-terminus of IRF-3.
Furthermore, while modification of functionally relevant (phosphoacceptor) serine and threonine sites may be by phosphorylation, the modification may also be a mutation represented by replacement of at least one of these functionally relevant serine or threonine residues with an amino acid having a carboxylic acid in its side chain, preferably aspartic acid or glutamic acid, more preferably aspartic acid. The preferred mutant form of IRF-3 is that 05-06-2000 : . ... ., a : . r . . ;; CA 009900314 _ s-: .. : .: . . . ..
76023-19 s . ~~ . ~ a.~ . : ::.. : ~ a c ~ . ~ . . . . .
. . . . . . . . . : .
s .. .:. .. .. . .. .:
having aspartic acid residues in at least one of positions 396, 398, 402, 404 and 405 of the sequence, more preferably in positions 396, 398, 402, 404 and 405 of the sequence (IRF-3 (5D) ) (Figure 10) . The preferred mutant form of IRF-7 is that having asparatic acid residues in at least one of positions 477 and 479 of the sequence, more preferable in positions 477 and 479 of the sequence (IRF-7(2D)) (Figure 12). ,; -Also within the scope of the invention are chirneric proteins comprising a carboxy-terminus domain of one modified IRF protein, modified as discussed above, and an amino-terminal domain of another IRF protein. Preferably, the amino-terminus of IRF-7 is fused to the carboxy-terminus of modified IRF-3.
It is more preferred that the carboxy-terminus of modified IRF-3 is that of IRF-3(5D). Even more preferred is a chimeric protein comprising residues 1 to 246 of IRF-7 and residues 132 to 427 of IRF-3(5D) (Figure 13). ;
Also within the scope of the invention are proteins which are substantially homologous to the above proteins and which retain the function of those proteins. This includes ;~ -proteins based on human IRF-3 and IRF-7, as well as corresponding IRF-3 and IRF-7 proteins of other~species.
Nucleotide sequences within the scope of the invention are those which encode a protein of the invention.
Preferably, the nucleotide sequence is a coding DNA sequence as defined in Figure 10 or a DNA sequence which is hybridizable under stringent conditions with the complement of the coding DNA sequence of Figure 10, which DNA encodes IRF-3(5D). Also, preferably, the nucleotide sequence is a coding DNA sequence as defined in Figure 12 or a DNA sequence which.is hybridizable under stringent conditions with the complement of the coding DNA sequence in Figure 12, which DNA encodes IRF-7(2D). Also , -~, , _ AMENDED SHEET

