US20030032609A1 - Methods of modulating of angiogenesis - Google Patents

Methods of modulating of angiogenesis Download PDF

Info

Publication number
US20030032609A1
US20030032609A1 US10/121,235 US12123502A US2003032609A1 US 20030032609 A1 US20030032609 A1 US 20030032609A1 US 12123502 A US12123502 A US 12123502A US 2003032609 A1 US2003032609 A1 US 2003032609A1
Authority
US
United States
Prior art keywords
epas1
seq
polypeptide
dna
amino acid
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
US10/121,235
Inventor
Mu-En Lee
Koji Maemura
Chung-Ming Hsieh
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Harvard College
Original Assignee
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
Application filed by Individual filed Critical Individual
Priority to US10/121,235 priority Critical patent/US20030032609A1/en
Assigned to PRESIDENT AND FELLOWS OF HARVARD COLLEGE reassignment PRESIDENT AND FELLOWS OF HARVARD COLLEGE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HSIEH, CHUNG-MING, LE, MU-EN, MAEMURA, KOJI
Assigned to PRESIDENT AND FELLOWS OF HARVARD COLLEGE reassignment PRESIDENT AND FELLOWS OF HARVARD COLLEGE CORRECTIVE ASSIGNMENT TO CORRECT THE NAME OF THE ASSIGNOR. FILED ON APRIL 12, 2002, RECORDED ON REEL 012796 FRAME 0037 ASSIGNOR HEREBY CONFIRMS THE ASSIGNMENT OF THE ENTIRE INTEREST. Assignors: HSIEH, CHUNG-MING, LEE, MU-EN, MAEMURA, KOJI
Publication of US20030032609A1 publication Critical patent/US20030032609A1/en
Assigned to NIH - DEITR reassignment NIH - DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: PRESIDENT AND FELLOWS OF HARVARD COLLEGE
Abandoned legal-status Critical Current

Links

Images

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
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06FELECTRIC DIGITAL DATA PROCESSING
    • G06F16/00Information retrieval; Database structures therefor; File system structures therefor
    • G06F16/20Information retrieval; Database structures therefor; File system structures therefor of structured data, e.g. relational data
    • G06F16/28Databases characterised by their database models, e.g. relational or object models
    • G06F16/289Object oriented databases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to vascular therapy.
  • Angiogenesis results from endothelial cell proliferation induced by angiogenic factors. Angiogenic factors bind to receptors on endothelial cells which line blood vessels. This event triggers signals which cause the cells to proliferate; the proliferating endothelial cells secrete proteases which digest the basement membrane surrounding a vessel. The junctions between the endothelial cells are altered, allowing projections from the cells to pass through the space created. These outgrowths then become new blood vessels, e.g., capillaries.
  • VEGF Vascular endothelial cell growth factor
  • VEGF-Rs VEGF receptors
  • VEGF signaling is mediated by two VEGF-Rs, the endothelial cell-specific tyrosine kinase receptors, flt-1 and KDR/flk-1.
  • the invention is based on the discovery that endothelial PAS domain protein-1 (EPAS1) binds to cis-acting regulatory sequences associated with genes encoding such angiogenic factors as VEGF and VEGF-Rs such as KDR/flk-1 and flt-1, thereby transactivating the promoters of such genes. Accordingly, the invention features a method of increasing the level of EPAS1 in a cell, e.g., an endothelial cell. An increase in the level of EPAS1 leads to increased promoter transactivation and increased transcription of genes encoding angiogenic factors which participate in the blood vessel formation.
  • EPAS1 endothelial PAS domain protein-1
  • the invention also includes a novel basic helix-loop-helix/Per-AhR-Arnt-Sim (bHLH/PAS) protein which binds to EPAS1 and forms a heterodimer which transactivates transcription of genes encoding angiogenic factors.
  • bHLH/PAS novel basic helix-loop-helix/Per-AhR-Arnt-Sim
  • Angiogenic factors are proteins or polypeptides and ligands thereof that participate in the process of new blood vessel formation.
  • angiogenic factors include VEGF, VEGF-Rs, and other signalling proteins such as intracellular tyrosine kinases which participate in the angiogenic process.
  • the angiogenic factors are expressed in endothelial cells, e.g., VEGF, VEGF-Rs such as KDR/flk-1 or flt-1, and tyrosine kinases such as Tie2.
  • a method of inhibiting angiogenesis in a mammal is carried out by administering to the mammal a compound which inhibits binding of EPAS1 to cis-acting transcription regulatory DNA associated with a gene encoding an angiogenic factor.
  • Angiogenesis is also inhibited by administering a compound which inhibits binding of EPAS1 to ARNT4, i.e., a compound which inhibits the formation of a functional heterodimer that can transactivate a promoter of gene encoding an angiogenic factor.
  • the angiogenic factor is preferably VEGF., a VEGF-R such as KDR/flk-1 or flt-1.
  • the compound inhibits transcription of the angiogenic factor by binding to a cis-acting regulatory sequence such as the sequence 5′GCCCTACGTGCTGTCTCA 3′ (SEQ ID NO: 1 ) in VEGF promoter DNA.
  • a cis-acting regulatory sequence such as the sequence 5′GCCCTACGTGCTGTCTCA 3′ (SEQ ID NO: 1 ) in VEGF promoter DNA.
  • the compound is an EPAS1 polypeptide that binds to a cis-acting regulatory sequence of a gene but fails to transactivate the promoter of the gene, e.g. a polypeptide lacking a transactivation domain (amino acids 486 - 690 of EPAS1).
  • the EPAS1 polypeptide contains the N-terminal binding domain (amino acids 14 - 67 of EPAS1; RRKEKSRDAARCRRSKETEVFYELAHELPLPHSVSSHLDKASIMRLEISFLRTH; SEQ ID NO: 3 ), more preferably the EPAS polypeptide contains amino acids 1 - 485 of human EPAS1.
  • EPAS1 dominant negative mutant 1 MTADKEKKRS SSERRKEKSR DAARCRRSKE TEVFYELAHE LPLPHSVSSH 51 LDKASIMRLE ISFLRTHKLL SSVCSENESE AEADQQMDNL YLKALEGFIA 101 VVTQDGDMIF LSENISKFMG LTQVELTGHS IFDFTHPCDH EEIRENLSLK 151 NGSGFGKKSK DMSTERDFFM RMKCTVTNRG RTVNLKSATW KVLHCTGQVK 201 VYNNCPPHNS LCGYKEPLLS CLIIMCEPIQ HPSHMDIPLD SKTFLSRHSM 251 DMKFTYCDDR ITELIGYHPE ELLGRSAYEF YHALDSENMT KSHQNLCTKG 301 QVVSGQYRML AKHGGYVWLE TQGT
  • the method may be carried out by administering DNA encoding such polypeptides.
  • the compound is a nucleic acid encoding an EPAS1 polypeptide lacking amino acids 486 - 690 of EPAS1.
  • the nucleic acid encodes a dominant negative mutant of EPAS1 which contains amino acids 1 - 485 of wild type EPAS1, i.e., SEQ ID NO: 5 .
  • the compound is a antisense nucleic acid molecule containing at least 10 nucleotides the sequence of which is complementary to an mRNA encoding all or part of a wild type EPAS1 polypeptide.
  • the compound e.g., an antisense oligonucleotide or antisense RNA produced from an antisense template, inhibits EPAS1 expression.
  • the compound may inhibit EPAS1 expression by inhibiting translation of EPAS1 mRNA.
  • antisense therapy is carried out by administering a single stranded nucleic acid complementary at least a portion of EPASI MRNA to interfere with the translation of MRNA into protein, thus reducing the amount of functional EPAS1 produced in the cell.
  • a reduction in the amount of functional transactivating EPAS1 reduces the level of transcription of angiogenic factors such as VEGF or VEGF-Rs, resulting in a decrease in new blood vessel formation.
  • the compound is an EPASI-specific intrabody, i.e., a recombinant single chain EPAS1-specific antibody that is expressed inside a target cell, e.g., a vascular endothelial cell.
  • a target cell e.g., a vascular endothelial cell.
  • Such an intrabody binds to endogenous intracellular EPAS1 and prevents it from binding to its target cis-acting regulatory sequence in the promoter region of a gene encoding an angiogenic factor such as VEGF or a VEGF-R.
  • An ARNT4-specific intrabody is also useful to inhibit angiogenesis.
  • Angiogenesis contributes to the progression of atherosclerotic lesions.
  • compounds are administered to a site of an atherosclerotic lesion in a mammal to inhibit growth of a lesion.
  • Compounds may also be locally administered to a tumor site to reduce blood vessel formation, thereby depriving a tumor of blood supply and inhibiting tumor growth.
  • VEGF itself is a growth factor for some tumors; the methods described above directly inhibit VEGF expression, and thus, are particularly useful for treating such tumor types.
  • the invention also includes an antibody which binds to EPAS1.
  • the antibody preferably binds to the C-terminal portion of EPAS1 (e.g., a polypeptide having the amino acid of SEQ ID NO: 17 or 18 ).
  • the antibody is a polyclonal or monoclonal antibody which specifically binds to the EPAS1.
  • the antibody binds to an epitope within the C-terminal transactivation domain (SEQ ID NO: 2 ).
  • the invention encompasses not only an intact monoclonal antibody, but also an immunologically-active antibody fragment, e.g., a Fab or (Fab) 2 fragment; an engineered single chain Fv molecule; or a chimeric molecule, e.g., an antibody which contains the binding specificity of one antibody, e.g., of murine origin, and the remaining portions of another antibody, e.g., of human origin.
  • an immunologically-active antibody fragment e.g., a Fab or (Fab) 2 fragment
  • an engineered single chain Fv molecule e.g., an antibody which contains the binding specificity of one antibody, e.g., of murine origin, and the remaining portions of another antibody, e.g., of human origin.
  • a compound e.g., DNA encoding EPASI or a functional fragment thereof, which increases expression of VEGF or a VEGF-R in an endothelial cell is administered to a mammal, e.g., an adult mammal which has been identified as being in need of therapy to promote angiogenesis such as a patient suffering from peripheral vascular disease.
  • a functional fragment of EPAS1 is one which binds to DNA in the promoter region of a gene encoding an angiogenic factor.
  • the invention also features an EPAS-binding element, ARNT4 and a nucleic acid which encodes ARNT4.
  • the nucleic acid includes a sequence which encodes the amino acid sequence a naturally-occurring human ARNT4 (SEQ ID NO: 19 ).
  • the DNA may encode a naturally-occurring mammalian ARNT4 polypeptide such as a human, rat, mouse, guinea pig, hamster, dog, cat, pig, cow, goat, sheep, horse, monkey, or ape ARNT4.
  • the DNA encodes a human ARNT4 polypeptide, e.g., a polypeptide which contains part or all of the amino acid sequence of SEQ ID NO: 19 .
  • the invention includes degenerate variants of the human cDNA (SEQ ID NO: 20 ) .
  • the DNA contains a nucleotide sequence having at least 50% sequence identity to SEQ ID NO: 20 .
  • the DNA contains a sequence which encodes a human ARNT4 polypeptide, such as the coding sequence of SEQ ID NO: 20 (nucleotides 220 to 2025 of SEQ ID NO: 20 ).
  • the DNA contains a strand which hybridizes at high stringency to a strand of DNA having the sequence of SEQ ID NO: 20 , or the complement thereof.
  • the DNA has at least 50% sequence identity to SEQ ID NO: 20 and encodes a polypeptide having the biological activity of a ARNT4 polypeptide, e.g, the ability to bind to EPAS1 to form a heterodimer.
  • the DNA has at least 75% identity, more preferably 85% identity, more referably 90% identity, more preferably 95% identity, more preferably 99% identity, and most preferably 100% identity to the coding sequence of SEQ ID NO: 20 .
  • Conservative substitutions typically include substitutions within the following groups: glycine and alanine; valine, isoleucine, and leucine; aspartic acid and glutamic acid; asparagine and glutamine; serine and threonine; lysine and arginine; and phenylalanine and tyrosine.
  • Hybridization is carried out using standard techniques, such as those described in Ausubel et al. (Current Protocols in Molecular Biology, John Wiley & Sons, 1989).
  • “High stringency” refers to nucleic acid hybridization and wash conditions characterized by high temperature and low salt concentration: wash conditions of 65° C. at a salt concentration of 0.1 ⁇ SSC.
  • “Low” to “moderate” stringency denotes DNA hybridization and wash conditions characterized by low temperature and high salt concentration: wash conditions of less than 60° C. at a salt concentration of 1.0 ⁇ SSC.
  • high stringency conditions include hybridization at 42° C., and 50% formamide; a first wash at 65° C., 2 ⁇ SSC, and 1% SDS; followed by a second wash at 65° C. and 0.1% ⁇ SSC.
  • Lower stringency conditions suitable for detecting DNA sequences having about 50% sequence identity to an ARNT4 gene are detected by, for example, hybridization at 42° C. in the absence of formamide; a first wash at 42° C., 6 ⁇ SSC, and 1% SDS; and a second wash at 50° C., 6 ⁇ SSC, and 1% SDS.
  • a vector containing an ARNT4-encoding DNA is also within the invention.
  • the DNA which includes an ARNT4-encoding DNA is less than 5 kilobases in length; more preferably, the DNA is less than 4 kilobases in length, more preferably the DNA is less than 3 kilobases in length, and most preferably the DNA is approximately 2 kilobases or less in length.
  • the invention also provides a method of directing cardiac-specific or smooth muscle cell-specific expression of a protein by introducing into a cell an isolated DNA containing a sequence encoding the protein operably linked to the tissue-specific promoter.
  • a cell containing the DNA or vector of the invention is also within the invention.
  • substantially pure DNA DNA that has a naturally-occurring sequence or that is free of the genes which, in the naturally-occurring genome of the organism from which the DNA of the invention is derived, flank the ARNT4 gene.
  • the term therefore includes, for example, a recombinant DNA which is incorporated into a vector, into an autonomously replicating plasmid or virus, or into the genomic DNA of a procaryote or eucaryote at a site other than its natural site; or which exists as a separate molecule (e.g., a cDNA or a genomic or cDNA fragment produced by PCR or restriction endonuclease digestion) independent of other sequences. It also includes a recombinant DNA which is part of a hybrid gene encoding additional polypeptide sequence.
  • ARNT4 polypeptide includes a polypeptide having the amino acid sequence and length of the naturally-occurring ARNT4 as well as fragments of the full-length naturally-occurring ARNT4.
  • the polypeptide contains the amino acid sequence of SEQ ID NO: 19 .
  • the polypeptide contains an amino acid sequence which is at least 50% identical to SEQ ID NO: 19 .
  • the amino acid sequence has at least 75% identity, more preferably 85% identity, more preferably 90% identity, more preferably 95% identity, more preferably 99% identity, and most preferably 100% identity to the amino acid sequence of SEQ ID NO: 19 .
  • the ARNT4 polypeptide may have the amino acid sequence of the naturally-occurring human polypeptide, e.g., a polypeptide which includes the amino acid sequence of SEQ ID NO: 19 .
  • the invention also encompasses a polypeptide with the amino acid sequence of a segment of SEQ ID NO: 17 which spans residues 75 to 128 , inclusive, or a segment spanning residues 155 to 207 , inclusive, of SEQ ID NO: 19 , or a segment spanning residues 232 to 384 of SEQ ID NO: 19 .
  • such a polypeptide has a biological activity of a naturally-occurring ARNT4 polypeptide, e.g, heterodimer formation with EPAS1 or the ability to transactivate transcription under the control of a VEGF promoter.
  • a substantially pure ARNT4 polypeptide is obtained by extraction from a natural source; by expression of a recombinant nucleic acid encoding a ARNT4 polypeptide; or by chemically synthesizing the protein.
  • a polypeptide or protein is substantially pure when it is separated from those contaminants which accompany it in its natural state (proteins and other naturally-occurring organic molecules).
  • the polypeptide is substantially pure when it constitutes at least 60%, by weight, of the protein in the preparation.
  • the protein in the preparation is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight, ARNT4.
  • substantially pure polypeptides include recombinant polypeptides derived from a eucaryote but produced in E. coli or another procaryote, or in a eucaryote other than that from which the polypeptide was originally derived.
  • the invention also includes a transgenic non-human mammal, the germ cells and somatic cells of which contain a null mutation in a gene encoding an ARNT4 polypeptide.
  • the null mutation is a deletion of part or all of an exon of ARNT4.
  • the mammal is a rodent such as a mouse.
  • An antibody which specifically binds to a ARNT4 polypeptide is also within the invention.
  • Angiogenesis is inhibited by administering to a mammal a compound which inhibits binding of EPAS1 to ARNT4 such as an ARNT4 polypeptide.
  • the compound is a polypeptide or peptide mimetic which contains the amino acid sequence of residues 75 to 128 , inclusive, of SEQ ID NO: 19 , the amino acid sequence of residues 155 to 207 , inclusive, of SEQ ID NO: 19 , or a the amino acid sequence of residues 232 to 384 of SEQ ID NO: 19 .
  • FIG. 1A is a bar graph showing dose-dependent transactivation of KDR/flk-1 promoter by EPAS1.
  • EPAS1 expression plasmid phEP-1 (0-6 ⁇ g), pcDNA3 (6-0 ⁇ g), and KDR/flk-1 reporter pGL2-4kb+296 (1 ⁇ g) were transfected into BAEC.
  • FIG. 1B is a bar graph showing that deletion of EPAS1 C-terminal region abolishes its ability to transactivate the KDR/flk-1 promoter.
  • Expression plasmids (6 ⁇ g each) and pGL2-4.Okb+296 (1 ⁇ g) were cotransfected into BAEC.
  • the plasmid pCMV- ⁇ GAL was cotransfected to correct for differences in transfection efficiency.
  • luciferase activity and ⁇ -galactosidase activity were measured, and normalized luciferase activity was calculated as described below.
  • the “fold induction” represents the ratio (mean ⁇ SE) of normalized luciferase activity in cells transfected with expression plasmid to that in cells transfected with empty vector (pcDNA3).
  • FIG. 2A is a bar graph showing transactivation of the KDR/flk-1 promoter by EPAS1 but not by HIF-1a (another member of the PAS family of transcription factors).
  • Expression plasmids (6 ⁇ g each) and KDR/flk-1 reporter pGL2-4kb+296 (1 ⁇ g) were cotransfected into the cell types indicated.
  • FIG. 2B is a bar graph showing transactivation of a VEGF promoter by EPAS1 and HIF-1 ⁇ .
  • Expression plasmids (6 ⁇ g each) and VEGF reporter pVR47/CAT (1 ⁇ g) were cotransfected into the cell types indicated.
  • the plasmid pCMV- GAL was cotransfected to correct for differences in transfection efficiency.
  • the “fold induction” represents the ratio (mean+SE) of normalized luciferase or CAT activity in cells transfected with expression plasmid to that in cells transfected with empty vector (pcDNA3).
  • FIG. 3A is a diagram showing an alignment of the amino acid sequence of human ARNT4 with human BMALlb and human ARNT.
  • FIG. 3B is a diagram of a phylogenetic tree of the ARNT family of proteins.
  • FIG. 4 is a bar graph showing the results of a yeast two-hybrid assay.
  • ARNT3 BMALlb
  • ARNT4 form heterodimers with EPAS1 as well as with CLOCK.
  • FIG. 5 is a bar graph showing that EPAS1 interacts with ARNT4 to form functional heterodimers which increase VEGF promoter activity and VEGF expression.
  • FIG. 6 is a bar graph showing that ARNT4 does not interact with HIF-1 ⁇ .
  • EPAS1 is a member of the transcription factor family characterized by a basic helix-loop-helix (bHLH) domain and a (Per-AhR-Arnt-Sim) PAS domain composed of two imperfect repeats.
  • Table 4 shows the amino acid sequence of human wild type EPAS1.
  • N-terminal bHLH domain which plays a role in DNA binding
  • C-terminal transactivation domain are highlighted (in bold and underlined type, respectively).
  • Table 5 shows the nucleotide sequence DNA encoding human wild type EPAS1. Nucleotides encoding the first amino acid of EPAS1 are underlined. TABLE 5 Nucleotide sequence of human EPAS1 cDNA 1 cctgactgcg cggggcgctc gggacctgcg cgcacctcgg accttcacca cccgccggg (SEQ ID NO:7) 61 ccgcggggag cggacgaggg ccacagccccccacccgcca gggagcccag gtgctcggcg 121 tctgaacgtc tcaaagggcc aca tg acagctga caaggagaag aaggagta 181 gctcggagag gaggaaggag aaaaggagta 181 gctc
  • Hypoxia inducible factor-1 ⁇ (HIF-1 ⁇ ) is another member of the PAS family to which EPAS1 belongs. Transcription factors of this family use the bHLH and PAS domains to form heterodimers that subsequently bind to target genes and regulate important biological processes.
  • EPAS1 plays a role in the regulation of angiogenic factors such as VEGF, VEGF-R such as KDR/flk-1 and flt-1, and Tie2.
  • VEGF vascular endothelial growth factor
  • VEGF-R vascular endothelial growth factor
  • Tie2 vascular endothelial growth factor
  • bHLH basic helix-loop-helix
  • EPAS1 transcription factor or DNA encoding all or part of EPAS1 is administered to individuals to promote angiogenesis.
  • EPAS1 antisense sequences are administered to cells to decrease intracellular production of EPAS1 gene product.
  • EPAS1-specific antibody e.g., EPAS1 intrabodies
  • EPAS1 dominant negative mutants can also be administered cells to inhibit EPAS1 function, e.g., by inhibiting binding of EPAS1 to cis-acting regulatory sequences of VEGF, VEGF-R, or Tie2 genes or by inhibiting EPAS1 transactivation of gene transcription.
  • EPAS1 is useful to modulate vasculogenesis and angiogenesis.
  • Anti-ARNT4 antibodies are obtained by techniques well known in the art. Such antibodies can be polyclonal or monoclonal. Polyclonal antibodies are obtained, for example, by the methods described in Ghose et al., Methods in Enzymology, Vol. 93, 326-327, 1983. For example, a ARNT4 polypeptide, or an antigenic fragment thereof, can be used as an immunogen to stimulate the production of ARNT4-reactive polyclonal antibodies in the antisera of animals such as rabbits, goats, sheep, or rodents. Antigenic polypeptides useful as immunogens include polypeptides which contain a bHLH domain/PAS domain.
  • Monoclonal antibodies are obtained by standard techniques such as those described by Milstein and Kohler in Nature, 256:495-97, 1975, or as modified by Gerhard, Monoclonal Antibodies, Plenum Press, 1980, pages 370-371.
  • Hybridomas are screened to identify those producing antibodies that are highly specific for an ARNT4 polypeptide.
  • the antibody will have an affinity of at least about 108 liters/mole and more preferably, an affinity of at least about 109 liters/mole.
  • ARNT4 knockout mice are generated by homologous recombination.
  • a gene targeting construct for generating ARNT4-deficient mice is made using a targeted gene deletion strategy using standard methods. The deletion in the ARNT4 gene renders the ARNT4 polypeptide non-functional.
  • the linearized targeting construct is transfected into murine D3 embryonic stem (ES) cells, and a clone with the correct homologous recombination (yielding the appropriately disrupted ARNT4 gene) is injected into blastocysts and used to generate ARNT4-deficient mice.
  • ES murine D3 embryonic stem
  • EPAS1 and KDR/flk-1 transcripts were found to colocalize in vascular endothelial cells in mouse embryonic and adult tissue.
  • a plasmid containing 4.0 kb of human KDR/flk-1 5′-flanking sequence linked to the luciferase reporter gene and a second vector containing DNA encoding either EPAS1 or another bHLH-PAS domain transcription factor HIF-1 ⁇ were cotransfected into bovine aortic endothelial cells (BAEC).
  • EPAS1 but not HIF-1 ⁇ markedly increased KDR/flk-1 promoter activity in a dose-dependent manner, and this induction of the KDR/flk-1 promoter by EPAS1 occurred preferentially in endothelial cells.
  • both EPAS1 and HIF-1 ⁇ activated the VEGF promoter in a non-endothelial cell-specific manner. This is the first demonstration of transactivation of the KDR/flk-1 promoter by EPAS1.
  • EPAS1 plays an important role in regulating vasculogenesis and angiogenesis.
  • BAEC were isolated and cultured in DME supplemented with 10% FCS (HyClone, Logan, Utah) and antibiotics according to known procedures. BAEC were passed every 3-5 days, and cells from passages 5-7 were used for the transfection experiments. The following cell lines were obtained from the American Type Culture Collection (ATCC) and were cultured in the same medium as BAEC: HeLa cells (human epidermoid carcinoma cells; ATCC #CRL7396 and NIH 3T3 cells (mouse fibroblasts; ATCC #CRL1888).
  • ATCC American Type Culture Collection
  • Total RNA (10 ⁇ g) was fractionated on a 1.3% formaldehyde-agarose gel and transferred to Nitropure filters (MSI, Westborough, Mass.). The filters were then hybridized with 32 P-labeled, randomly primed cDNA probes for 1 h at 68° C. in Quick-hyb solution (Stratagene, La Jolla, Calif.). The hybridized filters were washed in 30 mM NaCl, 3 mM sodium citrate, and 0.1% sodium dodecyl sulfate at 55° C.
  • the forward (5′ GAACTTGGATGCTCTTTGGAAA 3′; SEQ ID NO: 8 ) and reverse (5′ CACTTGCTGGCATCATAAGGC 3′; SEQ ID NO: 9 ) primers were used to generate PCR fragments that were subcloned into to a pCR 2.1 vector (Invitrogen, Carlsbad, Calif.). Nucleotide sequence authenticity was confirmed by the dideoxy chain termination method.
  • a 316 (771-1086) bp mouse EPAS1 CDNA and a 342 (2346-2687) bp mouse KDR/flk-1 cDNA from mouse lung total RNA was amplified by reverse transcriptase PCR with the following primers: EPAS1, forward 5° CATCATGTGTGAGCCAATCCA 3′ (SEQ ID NO: 10 ) and reverse 5′ GTTGTAGATGACCGTCCCCTG 3′ (SEQ ID NO: 11 ) KDR/flk-1, forward 5′ TGTACTGAGAGATGGGAACCG 3′ (SEQ ID NO: 12 ) and reverse 5′ CACTTGCTGGCATCATAAGGC 3′ (SEQ ID NO: 13 ). PCR fragments were subcloned into the pCR 2.1 vector in both orientations and the authenticity of the sequences was confirmed.
  • E9 mouse sections were purchased from Novagen (Madison, Wis.). E12 mice and various adult mouse organs were fixed in 4% paraformaldehyde, dehydrated, and embedded in paraffin. Tissue sections (6 ⁇ m thick) were hybridized with a 35 S-UTP-labeled antisense cRNA probe synthesized with the T7 RNA polymerase from linearized plasmids containing appropriate cDNA fragments using standard techniques. As a negative control, tissue sections were also hybridized with 35 S-UTP-labeled sense cRNA probes.
  • tissue sections were washed, and the dried tissue sections were then dipped into Kodak NTB2 emulsion (Eastman Kodak) and exposed for 10-15 days at 4° C. The sections were counterstained with hematoxylin and eosin.
  • pGL2-Basic and pGL2-Control contained the firefly luciferase reporter gene (Promega, Madison, Wis.).
  • pGL2-Basic had no promoter, whereas pGL2-Control contained the SV40 promoter and enhancer.
  • the pGL2-4kb+296 reporter plasmid was constructed by inserting the human KDR/flk-1 promoter from -4kb to +296 into pGL2-Basic.
  • pVR47/CAT which contains the human VEGF promoter from ⁇ 2362 to +61 and the chloramphenicol acetyltransferase (CAT) reporter gene sequence, was also constructed using standard techniques.
  • the plasmid phEP-lAS was made by cloning the antisense EPAS1 cDNA into pcDNA3.
  • phEP-1 ⁇ CT containing a C-terminal deletion mutant of the EPAS1 cDNA, was generated by subcloning a BamHI-XhoI restriction fragment encoding human EPAS1 amino acids 1 - 690 into pcDNA3.
  • phHIF-1 ⁇ a 2622 bp cDNA fragment containing the entire open reading frame of human HIF-1a was amplified using human leukocyte total RNA and pfu DNA polymerase (Stratagene, La Jolla, Calif.).
  • sequences of the forward (5′ GTGAAGACATCGCGGGGACC 3′; SEQ ID NO: 14 ) and reverse (5′ GTTTGTGCAGTATTGTAGCCAGG 3′; SEQ ID NO: 15 ) primers were based on human HIF-1a cDNA (Wang et al., 1995, Proc. Natl. Acad. Sci. USA. 92:5510-5514).
  • the PCR fragment was then cloned into pcDNA3, and the sequence was confirmed.
  • Cells were transfected with 1 ⁇ g of reporter construct and 6 ⁇ g of expression construct by the standard calcium phosphate method. To correct for variability in transfection efficiency against ⁇ -galactosidase, 1 ⁇ g of pCMV- ⁇ GAL was cotransfected in all experiments. Cell extracts were prepared 48 h after transfection by a detergent lysis method (Promega, Madison, Wis.). Luciferase activity was measured in duplicate for all samples with an EC&G Autolumat 953 (Gaithersburg, Md.) luminometer by the Promega luciferase assay. CAT activity was assayed by a two-phase fluor diffusion method.
  • ⁇ -galactosidase activity was assayed using standard methods. The ratio of luciferase or CAT activity to ⁇ -galactosidase activity in each sample served as a measure of normalized luciferase or CAT activity. Each construct was transfected at least four times, and each transfection was done in triplicate. Data for each construct are presented as the mean ⁇ SE.