CA 02325354 2000-io=io ' CA 009900314 05-06-2000 .Ei . : i i i : ~ if . i . . ~ ~ . i i Y Y .
76023-19 i : y . ..:i . . .~. : .. s . : . . . . : . : . . . .
~ . . . . . . . . ~
.. ... .. -.,. . .s .i lla , preferably, the nucleotide sequence is a coding DNA sequence as defined in Figure 13 or a DNA sequence which is ; , hybridizableunder stringent conditions with the complement of the coding DNA sequence of Figure 13, which DNA encodes IRF-7 (1-246) /IRF-3 (132-427) chimeric protein. ._ A combination of IRF-3 deletion and point mutations localized the inducible phosphorylation sites to the region -ISNSHPLSLTSDQ- between amino acids 395 and 407; point mutation AMENDED SHEET
of Ser-396 and Ser-398 residues eliminated virus-induced phosphorylation of IRF-3 protein, although residues Ser-402, Thr-404 and Ser-405 were also targets. Phosphorylation results in the cytoplasmic to nuclear translocation of IRF-3, DNA
S binding and increased transcriptional activation. Substitution of the Ser/Thr sites with the phosphomimetic Asp generated a constitutively active form of IRF-3 that functioned as a very strong activator of promoters containing PRDI/PRDIII or ISRE
regulatory elements. Use of phosphomimetic Glu for this purpose is also possible. Phosphorylation also appears to represent a signal for virus mediated degradation, since the virus induced turnover of IRF-3 was prevented by mutation of the IRF-3 Ser/Thr cluster or by proteasome inhibitors.
Interestingly, virus infection resulted in the association of IRF-3 with the CBP coactivator, as detected by co-immunoprecipitation with anti-CBP antibody, an interaction mediated by the C-terminal domains of both proteins. Mutation of the residues Ser-396 and Ser-398 in IRF-3 abrogated its binding to CBP. These results are discussed in terms of a model in which virus-inducible C-terminal phosphorylation of IRF-3 alters protein conformation to permit nuclear translocation, association with transcriptional partners and primary activation of IFN- and IFN-responsive genes.
Sendai virus dependent phosphorylation of IRF-3 was detected, occurring in a cluster of Ser and Thr sites in the carboxyl-terminal end of the protein. The residues implicated in this regulatory phosphorylation event are Ser-396/Ser-398/Ser-402/Thr-404/Ser-405, particularly the Ser-396/Ser-398 amino acids. 2) Phosphorylation of the IRF-3 in the Ser-Thr cluster resulted in the cytoplasmic to nuclear translocation of IRF-3; nuclear translocation was blocked by mutation of the phosphorylated amino acids. 3) Sendai virus infection induced the DNA binding and transactivation potential of IRF-3. Furthermore, IRF-3 containing the phosphomimetic Asp at the sites of C-terminal phosphorylation was an exceptionally strong transactivator of PRDI/PRDIII and ISRE containing promoters. 4) Phosphorylation was also required for the association of IRF-3 with the CBP co-activator protein. 5) Sendai virus infection resulted in IRF-3 degradation; again, phosphorylation was required as a signal for inducer mediated degradation since mutation of Ser/Thr cluster also blocked virus induced degradation.
Cytoplasmic to nuclear translocation of IRF-3 as a consequence of virus infection was inhibited by mutation of the Ser/Thr cluster, indicating an important regulatory role for C-terminal phosphorylation in the activation of IRF-3. Also strikingly, the conversion of the phosphorylation sites to the phosphomimetic Asp altered the subcellular localization of IRF-3 in uninfected cells. A proportion of IRF-3(5D) was localized to the nucleus of uninfected cells, suggesting that some IRF-3 may shuttle to and from the nucleus constitutively;
this observation is consistent with the identification of a nuclear export signal in IRF-3. Mutation of L144A/L145A in the NES element produced the most impressive alterations in subcellular localization. In uninfected cells, IRF-3 was partitioned in both the nucleus and cytoplasm; virus infection changed the nuclear pattern of staining from extra-nucleolar.
homogeneous staining as observed for wtIRF-3 to an intense nuclear speckling. At this stage, the nature of the subnuclear changes in IRF-3 localization are not explained, although it is possible that IRF-3(NES) translocates efficiently into the nucleus but becomes trapped in the nuclear pore complex during the export process.
One of the striking results of the mutagenesis of the C-terminal domain of IRF-3 was the generation of IRF-3(5D), an exceptionally strong activator of IFN-~i and ISG-15 gene expression. The phosphomimetic form of IRF-3 alone was able to stimulate IFN-~i expression as strongly as virus infection, a level of stimulation not previously observed in co-expression experiments (24,61). In previous experiments, it has been demonstrated that IRF-3 was able to bind the ISRE element of ISG-15, as well as the PRDIII/PRDI and IE regions of the IFNB
and IFNA promoters, respectively (2,56). Virus induction results in the appearance of two new protein-DNA complexes;
supershift experiments confirmed that both complexes contain IRF-3; it is not clear at this stage whether the upper complex also contains other proteins such as in the VIC (10,29) and DRAF (16) complexes or whether the lower complex represents a breakdown product of IRF-3. Strikingly, the same complexes appeared following co-transfection of IRF-3(5D) expression plasmid in the absence of virus induction, indicating that IRF-3(5D) represented a constitutive DNA binding form of IRF-3.
Thus, in uninfected cells, IRF-3(5D) localized in part to the nucleus (Fig. 5), interacted with DNA constitutively and was a strong activator of gene expression (Fig. 6).
The recent crystal structure of the related IRF-1 protein bound to PRDI provides evidence for a novel helix-turn-helix motif that latches onto a GAAA core sequence via three of the five conserved tryptophan amino acids of the DNA binding domain (20). By analogy with IRF-3, two GAAANN
sequences present in PRDIII of IFN-~i and another GAAANN element present in PRDI may serve as DNA contacts for multiple IRF-3(5D) proteins with strong activating potential.
Similarly, the ISRE element of the ISG-15 promoter also contains several GAAANN anchors for potential IRF binding.
Given the range of promoters that possess this hexameric sequence (48), it will be of interest to determine the capacity of IRF-3(5D) to stimulate expression of different cytokine and chemokine genes.
IRF-3 joins a growing list of cellular and viral proteins that functionally interact with CBP/p300 proteins, highly homologous proteins originally identified through their interactions with adenovirus ElA and CREB proteins (1,13). As a critical determinant of its global transcriptional coactivator activity, CBP/p300 possesses histone acetyltransferase activity (5,50). Acetylation of histones is involved in the destabilization and remodelling of nucleosomes, a crucial step in permitting the accessibility of transcriptional factors to DNA templates. Several studies have now demonstrated that CBP/p300 participates in the transcriptional process by providing a scaffold for different classes of transcriptional regulators on specific chromatin domains (12,50}. A growing body of biochemical and genetic evidence also implicates CBP/p300 as a negative regulator of cell growth, based on its interactions with adenovirus Ela, 5 SV40 large T antigen and the tumour suppressor p53, among others. With regard to p53-CBP/p300 complex formation, functional interaction between these two important growth regulatory proteins accounts for several of the known activities of p53 (3,28,40); interestingly, CBP/p300 was shown 10 recently to acetylate p53 and stimulate its transactivation potential (27) .
It will be of interest to determine whether IRF-3 is similarly modified by CBP association. The functional consequences of IRF-3 interaction with CBP/p300 remain to be 15 elucidated, although recent studies demonstrated that CBP/p300 also functionally interacts with STAT 1 (68) and STAT 2 (7) and may contribute to IFNa and IFN~y nuclear signalling. Recently published studies have demonstrated that synergistic activation of the IFNR promoter requires recruitment of CBP/p300 to the enhanceosome, via a new activating surface assembled from the activation domains of all the transcription factors in the enhanceosome (37,45). Alterations in any of the activation domains decreased both CBP recruitment and transcriptional synergy. By analogy, recruitment of CBP/p300 to DNA bound IRF-3 is likely required for maximal transcriptional activation. Association requires the interaction of the C-terminal domain of IRF-3 and the C-terminal interaction domain of CBP, a region previously shown to associate with the p53 tumour suppressor, whereas STAT1 and STAT2 associate with different regions of CBP (7, 68} .
Virus induced phosphorylation of IRF-3 also represents a signal for proteasome mediated degradation of IRF-3, since mutation of the Ser-396/Ser-398 or the use of proteasome inhibitors prevented the post infection degradation of IRF-3. Virus induced degradation of IRF-3 is reminiscent of the virus-induced turnover of another member of the IRF family - IRF-2. In response to dsRNA or viral induction, the 50 kD
IRF-2 protein is proteolytically processed into a smaller, 24-27 kDa protein (51) comprising the 160 as DBD of IRF-2, termed TH3 (14) or In4 (65). Although TH3 has been shown to bind DNA and repress transcription more efficiently than the full length IRF-2 protein (42), its physiological role is not clear. Since the induction kinetics of TH3 are slower than that of IFN-~i in response to dsRNA or viral infection (14), it has been suggested that the IRF-2 cleavage product may be a post-induction repressor of IFN-~3 gene expression (65).
Virus induced phosphorylation of IRF-3 at the C-terminal Ser/Thr residues and its subsequent degradation by a proteasome dependent pathway are also similar to the well studied phosphorylation and degradation of IrcBa which leads to activation of NF-xB binding activity (reviewed in 4,6). In unstimulated cells, NF-KB heterodimers are retained in the cytoplasm by inhibitory IrcB proteins. Upon stimulation by many activating agents, including cytokines, viruses and dSRNA, IKBa is rapidly phosphorylated and degraded, resulting in the release and nuclear translocation of NF-xB. The amino-terminus of IrcBa represents a signal response domain for activation of NF-~cB and substitution of alanine for either Ser-32 or Ser-36 completely abolished the signal-induced phosphorylation and degradation of IrcBa, and blocked the activation of NF-KH.
These mutations also blocked in vitro ubiquitination of the IKBa protein. The amino-terminus of IKBcx is necessary for signal-induced phosphorylation and ubiquitination, but for degradation to occur, there is an absolute requirement for the C-terminal PEST domain (reviewed in 4,6).
Similarities and differences exist between the observed degradation of IRF-3 and the mechanism of IKBa degradation. The C-terminal phosphorylation of IRF-3 as a consequence of virus infection is required for its subsequent degradation based on the deletion and point mutation analysis of the region -ISNSHPLSLTSDQ- between amino acids 395 and 407.
Minimally, phosphorylation of Ser-396 and Ser-398 are required for subsequent degradation, although Ser-402, Ser-404 and Ser-405 may represent secondary phosphorylation sites.
Likewise, in the case of IKBa, phosphorylation and Ser-32 and Ser-36 are required for inducer mediated degradation.
Furthermore, the protease inhibitor calpain inhibitor I and the more specific proteasome inhibitor MG132 block IRF-3 turnover.
S A major difference in the mechanisms of IKBa and IRF-3 turnover lies in the nature of the inducing stimuli.
Multiple inducers - cytokines such as TNF and IL-1, viruses, LPS, oxidative stress, etc (6) - all lead to the induction of IkBa phosphorylation and degradation whereas IRF-3 phosphorylation appears to be induced only by virus infection and dsRNA addition; other inducers have not resulted in IRF-3 turnover.
A significant temporal difference also exists between IrcBa phosphorylation/turnover and IRF-3 phosphorylation/degradation. Many activators of NF-rcB
stimulate IrcBa phosphorylation within minutes and TNF induced degradation occurs within the first 15-30 minute after treatment. In the case of IRF-3, phosphorylation is not detected until 6-8 hours after infection and continues in a heterogenous manner over the next 10-12 hours. Previous experiments have, however, demonstrated that Sendai virus-induced turnover of IKBa also occurs slowly over several hours (24) .
Based on the data presented herein and by analogy with the properties of other IRF family members (48), the following model is proposed to explain several observations.
IRF-3 exists in a latent state in the cytoplasm of uninfected cells; the C-terminus may physically interact with the DNA
binding domain in such a way as to obscure both the DBD and the IAD regions of the protein; the presence of an autoinhibitory domain within the C-terminal 20aa (407-427) would explain the activating effect of this deletion, as seen previously with IRF-4 (11,19). Virus induced phosphorylation at the Ser/Thr at 396-405aa cluster leads to a conformational change in IRF-3, exposing both the DBD and IAD and relieving C-terminal autoinhibition. Translocation to the nucleus, occurring via an unidentified nuclear localization sequence or in conjunction with a transcriptional partner associating through the IAD
region, leads to DNA binding at ISRE- and PRDI/PRDIII-containing promoters. Phosphorylation is also necessary for IRF-3 association with the chromatin remodelling activity of CBP/p300. The presence of a NES element ultimately shuttles IRF-3 from the nucleus and terminates the initial activation of IFN responsive promoters. The phosphorylated form of IRF-3 exported from the nucleus may now be susceptible to proteasome mediated degradation. This scenario shares several features with the protein synthesis independent activation of NF-KB, and further suggests that IRF-3 may represent a component of virus- or dsRNA-inducible complexes such as DRAF (16) or VIC (10,29) that could play a primary role in the induction of IFN- or IFN responsive genes.
In view of the above-mentioned properties, and in particular its ability to stimulate an immune response, IRF
protein is useful as a tumour suppresser.
The invention is described in more detail in the following examples.
Example 1: Plasmid constructions and Mutacrenesis.
The IRF-3 expression plasmid was prepared by cloning the EcoRI-XhoI fragment containing the IRF-3 cDNA from the pSKIRF-3 plasmid downstream of the CMV promoter of CMVBL
vector. CMVt-IRF-3 was constructed by cloning of IRF-3 cDNA
downstream of the doxycycline-responsive promoter CMVt at the BamHI site of the nee CMVt BL vector (49). cDNAs encoding IRF-3 carboxyl terminal deletion mutations were generated by 28 cycles of PCR amplification with Vent DNA polymerase. DNA
oligonucleotide primers were synthesized using an Applied Biosystems DNA/RNA synthesizer. The amino-terminal primer was synthesized with an EcoRI restriction enzyme site and the carboxyl-terminal primers were synthesized with XbaI
restriction enzyme sites at their ends. The PCR products were purified by phenol/chloroform extraction and ethanol precipitation, digested with EcoRI and XbaI, and inserted into EcoRI/XbaI sites of CMVBL vector.
The point mutations of IRF-3 were generated by overlap PCR mutagenesis using Vent DNA polymerase. Mutations were confirmed by sequencing.
The N-terminal deletion mutations (~N, ~N2A, AN3A and S ~NSA) of IRF-3 were generated by digestion of the related IRF-3/CMVBL plasmid with BamHI (filled in with Klenow enzyme), partial digestion with ScaI, and re-ligation. GFP-IRF-3 expression plasmids were generated by cloning of cDNAs encoding wild type or mutated forms of IRF-3 into the downstream of EGFP
in the pEGFP-C1 vector (Clonetech). For construction of plasmids encoding myc-tagged CBP truncated proteins, the cDNAs coding for CBP were generated from the pRC-RSV/mCBP plasmid (provided by Dr. Dimitris Thanos) by PCR amplification. The cDNA fragments were cloned in the downstream of myc-tag in 5' myc-PCDNA3 vector {provided by Dr. Stephane Richard).
For the construction of pFlag-IRF-7, the IRF-7 cDNA
was created by PCR and the resulting product was cloned into pFlag CMV-2 vector. To generate the IRF-7(aal-246)-IRF-3(5D) (aa132-427) chimera, the cDNA encoding IRF-3 (5D) {aa132-427) was cut out from IRF-3 (5D)/CMVBL plasmid with ScaI and NotI
{blunted with Klenow enzyme) and was cloned into pFlag-IRF-7 (digested with SmaI, which removed the C-terminal region of IRF-7 from 247-503) in frame with the IRF-7 N-terminal amino acid sequence (1-246). The point mutations of IRF-7 (D477-D479) were generated by overlap PCR mutagenesis essentially as described above for IRF-3 using Vent DNA polymerase. Codon AGC
encoding residues Ser 477 and Ser 479 were mutated to GAC
{Asp). Mutations were confirmed by sequencing.
Example 2: Generation of IRF-3 cell lines.
Plasmid CMVt-rtTA (49) was introduced into 293 cells by a calcium phosphate-based method. Cells were selected beginning at 48h after transfection for about one week in aMEM
media (GIBCO-BRL) containing 10~ heat-inactivated calf serum, glutamine, antibiotics and 2.5 ng/~.1 puromycin (Sigma).
Resistant cells carrying the CMVt-rtTA plasmid (rtTA-293 cells) were then transfected with the CMVt-IRF-3 plasmid. Cells were selected beginning at 48h for a period of approximately 2 weeks in aMEM containing 10~ heat-inactivated calf serum, glutamine, antibiotics, 2.5 ng/~1 puromycin and 400 ~g/ml 6418 (Life Technologies, Inc.).
Example 3: Cell culture and transfections.
S All transfections for CAT assay were carried out in human embryonic kidney 293 cells or NIH3T3 cells grown in aMEM
(293) or Dulbecco's MEM (NIH3T3) media (GIBCO-BRL) supplemented with 10~ calf serum, glutamine and antibiotics. Subconfluent cells were transfected with 5 ~g of CsCl purified 10 chloramphenicol acetyltransferase (CAT) reporter and expression plasmids by calcium phosphate coprecipitation method (293 cells) or lipofectamine (NIH3T3 cells). The reporter plasmids were the SVo,Q CAT and ISG15 CAT reporter genes (56); also the transfection procedures were previously described (41,56). For 15 individual transfections, 100 ~,g (SVo~i CAT) or 10 ~,g (ISG15 CAT) of total protein extract was assayed for 1-2h at 37°C. The CAT activity was normalized with ~i-Gal assay. All transfections were performed 3-6 times.
Example 4: Western blot analysis of IRF-3 modification and 20 degradation.
To characterize the posttranslational regulation of IRF-3 protein, stable or transiently transfected IRF-3 expressing cells were infected with Sendai Virus (80 HAU/ml) or treated with 5 ng/ml TNF-a, either with or without addition of 50 ~g/ml cycloheximide. In some experiments, cells were treated with either 100 ~,M calpain inhibitor I (ICN), 40 ~.M
MG132 proteasome inhibitor, or an equivalent volume of their respective solvent (ethanol) as control. Cells were washed with phosphate-buffered saline and lysed in 10 mm Tris-C1 pH
8.0, 200 mm NaCl, 1 mm EDTA, 1 mm dithiothreitol (DTT), 0.5~
Nonidet P-40 {NP-40), 0.5 mm phenylmethysulfonyl fluoride (PMSF), 5 ~,g/ml leupeptin, 5 ~g/ml pepstatin, and 5 ~.g/mI
aprotinin. Equivalent amounts of whole cell extract (20 ~.g) were subject to SDS-polyacrylamide gel electrophoresis (SDS-PAGE) in a lOg polyacrylamide gel. After electrophoresis, the proteins were transferred to Hybond transfer membrane (Amersham) in a buffer containing 30 mm This, 200 mm glycine and 20% methanol for lh. The membrane was blocked by incubation in phosphate-buffered saline (PBS) containing 5%
dried milk for lh and then probed with IRF-3 antibody in 5%
milk/PHS, at a dilution of 1:3000. These incubations were done at 4°C overnight or at RT for 1-3h. After four 10 minute washes with PHS, membranes were reacted with a peroxidase-conjugated secondary goat anti-rabbit antibody (Amersham) at a dilution of 1:2500. The reaction was then visualized with the enhanced chemiluminescence detection system (ECL) as recommended by the manufacturer (Amersham Corp.).
Example 5: Phosphatase treatment.
Twenty to sixty ~g of whole cell extract were treated with 0.3 units of potato acidic phosphatase (Sigma) in a final volume of 30 ~,1 PIPES buffer (10 mm PIPES pH 6.0, 0.5 mm PMSF, 5 ~,g/ml aprotinin, 1 ~g/ml leupeptin, and 1 ~g/ml pepstatin) or 5 units of calf intestine alkaline phosphatase (Pharmacia) in 30 ~,l CIP buffer. The phosphatase inhibitor mix contained 10 mm NaF, 1.5 mm Na2Mo04, 1 mm (3-glycerophosphate, 0.4 mm Na3V04 and 0.1 ~.g/ml okadaic acid.
Example 6: Subcellular localization of GFP-IRF-3 t~roteins.
To analyse the subcellular localization of wild type and mutated forms of IRF-3 proteins in uninfected and virus infected cells, the GFP-IRF-3 expression plasmids (5 fig) were transiently transfected into COS-7 cells by the calcium phosphate coprecipitation method. For virus infection, transfected cells were infected with Sendai virus (80 hemagglutinating units per mL for 2h) at 24h post transfection.
GFP fluorescence was analyzed in living cells with a Leica fluorescence microscope using a 40x objective.
Example 7: Electromobilitv Shift Assay.
Nuclear extracts were prepared from 293 cells at different times after infection with Sendai virus (80HAU/ml).
In some experiments, extracts were prepared from cells transfected with different IRF-3 expression plasmids, as indicated in individual experiments. Cells were washed in Buffer A [10 mM HEPES, pH 7.9; 1.5 mm MgCl2; 10 mM KC1; 0.5 mM
dithiothreitol (DTT); and 0.5 mM phenylmethylsulfonyl fluoride WO 99/5173? PCT/CA99/00314 (PMSF)3 and were resuspended in Buffer A containing 0.1% NP-40.
Cells were then chilled on ice for 10 minutes before centrifugation at 10,000 g. Pellets were then resuspended in Buffer B (20mM HEPES, pH 7.9; 25% glycerol; 0.42 M NaCl; 1.5 mM
MgClz; 0.2 mM EDTA; 0.5 mM DTT; 0.5 mM PMSF; 5 ~.g/ml leupeptin;
5 ~,g/ml pepstatin; 0.5 mM spermidine; 0.15 mM spermine; and 5 ~g/ml aprotinin). Samples were incubated on ice for 15 minutes before being centrifuged at 10,000 g. Nuclear extract supernatants were diluted with Buffer C (20 mM HEPES, pH 7.9;
20% glycerol; 0.2 mM EDTA; 50 mM KC1; 0.5 mM DTT; and 0.5 mM
PMSF). Nuclear extracts were subjected to EMSA by using a 32P-labelled probe corresponding to the PRDIII region of the IFN-~i promoter (5'-GGAAAACTGAAAGGG-3') or the ISRE region of the ISG-15 promoter (5'-GATCGGGAAAGGGAAACCGAAACTGAAGCC-3').
The resulting protein-DNA complexes were resolved by 5%
polyacrylamide gel and exposed to X-ray film. To demonstrate the specificity of protein-DNA complex formation, 125-fold molar excess of unlabelled oligonucleotide was added to the nuclear extract before adding labelled probe.
Example 8: Immunoprecipitation and Western analysis of CBP
associated proteins.
Whole cell extract (300 fig) were prepared from either transfected or untransfected cells and precleared with 5 ~1 of preimmune rabbit serum and 20 ~cl of protein A-Sepharose beads (Pharmacia) for 1 hour at 4°C. The extract was incubated with 10 ~,1 of anti-CBP antibody A-22 (Santa Cruz) or 2 ~,l anti-myc antibody 9E10 (21) and 30 ~1 of protein A-Sepharose beads for 2-3 hours at 4°C. Precipitates were washed 5 times with lysis buffer, eluted by boiling the beads 3 minutes in lx SDS sample buffer. Eluted proteins were separated by SDS PAGE, transferred to Hybond transfer membrane. Membranes were incubated with anti-IRF-3 (1:3000) or anti-myc antibody 9E10 (1:1000). Immunocomplexes were detected by using a chemiluminescence-based system.
The results of the above examples are summarized below.
Virus induced phosphorvlation of IRF-3 protein.
IRF-3 is expressed constitutively in various cells and its expression is not enhanced by viral infection or by IFN