  • Tissue distribution of EPAS1 and KDR/flk-1 in adult mice Northern blot analysis was performed with RNA prepared from various adult mouse tissues. EPAS1 mRNA was abundant in the lung, heart, and aorta, organs known to be rich in vascular endothelial cells. When the same blot was hybridized to a mouse KDR/flk-1 probe, the expression pattern of KDR/flk-1 was identical to that of EPAS1. In situ hybridization was performed using an antisense mouse EPAS1 probe to determine which cells in the aorta expressed EPAS1. The EPAS1 message localized to the luminal layer, and the antisense EPAS1 probe but not the sense EPAS1 probe hybridized to the endothelial cells of the aorta.
  • EPAS1 and KDR/flk-1 were expressed in endothelial cells of the aorta and other organs.
  • E12.5 EPAS1 mRNA was visible in the intervertebral blood vessels, heart, vascular plexuses in the meninges surrounding both the spinal cord and the brain, and choroid plexus.
  • the distribution of KDR/flk-1 MRNA at E12.5 was strikingly similar.
  • the EPAS1 and KDR/flk-1 mRNAs were both detected in endothelial cells of the blood vessels at higher magnification as well.
  • plasmid phEP-lACT was constructed to express a truncated form of EPAS1 lacking its 180 C-terminal amino acids. Deletion of the 180 C-terminal amino acids of EPAS1 completely abolished its ability to transactivate the KDR/flk-1 promoter (FIG. 1B). These data indicate that the 180 C-terminal amino acids of EPAS1 are necessary for transactivation of the KDR/flk-1 promoter.
  • the target gene for EPAS1 in adults is a VEGF-R such as KDR/flk-1 or flt-1 (as well as VEGF) as evidenced by the data showing that EPAS1 markedly induces KDR/flk-1 promoter activity.
  • VEGF-R such as KDR/flk-1 or flt-1 (as well as VEGF)
  • the EPAS1 or HIF-1 ⁇ expression plasmid and the KDR/flk-1 plasmid pGL2-4kb+296 were cotransfected into BAEC, HeLa cells, and NIH 3T3 cells.
  • EPAS1 expression plasmids in the sense (phEP-1) but not the antisense (phEP-1AS) orientation activated the KDR/flk-1 promoter (FIG. 2A), indicating that the transactivating effect is cell-specific.
  • the EPAS1 plasmid markedly increased KDR/flk-1 promoter activity in vascular endothelial cells, it had little effect on KDR/flk-1 promoter activity in HeLa or NIH 3T3 cells (FIG. 2A).
  • HIF-1 ⁇ had no effect on KDR/flk-1 promoter activity in all three cell types.
  • the EPAS1 or HIF-1 ⁇ expression plasmid was then cotransfected with a reporter plasmid containing the VEGF promoter, pVR47/CAT, to determine whether the differential effects of EPAS1 and HIF-1a were unique to the KDR/flk-1 promoter. In contrast to its cell-specific effect on the KDR/flk-1 promoter (FIG.
  • EPAS1 transactivated the VEGF promoter in all three cell types (FIG. 2B). Induction was highest in HeLa cells. Furthermore, HIF-1a increased VEGF promoter activity in BAEC and HeLa cells (FIG. 2B). These data indicate that the transactivating effect of EPAS1 depends on both the promoter and the cell type.
  • EPAS1 transactivated the KDR/flk promoter preferentially in endothelial cells (FIG. 2A), it activated the VEGF promoter in a non-endothelial cell-specific manner (FIG. 2B).
  • HIF-1 ⁇ is 48% homologous to EPAS1, HIF-1 ⁇ had no effect on the KDR/flk-1 promoter.
  • HIF-1 ⁇ transactivated the VEGF promoter.
  • the effect of EPAS1 on the KDR/flk-1 promoter is specific and cannot be replaced by other members of the PAS family of transcription factors.
  • EPAS1 heterodimerizes with the aryl hydrocarbon receptor nuclear translocator and transactivates the promoter of Tie2.
  • EPAS1 also markedly increases the promoter activity of KDR/flk-1 and VEGF. Mice deficient in the aryl hydrocarbon receptor nuclear translocator are not viable past E10.5, and the yolk sac shows defective angiogenesis.
  • EPAS1 Functional domains of EPAS1 were identified as follows.
  • the gene encoding VEGF has a cis-acting regulatory sequence to which EPAS1 binds (GCCCTACGTGCTGTCTCA; SEQ ID NO: 1 ) in its 5′ flanking region.
  • GCCCTACGTGCTGTCTCA cis-acting regulatory sequence to which EPAS1 binds
  • SEQ ID NO: 1 cis-acting regulatory sequence to which EPAS1 binds
  • the EPAS1 expression plasmid activated by 30-fold a CAT reporter plasmid containing 2.3 kb of VEGF 5′ flanking sequence (containing SEQ ID NO: 1 ) but not a similar plasmid differing only by a mutation in an amino acid of SEQ ID NO: 1 .
  • EPAS1 activates the VEGF promoter by binding to DNA containing the sequence of SEQ ID NO: 1 .
  • BAEC were cotransfected with expression plasmids encoding EPAS1 mutants and the reporter plasmid. Eight mutants were tested.
  • transactivation domain of EPAS1 spans amino acids 486 - 639 .
  • An EPAS1 mutant polypeptide lacking the amino acid sequence of SEQ ID NO: 2 e.g., an EPAS1 with the amino acid sequence of SEQ ID NO: 4 , functions as a dominant negative mutant EPAS1 because it inhibited transactivation of the VEGF promoter by wild type EPAS1 in a dose-dependent manner.
  • Deletion analysis is also used to identify domains of EPAS1 which participate in heterodimer formation with ARNT4.
  • This assay is also used to identify compounds which inhibit or decrease formation of functional ARNT4/EPAS1 heterodimers, and thus, inhibit angiogenesis.
  • expression plasmids which encode wild type or functional fragments of ARNT4 and EPAS1 are cotransfected with a VEGF/luciferase reporter plasmid into an endothelial cell in the presence and absence of a candidate compound.
  • a decrease in the amount of transactivation of the VEGF promoter (e.g., as measured by a standard luciferase assay) in the presence of the compound compared to the amount in the absence of the candidate compound indicates that the compound inhibits angiogenesis (by inhibiting ARNT4/EPAS1 transactivation of the VEGF promoter).
  • EPAS1 An adenoviral construct which expresses EPAS1 was generated. Overexpression of EPAS1 dramatically induced VEGF mRNA in human umbilical endothelial cells. In cotransfection experiments, EPAS1 transactivated the VEGF promoter via the HIF-1 binding site. This transactivation was further enhanced by hypoxia. Cotransfection of an aryl hydrocarbon receptor nuclear translocator (ARNT) expression plasmid and EPAS1 expression plasmid synergistically transactivated the VEGF promoter, indicating that heterodimerization of EPAS1 and ARNT is crucial for the transactivation of the VEGF promoter (FIG. 5). Using a gel shift analysis, EPAS1 (but not HIF-1) formed dimers with ARNT4 and bound to the HIF-1 binding site of the VEGF promoter.
  • RNT aryl hydrocarbon receptor nuclear translocator
  • EPAS1 Deletion analysis of EPAS1 further defined a potent transactivation domain to span amino acids 486 - 639 of human EPAS1 (SEQ ID NO: 6 ).
  • the transactivation domain is essential for EPAS1 to transactivate the VEGF promoter. The ability of this domain to activate transcription was confirmed using the GAL4 fusion protein system.
  • a truncated EPAS1 lacking the transactivation domain e.g., an EPAS1 polypeptide lacking amino acids 486 - 690 of SEQ ID NO: 6 or an EPASI polypeptide lacking amino acids 486 - 639 of SEQ ID NO: 6 ) retained its ability to form heterodimers and to bind the HIF-1 binding site.
  • mutated EPAS1 polypeptides with lack amino acids in the transactivation domain are dominant negative mutants because they sequester ARNT and prevent the formation of functional EPAS1/ARNT and HIF-1 ⁇ /ARNT heterodimers.
  • the EPAS1 polypeptide which lacked amino acids 486 - 639 of SEQ ID NO: 6 potently inhibited the induction of the VEGF promoter by EPAS1 and HIF-1 ⁇ .
  • Transfection of endothelial cells with an adenovirus construct encoding this mutant inhibited VEGF MRNA induction by hypoxia.
  • EPAS1 is an important regulator of VEGF gene expression and that dominant negative EPAS1 mutants (e.g., EPAS1 polypeptides lacking all or part of the transactivation domain (SEQ ID NO: 2 )) inhibit VEGF promoter activity, and in turn, VEGF expression and angiogenesis.
  • dominant negative EPAS1 mutants e.g., EPAS1 polypeptides lacking all or part of the transactivation domain (SEQ ID NO: 2 )
  • SEQ ID NO: 2 the transactivation domain
  • Modulation of the angiogenesis is achieved by contacting the vascular cells such as vascular endothelial cells with a compound that blocks or enhances EPAS1 binding to cis-acting regulatory sequences of VEGF, VEGF-Rs, or other angiogenic factors in endothelial cells such as Tie2.
  • a compound that blocks or enhances EPAS1 binding to cis-acting regulatory sequences of VEGF, VEGF-Rs, or other angiogenic factors in endothelial cells such as Tie2.
  • a compound that blocks or enhances EPAS1 binding to cis-acting regulatory sequences of VEGF, VEGF-Rs, or other angiogenic factors in endothelial cells such as Tie2.
  • a compound can be identified by methods ranging from rational drug design to screening of random compounds. The latter method is preferable, as simple and rapid assays for testing such compounds are available. oligonucleotides and small organic molecules are desirable candidate compounds for this
  • the screening of compounds for the ability to modulate angiogenesis by affecting EPAS1 transactivation of transcription of angiogenic factors may be carried out using in vitro biochemical assays, cell culture assays, or animal model systems.
  • labeled EPAS1 e.g., EPAS1 labeled with a fluorochrome or a radioisotope
  • ARNT4 is immobilized on the column. In this manner, compounds which inhibit ARNT4/EPAS1 heterodimerization may be identified.
  • a candidate compound is applied to the column before, after, or simultaneously with the labeled EPAS1, and the amount of labeled protein bound to the column in the presence of the compound is determined by conventional methods.
  • a compound tests positive for inhibiting EPAS1 binding (thereby having the effect of inhibiting angiogenesis) if the amount of labeled protein bound in the presence of the compound is lower than the amount bound in its absence.
  • a compound tests positive for enhancing EPAS1 binding (thereby having the effect of enhancing angiogenesis) if the amount of labeled protein bound in the presence of the compound is greater than the amount bound in its absence.
  • binding of labeled DNA to immobilized EPAS1 is measured.
  • candidate compounds may also be screened using cell culture assays.
  • Cells expressing EPAS1, either naturally or after introduction into the cells of genes encoding EPAS1 are cultured in the presence of the candidate compound.
  • the level of EPAS1 binding in the cell may be inferred using any of several assays.
  • levels of expression of EPAS1 regulated genes e.g., genes encoding VEGF, VEGF-Rs such as KDR/flk-1 or flt-1
  • levels of expression of EPAS1 regulated genes e.g., genes encoding VEGF, VEGF-Rs such as KDR/flk-1 or flt-1
  • Compounds identified as having the desired effect, either enhancing or inhibiting EPAS1 binding can be tested further in appropriate animal models, e.g., an animal with a tumor or atherosclerotic lesion.
  • Compounds found to inhibit EPAS1 binding to cis-acting regulatory sequences of geres encoding angiogenic factors may be used in methods for nhibiting pathogenic angiogenesis in order to, e.g., prevent or treat tumor progression or the progression of an atherosclerotic lesion.
  • Compounds found to enhance EPAS1 binding may be used in methods to therapeutically promote new blood vessel formation in adult mammals as discussed above.
  • the therapeutic compounds identified using the methods of the invention may be administered to a patient by any appropriate method for the particular compound, e.g., orally, intravenously, parenterally, transdermally, transmucosally, by inhalation, or by surgery or implantation at or near the site where the effect of the compound is desired (e.g., with the compound being in the form of a solid or semi-solid biologically compatible and resorbable matrix).
  • Therapeutic doses are determined specifically for each compound, most being administered within the range of 0.001 to 100.0 mg/kg body weight, or within a range that is clinically determined to be appropriate by one skilled in the art.
  • compositions which interact with EPAS1 were identified by screening for endothelial cell proteins which bind to EPAS1.
  • Yeast two hybrid screening of a human umbilical endothelial cell cDNA library was carried out using EPAS1 as a bait.
  • One of the clones isolated encoded a novel bHLH/PAS protein which was found to have similarity with arylhydrocarbon nuclear translocator 3 (Arnt3), a member of bHLH/PAS protein which heterodimerizes with Clock, a gene product involved in regulation of mammalian circadian rhythm.
  • the isolated clone was named ARNT4.
  • the amino acid sequences of hARNT and hARNT4 were found to be 23% identical; the sequences of hBMAL lb and hARNT4 were found to be 49% identical; and the sequences of hARNT and hBMAL lb were found to be 26% identical.
  • ARNT4 was shown to interact with EPASI using the yeast two-hybrid assay (FIG. 4). In a gel mobility shift assay using hypoxia responsive element of VEGF gene as the probe, ARNT4 formed a heterodimer with EPAS1 and bound to the hypoxia responsive element of the VEGF gene.
  • An expression plasmid encoding EPAS1 and an expression plasmid encoding ARNT4 were cotransfected with a VEGF/luciferase reporter plasmid into bovine aortic endothelial cells. Coexpression of ARNT4 and EPAS1 markedly transactivated the VEGF promoter (FIG. 5), and this transactivation was further enhanced by hypoxia. These data indicate that the heterodimer EPAS1/ARNT4 is activated under hypoxic conditions. Taken together, these results indicate that ARNT4, a novel bHLH/PAS protein, is an important regulator of VEGF gene expression especially in vascular system.
  • ARNT4 coding sequence in Table 7 is indicated by upper case letters (nucieotides 220 to 2025 ) with-the termination codon underlined.
  • ARNT4 is involved in regulating circadian rhythm, e.g., by forming a heterodimer with Clock, a protein that regulates the timing of fatigue and alertness.
  • Individuals with circadian rhythm disorders are screened for mutations in the an ARNT4 gene product or ARNT4 gene, e.g,. by detecting restriction fragment length polymorphisms (RFLPs) or by PCR.
  • RFLPs restriction fragment length polymorphisms
  • Individuals with symptoms of circadian rhythm disorders and identified as having a mutated ARNT4 gene are treated by administering DNA encoding a normal ARNT4 gene product. For example, DNA containing the coding sequence of SEQ ID NO: 20 is administered to such individuals using standard gene therapy techniques described herein.
  • ARNT4 protein or transcript is detected in an individual suffering from such disorders, the levels can be normalized by antisense therapy to inhibit ARNT4 production or gene therapy to augment production.
  • ARNT4 levels may also be altered to artificially regulate circadian rhythm, e.g., to induce long periods of sleep in patients to improve the healing process or in individuals travelling long distances such astronauts during space travel.
  • Nucleic acids complementary to all or part of the human EPAS1 CDNA may be used in methods for antisense treatment to inhibit expression of EPAS1.
  • Nucleic acids complementary to all or part of the human ARNT4 cDNA (SEQ ID NO: 20 ) may be used in methods for antisense treatment to inhibit expression of ARNT4.
  • Antisense treatment may be carried out by administering to a mammal, such as a human, DNA containing a promoter, e.g., an endothelial cell-specific promoter, operably linked to a DNA sequence (an antisense template), which is transcribed into an antisense RNA.
  • antisense oligonucleotides may be introduced directly into vascular cells.
  • the antisense oligonucleotide may be a short nucleotide sequence (generally at least 10, preferably at least 14, more preferably at least 20 (e.g., at least 30), and up to 100 or more nucleotides) formulated to be complementary to a portion, e.g., the coding sequence, or all of EPAS1 MRNA or ARNT4 mRNA.
  • the sequence is complementary some or all of the C-terminal activation domain; alternatively, the sequence may be complementary to all or part of the N-terminal DNA binding domain.
  • the antisense sequence is complementary to DNA encoding residues 75 to 128 , inclusive, of SEQ ID NO: 19 ; the antisense sequence.
  • the antisense sequence is complementary to DNA encoding residues 155 to 207 , inclusive, of SEQ ID NO: 19 , or encoding residues 232 to 384 of SEQ ID NO: 19 .
  • the antisense RNA binds to its target nucleic acid molecule, such as an MRNA molecule, thereby inhibiting expression of the target nucleic acid molecule.
  • target nucleic acid molecule such as an MRNA molecule
  • an antisense sequence complementary to a portion or all of EPAS1 MRNA could be used to inhibit the expression of EPAS1, thereby decreasing the level of transcription of angiogenic factors such as VEGF or VEGF-Rs, which in turn leads to a decrease in new blood vessel formation.
  • Oligonucleotides complementary to various portions of EPAS1 mRNA or ARNT4 MRNA can readily be tested in in vitro for their ability to decrease production of their respective gene products, using assays similar to those described herein. Sequences which decrease production of EPAS1 message or ARNT4 message in vitro cell-based or cell-free assays can then be tested in vivo in rats or mice to determine whether blood vessel formation is decreased.
  • Preferred vectors for antisense templates are viral vectors, including those derived from replication-defective hepatitis viruses (e.g., HBV and HCV), retroviruses (see, e.g., WO 89/07136; Rosenberg et al., 1990, N. Eng. J. Med. 323(9):570-578), adenovirus (see, e.g., Morsey et al., 1993, J. Cell. Biochem., Supp. 17E,), adeno-associated virus (Kotin et al., 1990, Proc. Natl. Acad. Sci.
  • viral vectors including those derived from replication-defective hepatitis viruses (e.g., HBV and HCV), retroviruses (see, e.g., WO 89/07136; Rosenberg et al., 1990, N. Eng. J. Med. 323(9):570-578), adenovirus (see, e
  • the invention may utilize any other delivery system which accomplishes in vivo transfer of nucleic acids into eucaryotic cells.
  • the nucleic acids may be packaged into liposomes, receptor-mediated delivery systems, non-viral nucleic acid-based vectors, erythrocyte ghosts, or microspheres (e.g., microparticles; see, e.g., U.S. Pat. No. 4,789,734; U.S.
  • nucleic acids may be administered. Delivery of nucleic acids to a specific site in the body for antisense therapy may also be accomplished using a biolistic delivery system, such as that described by Williams et al., 1991, Proc. Natl. Acad. Sci. USA 88:2726-2729.
  • Antisense oligonucleotides may consist of DNA, RNA, or any modifications or combinations thereof.
  • modifications that the oligonucleotides may contain, inter-nucleotide linkages other than phosphodiester bonds, such as phosphorothioate, methylphosphonate, methylphosphodiester, phosphorodithioate, phosphoramidate, phosphotriester, or phosphate ester linkages (Uhlman et al., 1990, Chem. Rev. 90(4) :544-584; Anticancer Research, 1990, 10:1169) may be present in the oligonucleotides, resulting in their increased stability.
  • oligonucleotide stability may also be increased by incorporating 3′-deoxythymidine or 2′-substituted nucleotides (substituted with, e.g., alkyl groups) into the oligonucleotides during synthesis, by providing the oligonucleotides as phenylisourea derivatives, or by having other molecules, such as aminoacridine or poly-lysine, linked to the 3′ ends of the oligonucleotides e.g., Anticancer Research, 1990, 10:1169-1182).
  • RNA and/or DNA nucleotides may be present throughout the oligonucleotide, or in selected regions of the oligonucleotide, e.g., in the 5′ and/or 3′ ends.
  • the antisense oligonucleotides may also be modified so as to increase their ability to penetrate the target tissue by, e.g., coupling the oligonucleotides to lipophilic compounds.
  • Antisense oligonucleotides based on the human EPAS1 nucleotide sequence (SEQ ID NO: 7 ) or the human ARNT4 nucleotide sequence (SEQ ID NO: 20 ) can be made by any method known in the art, including standard chemical synthesis, ligation of constituent oligonucleotides, and transcription of DNA complementary to the all or part of the EPASI CDNA or ARNT4 CDNA.
  • EPAS1 is naturally expressed in vascular endothelial cells. These cells are, therefore, the preferred cellular targets for antisense therapy. Targeting of antisense oligonucleotides to endothelial cells is not critical to the invention, but may be desirable in some instances, e.g. systemic administration of antisense compositions. Targeting may be achieved, for example, by coupling the oligonucleotides to ligands of endothelial cell surface receptors. Similarly, oligonucleotides may be targeted to endothelial cells by being conjugated to monoclonal antibodies that specifically bind to endothelial-specific cell surface proteins. Antisense compositions may also be administered locally, e.g., at the site of an atherosclerotic lesion or at the site of a tumor.
  • Antisense nucleic acids may be used alone or combined with one or more materials, including other antisense oligonucleotides or recombinant vectors, materials that increase the biological stability of the oligonucleotides or the recombinant vectors, or materials that increase the ability of the therapeutic compositions to penetrate endothelial cells selectively.
  • compositions e.g., inhibitors of EPAS1 and/or ARNT4 transcription or transactivating function
  • pharmaceutically acceptable carriers e.g., physiological saline
  • Suitable pharmaceutical carriers, as well as pharmaceutical necessities for use in pharmaceutical formulations, are described in Remington's Pharmaceutical Sciences, a standard reference text in this field, and in the USP/NF.
  • the compound may be administered with intravenous fluids as well as in combination with other anti-inflammatory agents, e.g., antibiotics; glucocorticoids, such as dexamethasone (Dex), or other chemotherapeutic drugs for the treatment of atherosclerotic lesions and tumors, respectively.
  • antibiotics e.g., antibiotics
  • glucocorticoids such as dexamethasone (Dex)
  • Dex dexamethasone
  • a therapeutically effective amount is an amount which is capable of producing a medically desirable result in a treated animal.
  • dosage for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently. Dosages will vary, but a preferred dosage for intravenous administration of DNA is approximately 10 6 to 10 22 copies of the DNA molecule.
  • the compositions of the invention may be administered locally or systemically. Administration will generally be parenterally, e.g., intravenously.
  • DNA may also be administered directly to the target site, e.g., by biolistic delivery to an internal or external target site or by catheter to a site in an artery.
  • compositions which enhance intracellular production of EPAS1 may be used in methods to promote new blood vessel formation, e.g., to promote angiogenesis in wound healing (e.g., healing of broken bones, burns, diabetic ulcers, or traumatic or surgical wounds) and organ transplantation.
  • wound healing e.g., healing of broken bones, burns, diabetic ulcers, or traumatic or surgical wounds
  • Such compounds may be used to treat peripheral vascular disease, cerebral vascular disease, hypoxic tissue damage (e.g., hypoxic damage to heart tissue), or coronary vascular disease as well as to treat patients who have, or have had, transient ischemic attacks, vascular graft surgery, balloon angioplasty, frostbite, gangrene, or poor circulation.
  • hypoxic tissue damage e.g., hypoxic damage to heart tissue
  • coronary vascular disease e.g., coronary vascular disease
  • EPAS1 and ARNT4 are nuclear proteins
  • a preferred method of increasing the levels of these proteins or polypeptides in a cell is intracellular expression of recombinant EPAS1 or ARNT4 or active fragments thereof, e.g., transactivating fragments.
  • DNA encoding EPAS1 or ARNT4 is administered alone or as part of an expression vector as described above.
  • gene therapy are also used to introduce administer DNA encoding a dominant negative mutant of EPAS1 such as DNA encoding a polypeptide with the amino acid sequence of SEQ ID NO: 4 or a polypeptide with the amino acid sequence of residues 486-639 of SEQ ID NO: 6 .
  • Anti-EPAS1 antibodies were obtained using techniques well known in the art. Such antibodies can be polyclonal or monoclonal. Polyclonal antibodies can be obtained, for example, by the methods described in Ghose et al., Methods in Enzymology, Vol. 93, 326-327, 1983. An EPAS1 polypeptide, or an antigenic fragment thereof, was used as the immunogen to stimulate the production of EPAS1-reactive polyclonal antibodies in the antisera of animals such as rabbits, goats, sheep, rodents and the like.
  • EPAS1-specific antibodies were raised by immunizing animals with a C-terminal EPAS1 polypeptide spanning amino acids 668 - 829 of human EPAS (PGGSTSHLMWKRMKNLRGGSCPLMPDKPLSANVPNDKFTQNPMRGL HPLRHLPLPQPPSAISPGENSKSRFPPQCYATQYQDYSLSSAHKVSGMASRLLGP; (SEQ ID NO: 17 ) and a C-terminal EPAS polypeptide spanning amino acids 641 - 875 of mouse EPAS1 DPPLHFGPTKWPVGDQSAE SLGALPVGSWQLELPSAPLHVSMFKMRSAKDFGARGPYMMSPAMIALSNK LKLKRQLEYEEQAFQDTSGGDPPGTSSSHLMWKRMKSLMGGTCPLMPDKT ISANMAPDEFTQKSMRGLGQPLRHLPPPQPPSTRSSGENAKTGFPPQCYA SQFQDYGPPGAQKVSGVASRLLGPSFEPYLLPEL
  • Monoclonal antibodies are obtained by the process described by Milstein and Kohler in Nature, 256:495-97, 1975, or as modified by Gerhard, Monoclonal Antibodies, Plenum Press, 1980, pages 370-371. Hybridomas are screened to identify those producing antibodies that are highly specific for an EPAS1 polypeptide. Preferably, the antibody will have an affinity of at least about 10 8 liters/mole and more preferably, an affinity of at least about 10 9 liters/mole. Monoclonal antibodies can be humanized by methods known in the art, e.g, MAbs with a desired binding specificity can be commercially humanized (Scotgene, Scotland; Oxford Molecular, Palo Alto, Calif.).
  • DNA encoding the antibody is cloned.
  • DNA encoding a single chain EPAS1-specific antibody in which heavy and light chain variable domains (separated by a flexible linker peptide such as Gly 4 -Ser 3 (SEQ ID NO: 16 ) is cloned into an expression vector using known methods (e.g., Marasco et al., 1993, Proc. Natl. Acad. Sci. USA 90:7889-7893 and Marasco et al., 1997, Gene Therapy 4:11-15)
  • Such constructs are introduced into cells, e.g., using gene therapy techniques described herein, for intracellular production of the antibodies.
  • Intracellular antibodies i.e., intrabodies
  • Intracellular antibodies are used to inhibit binding of endogenous EPAS1 to its target DNA (e.g., cis-acting regulatory sequences of genes encoding VEGF or VEGF-Rs), which in turn, decreases production of these angiogenic factors and decreases new blood vessel formation in the treated mammal.
  • Intrabodies which bind to a C-terminal transactivation domain of EPAS1 inhibit the ability of EPAS1 to induce transcription of a gene encoding an angiogenic factor such as VEGF or a VEGF-R.
  • a similar strategy is used to make intrabodies which bind to intracellular ARNT4.
  • Such intrabodies bind to ARNT4 and prevent heterodimeriation with EPAS1, and as a result, inhibit transactivation of the VEGF promoter. Inhibition of VEGF promoter activity, in turn, leads to inhibition of new blood vessel formation.

Abstract

A method of inhibiting angiogenesis in a mammal by administering to the mammal a compound which inhibits binding of endothelial PAS domain protein-1 to cis-acting transcription regulatory sequence in the promoter region of a gene encoding an angiogenic factor.

Description

    BACKGROUND OF THE INVENTION
  • This invention relates to vascular therapy. [0001]
  • Angiogenesis results from endothelial cell proliferation induced by angiogenic factors. Angiogenic factors bind to receptors on endothelial cells which line blood vessels. This event triggers signals which cause the cells to proliferate; the proliferating endothelial cells secrete proteases which digest the basement membrane surrounding a vessel. The junctions between the endothelial cells are altered, allowing projections from the cells to pass through the space created. These outgrowths then become new blood vessels, e.g., capillaries. [0002]
  • Vascular endothelial cell growth factor (VEGF) and VEGF receptors (VEGF-Rs) play a role in vasculogenesis and angiogenesis. Although VEGF is secreted by a variety of cell types, including vascular smooth muscle cells, osteoblasts, fibroblasts, and macrophages, its proliferative and chemotactic activities are restricted to endothelial cells. VEGF signaling is mediated by two VEGF-Rs, the endothelial cell-specific tyrosine kinase receptors, flt-1 and KDR/flk-1. Despite its importance in VEGF signaling, the molecular mechanisms of VEGF and VEGF-R expression have not been elucidated. [0003]
  • SUMMARY OF THE INVENTION
  • The invention is based on the discovery that endothelial PAS domain protein-1 (EPAS1) binds to cis-acting regulatory sequences associated with genes encoding such angiogenic factors as VEGF and VEGF-Rs such as KDR/flk-1 and flt-1, thereby transactivating the promoters of such genes. Accordingly, the invention features a method of increasing the level of EPAS1 in a cell, e.g., an endothelial cell. An increase in the level of EPAS1 leads to increased promoter transactivation and increased transcription of genes encoding angiogenic factors which participate in the blood vessel formation. [0004]
  • The invention also includes a novel basic helix-loop-helix/Per-AhR-Arnt-Sim (bHLH/PAS) protein which binds to EPAS1 and forms a heterodimer which transactivates transcription of genes encoding angiogenic factors. Increasing the level of ARNT4 in a cell, e.g., an endothelial cell also leads to increased promoter transactivation and increased expression of angiogenic factors which participate in the blood vessel formation. [0005]
  • Angiogenic factors are proteins or polypeptides and ligands thereof that participate in the process of new blood vessel formation. For example, angiogenic factors include VEGF, VEGF-Rs, and other signalling proteins such as intracellular tyrosine kinases which participate in the angiogenic process. Preferably, the angiogenic factors are expressed in endothelial cells, e.g., VEGF, VEGF-Rs such as KDR/flk-1 or flt-1, and tyrosine kinases such as Tie2. [0006]
  • A method of inhibiting angiogenesis in a mammal is carried out by administering to the mammal a compound which inhibits binding of EPAS1 to cis-acting transcription regulatory DNA associated with a gene encoding an angiogenic factor. Angiogenesis is also inhibited by administering a compound which inhibits binding of EPAS1 to ARNT4, i.e., a compound which inhibits the formation of a functional heterodimer that can transactivate a promoter of gene encoding an angiogenic factor. The angiogenic factor is preferably VEGF., a VEGF-R such as KDR/flk-1 or flt-1. For example, the compound inhibits transcription of the angiogenic factor by binding to a cis-acting regulatory sequence such as the [0007] sequence 5′GCCCTACGTGCTGTCTCA 3′ (SEQ ID NO:1) in VEGF promoter DNA. For example, the compound is an EPAS1 polypeptide that binds to a cis-acting regulatory sequence of a gene but fails to transactivate the promoter of the gene, e.g. a polypeptide lacking a transactivation domain (amino acids 486-690 of EPAS1).
    TABLE 1
    Transactivation domain of human EPAS1
    EDYYTSLDNDLKIEVIEKLFAMDTEAKDQCSTQTDFNELDLETLAPYIPMDGEDFQLSPI (SEQ ID NO:2)
    CPEERLLAENPQSTPQHCFSAMTNIFQPLAPVAPHSPFLLDKFQQQLESKKTEPEHRPMS
    SIFFDAGSKASLPPCCGQASTPLSSMGGRSNTQWPPDPPLHFGPTKWAVGDQRTEFLGAA
    PLGPPVSPPHVSTFKTRSAKGFGAR
  • When such an EPAS1 mutant is bound to a cis-acting regulatory DNA, it prevents wild type EPAS1 binding and thereby inhibits transcription of a gene encoding an angiogenic factor (and, in turn, angiogenesis). For example, the EPAS1 polypeptide contains the N-terminal binding domain (amino acids [0008] 14-67 of EPAS1; RRKEKSRDAARCRRSKETEVFYELAHELPLPHSVSSHLDKASIMRLEISFLRTH; SEQ ID NO: 3), more preferably the EPAS polypeptide contains amino acids 1-485 of human EPAS1. The amino acid sequence of such an EPAS1 dominant negative mutant polypeptide and DNA encoding such a mutant polypeptide is provided below.
    TABLE 2
    EPAS1 dominant negative mutant
    1 MTADKEKKRS SSERRKEKSR DAARCRRSKE TEVFYELAHE LPLPHSVSSH
    51 LDKASIMRLE ISFLRTHKLL SSVCSENESE AEADQQMDNL YLKALEGFIA
    101 VVTQDGDMIF LSENISKFMG LTQVELTGHS IFDFTHPCDH EEIRENLSLK
    151 NGSGFGKKSK DMSTERDFFM RMKCTVTNRG RTVNLKSATW KVLHCTGQVK
    201 VYNNCPPHNS LCGYKEPLLS CLIIMCEPIQ HPSHMDIPLD SKTFLSRHSM
    251 DMKFTYCDDR ITELIGYHPE ELLGRSAYEF YHALDSENMT KSHQNLCTKG
    301 QVVSGQYRML AKHGGYVWLE TQGTVIYNPR NLQPQCIMCV NYVLSEIEKN
    351 DVVFSMDQTE SLFKPHLMAM NSIFDSSGKG AVSEKSNFLF TKLKEEPEEL
    401 AQLAPTPGDA IISLDFGNQN FEESSAYGKA ILPPSQPWAT ELRSHSTQSE
  • 451 AGSLPAFTVP QAAAPGSTTP SATSSSSSCS TPNSP (SEQ ID NO: [0009] 4)
    TABLE 3
    DNA encoding EPAS1 Dominant Negative Mutant
    cctgactgcgcggggcgctcgggacctgcgcgcacctcggaccttcaccacccgcccggg (SEQ ID NO:5)
    ccgcggggagcggacgagggccacagccccccacccgccagggagcccaggtgctcggcg
    tctgaacgtctcaaagggccacagcgacaatgacagctgacaaggagaagaaaaggagta
    gctcggagaggaggaaggagaagtcccgggatgctgcgcggtgccggcggagcaaggaga
    cggaggtgttctatgagctggcccatgagctgcctctgccccacagtgtgagctcccatc
    tggacaaggcctccatcatgcgactggaaatcagcttcctgcgaacacacaagctcctct
    cctcagtttgctctgaaaacgagtccgaagccgaagctgaccagcagatggacaacttgt
    acctgaaagccttggagggtttcattgccgtggtgacccaagatggcgacatgatctttc
    tgtcagaaaacatcagcaagttcatgggacttacacaggtggagctaacaggacatagta
    tctttgacttcactcatccctgcgaccatgaggagattcgtgagaacctgagtctcaaaa
    atggctctggttttgggaaaaaaagcaaagacatgtccacagagcgggacttcttcatga
    ggatgaagtgcacggtcaccaacagaggccgtactgtcaacctcaagtcagccacctgga
    aggtcttgcactgcacgggccaggtgaaagtctacaacaactgccctcctcacaatagtc
    tgtgtggctacaaggagcccctgctgtcctgcctcatcatcatgtgtgaaccaatccagc
    acccatcccacatggacatccccctggatagcaagaccttcctgagccgccacagcatgg
    acatgaagttcacctactgtgatgacagaatcacagaactgattggttaccaccctgagg
    agctgcttggccgctcagcctatgaattctaccatgcgctagactccgagaacatgacca
    agagtcaccagaacttgtgcaccaagggtcaggtagtaagtggccagtaccggatgctcg
    caaagcatgggggctacgtgtggctggagacccaggggacggtcatctacaaccctcgca
    acctgcagccccagtgcatcatgtgtgtcaactacgtcctgagtgagattgagaagaatg
    acgtggtgttctccatggaccagactgaatccctgttcaagccccacctgatggccatga
    acagcatctttgatagcagtggcaagggggctgtgtctgagaagagtaacttcctattca
    ccaagctaaaggaggagcccgaggagctggcccagctggctcccaccccaggagacgcca
    tcatctctctggatttcgggaatcagaacttcgaggagtcctcagcctatggcaaggcca
    tcctgcccccgagccagccatgggccacggagttgaggagccacagcacccagagcgagg
    ctgggagcctgcctgccttcaccgtgccccaggcagctgccccgggcagcaccaccccca
    gtgccaccagcagcagcagcagctgctccacgcccaatagcccttga
  • Rather than administering EPAS1 polypeptides or ARNT4 polypeptides, the method may be carried out by administering DNA encoding such polypeptides. For example, the compound is a nucleic acid encoding an EPAS1 polypeptide lacking amino acids [0010] 486-690 of EPAS1. For example, the nucleic acid encodes a dominant negative mutant of EPAS1 which contains amino acids 1-485 of wild type EPAS1, i.e., SEQ ID NO: 5.
  • For antisense therapy, the compound is a antisense nucleic acid molecule containing at least 10 nucleotides the sequence of which is complementary to an mRNA encoding all or part of a wild type EPAS1 polypeptide. Preferably, the compound, e.g., an antisense oligonucleotide or antisense RNA produced from an antisense template, inhibits EPAS1 expression. For example, the compound may inhibit EPAS1 expression by inhibiting translation of EPAS1 mRNA. For example, antisense therapy is carried out by administering a single stranded nucleic acid complementary at least a portion of EPASI MRNA to interfere with the translation of MRNA into protein, thus reducing the amount of functional EPAS1 produced in the cell. A reduction in the amount of functional transactivating EPAS1 reduces the level of transcription of angiogenic factors such as VEGF or VEGF-Rs, resulting in a decrease in new blood vessel formation. [0011]
  • Alternatively, the compound is an EPASI-specific intrabody, i.e., a recombinant single chain EPAS1-specific antibody that is expressed inside a target cell, e.g., a vascular endothelial cell. Such an intrabody binds to endogenous intracellular EPAS1 and prevents it from binding to its target cis-acting regulatory sequence in the promoter region of a gene encoding an angiogenic factor such as VEGF or a VEGF-R. An ARNT4-specific intrabody is also useful to inhibit angiogenesis. [0012]
  • Angiogenesis contributes to the progression of atherosclerotic lesions. Thus, compounds are administered to a site of an atherosclerotic lesion in a mammal to inhibit growth of a lesion. Compounds may also be locally administered to a tumor site to reduce blood vessel formation, thereby depriving a tumor of blood supply and inhibiting tumor growth. VEGF itself is a growth factor for some tumors; the methods described above directly inhibit VEGF expression, and thus, are particularly useful for treating such tumor types. [0013]
  • The invention also includes an antibody which binds to EPAS1. The antibody preferably binds to the C-terminal portion of EPAS1 (e.g., a polypeptide having the amino acid of SEQ ID NO: [0014] 17 or 18). The antibody is a polyclonal or monoclonal antibody which specifically binds to the EPAS1. Preferably, the antibody binds to an epitope within the C-terminal transactivation domain (SEQ ID NO: 2). The invention encompasses not only an intact monoclonal antibody, but also an immunologically-active antibody fragment, e.g., a Fab or (Fab)2 fragment; an engineered single chain Fv molecule; or a chimeric molecule, e.g., an antibody which contains the binding specificity of one antibody, e.g., of murine origin, and the remaining portions of another antibody, e.g., of human origin.
  • To promote angiogenesis in a mammal, a compound, e.g., DNA encoding EPASI or a functional fragment thereof, which increases expression of VEGF or a VEGF-R in an endothelial cell is administered to a mammal, e.g., an adult mammal which has been identified as being in need of therapy to promote angiogenesis such as a patient suffering from peripheral vascular disease. A functional fragment of EPAS1 is one which binds to DNA in the promoter region of a gene encoding an angiogenic factor. [0015]
  • The invention also features an EPAS-binding element, ARNT4 and a nucleic acid which encodes ARNT4. For example, the nucleic acid includes a sequence which encodes the amino acid sequence a naturally-occurring human ARNT4 (SEQ ID NO: [0016] 19). The DNA may encode a naturally-occurring mammalian ARNT4 polypeptide such as a human, rat, mouse, guinea pig, hamster, dog, cat, pig, cow, goat, sheep, horse, monkey, or ape ARNT4. Preferably, the DNA encodes a human ARNT4 polypeptide, e.g., a polypeptide which contains part or all of the amino acid sequence of SEQ ID NO: 19. The invention includes degenerate variants of the human cDNA (SEQ ID NO: 20) . The DNA contains a nucleotide sequence having at least 50% sequence identity to SEQ ID NO: 20. For example, the DNA contains a sequence which encodes a human ARNT4 polypeptide, such as the coding sequence of SEQ ID NO: 20 (nucleotides 220 to 2025 of SEQ ID NO: 20). The DNA contains a strand which hybridizes at high stringency to a strand of DNA having the sequence of SEQ ID NO: 20, or the complement thereof. The DNA has at least 50% sequence identity to SEQ ID NO: 20 and encodes a polypeptide having the biological activity of a ARNT4 polypeptide, e.g, the ability to bind to EPAS1 to form a heterodimer. Preferably, the DNA has at least 75% identity, more preferably 85% identity, more referably 90% identity, more preferably 95% identity, more preferably 99% identity, and most preferably 100% identity to the coding sequence of SEQ ID NO: 20.
  • Nucleotide and amino acid comparisons are carried out using the CLUSTAL W sequence alignment system with (Thompson et al., 1994, Nucleic Acids Research 22:4673-4680 or http://www.infobiogen.fr/docs/ClustalW/clustalw.html). Amino acid sequences were compared using CLUSTAL W with the PAM250 residue weight table. “Per cent sequence identity”, as that term is used herein, is determined using the CLUSTAL W sequence alignment system referenced above, with the parameters described herein. In the case of polypeptide sequences which are less than 100% identical to a reference sequence, the non-identical positions are preferably, but not necessarily, conservative substitutions for the reference sequence. Conservative substitutions typically include substitutions within the following groups: glycine and alanine; valine, isoleucine, and leucine; aspartic acid and glutamic acid; asparagine and glutamine; serine and threonine; lysine and arginine; and phenylalanine and tyrosine. [0017]
  • Hybridization is carried out using standard techniques, such as those described in Ausubel et al. (Current Protocols in Molecular Biology, John Wiley & Sons, 1989). “High stringency” refers to nucleic acid hybridization and wash conditions characterized by high temperature and low salt concentration: wash conditions of 65° C. at a salt concentration of 0.1×SSC. “Low” to “moderate” stringency denotes DNA hybridization and wash conditions characterized by low temperature and high salt concentration: wash conditions of less than 60° C. at a salt concentration of 1.0×SSC. For example, high stringency conditions include hybridization at 42° C., and 50% formamide; a first wash at 65° C., 2×SSC, and 1% SDS; followed by a second wash at 65° C. and 0.1%×SSC. Lower stringency conditions suitable for detecting DNA sequences having about 50% sequence identity to an ARNT4 gene are detected by, for example, hybridization at 42° C. in the absence of formamide; a first wash at 42° C., 6×SSC, and 1% SDS; and a second wash at 50° C., 6×SSC, and 1% SDS. [0018]
  • A vector containing an ARNT4-encoding DNA is also within the invention. Preferably the DNA which includes an ARNT4-encoding DNA is less than 5 kilobases in length; more preferably, the DNA is less than 4 kilobases in length, more preferably the DNA is less than 3 kilobases in length, and most preferably the DNA is approximately 2 kilobases or less in length. The invention also provides a method of directing cardiac-specific or smooth muscle cell-specific expression of a protein by introducing into a cell an isolated DNA containing a sequence encoding the protein operably linked to the tissue-specific promoter. A cell containing the DNA or vector of the invention is also within the invention. [0019]
  • By “substantially pure DNA” is meant DNA that has a naturally-occurring sequence or that is free of the genes which, in the naturally-occurring genome of the organism from which the DNA of the invention is derived, flank the ARNT4 gene. The term therefore includes, for example, a recombinant DNA which is incorporated into a vector, into an autonomously replicating plasmid or virus, or into the genomic DNA of a procaryote or eucaryote at a site other than its natural site; or which exists as a separate molecule (e.g., a cDNA or a genomic or cDNA fragment produced by PCR or restriction endonuclease digestion) independent of other sequences. It also includes a recombinant DNA which is part of a hybrid gene encoding additional polypeptide sequence. [0020]
  • Also within the invention is a substantially pure human ARNT4 polypeptide. The term ARNT4 polypeptide includes a polypeptide having the amino acid sequence and length of the naturally-occurring ARNT4 as well as fragments of the full-length naturally-occurring ARNT4. The polypeptide contains the amino acid sequence of SEQ ID NO: [0021] 19. Preferably the polypeptide contains an amino acid sequence which is at least 50% identical to SEQ ID NO: 19. Preferably, the amino acid sequence has at least 75% identity, more preferably 85% identity, more preferably 90% identity, more preferably 95% identity, more preferably 99% identity, and most preferably 100% identity to the amino acid sequence of SEQ ID NO: 19. For example, the ARNT4 polypeptide may have the amino acid sequence of the naturally-occurring human polypeptide, e.g., a polypeptide which includes the amino acid sequence of SEQ ID NO: 19. The invention also encompasses a polypeptide with the amino acid sequence of a segment of SEQ ID NO: 17 which spans residues 75 to 128, inclusive, or a segment spanning residues 155 to 207, inclusive, of SEQ ID NO: 19, or a segment spanning residues 232 to 384 of SEQ ID NO: 19. Preferably, such a polypeptide has a biological activity of a naturally-occurring ARNT4 polypeptide, e.g, heterodimer formation with EPAS1 or the ability to transactivate transcription under the control of a VEGF promoter.
  • A substantially pure ARNT4 polypeptide is obtained by extraction from a natural source; by expression of a recombinant nucleic acid encoding a ARNT4 polypeptide; or by chemically synthesizing the protein. A polypeptide or protein is substantially pure when it is separated from those contaminants which accompany it in its natural state (proteins and other naturally-occurring organic molecules). Typically, the polypeptide is substantially pure when it constitutes at least 60%, by weight, of the protein in the preparation. Preferably, the protein in the preparation is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight, ARNT4. Purity is measured by any appropriate method, e.g., column chromatography, polyacrylamide gel electrophoresis, or HPLC analysis. Accordingly, substantially pure polypeptides include recombinant polypeptides derived from a eucaryote but produced in [0022] E. coli or another procaryote, or in a eucaryote other than that from which the polypeptide was originally derived.
  • The invention also includes a transgenic non-human mammal, the germ cells and somatic cells of which contain a null mutation in a gene encoding an ARNT4 polypeptide. For example, the null mutation is a deletion of part or all of an exon of ARNT4. Preferably, the mammal is a rodent such as a mouse. An antibody which specifically binds to a ARNT4 polypeptide is also within the invention. [0023]
  • Angiogenesis is inhibited by administering to a mammal a compound which inhibits binding of EPAS1 to ARNT4 such as an ARNT4 polypeptide. For example, the compound is a polypeptide or peptide mimetic which contains the amino acid sequence of residues [0024] 75 to 128, inclusive, of SEQ ID NO: 19, the amino acid sequence of residues 155 to 207, inclusive, of SEQ ID NO: 19, or a the amino acid sequence of residues 232 to 384 of SEQ ID NO: 19.
  • Other features and advantages of the invention will be apparent from the following detailed description and from the claims. [0025]
  • DETAILED DESCRIPTION
  • The drawings will first be described.[0026]
  • DRAWINGS
  • FIG. 1A is a bar graph showing dose-dependent transactivation of KDR/flk-1 promoter by EPAS1. EPAS1 expression plasmid phEP-1 (0-6 μg), pcDNA3 (6-0 μg), and KDR/flk-1 reporter pGL2-4kb+296 (1 μg) were transfected into BAEC. [0027]
  • FIG. 1B is a bar graph showing that deletion of EPAS1 C-terminal region abolishes its ability to transactivate the KDR/flk-1 promoter. Expression plasmids (6 μg each) and pGL2-4.Okb+296 (1 μg) were cotransfected into BAEC. For all constructs in FIGS. [0028] 1A-B, the plasmid pCMV-βGAL was cotransfected to correct for differences in transfection efficiency. In both FIGS. 1A and 1B, luciferase activity and β-galactosidase activity were measured, and normalized luciferase activity was calculated as described below. The “fold induction” represents the ratio (mean±SE) of normalized luciferase activity in cells transfected with expression plasmid to that in cells transfected with empty vector (pcDNA3).