treatment. To investigate whether the IRF-3 protein is regulated by post-translational modification after virus infection, 293 cells were transiently transfected with an IRF-3 expression plasmid and subsequently infected with Sendai virus 24h later. In cells transfected with CMVBL vector alone, endogenous IRF-3 protein was easily detected using a polyclonal IRF-3 antibody and in cells transfected with the IRF-3 expression plasmid, IRF-3 protein levels were significantly increased (Fig.i, lanes 1 and 3). Interestingly, Sendai virus infection resulted in two alterations in the expression of IRF-3: 1) an overall decrease in the amount of IRF-3 in transfected and control cells (Fig. 1, lanes 2 and 4) and the generation of a more slowly migrating form of IRF-3 (Fig. 1, compare lanes 1 and 2). In all experiments, the turnover of IRF-3 after virus infection was more pronounced with the endogenous protein than with the transfected proteins (see Fig. l, as well as others). Because the transfected proteins were driven by the CMV promoter, ongoing synthesis of transfected IRF-3 may partially obscure the turnover of IRF-3.
The kinetics of virus-induced modification of IRF-3 were characterized in a 293 cell line that expressed IRF-3 inducibly under the control of the tetracycline responsive promoter CMVt (25,26). Infection of this cell line (designated rtTA-IRF-3) with Sendai virus resulted in a decrease in the amount of IRF-3 between 12 and 24h after infection (Fig. 2A).
Two forms of IRF-3 protein (designated I and II) were detected in uninfected cells (Fig. 2A, lane 1) and following virus infection, a third slowly migrating form of IRF-3 was also detected (Fig.2A, lanes 4-7). To determine whether the slowest form of IRF-3 was due to virus-induced phosphorylation (P-IRF-3), the different forms of IRF-3 were subjected to treatment in vitro with potato acidic phosphatase {PPA) or calf intestine alkaline phosphatase (CIP) and/or phosphatase inhibitors (Fig. 2B). These treatments did not affect the mobilities of forms I and II in uninfected cells (Fig. 2B, lanes 1-3). However, in rtTA-IRF-3 expressing 293 cells infected with Sendai virus for 12h, an additional slowly migrating, presumably phosphorylated form of IRF-3 was also detected (Fig. 2B, lane 6); this form of IRF-3 completely disappeared following CIP or PPA treatment (Fig.2B, lanes 6 and 7) but was maintained in the presence of CIP/PPA when phosphatase inhibitors were also added to the reaction (Fig.
2B, lanes 5 and 8).
Mapping the IRF-3 phosphorylation sites.
A series of deletions of IRF-3 were generated to identify the virus-induced phosphorylation sites) of IRF-3 (Fig. 3A). 293 cells were transiently transfected with IRF-3 deletion mutants and the virus mediated phosphorylation was measured by immunoblotting (Fig. 3B). The results indicated that a virus-induced phosphorylation of IRF-3 occurs at the C-terminal end of IRF-3 since the mutations that contained only the N-terminal part of IRF-3 protein (133, 240, 328, 357 or 394aa) were not phosphorylated (Fig. 3B). Full length and 407aa forms of IRF-3 were phosphorylated as a consequence of virus infection (Fig. 3B, lanes 1-4). C-terminal truncation of IRF-3 to a protein of 394 or 357aa removed the sites) of inducible phosphorylation (Fig. 3B, lanes 5-8), although the shortened versions of forms I and II were still observed. Also in the IRF-3 D9-133 mutation (AN) which had the DNA binding, N-terminal amino acids (aa9 to aa133) removed, both virus induced phosphorylation of IRF-3 and the differential migration of the shortened forms I and II were easily detected (Fig. 3B, lanes 9 and 10). Degradation of the endogenous forms of IRF-3 by virus infection was also detected in this experiment (compare Fig. 3B, lanes 7 and 9 with lanes 8 and 10).
Thus, by deletion analysis, a phosphorylation domain of IRF-3 protein was localized to the region -ISNSHPLSLTSDQ-between amino acids 395 and 407. Point mutations in the several putative Ser and Thr phosphorylation residues within this region were generated in the full length protein and the D9-133 (ON} protein (Fig. 4A). In the IRF-3 cDNA encoding these proteins, the Ser-396/Ser398/Ser-402/Thr-404/Ser-405 residues were replaced by alanine (5A), as were the three residues Ser-402/Thr-404/Ser-405 (3A) and the two residues Ser-396/Ser-398 (2A). Transfection of these plasmids into 293 5 cells and subsequent virus infection revealed that full length wild type IRF-3 was phosphorylated (Fig. 4B, lanes 4 and 8), whereas the IRF-3 proteins containing 2A and 5A mutations were no longer phosphorylated in virus infected cells (Fig. 4B, lanes 6 and 10). Interestingly, IRF-3-3A was also very weakly 10 phosphorylated as a consequence of virus infection, thus implicating Ser-402/Thr-404/Ser-405 as potential secondary sites of phosphorylation. Using the ON IRF-3 protein and the relevant point mutations, phosphorylation was detected with ~N
(Fig. 4B, lane 12) but not with ~N-2A and ~N-5A (Fig. 4B, 15 lanes 14 and 18); likewise, 0N-3A displayed very weak phosphorylation (Fig. 4B, lane 16).
These experiments thus implicate Ser-396 and Ser-398 as critical sites of virus-induced phosphorylation of IRF-3;
however, Ser-402/Thr-404/Ser-405 residues also contribute to 20 the observed phosphorylation, since the migration of phosphorylated ~N-3A is significantly faster than ON and the phosphorylation level is decreased (Fig. 4B, lanes 12 and 16).
Another study suggested the involvement of the Ser residues at aa385 and 386 as potential phosphoacceptor sites (67).
25 However, in studies with the S385A/S386A mutation, no evidence was found for inducible phosphorylation at these sites.
Nevertheless, since these sites represent consensus sites for CKI and CKII, constitutive phosphorylation is a possibility.
IRF-3 phosphorylation induces cytoplasmic to nuclear translocation of IRF-3.
Initial studies indicated that IRF-3 was localized in the cytoplasm of uninfected cells (67); to investigate the role of phosphorylation on IRF-3 localization, wild type and point mutated forms of IRF-3 were linked to green fluorescent protein (GFP), transfected into COS-7 cells and examined for Sendai virus induced changes in subcellular localization (Fig. 5). In uninfected cells, GFP-IRF-3 localized exclusively to the cytoplasm; Sendai virus infection resulted in translocation of IRF-3 to the nucleus within 8h in 90-95% of the cells (Fig. 5A
and B). Mutation of the Ser/Thr cluster in GFP-IRF-3(5A) completely abrogated virus-induced cytoplasmic to nuclear translocation (Fig. 5, C and D). Interestingly, the substitution of the Ser/Thr cluster with the phosphomimetic Asp in GFP-IRF-3(5D) likewise altered subcellular localization.
IRF-3(5D) localized both to the nucleus and cytoplasm in uninfected cells (Fig. 5E), while virus infection resulted in an intense nuclear pattern of IRF-3(5D) fluorescence (Fig. 5F).
Point mutation of a putative nuclear export signal in IRF-3, the L145A/L146A modification - termed IRF-3(NES) - also changed subcellular localization of IRF-3. In uninfected cells, GFP-IRF-3(NES) was localized to the nucleus and cytoplasm, with a homogeneous, extra-nucleolar pattern of nuclear staining.
After virus infection, GFP-IRF-3(NES) localized to the nucleus with an intense speckled pattern of nuclear fluorescence in greater than 95% of the cells, suggesting that IRF-3(NES) may be trapped in the nucleus associated with the nuclear pore complex.
Transactivation of PRDI/PRDIII and ISRE promoters by IRF-3.
Next, the capacity of IRF-3 to regulate gene expression was analysed by transient transfection in human 293 and murine NIH3T3 cells using the IFN~i and ISG-15 promoters in reporter gene assays. Expression of NF-rcB RelA(p65), IRF-1 and IRF-3 alone minimally induced IFN~3 promoter activity between 3 to 4 fold (Fig. 6A and B), as shown previously (24,56,61).
Introduction of the C-terminal point mutants - IRF-3(2A), IRF-3(3A) IRF-3(5A) - reduced the low transactivation capacity of IRF-3 to control levels (Fig. 6A). Interestingly, deletion of the C-terminal 20aa of IRF-3 to IRF-3(407) stimulated IFNQ
activity about 6 fold, indicative of the removal of an inhibitory domain in IRF-3. However, further deletion to 394, 357 or 240 abrogated transactivation potential (Fig. 6A).
Mutation of the NES element was not sufficient to stimulate IFN~i activity. Strikingly, the substitution of the Ser/Thr cluster at aa397-405 in IRF-3 with the phosphomimetic Asp generated a very strong, constitutive transactivator protein that alone stimulated the IFN~i promoter 90 fold.
As shown previously, high level induction of the IFN~i promoter requires synergistic activation by NF-xB and IRF
proteins (24,61). To analyse the properties of IRF-3 in synergistic activation of the IFN/3 promoter, co-expression studies were performed using RelA(p65) expression plasmid and different wild type and mutant forms of IRF-3 (Fig. 6B).
Co-expression of RelA and IRF-1 or RelA and IRF-3 stimulated IFN~i-CAT activity by 20-25 fold. IRF-3 (407) and RelA(p65) stimulated IFN~i activity about 40 fold, supporting the idea of the removal of an inhibitory domain in IRF-3, whereas both the IRF-3(394) and the IRF-3(NES) failed to synergise with RelA in the activation of the IFN~i promoter. RelA and IRF-3{NES) produced a relatively weak 8 fold induction of IFN~i expression, indicating that nuclear localization is not sufficient for IRF-3 activation. The combination of RelA and IRF-3(5D) produced an 80 fold stimulation of IFN~i promoter activity (Fig.
6B); together with the above data, IRF-3(5D) alone appears to be capable of full stimulation of the IFN~i promoter and further synergy with RelA is not observed (compare Fig. 6A and B).
Surprisingly, IRF-3(5A) and RelA produced a 30 fold stimulation, suggesting that 5A can still synergise with RelA, despite mutation of the Ser/Thr cluster.
The transactivation potential of IRF-3 was also analysed using the ISG-15 promoter, an ISRE containing regulatory element (Fig. 6C). As shown previously (2), and above for the IFN~i promoter, IRF-3 alone weakly activated the ISG-15 promoter; in the context of this regulatory element, IRF-3 was weaker than IRF-1, which produced a 9 fold stimulation. Again deletion of the C-terminal 20aa of IRF-3 generated a protein that stimulated gene expression; with the ISG-15 promoter, a 12 fold induction was observed; IRF-3(394) and IRF-3(357) did not stimulate gene expression but rather slightly repressed ISG-15. Again remarkably, IRF-3(5D) produced a 50 fold induction of the ISG-15 promoter (Fig. 6C), thus demonstrating that substitution of the Ser/Thr sites with WO 99/51737 PCT/CA99/003i4 the phosphomimetic Asp generated a constitutively active form of IRF-3 that functioned as a very strong activator of promoters containing PRDI/PRDIII or ISRE regulatory elements.
Activation of RANTES Transcription by IRF-3 and Virus Chemokine expression is demonstrated in Figure 7, the chemokine being RANTES (Regulated on Activation Normal T-cell Expressed and Secreted) protein. IRF-3-inducible cells were used to determine whether other cytokine-chemokine genes may be regulated by IRF-3; an (Rnase Protection Analysis (RPA) with multiple human cytokine-chemokine probes (Pharmingen) was used to examine RNA derived from rtTA-IRF-3 or rtTA-IRF-3(5D) cells.
Strikingly, the RANTES gene was highly expressed in the IRF-3(5D)-inducible cells, as well as in virus-infected cells (Fig.
7A, lanes 3, 5, and 7) but not in uninfected rtTA- or wt IRF-3-expressing cells (Fig. 7A, lanes 1 and 4). Since IRF-3(5D) was a strong transactivator of the IFN-~i promoter in transient transfection assays, the possibility of an autoregulatory effect of IFN-a/,~ expression on transcription of RANTES
promoter via JAK-STAT activation was considered. Activation of RANTES did not occur secondary to the production of IFN-a/~i, since RANTES mRNA was not detected in control rtTA-expressing cells treated directly with IFN-a/(3 (Fig. 7A, lane 2);
furthermore, addition of neuralizing antibody directed against type I IFN did not block the stimulation of RANTES gene expression by IRF-3(5D) (Fig. 7A, lane 8). Other experiments also demonstraed that IRF-3 itself was not activated by IFN
treatment (13a). Inducible expression of RANTES in cells stably expressing a dominant-negative form of IRF-3 which lacks the N-terminal amino acids 9 to 133 and does not bind to DNA
was also examined. As shown in Fig. 7B, RANTES gene transcription was indcued by Sendai virus in control (rtTA) cells (Fig. 7H) but not in IRF-3 (oN)-expressing cells (Fig.
7B). This experiment indicates that a non-DNA binding, dominant-negative mutant of IRF-3 is able to block completely virus-induced activation of RANTES transcription.
The kinetics of IRF-3 transgene induction and RANTES
mRNA expression were characterized at various times following Dox induction. IRF-3(5D) was detected at 8 to 12 hours with peak levels at 24 hours following Dox addition. RANTES mRNA
was first detectable at 18 hours after Dox induction with peak levels at 40 hours (Fig. 7C,.lanes 5 to 10). Induction of RANTES protein expression as detected by ELISA (Fig. 7D) was first observed at 12 hours after Dox induction of IRF-3, in good agreement with the mRNA levels, and accumulated thereafter with a dramatic increase between 24 and 32 hours after stimulation, also in agreement with mRNA levels. The possibility that IRF-3(5D) may be directly activating another transcription factor such as NF-xB, which in turn would stimulte RANTES transciption, was also considered. No evidence for IRF-3(5D)-mediated activation of NF-rcB DNA binding activigy was observed. Similarly, IRF-3(5D) expression did not activate the human immunodeficiency virus (HIV)-long terminal repeat, a complex promoter controlled by NF-xB and other transcription factors (data not shown) .
Inhibition of IRF-3 degradation.
Another consequence of virus infection is the degradation of the IRF-3. Since phosphorylation of proteins is functionally associated with the process of protein degradation via the ubiquitin-dependent proteasome pathway (53,57,60), the effect of proteasome inhibitors on virus-induced turnover of IRF-3 was examined. In cells transfected with the ON and ONSA
forms of IRF-3, virus induced degradation of full length (endogenous) forms of IRF-3 (Fig. 8A, lanes 1 and 4) and the truncated ~N (Fig. 8B, lanes 1 and 4) was detected. Addition of the protease inhibitor calpain inhibitor I or the proteasome inhibitor MG132 blocked virus-induced IRF-3 degradation (Fig.
8A and 8B, lanes 4-6). Particularly with the ~N protein, the accumulation of the phosphorylated form of ~N was also detected in virus infected cells (Fig. 8B, lanes 5 and 6), suggesting that phosphorylation of IRF-3 may represent a signal for subsequent degradation by the proteasome pathway. To confirm this idea, the 5A point mutated form of IRF-3 was analysed; the IRF-3-~NSA protein was resistant to virus induced degradation (Fig. 8C, lanes 1 and 4); no further stabilization of IRF-3-ANSA occurred with calpain inhibitor I or MG132 addition and no phosphorylated IRF-3 was detected (Fig. 8C, lanes 4-6).
These experiments demonstrate that virus dependent phosphorylation of the C-terminal of IRF-3 represents a signal 5 for subsequent proteasome mediated degradation.
Interaction between IRF-3 and CBP in virus infected cells.
To examine the possibility that IRF-3 associated with the co-activator CBP/p300 (Fig. 9A) following Sendai virus infection, CBP was immunoprecipitated from virus-infected cells 10 with anti-CBP antibody; an immunoblot for IRF-3 revealed that IRF-3 was co-precipitated from virus-infected cells but not from uninfected cells (Fig. 9B, lanes 2 and 3). This interaction was observed clearly in cells co-transfected with the IRF-3 expression plasmid (Fig. 9B, lane 3 ) but was not 15 seen when the immunoprecipitation was performed with pre-immune serum (Fig. 9B, lane 7). The endogenous IRF-3 also co-precipitated from virus-infected cells (Fig. 9B, lane 1).
However, mutation of the Ser/Thr residues identified as the virus inducible phosphorylation sites abrogated the association 20 of IRF-3 with CBP. In particular, IRF-3(2A) and IRF-3(5A) were detected in whole cell extract immunoblot but not in the CBP
immunoprecipitate (Fig. 9B, compare lanes 4 and 6 with lanes 11 and 13). With the IRF-3(3A) mutant, interaction with CHP was still observed (Fig. 9B, lane 5). The high background in all 25 lanes represents secondary antibody reactivity with rabbit IgG
from the immunoprecipitation. Immunoblot analysis of the whole cell extracts revealed that phosphorylated IRF-3, as well as forms I and II were present in virus infected cells (Fig. 9B, lane 10) and in cells transfected with 2A, 3A and 5A the forms 30 I and II were observed but not the phosphorylated form of IRF-3 (Fig. 9B, lanes 11-13).
CBP has several domains that bind transcription factors, designated CBPl, CBP2, and CBP3 respectively (Fig. 9A, reviewed in (28)). To determine which domain of CBP interacts with IRF-3, the three specific subdomains were myc-tagged at the 5' end by subcloning into the pCDNA3 vector (Fig. 9A). 293 cells were co-transfected with these myc-tagged CBP expression plasmids together with the IRF-3 ON (D9-133) expression plasmid. At 24h after transfection, cells were infected with Sendai virus, co-immunopreciptated with anti-myc antibody 16h later (21) and then immunoblotted for IRF-3. Endogenous IRF-3 and transfected IRF-3 ~N proteins co-precipitated with CBP-3 from virus-infected cells but not from uninfected cells (Fig.
9C, lane 6). In cells co-transfected with CBP-1 and CBP-2, no endogenous or transfected ~N IRF-3 was detected (Fig. 9C, lanes 1-4). Immunoblot analysis of the whole cell extracts revealed that all three myc-tagged CBP proteins were efficiently expressed in uninfected and virus infected cells (Fig. 9D).
These results demonstrate that IRF-3 binds to the C-terminal domain of CBP in virus infected cells and interaction with CBP
requires Ser-396/Ser-398 phosphorylation of IRF-3 but not at Ser-402/Thr-404/Ser-405.
Figure 11 shows the relative activity of various forms of IRF-3 and IRF-7, and binary mixtures thereof, in transactivation studies. Both the IRF-3(5D) and IRF-7(2D) mutants show increased activity relative to their corresponding wild-type proteins. There is a synegistic effect present when both proteins are present, and this effect is most pronounced in a mixture of the IRF-3(5D) and IRF-7(2D) (D477/479) mutants.
Figure 14 shows that the chimeric protein IRF-7{1-246)/IRF-3(5D)(132-427) has a markedly increased activity over the mixture of the IRF-3(5D) and IRF-7(2D) (D477/479) mutants.
A pharmaceutical composition may be prepared, with a protein of the invention as active ingredient, for the treatment of a viral infection, such as an influenza infection, a herpes infection or an HIV infection.
The pharmaceutical compositions of the present invention may be formulated in a conventional manner using one or more pharmaceutically acceptable carriers. Thus, the active compounds of the invention may be formulated for oral, buccal, transdermal (e. g., patch), intranasal, parenteral (e. g., intravenous, intramuscular or subcutaneous) or rectal administration or in a form suitable for administration by inhalation or insufflation.
For oral administration, the pharmaceutical compositions may take the form of, for example, tablets or S capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e. g.
pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e. g., lactose, microcrystalline cellulose or calcium phosphate); lubricants (e. g., magnesium stearate, talc or silica); disintegrants (e. g., potato starch or sodium starch glycollate); or wetting agents (e.g., sodium lauryl sulphate). The tablets may be coated by methods well known in the art. Liquid preparations for oral administration may take the form of, for example, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e. g., sorbitol syrup, methyl cellulose or hydrogenated edible fats); emulsifying agents (e. g., lecithin or acacia); non-aqueous vehicles (e. g., almond oil, oily esters or ethyl alcohol); and preservatives (e. g., methyl or propyl p-hydroxybenzoates or sorbic acid).
For buccal administration the composition may take the form of tablets or lozenges formulated in conventional manner.
The active compounds of the invention may be formulated for parenteral administration by injection, including using conventional catherization techniques or infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulating agents such as suspending, stabilizing and/or dispersing agents.
Alternatively, the active ingredient may be in powder form for reconstitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
The active compounds of the invention may also be formulated in rectal compositions such as suppositories or S retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
For intranasal administration or administration by inhalation, the active compounds of the invention are conveniently delivered in the form of a solution or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. The pressurized container or nebulizer may contain a solution or suspension of the active compound. Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated containing a powder mix of a compound of the invention and a suitable powder base such as lactose or starch.
The protein of the invention can also be made available using gene therapy. The DNA encoding the protein can be introduced to cells of an organism at a target site, for example, by a viral vector, by electroporation, by co-transfection with a selectable marker, or by DNA vaccine.
REFERENCES
1. Arany, Z., Sellers, W.R., Livingston, D.M. and Eckner, R.
1994. ElA-associated p300 and CREB-associated CBP belong to a conserved family of coactivators. Cell 77:799-800.
2. Au, W.-C., Moore, P.A., Lowther, W., Juang, Y.-T, and Piths, P.M. 1995. Identification of a member of the interferon regulatory factor family that binds to the interferon-stimulated response element and activates expression of interferon-induced genes. Proc.Natl.Acad.Sci.USA
92:11657-11661.
3. Avantaggiati, M.L., Ogryzko, V., Gardner, K., Giordano, A., Levine, A.S. and Kelly, K. 1997. Recruitment of p300/CBP in p53-dependent signal pathways. Cell 89:1175-1184.
4. Baldwin, A.S.Jr. 1996. The NF-rcB and IrcB proteins: new discoveries and insights. Annu.Rev.Immunol. 14:649-681.
5. Bannister, A.J. and Kouzarides, T. 1996. The CBP
coactivator is a histone acetyltransferase. Nature 384:641-643.
6. Beauparlant, P. and Hiscott, J. 1996. Biological and biochemical inhibitors of the NF-KB/Rel proteins and cytokine synthesis. CytGrowthFactRev 7:175-190.
7. Bhattacharya, S., Eckner, R., Grossman, S., Oldread, E., Arany, Z., D'Andrea, A. and Livingston, D.M. 1996.
Cooperation of Stat2 and p300/CBP by interferon-a. Nature 383:344-347.
8. Hluyssen, H.A.R., Durbin, J.E. and Levy, D.E. 1996.
ISGF3~y p48, a specificity switch for interferon activated transcription factors. CytGrowthFactRev 7:11-17.
9. Bovolenta, C., Driggers, P.H., Marks, M.S., Medin, J.A., Politis, A.D., Vogel, S.N., Levy, D.E., Sakaguchi, K., Appella, E., Coligan, J.E. and Ozato, K. 1994. Molecular interactions between interferon consensus sequence binding protein and members of the interferon regulatory factor family.
Proc.Natl.Acad.Sci.USA 91:5046-5050.
10. Bragan~a, J., G~nin, P., Bandu, M.-T., Darracq, N., Vignal, M., Casse, C., Doly, J. and Civas, A. 1997. Synergism between multiple virus-induced-factor-binding elements involved in the differential expression of IFN-A genes. J.Biol.Chem. 272:
22154-22162.
11. Brass, A.L., Kehrli, E., Eisenbeis, C.F., Storb, U. and Singh, H. 1996. Pip, a lymphoid-restricted IRF, contains a regulatory domain that is important for autoinhibition and ternary complex formation with the Ets factor PU.l. Genes Dev.
10:2335-2347.