  • FIG. 2A is a bar graph showing transactivation of the KDR/flk-1 promoter by EPAS1 but not by HIF-1a (another member of the PAS family of transcription factors). Expression plasmids (6 μg each) and KDR/flk-1 reporter pGL2-4kb+296 (1 μg) were cotransfected into the cell types indicated. [0029]
  • FIG. 2B is a bar graph showing transactivation of a VEGF promoter by EPAS1 and HIF-1α. Expression plasmids (6 μg each) and VEGF reporter pVR47/CAT (1 μg) were cotransfected into the cell types indicated. For all constructs in FIGS. [0030] 2A-2B, the plasmid pCMV- GAL was cotransfected to correct for differences in transfection efficiency. The “fold induction” represents the ratio (mean+SE) of normalized luciferase or CAT activity in cells transfected with expression plasmid to that in cells transfected with empty vector (pcDNA3).
  • FIG. 3A is a diagram showing an alignment of the amino acid sequence of human ARNT4 with human BMALlb and human ARNT. [0031]
  • FIG. 3B is a diagram of a phylogenetic tree of the ARNT family of proteins. [0032]
  • FIG. 4 is a bar graph showing the results of a yeast two-hybrid assay. ARNT3 (BMALlb) and ARNT4 form heterodimers with EPAS1 as well as with CLOCK. [0033]
  • FIG. 5 is a bar graph showing that EPAS1 interacts with ARNT4 to form functional heterodimers which increase VEGF promoter activity and VEGF expression. [0034]
  • FIG. 6 is a bar graph showing that ARNT4 does not interact with HIF-1α. [0035]
  • DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • EPAS1 is a member of the transcription factor family characterized by a basic helix-loop-helix (bHLH) domain and a (Per-AhR-Arnt-Sim) PAS domain composed of two imperfect repeats. Table 4 shows the amino acid sequence of human wild type EPAS1. [0036]
    TABLE 4
    Amino acid sequence of human EPAS1
    MTADKEKKRSSSERRKEKSRDAARCRRSKETEVFYELAHELPLPHSVSSHLDKASIMRLE (SEQ ID NO:6)
    ISFLRTHKLLSSVCSENESEAEADQQMDNLKALEGFIAVVTQDGDMIFLSENISKFMG
    LTQVELTGHSIFDFTHPCDHEEIRENLSLKNGSGFGKKSKDMSTERDFFMRMKCTVTNRG
    RTVNLKSATWKVLHCTGQVKVYNNCPPHNSLCGYKEPLLSCLIIMCEPIQHPSHMDIPLD
    SKTFLSRHSMDMKFTYCDDRITELIGYHPEELLGRSAYEFYHALDSENMTKSHQNLCTKG
    QVVSGQYRMLAKHGGYVWLETQGTVIYNPRNLQPQCIMCVNYVLSEIEKNDVVFSMDQTE
    SLFKPHLMAMNSIFDSSGKGAVSEKSNFLFTKLKEEPEELAQLAPTPGDAIISLDFGNQN
    FEESSAYGKAILPPSQPWATELRSHSTQSEAGSLPAFTVPQAAAPGSTTPSATSSSSSCS
    TPNSPEDYYTSLDNDLKIEVIEKLFAMDTEAKDOCSTQTDFNELDLETLAPYIPMDGEDF
    QLSPICPEERLLAENPQSTPQHCFSAMTNIFQPLAPVAPHSPFLLDKFQQQLESKKTEPE
    HRPMSSIFFDAGSKASLPPCCGQASTPLSSMGGRSNTQWPPDPPLHFGPTKWAVGDQRTE
    FLGAAPLGPPVSPPHVSTFKTRSAKGFGARGPDVLSPAMVALSNKLKLKRQLEYEEQAFQ
    DLSGGDPPGGSTSHLMWKRMKNLRGGSCPLMPDKPLSANVPNDKFTQNPMRGLGHPLRHL
    PLPQPPSAISPGENSKSRFPPQCYATQYQDYSLSSAHKVSGMASRLLGPSFESYLLPELT
    RYDCEVNVPVLGSSTLLQGGDLLRALDQAT
  • The N-terminal bHLH domain (which plays a role in DNA binding) and the C-terminal transactivation domain are highlighted (in bold and underlined type, respectively). [0037]
  • Table 5 shows the nucleotide sequence DNA encoding human wild type EPAS1. Nucleotides encoding the first amino acid of EPAS1 are underlined. [0038]
    TABLE 5
    Nucleotide sequence of human EPAS1 cDNA
    1 cctgactgcg cggggcgctc gggacctgcg cgcacctcgg accttcacca cccgcccggg (SEQ ID NO:7)
    61 ccgcggggag cggacgaggg ccacagcccc ccacccgcca gggagcccag gtgctcggcg
    121 tctgaacgtc tcaaagggcc acagcgacaa tgacagctga caaggagaag aaaaggagta
    181 gctcggagag gaggaaggag aagtcccggg atgctgcgcg gtgccggcgg agcaaggaga
    241 cggaggtgtt ctatgagctg gcccatgagc cgcctctgcc ccacagtgtg agctcccatc
    301 tggacaaggc ctccatcatg cgactggaaa tcagcttcct gcgaacacac aagctcctct
    361 cctcagtttg ctctgaaaac gagtccgaag ccgaagctga ccagcagatg gacaacttgt
    421 acctgaaagc cttggagggt ttcattgccg tggtgaccca agatggcgac atgatctttc
    481 tgtcagaaaa catcagcaag ttcatgggac ttacacaggt ggagctaaca ggacatagta
    541 tctttgactt cactcatccc tgcgaccatg aggagattcg tgagaacctg agtctcaaaa
    601 atggctctgg ttttgggaaa aaaagcaaag acatgtccac agagcgggac ttcttcatga
    661 ggatgaagtg cacggtcacc aacagaggcc gtactgtcaa cctcaagtca gccacctgga
    721 aggtcttgca ctgcacgggc caggtgaaag tctacaacaa ctgccctcct cacaatagtc
    781 tgtgtggcta caaggagccc ctgctgtcct gcctcatcat catgtgtgaa ccaatccagc
    841 acccatccca catggacatc cccctggata gcaagacctt cctgagccgc cacagcatgg
    901 acatgaagtt cacctactgt gatgacagaa tcacagaact gattggttac caccctgagg
    961 agctgcttgg ccgctcagcc tatgaattct accatgcgct agactccgag aacatgacca
    1021 agagtcacca gaacctgtgc accaagggtc aggtagtaag tggccagtac cggatgctcg
    1081 caaagcatgg gggctacgtg tggctggaga cccaggggac ggtcatctac aaccctcgca
    1141 acctgcagcc ccagtgcatc atgtgtgtca actacgtcct gagtgagatt gagaagaatg
    1201 acgtggtgtt ctccatggac cagactgaat ccctgttcaa gccccacctg atggccatga
    1261 acagcatctt tgatagcagt ggcaaggggg ctgtgtctga gaagagtaac ttcctattca
    1321 ccaagctaaa ggaggagccc gaggagctgg cccagctggc tcccacccca ggagacgcca
    1381 tcatctctct ggatttcggg aatcagaact tcgaggagtc ctcagcctat ggcaaggcca
    1441 tcctgccccc gagccagcca tgggccacgg agttgaggag ccacagcacc cagagcgagg
    1501 ctgggagcct gcctgccttc accgtgcccc aggcagctgc cccgggcagc accaccccca
    1561 gtgccaccag cagcagcagc agctgctcca cgcccaatag ccctgaagac tattacacat
    1621 ctttggacaa cgacctgaag attgaagtga ttgagaagct cttcgccatg gacacagagg
    1681 ccaaggacca atgcagtacc cagacggatt tcaatgagct ggacttggag acactggcac
    1741 cctatatccc catggacggg gaagacttcc agctaagccc catctgcccc gaggagcggc
    1801 tcttggcgga gaacccacag tccacccccc agcactgctt cagtgccatg acaaacatct
    1861 tccagccact ggcccctgta gccccgcaca gtcccttcct cctggacaag tttcagcagc
    1921 agctggagag caagaagaca gagcccgagc accggcccat gtcctccatc ttctttgatg
    1981 ccggaagcaa agcatccctg ccaccgtgct gtggccaggc cagcacccct ctctcttcca
    2041 tggggggcag atccaatacc cagtggcccc cagatccacc attacatttt gggcccacaa
    2101 agtgggccgt cggggatcag cgcacagagt tcttgggagc agcgccgttg gggccccctg
    2161 tctctccacc ccatgtctcc accttcaaga caaggtctgc aaagggtttt ggggctcgag
    2221 gcccagacgc gctgagtccg gccatggtag ccctctccaa caagctgaag ctgaagcgac
    2281 agctggagta tgaagagcaa gccttccagg acctgagcgg gggggaccca cctggtggca
    2341 gcacctcaca tttgatgtgg aaacggatga agaacctcag gggtgggagc tgccctttga
    2401 tgccggacaa gccactgagc gcaaatgtac ccaatgataa gttcacccaa aaccccatga
    2461 ggggcctggg ccatcccctg agacatctgc cgctgccaca gcctccatct gccatcagtc
    2521 ccggggagaa cagcaagagc aggttccccc cacagtgcta cgccacccag taccaggact
    2581 acagcctgtc gtcagcccac aaggtgtcag gcatggcaag ccggctgctc gggccctcat
    2641 ttgagtccta cctgctgccc gaactgacca gatatgactg tgaggtgaac gtgcccgtgc
    2701 tgggaagctc cacgctcctg caaggagggg acctcctcag agccctggac caggccacct
    2761 gagccaggcc ttctacctgg gcagcacctc tgccgacgcc gtcccaccag cttcaccc
  • Hypoxia inducible factor-1α (HIF-1α) is another member of the PAS family to which EPAS1 belongs. Transcription factors of this family use the bHLH and PAS domains to form heterodimers that subsequently bind to target genes and regulate important biological processes. [0039]
  • EPAS1 plays a role in the regulation of angiogenic factors such as VEGF, VEGF-R such as KDR/flk-1 and flt-1, and Tie2. EPAS1, a nuclear protein with a basic helix-loop-helix (bHLH)/PAS domain, is expressed preferentially in endothelial cells. EPAS1 transcription factor or DNA encoding all or part of EPAS1 (e.g., a fragment containing the C-terminal activation domain) is administered to individuals to promote angiogenesis. To inhibit angiogenesis, EPAS1 antisense sequences are administered to cells to decrease intracellular production of EPAS1 gene product. Administration of DNA encoding an EPAS1-specific antibody (e.g., EPAS1 intrabodies) or EPAS1 dominant negative mutants can also be administered cells to inhibit EPAS1 function, e.g., by inhibiting binding of EPAS1 to cis-acting regulatory sequences of VEGF, VEGF-R, or Tie2 genes or by inhibiting EPAS1 transactivation of gene transcription. By regulating transcription of VEGF, VEGF-Rs, and Tie2, EPAS1 is useful to modulate vasculogenesis and angiogenesis. [0040]
  • Production of ARNT4-Specific Antibodies
  • Anti-ARNT4 antibodies are obtained by techniques well known in the art. Such antibodies can be polyclonal or monoclonal. Polyclonal antibodies are obtained, for example, by the methods described in Ghose et al., Methods in Enzymology, Vol. 93, 326-327, 1983. For example, a ARNT4 polypeptide, or an antigenic fragment thereof, can be used as an immunogen to stimulate the production of ARNT4-reactive polyclonal antibodies in the antisera of animals such as rabbits, goats, sheep, or rodents. Antigenic polypeptides useful as immunogens include polypeptides which contain a bHLH domain/PAS domain. [0041]
  • Monoclonal antibodies are obtained by standard techniques such as those described by Milstein and Kohler in Nature, 256:495-97, 1975, or as modified by Gerhard, Monoclonal Antibodies, Plenum Press, 1980, pages 370-371. Hybridomas are screened to identify those producing antibodies that are highly specific for an ARNT4 polypeptide. Preferably, the antibody will have an affinity of at least about 108 liters/mole and more preferably, an affinity of at least about 109 liters/mole. [0042]
  • ARNT4-Deficient Mice
  • To further investigate the role of ARNT4 in vivo, ARNT4 knockout mice (ARNT4-deficient mice) are generated by homologous recombination. A gene targeting construct for generating ARNT4-deficient mice is made using a targeted gene deletion strategy using standard methods. The deletion in the ARNT4 gene renders the ARNT4 polypeptide non-functional. The linearized targeting construct is transfected into murine D3 embryonic stem (ES) cells, and a clone with the correct homologous recombination (yielding the appropriately disrupted ARNT4 gene) is injected into blastocysts and used to generate ARNT4-deficient mice. [0043]
  • Activation of the KDR/flk-1 Promoter by EPAS1
  • EPAS1 and KDR/flk-1 transcripts were found to colocalize in vascular endothelial cells in mouse embryonic and adult tissue. To study the expression of EPAS1 relative to KDR/flk-1, a plasmid containing 4.0 kb of human KDR/flk-1 5′-flanking sequence linked to the luciferase reporter gene and a second vector containing DNA encoding either EPAS1 or another bHLH-PAS domain transcription factor HIF-1α were cotransfected into bovine aortic endothelial cells (BAEC). EPAS1 but not HIF-1α markedly increased KDR/flk-1 promoter activity in a dose-dependent manner, and this induction of the KDR/flk-1 promoter by EPAS1 occurred preferentially in endothelial cells. In contrast, both EPAS1 and HIF-1α activated the VEGF promoter in a non-endothelial cell-specific manner. This is the first demonstration of transactivation of the KDR/flk-1 promoter by EPAS1. By regulating transcription of KDR/flk-1 and VEGF, EPAS1 plays an important role in regulating vasculogenesis and angiogenesis. [0044]
  • Cell Culture
  • BAEC were isolated and cultured in DME supplemented with 10% FCS (HyClone, Logan, Utah) and antibiotics according to known procedures. BAEC were passed every 3-5 days, and cells from passages 5-7 were used for the transfection experiments. The following cell lines were obtained from the American Type Culture Collection (ATCC) and were cultured in the same medium as BAEC: HeLa cells (human epidermoid carcinoma cells; ATCC #CRL7396 and NIH 3T3 cells (mouse fibroblasts; ATCC #CRL1888). [0045]
  • RNA Isolation and Northern Analysis
  • Total RNA was isolated from mouse organs by guanidinium isothiocyanate extraction and centrifugation through cesium chloride according to standard protocols. Total RNA (10 μg) was fractionated on a 1.3% formaldehyde-agarose gel and transferred to Nitropure filters (MSI, Westborough, Mass.). The filters were then hybridized with [0046] 32P-labeled, randomly primed cDNA probes for 1 h at 68° C. in Quick-hyb solution (Stratagene, La Jolla, Calif.). The hybridized filters were washed in 30 mM NaCl, 3 mM sodium citrate, and 0.1% sodium dodecyl sulfate at 55° C. and autoradiographed for 20 h on Kodak XAR film at −80° C. To correct for differences in RNA loading, the filters were rehybridized with a radiolabeled ribosomal 18S-specific oligonucleotide. A 1.8 kb AccI-AccI fragment of mouse EPAS1 (GENBANK Accession # U81983) was used as a probe. The 667 (382-1086) bp mouse KDR/flk-1 cDNA fragment was amplified by the reverse transcriptase PCR by using mouse lung total RNA. The forward (5′ GAACTTGGATGCTCTTTGGAAA 3′; SEQ ID NO: 8) and reverse (5′ CACTTGCTGGCATCATAAGGC 3′; SEQ ID NO: 9) primers were used to generate PCR fragments that were subcloned into to a pCR 2.1 vector (Invitrogen, Carlsbad, Calif.). Nucleotide sequence authenticity was confirmed by the dideoxy chain termination method.
  • In Situ Hybridization
  • To generate probes for in situ hybridization, a 316 (771-1086) bp mouse EPAS1 CDNA and a 342 (2346-2687) bp mouse KDR/flk-1 cDNA from mouse lung total RNA was amplified by reverse transcriptase PCR with the following primers: EPAS1, forward 5° CATCATGTGTGAGCCAATCCA 3′ (SEQ ID NO: [0047] 10) and reverse 5′ GTTGTAGATGACCGTCCCCTG 3′ (SEQ ID NO: 11) KDR/flk-1, forward 5′ TGTACTGAGAGATGGGAACCG 3′ (SEQ ID NO: 12) and reverse 5′ CACTTGCTGGCATCATAAGGC 3′ (SEQ ID NO: 13). PCR fragments were subcloned into the pCR 2.1 vector in both orientations and the authenticity of the sequences was confirmed.
  • Slides of E9 mouse sections were purchased from Novagen (Madison, Wis.). E12 mice and various adult mouse organs were fixed in 4% paraformaldehyde, dehydrated, and embedded in paraffin. Tissue sections (6 μm thick) were hybridized with a [0048] 35S-UTP-labeled antisense cRNA probe synthesized with the T7 RNA polymerase from linearized plasmids containing appropriate cDNA fragments using standard techniques. As a negative control, tissue sections were also hybridized with 35S-UTP-labeled sense cRNA probes. After hybridization the tissue sections were washed, and the dried tissue sections were then dipped into Kodak NTB2 emulsion (Eastman Kodak) and exposed for 10-15 days at 4° C. The sections were counterstained with hematoxylin and eosin.
  • Construction of Plasmids
  • pGL2-Basic and pGL2-Control contained the firefly luciferase reporter gene (Promega, Madison, Wis.). pGL2-Basic had no promoter, whereas pGL2-Control contained the SV40 promoter and enhancer. The pGL2-4kb+296 reporter plasmid was constructed by inserting the human KDR/flk-1 promoter from -4kb to +296 into pGL2-Basic. pVR47/CAT, which contains the human VEGF promoter from −2362 to +61 and the chloramphenicol acetyltransferase (CAT) reporter gene sequence, was also constructed using standard techniques. [0049]
  • The plasmid phEP-lAS was made by cloning the antisense EPAS1 cDNA into pcDNA3. phEP-1ΔCT, containing a C-terminal deletion mutant of the EPAS1 cDNA, was generated by subcloning a BamHI-XhoI restriction fragment encoding human EPAS1 amino acids [0050] 1-690 into pcDNA3. To generate phHIF-1α, a 2622 bp cDNA fragment containing the entire open reading frame of human HIF-1a was amplified using human leukocyte total RNA and pfu DNA polymerase (Stratagene, La Jolla, Calif.). The sequences of the forward (5′ GTGAAGACATCGCGGGGACC 3′; SEQ ID NO: 14) and reverse (5′ GTTTGTGCAGTATTGTAGCCAGG 3′; SEQ ID NO: 15) primers were based on human HIF-1a cDNA (Wang et al., 1995, Proc. Natl. Acad. Sci. USA. 92:5510-5514). The PCR fragment was then cloned into pcDNA3, and the sequence was confirmed. Expression of phEP-1, phEP-1ΔCT, and phHIF-1a was confirmed by in vitro transcription and translation in the TNT-coupled reticulocyte lysate system (Promega, Madison, Wis.) according to the manufacturer's instructions.
  • Transient Transfection Assays
  • Cells were transfected with 1 μg of reporter construct and 6 μg of expression construct by the standard calcium phosphate method. To correct for variability in transfection efficiency against β-galactosidase, 1 βg of pCMV-βGAL was cotransfected in all experiments. Cell extracts were prepared 48 h after transfection by a detergent lysis method (Promega, Madison, Wis.). Luciferase activity was measured in duplicate for all samples with an EC&G Autolumat 953 (Gaithersburg, Md.) luminometer by the Promega luciferase assay. CAT activity was assayed by a two-phase fluor diffusion method. β-galactosidase activity was assayed using standard methods. The ratio of luciferase or CAT activity to β-galactosidase activity in each sample served as a measure of normalized luciferase or CAT activity. Each construct was transfected at least four times, and each transfection was done in triplicate. Data for each construct are presented as the mean ± SE. [0051]
  • Statistics
  • Comparisons between groups were made by a factorial analysis of variance followed by Fisher's least significant difference test when appropriate. Statistical significance was accepted at p<0.05. [0052]
  • Tissue distribution of EPAS1 and KDR/flk-1 in adult mice Northern blot analysis was performed with RNA prepared from various adult mouse tissues. EPAS1 mRNA was abundant in the lung, heart, and aorta, organs known to be rich in vascular endothelial cells. When the same blot was hybridized to a mouse KDR/flk-1 probe, the expression pattern of KDR/flk-1 was identical to that of EPAS1. In situ hybridization was performed using an antisense mouse EPAS1 probe to determine which cells in the aorta expressed EPAS1. The EPAS1 message localized to the luminal layer, and the antisense EPAS1 probe but not the sense EPAS1 probe hybridized to the endothelial cells of the aorta. [0053]
  • Tissue Distribution of EPAS1 and KDR/flk-1 in Developing Mouse Embryos
  • To characterize the temporal and spatial patterns of EPAS1 and KDR/flk-1 expression in developing mouse embryos, in situ hybridization was performed with EPAS1 and KDR/flk-1 probes. In embryonic-day (E)9 mice, EPAS1 mRNA was visible in the dorsal aorta, the endocardium of the developing outflow tract, the ventricle, and the perineural vascular plexus. KDR/flk-1 mRNA was expressed similarly in the same organs. At the E9 stage of development, the mouse aorta is composed mainly of a single layer of endothelial cells. Both EPAS1 and KDR/flk-1 were expressed in endothelial cells of the aorta and other organs. At E12.5, EPAS1 mRNA was visible in the intervertebral blood vessels, heart, vascular plexuses in the meninges surrounding both the spinal cord and the brain, and choroid plexus. The distribution of KDR/flk-1 MRNA at E12.5 was strikingly similar. The EPAS1 and KDR/flk-1 mRNAs were both detected in endothelial cells of the blood vessels at higher magnification as well. [0054]
  • Transactivation of the KDR/flk-1 Promoter by EPAS1 in a Dose-Dependent Manner
  • The colocalization of EPAS1 and KDR/flk-1 indicates that EPAS1 is important in regulating KDR/flk-1 expression. To test the role of EPAS1 in regulation of protein expression, a human EPAS1 expression plasmid (phEP-1) and a reporter plasmid (pGL2-4kb+296) containing approximately 4.0 kb of the human KDR/flk-1 5′-flanking sequence linked to a luciferase reporter gene were cotransfected into BAEC. EPAS1 increased KDR/flk-1 promoter activity in a dose-dependent manner (FIG. 1A). As little as 2 μg of EPAS1 expression vector phEP-1 increased the promoter activity of KDR/flk-1 by 3-fold, and 6 μg of the EPAS1 vector increased luciferase activity by 12.9-fold. Upregulation of KDR/flk-1 promoter activity by EPAS1 was specific, since cotransfection of the EPAS1 expression vector had no effect on the activity of pGL2-Control vector driven by the potent SV40 promoter and enhancer. [0055]
  • To identify the EPAS1 domain which participates in transactivation of the KDR/flk-1 promoter, plasmid phEP-lACT was constructed to express a truncated form of EPAS1 lacking its 180 C-terminal amino acids. Deletion of the 180 C-terminal amino acids of EPAS1 completely abolished its ability to transactivate the KDR/flk-1 promoter (FIG. 1B). These data indicate that the 180 C-terminal amino acids of EPAS1 are necessary for transactivation of the KDR/flk-1 promoter. [0056]
  • These data indicate that induction of the mRNA for KDR/flk-1 colocalizes with that of the mRNA for EPAS1 in vascular endothelial cells from fetal as well as adult mice. EPAS1 also transactivates the promoter of Tie2, which, like KDR/flk-1, is an endothelial cell-specific tyrosine kinase. Expression of Tie2 in endothelial cells is high during fetal development but barely detectable in adulthood. In contrast, expression of EPAS1 in endothelial cells is high in fetuses as well as adults. Thus, the target gene for EPAS1 in adults is a VEGF-R such as KDR/flk-1 or flt-1 (as well as VEGF) as evidenced by the data showing that EPAS1 markedly induces KDR/flk-1 promoter activity. [0057]
  • EPAS1 but not HIF-1αTransactivates the KDR/flk-1 Promoter Preferentially in Vascular Endothelial Cells
  • To determine whether another member of the bHLH/PAS family transactivated the KDR/flk-1 promoter, the EPAS1 or HIF-1α expression plasmid and the KDR/flk-1 plasmid pGL2-4kb+296 were cotransfected into BAEC, HeLa cells, and NIH 3T3 cells. EPAS1 expression plasmids in the sense (phEP-1) but not the antisense (phEP-1AS) orientation activated the KDR/flk-1 promoter (FIG. 2A), indicating that the transactivating effect is cell-specific. Although the EPAS1 plasmid markedly increased KDR/flk-1 promoter activity in vascular endothelial cells, it had little effect on KDR/flk-1 promoter activity in HeLa or NIH 3T3 cells (FIG. 2A). HIF-1α had no effect on KDR/flk-1 promoter activity in all three cell types. The EPAS1 or HIF-1α expression plasmid was then cotransfected with a reporter plasmid containing the VEGF promoter, pVR47/CAT, to determine whether the differential effects of EPAS1 and HIF-1a were unique to the KDR/flk-1 promoter. In contrast to its cell-specific effect on the KDR/flk-1 promoter (FIG. 2A), EPAS1 transactivated the VEGF promoter in all three cell types (FIG. 2B). Induction was highest in HeLa cells. Furthermore, HIF-1a increased VEGF promoter activity in BAEC and HeLa cells (FIG. 2B). These data indicate that the transactivating effect of EPAS1 depends on both the promoter and the cell type. [0058]
  • Although EPAS1 transactivated the KDR/flk promoter preferentially in endothelial cells (FIG. 2A), it activated the VEGF promoter in a non-endothelial cell-specific manner (FIG. 2B). Despite the fact that HIF-1α is 48% homologous to EPAS1, HIF-1α had no effect on the KDR/flk-1 promoter. In contrast, HIF-1α transactivated the VEGF promoter. Thus, the effect of EPAS1 on the KDR/flk-1 promoter is specific and cannot be replaced by other members of the PAS family of transcription factors. [0059]
  • EPAS1 heterodimerizes with the aryl hydrocarbon receptor nuclear translocator and transactivates the promoter of Tie2. EPAS1 also markedly increases the promoter activity of KDR/flk-1 and VEGF. Mice deficient in the aryl hydrocarbon receptor nuclear translocator are not viable past E10.5, and the yolk sac shows defective angiogenesis. These data indicate that EPAS1 functions as a nodal transcription factor by regulating expression of VEGF, KDR/flk-1, and Tie2 during vasculogenesis and angiogenesis. [0060]
  • Characterization of Functional Domains of EPAS1
  • Functional domains of EPAS1 were identified as follows. The gene encoding VEGF has a cis-acting regulatory sequence to which EPAS1 binds (GCCCTACGTGCTGTCTCA; SEQ ID NO:[0061] 1) in its 5′ flanking region. In cotransfection experiments in BAEC, the EPAS1 expression plasmid activated by 30-fold a CAT reporter plasmid containing 2.3 kb of VEGF 5′ flanking sequence (containing SEQ ID NO: 1) but not a similar plasmid differing only by a mutation in an amino acid of SEQ ID NO: 1. These data indicate that EPAS1 activates the VEGF promoter by binding to DNA containing the sequence of SEQ ID NO: 1. To further characterize domains of EPAS1 which function to activate promoters of angiogenic factors in endothelial cells, e.g., the VEGF promoter or VEGF-R promoters, BAEC were cotransfected with expression plasmids encoding EPAS1 mutants and the reporter plasmid. Eight mutants were tested. Deletion of the basic region (bHLH region) of EPAS1 (SEQ ID NO: 3) completely abolished its ability to induce transcription from the VEGF promoter, indicating that binding of EPAS1 to the cis-acting element though this basic region is critical. Deletion of 180 amino acids from the C-terminus of EPAS1 has little or no effect on the transcriptional transactivation activity of EPAS1 for the VEGF promoter; however, a deletion of the C-terminal 385 amino acids abolished the ability of EPAS1 to activate the VEGF promoter, indicating the presence of a transactivation domain in the portion of EPAS1 spanning amino acids 486-690. Further fine deletion analyses indicated that the transactivation domain of EPAS1 spans amino acids 486-639. An EPAS1 mutant polypeptide lacking the amino acid sequence of SEQ ID NO: 2, e.g., an EPAS1 with the amino acid sequence of SEQ ID NO: 4, functions as a dominant negative mutant EPAS1 because it inhibited transactivation of the VEGF promoter by wild type EPAS1 in a dose-dependent manner. Deletion analysis is also used to identify domains of EPAS1 which participate in heterodimer formation with ARNT4.
  • To characterize domains of ARNT4 which function to heterodimerize with EPAS1 and activate promoters of angiogenic factors in endothelial cells, e.g., the VEGF promoter or VEGF-R promoters, BAEC are cotransfected with expression plasmids encoding EPASI and ARNT4 deletion mutants and the reporter plasmid as described above. Domains of ARNT4 which participate in EPAS1 heterodimer formation with EPAS1 are identified using the yeast two-hybrid assay or a gel mobility assay. For example, those mutants which fail to activate the VEGF/luciferase promoter cannot form functional dimers with EPAS1. [0062]
  • This assay is also used to identify compounds which inhibit or decrease formation of functional ARNT4/EPAS1 heterodimers, and thus, inhibit angiogenesis. In such an assay, expression plasmids which encode wild type or functional fragments of ARNT4 and EPAS1 are cotransfected with a VEGF/luciferase reporter plasmid into an endothelial cell in the presence and absence of a candidate compound. A decrease in the amount of transactivation of the VEGF promoter (e.g., as measured by a standard luciferase assay) in the presence of the compound compared to the amount in the absence of the candidate compound indicates that the compound inhibits angiogenesis (by inhibiting ARNT4/EPAS1 transactivation of the VEGF promoter). [0063]
  • Generation of a Dominant-Negative EPAS1 Mutants
  • An adenoviral construct which expresses EPAS1 was generated. Overexpression of EPAS1 dramatically induced VEGF mRNA in human umbilical endothelial cells. In cotransfection experiments, EPAS1 transactivated the VEGF promoter via the HIF-1 binding site. This transactivation was further enhanced by hypoxia. Cotransfection of an aryl hydrocarbon receptor nuclear translocator (ARNT) expression plasmid and EPAS1 expression plasmid synergistically transactivated the VEGF promoter, indicating that heterodimerization of EPAS1 and ARNT is crucial for the transactivation of the VEGF promoter (FIG. 5). Using a gel shift analysis, EPAS1 (but not HIF-1) formed dimers with ARNT4 and bound to the HIF-1 binding site of the VEGF promoter. [0064]
  • Deletion analysis of EPAS1 further defined a potent transactivation domain to span amino acids [0065] 486-639 of human EPAS1 (SEQ ID NO: 6). The transactivation domain is essential for EPAS1 to transactivate the VEGF promoter. The ability of this domain to activate transcription was confirmed using the GAL4 fusion protein system. Finally, a truncated EPAS1 lacking the transactivation domain (e.g., an EPAS1 polypeptide lacking amino acids 486-690 of SEQ ID NO: 6 or an EPASI polypeptide lacking amino acids 486-639 of SEQ ID NO: 6) retained its ability to form heterodimers and to bind the HIF-1 binding site. These data indicate that the mutated EPAS1 polypeptides with lack amino acids in the transactivation domain are dominant negative mutants because they sequester ARNT and prevent the formation of functional EPAS1/ARNT and HIF-1α/ARNT heterodimers. For example, the EPAS1 polypeptide which lacked amino acids 486-639 of SEQ ID NO: 6 potently inhibited the induction of the VEGF promoter by EPAS1 and HIF-1α. Transfection of endothelial cells with an adenovirus construct encoding this mutant inhibited VEGF MRNA induction by hypoxia. These results indicate that EPAS1 is an important regulator of VEGF gene expression and that dominant negative EPAS1 mutants (e.g., EPAS1 polypeptides lacking all or part of the transactivation domain (SEQ ID NO: 2)) inhibit VEGF promoter activity, and in turn, VEGF expression and angiogenesis.
  • Identification of Compounds which Modulate EPAS1 Binding to Cis-Regulatory Sequences
  • Modulation of the angiogenesis is achieved by contacting the vascular cells such as vascular endothelial cells with a compound that blocks or enhances EPAS1 binding to cis-acting regulatory sequences of VEGF, VEGF-Rs, or other angiogenic factors in endothelial cells such as Tie2. Such a compound can be identified by methods ranging from rational drug design to screening of random compounds. The latter method is preferable, as simple and rapid assays for testing such compounds are available. oligonucleotides and small organic molecules are desirable candidate compounds for this analysis. [0066]
  • The screening of compounds for the ability to modulate angiogenesis by affecting EPAS1 transactivation of transcription of angiogenic factors may be carried out using in vitro biochemical assays, cell culture assays, or animal model systems. For example, in a biochemical assay, labeled EPAS1 (e.g., EPAS1 labeled with a fluorochrome or a radioisotope) is applied to a column containing immobilized DNA containing the cis-acting regulatory sequence. Alternatively, ARNT4 is immobilized on the column. In this manner, compounds which inhibit ARNT4/EPAS1 heterodimerization may be identified. A candidate compound is applied to the column before, after, or simultaneously with the labeled EPAS1, and the amount of labeled protein bound to the column in the presence of the compound is determined by conventional methods. A compound tests positive for inhibiting EPAS1 binding (thereby having the effect of inhibiting angiogenesis) if the amount of labeled protein bound in the presence of the compound is lower than the amount bound in its absence. Conversely, a compound tests positive for enhancing EPAS1 binding (thereby having the effect of enhancing angiogenesis) if the amount of labeled protein bound in the presence of the compound is greater than the amount bound in its absence. In a variation of the above-described biochemical assay, binding of labeled DNA to immobilized EPAS1 is measured. [0067]
  • As mentioned above, candidate compounds may also be screened using cell culture assays. Cells expressing EPAS1, either naturally or after introduction into the cells of genes encoding EPAS1 are cultured in the presence of the candidate compound. The level of EPAS1 binding in the cell may be inferred using any of several assays. For example, levels of expression of EPAS1 regulated genes (e.g., genes encoding VEGF, VEGF-Rs such as KDR/flk-1 or flt-1) in the cell may determined using, e.g., Northern blot analysis, RNAse protection analysis, immunohistochemistry, or other standard methods. Compounds identified as having the desired effect, either enhancing or inhibiting EPAS1 binding, can be tested further in appropriate animal models, e.g., an animal with a tumor or atherosclerotic lesion. [0068]
  • Compounds found to inhibit EPAS1 binding to cis-acting regulatory sequences of geres encoding angiogenic factors may be used in methods for nhibiting pathogenic angiogenesis in order to, e.g., prevent or treat tumor progression or the progression of an atherosclerotic lesion. Compounds found to enhance EPAS1 binding may be used in methods to therapeutically promote new blood vessel formation in adult mammals as discussed above. [0069]
  • The therapeutic compounds identified using the methods of the invention may be administered to a patient by any appropriate method for the particular compound, e.g., orally, intravenously, parenterally, transdermally, transmucosally, by inhalation, or by surgery or implantation at or near the site where the effect of the compound is desired (e.g., with the compound being in the form of a solid or semi-solid biologically compatible and resorbable matrix). Therapeutic doses are determined specifically for each compound, most being administered within the range of 0.001 to 100.0 mg/kg body weight, or within a range that is clinically determined to be appropriate by one skilled in the art. [0070]
  • Identification and Molecular Cloning of the EPAS1 Binding Partner, ARNT-4
  • Compositions which interact with EPAS1 were identified by screening for endothelial cell proteins which bind to EPAS1. Yeast two hybrid screening of a human umbilical endothelial cell cDNA library was carried out using EPAS1 as a bait. One of the clones isolated encoded a novel bHLH/PAS protein which was found to have similarity with arylhydrocarbon nuclear translocator [0071] 3 (Arnt3), a member of bHLH/PAS protein which heterodimerizes with Clock, a gene product involved in regulation of mammalian circadian rhythm. The isolated clone was named ARNT4. As described above, the CLUSTAL W sequence alignment system was used to compare the sequences of ARNT4 with the most closely related known DNA and/or amino acid sequences. With respect to DNA (comparison of coding sequences; untranslated regions excluded), the sequences of hARNT and hARNT4 were found to be 35% identical; the sequences of hBMAL lb and hARNT4 were found to be 56% identical; and the sequences of hARNT and hBMAL lb were found to be 37% identical. Nucleotide sequence comparisons using the CLUSTAL W system were carried out using the following parameters: KTUP=2; gap penalty=5; top diagonals=4; and window size=4. With respect to the proteins, the amino acid sequences of hARNT and hARNT4 were found to be 23% identical; the sequences of hBMAL lb and hARNT4 were found to be 49% identical; and the sequences of hARNT and hBMAL lb were found to be 26% identical. Amino acid sequence comparisons using the CLUSTAL W system were carried out using the following parameters: KTUP=1; gap penalty=3; top diagonals=5; and window size=5.
  • Northern analysis of human tissue revealed that this gene is highly expressed in brain, heart and placenta. In the brain, expression was high in the thalamus and amygdala, an almond-shaped component of the limbic system located in the temporal lobe of the brain. [0072]
  • Expression within human cultured cells demonstrated highest mRNA levels in vascular endothelial cells and smooth muscle cells. ARNT4 was shown to interact with EPASI using the yeast two-hybrid assay (FIG. 4). In a gel mobility shift assay using hypoxia responsive element of VEGF gene as the probe, ARNT4 formed a heterodimer with EPAS1 and bound to the hypoxia responsive element of the VEGF gene. [0073]
  • An expression plasmid encoding EPAS1 and an expression plasmid encoding ARNT4 were cotransfected with a VEGF/luciferase reporter plasmid into bovine aortic endothelial cells. Coexpression of ARNT4 and EPAS1 markedly transactivated the VEGF promoter (FIG. 5), and this transactivation was further enhanced by hypoxia. These data indicate that the heterodimer EPAS1/ARNT4 is activated under hypoxic conditions. Taken together, these results indicate that ARNT4, a novel bHLH/PAS protein, is an important regulator of VEGF gene expression especially in vascular system. [0074]
    TABLE 6
    Human ARNT4 amino acid sequence
    M A A E E 5 (SEQ ID NO:19)
    6 E A A A G G K V L R E E N Q C I A P V V 25
    26 S S R V S P G T R P T A M G S F S S H M 45
    46 T E F P R K R K G S D S D P S Q V E D G 65
    66 E H Q V K M K A F R E A H S Q T E K R R 85
    86 R D K M N N L I E E L S A M I P Q C N P 105
    106 M A R K L D K L T V L R M A V Q H L R S 125
    126 L K G L T N S Y V G S N Y R P S F L Q D 145
    146 N E L R H L I L K T A E G F L F V V G C 165
    166 E R G K I L F V S K S V S K I L N Y D Q 185
    186 A S L T G Q S L F D F L H P K D V A K V 205
    206 K E Q L S S F D I S P R E K L I D A K T 225
    226 G L Q V H S N L H A G R T R V Y S G S R 245
    246 R S F F C R I K S C K I S V K E E H G C 265
    266 L P N S K K K E H R K F Y T I H C T G Y 285
    286 L R S W P P N I V G M E E E R N S K K D 305
    306 N S N F T C L V A I G R L Q P Y I V P Q 325
    326 N S G E I N V K P T E F I T R F A V N G 345
    346 K F V Y V D Q R A T A I L G Y L P Q E L 365
    366 L G T S C Y E Y F H Q D D H N N L T D K 385
    386 H K A V L Q S K E K I L T D S Y K F R A 405
    406 K D G S F V T L K S Q W F S F T N P W T 425
    426 K E L E Y I V S V N T L V L G H S E P G 445
    446 E A S F L P C S S Q S S E E S S R Q S C 465
    466 M S V P G M S T G T V L G A G S I G T D 485
    486 I A N E I L D L Q R L Q S S S Y L D D S 505
    506 S P T G L M K D T H T V N C R S M S N K 525
    526 E L F P P S P S E M G E L E A T R Q N Q 545
    546 S T V A V H S H E P L L S D G A Q L D F 565
    566 D A L C D N D D T A M A A F M N Y L E A 585
    586 E G G L G D P G D F S D I Q W T L 602
  • [0075]
    TABLE 7
    Human ARNT4 cDNA
    ctccagtccgcatgctcagtagctgctgccggccgggctgcggggcggcgtccgctgcgc (SEQ ID NO:20)
    gcctacgggctgcggtggcggccgccgcggcacccggcagggcccgccagtccccgcttc
    cctgctccagagccgccgcctgggccggggcagggcgggcccggggctcctccatgctgc
    cagccgccgggctgcggagccgaccaagtggctcctgcg ATG GCG GCG GAA GAG
    GAG GCT GCG GCG GGA GGT AAA GTG TTG AGA GAG GAG AAC CAG TGC
    ATT GCT CCT GTG GTT TCC AGC CGC GTG AGT CCA GGG ACA AGA CCA
    ACA GCT ATG GGG TCT TTC AGC TCA CAC ATG ACA GAG TTT CCA CGA
    AAA CGC AAA GGA AGT GAT TCA GAC CCA TCC CAA GTG GAA GAT GGT
    GAA CAC CAA GTT AAA ATG AAG GCC TTC AGA GAA GCT CAT AGC CAA
    ACT GAA AAG CGG AGG AGA GAT AAA ATG AAT AAC CTG ATT GAA GAA
    CTG TCT GCA ATG ATC CCT CAG TGC AAC CCC ATG GCG CGT AAA CTG
    GAC AAA CTT ACA GTT TTA AGA ATG GCT GTT CAA CAC TTG AGA TCT
    TTA AAA GGC TTG ACA AAT TCT TAT GTG GGA AGT AAT TAT AGA CCA
    TCA TTT CTT CAG GAT AAT GAG CTC AGA CAT TTA ATC CTT AAG ACT
    GCA GAA GGC TTC TTA TTT GTG GTT GGA TGT GAA AGA GGA AAA ATT
    CTC TTC GTT TCT AAG TCA GTC TCC AAA ATA CTT AAT TAT GAT CAG
    GCT AGT TTG ACT GGA CAA AGC TTA TTT GAC TTC TTA CAT CCA AAA
    GAT GTT GCC AAA GTA AAG GAA CAA CTT TCT TCT TTT GAT ATT TCA
    CCA AGA GAA AAG CTA ATA GAT GCC AAA ACT GGT TTG CAA GTT CAC
    AGT AAT CTC CAC GCT GGA AGG ACA CGT GTG TAT TCT GGC TCA AGA
    CGA TCT TTT TTC TGT CGG ATA AAG AGT TGT AAA ATC TCT GTC AAA
    GAA GAG CAT GGA TGC TTA CCC AAC TCA AAG AAG AAA GAG CAC AGA
    AAA TTC TAT ACT ATC CAT TGC ACT GGT TAC TTG AGA AGC TGG CCT
    CCA AAT ATT GTT GGA ATG GAA GAA GAA AGG AAC AGT AAG AAA GAC
    AAC AGT AAT TTT ACC TGC CTT GTG GCC ATT GGA AGA TTA CAG CCA
    TAT ATT GTT CCA CAG AAC AGT GGA GAG ATT AAT GTG AAA CCA ACT
    GAA TTT ATA ACC CGG TTT GCA GTG AAT GGA AAA TTT GTC TAT GTA
    GAT CAA AGG GCA ACA GCG ATT TTA GGA TAT CTG CCT CAG GAA CTT
    TTG GGA ACT TCT TGT TAT GAA TAT TTT CAT CAA GAT GAC CAC AAT
    AAT TTG ACT GAC AAG CAC AAA GCA GTT CTA CAG AGT AAG GAG AAA
    ATA CTT ACA GAT TCC TAC AAA TTC AGA GCA AAA GAT GGC TCT TTT
    GTA ACT TTA AAA AGC CAA TGG TTT AGT TTC ACA AAT CCT TGG ACA
    AAA GAA CTG GAA TAT ATT GTA TCT GTC AAC ACT TTA GTT TTG GGA
    CAT AGT GAG CCT GGA GAA GCA TCA TTT TTA CCT TGT AGC TCT CAA
    TCA TCA GAA GAA TCC TCT AGA CAG TCC TGT ATG AGT GTA CCT GGA
    ATG TCT ACT GGA ACA GTA CTT GGT GCT GGT AGT ATT GGA ACA GAT
    ATT GCA AAT GAA ATT CTG GAT TTA CAG AGG TTA CAG TCT TCT TCA
    TAC CTT GAT GAT TCG AGT CCA ACA GGT TTA ATG AAA GAT ACT CAT
    ACT GTA AAC TGC AGG AGT ATG TCA AAT AAG GAG TTG TTT CCA CCA
    AGT CCT TCT GAA ATG GGG GAG CTA GAG GCT ACC AGG CAA AAC CAG
    AGT ACT GTT GCT GTC CAC AGC CAT GAG CCA CTC CTC AGT GAT GGT
    GCA CAG TTG GAT TTC GAT GCC CTA TGT GAC AAT GAT GAC ACA GCC
    ATG GCT GCA TTT ATG AAT TAC TTA GAA GCA GAG GGG GGC CTG GGA
    GAC CCT GGG GAC TTC AGT GAC ATC CAG TOG ACC CTC