12. Chen, H., Lin, R.J., Schiltz, R.L., Chakravarti, D., Nash, A., Nagy, L., Privalsky, M.L., Nakatani, Y. and Evans, R.M.
1997. Nuclear receptor coactivator ACTR is a noval histone acetyltransferases and forms a multimeric activation complex 5 with P/CAF and CBP/p300. Cell 90:569-580.
13. Chrivia, J.C., Kwok, R.P.S., Lamb, N., Hagiwara, M., Montminy, M.R. and Goodman, R.H. 1993. Phosphorylated CREB
binds specifically to the nuclear protein CBP. Nature 365:855-859.
10 14. Cohen, L. and Hiscott, J. 1992. Characterization of TH3, an induction specific protein interacting with the interferon-~i promoter. Virol. 191:589-599.
15. Crepieux, P., Coll, J. and Stehelin, D. 1994. The Ets family of proteins: weak modulators of gene expression in 15 quest for transcriptional partners. CritRevOncogen 5:615-638.
16. Daly, C. and Reich, N.C. 1993. Double-stranded RNA
activates novel factors that bind to the interferon stimulated response element. Mol.Cell.Biol. 13:3756-3764.
17. Darnell Jr., J.E., Kerr, I.M. and Stark, G.R. 1994.
20 Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 264:1415-1421.
18. Driggers, P.H., Ennist, D.L., Gleason, S.L., Mak, W.-H., Marks, M.S., Levi, B.-Z., Flanagan, J.R., Appella, E. and 25 Ozato, K. 1990. An interferon y-regulated protein that binds the interferon-inducible enhancer element of major histocompatibility complex class I genes.
Proc.Natl.Acad.Sci.USA 87:3743-3747.
19. Eisenbeis, C.F., Singh, H. and Storb, U. 1995. Pip, a 30 novel IRF family member, is a lymphoid-specific, PU.1-dependent transcriptional activator. Genes Dev. 9:1377-1387.
20. Escalante, C.R., Yie, J., Thanos, D. and Aggarwal, A.K.
1998. Structure of IRF-1 with bound DNA reveals determinants of interferon regulation. Nature 391:103-106.
35 21. Evan, G.I. and Bishop, J.M. 1985. Isolation of monoclonal antibodies specific for the human c-myc proto-oncogene product.
Mol.Cell.Biol. 4:2843-2850.
22. Fujita, T., Kimura, Y., Miyamoto, M., Barsoumian, E.L. and Taniguchi, T. 1989. Induction of endogenous IFN-a and IFN-~i genes by a regulatory transcription factor IRF-1. Nature 337:270-272.
23. Fujita, T., Sakakibara, J., Sudo, Y., Miyamoto, M., Kimura, Y. and Taniguchi, T. 1988. Evidence for a nuclear factor(s), IRF-1, mediating induction and silencing properties to human IFN-~i gene regulatory elements. EMBO J. 7:3397-3405.
24. Garoufalis, E., Kwan, I., Lin, R., Mustafa, A., Pepin, N., Roulston, A., Lacoste, J. and Hiscott, J. 1994. Viral induction of the human interferon ~3 promoter: modulation of transcription by NF-KB/rel proteins and interferon regulatory factors. J.Virol. 68:4707-4715.
25. Gossen, M. and Bujard, H. 1992. Tight control of gene expression in mammalian cells by tetracycline-responsive promoters. Proc.Natl.Acad.Sci.USA 89:5547-5551.
26. Gossen, M., Freundlieb, S., Bender, G., Muller, G., Hillen, W. and Bujard, H. 1995. Transcriptional activation by tetracyclines in mammalian cells. Science 268:1766-1769.
27. Gu, W. and Roeder, R.G. 1997. Activation of p53 sequence-specific DNA binding by acetylation of p53 C-terminal domain. Cell 90:595-606.
28. Gu, W., Shi, X.L. and Roeder, R.G. 1997. Synergistic activation of transcription by CBP and p53. Nature 387:819-823.
29. G~nin, P., Bragan~a, J., Darracq, N., Doly, J. and Civas, A. 1995. A novel PRDI and TG binding activity involved in virus-induced transcription of IFN-A genes. NuclAcidRes 23:5055-5063.
30. Harada, H., Fujita, T., Miyamoto, M., Kimura, Y., Maruyama, M., Furia, A., Miyata, T. and Taniguchi, T. 1989.
Structurally similar but functionally distinct factors, IRF-1 and IRF-2, bind to the same regulatory elements of IFN and IFN-inducible genes. Cell 58:729-739.
31. Harada, H., Matsumoto, M., Sato, M., Kashiwazaki, Y., Kimura, T., Kitagawa, M., Yokochi, T., Tan, R.S.-P., Takasugi, T., Kadokawa, Y., Schindler, C., Schreiber, R.D., Noguchi, S.
and Taniguchi, T. 1996. Regulation of IFN-a/(3 genes: evidence for a dual function of the transcription factor complex ISGF3 in the production and action of IFN-a/Q. GenestoCells 1:995-1005.
S 32. Harada, H., Willison, K., Sakakibara, J., Miyamoto, M., Fujita, T. and Taniguchi, T. 1990. Absence of type I IFN
system in EC cells: transcriptional activator (IRF-1) and repressor (IRF-2) genes are developmentally regulated. Cell 63:903-913.
33. Hiscott, J., Nguyen, H. and Lin, R. 1995. Molecular mechanisms of interferon ~3 gene induction. SeminVirol 6:161-173.
34. Holtschke, T., Lohler, J., Kanno, Y., Fehr, T., Giese, N., Rosenbauer, F., Lou, J., Knobeloch, K.-P., Gabriele, L., blaring, J.F., Bachmann, M.F., Zingernagel, R.M., Morse III, H.C., Ozato, K. and Horak, I. 1996. Immunodeficiency and chronic myelogenous leukemia-like syndrome in mice with a targeted mutation of the ICSBP gene. Cell 87:307-317.
35. Ihle, J.N. 1996. STATs: signal transducers and activators of transcription. Cell 84:331-334.
36. Kawakami, T., Matsumoto, M., Sato, M., Harada, H., Taniguchi, T. and Kitigawa, M. 1995. Possible involvement of the transcription factor ISGF3~y in virus-induced expression of the IFN-~i gene. FEBS Lett. 358:225-229.
37. Kim, T.K. and Maniatis, T. 1998. The mechanism of transcriptional synergy of an in vitro assembled interferon ,Q
enhanceosome. Mol.Cel1 1:119-129.
38. Kimura, T., Kadokawa, Y., Harada, H., Matsumoto, M., Sato, M., Kashiwazaki, Y., Tarutani, M., Tan, R.S-P., Takasugi, T., Matsuyama, T., Mak, T.M., Noguchi, S. and Taniguchi, T. 1996.
Essential and non-redundant roles of p48 (ISGF3~y) and IRF-1 in both type I and type II interferon responses, as revealed by gene targeting studies. GenestoCells 1:115-124.
39. Levy, D.E. 1995. Interferon induction of gene expression through the Jak-Stat pathway. SeminVirol 6:181-190.
40. Lill, N.L., Grossman, S.R., Ginsberg, D., DeCaprio, J. and Livingston, D.M. 1997. Binding and modulation of p53 by p300/CBP coactivators. Nature 387:823-827.
41. Lin, R., Beauparlant, P., Makris, C., Meloche, S. and Hiscott, J. 1996. Phosphorylation of IrcBa in the C-terminal PEST domain by casein kinase II affects intrinsic protein stability. Mol.Cell.Biol. 16:1401-1409.
42. Lin, R., Mustafa, A., Nguyen, H. and Hiscott, J. 1994.
Mutational analysis of interferon (IFN) regulatory factors 1 and 2: Effects on the induction of IFN-~i gene expression.
J.Biol.Chem. 269:17542-17549.
43. Matsuyama, T., Grossman, A., Mittrfcker, H.-W., Siderovski, D.P., Kiefer, F., Kawakami, T., Richardson, C.D., Taniguchi, T., Yoshinaga, S.K. and Mak, T.W. 1995. Molecular cloning of LSIRF, a lymphoid-specific member of the interferon regulatory factor family that binds the interferon-stimulated response element (ISRE). NuclAcidRes 23:2127-2136.
44. Matsuyama, T., Kimura, T., Kitagawa, M., Watanabe, N., Kundig, T., Amakawa, R., Kishihara, K., Wakeham, A., Potter, J., Furlonger, C., Narendran, A., Suzuki, H., Ohashi, P., Paige, C., Taniguchi, T. and Mak, T. 1993. Targeted disruption of IRF-1 or IRF-2 results in abnormal type I IFN
induction and aberrant lymphocyte development. Cell 75:83-97.
45. Merika, M., Williams, A., Chen, G., Collins, T. and Thanos, D. 1998. Recruitment of CBP/p300 by the IFN~i enhanceosome is required for synergistic activation of transcription. Mol.Cell 1:277-287.
46. Mittrucker, H.-W., Matsuyama, T., Grossman, A., Kundig, T.M., Potter, J., Shahinian, A., Wakeham, A., Patterson, B., Ohashi, P.S. and Mak, T.W. 1997. Requirement for the transcription factor LSIRF/IRF4 for mature B and T lymphocyte function. Science 275:540-543.
47. Miyamoto, M., Fujita, T., Kimura, Y., Maruyama, M., Harada, H., Sudo, Y., Miyata, T. and Taniguchi, T. 1988. Regulated expression of a gene encoding a nuclear factor, IRF-1, that specifically binds to the IFN-Q gene regulatory elements. Cell 54:903-913.
48. Nguyen, H., Hiscott, J. and Pitha, P.M. 1997. The growing family of IRF transcription factors. CytGrowthFactRev 8: in press.
49. Nguyen, H., Lin, R. and Hiscott, J. 1997. Activation of multiple growth regulatory genes following inducible expression of IRF-1 or IRF/RelA fusion proteins. Oncogene 15:1425-1435.
50. Ogryzko, V.V., Schiltz, R.L., Russanova, V., Howard, B.H.
and Nakatani, Y. 1996. The transcriptional coactivators p300 and CBP are histone acetyltransferases. Cell 87:953-959.
51. Palombella, V. and Maniatis, T. 1992. Inducible processing of interferon regulatory factor-2. Mol.Cell.Biol.
12:3325-3336.
52. Pitha, P.M. and Au, W.-C. 1995. Induction of interferon a gene expression. SeminVirol 6:151-159.
53. Read, M.A., Neish, A.S., Luscinskas, F.W., Palombella, V.J., Maniatis, T. and Collins, T. 1995. The proteasome pathway is required for cytokine-induced endothelial-leukocyte adhesion molecule expression. Immunity 2:493-506.
54. Reis, L.F.L., Harada, H., Wolchok, J.D., Taniguchi, T. and Vilcek, J. 1992. Critical role of a common transcription factor, IRF-1, in the regulation of IFN-~i and IFN-inducible genes. EMBO J. 11:185-193.
55. Russo, J.J., Bohenzky, R.A., Chien, M.-C., Chen, J., Yan, M., Maddalena, D., Parry, J.P., Peruzzi, D., Edelman, I.S., Chang, Y. and Moore, P. 1996. Nucleotide sequence of the kaposi sarcoma-associated hexpesvirus (HHV8).
Proc.Natl.Acad.Sci.USA 93:14862-14867.
56. Schafer, S., Lin, R., Moore, P., Hiscott, J. and Pitha, P.M. 1998. Regulation of type 1 interferon gene expression by interferon regulatory factor 3. J.Biol.Chem. 273:2714-2720.
57. Scherer, D.C., Brockman, J.A., Chen, Z., Maniatis, T. and Ballard, D.W. 1995. Signal-induced degradation of IKBa requires site-specific ubiquitination. Proc.Natl.Acad.Sci.USA
92:11259-11263.
58. Schindler, C. and Darnell Jr., J.E. 1995. Transcriptional responses to polypeptide ligands: the JAK-STAT pathway.
Ann. Rev.Biochem. 64:621-651.
59. Sharf, R., Meraro, D., Azriel, A., Thornton, A.M., Ozato, K., Petricoin, E.F., Larner, A.C., Schaper, F., Hauser, H. and Levi, B.-Z. 1997. Phosphorylation events modulate the ability of interferon consensus sequence binding protein to interact 5 with interferon regulatory factors and to bind DNA.
J.Biol.Chem. 272:9785-9792.
60. Thanos, D. and Maniatis, T. 1995. NF-KB: a lesson in family values. Cell 80:529-532.
61. Thanos, D. and Maniatis, T. 1995. Identification of the 10 rel family members required for virus induction of the human ~i interferon gene. Mo1Ce11Bio1 15:152-164.
62. Veals, S.A., Schindler, C., Leonard, D., Fu, X.-Y., Aebersold, R., Darnell Jr., J.E. and Levy, D.E. 1992. Subunit of an a-interferon-responsive transcription factor is related 15 to interferon regulatory factor and myb families of DNA-binding proteins. Mo1Ce11Bio1 12:3315-3324.
63. Vilcek, J. and Sen, G. Interferons and other cytokines.
In: Virology, edited by Fields, B., Knipe, D.M. and Howley, P.M. Philadelphia: Lippincott-Raven, 1996, p. 375-399.
20 64. Weisz, A., Marx, P., Sharf, R., Appella, E., Driggers, P.H., Ozato, K. and Levi, B.-Z. 1992. Human interferon consensus sequence binding protein is a negative regulator of enhancer elements common to interferon-inducible genes.
J.Biol.Chem. 267:25589-25596.
2S 65. Whiteside, S.T., King, P. and Goodbourn, S. 1994. A
truncated form of the IRF-2 transcription factor has the properties of a postinduction repressor of interferon-(3 gene expression. J.Biol.Chem. 269:27059-27065.
66. Yamagata, T., Nishida, J., Tanaka, T., Sakai, R., Mitani, 30 K., Yoshida, M., Taniguchi, T., Yazaki, Y. and Hirai, H. 1996.
A novel interferon regulatory factor family transcription factor, ICSAT/Pip/LSIRF, that negatively regulates the activity of interferon-regulated genes. Mo1Ce11Bio1 16:1283-1294.
67. Yoneyama, M., Suhara, W., Fukuhara, Y. and Fujita, T.
35 1997. Direct activation of a factor complex composed of IRF-3 and CBP/p300 by virus infection. J.Interferon Cytokine Res.
17:553.