    tagcctttgatttttaactccaaaaatgagaaacattttaaagcattatttacgaaaaaa
    ctgtctcaactattcttaagtactgtattgatattgtttgtatcttttattaatgttcta
    ccactttttatagatttgcatcttcctgtcacagggatgtggggaaatacgttttcctcc
    caagagaaccaagtttattatagactcctttattcagtgaaatggcttataatccactag
    ttgccatatttttgctaaaatatttctaaccaagaatactacttacatattgttttggct
    ttgttttatttttgatgcagttttttttagttgaggtaatgtaatatattgatgttttcc
    tttgtgtctaagattgatttataatagtaggtttgtataatttggaacattttccatgcc
    ttgcgaatttccttaattgaggatagggcttacacactttaagaaaacagtgagtacttg
    aacatttaaagggacagtgcaatttatagtcataatcacattgaatactgtatttgatct
    ttggagacttaggcaagcacagagctgggatatttatgctcagttgagcactttaagatg
    aattttaagtgagatgatttcttgcttaaaactcagaaagtcaaaagagtttcagctttc
    cttacagaaaaggaaggatcttgggccctagatcttggggattaacctctgcatataaga
    tttactcttaataggccagacgtggtgctcacgcctgtaatcccagtactttgggaggct
    gagacgggcagatcacttgaggtcaggagttcaagaccagcctggccaatatggtgaaac
    cccgtttctactaaaaatacaaaaaaaattacccaggcactcactcttgaggtaactaac
    caactcccacgataatgacagtccattcatgagcgcaaaggcctcatgacctaatggcac
    acacctgtaatcccaactgcttgggaggctgaggcgagaggattgcttgaacctgggagg
    cagaggttgcagtgagccgagatcgcaccactgcactccagtctgggcaacagagtgaga
    cttcatctcaaaaaaagtaaaaaaaaagatttaatataatcactgaagatctctattata
    gatagattaggtttttgacattggaaacatacttagggatagatttgtcctaaaggaaaa
    aagtaggcccgggcagattaaatgtcttgtgtaaagtcacacattaaattcagtcacaca
    ttaaattcatagagttttaaatgtttaatgtatataaaccagtttctttatacacatttg
    ggaaaacattggtctcacagattaaatgattaactaactgacccaggaactagttgtagc
    tttctaagtaattaggcaattacagttattgcctgtaaccaaaggtaataaaacaaaatg
    acaagtacatgtttaaaattatgaggcaatgagaaataatttaaaaaccaattttctagt
    tataatttaaaatttggagagcatttttaacagtaattaatccagaggtggctcaaattg
    agtataagaattaagattatttaaaatactgcatgtctaccttctcggggatcatacttt
    ataacactttctgcttcagtagctcttcatagcttgccaagtatgctcccatattttctc
    tctcgtgcctcgcaaatgaaagtcagataggctgggaactcatggggcagccctcagact
    tcaatgtgggcttcaaatccagtttcctgttctatatggtgctacatctttccagaaaat
    ttccctcagagcccctcgccaaaacaaagcattattttgaccctgcatgctatttcttta
    gctgtaggtgatagattagaacttctgtcagacatgttaatgacaaacataccaacagac
    aataaccaaagcaaatgtttccttcaagtgtgaaatgtgcaggggctcgtgggcaaggat
    gtattggcacactgtcctcttgaactgatagtgtcccaqcaatgttggaggttggcacca
    ttcctggtccgacacttgaggacctgagagacatcaggtttagaatgagccaaagaaatc
    ctacaagatggggagaattggtgtgcagcagcctaagtgttatagttaagtctaaagaag
    tatgaaagatcccctgtgttctctaaattgagcagaggggcctgcctaccaatatcactt
    tttaggggactgaaccattgcaggttagacttggcttccaaagagtctgcctaagccagg
    ggtggcagggtaggccatcatagctggatggcctcaaaagcagatgggggcagacttgcc
    ctcgtgatgccaggatttgagaggcagagtttctagagggagaccagtgctgcctctcac
    agtggcagttttttctctttgcaagaggaggggctgttcaattccatagaccagtgggca
    gatagccagttgaatactctgtgcatggtttgatcctttattagttcgctctaatatttt
    tctgtagatccttttgtcctggactcaaaatctaatccatgcattgtatgataccgtagc
    tctcctaaggtttgtgtttccttcaaaatgttttagttttcttcaactaaatttgatttt
    tgctgttagaagtgacatatttttatggtatacactatgttccttttttctactgcgagt
    caattttttgaattttcgtgagaaagaatatatctacaaattgcacgaaagtatcataaa
    aacagtactctagagcagcgctgtccaatagaaatataatctgagccacatgtataattt
    tattttcttctagccacattaaagaagtaaaaagatacaagtagaactaattttaatgtt
    ttaattcagtatatccaaaatatcatttgaacatgtaattaatataaaattattaatgtg
    atattttacattcttttggtaatactagtcttcaaaatctggtatgtatcttacattgat
    agcacatctcactttgtactagccacattgcaagtgctcagtagccacatgtggctagtg
    gctactgcactggacagcacagttctaggttccaccctaacacccaagtcctgtggatta
    gaatcccagaatcagagctggaagtaaacatagagatcaaacctccttttaaaaatgagg
    acgctgaggcacagagtttaaatggcttgcatgaggtcatacagctaaattcagcctcaa
    cagggtcttctgattccaggcactcttcccactccactacattactgtagtggtaattct
    tagggttaaaaaaagtgtagagtaggccgggcgcagtggctcatgcctgtaatcccagca
    ctttgggaggccgaagtgggcggatcacgaggtcagg&gatcgagaccatcctggccaac
    atggtgaaaccccgtctctactgaaaatacaaagcaaaattagccaggtgtggtggcggg
    cgcctgtggtcccagctgctctggaggctgaggcagaatggcgtgaacccaggaggcaga
    gatggcagtgagccaagatcgcgccactgcaccccagcctgggcgacagagcgagactcc
    atctcaaaaaaaaaaaaaaaaaaaaaagaaaagaaaagaaaagtctagagaacattatat
    taagtggttattattgaagtagaccaaagtttataccataaggatatttttccttaaata
    ccatgtttgaagaacaattatttattgatccttgaatctgtaagatcaaataacaagtct
    ctatccatgttaccaaatttaaccttttgaaaataataaactttaaaatatcagatgtgt
    tattacaggatgatacttggaatcaagtgaaatgagttatatggtcatcactaaatttag
    aaatctattgtgaaacaaagacaaacaggaaagtacagaatagagacttttagtaaataa
    atggaatttaaaagaaagtgtttatttacagtgtcacgacagaaaaggatgtctttgttg
    tcatagtctttgagggatctccgtaaaatctggggcacaggtacaagaaatagccaatat
    ttagttcccagaccatgtttagtagtgtccagtttcagatcatgctgccaagaggtatct
    ccccctcaggtgggtcatcactgagccctggaattggagactcatacttgcccagcacaa
    tgttacgggcagacaggccgacatctatgattagctagaagccataaagaaaagctgcta
    agtggccactaggtgccacttttctgtttttgtaatgctttcattagcagatcttttttt
    tccaagctccatggggcctatgagaggcatttatgatttttgtgcctacaataagtcagc
    ctgtctggtgtgagttgttttatgagaaatgctttccaagggaggtctaggaagatcctg
    acacataagaactttggcttagagagctttccaggtgtagtgccaataaaaactgacctg
    gaaagaaaacctgcccagcacggaacatgctttctgaactcacttgagagtgtatggtgt
    atgtcacttctcatatattcttgagtttagatttgtcttttatacaatttttagctcttt
    tccagttcacttgtgctcgtctgtatattggtatttttaaatttttgtggtaaataatga
    aaagagtgaaattatattttataattactcatttgtagtttttttttttaatttaataaa
    cttcctccaaaaagtgctcccttaaaa
  • ARNT4 coding sequence in Table 7 is indicated by upper case letters (nucieotides [0076] 220 to 2025) with-the termination codon underlined.
  • Diagnosis and Treatment of Circadian Rhythm Disorders
  • ARNT4 is involved in regulating circadian rhythm, e.g., by forming a heterodimer with Clock, a protein that regulates the timing of fatigue and alertness. Individuals with circadian rhythm disorders are screened for mutations in the an ARNT4 gene product or ARNT4 gene, e.g,. by detecting restriction fragment length polymorphisms (RFLPs) or by PCR. Individuals with symptoms of circadian rhythm disorders and identified as having a mutated ARNT4 gene are treated by administering DNA encoding a normal ARNT4 gene product. For example, DNA containing the coding sequence of SEQ ID NO: [0077] 20 is administered to such individuals using standard gene therapy techniques described herein. Similarly, an abnormally low or high level of ARNT4 protein or transcript is detected in an individual suffering from such disorders, the levels can be normalized by antisense therapy to inhibit ARNT4 production or gene therapy to augment production. ARNT4 levels may also be altered to artificially regulate circadian rhythm, e.g., to induce long periods of sleep in patients to improve the healing process or in individuals travelling long distances such astronauts during space travel.
  • Antisense Therapy
  • Nucleic acids complementary to all or part of the human EPAS1 CDNA (GenBank Accession # U81984; SEQ ID NO: [0078] 7) may be used in methods for antisense treatment to inhibit expression of EPAS1. Nucleic acids complementary to all or part of the human ARNT4 cDNA (SEQ ID NO: 20) may be used in methods for antisense treatment to inhibit expression of ARNT4. Antisense treatment may be carried out by administering to a mammal, such as a human, DNA containing a promoter, e.g., an endothelial cell-specific promoter, operably linked to a DNA sequence (an antisense template), which is transcribed into an antisense RNA. Alternatively, as mentioned above, antisense oligonucleotides may be introduced directly into vascular cells. The antisense oligonucleotide may be a short nucleotide sequence (generally at least 10, preferably at least 14, more preferably at least 20 (e.g., at least 30), and up to 100 or more nucleotides) formulated to be complementary to a portion, e.g., the coding sequence, or all of EPAS1 MRNA or ARNT4 mRNA. For example, the sequence is complementary some or all of the C-terminal activation domain; alternatively, the sequence may be complementary to all or part of the N-terminal DNA binding domain. The antisense sequence is complementary to DNA encoding residues 75 to 128, inclusive, of SEQ ID NO: 19; the antisense sequence. Alternatively, the antisense sequence is complementary to DNA encoding residues 155 to 207, inclusive, of SEQ ID NO: 19, or encoding residues 232 to 384 of SEQ ID NO: 19.,
  • Standard methods of administering antisense therapy have been described (see, e.g., Melani et al., 1991, Cancer Res. 51:2897-2901). Following transcription of a DNA sequence into an antisense RNA, the antisense RNA binds to its target nucleic acid molecule, such as an MRNA molecule, thereby inhibiting expression of the target nucleic acid molecule. For example, an antisense sequence complementary to a portion or all of EPAS1 MRNA could be used to inhibit the expression of EPAS1, thereby decreasing the level of transcription of angiogenic factors such as VEGF or VEGF-Rs, which in turn leads to a decrease in new blood vessel formation. Oligonucleotides complementary to various portions of EPAS1 mRNA or ARNT4 MRNA can readily be tested in in vitro for their ability to decrease production of their respective gene products, using assays similar to those described herein. Sequences which decrease production of EPAS1 message or ARNT4 message in vitro cell-based or cell-free assays can then be tested in vivo in rats or mice to determine whether blood vessel formation is decreased. [0079]
  • Preferred vectors for antisense templates are viral vectors, including those derived from replication-defective hepatitis viruses (e.g., HBV and HCV), retroviruses (see, e.g., WO 89/07136; Rosenberg et al., 1990, N. Eng. J. Med. 323(9):570-578), adenovirus (see, e.g., Morsey et al., 1993, J. Cell. Biochem., Supp. 17E,), adeno-associated virus (Kotin et al., 1990, Proc. Natl. Acad. Sci. USA 87:2211-2215,), replication defective herpes simplex viruses (HSV; Lu et al:, 1992, Abstract, page 66, Abstracts of the Meeting on Gene Therapy, Sept. 22-26, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y.), and any modified versions of these vectors. The invention may utilize any other delivery system which accomplishes in vivo transfer of nucleic acids into eucaryotic cells. For example, the nucleic acids may be packaged into liposomes, receptor-mediated delivery systems, non-viral nucleic acid-based vectors, erythrocyte ghosts, or microspheres (e.g., microparticles; see, e.g., U.S. Pat. No. 4,789,734; U.S. Pat. No. 4,925,673; U.S. Pat. No. 3,625,214; Gregoriadis, 1979, Drug Carriers in Biology and Medicine, pp. 287-341 (Academic Press,). Alternatively, naked DNA may be administered. Delivery of nucleic acids to a specific site in the body for antisense therapy may also be accomplished using a biolistic delivery system, such as that described by Williams et al., 1991, Proc. Natl. Acad. Sci. USA 88:2726-2729. [0080]
  • Antisense oligonucleotides may consist of DNA, RNA, or any modifications or combinations thereof. As an example of the modifications that the oligonucleotides may contain, inter-nucleotide linkages other than phosphodiester bonds, such as phosphorothioate, methylphosphonate, methylphosphodiester, phosphorodithioate, phosphoramidate, phosphotriester, or phosphate ester linkages (Uhlman et al., 1990, Chem. Rev. 90(4) :544-584; Anticancer Research, 1990, 10:1169) may be present in the oligonucleotides, resulting in their increased stability. oligonucleotide stability may also be increased by incorporating 3′-deoxythymidine or 2′-substituted nucleotides (substituted with, e.g., alkyl groups) into the oligonucleotides during synthesis, by providing the oligonucleotides as phenylisourea derivatives, or by having other molecules, such as aminoacridine or poly-lysine, linked to the 3′ ends of the oligonucleotides e.g., Anticancer Research, 1990, 10:1169-1182). Modifications of the RNA and/or DNA nucleotides may be present throughout the oligonucleotide, or in selected regions of the oligonucleotide, e.g., in the 5′ and/or 3′ ends. The antisense oligonucleotides may also be modified so as to increase their ability to penetrate the target tissue by, e.g., coupling the oligonucleotides to lipophilic compounds. Antisense oligonucleotides based on the human EPAS1 nucleotide sequence (SEQ ID NO: [0081] 7) or the human ARNT4 nucleotide sequence (SEQ ID NO: 20) can be made by any method known in the art, including standard chemical synthesis, ligation of constituent oligonucleotides, and transcription of DNA complementary to the all or part of the EPASI CDNA or ARNT4 CDNA.
  • EPAS1 is naturally expressed in vascular endothelial cells. These cells are, therefore, the preferred cellular targets for antisense therapy. Targeting of antisense oligonucleotides to endothelial cells is not critical to the invention, but may be desirable in some instances, e.g. systemic administration of antisense compositions. Targeting may be achieved, for example, by coupling the oligonucleotides to ligands of endothelial cell surface receptors. Similarly, oligonucleotides may be targeted to endothelial cells by being conjugated to monoclonal antibodies that specifically bind to endothelial-specific cell surface proteins. Antisense compositions may also be administered locally, e.g., at the site of an atherosclerotic lesion or at the site of a tumor. [0082]
  • Therapeutic applications of antisense oligonucleotides in general are described, e.g., in the following review articles: Le Doan et al., Bull. Cancer 76:849-852, 1989; Dolnick, Biochem. Pharmacol. 40:671-675, 1990; Crooke, Annu. Rev. Pharmacol. Toxicol. 32, 329-376, 1992. Antisense nucleic acids may be used alone or combined with one or more materials, including other antisense oligonucleotides or recombinant vectors, materials that increase the biological stability of the oligonucleotides or the recombinant vectors, or materials that increase the ability of the therapeutic compositions to penetrate endothelial cells selectively. [0083]
  • Therapeutic compositions, e.g., inhibitors of EPAS1 and/or ARNT4 transcription or transactivating function, may be administered in pharmaceutically acceptable carriers (e.g., physiological saline), which are selected on the basis of the mode and route of administration and standard pharmaceutical practice. Suitable pharmaceutical carriers, as well as pharmaceutical necessities for use in pharmaceutical formulations, are described in Remington's Pharmaceutical Sciences, a standard reference text in this field, and in the USP/NF. The compound may be administered with intravenous fluids as well as in combination with other anti-inflammatory agents, e.g., antibiotics; glucocorticoids, such as dexamethasone (Dex), or other chemotherapeutic drugs for the treatment of atherosclerotic lesions and tumors, respectively. [0084]
  • A therapeutically effective amount is an amount which is capable of producing a medically desirable result in a treated animal. As is well known in the medical arts, dosage for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently. Dosages will vary, but a preferred dosage for intravenous administration of DNA is approximately 10[0085] 6 to 1022 copies of the DNA molecule. The compositions of the invention may be administered locally or systemically. Administration will generally be parenterally, e.g., intravenously. As mentioned above, DNA may also be administered directly to the target site, e.g., by biolistic delivery to an internal or external target site or by catheter to a site in an artery.
  • Gene Therapy
  • Compositions which enhance intracellular production of EPAS1 (or its binding to a cis-acting regulatory region of a gene encoding VEGF or a VEGF-R) or ARNT4 may be used in methods to promote new blood vessel formation, e.g., to promote angiogenesis in wound healing (e.g., healing of broken bones, burns, diabetic ulcers, or traumatic or surgical wounds) and organ transplantation. Such compounds may be used to treat peripheral vascular disease, cerebral vascular disease, hypoxic tissue damage (e.g., hypoxic damage to heart tissue), or coronary vascular disease as well as to treat patients who have, or have had, transient ischemic attacks, vascular graft surgery, balloon angioplasty, frostbite, gangrene, or poor circulation. [0086]
  • Since EPAS1 and ARNT4 are nuclear proteins, a preferred method of increasing the levels of these proteins or polypeptides in a cell (to increase transcription of such angiogenic factors as VEGF or VEGF-Rs) is intracellular expression of recombinant EPAS1 or ARNT4 or active fragments thereof, e.g., transactivating fragments. DNA encoding EPAS1 or ARNT4 is administered alone or as part of an expression vector as described above. The DNA introduced into its target cells, e.g., endothelial cells at an anatomical site in need of angiogenesis, directs the production of recombinant EPAS1 or ARNT4 or fragments thereof in the target cell, to enhance production of new blood vessels. For inhibition of angiogenesis, gene therapy are also used to introduce administer DNA encoding a dominant negative mutant of EPAS1 such as DNA encoding a polypeptide with the amino acid sequence of SEQ ID NO: [0087] 4 or a polypeptide with the amino acid sequence of residues 486-639 of SEQ ID NO: 6.
  • Antibodies and Intrabodies
  • Anti-EPAS1 antibodies were obtained using techniques well known in the art. Such antibodies can be polyclonal or monoclonal. Polyclonal antibodies can be obtained, for example, by the methods described in Ghose et al., Methods in Enzymology, Vol. 93, 326-327, 1983. An EPAS1 polypeptide, or an antigenic fragment thereof, was used as the immunogen to stimulate the production of EPAS1-reactive polyclonal antibodies in the antisera of animals such as rabbits, goats, sheep, rodents and the like. EPAS1-specific antibodies were raised by immunizing animals with a C-terminal EPAS1 polypeptide spanning amino acids [0088] 668-829 of human EPAS (PGGSTSHLMWKRMKNLRGGSCPLMPDKPLSANVPNDKFTQNPMRGL HPLRHLPLPQPPSAISPGENSKSRFPPQCYATQYQDYSLSSAHKVSGMASRLLGP; (SEQ ID NO: 17) and a C-terminal EPAS polypeptide spanning amino acids 641-875 of mouse EPAS1 DPPLHFGPTKWPVGDQSAE SLGALPVGSWQLELPSAPLHVSMFKMRSAKDFGARGPYMMSPAMIALSNK LKLKRQLEYEEQAFQDTSGGDPPGTSSSHLMWKRMKSLMGGTCPLMPDKT ISANMAPDEFTQKSMRGLGQPLRHLPPPQPPSTRSSGENAKTGFPPQCYA SQFQDYGPPGAQKVSGVASRLLGPSFEPYLLPELTRYDCEVNVPVPGSST LLQGRDLLRALDQAT (SEQ ID NO: 18).
  • Monoclonal antibodies are obtained by the process described by Milstein and Kohler in Nature, 256:495-97, 1975, or as modified by Gerhard, Monoclonal Antibodies, Plenum Press, 1980, pages 370-371. Hybridomas are screened to identify those producing antibodies that are highly specific for an EPAS1 polypeptide. Preferably, the antibody will have an affinity of at least about 10[0089] 8 liters/mole and more preferably, an affinity of at least about 109 liters/mole. Monoclonal antibodies can be humanized by methods known in the art, e.g, MAbs with a desired binding specificity can be commercially humanized (Scotgene, Scotland; Oxford Molecular, Palo Alto, Calif.).
  • Following identification of a hybridoma producing a suitable monoclonal antibody, DNA encoding the antibody is cloned. DNA encoding a single chain EPAS1-specific antibody in which heavy and light chain variable domains (separated by a flexible linker peptide such as Gly[0090] 4-Ser3 (SEQ ID NO: 16) is cloned into an expression vector using known methods (e.g., Marasco et al., 1993, Proc. Natl. Acad. Sci. USA 90:7889-7893 and Marasco et al., 1997, Gene Therapy 4:11-15) Such constructs are introduced into cells, e.g., using gene therapy techniques described herein, for intracellular production of the antibodies. Intracellular antibodies, i.e., intrabodies, are used to inhibit binding of endogenous EPAS1 to its target DNA (e.g., cis-acting regulatory sequences of genes encoding VEGF or VEGF-Rs), which in turn, decreases production of these angiogenic factors and decreases new blood vessel formation in the treated mammal. Intrabodies which bind to a C-terminal transactivation domain of EPAS1 inhibit the ability of EPAS1 to induce transcription of a gene encoding an angiogenic factor such as VEGF or a VEGF-R. A similar strategy is used to make intrabodies which bind to intracellular ARNT4. Such intrabodies bind to ARNT4 and prevent heterodimeriation with EPAS1, and as a result, inhibit transactivation of the VEGF promoter. Inhibition of VEGF promoter activity, in turn, leads to inhibition of new blood vessel formation.
  • Other embodiments are within the following claims.[0091]