WO 99/51737 PCT/CA99/bb314 68. Zhang, J.J., Vinkemeier, U., Gu, W., Chakravarti, D., Horvath, C.M. and Darnell, J.E. 1996. Two contact regions between STAT1 and CBP/p300 in interferon 'y signalling.
Proc.Natl.Acad.Sci.USA 93:15092-15096.
69. Zhang, L. and Pagano, J.S. 1997. IRF-7, a new interferon regulatory factor associated with Epstein Barr Virus latency.
Mol.Cell.Biol. 17:5748-5757.
70. Au, W.C., Moore, P.A., LaFleur, D.W., Tombal, B. and Pitha, P.M. (1998). Characterization of the interferon regulatory factor-7 and its potential role in the transcription activation of interferon A gene. J. Biol. Chem. 273, 29210-29217 .
71. Marie, I., Durbin, J.B. and Levy, D.E. (1998).
Differential viral induction of distinct interferon-a genes by positive feedback through interferon regulatory factor-7. E1~0 J. 17, 6660-6669.
72. Nonkwello C, Ruf IK, Sample J. 1997. Interferon-independent and -induced regulation of Epstein-Barr Virus EBNA-1 gene transcription in Burkitt lymphoma. J. Virol. 71, 6887-6897.

SEQUENCE LISTING
<110> THE SIR MORTIMER B. DAVIS-JEWISH GENERAL HOSPITAL
<120> HIGHLY ACTIVE FORMS OF INTERFERON REGULATORY FACTOR
PROTEINS
<130> IRF-3 <140>
<141>
<150> CA 2,234,588 <151> 1998-04-07 <160> 11 <170> PatentIn Ver. 2.0 <210> 1 <211> 1284 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (1)..(1281) <400> 1 atg gga acc cca aag cca cgg atc ctg ccc tgg ctg gtg tcg cag ctg 48 Met Gly Thr Pro Lys Pro Arg Ile Leu Pro Trp Leu Val Ser Gln Leu gac ctg ggg caa ctg gag ggc gtg gcc tgg gtg aac aag agc cgc acg 96 Asp Leu Gly Gln Leu Glu Gly Val Ala Trp Val Asn Lys Ser Arg Thr cgc ttc cgc atc cct tgg aag cac ggc cta cgg cag gat gca cag cag 144 Arg Phe Arg Ile Pro Trp Lys His Gly Leu Arg Gln Asp Ala Gln Gln gag gat ttc gga atc ttc cag gcc tgg gcc gag gcc act ggt gca tat 192 Glu Asp Phe Gly Ile Phe Gln Ala Trp Ala Glu Ala Thr Gly Ala Tyr gtt ccc ggg agg gat aag cca gac ctg cca acc tgg aag agg aat ttc 240 Val Pro Gly Arg Asp Lys Pro Asp Leu Pro Thr Trp Lys Arg Asn Phe 05-06-2000 CA 02325354 2000-io-io.i i~ .; ;, CA 009900314 i "~'' . : . . r .
i : ? . i v.~i . . v . i . f i i _... . i . . ~. . i . G
. i . . ~ ' . . . . . . i :
2/13 ~.. ._.. .. .. . .:- .:
cgc tct gcc ctc aac cgc aaa gaa ggg ttg cgt tta gca gag gac cgg 288 Arg Ser Ala Leu Asn Arg Lys Glu Gly Leu Arg Leu Ala Glu Asp Arg agc aag gac cct cac gac cca cat aaa atc tac gag ttt gtg aac tca 336 Ser Lys Asp Pro His Asp Pro His Lys Ile Tyr Glu Phe Val Asn Ser 100 ~ 105 110 gga gtt ggg gac ttt tcc cag cca gac acc tct ccg gac acc aat ggt 384 1 0 Gly Val Gly Asp Phe Ser Gln Pro Asp Thr Ser Pro Asp Thr Asn Gly gga ggc agt act tct gat acc cag gaa gac att ctg gat gag tta ctg 932 Gly Gly Ser Thr Ser Asp Thr Gln Glu Asp Ile Leu Asp Glu Leu Leu ' ggt aac atg gtg ttg gcc cca ctc cca gat ccg gga ccc cca agc ctg 480 Gly Asn Met Val Leu Ala Pro Leu Pro Asp Pro Gly Pro Pro 5er Leu 14s 150 ls5 160 get gta gcc cct gag ccc tgc cct cag ccc ctg cgg agc ccc agc ttg 528 Ala Val Ala Pro Glu Pro Cys Pro Gln Pro Leu Arg Ser Pro Ser Leu gac aat ccc act ccc ttc cca aac ctg ggg ccc tct gag aac cca ctg 576 Asp Asn Pro Thr Pro Phe Pro Asn Leu Gly Pro Ser Glu Asn Pro Leu aag cgg ctg ttg gtg ccg ggg gaa gag tgg gag ttc gag gtg aca gcc 624 3 0 Lys Arg Leu Leu Val Pro Gly Glu Glu Trp Glu Phe Glu Val Thr Ala ttc tac cgg ggc cgc caa gtc ttc cag cag acc atc tcc tgc ccg gag 672 Phe Tyr Arg Gly Arg Gln Val Phe Gln Gln Thr Ile Ser Cys Pro Glu ggc ctg cgg ctg gtg ggg tcc gaa gtg gga gac agg acg ctg cct gga 720 Gly Leu Arg Leu Val Gly 5er Glu Val Gly Asp Arg Thr Leu Pro Gly tgg cca gtc aca ctg cca gac cct ggc atg tcc ctg aca gac agg gga 768 Trp Pro Val Thr Leu Pro Asp Pro Gly Met Ser Leu Thr Asp Arg Gly gtg atg agc tac gtg agg cat gtg ctg agc tgc ctg ggt ggg gga ctg 816 Val Met Ser Tyr Val Arg His Val Leu Ser Cys Leu Gly Gly Gly Leu get ctc tgg cgg gcc ggg cag tgg ctc tgg gcc cag cgg ctg ggg cac 864 5 0 Ala Leu Trp Arg Ala Gly Gln Trp Leu Trp Ala Gln Arg Leu Gly His tgc cac aca tac tgg gca gtg agc gag gag ctg ctc ccc aac agc ggg 912 Cys His Thz Tyr Trp Ala Val Ser Glu Glu Leu Leu Pro Asn Ser Gly cat ggg cct gat ggc gag gtc ccc aag gac aag gaa gga ggc gtg ttt 960 His Gly Pro Asp Gly Glu Val Pro Lys Asp Lys G1u Gly Gly Val Phe gac ctg ggg ccc ttc att gta gat ctg att acc ttc acg gaa gga agc 1008 Asp Leu Gly Pro Phe Ile Val Asp Leu Ile Thr Phe Thr Glu Gly Ser gga cgc tca cca cgc tat gcc ctc tgg ttc tgt gtg ggg gag tca tgg 1056 Gly Arg Ser Pro Arg Tyr Ala Leu Trp Phe Cys Val Gly Glu Ser Trp -..
AMENDED SHEET

.: ~. i 1 . i i i i s , , i-1 CA 009900314 i.~ .. a . 1 . : : . r a y : -i i ~~ii ~ i~~v 1 1~ i i 1 ~ ~~~ . i . i~ v 1 i i i 1 i ~ 1 ~ 1 . . i 1 1 ~ i ~ 313 ~. ... .. .. . ..
ccc cag gac cag ccg tgg acc aag agg ctc gtg atg gtc aag gtt gtg 1104 Pro Gln Asp Gln Pro Trp Thr Lys Arg Leu Val Met Val Lys Val Val ccc acg tgc ctc agg gcc ttg gta gaa atg gcc cgg gta ggg ggt gcc 1152 Pro Thr Cys Leu Arg Ala Leu Val Glu Met Ala Arg Val Gly Gly Ala tcc tcc ctg gag aat act gtg gac ctg cac att gac aac gac cac cca 1200 1 0 Ser Ser Leu Glu Asn Thr Val Asp Leu His Ile Asp Asn Asp His Pro ctc gac ctc gac gac gac cag tac aag gcc tac ctg cag gac ttg gtg 1298 Leu Asp Leu Asp Asp Asp Gln Tyr Lys Ala Tyr Leu Gln Asp Leu Val gag ggc atg gat ttc cag ggc cct ggg gag agc tga 1284 Glu Gly Met Asp Phe Gln Gly Pro Gly Glu Ser <210> 2 <211> 427 <212> PRT
<213> Homo sapiens <400> 2 Met Gly Thr Pro Lys Pro Arg Ile Leu Pro Trp Leu Val Ser Gln Leu Asp Leu Gly Gln Leu Glu Gly Val Ala Trp Val Asn Lys Ser Arg Thr zo z5 30 Arg Phe Arg Ile Pro Trp Lys His Gly Leu Arg Gln Asp Ala Gln Gln Glu Asp Phe Gly Ile Phe Gln Ala Trp Ala Glu Ala Thr Gly Ala Tyr Val Pro Gly Arg Asp Lys Pro Asp Leu Pro Thr Trp Lys Arg Asn Phe Arg SerAlaLeuAsnArgLysGluGlyLeuArgLeuAlaGluAspArg Ser LysAspProHisAspProHisLysIleTyrGluPheValAsnSer Gly ValGlyAspPheSerGlnProAspThrSerProAspThrAsnGly 5 Gly GlySerThrSerAspThrGlnGluAspIleLeuAspGluLeuLeu Gly AsnMetValLeuAlaProLeuProAspProGlyProProSerLeu Ala ValAlaProGluProCysProGlnProLeuArgSerProSerLeu Asp AsnProThrProPheProAsnLeuGlyProSerGluAsnProLeu AMENDED SHEET

0~-06-2000 ;. ; ~,. ",.; " :, , r CA 009900314 , is .. . . ,-. . ~ ..: : . r ~
~ f . i 1 . ..i . i a:v i . I i t . i i 1 . . . . - .. . . . i . i . . . . . i . . ~ i i x/13 ~~ ... .. .. . ..
Lys Arg Leu Leu Val Pro Gly Glu Glu Trp Glu Phe Glu Val Thr Ala Phe ArgGlyArgGlnValPheGlnGlnThrIleSerCysProGlu Tyr 210 21s 220 Gly ArgLeuValGlySerGluValGlyAspArgThrLeuProGly Leu 1 Trp ValThrLeuProAspProGlyMetSerLeuThrAspArgGly 0 Pro Val SerTyrValArgHisValLeuSerCysLeuGlyGlyGlyLeu Met Ala TrpArgAlaGlyGlnTrpLeuTrpAlaGlnArgLeuGlyHis Leu Cys ThrTyrTrpAlaValSerGluGluLeuLeuProAsnSerGly His His ProAspGlyGluValProLysAspLysGluGlyGlyValPhe Gly Asp GlyProPheIleValAspLeuIleThrPheThrGluGlySer Leu Gly SerProArgTyrAlaLeuTrpPheCysValGlyGlu5erTrp Arg Pro AspGlnProTrpThrLysArgLeuValMetValLysValVal Gln Pro CysLeuArgAlaLeuValGluMetAlaArgValGlyGlyAla Thr Ser LeuGluAsnThrValAspLeuHisIleAspAsnAspHisPro 5er 4 Leu LeuAspAspAspGlnTyrLysAlaTyrLeuGlnAspLeuVal 0 Asp Glu MetAspPheGlnGlyProGlyGluSer Gly <210>

<211>

<212>
PRT

<213>
Homo sapiens 50 <900>

Ile AsnSerHisProLeuSerLeuThr5erAspGln 5er <210>

<211>

<212>
PRT

<213> 444 Homo Sapiens <400>

-.~

AMENDED SHEET

Gly Ala Ala Ala <210> 5 <211> 6 <212> PRT
<213> Homo sapiens <400> 5 Gly Ala Ala Ala Asn Asn <210>6 <211>15 <2I2>DNA