Claims (56)

What is claimed is:
1. A method of inhibiting angiogenesis in a mammal comprising administering to said mammal a compound which inhibits binding of endothelial PAS domain protein-1 (EPAS1) to cis-acting transcription regulatory DNA of an angiogenic factor.
2. The method of claim 1, wherein said angiogenic factor is a vascular endothelial growth factor receptor (VEGF-R).
3. The method of claim 2, wherein said receptor is KDR/flk-1.
4. The method of claim 2, wherein said receptor is flt-1.
5. The method of claim 1, wherein said angiogenic factor is vascular endothelial growth factor (VEGF).
6. The method of claim 1, wherein said angiogenic factor is Tie2.
7. The method of claim 1, wherein said compound inhibits transcription of said angiogenic factor.
8. The method of claim 1, wherein said regulatory DNA comprises GCCCTACGTGCTGTCTCA (SEQ ID NO:1).
9. The method of claim 1, wherein said compound is an EPAS1 polypeptide lacking a transactivation domain.
10. The method of claim 9, wherein said transactivation domain comprises the amino acid sequence of SEQ ID NO: 2.
11. The method of claim 9, wherein said transactivation domain comprises the amino acids 486-639 of SEQ ID NO: 6.
12. The method of claim 9, wherein said polypeptide comprises the amino acid sequence of SEQ ID NO: 4.
13. The method of claim 1, wherein said compound is a nucleic acid encoding an EPAS1 polypeptide lacking the amino acid sequence of SEQ ID NO: 2.
14. The method of claim 1, wherein said compound is a nucleic acid encoding an EPAS1 polypeptide lacking amino acids 486-639 of SEQ ID NO: 6.
15. The method of claim 1, wherein said compound is a antisense nucleic acid molecule comprising at least 10 nucleotides, wherein the sequence of said molecule is complementary to part of or all ofan MRNA encoding EPAS1 polypeptide.
16. The method of claim 1, wherein said compound is an EPAS1-specific intrabody.
17. The method of claim 1, wherein said compound is administered to a site of an atherosclerotic lesion in said mammal.
18. The method of claim 1, wherein said compound is administered to a tumor site in said mammal.
19. An antibody which binds to EPAS1 .
20. The antibody of claim 19, wherein said antibody binds to a C-terminal activation domain of EPAS1.
21. The antibody of claim 20, wherein said activation domain comprises SEQ ID NO: 2.
22. A method of promoting angiogenesis in a mammal comprising administering to said mammal a compound which increases expression of VEGF or a VEGF-R in an endothelial cell.
23. The method of claim 19, wherein said VEGF-R is KDR/flk-1 or flt-1.
24. A substantially pure DNA comprising a sequence encoding a aryl hydrocarbon receptor nuclear translocator-4 (ARNT4) polypeptide.
25. The DNA of claim 24, wherein said DNA encodes a human ARNT4 polypeptide.
26. The DNA of claim 24, wherein said polypeptide comprises the amino acid sequence of residues 75 to 128, inclusive, of SEQ ID NO: 19.
27. The DNA of claim 24, wherein said polypeptide comprises the amino acid sequence of residues 155 to 207, inclusive, of SEQ ID NO: 19.
28. The DNA of claim 24, wherein said polypeptide comprises the amino acid sequence of residues 232 to 384, inclusive, of SEQ ID NO: 19.
29. A substantially pure DNA comprising a nucleotide sequence having at least 50% sequence identity to SEQ ID NO: 20.
30. The DNA of claim 24, wherein said DNA comprises the coding sequences of SEQ ID NO: 20.
31. The DNA of claim 24, wherein said polypeptide comprises the amino acid sequence of SEQ ID NO: 19.
32. A substantially pure DNA comprising a strand which hybridizes at high stringency to a strand of DNA consisting of the coding sequence of SEQ ID NO: 20, or the complement thereof.
33. A substantially pure DNA comprising a sequences at least 50% sequence identity to the coding sequence of SEQ ID NO: 20, and encoding a polypeptide having the biological activity of an ARNT4 polypeptide.
34. A substantially pure ARNT4 polypeptide.
35. The polypeptide of claim 34, wherein said polypeptide is human ARNT4.
36. The polypeptide of claim 34, wherein said polypeptide comprises the amino acid sequence of residues 75 to 128, inclusive, of SEQ ID NO: 19.
37. The polypeptide of claim 34, wherein said polypeptide comprises the amino acid sequence of residues 155 to 207, inclusive, of SEQ ID NO: 19.
38. The polypeptide of claim 34, wherein said polypeptide comprises the amino acid sequence of residues 232 to 384, inclusive, of SEQ ID NO: 19.
39. The polypeptide of claim 34, wherein said polypeptide comprises the amino acid sequence of SEQ ID NO: 19.
40. The polypeptide of claim 34, wherein said polypeptide comprises an amino acid sequence at least 50% identical to SEQ ID NO: 19.
41. The polypeptide of claim 34, wherein said polypeptide comprises the amino acid sequence of SEQ ID NO: 19.
42. A vector comprising the DNA of claim 24.
43. A host cell comprising the DNA of claim 24.
44. A transgenic non-human animal the germ cells and nucleated somatic cells of which comprise a null mutation in a gene encoding ARNT4.
45. A method of inhibiting angiogenesis in a mammal comprising administering to said mammal a compound which inhibits binding of EPAS1 to ARNT4.
46. The method of claim 45, wherein said compound is an ARNT4 polypeptide.
47. The method of claim 45, wherein said polypeptide comprises the amino acid sequence of residues 75 to 128, inclusive, of SEQ ID NO: 19.
48. The method of claim 45, wherein said polypeptide comprises the amino acid sequence of residues 155 to 207, inclusive, of SEQ ID NO: 19.
49. The method of claim 45, wherein said polypeptide comprises the amino acid sequence of residues 232 to 384, inclusive, of SEQ ID NO: 19.
50. The method of claim 45, wherein said polypeptide comprises the amino acid sequence of SEQ ID NO: 19.
51. An EPAS1 polypeptide lacking a transactivation domain.
52. The polypeptide of claim 51, wherein said transactivation domain comprises the amino acid sequence of SEQ ID NO: 2.
53. The polypeptide of claim 51, wherein said transactivation domain comprises the amino acids 486-639 of SEQ ID NO: 6.
54. The polypeptide of claim 51, wherein said polypeptide comprises the amino acid sequence of SEQ ID NO: 4.
55. A nucleic acid encoding an EPAS1 polypeptide lacking the amino acid sequence of SEQ ID NO: 2.
56. The nucleic acid of claim 55, wherein said nucleic acid encodes an EPASI polypeptide lacking amino acids 486-639 of SEQ ID NO: 6.
US10/121,235 1998-08-14 2002-04-12 Methods of modulating of angiogenesis Abandoned US20030032609A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/121,235 US20030032609A1 (en) 1998-08-14 2002-04-12 Methods of modulating of angiogenesis

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US9651598P 1998-08-14 1998-08-14
US09/374,454 US6395548B1 (en) 1998-08-14 1999-08-13 Methods of modulating of angiogenesis
US10/121,235 US20030032609A1 (en) 1998-08-14 2002-04-12 Methods of modulating of angiogenesis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/374,454 Continuation US6395548B1 (en) 1998-08-14 1999-08-13 Methods of modulating of angiogenesis

Publications (1)

Publication Number Publication Date
US20030032609A1 true US20030032609A1 (en) 2003-02-13

Family

ID=22257713

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/374,454 Expired - Fee Related US6395548B1 (en) 1998-08-14 1999-08-13 Methods of modulating of angiogenesis
US10/121,235 Abandoned US20030032609A1 (en) 1998-08-14 2002-04-12 Methods of modulating of angiogenesis

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/374,454 Expired - Fee Related US6395548B1 (en) 1998-08-14 1999-08-13 Methods of modulating of angiogenesis

Country Status (3)

Country Link
US (2) US6395548B1 (en)
AU (1) AU5562999A (en)
WO (1) WO2000009657A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070286848A1 (en) * 2003-06-05 2007-12-13 Guy Louis-Georges Epas1 Gene Transfer to Improve Cell Therapy
WO2013074814A2 (en) * 2011-11-15 2013-05-23 University Of Utah Research Fourdation Morpholinos, morpholino upregulating, and associated methods
WO2013159051A1 (en) * 2012-04-19 2013-10-24 University Of Utah Research Foundation Morpholino-mediated increase in soluble flt-1 expression results in decreased ocular and tumor neovascularization

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6902890B1 (en) 1999-11-04 2005-06-07 Diadexus, Inc. Method of diagnosing monitoring, staging, imaging and treating cancer
CN1311218A (en) * 2000-03-02 2001-09-05 上海博德基因开发有限公司 New polypeptide-human nucleotransfer protein 12 containing ATP/GTP combination structure zone and polynucleotide coding for said polypeptide
WO2001092578A2 (en) * 2000-05-26 2001-12-06 Board Of Trustees Of The University Of Illinois Reagents and methods for identifying and modulating expression of genes regulated by retinoids
SE0002551D0 (en) * 2000-07-06 2000-07-06 Pharmacia & Upjohn Ab Screening methods
US7214770B2 (en) 2000-07-06 2007-05-08 Angiogenetics Sweden Ab Screening Methods
DE10104584A1 (en) * 2001-02-01 2002-08-22 Gsf Forschungszentrum Umwelt Medane genes and proteins
JP3769465B2 (en) 2001-02-13 2006-04-26 独立行政法人科学技術振興機構 New clock gene Bmal2
AU2002257794B8 (en) * 2001-04-24 2007-02-15 D. Collen Research Foundation Use of hypoxia inducible factor 2alpha for curing neonatal respiratory distress syndrome and as a target for the treatment of pulmonary hypertension
CA2447958A1 (en) 2001-05-23 2002-11-28 Angiogene Inc. Hypoxia inducible factors and uses thereof for inducing angiogenesis and improving muscular functions
AU2002365374A1 (en) * 2001-11-28 2003-06-10 Angiogenetics Sweden Ab Regulation of hypoxia-inducible gene expression with antisense inhibitory pas domain protein
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
US7060684B1 (en) 2002-12-16 2006-06-13 Quijano Rodolfo C Device for treating diabetes and methods thereof
FR2865736B1 (en) * 2004-02-02 2006-07-14 Synt Em INHIBITORS OF ANGIOGENESIS, COMPOSITIONS CONTAINING SAME AND USE THEREOF FOR THE TREATMENT OF DISEASES ASSOCIATED WITH DEREGULATION OF ANGIOGENESIS
FR2865735B1 (en) * 2004-02-02 2006-08-11 Synt Em INHIBITORS OF ANGIOGENESIS, COMPOSITIONS CONTAINING SAME AND USE THEREOF FOR THE TREATMENT OF DISEASES ASSOCIATED WITH DEREGULATION OF ANGIOGENESIS
CN100418574C (en) * 2005-10-26 2008-09-17 王成球 chromatin peptide for blocking human HIF-1 alpha gene and regulating network downstream related gene
GB0818080D0 (en) 2008-10-02 2008-11-05 Isis Innovation Immunogenic peptides

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5843683A (en) 1993-07-14 1998-12-01 Brandeis University PAS domain proteins
US5786171A (en) 1995-06-22 1998-07-28 President And Fellows Of Harvard University Aortic preferentially expressed gene and uses thereof
US5846773A (en) 1995-06-22 1998-12-08 President And Fellows Of Harvard College Single gene encoding aortic-specific and striated-specific muscle cell isoforms and uses thereof
WO1997015301A1 (en) 1995-10-24 1997-05-01 Gruenenthal Gmbh Method of adjusting the circadian rhythm of a mammal
US5695963A (en) * 1997-01-17 1997-12-09 Board Of Regents, The University Of Texas System Endothelial PAS domain protein
US5874241A (en) 1997-03-13 1999-02-23 Northwestern University Clock gene and gene product

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070286848A1 (en) * 2003-06-05 2007-12-13 Guy Louis-Georges Epas1 Gene Transfer to Improve Cell Therapy
WO2013074814A2 (en) * 2011-11-15 2013-05-23 University Of Utah Research Fourdation Morpholinos, morpholino upregulating, and associated methods
WO2013074814A3 (en) * 2011-11-15 2013-08-15 University Of Utah Research Foundation Morpholinos, morpholino upregulating, and associated methods
US9534222B2 (en) 2011-11-15 2017-01-03 University Of Utah Research Foundation Morpholinos, morpholino upregulating, and associated methods
WO2013159051A1 (en) * 2012-04-19 2013-10-24 University Of Utah Research Foundation Morpholino-mediated increase in soluble flt-1 expression results in decreased ocular and tumor neovascularization
US9506069B2 (en) 2012-04-19 2016-11-29 University Of Utah Research Foundation Morpholino-mediated increase in soluble Flt-1 expression results in decreased ocular and tumor neovascularization

Also Published As

Publication number Publication date
US6395548B1 (en) 2002-05-28
AU5562999A (en) 2000-03-06
WO2000009657A2 (en) 2000-02-24
WO2000009657A3 (en) 2000-08-31

Similar Documents

Publication Publication Date Title
US6395548B1 (en) Methods of modulating of angiogenesis
US7838294B2 (en) Inhibitors of the JNK signal transduction pathway and methods of use
US6331527B1 (en) Promoter smooth muscle cell expression
US6620914B1 (en) Transcription factor islet-brain 1 (IB1)
JPH11507541A (en) Hypoxia-inducing factor-1 and method of use
US20050287530A1 (en) TCL-1b gene and protein and related methods and compositions
JP2002502590A (en) Novel molecules of the CARD-related protein family and uses thereof
US6323329B1 (en) Nucleic acid sequences of genes encoding high mobility group proteins
EP1115733A1 (en) Sequences characteristic of hypoxia-regulated gene transcription
US6514935B1 (en) Methods of treating hypertension
US6552177B2 (en) EH domain containing genes and proteins
WO1994005776A1 (en) Myocyte-specific transcription enhancing factor 2
WO1994005776A9 (en) Myocyte-specific transcription enhancing factor 2
US20030054446A1 (en) Novel retina-specific human proteins C7orf9, C12orf7, MPP4 and F379
JP2004502444A (en) Methods and compositions related to muscle selective calcineurin interacting protein (MCIP)
US6258557B1 (en) Smooth muscle cell LIM promoter
US7071318B2 (en) Methods and compositions for stabilizing microtubules and intermediate filaments in striated muscle cells
US7070957B2 (en) STARS—a muscle-specific actin-binding protein
US20030026793A1 (en) Hypoxia inducing factors and uses thereof for inducing angiogenesis and improving muscular functions
US7041497B2 (en) Nuclear factor κB inducing factor
US6268216B1 (en) Reagents and methods for the screening of compounds useful in the treatment of neurological diseases
US20030027152A1 (en) Generation of diagnostic tools to assay the human LHX3/P-LIM/LIM-3 factor
WO1997033900A1 (en) E2a-binding protein
US20030176649A1 (en) Vmglom gene and its mutations causing disorders with a vascular component
US20040009502A1 (en) Identification and tissue distribution of two novel spliced variants of the mouse LATS2 gene

Legal Events

Date Code Title Description
AS Assignment

Owner name: PRESIDENT AND FELLOWS OF HARVARD COLLEGE, MASSACHU

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LE, MU-EN;MAEMURA, KOJI;HSIEH, CHUNG-MING;REEL/FRAME:012796/0037;SIGNING DATES FROM 19991003 TO 19991004

AS Assignment

Owner name: PRESIDENT AND FELLOWS OF HARVARD COLLEGE, MASSACHU

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE NAME OF THE ASSIGNOR. FILED ON APRIL 12, 2002, RECORDED ON REEL 012796 FRAME 0037;ASSIGNORS:LEE, MU-EN;MAEMURA, KOJI;HSIEH, CHUNG-MING;REEL/FRAME:012881/0084;SIGNING DATES FROM 19991003 TO 19991004

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NIH - DEITR, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:PRESIDENT AND FELLOWS OF HARVARD COLLEGE;REEL/FRAME:048451/0294

Effective date: 20190226