<213>Homo sapiens <400> 6 ggaaaactga aaggg 15 <210> 7 <211> 30 <212> DNA
<213> Homo sapiens <400> 7 gatcgggaaa gggaaaccga aactgaagcc 30 <210> 8 <211> 1512 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (1)..(1509) <400> 8 atg gcc ttg get cct gag agg gca gcc cca cgc gtg ctg ttc gga gag 48 Met Ala Leu Ala Pro Glu Arg Ala Ala Pro Arg Val Leu Phe Gly Glu tgg ctc ctt gga gag atc agc agc ggc tgc tat gag ggg ctg cag tgg 96 Trp Leu Leu Gly Glu Ile Ser Ser Gly Cys Tyr Glu Gly Leu Gln Trp ctg gac gag gcc cgc acc tgt ttc cgc gtg ccc tgg aag cac ttc gcg 144 Leu Asp Glu Ala Arg Thr Cys Phe Arg Val Pro Trp Lys His Phe Ala cgc aag gac ctg agc gag gcc gac gcg cgc atc ttc aag gcc tgg get 192 Arg Lys Asp Leu Ser Glu Ala Asp Ala Arg Ile Phe Lys Ala Trp Ala gtg gcc cgc ggc agg tgg ccg cct agc agc agg gga ggt ggc ccg ccc 240 Val Ala Arg Gly Arg Trp Pro Pro Ser Ser Arg Gly Gly Gly Pro Pro ccc gag get gag act gcg gag cgc gcc ggc tgg aaa acc aac ttc cgc 288 Pro Glu Ala Glu Thr Ala Glu Arg Ala Gly Trp Lys Thr Asn Phe Arg tgc gca ctg cgc agc acg cgt cgc ttc gtg atg ctg cgg gat aac tcg 336 Cys Ala Leu Arg Ser Thr Arg Arg Phe Val Met Leu Arg Asp Asn Ser ggg gac ccg gcc gac ccg cac aag gtg tac gcg ctc agc cgg gag ctg 384 Gly Asp Pro Ala Asp Pro His Lys Val Tyr Ala Leu Ser Arg Glu Leu tgc tgg cga gaa ggc cca ggc acg gac cag act gag gca gag gcc ccc 432 Cys Trp Arg Glu Gly Pro Gly Thr Asp Gln Thr Glu Ala Glu Ala Pro gca get gtc cca cca cca cag ggt ggg ccc cca ggg cca ttc ttg gca 480 Ala Ala Val Pro Pro Pro Gln Gly Gly Pro Pro Gly Pro Phe Leu Ala cac aca cat get gga ctc caa gcc cca ggc ccc ctc cct gcc cca get 528 His Thr His Ala Gly Leu Gln Ala Pro Gly Pro Leu Pro Ala Pro Ala ggt gac aag ggg gac ctc ctg ctc cag gca gtg caa cag agc tgc ctg 576 Gly Asp Lys Gly Asp Leu Leu Leu Gln Ala Val Gln Gln Ser Cys Leu gca gac cat ctg ctg aca gcg tca tgg ggg gca gat cca gtc cca acc 624 Ala Asp His Leu Leu Thr Ala Ser Trp Gly Ala Asp Pro Val Pro Thr aag get cct gga gag gga caa gaa ggg ctt ccc ctg act ggg gcc tgt 672 Lys Ala Pro Gly Glu Gly Gln Glu Gly Leu Pro Leu Thr Gly Ala Cys get gga ggc cca ggg ctc cct get ggg gag ctg tac ggg tgg gca gta 720 Ala Gly Gly Pro Gly Leu Pro Ala Gly Glu Leu Tyr Gly Trp Ala Val gag acg acc ccc agc ccc ggg ccc cag ccc gcg gca cta acg aca ggc 768 Glu Thr Thr Pro Ser Pro Gly Pro Gln Pro Ala Ala Leu Thr Thr Gly gag gcc gcg gcc cca gag tcc ccg cac cag gca gag ccg tac ctg tca 816 Glu Ala Ala Ala Pro Glu Ser Pro His Gln Ala Glu Pro Tyr Leu Ser ccc tcc cca agc gcc tgc acc gcg gtg caa gag ccc agc cca ggg gcg 864 Pro Ser Pro Ser Ala Cys Thr Ala Val Gln Glu Pro Ser Pro Gly Ala ctg gac gtg acc atc atg tac aag ggc cgc acg gtg ctg cag aag gtg 912 Leu Asp Val Thr Ile Met Tyr Lys Gly Arg Thr Val Leu Gln Lys Val gtgggacac ccgagctgcacg ttcctatac ggcccccca gacccaget 960 ValGlyHis ProSerCysThr PheLeuTyr GlyProPro AspProAla gtccgggcc acagacccccag caggtagca ttccccagc cctgccgag 1008 ValArgAla ThrAspProGln GlnValAla PheProSer ProAlaGlu ctcccggac cagaagcagctg cgctacacg gaggaactg ctgcggcac 1056 LeuProAsp GlnLysGlnLeu ArgTyrThr GluGluLeu LeuArgHis gtggcccct gggttgcacctg gagcttcgg gggccacag ctgtgggcc 1104 ValAlaPro GlyLeuHisLeu GluLeuArg GlyProGln LeuTrpAla cggcgcatg ggcaagtgcaag gtgtactgg gaggtgggc ggaccccca 1152 ArgArgMet GlyLysCysLys ValTyrTrp GluValGly GlyProPro ggctccgcc agcccctccacc ccagcctgc ctgctgcct cggaactgt 1200 GlySerAla SerProSerThr ProAlaCys LeuLeuPro ArgAsnCys gacaccccc atcttcgacttc agagtcttc ttccaagag ctggtggaa 1248 AspThrPro IlePheAspPhe ArgValPhe PheGlnGlu LeuValGlu ttccgggca cggcagcgccgt ggctcccca cgctatacc atctacctg 1296 PheArgAla ArgGlnArgArg GlySerPro ArgTyrThr IleTyrLeu ggcttcggg caggacctgtca getgggagg cccaaggag aagagcctg 1344 GlyPheGly GlnAspLeuSer AlaGlyArg ProLysGlu LysSerLeu gtcctggtg aagctggaaccc tggctgtgc cgagtgcac ctagagggc 1392 ValLeuVal LysLeuGluPro TrpLeuCys ArgValHis LeuGluGly acgcagcgt gagggtgtgtct tccctggat agcagcgac ctcgacctc 1440 ThrGlnArg GluGlyValSer SerLeuAsp SerSerAsp LeuAspLeu tgcctgtcc agcgccaacagc ctctatgac gacatcgag tgcttcctt 1488 CysLeuSer SerAlaAsnSer LeuTyrAsp AspIleGlu CysPheLeu atggagctg gagcagcccgcc tag 1512 MetGluLeu GluGlnProAla <210> 9 <211> 503 <212 > PRT
<213> Homo sapiens <400> 9 Met Ala Leu Ala Pro Glu Arg Ala Ala Pro Arg Val Leu Phe Gly Glu Trp Leu Leu Gly Glu Ile Ser Ser Gly Cys Tyr Glu Gly Leu Gln Trp Leu Asp Glu Ala Arg Thr Cys Phe Arg Val Pro Trp Lys His Phe Ala Arg Lys Asp Leu Ser Glu Ala Asp Ala Arg Ile Phe Lys Ala Trp Ala Val Ala Arg Gly Arg Trp Pro Pro Ser Ser Arg Gly Gly Gly Pro Pro Pro Glu Ala Glu Thr Ala Glu Arg Ala Gly Trp Lys Thr Asn Phe Arg Cys Ala Leu Arg Ser Thr Arg Arg Phe Val Met Leu Arg Asp Asn Ser Gly Asp Pro Ala Asp Pro His Lys Val Tyr Ala Leu Ser Arg Glu Leu Cys Trp Arg Glu Gly Pro Gly Thr Asp Gln Thr Glu Ala Glu Ala Pro Ala Ala Val Pro Pro Pro Gln Gly Gly Pro Pro Gly Pro Phe Leu Ala His Thr His Ala Gly Leu Gln Ala Pro Gly Pro Leu Pro Ala Pro Ala Gly Asp Lys Gly Asp Leu Leu Leu Gln Ala Val Gln Gln Ser Cys Leu Ala Asp His Leu Leu Thr Ala Ser Trp Gly Ala Asp Pro Val Pro Thr Lys Ala Pro Gly Glu Gly Gln Glu Gly Leu Pro Leu Thr Gly Ala Cys Ala Gly Gly Pro Gly Leu Pro Ala Gly Glu Leu Tyr Gly Trp Ala Val Glu Thr Thr Pro Ser Pro Gly Pro Gln Pro Ala Ala Leu Thr Thr Gly Glu Ala Ala Ala Pro Glu Ser Pro His Gln Ala Glu Pro Tyr Leu Ser Pro Ser Pro Ser Ala Cys Thr Ala Val Gln Glu Pro Ser Pro Gly Ala Leu Asp Val Thr Ile Met Tyr Lys Gly Arg Thr Val Leu Gln Lys Val Val Gly His Pro Ser Cys Thr Phe Leu Tyr Gly Pro Pro Asp Pro Ala Val Arg Ala Thr Asp Pro Gln Gln Val Ala Phe Pro Ser Pro Ala Glu Leu Pro Asp Gln Lys Gln Leu Arg Tyr Thr Glu Glu Leu Leu Arg His Val Ala Pro Gly Leu His Leu Glu Leu Arg Gly Pro Gln Leu Trp Ala Arg Arg Met Gly Lys Cys Lys Val Tyr Trp Glu Val Giy Gly Pro Pro Gly Ser Ala Ser Pro Ser Thr Pro Ala Cys Leu Leu Pro Arg Asn Cys Asp Thr Pro Ile Phe Asp Phe Arg Val Phe Phe Gln Glu Leu Val Glu Phe Arg Ala Arg Gln Arg Arg Gly Ser Pro Arg Tyr Thr Ile Tyr Leu Gly Phe Gly Gln Asp Leu Ser Ala Gly Arg Pro Lys Glu Lys Ser Leu Val Leu Val Lys Leu Glu Pro Trp Leu Cys Arg Val His Leu Glu Gly Thr Gln Arg Glu Gly Val Ser Ser Leu Asp Ser Ser Asp Leu Asp Leu Cys Leu Ser Ser Ala Asn Ser Leu Tyr Asp Asp Ile Glu Cys Phe Leu Met Glu Leu Glu Gln Pro Ala <210> 10 <211> 1629 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (1)..(1626) <400> 10 atggccttggetcct gagagggca gccccacgc gtgctgttc ggagag 48 MetAlaLeuAlaPro GluArgAla AlaProArg ValLeuPhe GlyGlu tggctccttggagag atcagcagc ggctgctat gaggggctg cagtgg 96 TrpLeuLeuGlyGlu IleSerSer GlyCysTyr GluGlyLeu GlnTrp ctggacgaggcccgc acctgtttc cgcgtgccc tggaagcac ttcgcg 144 LeuAspGluAlaArg ThrCysPhe ArgValPro TrpLysHis PheAla cgcaaggacctgagc gaggccgac gcgcgcatc ttcaaggcc tggget 192 ArgLysAspLeuSer GluAlaAsp AlaArgIle PheLysAla TrpAla gtggcccgcggcagg tggccgcct agcagcagg ggaggtggc ccgccc 240 ValAlaArgGlyArg TrpProPro SerSerArg GlyGlyGly ProPro cccgaggetgagact gcggagcgc gccggctgg aaaaccaac ttccgc 288 ProGluAlaGluThr AlaGluArg AlaGlyTrp LysThrAsn PheArg tgcgcactgcgcagc acgcgtcgc ttcgtgatg ctgcgggat aactcg 336 CysAlaLeuArgSer ThrArgArg PheValMet LeuArgAsp AsnSer ggg gacccggcc gacccgcacaag gtgtacgcg ctcagccgg gagctg 384 Gly AspProAla AspProHisLys ValTyrAla LeuSerArg GluLeu tgc tggcgagaa ggcccaggcacg gaccagact gaggcagag gccccc 432 Cys TrpArgGlu GlyProGlyThr AspGlnThr GluAlaGlu AlaPro gca getgtccca ccaccacagggt gggccccca gggccattc ttggca 480 Ala AlaValPro ProProGlnGly GlyProPro GlyProPhe LeuAla cac acacatget ggactccaagcc ccaggcccc ctccctgcc ccaget 528 His ThrHisAla GlyLeuGlnAla ProGlyPro LeuProAla ProAla ggt gacaagggg gacctcctgctc caggcagtg caacagagc tgcctg 576 Gly AspLysGly AspLeuLeuLeu GlnAlaVal GlnGlnSer CysLeu gca gaccatctg ctgacagcgtca tggggggca gatccagtc ccaacc 624 Ala AspHisLeu LeuThrAlaSer TrpGlyAla AspProVal ProThr aag getcctgga gagggacaagaa gggcttccc ctgactggg gcctgt 672 Lys AlaProGly GluGlyGlnGlu GlyLeuPro LeuThrGly AlaCys get ggaggccca gggctccctget ggggagctg tacgggtgg gcagta 720 Ala GlyGlyPro GlyLeuProAla GlyGluLeu TyrGlyTrp AlaVal gag acgaccccc agccccacttct gatacccag gaagacatt ctggat 768 Glu ThrThrPro SerProThrSer AspThrGln GluAspIle LeuAsp gag ttactgggt aacatggtgttg gccccactc ccagatccg ggaccc 816 Glu LeuLeuGly AsnMetValLeu AlaProLeu ProAspPro GlyPro cca agcctgget gtagcccctgag ccctgccct cagcccctg cggagc 864 Pro SerLeuAla ValAlaProGlu ProCysPro GlnProLeu ArgSer ccc agcttggac aatcccactccc ttcccaaac ctggggccc tctgag 912 Pro SerLeuAsp AsnProThrPro PheProAsn LeuGlyPro SerGlu aac ccactgaag cggctgttggtg ccgggggaa gagtgggag ttcgag 960 Asn ProLeuLys ArgLeuLeuVal ProGlyGlu GluTrpGlu PheGlu gtg acagccttc taccggggccgc caagtcttc cagcagacc atctcc 1008 Val ThrAlaPhe TyrArgGlyArg GlnValPhe GlnGlnThr IleSer tgc ccggagggc ctgcggctggtg gggtccgaa gtgggagac aggacg 1056 Cys ProGluGly LeuArgLeuVal GlySerGlu ValGlyAsp ArgThr ctg cctggatgg ccagtcacactg ccagaccct ggcatgtcc ctgaca 1104 Leu ProGlyTrp ProValThrLeu ProAspPro GlyMetSer LeuThr gac aggggagtg atgagctacgtg aggcatgtg ctgagctgc ctgggt 1152 Asp ArgGlyVal MetSerTyrVal ArgHisVal LeuSerCys LeuGly WO 99/51737 PCT/CA99/003i4 gggggactgget ctctggcgg gccgggcag tggctctgg gcccagcgg 1200 GlyGlyLeuAla LeuTrpArg AlaGlyGln TrpLeuTrp AlaGlnArg ctggggcactgc cacacatac tgggcagtg agcgaggag ctgctcccc 1248 LeuGlyHisCys HisThrTyr TrpAlaVal SerGluGlu LeuLeuPro aacagcgggcat gggcctgat ggcgaggtc cccaaggac aaggaagga 1296 AsnSerGlyHis GlyProAsp GlyGluVal ProLysAsp LysGluGly ggcgtgtttgac ctggggccc ttcattgta gatctgatt accttcacg 1344 GlyValPheAsp LeuGlyPro PheIleVal AspLeuIle ThrPheThr gaaggaagcgga cgctcacca cgctatgcc ctctggttc tgtgtgggg 1392 GluGlySerGly ArgSerPro ArgTyrAla LeuTrpPhe CysValGly gagtcatggccc caggaccag ccgtggacc aagaggctc gtgatggtc 1440 GluSerTrpPro GlnAspGln ProTrpThr LysArgLeu ValMetVal aaggttgtgccc acgtgcctc agggccttg gtagaaatg gcccgggta 1488 LysValValPro ThrCysLeu ArgAlaLeu ValGluMet AlaArgVal gggggtgcctcc tccctggag aatactgtg gacctgcac attgacaac 1536 GlyGlyAlaSer SerLeuGlu AsnThrVal AspLeuHis IleAspAsn gaccacccactc gacctcgac gacgaccag tacaaggcc tacctgcag 1584 AspHisProLeu AspLeuAsp AspAspGln TyrLysAla TyrLeuGln gacttggtggag ggcatggat ttccagggc cctggggag agctga 1629 AspLeuValGlu GlyMetAsp PheGlnGly ProGlyGlu Ser <210> 11 <211> 542 <212> PRT
<213> Homo sapiens <400> 11 Met Ala Leu Ala Pro Glu Arg Ala Ala Pro Arg Val Leu Phe Gly Glu Trp Leu Leu Gly Glu Ile Ser Ser Gly Cys Tyr Glu Gly Leu Gln Trp Leu Asp Glu Ala Arg Thr Cys Phe Arg Val Pro Trp Lys His Phe Ala Arg Lys Asp Leu Ser Glu Ala Asp Ala Arg Ile Phe Lys Ala Trp Ala Val Ala Arg Gly Arg Trp Pro Pro Ser Ser Arg Gly Gly Gly Pro Pro Pro Glu Ala Glu Thr Ala Glu Arg Ala Gly Trp Lys Thr Asn Phe Arg Cys Ala Leu Arg Ser Thr Arg Arg Phe Val Met Leu Arg Asp Asn Ser Gly Asp Pro Ala Asp Pro His Lys Val Tyr Ala Leu Ser Arg Glu Leu Cys Trp Arg Glu Gly Pro Gly Thr Asp Gln Thr Glu Ala Glu Ala Pro Ala Ala Val Pro Pro Pro Gln Gly Gly Pro Pro Gly Pro Phe Leu Ala His Thr His Ala Gly Leu Gln Ala Pro Gly Pro Leu Pro Ala Pro Ala Gly Asp Lys Gly Asp Leu Leu Leu Gln Ala Val Gln Gln Ser Cys Leu Ala Asp His Leu Leu Thr Ala Ser Trp Gly Ala Asp Pro Val Pro Thr Lys Ala Pro Gly Glu Gly Gln Glu Gly Leu Pro Leu Thr Gly Ala Cys Ala Gly Gly Pro Gly Leu Pro Ala Gly Glu Leu Tyr Gly Trp Ala Val Glu Thr Thr Pro Ser Pro Thr Ser Asp Thr Gln Glu Asp Ile Leu Asp Glu Leu Leu Gly Asn Met Val Leu Ala Pro Leu Pro Asp Pro Gly Pro Pro Ser Leu Ala Val Ala Pro Glu Pro Cys Pro Gln Pro Leu Arg Ser Pro Ser Leu Asp Asn Pro Thr Pro Phe Pro Asn Leu Gly Pro Ser Glu Asn Pro Leu Lys Arg Leu Leu Val Pro Gly Glu Glu Trp Glu Phe Glu Val Thr Ala Phe Tyr Arg Gly Arg Gln Val Phe Gln Gln Thr Ile Ser Cys Pro Glu Gly Leu Arg Leu Val Gly Ser Glu Val Gly Asp Arg Thr Leu Pro Gly Trp Pro Val Thr Leu Pro Asp Pro Gly Met Ser Leu Thr Asp Arg Gly Val Met Ser Tyr Val Arg His Val Leu Ser Cys Leu'Gly Gly Gly Leu Ala Leu Txp Arg Ala Gly Gln Trp Leu Trp Ala Gln Arg Leu Gly His Cys His Thr Tyr Trp Ala Val Ser Glu Glu Leu Leu Pro Asn Ser Gly His Gly Pro Asp Gly Glu Val Pro Lys Asp Lys Glu Gly Gly Val Phe Asp Leu Gly Pro Phe Ile Val Asp Leu Ile Thr Phe Thr Glu Gly Ser Gly Arg Ser Pro Arg Tyr Ala Leu Trp Phe Cys Val Gly Glu Ser Trp Pro Gln Asp Gln Pro Trp Thr Lys Arg Leu Val Met Val Lys Val Val Pro Thr Cys Leu Arg Ala Leu Val Glu Met Ala Arg Val Gly Gly Ala Ser Ser Leu Glu Asn Thr Val Asp Leu His Ile Asp Asn Asp His Pro Leu Asp Leu Asp Asp Asp Gln Tyr Lys Ala Tyr Leu Gln Asp Leu Val Glu Gly Met Asp Phe Gln Gly Pro Gly Glu Ser

Claims (34)

CLAIMS:
1. A modified interferon regulatory factor (IRF) protein, the protein comprising at least one modified serine or threonine phosphoacceptor site in the carboxy-terminus domain, with the proviso that where said IRF protein is IRF-3, said at least one modified phosphoacceptor site does not comprise Ser-385 or Ser-386.
2. The interferon regulatory factor (IRF) protein according to claim 1, wherein cytokine gene activation by the modified IRF is increased relative to cytokine gene activation by a corresponding wild type IRF protein.
3. The interferon regulatory factor (IRF) protein according to claim 1 or 2, wherein the modified IRF is an IRF-3 protein modified at at least one serine or threonine phosphoacceptor site.
4. The interferon regulatory factor (IRF) protein according to claim 1 or 2, wherein the modified IRF is an IRF-7 protein modified at at least one serine or threonine phosphoacceptor site.
5. The interferon regulatory factor (IRF) protein according to any one of claims 1 to 4, wherein the at least one modified phosphoacceptor site is modified by phosphorylation.
6. The interferon regulatory factor (IRF) protein according to any one of claims 1 to 4, wherein the at least one modified phosphoacceptor site comprises an amino acid residue having an acidic side chain.
7. The interferon regulatory factor (IRF) protein according to claim 6, wherein the amino acid residue is aspartic acid.
8. The interferon regulatory factor (IRF) protein according to claim 5, wherein the modified IRF is IRF-3 modified at a site selected from at least one of Ser-396, Ser-398, Ser-402, Thr-404 and Ser-405.
9. The interferon regulatory factor (IRF) protein according to claim 8, wherein the modified IRF is IRF-3 modified at Ser-396, Ser-398, Ser-402, Thr-404 and Ser-405 sites.
10. The interferon regulatory factor (IRF) protein according to claim 9, wherein the modified IRF comprises a carboxy-terminus domain of 5er-396, Ser-398, Ser-402, Thr-404 and Ser-405 and an amino-terminus domain from IRF-7.
11. The interferon regulatory factor (IRF) protein according to claim 6 or 7, wherein the modified IRF is IRF-3 modified at a site selected from at least one of Ser-396, Ser-398, Ser-402, Thr-404 and Ser-405.
12. The interferon regulatory factor (IRF) protein according to claim 11, wherein the modified IRF is IRF-3 modified at Ser-396, Ser-398, Ser-402, Thr-404 and Ser-405 sites.
13. The interferon regulatory factor (IRF) protein according to claim 12 having SEQ ID NO. 2 (IRF-3 (5D)).
14. The interferon regulatory factor (IRF) protein according to claim 12, wherein the modified IRF comprises a carboxy-terminus domain of Ser-396, Ser-398, Ser-402, Thr-404 and Ser-405 and an amino-terminus domain from IRF-7.
15. The interferon regulatory factor (IRF) protein according to claim 14, wherein the modified IRF has an amino-terminal domain comprising residues 1 to 246 of IRF-7 and a carboxy-terminal domain comprising residues 132 to 427 of IRF-3 modified by replacement each of Ser-396, Ser-398, Ser-402, Thr-404 and Ser-405 by an aspartic acid residue.
16. The interferon regulatory factor (IRF) protein according to claim 15 having SEQ ID NO. 11 (IRF-7{1-246)/
IRF-3(5D) (132-427)).
17. The interferon regulatory factor (IRF) protein according to claim 5, wherein the modified IRF is IRF-7 modified at a site selected from at least one of Ser-477 and Ser-479.
18. The interferon regulatory factor (IRF) protein according to claim 17, wherein the modified IRF-7 is modified at Ser-477 and Ser-479 sites.
19. The interferon regulatory factor (IRF) protein according to claims 6 or 7, wherein the modified IRF is IRF-7 modified at a site selected from at least one of Ser-477 and Ser-479.
20. The interferon regulatory factor (IRF) protein according to claim 19, wherein the modified IRF-7 is modified at Ser-477 and Ser-479 sites.
21. The interferon regulatory factor (IRF) protein according to claim 20 having SEQ ID NO. 9 (IRF-7(2D)).
22. A nucleotide sequence selected from:
(a) a first nucleotide sequence which encodes the interferon regulatory factor (IRF) protein according to any one of claims 6, 7, 11 to 16, 19, 20 or 21, or (b) a second nucleotide sequence that is hybridizable under stringent conditions with the complement of the first nucleotide sequence, wherein said second nucleotide sequence encodes an IRF protein wherein at least one serine or threonine phosphoacceptor site comprises an amino acid residue having an acidic side chain.
23. The nucleotide sequence according to claim 22, having SEQ ID NO. 1.
24. The nucleotide sequence according to claim 22, having SEQ ID NO. 8.
25. The nucleotide sequence according to claim 22, having SEQ ID NO. 10.
26. A pharmaceutical composition comprising an effective amount of the interferon regulatory factor (IRF) protein according to any one of claims 1 to 21, together with a pharmaceutically acceptable carrier, for the treatment of a viral infection.
27. The pharmaceutical composition according to claim 26, wherein the viral infection is selected from an influenza infection, a herpes infection, a hepatitis infection and an HIV
infection.
28. Use of the interferon regulatory factor (IRF) protein according to any one of claims 1 to 21 to activate a cytokine gene.
29. The use according to claim 28, wherein the cytokine gene is an interferon gene or a chemokine gene.
30. Use of the interferon regulatory factor (IRF) protein according to any one of claims 1 to 21 in cancer treatment.
31. Use of the nucleotide sequence according to any one of claims 22 to 25 to modify a target cell of an organism.
32. A commercial package containing as an active pharmaceutical ingredient the pharmaceutical composition according to claim 26 together with instructions for its use for the treatment of a viral infection.
33. The commercial package according to claim 32, wherein the viral infection is selected from an influenza infection, a herpes infection, a hepatitis infection and an HIV infection.
34. A commercial package containing as an active pharmaceutical ingredient the interferon regulatory factor (IRF) protein according to any one of claims 1 to 21 together with instructions for its use for the treatment of cancer.
CA002325354A 1998-04-07 1999-04-07 Highly active forms of interferon regulatory factor proteins Abandoned CA2325354A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA002325354A CA2325354A1 (en) 1998-04-07 1999-04-07 Highly active forms of interferon regulatory factor proteins

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CA002234588A CA2234588A1 (en) 1998-04-07 1998-04-07 Virus induced phosphorylation of interferon regulatory factor-3
CA2,234,588 1998-04-07
CA002325354A CA2325354A1 (en) 1998-04-07 1999-04-07 Highly active forms of interferon regulatory factor proteins
PCT/CA1999/000314 WO1999051737A1 (en) 1998-04-07 1999-04-07 Highly active forms of interferon regulatory factor proteins

Publications (1)

Publication Number Publication Date
CA2325354A1 true CA2325354A1 (en) 1999-10-14

Family

ID=25680127

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002325354A Abandoned CA2325354A1 (en) 1998-04-07 1999-04-07 Highly active forms of interferon regulatory factor proteins

Country Status (1)

Country Link
CA (1) CA2325354A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114874313A (en) * 2015-11-16 2022-08-09 Ubi蛋白公司 Method for extending protein half-life
EP3740198A4 (en) * 2018-01-18 2023-08-09 Fred Hutchinson Cancer Center Altering inflammatory states of immune cells in vivo by modulating cellular activation states
US11872195B2 (en) 2016-04-14 2024-01-16 Fred Hutchinson Cancer Center Compositions and methods to program therapeutic cells using targeted nucleic acid nanocarriers

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114874313A (en) * 2015-11-16 2022-08-09 Ubi蛋白公司 Method for extending protein half-life
US11872195B2 (en) 2016-04-14 2024-01-16 Fred Hutchinson Cancer Center Compositions and methods to program therapeutic cells using targeted nucleic acid nanocarriers
EP3740198A4 (en) * 2018-01-18 2023-08-09 Fred Hutchinson Cancer Center Altering inflammatory states of immune cells in vivo by modulating cellular activation states

Similar Documents

Publication Publication Date Title
Lin et al. Virus-dependent phosphorylation of the IRF-3 transcription factor regulates nuclear translocation, transactivation potential, and proteasome-mediated degradation
Sangfelt et al. Induction of Cip/Kip and Ink4 cyclin dependent kinase inhibitors by interferon-α in hematopoietic cell lines
Mamane et al. Interferon regulatory factors: the next generation
Matsumura et al. Thrombopoietin-induced differentiation of a human megakaryoblastic leukemia cell line, CMK, involves transcriptional activation of p21 WAF1/Cip1 by STAT5
Pitha et al. Role of the interferon regulatory factors (IRFs) in virus-mediated signaling and regulation of cell growth
D'Souza et al. Nuclear factor κB-dependent activation of the antiapoptotic bfl-1 gene by the Epstein-Barr virus latent membrane protein 1 and activated CD40 receptor
US20060286091A1 (en) RelA-ASSOCIATED INHIBITOR, PROCESS FOR PRODUCING THE SAME AND UTILIZATION THEREOF
Roulston et al. Chronic human immunodeficiency virus type 1 infection stimulates distinct NF-kappa B/rel DNA binding activities in myelomonoblastic cells
US20050181420A1 (en) Polypeptide that interacts with heat shock proteins
US20030077262A1 (en) Methods and compositions for modulating apoptosis
US6338949B1 (en) Nucleic acids encoding receptor recognition factor stat4 and methods of use thereof
Kieser et al. Protein kinase C-zeta reverts v-raf transformation of NIH-3T3 cells.
EP1506784A1 (en) Identification of novel factors that block programmed cell death or apoptosis by targeting JNK
CA2325354A1 (en) Highly active forms of interferon regulatory factor proteins
US20130330738A1 (en) Sumoylation Control Agent and Uses Thereof
AU2001274772B2 (en) Inhibitory PAS domain protein (IPAS) and screening methods related to angiogenesis and tumor progression
US20020147147A1 (en) Nucleic acid molecules encoding a protein interacting with the chemokine receptor CCR5 or other chemokine receptor family members
US7053194B2 (en) Compositions and methods for p53-mediated repression of gene expression
WO1999051737A1 (en) Highly active forms of interferon regulatory factor proteins
US6680366B1 (en) Nucleotide sequence encoding a modulator of NF-κB
AU8072194A (en) Receptor recognition factors, protein sequences and methods of use thereof
CA2132317A1 (en) Ifn receptors recognition factors, protein sequences and methods of use thereof
AU2001274772A1 (en) Inhibitory PAS domain protein (IPAS) and screening methods related to angiogenesis and tumor progression
US7094873B1 (en) Polypeptide that interacts with heat shock proteins
US7638304B2 (en) Hybrid fusion protein transcription regulator to induce interferon target gene expression

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued